You are on page 1of 18

755629

research-article2018
JBXXXX10.1177/2472555218755629SLAS DISCOVERY: Advancing Life Sciences R&DBröer

Review
SLAS Discovery

Amino Acid Transporters as Disease


2018, Vol. 23(4) 303­–320
© 2018 Society for Laboratory
Automation and Screening
Modifiers and Drug Targets DOI: 10.1177/2472555218755629
https://doi.org/10.1177/2472555218755629
journals.sagepub.com/home/jbx

1
Stefan Bröer

Abstract
Amino acids perform a variety of functions in cells and organisms, particularly in the synthesis of proteins, as energy
metabolites, neurotransmitters, and precursors for many other molecules. Amino acid transport plays a key role in all
these functions. Inhibition of amino acid transport is pursued as a therapeutic strategy in several areas, such as diabetes
and related metabolic disorders, neurological disorders, cancer, and stem cell biology. The role of amino acid transporters
in these disorders and processes is well established, but the implementation of amino acid transporters as drug targets
is still in its infancy. This is at least in part due to the underdeveloped pharmacology of this group of membrane proteins.
Recent advances in structural biology, membrane protein expression, and inhibitor screening methodology will see an
increased number of improved and selective inhibitors of amino acid transporters that can serve as tool compounds for
further studies.

Keywords
solute carrier, diabetes, cancer, drug development

Introduction They are several aspects of human physiology and


pathophysiology where amino acid transporters are consid-
Amino acids are key metabolites that are essential for cells ered disease modifiers and drug targets (Fig. 1 and Table
and organisms. They form the building blocks for the syn- 1).5–9 These are
thesis of proteins, they are used as energy metabolites, and
they are or contribute to the biosynthesis of signaling mol- -  Metabolic disorders, such as diabetes and obesity
ecules, such as peptide hormones and neurotransmitters. -  Neurological disorders
They also contribute to the synthesis of many other mole- - Cancer
cules, such as nucleotides, creatine, polyamines, glucos- -  Embryo development and stem cells
amines, and related carbohydrates, and are the principal
generators of C1 compounds.1 Three approaches are generally used to influence amino
In view of this diversity of roles, it is not surprising that acid homeostasis (Fig. 1). First is the blockage of transport-
amino acid homeostasis is highly regulated and that all cells ers to induce amino acid restriction or starvation. This is
maintain a pool of the 20 proteinogenic amino acids at any used as a possible treatment to reduce the growth of cancer
given time.2 Mammalian cells can synthesize nonessential cells,10–12 but also to induce protein restriction as a novel
amino acids (alanine, arginine, asparagine, aspartate, cyste- attempt to treat diabetes.13 Second is the blocking of trans-
ine, glutamate, glutamine, glycine, proline, and serine), but porters to increase the concentration of amino acids in the
need to acquire essential amino acids from nutrition (histi- interstitial space. This is often used to treat neurological dis-
dine, isoleucine, leucine, lysine, methionine, phenylalanine, orders, where amino acids such as glutamate and glycine
threonine, tryptophan, and valine). Cellular protein, in par- act as neurotransmitters.7,8,14 Third is the use of transporters
ticular muscle protein, is the storage site for amino acids in
organisms. Most cells can survive several hours without
1
amino acids, inducing autophagy to degrade cellular protein Research School of Biology, College of Science, The Australian National
for the synthesis of essential proteins.3 Protein degradation University, Canberra, ACT, Australia
is combined with efficient mechanisms to recycle the gener- Received Oct 11, 2017, and in revised form Jan 4, 2018. Accepted for
ated amino acids; thus, net loss of amino acids is limited. publication Jan 5, 2018.
Transporters play a key role in the acquisition of amino
Corresponding Author:
acids by the whole organism from the nutrition and in the Stefan Bröer, Research School of Biology, College of Science, Building
acquisition of amino acids by individual cells from blood 134, The Australian National University, Canberra, ACT 2601, Australia.
plasma.2,4,5 Email: stefan.broeer@anu.edu.au
304 SLAS Discovery 23(4)

Organism
Brain cell
Increase
extracellular
AA concentraon C
Cellular
and signalling AA absorpon

O
Organismic Malignant cell
AA absorpon Figure 1.  Current concepts of
- Protein restricon amino acid transporters as drug
to improve metabolic Cellular AA absorpon
C targets. Reducing uptake of amino
health - Reduce cell growth acids in the intestine causes protein
restriction and can improve
metabolic health. Uptake into
cells can be blocked to increase
signaling, particularly when amino
acids serve as neurotransmitters
Stem cell/Embryo or neuromodulators. Blocking
uptake of essential amino acids can
be used to reduce the growth of
malignant cells. In vitro amino acid
Cellular AA absorpon supplementation and transporter
- Opmize AA composion modulation can be used to optimize
conditions for embryo transfer and
implantation.

Table 1.  Amino Acid Transporters Covered in This Review and Their Function..

Solute Carrier ID Common Name Function Potential Indication


SLC1A2 GLT-1 Glutamate reuptake at the synapse ALS
SLC1A5 ASCT2 Glutamine transport, intracellular amino acid Cancer
homeostasis
SLC6A5 GlyT2 Glycine reuptake at the synapse Neuropathic pain
SLC6A9 GlyT1 Glycine homeostasis at the synapse Schizophrenia
SLC6A14 ATB0,+ General amino acid transporter Cancer
SLC6A19 B0AT1 Neutral amino acid transporter Diabetes
SLC6A20 SIT1 Proline, betaine transporter In vitro fertilization
SLC7A2 CAT2B Cationic amino acid transporter Inflammation
SLC7A5 LAT1 Neutral amino acid transporter Cancer
SLC7A9 b0,+AT Cationic amino acid transporter Unclear
SLC7A11 xCT Glutamate/cystine transporter Cancer
SLC16A10 TAT1 Aromatic amino acid transporter Autoimmune disorders
SLC38A1 SNAT1 Neutral amino acid transporter Cancer
SLC38A2 SNAT2 Neutral amino acid transporter Cancer
SLC38A3 SNAT3 Glutamine transporter Diabetes
SLC38A4 SNAT4 Neutral amino acid transporter Diabetes
SLC38A5 SNAT5 Glutamine transporter Diabetes
SLC43A1 LAT3 BCAA transporter Cancer
Bröer 305

Figure 2.  Structure of amino


acid transporters and their
binding sites. (A) The outward-
open conformation of the leucine
transporter from Aquifex aeolicus
(LeuTAa) is shown (PDB ID 3TT1).
The cavity that leads to the
substrate binding site is visible in
the center of the molecule. (B) The
occluded state of LeuTAa (PDB ID
2A65) is shown from the side and
sliced to reveal the substrate binding
site, which encloses the substrate
(magenta) tightly. The vestibule is
visible at the top.

to selectively provide amino acids to certain cells. This large aromatic fragment, which blocks the transition of the
approach is used less frequently but is particularly interest- transporter to the occluded conformation. The substrate-
ing with regard to embryo development and stem cell biol- like fragment ensures recognition and specificity; the wedge
ogy.15–22 The latter could be complemented by compounds fragment provides additional interactions with the trans-
to induce transporter function or expression, but this has not porter to increase affinity and at the same time blocks adop-
been translated to date. tion of the transition state. This design is well suited to a
medicinal chemistry approach.34 However, transport inhibi-
tors based on substrate analogues have two potential prob-
Principles of Transporter Inhibition lems. First, inhibitors may have problems binding to the
Biomembrane transporters, including amino acid transport- transporter in vivo in the presence of high concentrations of
ers, are attractive drug targets for several reasons. First, they substrates. The combined concentration of amino acid sub-
are located at the cell surface and, similar to receptors, are strates of a specific transporter in blood serum can easily
easily accessible by drugs from the bloodstream.23 Notable reach 1 mM, against which a competitive inhibitor has to
exceptions are transporters in the brain, which are shielded find its target. Second, substrate-like inhibitors may show
by the blood–brain barrier against most drugs,24–26 and lack of isoform specificity. For instance, dl-threo-β-
transporters in organelles, such as lysosomes or synaptic benzoylaspartate (TBOA) inhibits all glutamate transporter
vesicles. Second, as part of their function, amino acid trans- isoforms (Fig. 3).35 Allosteric inhibitors, by contrast, are
porters have deep substrate binding pockets, which often not displaced by substrates and can be isoform specific. For
reach halfway through the membrane (Fig. 2A). The pres- instance, UCPH101 (Fig. 4A) is a specific inhibitor of the
ence of a substrate binding site with a vestibule allows man- glutamate transporter EAAT1, due to its binding between
ifold interactions and bond formation to stabilize binding of the scaffold and transporter domain of EAAT1 (Fig. 4B),
an inhibitor.27 In contrast to enzymes, where transition-state which has a less conserved sequence than the substrate
analogues are considered to be the best inhibitors,28 ground- binding site.36 Allosteric inhibitors are more likely to arise
state analogues are preferred as transport inhibitors.29 The from high-throughput screening (HTS) or specialized
ground state of a transporter for drug binding is the outside screens,37 but can be further modified through medicinal
open conformation.30 In contrast to the majority of enzymes chemistry. The betain-γ-amino butyric acid (GABA) trans-
where the substrate adopts a transition state through nonco- porter BGT1, for instance, can be specifically inhibited by
valent interactions, transporters form a transition state N-(1-benzyl-4-piperidinyl)-2,4-dichlorobenzamide (Fig.
around the substrate (induced fit), while the substrate 4A), which binds to the vestibule of the transporter without
remains in its original conformation.31,32 The transition state penetrating the binding site.38 Allosteric transporter inhibi-
is called the occluded conformation, in which the trans- tors have also been identified by screening cyclic peptide
porter protein fully encloses the substrate (Fig. 2B).33 Thus, libraries.39
only compounds that are very similar to the substrate itself
fit into this cavity. Accordingly, transport inhibitors typi- Methods to Identify Transporter
cally contain a substrate-like fragment to which a large
fragment is coupled, often containing aromatic rings.
Inhibitors
Examples of this design are shown in Figure 3. The ratio- Medicinal chemistry is one of the major tools to design trans-
nale behind this design is the wedge effect exerted by the port inhibitors, usually starting with one of the substrates of the
306 SLAS Discovery 23(4)

Figure 3.  Examples of transport inhibitors based on substrate analogues. Three amino acid transport inhibitors are shown. The
substrate-like part of each inhibitor is highlighted in magenta. Bulky side chains (black) prevent the occlusion of the substrate by the
transporter. See references 12, 129, and 181 for the depicted inhibitors.

