Comprehensive Physiology Wiley Online Library

Transport of Amino Acids in the Kidney

Full Article on Wiley Online Library



Abstract

Amino acids are the building blocks of proteins and key intermediates in the synthesis of biologically important molecules, as well as energy sources, neurotransmitters, regulators of cellular metabolism, etc. The efficient recovery of amino acids from the primary filtrate is a well‐conserved key role of the kidney proximal tubule. Additionally, renal metabolism participates in the whole body disposition of amino acids. Therefore, a wide array of axially heterogeneously expressed transporters is localized on both epithelial membranes. For transepithelial transport, luminal uptake, which is carried out mainly by active symporters, is coupled with a mostly passive basolateral efflux. Many transporters require partner proteins for appropriate localization, or to modulate transporter activity, and/or increase substrate supply. Interacting proteins include cell surface antigens (CD98), endoplasmic reticulum proteins (GTRAP3‐18 or 41), or enzymes (ACE2 and aminopeptidase N). In the past two decades, the molecular identification of transporters has led to significant advances in our understanding of amino acid transport and aminoacidurias arising from defects in renal transport. Furthermore, the three‐dimensional crystal structures of bacterial homologues have been used to yield new insights on the structure and function of mammalian transporters. Additionally, transgenic animal models have contributed to our understanding of the role of amino acid transporters in the kidney and other organs and/or at critical developmental stages. Progress in elucidation of the renal contribution to systemic amino acid homeostasis requires further integration of kinetic, regulatory, and expression data of amino acid transporters into our understanding of physiological regulatory networks controlling metabolism. © 2014 American Physiological Society. Compr Physiol 4:367‐403, 2014.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. The role of the kidney in total body amino acid homeostasis. This schematic representation of the main amino acid fluxes through the body shows that amino acids absorbed in the intestine reach the systemic circulation via the liver. Following a single meal the amount ingested may exceed the quantity found in the extracellular space by several fold. Therefore, the rapid cellular uptake and metabolic use of amino acids play central homeostatic roles. Liver, muscles, intestine, and kidney are major sites of amino acid metabolism. Additionally, to prevent their loss the kidneys reabsorb approximately 50 g/day of amino acids from the primary urine.
Figure 2. Figure 2. Scheme of nephron segments involved in amino acid reabsorption. Amino acids are freely filtered at the glomerulus and approximately 99.5% are reabsorbed in the proximal tubule (PT, blue color), mainly in the first and second segments (S1 and S2). Notable exceptions in humans are serine, glycine, histidine, and taurine, which lose a higher fraction in the urine.
Figure 3. Figure 3. Cellular model for the reabsorption of amino acids across a proximal tubule cells. Panel A shows a schematic representation of the luminal and basolateral amino acid transporters. Most amino acids are transported across the luminal membrane by symporters that use the electro‐chemical gradient of Na+ or H+ for driving the influx of amino acids. Neutral amino acids may be recycled to the lumen by the exchanger b0,+AT‐rBAT (SLC7A9‐SLC3A1) to allow the uptake of cationic amino acids and cystine. The basolateral efflux of most nonessential neutral amino acids and of cationic amino acids is mediated by antiporters. Essential amino acids taken up in exchange for cationic amino acid efflux may recycle to the extracellular space via selective antiporters. Panel B depicts most of the known renal transport proteins; their protein and human gene names are indicated. In mice most of the transporters are expressed mainly in the early segments of the proximal tubule (S1 and S2). However, SIT1 (SLC6A20) is expressed along the entire proximal tubule, and B0AT3 (SLC6A18) and EAAT3 (SLC1A1), are expressed in the later proximal tubule (S2 and S3). Key: AA0 neutral amino acid, AA anionic amino acid, AA+ cationic amino acid, TMEM27 (collectrin).
Figure 4. Figure 4. Transport mechanisms: symporters, antiporters, and uniporters. Solutes are transported by secondary active transporters by an alternated access mechanism. The number of steps and solutes involved depends on the transport mechanism (uniporter, symporter, or antiporter). Transport mechanisms are classified based on the number of substrates simultaneously transported and the relative direction of substrate movements. (A and B) Alternative access mechanism for symporters and antiporters. (A) Symporters: Upon substrate (red circle) and ion (blue square with red center) binding to the open outward‐facing state (2), the substrate‐ion‐bound transporter changes to the occluded and inward‐facing states (172) releasing substrates. A transition between the unbound inward‐ and outward‐facing conformations is required to renew the transport cycle (4 and 1). For antiporters (B), substrate binding to the outward‐facing state (blue diamond) results in the transition to the inward‐facing states (172) and substrate release. Restoration of the outward‐facing state (4 to 1) requires the binding of a substrate to the inward‐facing transporter state (red circle) (5) to allow the transporter to return to the outward‐facing conformation and release the substrate (6). (C) Chart of each of transporter mechanism. Uniporters, move one solute along the solute concentration gradient; Symporters, translocate two or more substrates in the same direction, one solute (e.g., AA) eventually against its concentration gradient and the other solute (e.g., Na+) along its concentration gradient; Antiporters, translocate two substrates in opposite directions against the gradient concentration of one of the substrates. For symporters and antiporters, the presence of both substrates involved in the transport cycle is required for the translocation. All three mechanisms are utilitzed by amino acid and oligopeptide transporters. TAT1 (SLC16A10) is an example of an aromatic amino acid uniporter. B0AT1 (SLC6A19) is a symporter, cotranslocating neutral amino acids with sodium. PEPT1 (SLC15A1) is another symporter that cotransports peptides and protons. The heterodimeric amino acid transporter LAT2‐4F2hc (SLC7A8‐ SLC3A2) is an antiporter that exchanges neutral amino acids.
Figure 5. Figure 5. Chimeras and cysteine scanning experimental strategies to study the structure function relationship of solute transporters. Functional heterodimeric amino acid transporters (HATs) are formed by SLC7 transporters interacting covalently with accessory proteins of the SLC3 family, rBAT (SLC3A1) or 4F2hc (SLC3A2). To date rBAT has only been confirmed to interact with one SLC7 family member, b0,+ (SLC7A9); while 4F2hc interacts with several members including LAT1 (Slc7a5). (A) Schematic representation of chimeras and truncations of rBAT and 4F2hc. To define the domain(s) of rBAT and 4F2hc involved in the recognition and interaction with the Slc7 transporters, chimeras, and truncations of rBAT and 4F2hc were constructed and in combination with b0,+ and LAT1 functionally assayed. The chimeras were prepared by combining the following protein regions: cytoplasmic tail, transmembrane domain, neck, and glycosidase‐like domain. rBAT is represented in blue and 4F2hc in yellow. (B) L‐Arginine transport by b+0AT coexpressed with various chimeras. When coexpressed with b0,+AT, chimeras containing rBAT cytoplasmic tail (BB44, BB4B) with or without the transmembrane domain (BBB4) transport L‐arginine indicating a role in the interaction between rBAT with b0,+AT [adapted, with permission, from (70)]. (C) Schematic representation of targeted SERT residues. “Cysteine scanning” or the systematic introduction of cysteine or lysine mutations is another method for studying the relationship of specific residues to transporter function. To probe conformational changes occurring during the transport cycle the targeted residues are evaluated for cytoplasmic or extracellular accessibility and/or impact on transport function or protein expression. Rudnick and collaborators [adapted from reference (161) with permission] extensively studied the serotonine transporter (SERT/ SLC6A2), which is responsible for reuptake of 5‐hydroxytryptamine (5‐HT, Serotonin) in the postsynaptic membrane and is a target for antidepressants such as Fluoxetine (Prozac). The scheme for SERT is based on the LeuT model (discussed in the text). Residues studied by cysteine scanning are labeled in red. Combining the structural predictions based on the bacterial homologs and the cysteine scan, the authors suggested several possible protein conformations during the transport cycle that allow substrate binding and dissociation from both sides of the membrane.
Figure 6. Figure 6. The heterodimeric amino acid family. The HAT family is formed by glycoproteins from the SLC3 (rBAT and 4F2hc) family and amino acid transporters from the SLC7 family. (A) Chart of known SLC3 transporter partners. 4F2 interacts with several SLC7 transporters while rBAT is only known to interact with b0,+AT. (B) Schematic representation of covalent interaction between SLC3 and SLC7 members. The SLC3 heavy subunits (pink), which are type II membrane glycoproteins with an intracellular NH2 terminus and a single transmembrane domain, and the SLC7 transporters [light subunit (blue)] are linked by a disulfide bridge (yellow) with conserved cysteine residues (e.g., cysteine 158 for the human xCT and cysteine 109 for human 4F2hc). [Adapted, with permission, from (140).]
Figure 7. Figure 7. Interaction of SLC6 family members with the Renin‐Angiotensin system proteins. (A) Schematic representation of B0AT1 interaction with RAS members ACE2 and TMEM27. ACE2 and TMEM27/Collectrin are type I integral membrane proteins with N‐terminal (N), transmembrane, and short C‐terminal (C) domains. ACE2 has one zinc‐binding motif (HEMGH) (bm) in the extracellular domain and is a carboxipeptidase with the consensus sequence P(Φ)1‐3PΦ/+. The bradikinin‐(1‐8) peptide depicted (reversed) is one of the best in vivo targets of ACE2 (84). (B‐J) ACE2 and Tmem27/collectrin tissue specific colocalization with B0AT1. Representative immunofluorescent images of mouse tissue sections labeled with antibodies against B0AT1 (red) in small intestine (C) and kidney (E), and against Ace2 in small intestine (B) and Tmem27/collectrin in kidney (D). Panel (F) shows colocalization of B0AT1 and Ace2 in small intestine enterocytes (G) the colocalization of B0AT1 with Tmem27/collectrin in the proximal tubules. (H and I) Ace2 and Tmem27/collectrin knockout ablates B0AT1 expression in the small intestine or kidney, respectively. ACE2 and TMEM27 are encoded by X chromosome‐linked genes. Western blots of small intestine (H) and kidney (I) tissue lysates from wild‐type vs ace2−/y and coll−/y knockout mice probed with anti‐B0AT1 and anti‐β‐actin (loading control) antibodies (36,52). (J‐M) SIT1 colocalizes and functionally interacts with ACE2. SIT1 (SLC6A20) colocalizes with ACE2 in human small intestine (J, K, and L) and its coexpression with ACE2 in Xenopus laevis oocytes stimulates SIT1 transport (M).
Figure 8. Figure 8. Functional interaction between LAT2‐4F2 and TAT1. (A) Schematic representation of the functional interaction between LAT2‐4F2 and TAT1. LAT2‐4F2 recycles aromatic amino acids effluxed by TAT1 in exchange for efflux of neutral amino acids, which are not TAT1 substrates. (B) TAT1 does not physically associate with 4F2hc or LAT2. Coimmunoprecipitation was performed using lysates of biosynthetically 35S‐met labeled oocytes coinjected with 4F2hc, LAT2, and TAT1 cRNAs (LT) or noninjected oocytes (NI) and analyzed by autoradiograph. Western blot (WB) of total lysates, or lysates immunoprecipitated using anti‐4F2 (IP‐4F2hc) antibodies detected LAT2 but not TAT1 coimmunopreciptated with 4F2hc. (C) Coexpression of TAT1 and LAT2‐4F2hc stimulates efflux of LAT2‐4F2 substrates that are not TAT1 substrates (e.g., L‐Gln). The amount of amino acids accumulating in the extracellular bath of oocytes with and without expression of LAT2‐4F2 and/or TAT1 was analyzed by UPLC [for details, see (150)]. The efflux of LAT2‐4F2 substrates (yellow) such as L‐glutamine, asparagine, serine and alanine was increased in the presence of TAT1. However, TAT1 (blue) substrate concentrations were not altered by coexpression of 4F2hc with TAT1. [Figure adapted from reference (150) with permission].
Figure 9. Figure 9. Expression of amino acid and oligopeptide transporters in kidney by region. (A) Scheme of proximal tubule. The proximal tubule segments extend from the kidney cortex to the outer medullary stripe (OM) of the medulla. The renal arcuate artery (red line) and vein (blue line) demarking the separation between the cortex and medulla are indicated. The glomerulus is indicated with (G). The proximal convoluted (S1) and (S2), and the proximal straight (S3) segments are separated with straight dotted lines. (B‐E) Reported expression of accessory proteins and transporters in proximal tubule segments S1 to S3. Panels B and C give expression data for apical, and panels D and E for basolateral localized proteins. (F) Scheme of distal nephron regions. The arcuate artery and veins separating cortex and medulla are indicated as in panel A, and the outer and inner medulla is separated by curved dotted lines. The thick ascending limb, distal convoluted tubule (DCT), connecting tubule (CNT) and collecting duct are indicated. (G and H) Reported expression of accessory proteins and transporters in distal nephron regions. The data shown for transporter and accessory protein expression in distal segments are incomplete and reflect cases in which the level of expression reported suggests a significant physiological role. No subcellular localization data are provided. Where no protein data are available, the mRNA expression is represented. For all panels (B‐E, G, and H) the data are represented using an arbitary scale of 0 to 100% expression—white being no reported expression with increasing expression indicated by darker colors. The relative expression of different genes is not represented. For all genes, a reference is given for the expression data represented.
Figure 10. Figure 10. Transporter basolateral versus apical membrane localization in kidney proximal tubule segments. (A‐I) Representative immunofluorescence images from labeled mouse kidney tissue sections of apical membrane amino acid transporters. Tissue sections were stained as follows: (A) B0AT1, (B) B0AT3, and (C) SIT1 are labeled in red, and in (A‐C) the basolateral membrane transporter, 4F2hc, is labeled with green (157). For panel (D), collectrin is labeled in green and shown without a labeled basolateral transporter (180). (E and F) The opposing axial distribution along proximal kidney tubule of mRNA for the HAT catalytic subunits, b0,+AT, and rBAT, its glycoprotein partner. In situ hybridization of kidney section shown at a low magnification to demonstrate the individual HAT subunit mRNA gradients from cortex (c) to the medullary outer strip (os) and inner stripe (is) kidney regions (145). (G) Scheme of the apical versus basolateral localization of transporters expressed in kidney proximal tubules. Direction of transport and transporter mechanism is represented by arrows. Evidence exists for the expression of an as yet unidentified basolateral symporter and facilitative diffusion transporter(s) indicated as unlabeled transporters. (H‐I) Representative immunofluorescence images from stained mouse kidney tissue sections of basolateral localized amino acid transporters. The basolateral localized (H) SNAT3 (126) is labeled in red, (I) TAT1 (151) is labled in green, (J) LAT2 localization and (K) y+LAT1 transporters are labeled in red and (L) 4F2hc is labeled in green (126).
Figure 11. Figure 11. Renal amino acid metabolism in the proximal tubule. These diagrams illustrate some of the enzymes and transporters involved in the proximal tubule metabolism of Gln and Glu in panel A, and Arg, Cit, and NO in panel B. While the subcellular localization of proteins is indicated, the relative contributions of individual pathways to amino acid metabolism and transport is not shown (please refer to the text for more information). (A) Overview of L‐glutamine and L‐glutamate metabolic pathways and transporters in renal proximal tubule. Briefly L‐glutamine (Q) is taken up on the apical membrane by B0AT1 (SLC6A19) and B0AT3 (SLCA18) and L‐glutamate (E) is transported by EAAT3 (SLC1A1). Basolateral SNAT3 (SLC38A3) transports Q bidirectionally depending on the relative cytosolic versus extracellular Q concentration and pH. In the proximal tubule ammonia and ammonium (H+/NH4 +/NH3,) are preferentially exported to the lumen by a number of systems. Here the apical NHE3 (SLC9A3) transporter is shown. NHE3 mediates H+ efflux, which may combine with NH3 in the lumen to form NH4 +. NHE3 has also been reported to transport NH4 +. On the apical membrane Q of distal proximal tubule segments can be transaminated to E by γ‐glutamyl transferase (γ‐GT; EC 2.3.2.2). A number of SLC25 family transporters mediate mitochondrial transport of amino acids and metabolites. Internalized Q is taken up by unknown mitochondrial transporters where the mitochondrial phosphate‐dependent glutaminase (PDG; EC 3.5.1.2) converts Q to E and NH4 +. Recently, NH4 + has been shown to be potentially transported by Aquaporin 8, which localized to the inner mitochondrial membrane. A further conversion of E to α‐KG and a NH4 + can be catalyzed by glutamate dehydrogenase 1 (GDH1; EC 1.4.1.3) or E can be decarboxylated by glutamate decarboxylase (GAD; EC 4.1.1.15) to GABA and a bicarbonate ion (HCO3 ). Alternatively glutamic‐oxaloacetic transminase (GOT; EC 2.6.1.1) produces from E and pyruvate α‐KG and L‐aspartate (D). Mitochondrial TCA metabolism of α‐KG produces intermediates such as malate which can be exported to the cytoplasm for gluconeogeneisis. In the cytosol phosphenolpyruvate carboxykinase (PEPCK; EC 4.1.1.32) produces phosphoenolpyruvate (PEP) and CO2 for gluconeogenesis and pyruvate formation. Finally, cytosolic glutamine synthetase (GS; E.C. 6.3.1.2) can convert E and NH4 + to Q. (B) Proximal tubule transporters and enzymatic pathways in renal arginine metabolism. To summarize, apical b0+‐rBAT (SLC7A9‐SLC3A2) transporters uptake L‐arginine (R) in exchange for neutral amino acids (AA0), one source of AA0 is luminal B0AT1 (SLC6A19), which also takes up citrulline. On the basolateral membrane the anionic exchanger OAT1 (SLC22A6) is likely responsible for a large part of the citrulline uptake. The basolateral transporter TAT1 (SLC16A10) provides a facilitative diffusion pathway for aromatic AA0 which can be recycled by y+LAT1 (SLC7A7) in exchange for R efflux. Additionally, R may be exported by basolateral CAT1 (SLC7A1). In the proximal tubule cytoplasm, citrulline together with L‐aspartate (D) is converted to R and fumarate via the sequential action of arginosuccinate synthetase (ASS1; EC 6.3.4.5) and arginosuccinate lyase (ASL; EC 4.3.2.1). Nitric oxide synthase (NOS; EC 1.14.13.39) catalyses the production of nitric oxide (NO) and citrulline from R. Mitochondrial SLC25 family transport proteins exchange ornithine and R for citrulline. Within the mitorchondrial matrix ornithine can be converted to citrulline by ornithine transcarbamoylase (OTC: EC 2.1.3.3). Intramitochrondrial metabolism of R by arginine:glycine amidotransferase (AGAT; EC 2.1.4.1) produces ornithine and guanidinoacetate (GAA). R can also be decarboxylated by arginine decarboxylase (ADC; EC 4.1.19) to agmatine and CO2. GAA can also be produced in the cytoplasm from R by cytosolic AGAT. GAA is released in the blood stream for creatine production. Mitochondrial argininase II (AII; EC 3.5.1.3) hydrolyzes R to produce ornithine. Transporters are indicated with ovals. Plasma membrane transporters, when known, are labeled with common and SLC names and the substrates translocation indicated with solid lines. Mitochondrial transporters are not labeled. Enyzmatic pathways are schematic and not all substrate intermediates are shown. Direct enzymatic pathways are shown with solid lines. Dotted lines indicate either translocation of substrates, for example, through the cytoplasm or extracellular space, or multiple metabolic steps. For more information regarding specific enzymes please refer to the Enzyme Commission (EC) numbers indicated in the figure legend.