Figure 4.  Allosteric inhibitors of amino acid transporters. (A) Structures of allosteric transport inhibitors are unrelated to the
substrate. Top: UCPH101, a specific EAAT1 inhibitor. Bottom: The BGT-1 inhibitor BPDBA is thought to bind an allosteric secondary
binding site observed in some transporters of the SLC6 family. (B) The binding site of the EAAT1 inhibitor (UCPH101) was resolved
by x-ray crystallography and is located at a distance from the substrate binding site.182 The substrate is shown in the center of the
transporter, while the inhibitor is located between the translocation and scaffold domain of the transporter (lower right). The
inhibitor glues the two domains together, thereby inhibiting transport.
Bröer 307

transporter or a close analogue. The reader is referred to a com- have been used for many years to study transport and ion
prehensive review of the medicinal chemistry of α-amino acids channels in membrane vesicles and adherent cells.45 The
for further information.34 To identify substrate-unrelated start- dyes are membrane permeable and therefore can be washed
ing points for medicinal chemistry, HTS is increasingly used in out. The assay has been improved by the addition of spe-
academic environments to identify transport inhibitors. The cific dyes to quench signals occurring outside the cell (U.S.
most frequently used techniques for transporters are briefly patent 6,420,183, EPO patent 0 906 572) and is available
described in the following. commercially as FLIPR Membrane Potential Assay. A cell
expressing the selected transporter is required, although
expression can be endogenous or heterologous. Endogenous
Accumulation of Radiolabeled Substrate transporters may interfere with the signal generated by het-
Although radiolabeled compounds are less frequently used in erologous expressed transporters. The assay has been suc-
HTS environments due to regulatory reasons, inhibition of cessfully used to screen for inhibitors of several amino acid
radiolabeled substrate uptake is still the most robust and quan- transporters, such as the neutral amino acid transporter
titative assay to study transport inhibitors. Moreover, it is the SLC6A19,46 glycine transporters GlyT147 and GlyT2,48 and
gold standard to identify false-positive hits from other screen- glutamate transporters EAAT1–3.49 In the case of SLC6A19,
ing methods described below and to characterize the mode of several factors were important to optimize the signal. First,
inhibition. Typically, a cell line stably expressing the trans- heterologous expression of both SLC6A19 and its traffic
porter of choice is washed with isosmotic buffer, and subse- subunit TMEM27 was required for significant expression.
quently incubated with radiolabeled substrate for 5–30 min in CHO cells were used as a host cell line because it had no
the presence or absence of inhibitors. After washing the cells to measurable Na+-dependent uptake for isoleucine, one of the
remove extracellular radiolabel, the amount of radioactivity is preferred substrates of SLC6A19. While a tailored platform
measured in cell extracts. This method has been used to screen exists for the FLIPR assay, it can be implemented on any
for inhibitors of glycine transporter (GlyT1)40 and neutral suitable fluorescent plate reader.
amino acid transporter asc-1.41 The method can also be used in
conjunction with reconstituted protein in proteoliposomes, in
Ion-Sensitive Dyes
which case filtration is used to separate the proteoliposomes
from radiolabeled compounds after incubation.42 Three different sodium dyes have been evaluated in prostate
cancer cell lines, namely, benzofuran isophthalate (SBFI),
CoroNa Green (Corona), and Asante NaTRIUM Green-2
Scintillation Proximity Assay (ANG-2).50 While calibration was robust in the three cho-
In this assay, the radioactive substrate of a transporter is sen cell lines, the response of each cell line to changes in the
brought into proximity with a scintillant. Weak β-particles are extracellular Na+ concentration was quite different.
strongly quenched in water; thus, only radioactive compounds Inhibition of the Na+/K+-ATPase by Ouabain caused a sig-
in close proximity to the scintillant generate light flashes, nificant increase of intracellular Na+ that was readily
which can be recorded with a normal scintillation counter. detected by SBFI and ANG-2, but not by Corona.
Several variations of this assay are available, depending on Interestingly, the addition of veratridine to open Na+ chan-
whether cell lines, membrane preparations, or reconstituted nels did not result in a detectable increase of the intracellu-
transporters are used. Cells can be seeded out onto specific lar Na+ concentration. Although principally suitable for
multiwell plates with a scintillating base.43 Uptake of the radio- HTS, the author is not aware of any study that has used
labeled substrate into the cells brings the radioactivity into Na+-sensitive dyes to screen for inhibitors of Na+ cotrans-
proximity of the scintillant generating light emissions, which porters. The pH-sensitive dye BCECF has been used in
are recorded. In a variation of this procedure, suspended cells Caco-2 cells to monitor H+-peptide cotransport via the pep-
are brought into contact with polylysine-coated scintillation tide transporter SLC15A1.51 BCECF is a ratiometric dye
proximity assay (SPA) beads, also resulting in an uptake- and can be preloaded as an AM ester. The ratiometric mode
dependent scintillation signal.41 For membrane fragments or of measurement is useful when BCECF is leaking out of the
reconstituted protein, beads are used that are coated with poly- cells. BCECF has not been used in combination with amino
lysine to bind membrane lipids. Purified transporters in deter- acid transporters.
gent micelles have also been characterized by this method
using copper chelate–coated beads to immobilize a tyrosine
Biosensors
transporter through its His tag.44
Glutamine transport has been analyzed in living cells using
a genetically encoded biosensor, for instance, in Cos-7 cells
Membrane Potential–Sensitive Dyes transiently expressing the glutamine transporter SLC1A5.
These can only be applied to electrogenic transporters and The biosensor is based on the Escherichia coli glutamine
require significant transport activity. Voltage-sensitive dyes binding protein, glnH.52 Förster resonance energy transfer
308 SLAS Discovery 23(4)

(FRET) occurs between monomeric teal fluorescent protein Thus far, only low-affinity compounds have been identified
(mTFP) 1 and the yellow fluorescent protein venus. FRET- through computational screens for amino acid transport
based arginine biosensors have been generated based on the inhibitors, which require significant modification to
arginine repressor/activator ahrC gene from Bacillus subti- improve binding.46,60,61
lis53 and through protein engineering of bacterial amino
acid binding proteins.54 A biosensor for methionine and
Methods to Characterize Inhibitor Action in
branched-chain amino acids (BCAAs) has been developed
using the leucine-responsive protein Lrp of Corynebacterium Detail
glutamicum.55 The methods described above are frequently used to screen for
inhibitors. Further characterization of the mechanistic action of
Mass Spectrometry inhibitors can require additional methods. As outlined above,
noncompetitive inhibitors can provide better specificity and
Amino acid uptake via a transporter can be monitored by mass resistance against substrate competition. Electrophysiological
spectrometry (MS) either detecting accumulation of the natu- techniques, such as voltage-clamp recordings, are frequently
ral amino acid or using stable isotopes. Classical techniques, used to monitor electrogenic transporters, but on occasion can
such as gas chromatography–mass spectrometry (GC-MS) or also be used to monitor transport-associated currents in elec-
liquid chromatography–mass spectrometry (LC-MS), are less troneutral transporters. Electrophysiological techniques can be
suitable for large-scale inhibitor screening due to the slow applied to transfected cells and Xenopus laevis oocytes
chromatography step. With the development of high-resolu- expressing recombinant transporter mRNA. Rapid perfusion
tion mass spectrometers, complex mixtures of metabolites techniques and the use of caged compounds that can be
from cell extracts can be analyzed in a single step or following released upon light exposure are particularly valuable to ana-
simple extraction. Typically, electrospray ionization or matrix- lyze binding events during the transport cycle. Moreover, elec-
assisted laser desorption ionization are used in combination trophysiology can be combined with fluorescent reporter
with a quadrupole time-of-flight (Q-TOF) mass spectrometer groups that are attached to mobile parts of the transporter struc-
to analyze liquid samples directly or after extraction using ture. An extensive review of these methods has been compiled
solid-phase extraction cartridges. This methodology has been by Grewer et al.65 In the case of transporters, electrophysiolog-
used to identify novel inhibitors of P-glycoprotein.56 The ical methods report currents from a large number of transport-
development of echo acoustic liquid handling by Labcyte Inc. ers, particularly in oocytes. For reasons that are not fully
(San Jose, CA) will provide further improvement of sample elucidated, transporters expressed in X. laevis oocytes are often
preparation and injection for MS.57 refractory to inhibition by hydrophobic compounds, thus
resulting in IC50 values that are significantly higher than in cul-
Docking Studies tured cells.46,66,67 If purified protein is available, proteolipo-
somes can be used to study transporter inhibition.
Computational screening has become more feasible due to Proteoliposomes are obtained by detergent removal from lipo-
the increasing number of available transporter structures.58 some–detergent–protein mixtures.42,68 Due to the curvature of
Several obstacles need to be overcome for computational the vesicles, incorporation of protein is often asymmetric,
approaches to become competitive to HTS. First, very few allowing the design of efflux or influx experiments.
mammalian amino acid transporter structures have been More recently, single-molecule techniques have been
resolved, particularly in the outside open conformation, which developed to follow transport processes in individual mol-
is the most relevant structure for inhibitor identification.36,59 As ecules using FRET.69 Typically, these require the introduc-
a result, most docking studies rely on homology mod- tion of cysteine residues into cys-less or cys-reduced
els.46,60,61 Second, docking studies are still relatively slow. versions of the native transporter, which are then modified
Even with generous computation time, only small libraries with donor and acceptor fluorophores. These methods can
(tens of thousands of compounds) can be screened. Larger be used to study transport inhibitors. Inhibitors can provide
screens make use of a prescreen using a pharmacophore valuable insight for these studies because they can lock
similarity screen.62 This allows reducing very large libraries transporters in a particular state, which is recognizable due
to smaller compound libraries, but at the same time to the movement of fluorophores or the lack thereof.69
decreases the likelihood of finding novel structures. Third,
docking studies have a strong bias toward larger molecules,
because an increase of interaction surface automatically Role of Transporters in Nutritional
increases the docking score. Larger molecules often do not Disorders, Such as Diabetes and
comply with Lipinski’s rule of five, thereby reducing their
Obesity
suitability as starting points for medicinal chemistry.63
Moreover, molecular dynamics simulations are required to Several studies have reported an association between obe-
test accessibility of the compounds to the binding site.64 sity and polymorphisms in the gene encoding the general
Bröer 309

amino acid transporter SLC6A14.70–72 The contribution to coincidental or caused by poor nutrition. Thus, it appears
obesity of this transporter appears to be weak, with allele likely that inhibition of the transporter is generally safe in
frequencies showing only a few percent differences in nor- mature individuals with overnutrition.
mal and obese individuals. The result suggests only a small Through the use of computational approaches and screen-
influence of SLC6A14 on the obesity phenotype in general. ing assays based on membrane potential–sensitive dyes, benz-
A mechanism has not been established but could be related tropine was identified as an inhibitor of SLC6A19.46 Through
to amino acids contributing to appetite regulation in the the use of SLC6A19 containing proteoliposomes, nimesu-
pituitary gland.73 Alternatively, it could provide tryptophan lide was identified as another inhibitor of SLC6A19.88 Both
for serotonin production, a neurotransmitter involved in compounds have other molecular targets, and the IC50 val-
appetite control. Another scenario relates to the function of ues are such that they can only serve as research tool
SLC6A14 in fetal development.74 Reduced amino acid pro- compounds.
vision to the fetus could result in epigenetic programming, SLC7A5 is the major transporter for BCAAs in nonepi-
which in turn could foster inappropriate eating behavior thelial cells.89 It is an amino acid exchanger that can use intra-
later in life. cellular glutamine and other amino acids as an exchange
SLC6A19 is the major transporter for neutral amino substrate to import essential amino acids.2,90 Transport exper-
acids in the small intestine and in the proximal tubule of the iments performed in X. laevis oocytes suggest that glutamine
kidney.75 Elevated levels of BCAAs have been detected in is a rather poor substrate of SLC7A5; however, studies in cell
individuals with type 2 diabetes and are thought to be strong lines are consistent with a significant role of glutamine as an
predictors of future onset of type 2 diabetes.76–79 Mechanistically exchange substrate, due to its abundance in the cytosol.91 The
elevated levels of BCAAs are thought to cause insulin resis- transporter is expressed in β cells, and leucine uptake via
tance over the long term by a variety of mechanisms.80,81 SLC7A5 is important for activation of mTORC192 and also
Leucine is an important regulator of mTORC1, which stimulates the release of insulin as an allosteric activator of
through p70S6 kinase can cause insulin resistance by phos- glutamate dehydrogenase.93 Inhibition of SLC7A5 using
phorylation of insulin receptor substrate. Probably more BCH reduces mTORC1 activation and insulin release.
relevant is the effect of BCAA metabolites on insulin resis- However, BCH has also been reported to stimulate insulin
tance.76,77,79 Carnitine esters of branched-chain fatty acids release through the same mechanism as leucine.94
have been identified as possible causes of insulin resis- Amino acids play an important role in the modulation of
tance79 and branched-chain keto acids that are generated insulin release from pancreatic β cells,94,95 and transporters
after transamination of BCAAs appears to reduce GLUT4- are involved in this process.92 It is thought that amino acids
mediated uptake of glucose into muscle after insulin stimu- stimulate insulin secretion through ATP production inside
lation.82 On a broader scale, low-protein diets promote mitochondria. Glutamine and glutamate, in particular, are
general metabolic health and possibly longevity,83 and so readily metabolized inside mitochondria to produce ATP.
does methionine restriction.84 More importantly, gastric Leucine, in addition, acts as an allosteric inducer of gluta-
bypass surgery has dramatic effects on insulin sensitivity mate dehydrogenase, which provides intermediates for the
within days of surgery, long before significant weight loss tricarboxylic acid (TCA) cycle derived from glutamine. As a
occurs.85 At least some of these effects are thought to arise result, a combination of leucine and glutamine potently
from reduced protein uptake. Together, this suggests that increases insulin secretion.94 Glutamine can enter β cells via
reducing amino acid absorption could be an avenue to treat the glutamine transporter SLC38A3,96 while leucine is trans-
type 2 diabetes and obesity. SLC6A19 is the major trans- ported by SLC7A5 (see above). Expression of glutamate
porter for BCAAs and methionine in the intestine, and mice transporters is too low to support significant metabolism.97
lacking this transporter show a host of responses that are Glutamine uptake via SLC38A5 supports α-cell prolifer-
similar to those observed after gastric bypass surgery.13 The ation. Glucagon is produced in α cells of the pancreas acting,
main responses were improved glycemic control, reduced among other cells, on hepatocytes to induce gluconeogene-
plasma insulin, increased secretion of GLP-1 and GIP, sis. Glutamine and alanine are the main precursors in the
increased production of FGF-21, reduced plasma fatty acids early hours of fasting. In diabetes, elevated levels of fasting
and liver triglycerides, browning of subcutaneous adipose glucose are observed, suggesting an overstimulation of glu-
tissue, and increased ketone body production. FGF-21 coneogenesis. However, attempts to reduce glucagon signal-
could be an important factor of this metabolic phenotype ing cause hyperglucagonemia and α-cell hyperplasia.98
causing ketone body production from plasma and liver fatty Mice with a liver-specific lack of the glucagon receptor
acids, and browning of adipose tissue. Humans with muta- show a 3- to 7-fold increased proliferation of α cells, sug-
tions in SLC6A19 have Hartnup disorder, a largely benign gesting a factor that is released by hepatocytes to induce
syndrome that is sometimes associated with photosensitive α-cell proliferation.99 Systematic metabolomic analysis
skin rash.86,87 Very rarely has cerebellar ataxia been reported revealed that l-glutamine was the hepatocyte factor, act-
in young individuals with Hartnup disorder, which may be ing through mTOR signaling and the FoxP transcription
310 SLAS Discovery 23(4)