Figure 1. The role of the kidney in total body amino acid homeostasis. This schematic representation of the main amino acid fluxes through the body shows that amino acids absorbed in the intestine reach the systemic circulation via the liver. Following a single meal the amount ingested may exceed the quantity found in the extracellular space by several fold. Therefore, the rapid cellular uptake and metabolic use of amino acids play central homeostatic roles. Liver, muscles, intestine, and kidney are major sites of amino acid metabolism. Additionally, to prevent their loss the kidneys reabsorb approximately 50 g/day of amino acids from the primary urine.


Figure 2. Scheme of nephron segments involved in amino acid reabsorption. Amino acids are freely filtered at the glomerulus and approximately 99.5% are reabsorbed in the proximal tubule (PT, blue color), mainly in the first and second segments (S1 and S2). Notable exceptions in humans are serine, glycine, histidine, and taurine, which lose a higher fraction in the urine.


Figure 3. Cellular model for the reabsorption of amino acids across a proximal tubule cells. Panel A shows a schematic representation of the luminal and basolateral amino acid transporters. Most amino acids are transported across the luminal membrane by symporters that use the electro‐chemical gradient of Na+ or H+ for driving the influx of amino acids. Neutral amino acids may be recycled to the lumen by the exchanger b0,+AT‐rBAT (SLC7A9‐SLC3A1) to allow the uptake of cationic amino acids and cystine. The basolateral efflux of most nonessential neutral amino acids and of cationic amino acids is mediated by antiporters. Essential amino acids taken up in exchange for cationic amino acid efflux may recycle to the extracellular space via selective antiporters. Panel B depicts most of the known renal transport proteins; their protein and human gene names are indicated. In mice most of the transporters are expressed mainly in the early segments of the proximal tubule (S1 and S2). However, SIT1 (SLC6A20) is expressed along the entire proximal tubule, and B0AT3 (SLC6A18) and EAAT3 (SLC1A1), are expressed in the later proximal tubule (S2 and S3). Key: AA0 neutral amino acid, AA anionic amino acid, AA+ cationic amino acid, TMEM27 (collectrin).


Figure 4. Transport mechanisms: symporters, antiporters, and uniporters. Solutes are transported by secondary active transporters by an alternated access mechanism. The number of steps and solutes involved depends on the transport mechanism (uniporter, symporter, or antiporter). Transport mechanisms are classified based on the number of substrates simultaneously transported and the relative direction of substrate movements. (A and B) Alternative access mechanism for symporters and antiporters. (A) Symporters: Upon substrate (red circle) and ion (blue square with red center) binding to the open outward‐facing state (2), the substrate‐ion‐bound transporter changes to the occluded and inward‐facing states (172) releasing substrates. A transition between the unbound inward‐ and outward‐facing conformations is required to renew the transport cycle (4 and 1). For antiporters (B), substrate binding to the outward‐facing state (blue diamond) results in the transition to the inward‐facing states (172) and substrate release. Restoration of the outward‐facing state (4 to 1) requires the binding of a substrate to the inward‐facing transporter state (red circle) (5) to allow the transporter to return to the outward‐facing conformation and release the substrate (6). (C) Chart of each of transporter mechanism. Uniporters, move one solute along the solute concentration gradient; Symporters, translocate two or more substrates in the same direction, one solute (e.g., AA) eventually against its concentration gradient and the other solute (e.g., Na+) along its concentration gradient; Antiporters, translocate two substrates in opposite directions against the gradient concentration of one of the substrates. For symporters and antiporters, the presence of both substrates involved in the transport cycle is required for the translocation. All three mechanisms are utilitzed by amino acid and oligopeptide transporters. TAT1 (SLC16A10) is an example of an aromatic amino acid uniporter. B0AT1 (SLC6A19) is a symporter, cotranslocating neutral amino acids with sodium. PEPT1 (SLC15A1) is another symporter that cotransports peptides and protons. The heterodimeric amino acid transporter LAT2‐4F2hc (SLC7A8‐ SLC3A2) is an antiporter that exchanges neutral amino acids.


Figure 5. Chimeras and cysteine scanning experimental strategies to study the structure function relationship of solute transporters. Functional heterodimeric amino acid transporters (HATs) are formed by SLC7 transporters interacting covalently with accessory proteins of the SLC3 family, rBAT (SLC3A1) or 4F2hc (SLC3A2). To date rBAT has only been confirmed to interact with one SLC7 family member, b0,+ (SLC7A9); while 4F2hc interacts with several members including LAT1 (Slc7a5). (A) Schematic representation of chimeras and truncations of rBAT and 4F2hc. To define the domain(s) of rBAT and 4F2hc involved in the recognition and interaction with the Slc7 transporters, chimeras, and truncations of rBAT and 4F2hc were constructed and in combination with b0,+ and LAT1 functionally assayed. The chimeras were prepared by combining the following protein regions: cytoplasmic tail, transmembrane domain, neck, and glycosidase‐like domain. rBAT is represented in blue and 4F2hc in yellow. (B) L‐Arginine transport by b+0AT coexpressed with various chimeras. When coexpressed with b0,+AT, chimeras containing rBAT cytoplasmic tail (BB44, BB4B) with or without the transmembrane domain (BBB4) transport L‐arginine indicating a role in the interaction between rBAT with b0,+AT [adapted, with permission, from (70)]. (C) Schematic representation of targeted SERT residues. “Cysteine scanning” or the systematic introduction of cysteine or lysine mutations is another method for studying the relationship of specific residues to transporter function. To probe conformational changes occurring during the transport cycle the targeted residues are evaluated for cytoplasmic or extracellular accessibility and/or impact on transport function or protein expression. Rudnick and collaborators [adapted from reference (161) with permission] extensively studied the serotonine transporter (SERT/ SLC6A2), which is responsible for reuptake of 5‐hydroxytryptamine (5‐HT, Serotonin) in the postsynaptic membrane and is a target for antidepressants such as Fluoxetine (Prozac). The scheme for SERT is based on the LeuT model (discussed in the text). Residues studied by cysteine scanning are labeled in red. Combining the structural predictions based on the bacterial homologs and the cysteine scan, the authors suggested several possible protein conformations during the transport cycle that allow substrate binding and dissociation from both sides of the membrane.


Figure 6. The heterodimeric amino acid family. The HAT family is formed by glycoproteins from the SLC3 (rBAT and 4F2hc) family and amino acid transporters from the SLC7 family. (A) Chart of known SLC3 transporter partners. 4F2 interacts with several SLC7 transporters while rBAT is only known to interact with b0,+AT. (B) Schematic representation of covalent interaction between SLC3 and SLC7 members. The SLC3 heavy subunits (pink), which are type II membrane glycoproteins with an intracellular NH2 terminus and a single transmembrane domain, and the SLC7 transporters [light subunit (blue)] are linked by a disulfide bridge (yellow) with conserved cysteine residues (e.g., cysteine 158 for the human xCT and cysteine 109 for human 4F2hc). [Adapted, with permission, from (140).]


Figure 7. Interaction of SLC6 family members with the Renin‐Angiotensin system proteins. (A) Schematic representation of B0AT1 interaction with RAS members ACE2 and TMEM27. ACE2 and TMEM27/Collectrin are type I integral membrane proteins with N‐terminal (N), transmembrane, and short C‐terminal (C) domains. ACE2 has one zinc‐binding motif (HEMGH) (bm) in the extracellular domain and is a carboxipeptidase with the consensus sequence P(Φ)1‐3PΦ/+. The bradikinin‐(1‐8) peptide depicted (reversed) is one of the best in vivo targets of ACE2 (84). (B‐J) ACE2 and Tmem27/collectrin tissue specific colocalization with B0AT1. Representative immunofluorescent images of mouse tissue sections labeled with antibodies against B0AT1 (red) in small intestine (C) and kidney (E), and against Ace2 in small intestine (B) and Tmem27/collectrin in kidney (D). Panel (F) shows colocalization of B0AT1 and Ace2 in small intestine enterocytes (G) the colocalization of B0AT1 with Tmem27/collectrin in the proximal tubules. (H and I) Ace2 and Tmem27/collectrin knockout ablates B0AT1 expression in the small intestine or kidney, respectively. ACE2 and TMEM27 are encoded by X chromosome‐linked genes. Western blots of small intestine (H) and kidney (I) tissue lysates from wild‐type vs ace2−/y and coll−/y knockout mice probed with anti‐B0AT1 and anti‐β‐actin (loading control) antibodies (36,52). (J‐M) SIT1 colocalizes and functionally interacts with ACE2. SIT1 (SLC6A20) colocalizes with ACE2 in human small intestine (J, K, and L) and its coexpression with ACE2 in Xenopus laevis oocytes stimulates SIT1 transport (M).


Figure 8. Functional interaction between LAT2‐4F2 and TAT1. (A) Schematic representation of the functional interaction between LAT2‐4F2 and TAT1. LAT2‐4F2 recycles aromatic amino acids effluxed by TAT1 in exchange for efflux of neutral amino acids, which are not TAT1 substrates. (B) TAT1 does not physically associate with 4F2hc or LAT2. Coimmunoprecipitation was performed using lysates of biosynthetically 35S‐met labeled oocytes coinjected with 4F2hc, LAT2, and TAT1 cRNAs (LT) or noninjected oocytes (NI) and analyzed by autoradiograph. Western blot (WB) of total lysates, or lysates immunoprecipitated using anti‐4F2 (IP‐4F2hc) antibodies detected LAT2 but not TAT1 coimmunopreciptated with 4F2hc. (C) Coexpression of TAT1 and LAT2‐4F2hc stimulates efflux of LAT2‐4F2 substrates that are not TAT1 substrates (e.g., L‐Gln). The amount of amino acids accumulating in the extracellular bath of oocytes with and without expression of LAT2‐4F2 and/or TAT1 was analyzed by UPLC [for details, see (150)]. The efflux of LAT2‐4F2 substrates (yellow) such as L‐glutamine, asparagine, serine and alanine was increased in the presence of TAT1. However, TAT1 (blue) substrate concentrations were not altered by coexpression of 4F2hc with TAT1. [Figure adapted from reference (150) with permission].


Figure 9. Expression of amino acid and oligopeptide transporters in kidney by region. (A) Scheme of proximal tubule. The proximal tubule segments extend from the kidney cortex to the outer medullary stripe (OM) of the medulla. The renal arcuate artery (red line) and vein (blue line) demarking the separation between the cortex and medulla are indicated. The glomerulus is indicated with (G). The proximal convoluted (S1) and (S2), and the proximal straight (S3) segments are separated with straight dotted lines. (B‐E) Reported expression of accessory proteins and transporters in proximal tubule segments S1 to S3. Panels B and C give expression data for apical, and panels D and E for basolateral localized proteins. (F) Scheme of distal nephron regions. The arcuate artery and veins separating cortex and medulla are indicated as in panel A, and the outer and inner medulla is separated by curved dotted lines. The thick ascending limb, distal convoluted tubule (DCT), connecting tubule (CNT) and collecting duct are indicated. (G and H) Reported expression of accessory proteins and transporters in distal nephron regions. The data shown for transporter and accessory protein expression in distal segments are incomplete and reflect cases in which the level of expression reported suggests a significant physiological role. No subcellular localization data are provided. Where no protein data are available, the mRNA expression is represented. For all panels (B‐E, G, and H) the data are represented using an arbitary scale of 0 to 100% expression—white being no reported expression with increasing expression indicated by darker colors. The relative expression of different genes is not represented. For all genes, a reference is given for the expression data represented.


Figure 10. Transporter basolateral versus apical membrane localization in kidney proximal tubule segments. (A‐I) Representative immunofluorescence images from labeled mouse kidney tissue sections of apical membrane amino acid transporters. Tissue sections were stained as follows: (A) B0AT1, (B) B0AT3, and (C) SIT1 are labeled in red, and in (A‐C) the basolateral membrane transporter, 4F2hc, is labeled with green (157). For panel (D), collectrin is labeled in green and shown without a labeled basolateral transporter (180). (E and F) The opposing axial distribution along proximal kidney tubule of mRNA for the HAT catalytic subunits, b0,+AT, and rBAT, its glycoprotein partner. In situ hybridization of kidney section shown at a low magnification to demonstrate the individual HAT subunit mRNA gradients from cortex (c) to the medullary outer strip (os) and inner stripe (is) kidney regions (145). (G) Scheme of the apical versus basolateral localization of transporters expressed in kidney proximal tubules. Direction of transport and transporter mechanism is represented by arrows. Evidence exists for the expression of an as yet unidentified basolateral symporter and facilitative diffusion transporter(s) indicated as unlabeled transporters. (H‐I) Representative immunofluorescence images from stained mouse kidney tissue sections of basolateral localized amino acid transporters. The basolateral localized (H) SNAT3 (126) is labeled in red, (I) TAT1 (151) is labled in green, (J) LAT2 localization and (K) y+LAT1 transporters are labeled in red and (L) 4F2hc is labeled in green (126).