factor.21,22 Expression of SLC38A5 was upregulated in α and their extracellular levels are controlled by specific
cells from glucagon receptor knockout mice, while it was transporters. In the case of glycine, GlyT1 is thought to
barely detectable in wild-type mice. Blocking glucagon control glycine levels in the synaptic cleft.9 The main
signaling in the liver, by contrast, reduced the expression transporter for d-serine is the neutral amino acid trans-
of amino acid transporters SLC38A3, SLC38A4, and porter asc-1;102 smaller contributions are mediated by
SLC7A2, which mediate the import of amino acids for amino acid transporters SLC1A4 and SLC1A5.103 In both
gluconeogenesis.22 cases, blocking the uptake of glycine or d-serine would
As an overall strategy, it is conceivable that insulin increase the concentration of these allosteric modulators
secretion and hepatic glucose output can be modulated by in the synaptic cleft, thereby enhancing NMDA receptor
blocking amino acid transporters. This could be combined activity. Surprisingly, inhibitors of asc-1 decreased extra-
with transport inhibitors that cause methionine restriction. cellular d-serine levels, suggesting that this transporter is
rather involved in the efflux of d-serine from astro-
Role of Amino Acid Transporters in cytes.104,105 While modulation of d-serine levels in schizo-
phrenia requires more research, GlyT1 inhibitor bitopertin
Neurological Disorders has been used in clinical trials (up to clinical trial phase 3).9
GABA, glutamate, glycine, d-serine, and glutamine are Unfortunately, the results were inconclusive and no separa-
neuroactive amino acids, the modulation of which can be tion between placebo and bitopertin-treated patient groups
exploited to treat several neurological pathologies. was observed.106 Bitopertin was well tolerated, overcoming
Glycine transporters GlyT1 (SLC6A9) and GlyT2 concerns that complete inhibition of GlyT1 could result in
(SLC6A5) are the major transporters for the neurotransmitter severe motor and respiratory deficits, as observed in GlyT1
glycine in the brain. Pain is a normal response to injury or knockout mice.107
inflammation and is mediated by nociceptive neurons.8,14 Despite years of intensive research on glutamate trans-
Acute adaptive changes to these circuits also lead to hyper- porters in the brain, a promising pharmacological approach
sensitivity that serves a protective function during healing. has yet to emerge. Glutamate transporters are essential for
However, nerve injury in the peripheral or central nervous the removal of the excitatory neurotransmitter glutamate
system can lead to chronic pain or hypersensitivity that from the synaptic cleft. Modulation of glutamatergic neuro-
persists beyond the healing process. Glycine and GABA are transmission is involved in many neurological and psychi-
the major inhibitory neurotransmitters in the nervous system atric diseases.7 It is thought that elevated levels of glutamate
and are released to dampen excitatory neurotransmission, are neurotoxic due to overactivation of glutamate receptors,
which is used to transmit signals to the brain, including pain. which in turn causes excessive calcium influx into neurons,
Physiologically, neurotransmitters are rapidly removed resulting in cell damage and death. As a result, pharmaco-
from the synaptic cleft to terminate the signal. It is thought logical approaches using glutamate transporters rely on
that chronic pain is caused by a reduction of glycinergic compounds that enhance glutamate transport rather than
inhibition in the spinal cord, where glycine transporters are inhibiting it. In a broad screen, Rothstein et al. identified
particularly abundant.8 A broad variety of glycine transport β-lactam antibiotics as compounds that increase transcrip-
inhibitors have been generated, for both GlyT1 and GlyT2.14 tion of the gene encoding glutamate transporter EAAT2
Inhibitors based on sarcosine (N-methyl-glycine) are selec- (SLC1A2).108 Activators of EAAT2 translation were identi-
tive for GlyT1. However, GlyT1 is widely expressed in fied by using an enzyme-linked immunosorbent assay
astrocytes of the nervous system, and its inhibition enhances (ELISA) in combination with antibodies that bind to the
excitatory neurotransmission by increasing extracellular C-terminus of EAAT2.109 In the used cell line, EAAT2
glycine as a co-agonist at NMDA receptors.9 GlyT2 inhibi- expression was driven by a cytomegalovirus (CMV) pro-
tors ALX1393 and ORG25543 were shown in a mouse moter to increase the likelihood of identifying translational
model of neuropathic pain to have antiallodynia effects,100 activators. In amyotrophic lateral sclerosis (ALS), motor
that is, preventing an innocuous stimulus from becoming neurons gradually degenerate, resulting in muscle atrophy,
painful. weakness, and eventually respiratory failure and death. The
Schizophrenia is a devastating neuropsychiatric disorder etiology of neurodegeneration in ALS is not fully under-
characterized by positive symptoms (i.e., symptoms that stood, but glutamate excitotoxicity may contribute to the
healthy people do not experience, such as delusions and disease progression.110 A clinical trial was conducted to
hallucinations), negative symptoms (i.e., deficits compared evaluate whether enhanced expression of EAAT2 by ceftriax-
with healthy individuals, such as lack of emotion and moti- one would delay the functional decline in patients with ALS.110
vation), and cognitive deficits.9,101 The cause of this disease It appears that ceftriaxone showed some beneficial effects in
remains unclear, but reduced activity of NMDA receptors earlier stages of the disease, but no differences were observed
may be involved in the negative symptoms. NMDA recep- in overall survival. The translational activator 3-([(2-methyl-
tors are allosterically modulated by d-serine and glycine, phenyl)methyl]thio)-6-(2-pyridinyl)-pyridazine was able to
Bröer 311

protect neurons from excitotoxicity and delayed functional in general, can only synthesize the nonessential amino
decline in an animal model of ALS.111 acids, while essential amino acids must be acquired from
nutrition or blood supply in the case of cancer cells.
Role of Amino Acid Transporters in SLC1A5 is an amino acid exchanger for small neutral
amino acids, including serine, threonine, and glutamine.122 It
Cancer is upregulated in many cancer cells and functionally the dom-
Alteration of metabolism is almost universally observed in inant glutamine transporter in most cancer cell lines.122
cancer cell lines.112 These adaptations are governed by the Silencing of SLC1A5 reduces cell growth in some cancer cell
needs of an anabolic cell to generate sufficient biomass to lines123–126 but not in others.121,126 Related to this, SLC1A5
produce new cells. Protein forms the majority of new cell silencing reduces mTORC1 activity in some cell lines90,123
mass, and cancer cells therefore show enhanced rates of but not in others.121 Pharmacological studies have been ham-
amino acid uptake. In addition, amino acids are used for a pered by the poor selectivity of initially published inhibitors
wide variety of metabolic purposes in cancer cells. One of and by the fact that most of them are competitive binders and
the earliest approaches to combat cancer relied on antifo- are easily displaced by amino acids in media.60,121,127,128
lates, which are essential to provide one-carbon compounds Potentially more powerful and selective inhibitors have been
to metabolic pathways.113 One-carbon compounds are par- published more recently (Fig. 3).129 The mechanism by which
ticularly important for the synthesis of nucleobases. blockage of SLC1A5 reduces tumor growth is still unclear.
5-Fluorouracil is an inhibitor of thymidylate synthase, As an exchanger, SLC1A5 cannot mediate net uptake of
which converts nucleotide dUMP to dTMP for DNA bio- amino acids, but it can quickly restore depleted amino acids
synthesis. In the process, N5,N10-methylene tetrahydrofolate at the expense of abundant amino acids.121 It appears likely
is converted to dihydrofolate. Dihydrofolate is recycled in that in cell lines where blocking SLC1A5 reduces mTORC1
two steps involving reduction and recharging of a C1 com- activity, reduced growth is observed, while in cells where
pound, which is derived from serine-to-glycine conversion mTORC1 signaling is unaffected, growth persists. Silencing
by serine hydroxymethyl transferase. Thus, serine is an of SLC1A5 induces amino acid restriction and activation of
important metabolite for cancer cells, which can be gener- the GCN2/ATF4 pathway, which in turn upregulates other
ated from the glycolytic intermediate 3-phosphoglycerate. amino acid transporters.121,126 The variability in the response
Serine depletion causes growth arrest in several cancer cell of cancer cell lines to blockage of SLC1A5 is also illustrated
lines through its role in C1 metabolism, but in addition, ser- by the antitumor efficacy evaluation of a monoclonal anti-
ine depletion increases respiration and production of reac- body against SLC1A5.130 Due to the lack of potent inhibitors,
tive oxygen species, by reducing the allosteric activation of clinical trials or pharmacological experiments in animals
pyruvate kinase PKM2.114 Equally important in cancer cells have not been reported. SLC1A5 knockout mice are viable
is the amino acid glutamine, which contributes to the bio- and fertile,131 suggesting that blocking of ASCT2 in tumor
synthesis of biomolecules in several ways. First, it provides cells is unlikely to affect differentiated cells. Immune cell
nitrogen from its amino group for nucleobase biosynthesis, populations were found to be normal even when proliferating
Second, it is used to synthesize aspartate (also required for intensively.131
nucleobase biosynthesis) and pyruvate in a pathway called SLC7A5 is the major transporter for many essential
glutaminolysis, a linearized version of the TCA cycle used amino acids in cancer cells.11,122,132 Like SLC1A5, it is an
in rapidly proliferating cells.115,116 Through this pathway, amino acid exchanger, which can mediate the uptake of
glutamine can sustain anaplerosis of the TCA cycle, which essential amino acids (BCAAs, aromatic amino acids) at the
provides citrate, for example, for fatty acid biosynthesis. expense of nonessential amino acids. Moreover, it is over-
Third, glutamine is essential for the hexosamine pathway expressed in many cancer cell lines,11,12,122,133 and high
that produces aminated carbohydrates for the purposes of expression is often correlated with poor clinical progno-
glycosylation, the amino group being derived from gluta- sis.134 Genetic deletion of LAT1 in human colon adenocar-
mine.117 Fourth, glutamine is an important precursor of glu- cinoma cells significantly reduced cell proliferation, the
tathione biosynthesis.118 Differentiated cells often produce formation of tumor spheroids, and the volume of tumor
glutamine to dispose of ammonia from amino acid metabo- xenografts.135 Similar to the deletion of SLC1A5, deletion
lism, while cancer cells become glutamine dependent. of SLC7A5 induced an amino acid stress response, as evi-
Many cancer cells cease to grow when glutamine is removed denced by phosphorylation of eIF2α and GCN2 and tran-
from the media.119 The glutamine dependence is a conse- scription of ATF4. This has also been observed in other
quence of the multiple uses of glutamine in the pathways cancer cell lines.136 It is well established that SLC7A5
mentioned above. Consequently, glutamine transport has forms a covalent heteromeric complex with CD98, which is
been considered a target for cancer therapy, but the approach required for trafficking to the plasma membrane,137,138 but is
is not straightforward due to the multitude of glutamine not involved in the transport process itself.139 However,
transporters in mammalian cells.6,10,120,121 Mammalian cells, genetic deletion of CD98 in human colon adenocarcinoma
312 SLAS Discovery 23(4)