Figure 11. Renal amino acid metabolism in the proximal tubule. These diagrams illustrate some of the enzymes and transporters involved in the proximal tubule metabolism of Gln and Glu in panel A, and Arg, Cit, and NO in panel B. While the subcellular localization of proteins is indicated, the relative contributions of individual pathways to amino acid metabolism and transport is not shown (please refer to the text for more information). (A) Overview of L‐glutamine and L‐glutamate metabolic pathways and transporters in renal proximal tubule. Briefly L‐glutamine (Q) is taken up on the apical membrane by B0AT1 (SLC6A19) and B0AT3 (SLCA18) and L‐glutamate (E) is transported by EAAT3 (SLC1A1). Basolateral SNAT3 (SLC38A3) transports Q bidirectionally depending on the relative cytosolic versus extracellular Q concentration and pH. In the proximal tubule ammonia and ammonium (H+/NH4 +/NH3,) are preferentially exported to the lumen by a number of systems. Here the apical NHE3 (SLC9A3) transporter is shown. NHE3 mediates H+ efflux, which may combine with NH3 in the lumen to form NH4 +. NHE3 has also been reported to transport NH4 +. On the apical membrane Q of distal proximal tubule segments can be transaminated to E by γ‐glutamyl transferase (γ‐GT; EC 2.3.2.2). A number of SLC25 family transporters mediate mitochondrial transport of amino acids and metabolites. Internalized Q is taken up by unknown mitochondrial transporters where the mitochondrial phosphate‐dependent glutaminase (PDG; EC 3.5.1.2) converts Q to E and NH4 +. Recently, NH4 + has been shown to be potentially transported by Aquaporin 8, which localized to the inner mitochondrial membrane. A further conversion of E to α‐KG and a NH4 + can be catalyzed by glutamate dehydrogenase 1 (GDH1; EC 1.4.1.3) or E can be decarboxylated by glutamate decarboxylase (GAD; EC 4.1.1.15) to GABA and a bicarbonate ion (HCO3 ). Alternatively glutamic‐oxaloacetic transminase (GOT; EC 2.6.1.1) produces from E and pyruvate α‐KG and L‐aspartate (D). Mitochondrial TCA metabolism of α‐KG produces intermediates such as malate which can be exported to the cytoplasm for gluconeogeneisis. In the cytosol phosphenolpyruvate carboxykinase (PEPCK; EC 4.1.1.32) produces phosphoenolpyruvate (PEP) and CO2 for gluconeogenesis and pyruvate formation. Finally, cytosolic glutamine synthetase (GS; E.C. 6.3.1.2) can convert E and NH4 + to Q. (B) Proximal tubule transporters and enzymatic pathways in renal arginine metabolism. To summarize, apical b0+‐rBAT (SLC7A9‐SLC3A2) transporters uptake L‐arginine (R) in exchange for neutral amino acids (AA0), one source of AA0 is luminal B0AT1 (SLC6A19), which also takes up citrulline. On the basolateral membrane the anionic exchanger OAT1 (SLC22A6) is likely responsible for a large part of the citrulline uptake. The basolateral transporter TAT1 (SLC16A10) provides a facilitative diffusion pathway for aromatic AA0 which can be recycled by y+LAT1 (SLC7A7) in exchange for R efflux. Additionally, R may be exported by basolateral CAT1 (SLC7A1). In the proximal tubule cytoplasm, citrulline together with L‐aspartate (D) is converted to R and fumarate via the sequential action of arginosuccinate synthetase (ASS1; EC 6.3.4.5) and arginosuccinate lyase (ASL; EC 4.3.2.1). Nitric oxide synthase (NOS; EC 1.14.13.39) catalyses the production of nitric oxide (NO) and citrulline from R. Mitochondrial SLC25 family transport proteins exchange ornithine and R for citrulline. Within the mitorchondrial matrix ornithine can be converted to citrulline by ornithine transcarbamoylase (OTC: EC 2.1.3.3). Intramitochrondrial metabolism of R by arginine:glycine amidotransferase (AGAT; EC 2.1.4.1) produces ornithine and guanidinoacetate (GAA). R can also be decarboxylated by arginine decarboxylase (ADC; EC 4.1.19) to agmatine and CO2. GAA can also be produced in the cytoplasm from R by cytosolic AGAT. GAA is released in the blood stream for creatine production. Mitochondrial argininase II (AII; EC 3.5.1.3) hydrolyzes R to produce ornithine. Transporters are indicated with ovals. Plasma membrane transporters, when known, are labeled with common and SLC names and the substrates translocation indicated with solid lines. Mitochondrial transporters are not labeled. Enyzmatic pathways are schematic and not all substrate intermediates are shown. Direct enzymatic pathways are shown with solid lines. Dotted lines indicate either translocation of substrates, for example, through the cytoplasm or extracellular space, or multiple metabolic steps. For more information regarding specific enzymes please refer to the Enzyme Commission (EC) numbers indicated in the figure legend.
References
 1. Abplanalp J , Laczko E , Philp NJ , Neidhardt J , Zuercher J , Braun P , Schorderet DF , Munier FL , Verrey F , Berger W , Camargo SM , Kloeckener‐Gruissem B . The cataract and glucosuria associated monocarboxylate transporter MCT12 is a new creatine transporter. Hum Mol Genet 22: 3218‐3226, 2013.
 2. Adibi SA . Renal assimilation of oligopeptides: Physiological mechanisms and metabolic importance. Am J Physiol 272: E723‐736, 1997.
 3. Aoyama K , Wang F , Matsumura N , Kiyonari H , Shioi G , Tanaka K , Kinoshita C , Kikuchi‐Utsumi K , Watabe M , Nakaki T . Increased neuronal glutathione and neuroprotection in GTRAP3‐18‐deficient mice. Neurobiol Dis 45: 973‐982, 2012.
 4. Aoyama K , Watabe M , Nakaki T . Modulation of neuronal glutathione synthesis by EAAC1 and its interacting protein GTRAP3‐18. Amino Acids 42: 163‐169, 2012.
 5. Auray‐Blais C , Cyr D , Drouin R . Quebec neonatal mass urinary screening programme: From micromolecules to macromolecules. J Inherit Metab Dis 30: 515‐521, 2007.
 6. Bailey CG , Ryan RM , Thoeng AD , Ng C , King K , Vanslambrouck JM , Auray‐Blais C , Vandenberg RJ , Broer S , Rasko JE . Loss‐of‐function mutations in the glutamate transporter SLC1A1 cause human dicarboxylic aminoaciduria. J Clin Invest 121: 446‐453, 2011.
 7. Bartoccioni P , Rius M , Zorzano A , Palacin M , Chillaron J . Distinct classes of trafficking rBAT mutants cause the type I cystinuria phenotype. Hum Mol Genet 17: 1845‐1854, 2008.
 8. Bauch C , Forster N , Loffing‐Cueni D , Summa V , Verrey F . Functional cooperation of epithelial heteromeric amino acid transporters expressed in madin‐darby canine kidney cells. J Biol Chem 278: 1316‐1322, 2003.
 9. Bauch C , Verrey F . Apical heterodimeric cystine and cationic amino acid transporter expressed in MDCK cells. Am J Physiol Renal Physiol 283: F181‐189, 2002.
 10. Bergeron M , Vadeboncoeur M . Microinjections of L‐leucine into tubules and peritubular capillaries of the rat. II. The maleic acid model. Nephron 8: 367‐374, 1971.
 11. Bitoun M , Levillain O , Tappaz M . Gene expression of the taurine transporter and taurine biosynthetic enzymes in rat kidney after antidiuresis and salt loading. Pflugers Arch 442: 87‐95, 2001.
 12. Bodoy S , Fotiadis D , Stoeger C , Kanai Y , Palacin M . The small SLC43 family: Facilitator system l amino acid transporters and the orphan EEG1. Mol Aspects Med 34: 638‐645, 2013.
 13. Bodoy S , Martin L , Zorzano A , Palacin M , Estevez R , Bertran J . Identification of LAT4, a novel amino acid transporter with system L activity. J Biol Chem 280: 12002‐12011, 2005.
 14. Bohmer C , Broer A , Munzinger M , Kowalczuk S , Rasko JE , Lang F , Broer S . Characterization of mouse amino acid transporter B0AT1 (slc6a19). Biochem J 389: 745‐751, 2005.
 15. Boll M , Foltz M , Rubio‐Aliaga I , Kottra G , Daniel H . Functional characterization of two novel mammalian electrogenic proton‐dependent amino acid cotransporters. J Biol Chem 277: 22966‐22973, 2002.
 16. Bourke E , Fine A , Scott JM . Glutaminase II pathway in human kidney. Nature: New biology 233: 249‐250, 1971.
 17. Braun D , Wirth EK , Wohlgemuth F , Reix N , Klein MO , Gruters A , Kohrle J , Schweizer U . Aminoaciduria, but normal thyroid hormone levels and signaling, in mice lacking the amino acid and thyroid hormone transporter Slc7a8. Biochem J 2011.
 18. Broer A , Albers A , Setiawan I , Edwards RH , Chaudhry FA , Lang F , Wagner CA , Broer S . Regulation of the glutamine transporter SN1 by extracellular pH and intracellular sodium ions. J Physiol 539: 3‐14, 2002.
 19. Broer A , Juelich T , Vanslambrouck JM , Tietze N , Solomon PS , Holst J , Bailey CG , Rasko JE , Broer S . Impaired nutrient signaling and body weight control in a Na+ neutral amino acid cotransporter (Slc6a19)‐deficient mouse. J Biol Chem 286: 26638‐26651, 2011.
 20. Broer A , Klingel K , Kowalczuk S , Rasko JE , Cavanaugh J , Broer S . Molecular cloning of mouse amino acid transport system B0, a neutral amino acid transporter related to Hartnup disorder. J Biol Chem 279: 24467‐24476, 2004.
 21. Broer S . Lysinuric protein intolerance: One gene, many problems. Am J Physiol Cell Physiol 293: C540‐541, 2007.
 22. Broer S . Amino acid transport across mammalian intestinal and renal epithelia. Physiol Rev 88: 249‐286, 2008.
 23. Broer S . Apical transporters for neutral amino acids: Physiology and pathophysiology. Physiology (Bethesda) 23: 95‐103, 2008.
 24. Broer S , Bailey CG , Kowalczuk S , Ng C , Vanslambrouck JM , Rodgers H , Auray‐Blais C , Cavanaugh JA , Broer A , Rasko JE . Iminoglycinuria and hyperglycinuria are discrete human phenotypes resulting from complex mutations in proline and glycine transporters. J Clin Invest 118: 3881‐3892, 2008.
 25. Broer S , Palacin M . The role of amino acid transporters in inherited and acquired diseases. Biochem J 436: 193‐211, 2011.
 26. Brosnan JT . Interorgan amino acid transport and its regulation. The Journal of nutrition 133: 2068S‐2072S, 2003.
 27. Brosnan JT , Brosnan ME . Creatine metabolism and the urea cycle. Mol Genet Metab 100(Suppl 1): S49‐52, 2010.
 28. Brosnan ME , Brosnan JT . Renal arginine metabolism. J Nutr 134: 2791S‐2795S; discussion 2796S‐2797S, 2004.
 29. Brown JLS , Samiy AH, Pitts RF . Localization of amino‐nitrogen reabsorption in the nephron of the dog. Am J Physiol 200: 370‐372, 1961.
 30. Bulus N , Feral C , Pozzi A , Zent R . CD98 increases renal epithelial cell proliferation by activating MAPKs. PloS one 7: e40026, 2012.
 31. Busque SM , Wagner CA . Potassium restriction, high protein intake, and metabolic acidosis increase expression of the glutamine transporter SNAT3 (Slc38a3) in mouse kidney. Am J Physiol Renal Physiol 297: F440‐450, 2009.