cells had a much milder phenotype than deletion of but less so in PC-3 cells, which appear to rely more on
SLC7A5, although transport activity was reduced by SLC7A5 activity.136 In prostate cancer cells, SLC7A5 and
90%.135 Blocking the residual transport activity with the SLC43A1 were regulated in a coordinated fashion. High
SLC7A5 inhibitor JPH203 completely abolished growth of expression of SLC43A1 is accompanied by low expression
the CD98 knockout cells. Reduced growth was accompa- of SLC7A5 and vice versa. The expression of SLC43A1 is
nied by reduced mTORC1 activity. Inhibitors for SLC7A5 regulated by androgen receptor binding, while SLC7A5 is
have been developed and reduce tumor growth in vivo12,140 regulated by an amino acid response element that increases
or in vitro.141–143 No human clinical trials have been transcription after amino acid depletion.136
reported. A potential concern is the role of SLC7A5 as the SLC38A1 and SLC38A2 are Na+-dependent transporters
major transporter for essential amino acids in the blood– for small and medium-sized neutral amino acids. When suf-
brain barrier.144 Global knockout of SLC7A5 and CD98 in ficient amino acids are present, SLC38A2 is not found at
mice has been reported as being embryonically lethal.145,146 the plasma membrane, but its expression increases mani-
SLC7A11 encodes a glutamate/cystine exchanger, which fold after amino acid depletion.121,158 The expression is
plays an important role in providing cysteine for the synthe- tightly regulated at the transcriptional level,159 mRNA
sis of glutathione in a variety of cell types.147 Triple-negative level,160 and protein level.161,162 Importantly, SLC38A1 and
breast cancer cell lines that were highly glutamine depen- SLC38A2 are involved in providing glutamine for glutami-
dent also showed elevated expression of SLC7A11, cystine nolysis.121 The pharmacology of SLC38A1 and A2 is ill-
uptake, and glutamate secretion. SLC7A11 expression is developed. The proline analogue N-methylamino-isobutyric
responsible for the methionine dependency of cancer cell acid is frequently used as an SLC38A1/2 inhibitor and
growth observed in some breast cancer cell lines.148 In these reduces growth of osteosarcoma cells.121 Silencing of
cells, cystine uptake reduces the use of methionine as a pre- SLC38A1 also reduced the growth of colon cancer cell lines
cursor for cysteine. SLC7A11 is highly regulated in cancer and pancreas cancer cell lines.163,164
cells, being downregulated by oncogenic PI3K148 and also These results suggest that amino acid transporters have a
by overexpression of mutated p53.149 Downregulation of significant potential in cancer therapy, but the field lacks,
SLC7A11 renders cells more susceptible to cell death with a few exceptions, highly specific inhibitors that could
caused by reactive oxygen species, such as ferroptosis.149 be used in animal models and clinical trials.
The anti-inflammatory pro-drug sulfasalazine, an inhibitor
of SLC7A11, reduced tumor size in xenografts.118 While Role of Amino Acid Transporters in
sulfasalazine is not used as a drug to treat cancer due to its
chemical instability, it provides proof of concept that target-
Inflammation and Immune Disorders
ing xCT could be a therapeutic strategy in certain subgroups Arginine is the precursor for the generation of nitric oxide.
of cancer, particularly in combination with anticancer drugs The inducible cationic amino acid transporter 2 (SLC7A2,
that increase oxidative stress.150 splice form CAT2B) provides arginine for nitric oxide syn-
SLC6A14 is a Na+- and Cl–-dependent amino acid trans- thase, particularly in macrophages.165 Nitric oxide is thought
porter, which accepts all amino acids, except aspartate and glu- to contribute to the pathology of inflammatory bowel dis-
tamate. The transporter is expressed in selected tissues, such as ease and Crohn’s disease. Provision of arginine has shown
lung, trachea, salivary gland, pituitary, distal ileum, and colon. beneficial effects in animal models of colonic inflamma-
Due to its tissue-specific expression, it is not generally upregu- tion.166 The beneficial effects were lost in mice lacking
lated in cancer but is found in colon cancer,151 cervical can- SLC7A2.167 Consistently, SLC7A2 knockout mice develop
cer,152 estrogen receptor–positive breast cancer,153 and chronic inflammation in the lungs, potentially because of
pancreatic cancer.120 The tumor-promoting role of SLC6A14 reduced NO production in macrophages.168
has been documented in estrogen receptor–positive breast can- SLC7A5 is the major transporter for BCAAs in activated
cer using α-methyl-tryptophan, a small-molecule blocker of lymphocytes.145 A global knockout of SLC7A5 is embry-
the transporter.154 SLC6A14 knockout mice, when crossed onically lethal, but a CD4-driven cre-lox SLC7A5 knock-
with the polyoma middle T oncoprotein mouse breast cancer out results in normal animal and normal immune cell
model, showed delays in the development of mammary number in unchallenged animals.145 However, immunologi-
tumors.155 More advanced inhibitors are required to evaluate cal activation of T cells was largely abolished in SLC7A5
this target in cancer treatment. knockout cells. Deletion of SLC7A5 not only prevented
SLC43A1 is a Na+-independent transporter for large neu- activation of mTORC1, but also interfered with metabolic
tral amino acids.156 In contrast to SLC7A5, which operates as reprogramming, such as upregulation of glucose and gluta-
an amino acid exchanger, SLC43A1 mediates facilitative dif- mine transport.145
fusion of its substrates.157 The gene is overexpressed in pros- SLC1A5 is an amino acid exchanger that is used to har-
tate cancer, together with SLC7A5. Silencing of SLC43A1 monize amino acid levels within rapidly proliferating cells.
reduced mTORC1 signaling and cell growth in LNCaP cells, As a result, it is highly expressed in cancer cells, but also in
Bröer 313

Development of
transcriptional/
translational/
allosteric Improved
Mutaon activator amino acid
associated balance Robust
with under amino acid Disease
disease metabolic homeostasis prevention
phenotype
stress
condions

Figure 5.  Future strategies to modulate metabolic diseases. Lack of transporter activity is associated with metabolic diseases.
Enhancing the activity of the transporter protects the cell against metabolic stress.

rapidly proliferating immune cell populations. Nakaya transporters. Amino acid transporters are highly regulated dur-
et al.169 reported that SLC1A5 knockout animals had attenu- ing early embryo development.16,173 For instance, GLYT1 is
ated inflammatory T-cell responses. Particularly, interferon-γ- essential for the regulation of oocyte size and is activated as the
producing T-helper cells were significantly reduced. oocyte undergoes meiosis,19 transporting the osmolyte glycine.
Masle-Farquhar et al., by contrast, reported normal B- and The osmolyte betaine is transported by SLC6A20, which is
T-cell development and normal B-cell function, suggesting a upregulated in one- and two-cell embryos.20 Cationic amino
limited effect of this transporter on immune function.131 acids are recruited through SLC7A9, while neutral amino
SLC16A10 is the major transporter for aromatic amino acids are taken up by SLC7A5.174 The sodium neutral amino
acids in many cell types.170 The transporter mediates facili- acid transporter SLC38A2 is expressed in pluripotent stem
tated diffusion of aromatic amino acids and may therefore cells, and its activity is required for cell differentiation by accu-
serve as a route for uptake (e.g., in the liver, where metabo- mulating l-proline.175 Competitive inhibition of SLC38A2 by
lism of aromatic amino acids occurs) or efflux (e.g., in epi- high concentrations of glycine and alanine in the oviductal
thelial cells, where aromatic amino acids are released across fluid may prevent development before implantation in the
the basolateral membrane). NOD (nonobese diabetic) mice uterus.16 Future research in this area is projected to focus on
are a widely used model of type 1 diabetes, an autoimmune modulation of epigenetic signatures,176 which could be
disorder, which results in the destruction of β cells by lym- achieved by media supplements, but also by activation or inhi-
phocytes infiltrating the islets of the pancreas.171 NOD mice bition of amino acid transporters. Embryonic stem cells, for
spontaneously develop type 1 diabetes at an elevated rate, instance, acquire epigenetic changes associated with the for-
with an onset at an age of more than 100 days. Transposon- mation of early primitive ectoderm-like cells, which are
mediated inactivation of SLC16A10 increased the suscepti- induced by l-proline.175,177
bility to T1D in NOD mice.172 The transporter is highly Amino acid supplementation and modulation of amino
expressed in macrophages, which play an important role in acid transport could be used to optimize the growth and dif-
antigen presentation and the release of pro-inflammatory ferentiation of stem cells and to optimize conditions for in
cytokines. The mechanism by which inactivation of vitro fertilized oocytes.
SLC16A10 increases the incidence of T1D remains to be
elucidated.
Conclusion and Future Outlook
Role of Amino Acid Transporters in Amino acid transporters are often found at the beginning of
metabolic pathways and influence a wide variety of biological
Embryo Development and Stem Cells processes, in particular, cell proliferation, but also cell signal-
In vitro fertilization is an important process in research (mouse ing and nutrient homeostasis. Thus far, very few amino acid
embryos), animal husbandry, and human reproduction. transport inhibitors have found their way into clinical trials.
Significant efforts have been made to optimizie media for However, this is largely the result of the limited development
embryo development in vitro. While embryos develop at phys- of high-affinity and selective inhibitors. With improvements of
iological osmolarity in vivo, in vitro development beyond the tools to express membrane transporters and HTS being increas-
two-cell stage requires reduced osmolarity. The two-cell block ingly available in academic environments, this is likely to
can, however, be overcome in normal osmolarity by the addi- change in the near future. Solute carriers are considered an
tion of osmolytes, such as glycine, betaine, proline, β-alanine, underdeveloped area of drug development.178 In addition to
and hypotaurine,19 which are accumulated by amino acid current approaches presented in this review, several strategies
314 SLAS Discovery 23(4)

Stress Response
(alternative
transcripts) Restored Growth
amino acid Migration
homeostasis Metastasis
Block of a
single
amino acid
transporter Amino acid
Well- imbalance
nourished Nutrient
tumor cell stress Figure 6.  Future strategies
combining two stress factors to
reduce cell growth. A significant
problem using amino acid
Inhibitors of
Breakdown transporters as drug targets is
Stress response Stagnation
redundancy. Blocking the adaptive
Pathways of cellular Apoptosis
homeostasis stress response or inducing an
Stress inducers independent stress can overcome
metabolic adaptation.