 32. Butchbach ME , Lai L , Lin CL . Molecular cloning, gene structure, expression profile and functional characterization of the mouse glutamate transporter (EAAT3) interacting protein GTRAP3‐18. Gene 292: 81‐90, 2002.
 33. Calonge MJ , Gasparini P , Chillaron J , Chillon M , Gallucci M , Rousaud F , Zelante L , Testar X , Dallapiccola B , Di Silverio F , et al. Cystinuria caused by mutations in rBAT, a gene involved in the transport of cystine. Nat Genet 6: 420‐425, 1994.
 34. Camargo SM , Bockenhauer D , Kleta R . Aminoacidurias: Clinical and molecular aspects. Kidney Int 73: 918‐925, 2008.
 35. Camargo SM , Makrides V , Virkki LV , Forster IC , Verrey F . Steady‐state kinetic characterization of the mouse B0AT1 sodium‐dependent neutral amino acid transporter. Pflugers Arch 451: 338‐348, 2005.
 36. Camargo SM , Singer D , Makrides V , Huggel K , Pos KM , Wagner CA , Kuba K , Danilczyk U , Skovby F , Kleta R , Penninger JM , Verrey F . Tissue‐specific amino acid transporter partners ACE2 and collectrin differentially interact with hartnup mutations. Gastroenterology 136: 872‐882, 2009.
 37. Cantor J , Browne CD , Ruppert R , Feral CC , Fassler R , Rickert RC , Ginsberg MH . CD98hc facilitates B cell proliferation and adaptive humoral immunity. Nature immunology 10: 412‐419, 2009.
 38. Cantor J , Slepak M , Ege N , Chang JT , Ginsberg MH . Loss of T cell CD98 H chain specifically ablates T cell clonal expansion and protects from autoimmunity. J Immunol 187: 851‐860, 2011.
 39. Castorino JJ , Gallagher‐Colombo SM , Levin AV , Fitzgerald PG , Polishook J , Kloeckener‐Gruissem B , Ostertag E , Philp NJ . Juvenile cataract‐associated mutation of solute carrier SLC16A12 impairs trafficking of the protein to the plasma membrane. Invest Ophth Vis Sci 52: 6774‐6784, 2011.
 40. Chairoungdua A , Kanai Y , Matsuo H , Inatomi J , Kim DK , Endou H . Identification and characterization of a novel member of the heterodimeric amino acid transporter family presumed to be associated with an unknown heavy chain. J Biol Chem 276: 49390‐49399, 2001.
 41. Chen Z , Kennedy DJ , Wake KA , Zhuang L , Ganapathy V , Thwaites DT . Structure, tissue expression pattern, and function of the amino acid transporter rat PAT2. Biochem Biophys Res Commun 304: 747‐754, 2003.
 42. Cheon CK , Lee BH , Ko JM , Kim HJ , Yoo HW . Novel mutation in SLC6A19 causing late‐onset seizures in Hartnup disorder. Pediatr Neurol 42: 369‐371.
 43. Cheval L , Pierrat F , Dossat C , Genete M , Imbert‐Teboul M , Duong Van Huyen JP , Poulain J , Wincker P , Weissenbach J , Piquemal D , Doucet A . Atlas of gene expression in the mouse kidney: New features of glomerular parietal cells. Physiol Genomics 43: 161‐173, 2011.
 44. Chillaron J , Estevez R , Mora C , Wagner CA , Suessbrich H , Lang F , Gelpi JL , Testar X , Busch AE , Zorzano A , Palacin M . Obligatory amino acid exchange via systems b0,+‐like and y+L‐like. A tertiary active transport mechanism for renal reabsorption of cystine and dibasic amino acids. J Biol Chem 271: 17761‐17770, 1996.
 45. Chillaron J , Font‐Llitjos M , Fort J , Zorzano A , Goldfarb DS , Nunes V , Palacin M . Pathophysiology and treatment of cystinuria. Nat Rev Nephrol 6: 424‐434, 2010.
 46. Christensen HN . Role of amino acid transport and countertransport in nutrition and metabolism. Physiol Rev 70: 43‐77, 1990.
 47. Closs EI , Simon A , Vekony N , Rotmann A . Plasma membrane transporters for arginine. J Nutr 134: 2752S‐2759S; discussion 2765S‐2767S, 2004.
 48. Crackower MA , Sarao R , Oudit GY , Yagil C , Kozieradzki I , Scanga SE , Oliveira‐dos‐Santos AJ , da Costa J , Zhang L , Pei Y , Scholey J , Ferrario CM , Manoukian AS , Chappell MC , Backx PH , Yagil Y , Penninger JM . Angiotensin‐converting enzyme 2 is an essential regulator of heart function. Nature 417: 822‐828, 2002.
 49. Dales NA , Gould AE , Brown JA , Calderwood EF , Guan B , Minor CA , Gavin JM , Hales P , Kaushik VK , Stewart M , Tummino PJ , Vickers CS , Ocain TD , Patane MA . Substrate‐based design of the first class of angiotensin‐converting enzyme‐related carboxypeptidase (ACE2) inhibitors. J Am Chem Soc 124: 11852‐11853, 2002.
 50. Daniel H , Herget M . Cellular and molecular mechanisms of renal peptide transport. Am J Physiol 273: F1‐8, 1997.
 51. Daniel H , Rubio‐Aliaga I . An update on renal peptide transporters. Am J Physiol Renal Physiol 284: F885‐892, 2003.
 52. Danilczyk U , Sarao R , Remy C , Benabbas C , Stange G , Richter A , Arya S , Pospisilik JA , Singer D , Camargo SM , Makrides V , Ramadan T , Verrey F , Wagner CA , Penninger JM . Essential role for collectrin in renal amino acid transport. Nature 444: 1088‐1091, 2006.
 53. Dave MH , Schulz N , Zecevic M , Wagner CA , Verrey F . Expression of heteromeric amino acid transporters along the murine intestine. J Physiol 558: 597‐610, 2004.
 54. Dello Strologo L , Pras E , Pontesilli C , Beccia E , Ricci‐Barbini V , de Sanctis L , Ponzone A , Gallucci M , Bisceglia L , Zelante L , Jimenez‐Vidal M , Font M , Zorzano A , Rousaud F , Nunes V , Gasparini P , Palacin M , Rizzoni G . Comparison between SLC3A1 and SLC7A9 cystinuria patients and carriers: A need for a new classification. J Am Soc Nephrol 13: 2547‐2553, 2002.
 55. Dent CE . A study of the behaviour of some sixty amino‐acids and other ninhydrin‐reacting substances on phenol‐;collidine' filter‐paper chromatograms, with notes as to the occurrence of some of them in biological fluids. Biochem J 43: 169‐180, 1948.
 56. Drgonova J , Liu QR , Hall FS , Krieger RM , Uhl GR . Deletion of v7‐3 (SLC6A15) transporter allows assessment of its roles in synaptosomal proline uptake, leucine uptake and behaviors. Brain Res 1183: 10‐20, 2007.
 57. Eisenbach GM , Weise M , Stolte H . Amino acid reabsorption in the rat nephron. Free flow micropuncture study. Pflugers Arch 357: 63‐76, 1975.
 58. Eslami B , Kinboshi M , Inoue S , Harada K , Inoue K , Koizumi A . A nonsense polymorphism (Y319X) of the solute carrier family 6 member 18 (SLC6A18) gene is not associated with hypertension and blood pressure in Japanese. Tohoku J Exp Med 208: 25‐31, 2006.
 59. Faham S , Watanabe A , Besserer GM , Cascio D , Specht A , Hirayama BA , Wright EM , Abramson J . The crystal structure of a sodium galactose transporter reveals mechanistic insights into Na+/sugar symport. Science 321: 810‐814, 2008.
 60. Fairweather SJ , Broer A , O'Mara ML , Broer S . Intestinal peptidases form functional complexes with the neutral amino acid transporter B(0)AT1. Biochem J 446: 135‐148, 2012.
 61. Fei YJ , Kanai Y , Nussberger S , Ganapathy V , Leibach FH , Romero MF , Singh SK , Boron WF , Hediger MA . Expression cloning of a mammalian proton‐coupled oligopeptide transporter. Nature 368: 563‐566, 1994.
 62. Fei YJ , Sugawara M , Nakanishi T , Huang W , Wang H , Prasad PD , Leibach FH , Ganapathy V . Primary structure, genomic organization, and functional and electrogenic characteristics of human system N 1, a Na+‐ and H+‐coupled glutamine transporter. J Biol Chem 275: 23707‐23717, 2000.
 63. Feliubadalo L , Arbones ML , Manas S , Chillaron J , Visa J , Rodes M , Rousaud F , Zorzano A , Palacin M , Nunes V . Slc7a9‐deficient mice develop cystinuria non‐I and cystine urolithiasis. Hum Mol Genet 12: 2097‐2108, 2003.
 64. Feliubadalo L , Font M , Purroy J , Rousaud F , Estivill X , Nunes V , Golomb E , Centola M , Aksentijevich I , Kreiss Y , Goldman B , Pras M , Kastner DL , Pras E , Gasparini P , Bisceglia L , Beccia E , Gallucci M , de Sanctis L , Ponzone A , Rizzoni GF , Zelante L , Bassi MT , George AL, Jr. , Manzoni M , De Grandi A , Riboni M , Endsley JK , Ballabio A , Borsani G , Reig N , Fernandez E , Estevez R , Pineda M , Torrents D , Camps M , Lloberas J , Zorzano A , Palacin M . Non‐type I cystinuria caused by mutations in SLC7A9, encoding a subunit (b0,+AT) of rBAT. Nat Genet 23: 52‐57, 1999.
 65. Filho JC , Bergstrom J , Stehle P , Furst P . Simultaneous measurements of free amino acid patterns of plasma, muscle and erythrocytes in healthy human subjects. Clin Nutr 16: 299‐305, 1997.
 66. Fogelstrand P , Feral CC , Zargham R , Ginsberg MH . Dependence of proliferative vascular smooth muscle cells on CD98hc (4F2hc, SLC3A2). The J Exp Med 206: 2397‐2406, 2009.
 67. Font‐Llitjos M , Feliubadalo L , Espino M , Cleries R , Manas S , Frey IM , Puertas S , Colell G , Palomo S , Aranda J , Visa J , Palacin M , Nunes V . Slc7a9 knockout mouse is a good cystinuria model for antilithiasic pharmacological studies. Am J Physiol Renal Physiol 293: F732‐740, 2007.
 68. Forrest LR , Kramer R , Ziegler C . The structural basis of secondary active transport mechanisms. Biochim Biophys Acta 1807: 167‐188, 2011.
 69. Forrest LR , Tavoulari S , Zhang YW , Rudnick G , Honig B . Identification of a chloride ion binding site in Na+/Cl ‐dependent transporters. Proc Natl Acad Sci U S A 104: 12761‐12766, 2007.
 70. Franca R , Veljkovic E , Walter S , Wagner CA , Verrey F . Heterodimeric amino acid transporter glycoprotein domains determining functional subunit association. Biochem J 388: 435‐443, 2005.
 71. Frey IM , Rubio‐Aliaga I , Siewert A , Sailer D , Drobyshev A , Beckers J , de Angelis MH , Aubert J , Bar Hen A , Fiehn O , Eichinger HM , Daniel H . Profiling at mRNA, protein, and metabolite levels reveals alterations in renal amino acid handling and glutathione metabolism in kidney tissue of Pept2−/− mice. Physiol Genomics 28: 301‐310, 2007.
 72. Fuchs BC , Bode BP . Amino acid transporters ASCT2 and LAT1 in cancer: Partners in crime? Semin Cancer Biol 15: 254‐266, 2005.
 73. Fukasawa Y , Segawa H , Kim JY , Chairoungdua A , Kim DK , Matsuo H , Cha SH , Endou H , Kanai Y . Identification and characterization of a Na(+)‐independent neutral amino acid transporter that associates with the 4F2 heavy chain and exhibits substrate selectivity for small neutral D‐ and L‐amino acids. J Biol Chem 275: 9690‐9698, 2000.
 74. Fukui K , Yang Q , Cao Y , Takahashi N , Hatakeyama H , Wang H , Wada J , Zhang Y , Marselli L , Nammo T , Yoneda K , Onishi M , Higashiyama S , Matsuzawa Y , Gonzalez FJ , Weir GC , Kasai H , Shimomura I , Miyagawa J , Wollheim CB , Yamagata K . The HNF‐1 target collectrin controls insulin exocytosis by SNARE complex formation. Cell Metab 2: 373‐384, 2005.