are emerging as new opportunities for targeting amino acid Funding


transport. One such strategy is augmentation (Fig. 5). Whole- The author disclosed receipt of the following financial support for
genome sequencing is increasingly used to identify mutations, the research, authorship, and/or publication of this article:
including those affecting amino acid transporters associated Research in the laboratory of the author is supported by grants of
with disease phenotypes. These mutations are most likely inac- the National Health and Medical Research Council ID 1128442
tivating transporters, and as a result, transport activators rather and 1105857 and the Australian Research Council DP180101702.
than inhibitors will be required. For instance, mutation in
the putative monocarboxylate transporter Slc16a11 was ORCID iD
identified as being associated with the development of type
Stefan Bröer https://orcid.org/0000-0002-8040-1634
2 diabetes in Mexican populations.179 This provides evi-
dence that an activator of Slc16a11 could be used to treat or
prevent type 2 diabetes.180 Transport activators can be iden- References
tified at three levels, namely, transcriptional, translational, 1. Wu, G. Amino Acids: Biochemistry and Nutrition; CRC Press:
and allosteric. Transcriptional and translational activators Boca Raton, FL, 2013.
have been successfully identified for amino acid transporters, 2. Broer, S.; Broer, A. Amino Acid Homeostasis and Signalling
as outlined above.108,109 Another strategy is combination (Fig. in Mammalian Cells and Organisms. Biochem. J. 2017, 474,
6). Amino acid transport is a tempting target for tumor chemo- 1935–1963.
therapy, but a better understanding of amino acid transporter 3. Kuma, A.; Mizushima, N. Physiological Role of Autophagy
redundancy and plasticity will be required. Tumor cells often as an Intracellular Recycling System: With an Emphasis on
escape inhibition of amino acid transporters through upregula- Nutrient Metabolism. Semin. Cell Dev. Biol. 2010, 21, 683–
690.
tion of alternative transporters via the GCN2/ATF4 path-
4. Taylor, P. M. Role of Amino Acid Transporters in Amino
way.121,136 This pathway reduces conventional Cap-dependent Acid Sensing. Am. J. Clin. Nutr. 2014, 99, 223S–230S.
translation and, at the same time, activates transcripts with 5. Broer, S.; Palacin, M. The Role of Amino Acid Transporters
complex secondary structures.2 As outlined above, transla- in Inherited and Acquired Diseases. Biochem. J. 2011, 436,
tional activators have been identified that appear to weaken 193–211.
complex secondary structures occurring in mRNA mole- 6. Pochini, L.; Scalise, M.; Galluccio, M.; et al. Membrane
cules.109 It appears feasible to identify compounds that stabilize Transporters for the Special Amino Acid Glutamine:
secondary structures, thereby inhibiting translation. Combining Structure/Function Relationships and Relevance to Human
amino acid transporter inhibition with inhibition of rescue Health. Front. Chem. 2014, 2, 61.
mechanisms could open new chemotherapeutic approaches. 7. Nakagawa, T.; Kaneko, S. SLC1 Glutamate Transporters and
Diseases: Psychiatric Diseases and Pathological Pain. Curr.
Mol. Pharmacol. 2013, 6, 66–73.
Declaration of Conflicting Interests
8. Vandenberg, R. J.; Ryan, R. M.; Carland, J. E.; et al. Glycine
The author declared no potential conflicts of interest with respect Transport Inhibitors for the Treatment of Pain. Trends
to the research, authorship, and/or publication of this article. Pharmacol. Sci. 2014, 35, 423–430.
Bröer 315

9. Harvey, R. J.; Yee, B. K. Glycine Transporters as Novel Hindrance in Drug Delivery to the Central Nervous System?
Therapeutic Targets in Schizophrenia, Alcohol Dependence F1000Res 2016, 5.
and Pain. Nat. Rev. Drug Discov. 2013, 12, 866–885. 26. Banks, W. A. From Blood-Brain Barrier to Blood-Brain

10. Bhutia, Y. D.; Babu, E.; Ramachandran, S.; et al. Amino Acid Interface: New Opportunities for CNS Drug Delivery. Nat.
Transporters in Cancer and Their Relevance to “Glutamine Rev. Drug Discov. 2016, 15, 275–292.
Addiction”: Novel Targets for the Design of a New Class of 27. Koldso, H.; Grouleff, J.; Schiott, B. Insights to Ligand Binding
Anticancer Drugs. Cancer Res. 2015, 75, 1782–1788. to the Monoamine Transporters—From Homology Modeling to
11. Wang, Q.; Holst, J. L-Type Amino Acid Transport and Cancer: LeuBAT and dDAT. Front. Pharmacol. 2015, 6, 208.
Targeting the mTORC1 Pathway to Inhibit Neoplasia. Am. J. 28. Wolfenden, R. Conformational Aspects of Inhibitor Design:
Cancer Res. 2015, 5, 1281–1294. Enzyme-Substrate Interactions in the Transition State. Bioorg.
12. Oda, K.; Hosoda, N.; Endo, H.; et al. L-Type Amino Acid Med. Chem. 1999, 7, 647–652.
Transporter 1 Inhibitors Inhibit Tumor Cell Growth. Cancer 29. Kramer, R. Functional Principles of Solute Transport

Sci. 2010, 101, 173–179. Systems: Concepts and Perspectives. Biochim. Biophys. Acta
13. Jiang, Y.; Rose, A. J.; Sijmonsma, T. P.; et al. Mice Lacking 1994, 1185, 1–34.
Neutral Amino Acid Transporter B(0)AT1 (Slc6a19) 30. Yan, N. A Glimpse of Membrane Transport through

Have Elevated Levels of FGF21 and GLP-1 and Improved Structures—Advances in the Structural Biology of the GLUT
Glycaemic Control. Mol. Metab. 2015, 4, 406–717. Glucose Transporters. J. Mol. Biol. 2017, 429, 2710–2725.
14. Dohi, T.; Morita, K.; Kitayama, T.; et al. Glycine Transporter 31. Kumar, H.; Kasho, V.; Smirnova, I.; et al. Structure of Sugar-
Inhibitors as a Novel Drug Discovery Strategy for Neuropathic Bound LacY. Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 1784–
Pain. Pharmacol. Ther. 2009, 123, 54–79. 1788.
15. Yoneda, Y. An L-Glutamine Transporter Isoform for
32. Ewers, D.; Becher, T.; Machtens, J. P.; et al. Induced Fit
Neurogenesis Facilitated by L-Theanine. Neurochem. Res. Substrate Binding to an Archeal Glutamate Transporter
2017, 42, 2686–2697. Homologue. Proc. Natl. Acad. Sci. U.S.A. 2013, 110, 12486–
16. Tan, B. S.; Rathjen, P. D.; Harvey, A. J.; et al. Regulation of 12491.
Amino Acid Transporters in Pluripotent Cell Populations in 33. Forrest, L. R.; Kramer, R.; Ziegler, C. The Structural Basis of
the Embryo and in Culture; Novel Roles for Sodium-Coupled Secondary Active Transport Mechanisms. Biochim. Biophys.
Neutral Amino Acid Transporters. Mech. Dev. 2016, 141, 32–39. Acta 2011, 1807, 167–188.
17. Kondou, H.; Kawai, M.; Tachikawa, K.; et al. Sodium-
34. Krogsgaard-Larsen, P.; Bunch, L. Medicinal Chemistry of
Coupled Neutral Amino Acid Transporter 4 Functions as a α-Amino Acids. In Amino Acids, Peptides and Proteins in
Regulator of Protein Synthesis during Liver Development. Organic Synthesis; Hughes, A. B., Ed.; Wiley-VCH Verlag:
Hepatol. Res. 2013, 43, 1211–1223. Weinheim, 2011; pp 151–174.
18. Richards, T.; Wang, F.; Liu, L.; et al. Rescue of
35. Kanai, Y.; Clemencon, B.; Simonin, A.; et al. The SLC1 High-
Postcompaction-Stage Mouse Embryo Development from Affinity Glutamate and Neutral Amino Acid Transporter
Hypertonicity by Amino Acid Transporter Substrates That Family. Mol. Aspects Med. 2013, 34, 108–120.
May Function as Organic Osmolytes. Biol. Reprod. 2010, 82, 36. Canul-Tec, J. C.; Assal, R.; Cirri, E.; et al. Structure and
769–777. Allosteric Inhibition of Excitatory Amino Acid Transporter 1.
19. Baltz, J. M.; Tartia, A. P. Cell Volume Regulation in Oocytes Nature 2017, 544, 446–451.
and Early Embryos: Connecting Physiology to Successful 37. Jensen, A. A.; Erichsen, M. N.; Nielsen, C. W.; et al. Discovery
Culture Media. Hum. Reprod. Update 2010, 16, 166–176. of the First Selective Inhibitor of Excitatory Amino Acid
20. Anas, M. K.; Lee, M. B.; Zhou, C.; et al. SIT1 Is a Betaine/ Transporter Subtype 1. J. Med. Chem. 2009, 52, 912–915.
Proline Transporter That Is Activated in Mouse Eggs 38. Jorgensen, L.; Al-Khawaja, A.; Kickinger, S.; et al. Structure-
after Fertilization and Functions until the 2-Cell Stage. Activity Relationship, Pharmacological Characterization,
Development 2008, 135, 4123–4130. and Molecular Modeling of Noncompetitive Inhibitors of the
21. Dean, E. D.; Li, M.; Prasad, N.; et al. Interrupted Glucagon Betaine/Gamma-Aminobutyric Acid Transporter 1 (BGT1).
Signaling Reveals Hepatic Alpha Cell Axis and Role for J. Med. Chem. 2017, 60, 8834–8846.
L-Glutamine in Alpha Cell Proliferation. Cell Metab. 2017, 39. Lewis, R. J. Discovery and Development of the Chi-

25, 1362–1373.e5. Conopeptide Class of Analgesic Peptides. Toxicon 2012, 59,
22. Kim, J.; Okamoto, H.; Huang, Z.; et al. Amino Acid
524–528.
Transporter Slc38a5 Controls Glucagon Receptor Inhibition- 40. Kopec, K.; Jones, B.; Thomas, J. C.; et al. Successful

Induced Pancreatic Alpha Cell Hyperplasia in Mice. Cell Identification of Glycine Transporter Inhibitors Using an
Metab. 2017, 25, 1348–1361.e8. Adaptation of a Functional Cell-Based Assay. J. Biomol.
23.
International Transporter Consortium; Giacomini, K. Screen. 2009, 14, 1185–1194.
M.; Huang, S. M.; et al. Membrane Transporters in Drug 41. Kutchukian, P. S.; Warren, L.; Magliaro, B. C.; et al. Iterative
Development. Nat. Rev. Drug Discov. 2010, 9, 215–236. Focused Screening with Biological Fingerprints Identifies
24. Kaisar, M. A.; Sajja, R. K.; Prasad, S.; et al. New Experimental Selective Asc-1 Inhibitors Distinct from Traditional High
Models of the Blood-Brain Barrier for CNS Drug Discovery. Throughput Screening. ACS Chem. Biol. 2017, 12, 519–527.
Expert Opin. Drug Discov. 2017, 12, 89–103. 42. Pochini, L.; Scalise, M.; Galluccio, M.; et al. OCTN Cation
25. Saunders, N. R.; Habgood, M. D.; Mollgard, K.; et al. The Transporters in Health and Disease: Role as Drug Targets and
Biological Significance of Brain Barrier Mechanisms: Help or Assay Development. J. Biomol. Screen. 2013, 18, 851–867.
316 SLAS Discovery 23(4)