 75. Ganapathy V , Leibach FH . Is intestinal peptide‐transport energized by a proton gradient. Am J Physiol 249: G153‐G160, 1985.
 76. Garibotto G , Sofia A , Robaudo C , Saffioti S , Sala MR , Verzola D , Vettore M , Russo R , Procopio V , Deferrari G , Tessari P . Kidney protein dynamics and ammoniagenesis in humans with chronic metabolic acidosis. J Am Soc Nephrol 15: 1606‐1615, 2004.
 77. Garibotto G , Sofia A , Saffioti S , Bonanni A , Mannucci I , Verzola D . Amino acid and protein metabolism in the human kidney and in patients with chronic kidney disease. Clin Nutr 29: 424‐433, 2010.
 78. Garrod AE . The Croonian lectures on inborn errors of metabolism. Lectures III. Lancet 172: 124‐148, 1908.
 79. Garrod AE . The Croonian lectures on inborn errors of metabolism. Lectures IV. Lancet 172: 214‐220, 1908.
 80. Gomeza J , Hulsmann S , Ohno K , Eulenburg V , Szoke K , Richter D , Betz H . Inactivation of the glycine transporter 1 gene discloses vital role of glial glycine uptake in glycinergic inhibition. Neuron 40: 785‐796, 2003.
 81. Gomeza J , Ohno K , Hulsmann S , Armsen W , Eulenburg V , Richter DW , Laube B , Betz H . Deletion of the mouse glycine transporter 2 results in a hyperekplexia phenotype and postnatal lethality. Neuron 40: 797‐806, 2003.
 82. Gropper SS , Smith JL , Groff JL . Advanced Nutrition and Human Metabolism. Belmont: Wadsworth Pub Co., 2009.
 83. Gunther R , Silbernagl S , Deetjen P . Maleic acid induced aminoaciduria, studied by free flow micropuncture and continuous microperfusion. Pflugers Arch 382: 109‐114, 1979.
 84. Guy JL , Jackson RM , Acharya KR , Sturrock ED , Hooper NM , Turner AJ . Angiotensin‐converting enzyme‐2 (ACE2): Comparative modeling of the active site, specificity requirements, and chloride dependence. Biochemistry 42: 13185‐13192, 2003.
 85. Hamming I , Cooper ME , Haagmans BL , Hooper NM , Korstanje R , Osterhaus AD , Timens W , Turner AJ , Navis G , van Goor H . The emerging role of ACE2 in physiology and disease. J Pathol 212: 1‐11, 2007.
 86. Hediger MA , Romero MF , Peng JB , Rolfs A , Takanaga H , Bruford EA . The ABCs of solute carriers: Physiological, pathological and therapeutic implications of human membrane transport proteins. 447: 465‐468, 2004.
 87. Heller‐Stilb B , van Roeyen C , Rascher K , Hartwig HG , Huth A , Seeliger MW , Warskulat U , Haussinger D . Disruption of the taurine transporter gene (taut) leads to retinal degeneration in mice. FASEB J 16: 231‐233, 2002.
 88. Houdebine LM . Transgenic animal models in biomedical research. Methods Mol Biol 360: 163‐202, 2007.
 89. Hu Y , Smith DE , Ma K , Jappar D , Thomas W , Hillgren KM . Targeted disruption of peptide transporter Pept1 gene in mice significantly reduces dipeptide absorption in intestine. Mol Pharm 5: 1122‐1130, 2008.
 90. Huang DY , Boini KM , Lang PA , Grahammer F , Duszenko M , Heller‐Stilb B , Warskulat U , Haussinger D , Lang F , Vallon V . Impaired ability to increase water excretion in mice lacking the taurine transporter gene TAUT. Pflugers Arch 451: 668‐677, 2006.
 91. Imai Y , Kuba K , Rao S , Huan Y , Guo F , Guan B , Yang P , Sarao R , Wada T , Leong‐Poi H , Crackower MA , Fukamizu A , Hui CC , Hein L , Uhlig S , Slutsky AS , Jiang C , Penninger JM . Angiotensin‐converting enzyme 2 protects from severe acute lung failure. Nature 436: 112‐116, 2005.
 92. Inigo C , Barber A , Lostao MP . Na+ and pH dependence of proline and beta‐alanine absorption in rat small intestine. Acta Physiol (Oxf) 186: 271‐278, 2006.
 93. Jardetzky O . Simple allosteric model for membrane pumps. Nature 211: 969‐970, 1966.
 94. Jiang J , Amara SG . New views of glutamate transporter structure and function: Advances and challenges. Neuropharmacology 60: 172‐181, 2011.
 95. Kanai Y , Hediger MA . Primary structure and functional characterization of a high‐affinity glutamate transporter. Nature 360: 467‐471, 1992.
 96. Kanai Y , Hediger MA . The glutamate and neutral amino acid transporter family: Physiological and pharmacological implications. Eur J Pharmacol 479: 237‐247, 2003.
 97. Kanai Y , Hediger MA . The glutamate/neutral amino acid transporter family SLC1: Molecular, physiological and pharmacological aspects. Pflugers Arch 447: 469‐479, 2004.
 98. Kanai Y , Nussberger S , Romero MF , Boron WF , Hebert SC , Hediger MA . Electrogenic properties of the epithelial and neuronal high affinity glutamate transporter. J Biol Chem 270: 16561‐16568, 1995.
 99. Karlsson RM , Tanaka K , Saksida LM , Bussey TJ , Heilig M , Holmes A . Assessment of glutamate transporter GLAST (EAAT1)‐deficient mice for phenotypes relevant to the negative and executive/cognitive symptoms of schizophrenia. Neuropsychopharmacology 34: 1578‐1589, 2009.
 100. Kennedy DJ , Gatfield KM , Winpenny JP , Ganapathy V , Thwaites DT . Substrate specificity and functional characterisation of the H+/amino acid transporter rat PAT2 (Slc36a2). Br J Pharmacol 144: 28‐41, 2005.
 101. 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 276: 17221‐17228, 2001.
 102. Kleemola M , Toivonen M , Mykkanen J , Simell O , Huoponen K , Heiskanen KM . Heterodimerization of y(+)LAT‐1 and 4F2hc visualized by acceptor photobleaching FRET microscopy. Biochim Biophys Acta 1768: 2345‐2354, 2007.
 103. Kleta R , Gahl WA . Collecting evidence: The case of collectrin (Tmem27) and amino acid transport. Am J Physiol Renal Physiol 292: F531‐532, 2007.
 104. Kleta R , Romeo E , Ristic Z , Ohura T , Stuart C , Arcos‐Burgos M , Dave MH , Wagner CA , Camargo SR , Inoue S , Matsuura N , Helip‐Wooley A , Bockenhauer D , Warth R , Bernardini I , Visser G , Eggermann T , Lee P , Chairoungdua A , Jutabha P , Babu E , Nilwarangkoon S , Anzai N , Kanai Y , Verrey F , Gahl WA , Koizumi A . Mutations in SLC6A19, encoding B0AT1, cause Hartnup disorder. Nat Genet 36: 999‐1002, 2004.
 105. Kleta R , Tinloy B , Aydin HI , Gunay‐Aygun M , Stanescu H , Bernardini I , Gahl WA , Coskun T . Iminoglycinuria. Unexpected molecular findings. J Am Soc Nephrol 17: 819A, 2006 (Abstract).
 106. Kowalczuk S , Broer A , Munzinger M , Tietze N , Klingel K , Broer S . Molecular cloning of the mouse IMINO system: An Na+‐ and Cl−‐dependent proline transporter. Biochem J 386: 417‐422, 2005.
 107. Kowalczuk S , Broer A , Tietze N , Vanslambrouck JM , Rasko JE , Broer S . A protein complex in the brush‐border membrane explains a Hartnup disorder allele. Faseb J 22: 2880‐2887, 2008.
 108. Kuba K , Imai Y , Rao S , Gao H , Guo F , Guan B , Huan Y , Yang P , Zhang Y , Deng W , Bao L , Zhang B , Liu G , Wang Z , Chappell M , Liu Y , Zheng D , Leibbrandt A , Wada T , Slutsky AS , Liu D , Qin C , Jiang C , Penninger JM . A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus‐induced lung injury. Nat Med 11: 875‐879, 2005.
 109. Lash LH . Renal glutathione transport: Identification of carriers, physiological functions, and controversies. Biofactors 35: 500‐508, 2009.
 110. Lash LH . Renal membrane transport of glutathione in toxicology and disease. Vet Pathol 48: 408‐419, 2011.
 111. Lash LH , Putt DA , Xu F , Matherly LH . Role of rat organic anion transporter 3 (Oat3) in the renal basolateral transport of glutathione. Chem‐Biol Interact 170: 124‐134, 2007.
 112. Levillain O . Expression and function of arginine‐producing and consuming‐enzymes in the kidney. Amino Acids 42: 1237‐1257, 2011.
 113. Liang R , Fei YJ , Prasad PD , Ramamoorthy S , Han H , Yang‐Feng TL , Hediger MA , Ganapathy V , Leibach FH . Human intestinal H+/peptide cotransporter. Cloning, functional expression, and chromosomal localization. J Biol Chem 270: 6456‐6463, 1995.
 114. Liu QR , Lopez‐Corcuera B , Nelson H , Mandiyan S , Nelson N . Cloning and expression of a cDNA encoding the transporter of taurine and beta‐alanine in mouse brain. Proc Natl Acad Sci U S A 89: 12145‐12149, 1992.
 115. Liu W , Liang R , Ramamoorthy S , Fei YJ , Ganapathy ME , Hediger MA , Ganapathy V , Leibach FH . Molecular cloning of PEPT 2, a new member of the H+/peptide cotransporter family, from human kidney. Biochim Biophys Acta 1235: 461‐466, 1995.
 116. Liu W , Lopez JM , VanderJagt DJ , Glew RH , Fry DE , Schermer C , Morris DM . Evaluation of aminoaciduria in severely traumatized patients. Clin Chim Acta 316: 123‐128, 2002.
 117. Mackenzie B , Erickson JD . Sodium‐coupled neutral amino acid (System N/A) transporters of the SLC38 gene family. Pflugers Arch 447: 784‐795, 2004.
 118. Malakauskas SM , Quan H , Fields TA , McCall SJ , Yu MJ , Kourany WM , Frey CW , Le TH . Aminoaciduria and altered renal expression of luminal amino acid transporters in mice lacking novel gene collectrin. Am J Physiol Renal Physiol 292: F533‐544, 2007.
 119. Mariotta L , Ramadan T , Singer D , Guetg A , Herzog B , Stoeger C , Palacin M , Lahoutte T , Camargo SM , Verrey F . T‐type amino acid transporter TAT1 (Slc16a10) is essential for extracellular aromatic amino acid homeostasis control. J Physiol 590: 6413‐6424, 2012.
 120. Matsuo H , Kanai Y , Kim JY , Chairoungdua A , Kim DK , Inatomi J , Shigeta Y , Ishimine H , Chaekuntode S , Tachampa K , Choi HW , Babu E , Fukuda J , Endou H . Identification of a novel Na+‐independent acidic amino acid transporter with structural similarity to the member of a heterodimeric amino acid transporter family associated with unknown heavy chains. J Biol Chem 277: 21017‐21026, 2002.
 121. Meier C , Ristic Z , Klauser S , Verrey F . Activation of system L heterodimeric amino acid exchangers by intracellular substrates. EMBO J 21: 580‐589, 2002.
 122. Melancon SB , Dallaire L , Lemieux B , Robitaille P , Potier M . Dicarboxylic aminoaciduria: An inborn error of amino acid conservation. J Pediatr 91: 422‐427, 1977.
 123. Mitsuoka K , Shirasaka Y , Fukushi A , Sato M , Nakamura T , Nakanishi T , Tamai I . Transport characteristics of L‐citrulline in renal apical membrane of proximal tubular cells. Biopharm Drug Dispos 30: 126‐137, 2009.
 124. Molinas SM , Trumper L , Marinelli RA . Mitochondrial aquaporin‐8 in renal proximal tubule cells: Evidence for a role in the response to metabolic acidosis. Am J Physiol Renal Physiol 303: F458‐466, 2012.
 125. Moller N , Meek S , Bigelow M , Andrews J , Nair KS . The kidney is an important site for in vivo phenylalanine‐to‐tyrosine conversion in adult humans: A metabolic role of the kidney. Proc Natl Acad Sci U S A 97: 1242‐1246, 2000.