43. Williams, J. B.; Mallorga, P. J.; Lemaire, W.; et al. Development High-Throughput MS Screening. J. Lab. Autom. 2016, 21,
of a Scintillation Proximity Assay for Analysis of Na+/ 19–26.
Cl–-Dependent Neurotransmitter Transporter Activity. Anal. 58. Colas, C.; Ung, P. M.; Schlessinger, A. SLC Transporters:
Biochem. 2003, 321, 31–37. Structure, Function, and Drug Discovery. Medchemcomm
44. Quick, M.; Javitch, J. A. Monitoring the Function of
2016, 7, 1069–1081.
Membrane Transport Proteins in Detergent-Solubilized Form. 59. Coleman, J. A.; Green, E. M.; Gouaux, E. X-Ray Structures
Proc. Natl. Acad. Sci. U.S.A. 2007, 104, 3603–3608. and Mechanism of the Human Serotonin Transporter. Nature
45. Waggoner, A. S. Dye Indicators of Membrane Potential.
2016, 532, 334–339.
Annu. Rev. Biophys. Bioeng. 1979, 8, 47–68. 60. Colas, C.; Grewer, C.; Otte, N. J.; et al. Ligand Discovery for
46. Cheng, Q.; Shah, N.; Broer, A.; et al. Identification of
the Alanine-Serine-Cysteine Transporter (ASCT2, SLC1A5)
Novel Inhibitors of the Amino Acid Transporter B(0) AT1 from Homology Modeling and Virtual Screening. PLoS
(SLC6A19), a Potential Target to Induce Protein Restriction Comput. Biol. 2015, 11, e1004477.
and to Treat Type 2 Diabetes. Br. J. Pharmacol. 2017, 174, 61. Geier, E. G.; Schlessinger, A.; Fan, H.; et al. Structure-

468–482. Based Ligand Discovery for the Large-Neutral Amino Acid
47. Allan, L.; Leith, J. L.; Papakosta, M.; et al. Development Transporter 1, LAT-1. Proc. Natl. Acad. Sci. U.S.A. 2013,
of a Novel High-Throughput Assay for the Investigation 110, 5480–5485.
of GlyT-1b Neurotransmitter Transporter Function. Comb. 62. Ruggeri, C.; Drinkwater, N.; Sivaraman, K. K.; et al.

Chem. High Throughput Screen. 2006, 9, 9–14. Identification and Validation of a Potent Dual Inhibitor of the
48. Benjamin, E. R.; Skelton, J.; Hanway, D.; et al. Validation P. falciparum M1 and M17 Aminopeptidases Using Virtual
of a Fluorescent Imaging Plate Reader Membrane Potential Screening. PLoS One 2015, 10, e0138957.
Assay for High-Throughput Screening of Glycine Transporter 63. Dobson, P. D.; Kell, D. B. Carrier-Mediated Cellular Uptake
Modulators. J. Biomol. Screen. 2005, 10, 365–373. of Pharmaceutical Drugs: An Exception or the Rule? Nat.
49. Faure, S.; Jensen, A. A.; Maurat, V.; et al. Stereoselective Rev. Drug Discov. 2008, 7, 205–220.
Chemoenzymatic Synthesis of the Four Stereoisomers of 64. Schlessinger, A.; Khuri, N.; Giacomini, K. M.; et al. Molecular
l-2-(2-Carboxycyclobutyl)Glycine and Pharmacological Modeling and Ligand Docking for Solute Carrier (SLC)
Characterization at Human Excitatory Amino Acid Transporters. Curr. Top. Med. Chem. 2013, 13, 843–856.
Transporter Subtypes 1, 2, and 3. J. Med. Chem. 2006, 49, 65. Grewer, C.; Gameiro, A.; Mager, T.; et al. Electrophysiological
6532–6538. Characterization of Membrane Transport Proteins. Annu. Rev.
50. Iamshanova, O.; Mariot, P.; Lehen’kyi, V.; et al. Comparison Biophys. 2013, 42, 95–120.
of Fluorescence Probes for Intracellular Sodium Imaging in 66. Kiehn, J.; Lacerda, A. E.; Wible, B.; et al. Molecular

Prostate Cancer Cell Lines. Eur. Biophys. J. 2016, 45, 765– Physiology and Pharmacology of HERG. Single-Channel
777. Currents and Block by Dofetilide. Circulation 1996, 94,
51. Wenzel, U.; Thwaites, D. T.; Daniel, H. Stereoselective
2572–2579.
Uptake of Beta-Lactam Antibiotics by the Intestinal Peptide 67. Ovens, M. J.; Davies, A. J.; Wilson, M. C.; et al. AR-C155858
Transporter. Br. J. Pharmacol. 1995, 116, 3021–3027. Is a Potent Inhibitor of Monocarboxylate Transporters MCT1
52. Gruenwald, K.; Holland, J. T.; Stromberg, V.; et al.
and MCT2 That Binds to an Intracellular Site Involving
Visualization of Glutamine Transporter Activities in Living Transmembrane Helices 7–10. Biochem. J. 2010, 425, 523–530.
Cells Using Genetically Encoded Glutamine Sensors. PLoS 68. Jung, H.; Tebbe, S.; Schmid, R.; et al. Unidirectional

One 2012, 7, e38591. Reconstitution and Characterization of Purified Na+/Proline
53. Vanoaica, L.; Behera, A.; Camargo, S. M.; et al. Real-
Transporter of Escherichia coli. Biochemistry 1998, 37,
Time Functional Characterization of Cationic Amino Acid 11083–11088.
Transporters Using a New FRET Sensor. Pflugers Arch. 69. Akyuz, N.; Altman, R. B.; Blanchard, S. C.; et al. Transport
2016, 468, 563–572. Dynamics in a Glutamate Transporter Homologue. Nature
54. Whitfield, J. H.; Zhang, W. H.; Herde, M. K.; et al.
2013, 502, 114–118.
Construction of a Robust and Sensitive Arginine Biosensor 70. Ohman, M.; Oksanen, L.; Kaprio, J.; et al. Genome-Wide
through Ancestral Protein Reconstruction. Protein Sci. 2015, Scan of Obesity in Finnish Sibpairs Reveals Linkage to
24, 1412–1422. Chromosome Xq24. J. Clin. Endocrinol. Metab. 2000, 85,
55. Mustafi, N.; Grunberger, A.; Kohlheyer, D.; et al. The
3183–3190.
Development and Application of a Single-Cell Biosensor for 71. Suviolahti, E.; Oksanen, L. J.; Ohman, M.; et al. The SLC6A14
the Detection of L-Methionine and Branched-Chain Amino Gene Shows Evidence of Association with Obesity. J. Clin.
Acids. Metab. Eng. 2012, 14, 449–457. Invest. 2003, 112, 1762–1772.
56. Wagner, A. D.; Kolb, J. M.; Ozbal, C. C.; et al. Ultrafast 72. Durand, E.; Boutin, P.; Meyre, D.; et al. Polymorphisms

Mass Spectrometry Based Bioanalytical Method for Digoxin in the Amino Acid Transporter Solute Carrier Family 6
Supporting an In Vitro P-Glycoprotein (P-gp) Inhibition (Neurotransmitter Transporter) Member 14 Gene Contribute
Screen. Rapid Commun. Mass Spectrom. 2011, 25, 1231– to Polygenic Obesity in French Caucasians. Diabetes 2004,
1240. 53, 2483–2486.
57. Sinclair, I.; Stearns, R.; Pringle, S.; et al. Novel Acoustic 73. Sloan, J. L.; Mager, S. Cloning and Functional Expression of
Loading of a Mass Spectrometer: Toward Next-Generation a Human Na(+) and Cl(–)-Dependent Neutral and Cationic
Bröer 317

Amino Acid Transporter B(0+). J. Biol. Chem. 1999, 274, 90. Nicklin, P.; Bergman, P.; Zhang, B.; et al. Bidirectional

23740–23745. Transport of Amino Acids Regulates mTOR and Autophagy.
74. Van Winkle, L. J.; Tesch, J. K.; Shah, A.; et al. System Cell 2009, 136, 521–534.
B0,+ Amino Acid Transport Regulates the Penetration 91. Khunweeraphong, N.; Nagamori, S.; Wiriyasermkul, P.;

Stage of Blastocyst Implantation with Possible Long-Term et al. Establishment of Stable Cell Lines with High
Developmental Consequences through Adulthood. Hum. Expression of Heterodimers of Human 4F2hc and Human
Reprod. Update 2006, 12, 145–157. Amino Acid Transporter LAT1 or LAT2 and Delineation
75. Broer, A.; Klingel, K.; Kowalczuk, S.; et al. Molecular Cloning of Their Differential Interaction with Alpha-Alkyl Moieties.
of Mouse Amino Acid Transport System B0, a Neutral Amino J. Pharmacol. Sci. 2012, 119, 368–380.
Acid Transporter Related to Hartnup Disorder. J. Biol. Chem. 92. Cheng, Q.; Beltran, V. D.; Chan, S. M.; et al. System-L
2004, 279, 24467–24476. Amino Acid Transporters Play a Key Role in Pancreatic Beta-
76. Giesbertz, P.; Daniel, H. Branched-Chain Amino Acids as Cell Signalling and Function. J. Mol. Endocrinol. 2016, 56,
Biomarkers in Diabetes. Curr. Opin. Clin. Nutr. Metab. Care 175–187.
2016, 19, 48–54. 93. Mourao, J. M.; McGivan, J. D.; Chappell, J. B. The Effects
77. Zhao, X.; Han, Q.; Liu, Y.; et al. The Relationship between L-Leucine on the Synthesis of Urea, Glutamate and Glutamine
Branched-Chain Amino Acid Related Metabolomic Signature by Isolated Rat Liver Cells. Biochem. J. 1975, 146, 457–464.
and Insulin Resistance: A Systematic Review. J. Diabetes 94. Henquin, J. C.; Dufrane, D.; Nenquin, M. Nutrient Control of
Res. 2016, 2016, 2794591. Insulin Secretion in Isolated Normal Human Islets. Diabetes
78. Lynch, C. J.; Adams, S. H. Branched-Chain Amino Acids 2006, 55, 3470–3407.
in Metabolic Signalling and Insulin Resistance. Nat. Rev. 95. Gao, Z. Y.; Li, G.; Najafi, H.; et al. Glucose Regulation of
Endocrinol. 2014, 10, 723–736. Glutaminolysis and Its Role in Insulin Secretion. Diabetes
79. Newgard, C. B. Interplay between Lipids and Branched-
1999, 48, 1535–1542.
Chain Amino Acids in Development of Insulin Resistance. 96. Gammelsaeter, R.; Jenstad, M.; Bredahl, M. K.; et al.

Cell Metab. 2012, 15, 606–614. Complementary Expression of SN1 and SAT2 in the Islets
80. Shah, O. J.; Wang, Z.; Hunter, T. Inappropriate Activation of Langerhans Suggests Concerted Action of Glutamine
of the TSC/Rheb/mTOR/S6K Cassette Induces IRS1/2 Transport in the Regulation of Insulin Secretion. Biochem.
Depletion, Insulin Resistance, and Cell Survival Deficiencies. Biophys. Res. Commun. 2009, 381, 378–382.
Curr. Biol. 2004, 14, 1650–1656. 97. Zhou, Y.; Waanders, L. F.; Holmseth, S.; et al. Proteome
81. Krebs, M.; Krssak, M.; Bernroider, E.; et al. Mechanism of Analysis and Conditional Deletion of the EAAT2 Glutamate
Amino Acid-Induced Skeletal Muscle Insulin Resistance in Transporter Provide Evidence against a Role of EAAT2 in
Humans. Diabetes 2002, 51, 599–605. Pancreatic Insulin Secretion in Mice. J. Biol. Chem. 2014,
82. Moghei, M.; Tavajohi-Fini, P.; Beatty, B.; et al. Ketoisocaproic 289, 1329–1344.
Acid, a Metabolite of Leucine, Suppresses Insulin-Stimulated 98. Campbell, J. E.; Drucker, D. J. Islet Alpha Cells and

Glucose Transport in Skeletal Muscle Cells in a BCAT2- Glucagon—Critical Regulators of Energy Homeostasis. Nat.
Dependent Manner. Am. J. Physiol. Cell Physiol. 2016, 311, Rev. Endocrinol. 2015, 11, 329–338.
C518–C527. 99. Longuet, C.; Robledo, A. M.; Dean, E. D.; et al. Liver-

83. Solon-Biet, S. M.; McMahon, A. C.; Ballard, J. W.; et al. Specific Disruption of the Murine Glucagon Receptor
The Ratio of Macronutrients, Not Caloric Intake, Dictates Produces Alpha-Cell Hyperplasia: Evidence for a Circulating
Cardiometabolic Health, Aging, and Longevity in Ad Alpha-Cell Growth Factor. Diabetes 2013, 62, 1196–1205.
Libitum-Fed Mice. Cell Metab. 2014, 19, 418–430. 100. Morita, K.; Motoyama, N.; Kitayama, T.; et al. Spinal

84. Ables, G. P.; Hens, J. R.; Nichenametla, S. N. Methionine Antiallodynia Action of Glycine Transporter Inhibitors in
Restriction Beyond Life-Span Extension. Ann. N.Y. Acad. Sci. Neuropathic Pain Models in Mice. J. Pharmacol. Exp. Ther.
2016, 1363, 68–79. 2008, 326, 633–645.
85. Batterham, R. L.; Cummings, D. E. Mechanisms of Diabetes 101. Javitt, D. C. Glycine Transport Inhibitors for the Treatment
Improvement Following Bariatric/Metabolic Surgery. Diabetes of Schizophrenia: Symptom and Disease Modification.
Care 2016, 39, 893–901. Curr. Opin. Drug Discov. Devel. 2009, 12, 468–478.
86. Seow, H. F.; Broer, S.; Broer, A.; et al. Hartnup Disorder Is 102. Rutter, A. R.; Fradley, R. L.; Garrett, E. M.; et al. Evidence
Caused by Mutations in the Gene Encoding the Neutral Amino from Gene Knockout Studies Implicates Asc-1 as the
Acid Transporter SLC6A19. Nat. Genet. 2004, 36, 1003–1007. Primary Transporter Mediating D-Serine Reuptake in the
87. Kleta, R.; Romeo, E.; Ristic, Z.; et al. Mutations in SLC6A19, Mouse CNS. Eur. J. Neurosci. 2007, 25, 1757–1766.
Encoding B0AT1, Cause Hartnup Disorder. Nat. Genet. 2004, 103. Foster, A. C.; Farnsworth, J.; Lind, G. E.; et al. D-Serine Is
36, 999–1002. a Substrate for Neutral Amino Acid Transporters ASCT1/
88. Pochini, L.; Seidita, A.; Sensi, C.; et al. Nimesulide Binding Site SLC1A4 and ASCT2/SLC1A5, and Is Transported by Both
in the B0AT1 (SLC6A19) Amino Acid Transporter. Mechanism Subtypes in Rat Hippocampal Astrocyte Cultures. PLoS One
of Inhibition Revealed by Proteoliposome Transport Assay and 2016, 11, e0156551.
Molecular Modelling. Biochem. Pharmacol. 2014, 89, 422–430. 104. Sakimura, K.; Nakao, K.; Yoshikawa, M.; et al. A Novel
89. Fotiadis, D.; Kanai, Y.; Palacin, M. The SLC3 and SLC7 Na(+)-Independent Alanine-Serine-Cysteine Transporter
Families of Amino Acid Transporters. Mol. Aspects Med. 1 Inhibitor Inhibits Both Influx and Efflux of D-Serine. J.
2013, 34, 139–158. Neurosci. Res. 2016, 94, 888–895.
318 SLAS Discovery 23(4)

105. Sason, H.; Billard, J. M.; Smith, G. P.; et al. Asc-1


121. Broer, A.; Rahimi, F.; Broer, S. Deletion of Amino Acid
Transporter Regulation of Synaptic Activity via the Tonic Transporter ASCT2 (SLC1A5) Reveals an Essential Role for
Release of D-Serine in the Forebrain. Cereb. Cortex 2017, Transporters SNAT1 (SLC38A1) and SNAT2 (SLC38A2)
27, 1573–1587. to Sustain Glutaminolysis in Cancer Cells. J. Biol. Chem.
106. Bugarski-Kirola, D.; Blaettler, T.; Arango, C.; et al.
2016, 291, 13194–13205.
Bitopertin in Negative Symptoms of Schizophrenia— 122. Fuchs, B. C.; Bode, B. P. Amino Acid Transporters ASCT2
Results from the Phase III FlashLyte and DayLyte Studies. and LAT1 in Cancer: Partners in Crime? Semin. Cancer
Biol. Psychiatry 2017, 82, 8–16. Biol. 2005, 15, 254–266.
107. Gomeza, J.; Hulsmann, S.; Ohno, K.; et al. Inactivation of 123. Fuchs, B. C.; Finger, R. E.; Onan, M. C.; et al. ASCT2
the Glycine Transporter 1 Gene Discloses Vital Role of Glial Silencing Regulates Mammalian Target-of-Rapamycin
Glycine Uptake in Glycinergic Inhibition. Neuron 2003, 40, Growth and Survival Signaling in Human Hepatoma Cells.
785–796. Am. J. Physiol. Cell Physiol. 2007, 293, C55–C63.
108. Rothstein, J. D.; Patel, S.; Regan, M. R.; et al. Beta-Lactam 124. Wang, Q.; Hardie, R. A.; Hoy, A. J.; et al. Targeting ASCT2-
Antibiotics Offer Neuroprotection by Increasing Glutamate Mediated Glutamine Uptake Blocks Prostate Cancer Growth
Transporter Expression. Nature 2005, 433, 73–77. and Tumour Development. J. Pathol. 2015, 236, 278–289.
109. Colton, C. K.; Kong, Q.; Lai, L.; et al. Identification of 125. Wang, Q.; Beaumont, K. A.; Otte, N. J.; et al. Targeting
Translational Activators of Glial Glutamate Transporter Glutamine Transport to Suppress Melanoma Cell Growth.
EAAT2 through Cell-Based High-Throughput Screening: Int. J. Cancer 2014, 135, 1060–1071.
An Approach to Prevent Excitotoxicity. J. Biomol. Screen. 126. van Geldermalsen, M.; Wang, Q.; Nagarajah, R.; et al.

2010, 15, 653–662. ASCT2/SLC1A5 Controls Glutamine Uptake and Tumour
110. Cudkowicz, M. E.; Titus, S.; Kearney, M.; et al. Safety and Growth in Triple-Negative Basal-Like Breast Cancer.
Efficacy of Ceftriaxone for Amyotrophic Lateral Sclerosis: Oncogene 2016, 35, 3201–3208.
A Multi-Stage, Randomised, Double-Blind, Placebo- 127. Grewer, C.; Grabsch, E. New Inhibitors for the Neutral
Controlled Trial. Lancet Neurol. 2014, 13, 1083–1091. Amino Acid Transporter ASCT2 Reveal Its Na+-Dependent
111. Kong, Q.; Chang, L. C.; Takahashi, K.; et al. Small-Molecule Anion Leak. J. Physiol. 2004, 557, 747–759.
Activator of Glutamate Transporter EAAT2 Translation 128. Esslinger, C. S.; Cybulski, K. A.; Rhoderick, J. F. Ngamma-
Provides Neuroprotection. J. Clin. Invest. 2014, 124, 1255– Aryl Glutamine Analogues as Probes of the ASCT2 Neutral
1267. Amino Acid Transporter Binding Site. Bioorg. Med. Chem.
112. Dang, C. V.; Hamaker, M.; Sun, P.; et al. Therapeutic
2005, 13, 1111–1118.
Targeting of Cancer Cell Metabolism. J. Mol. Med. (Berl.) 129. Schulte, M. L.; Khodadadi, A. B.; Cuthbertson, M. L.;

2011, 89, 205–212. et al. 2-Amino-4-Bis(Aryloxybenzyl)Aminobutanoic Acids: A
113. Zubrod, C. G. Historic Milestones in Curative Chemotherapy. Novel Scaffold for Inhibition of ASCT2-Mediated Glutamine
Semin. Oncol. 1979, 6, 490–505. Transport. Bioorg. Med. Chem. Lett. 2016, 26, 1044–1047.
114. Maddocks, O. D.; Berkers, C. R.; Mason, S. M.; et al. Serine 130. Kasai, N.; Sasakawa, A.; Hosomi, K.; et al. Anti-Tumor
Starvation Induces Stress and p53-Dependent Metabolic Efficacy Evaluation of a Novel Monoclonal Antibody
Remodelling in Cancer Cells. Nature 2013, 493, 542–546. Targeting Neutral Amino Acid Transporter ASCT2 Using
115. Newsholme, E. A.; Crabtree, B.; Ardawi, M. S. The Role Patient-Derived Xenograft Mouse Models of Gastric Cancer.
of High Rates of Glycolysis and Glutamine Utilization in Am. J. Transl. Res. 2017, 9, 3399–3410.
Rapidly Dividing Cells. Biosci. Rep. 1985, 5, 393–400. 131. Masle-Farquhar, E.; Broer, A.; Yabas, M.; et al.

116. DeBerardinis, R. J.; Mancuso, A.; Daikhin, E.; et al. Beyond ASCT2 (SLC1A5)-Deficient Mice Have Normal B-Cell
Aerobic Glycolysis: Transformed Cells Can Engage in Development, Proliferation, and Antibody Production.
Glutamine Metabolism That Exceeds the Requirement for Front. Immunol. 2017, 8, 549.
Protein and Nucleotide Synthesis. Proc. Natl. Acad. Sci. 132. Hayashi, K.; Anzai, N. Novel Therapeutic Approaches

U.S.A. 2007, 104, 19345–19350. Targeting L-Type Amino Acid Transporters for Cancer
117. Wellen, K. E.; Lu, C.; Mancuso, A.; et al. The Hexosamine Treatment. World J. Gastrointest. Oncol. 2017, 9, 21–29.
Biosynthetic Pathway Couples Growth Factor-Induced 133. Kaira, K.; Oriuchi, N.; Imai, H.; et al. L-Type Amino

Glutamine Uptake to Glucose Metabolism. Genes Dev. Acid Transporter 1 and CD98 Expression in Primary and
2010, 24, 2784–2799. Metastatic Sites of Human Neoplasms. Cancer Sci. 2008,
118. Timmerman, L. A.; Holton, T.; Yuneva, M.; et al. Glutamine 99, 2380–2386.
Sensitivity Analysis Identifies the xCT Antiporter as a 134. Kaira, K.; Oriuchi, N.; Imai, H.; et al. Prognostic Significance
Common Triple-Negative Breast Tumor Therapeutic Target. of L-Type Amino Acid Transporter 1 Expression in
Cancer Cell 2013, 24, 450–465. Resectable Stage I-III Nonsmall Cell Lung Cancer. Br. J.
119. Wise, D. R.; Thompson, C. B. Glutamine Addiction: A New Cancer 2008, 98, 742–748.
Therapeutic Target in Cancer. Trends Biochem. Sci. 2010, 135. Cormerais, Y.; Giuliano, S.; LeFloch, R.; et al. Genetic
35, 427–433. Disruption of the Multifunctional CD98/LAT1 Complex
120. Bhutia, Y. D.; Ganapathy, V. Glutamine Transporters in Demonstrates the Key Role of Essential Amino Acid
Mammalian Cells and Their Functions in Physiology and Transport in the Control of mTORC1 and Tumor Growth.
Cancer. Biochim. Biophys. Acta 2016, 1863, 2531–2539. Cancer Res. 2016, 76, 4481–4492.
Bröer 319

136. Wang, Q.; Bailey, C. G.; Ng, C.; et al. Androgen Receptor (SLC6A14) in Colorectal Cancer and Metastasis in Humans.
and Nutrient Signaling Pathways Coordinate the Demand Biochim. Biophys. Acta 2005, 1741, 215–223.
for Increased Amino Acid Transport during Prostate Cancer 152. Gupta, N.; Prasad, P. D.; Ghamande, S.; et al. Up-Regulation
Progression. Cancer Res. 2011, 71, 7525–7536. of the Amino Acid Transporter ATB(0,+) (SLC6A14) in
137. Wagner, C. A.; Lang, F.; Broer, S. Function and Structure Carcinoma of the Cervix. Gynecol. Oncol. 2006, 100, 8–13.
of Heterodimeric Amino Acid Transporters. Am. J. Physiol. 153. Karunakaran, S.; Umapathy, N. S.; Thangaraju, M.; et al.
Cell Physiol. 2001, 281, C1077–C1093. Interaction of Tryptophan Derivatives with SLC6A14
138. Kanai, Y.; Endou, H. Heterodimeric Amino Acid
(ATB0,+) Reveals the Potential of the Transporter as a Drug
Transporters: Molecular Biology and Pathological and Target for Cancer Chemotherapy. Biochem. J. 2008, 414,
Pharmacological Relevance. Curr. Drug Metab. 2001, 2, 343–355.
339–354. 154. Karunakaran, S.; Ramachandran, S.; Coothankandaswamy,
139. Napolitano, L.; Scalise, M.; Galluccio, M.; et al. LAT1 Is the V.; et al. SLC6A14 (ATB0,+) Protein, a Highly Concentrative
Transport Competent Unit of the LAT1/CD98 Heterodimeric and Broad Specific Amino Acid Transporter, Is a Novel and
Amino Acid Transporter. Int. J. Biochem. Cell Biol. 2015, Effective Drug Target for Treatment of Estrogen Receptor-
67, 25–33. Positive Breast Cancer. J. Biol. Chem. 2011, 286, 31830–
140. Kim, C. S.; Cho, S. H.; Chun, H. S.; et al. BCH, an Inhibitor 31838.
of System L Amino Acid Transporters, Induces Apoptosis in 155. Babu, E.; Bhutia, Y. D.; Ramachandran, S.; et al. Deletion
Cancer Cells. Biol. Pharm. Bull. 2008, 31, 1096–1100. of the Amino Acid Transporter Slc6a14 Suppresses Tumour
141. Napolitano, L.; Scalise, M.; Koyioni, M.; et al. Potent
Growth in Spontaneous Mouse Models of Breast Cancer.
Inhibitors of Human LAT1 (SLC7A5) Transporter Based on Biochem. J. 2015, 469, 17–23.
Dithiazole and Dithiazine Compounds for Development of 156. Babu, E.; Kanai, Y.; Chairoungdua, A.; et al. Identification
Anticancer Drugs. Biochem. Pharmacol. 2017, 143, 39–52. of a Novel System L Amino Acid Transporter Structurally
142. Marshall, A. D.; van Geldermalsen, M.; Otte, N. J.; et al. LAT1 Distinct from Heterodimeric Amino Acid Transporters. J.
Is a Putative Therapeutic Target in Endometrioid Endometrial Biol. Chem. 2003, 278, 43838–43845.
Carcinoma. Int. J. Cancer 2016, 139, 2529–2539. 157. Kim, D. K.; Kanai, Y.; Matsuo, H.; et al. The Human

143. Kongpracha, P.; Nagamori, S.; Wiriyasermkul, P.;
T-Type Amino Acid Transporter-1: Characterization, Gene
et al. Structure-Activity Relationship of a Novel Series Organization, and Chromosomal Location. Genomics 2002,
of Inhibitors for Cancer Type Transporter L-Type Amino 79, 95–103.
Acid Transporter 1 (LAT1). J. Pharmacol. Sci. 2017, 133, 158. Gazzola, R. F.; Sala, R.; Bussolati, O.; et al. The Adaptive
96–102. Regulation of Amino Acid Transport System A Is Associated
144. Boado, R. J.; Li, J. Y.; Nagaya, M.; et al. Selective
to Changes in ATA2 Expression. FEBS Lett. 2001, 490, 11–
Expression of the Large Neutral Amino Acid Transporter at 14.
the Blood-Brain Barrier. Proc. Natl. Acad. Sci. U.S.A. 1999, 159. Palii, S. S.; Chen, H.; Kilberg, M. S. Transcriptional Control
96, 12079–12084. of the Human Sodium-Coupled Neutral Amino Acid
145. Sinclair, L. V.; Rolf, J.; Emslie, E.; et al. Control of
Transporter System A Gene by Amino Acid Availability Is
Amino-Acid Transport by Antigen Receptors Coordinates Mediated by an Intronic Element. J. Biol. Chem. 2004, 279,
the Metabolic Reprogramming Essential for T Cell 3463–3471.
Differentiation. Nat. Immunol. 2013, 14, 500–508. 160. Gaccioli, F.; Huang, C. C.; Wang, C.; et al. Amino Acid

146. Tsumura, H.; Suzuki, N.; Saito, H.; et al. The Targeted Starvation Induces the SNAT2 Neutral Amino Acid Transporter
Disruption of the CD98 Gene Results in Embryonic by a Mechanism That Involves Eukaryotic Initiation Factor
Lethality. Biochem. Biophys. Res. Commun. 2003, 308, 2alpha Phosphorylation and Cap-Independent Translation. J.
847–851. Biol. Chem. 2006, 281, 17929–17940.
147. Bannai, S.; Tateishi, N. Role of Membrane Transport in 161. Jeon, Y. J.; Khelifa, S.; Ratnikov, B.; et al. Regulation of
Metabolism and Function of Glutathione in Mammals. J. Glutamine Carrier Proteins by RNF5 Determines Breast
Membr. Biol. 1986, 89, 1–8. Cancer Response to ER Stress-Inducing Chemotherapies.
148. Lien, E. C.; Ghisolfi, L.; Geck, R. C.; et al. Oncogenic PI3K Cancer Cell 2015, 27, 354–369.
Promotes Methionine Dependency in Breast Cancer Cells 162. Nardi, F.; Hoffmann, T. M.; Stretton, C.; et al. Proteasomal
through the Cystine-Glutamate Antiporter xCT. Sci. Signal. Modulation of Cellular SNAT2 (SLC38A2) Abundance and
2017, 10. Function by Unsaturated Fatty Acid Availability. J. Biol.
149. Jiang, L.; Kon, N.; Li, T.; et al. Ferroptosis as a p53-Mediated Chem. 2015, 290, 8173–8184.
Activity during Tumour Suppression. Nature 2015, 520, 163. Zhou, F. F.; Xie, W.; Chen, S. Q.; et al. SLC38A1 Promotes
57–62. Proliferation and Migration of Human Colorectal Cancer
150. Teppo, H. R.; Soini, Y.; Karihtala, P. Reactive Oxygen Cells. J. Huazhong Univ. Sci. Technolog. Med. Sci. 2017,
Species-Mediated Mechanisms of Action of Targeted 37, 30–36.
Cancer Therapy. Oxid. Med. Cell. Longev. 2017, 2017, 164. Xie, J.; Chen, Z.; Liu, L.; et al. shRNA-Mediated Slc38a1
1485283. Silencing Inhibits Migration, but Not Invasiveness of
151. Gupta, N.; Miyauchi, S.; Martindale, R. G.; et al.
Human Pancreatic Cancer Cells. Chin. J. Cancer Res. 2013,
Upregulation of the Amino Acid Transporter ATB0,+ 25, 514–519.
320 SLAS Discovery 23(4)

165. Closs, E. I.; Scheld, J. S.; Sharafi, M.; et al. Substrate Supply 174. Van Winkle, L. J.; Campione, A. L.; Gorman, J. M.; et al.
for Nitric-Oxide Synthase in Macrophages and Endothelial Changes in the Activities of Amino Acid Transport Systems
Cells: Role of Cationic Amino Acid Transporters. Mol. b0,+ and L during Development of Preimplantation Mouse
Pharmacol. 2000, 57, 68–74. Conceptuses. Biochim. Biophys. Acta 1990, 1021, 77–84.
166. Coburn, L. A.; Gong, X.; Singh, K.; et al. L-Arginine
175. Tan, B. S.; Lonic, A.; Morris, M. B.; et al. The Amino
Supplementation Improves Responses to Injury and Acid Transporter SNAT2 Mediates L-Proline-Induced
Inflammation in Dextran Sulfate Sodium Colitis. PLoS One Differentiation of ES Cells. Am. J. Physiol. Cell Physiol.
2012, 7, e33546. 2011, 300, C1270–C1279.
167. Singh, K.; Coburn, L. A.; Barry, D. P.; et al. Deletion of 176. Lee, M. B.; Kooistra, M.; Zhang, B.; et al. Betaine

Cationic Amino Acid Transporter 2 Exacerbates Dextran Homocysteine Methyltransferase Is Active in the Mouse
Sulfate Sodium Colitis and Leads to an IL-17-Predominant Blastocyst and Promotes Inner Cell Mass Development. J.
T Cell Response. Am. J. Physiol. Gastrointest. Liver Physiol. Biol. Chem. 2012, 287, 33094–33103.
2013, 305, G225–G240. 177. Phang, J. M.; Liu, W.; Hancock, C. Bridging Epigenetics
168. Rothenberg, M. E.; Doepker, M. P.; Lewkowich, I. P.; et al. and Metabolism: Role of Non-Essential Amino Acids.
Cationic Amino Acid Transporter 2 Regulates Inflammatory Epigenetics 2013, 8, 231–236.
Homeostasis in the Lung. Proc. Natl. Acad. Sci. U.S.A. 178. Cesar-Razquin, A.; Snijder, B.; Frappier-Brinton, T.; et al. A
2006, 103, 14895–14900. Call for Systematic Research on Solute Carriers. Cell 2015,
169. Nakaya, M.; Xiao, Y.; Zhou, X.; et al. Inflammatory T 162, 478–487.
Cell Responses Rely on Amino Acid Transporter ASCT2 179. Consortium, S. T. D.; Williams, A. L.; Jacobs, S. B.; et al.
Facilitation of Glutamine Uptake and mTORC1 Kinase Sequence Variants in SLC16A11 Are a Common Risk Factor
Activation. Immunity 2014, 40, 692–705. for Type 2 Diabetes in Mexico. Nature 2014, 506, 97–101.
170. Mariotta, L.; Ramadan, T.; Singer, D.; et al. T-Type
180. Rusu, V.; Hoch, E.; Mercader, J. M.; et al. Type 2 Diabetes
Amino Acid Transporter TAT1 (Slc16a10) Is Essential for Variants Disrupt Function of SLC16A11 through Two
Extracellular Aromatic Amino Acid Homeostasis Control. J. Distinct Mechanisms. Cell 2017, 170, 199–212.e20.
Physiol. 2012, 590, 6413–6424. 181. Shimamoto, K.; Lebrun, B.; Yasuda-Kamatani, Y.; et al.

171. Jayasimhan, A.; Mansour, K. P.; Slattery, R. M. Advances DL-Threo-Beta-Benzyloxyaspartate, a Potent Blocker of
in Our Understanding of the Pathophysiology of Type 1 Excitatory Amino Acid Transporters. Mol. Pharmacol. 1998,
Diabetes: Lessons from the NOD Mouse. Clin. Sci. (Lond.) 53, 195–201.
2014, 126, 1–18. 182. Reyes, N.; Ginter, C.; Boudker, O. Transport Mechanism of
172. Elso, C. M.; Chu, E. P. F.; Alsayb, M. A.; et al. Sleeping a Bacterial Homologue of Glutamate Transporters. Nature
Beauty Transposon Mutagenesis as a Tool for Gene 2009, 462, 880–885.
Discovery in the NOD Mouse Model of Type 1 Diabetes.
G3 2015, 5, 2903–2911.
173. Van Winkle, L. J. Amino Acid Transport in Developing
Animal Oocytes and Early Conceptuses. Biochim. Biophys.
Acta 1988, 947, 173–208.

You might also like