 126. Moret C , Dave MH , Schulz N , Jiang JX , Verrey F , Wagner CA . Regulation of renal amino acid transporters during metabolic acidosis. Am J Physiol Renal Physiol 292: F555‐566, 2007.
 127. Murer H , Evers J , Kinne R . Polarity of proximal tubular epithelial cells in relation to transepithelial transport. Curr Probl Clin Biochem 6: 173‐189, 1976.
 128. Nakakariya M , Shima Y , Shirasaka Y , Mitsuoka K , Nakanishi T , Tamai I . Organic anion transporter OAT1 is involved in renal handling of citrulline. Am J Physiol Renal Physiol 297: F71‐79, 2009.
 129. Nakamura E , Sato M , Yang H , Miyagawa F , Harasaki M , Tomita K , Matsuoka S , Noma A , Iwai K , Minato N . 4F2 (CD98) heavy chain is associated covalently with an amino acid transporter and controls intracellular trafficking and membrane topology of 4F2 heterodimer. J Biol Chem 274: 3009‐3016, 1999.
 130. Nakauchi J , Matsuo H , Kim DK , Goto A , Chairoungdua A , Cha SH , Inatomi J , Shiokawa Y , Yamaguchi K , Saito I , Endou H , Kanai Y . Cloning and characterization of a human brain Na(+)‐independent transporter for small neutral amino acids that transports D‐serine with high affinity. Neurosci Lett 287: 231‐235, 2000.
 131. Nassl AM , Rubio‐Aliaga I , Fenselau H , Marth MK , Kottra G , Daniel H . Amino acid absorption and homeostasis in mice lacking the intestinal peptide transporter PEPT1. Am J Physiol Gastrointest Liver Physiol 301: G128‐137, 2011.
 132. Newstead S , Drew D , Cameron AD , Postis VL , Xia X , Fowler PW , Ingram JC , Carpenter EP , Sansom MS , McPherson MJ , Baldwin SA , Iwata S . Crystal structure of a prokaryotic homologue of the mammalian oligopeptide‐proton symporters, PepT1 and PepT2. EMBO J 30: 417‐426, 2011.
 133. Nicholson B , Manner CK , Kleeman J , MacLeod CL . Sustained nitric oxide production in macrophages requires the arginine transporter CAT2. J Biol Chem 276: 15881‐15885, 2001.
 134. Nicklin P , Bergman P , Zhang B , Triantafellow E , Wang H , Nyfeler B , Yang H , Hild M , Kung C , Wilson C , Myer VE , MacKeigan JP , Porter JA , Wang YK , Cantley LC , Finan PM , Murphy LO . Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 136: 521‐534, 2009.
 135. Nishimura M , Naito S . Tissue‐specific mRNA expression profiles of human ATP‐binding cassette and solute carrier transporter superfamilies. Drug Metab Pharmacokinet 20: 452‐477, 2005.
 136. Nowik M , Lecca MR , Velic A , Rehrauer H , Brandli AW , Wagner CA . Genome‐wide gene expression profiling reveals renal genes regulated during metabolic acidosis. Physiol Genomics 32: 322‐334, 2008.
 137. Ocheltree SM , Shen H , Hu Y , Keep RF , Smith DE . Role and relevance of peptide transporter 2 (PEPT2) in the kidney and choroid plexus: In vivo studies with glycylsarcosine in wild‐type and PEPT2 knockout mice. J Pharmacol Exp Ther 315: 240‐247, 2005.
 138. Ottonello L , Gnerre P , Bertolotto M , Mancini M , Dapino P , Russo R , Garibotto G , Barreca T , Dallegri F . Leptin as a uremic toxin interferes with neutrophil chemotaxis. J Am Soc Nephrol 15: 2366‐2372, 2004.
 139. Palacin M , Estevez R , Bertran J , Zorzano A . Molecular biology of mammalian plasma membrane amino acid transporters. Physiol Rev 78: 969‐1054, 1998.
 140. Palacin M , Nunes V , Font‐Llitjos M , Jimenez‐Vidal M , Fort J , Gasol E , Pineda M , Feliubadalo L , Chillaron J , Zorzano A . The genetics of heteromeric amino acid transporters. Physiology (Bethesda) 20: 112‐124, 2005.
 141. Parks LD , Barfuss DW . Transepithelial transport and metabolism of glycine in S1, S2, and S3 cell types of the rabbit proximal tubule. Am J Physiol Renal Physiol 283: F1208‐1215, 2002.
 142. Peghini P , Janzen J , Stoffel W . Glutamate transporter EAAC‐1‐deficient mice develop dicarboxylic aminoaciduria and behavioral abnormalities but no neurodegeneration. EMBO J 16: 3822‐3832, 1997.
 143. Perkins CP , Mar V , Shutter JR , del Castillo J , Danilenko DM , Medlock ES , Ponting IL , Graham M , Stark KL , Zuo Y , Cunningham JM , Bosselman RA . Anemia and perinatal death result from loss of the murine ecotropic retrovirus receptor mCAT‐1. Gene Dev 11: 914‐925, 1997.
 144. Peters T , Thaete C , Wolf S , Popp A , Sedlmeier R , Grosse J , Nehls MC , Russ A , Schlueter V . A mouse model for cystinuria type I. Hum Mol Genet 12: 2109‐2120, 2003.
 145. Pfeiffer R , Loffing J , Rossier G , Bauch C , Meier C , Eggermann T , Loffing‐Cueni D , Kuhn LC , Verrey F . Luminal heterodimeric amino acid transporter defective in cystinuria. Mol Biol Cell 10: 4135‐4147, 1999.
 146. Pfeiffer R , Rossier G , Spindler B , Meier C , Kuhn L , Verrey F . Amino acid transport of y+L‐type by heterodimers of 4F2hc/CD98 and members of the glycoprotein‐associated amino acid transporter family. Embo J 18: 49‐57, 1999.
 147. Pineda M , Font M , Bassi MT , Manzoni M , Borsani G , Marigo V , Fernandez E , Rio RM , Purroy J , Zorzano A , Nunes V , Palacin M . The amino acid transporter asc‐1 is not involved in cystinuria. Kidney Int 66: 1453‐1464, 2004.
 148. Premen AJ . Potential mechanisms mediating postprandial renal hyperemia and hyperfiltration. FASEB J 2: 131‐137, 1988.
 149. Quan H , Athirakul K , Wetsel WC , Torres GE , Stevens R , Chen YT , Coffman TM , Caron MG . Hypertension and impaired glycine handling in mice lacking the orphan transporter XT2. Mol Cell Biol 24: 4166‐4173, 2004.
 150. Ramadan T , Camargo SM , Herzog B , Bordin M , Pos KM , Verrey F . Recycling of aromatic amino acids via TAT1 allows efflux of neutral amino acids via LAT2‐4F2hc exchanger. Pflugers Arch 454: 507‐516, 2007.
 151. Ramadan T , Camargo SM , Summa V , Hunziker P , Chesnov S , Pos KM , Verrey F . Basolateral aromatic amino acid transporter TAT1 (Slc16a10) functions as an efflux pathway. J Cell Physiol 206: 771‐779, 2006.
 152. Ramamoorthy S , Kulanthaivel P , Leibach FH , Mahesh VB , Ganapathy V . Solubilization and functional reconstitution of the human placental taurine transporter. Biochim Biophys Acta 1145: 250‐256, 1993.
 153. Ramamoorthy S , Leibach FH , Mahesh VB , Han H , Yang‐Feng T , Blakely RD , Ganapathy V . Functional characterization and chromosomal localization of a cloned taurine transporter from human placenta. Biochem J 300(Pt 3): 893‐900, 1994.
 154. Ramamoorthy S , Liu W , Ma YY , Yang‐Feng TL , Ganapathy V , Leibach FH . Proton/peptide cotransporter (PEPT 2) from human kidney: Functional characterization and chromosomal localization. Biochim Biophys Acta 1240: 1‐4, 1995.
 155. Reig N , Chillaron J , Bartoccioni P , Fernandez E , Bendahan A , Zorzano A , Kanner B , Palacin M , Bertran J . The light subunit of system b(0,+) is fully functional in the absence of the heavy subunit. Embo J 21: 4906‐4914, 2002.
 156. Ressl S , Terwisscha van Scheltinga AC , Vonrhein C , Ott V , Ziegler C . Molecular basis of transport and regulation in the Na(+)/betaine symporter BetP. Nature 458: 47‐52, 2009.
 157. Romeo E , Dave MH , Bacic D , Ristic Z , Camargo SM , Loffing J , Wagner CA , Verrey F . Luminal kidney and intestine SLC6 amino acid transporters of B0AT‐cluster and their tissue distribution in Mus musculus. Am J Physiol Renal Physiol 290: F376‐383, 2006.
 158. Rossier G , Meier C , Bauch C , Summa V , Sordat B , Verrey F , Kuhn LC . LAT2, a new basolateral 4F2hc/CD98‐associated amino acid transporter of kidney and intestine. J Biol Chem 274: 34948‐34954, 1999.
 159. Rubio‐Aliaga I , Daniel H . Peptide transporters and their roles in physiological processes and drug disposition. Xenobiotica 38: 1022‐1042, 2008.
 160. Ruderisch N , Virgintino D , Makrides V , Verrey F . Differential axial localization along the mouse brain vascular tree of luminal sodium‐dependent glutamine transporters Snat1 and Snat3. J Cereb Blood Flow Metab 31: 1637‐1647, 2011.
 161. Rudnick G . Serotonin transporters–structure and function. J Membr Biol 213: 101‐110, 2006.
 162. Ruggiero AM , Liu Y , Vidensky S , Maier S , Jung E , Farhan H , Robinson MB , Sitte HH , Rothstein JD . The endoplasmic reticulum exit of glutamate transporter is regulated by the inducible mammalian Yip6b/GTRAP3‐18 protein. J Biol Chem 283: 6175‐6183, 2008.
 163. Rutter AR , Fradley RL , Garrett EM , Chapman KL , Lawrence JM , Rosahl TW , Patel S . Evidence from gene knockout studies implicates Asc‐1 as the primary transporter mediating d‐serine reuptake in the mouse CNS. Eur J Neurosci 25: 1757‐1766, 2007.
 164. Sakamoto S , Chairoungdua A , Nagamori S , Wiriyasermkul P , Promchan K , Tanaka H , Kimura T , Ueda T , Fujimura M , Shigeta Y , Naya Y , Akakura K , Ito H , Endou H , Ichikawa T , Kanai Y . A novel role of the C‐terminus of b 0,+ AT in the ER‐Golgi trafficking of the rBAT‐b 0,+ AT heterodimeric amino acid transporter. Biochem J 417: 441‐448, 2009.
 165. Sallstrom J , Carlstrom M , Olerud J , Fredholm BB , Kouzmine M , Sandler S , Persson AE . High‐protein‐induced glomerular hyperfiltration is independent of the tubuloglomerular feedback mechanism and nitric oxide synthases. Am J Physiol Regul Integr Comp Physiol 299: R1263‐1268, 2010.
 166. Sato H , Shiiya A , Kimata M , Maebara K , Tamba M , Sakakura Y , Makino N , Sugiyama F , Yagami K , Moriguchi T , Takahashi S , Bannai S . Redox imbalance in cystine/glutamate transporter‐deficient mice. J Biol Chem 280: 37423‐37429, 2005.
 167. Scriver CR , Mahon B , Levy HL , Clow CL , Reade TM , Kronick J , Lemieux B , Laberge C . The Hartnup phenotype: Mendelian transport disorder, multifactorial disease. Am J Hum Genet 40: 401‐412, 1987.
 168. Sebastio G , Sperandeo MP , Andria G . Lysinuric protein intolerance: Reviewing concepts on a multisystem disease. Am J Med Genet C Semin Med Genet 157: 54‐62, 2011.
 169. Seow HF , Broer S , Broer A , Bailey CG , Potter SJ , Cavanaugh JA , Rasko JE . Hartnup disorder is caused by mutations in the gene encoding the neutral amino acid transporter SLC6A19. Nat Genet 36: 1003‐1007, 2004.
 170. Shayakul C , Kanai Y , Lee WS , Brown D , Rothstein JD , Hediger MA . Localization of the high‐affinity glutamate transporter EAAC1 in rat kidney. Am J Physiol 273: F1023‐1029, 1997.
 171. Shen H , Ocheltree SM , Hu Y , Keep RF , Smith DE . Impact of genetic knockout of PEPT2 on cefadroxil pharmacokinetics, renal tubular reabsorption, and brain penetration in mice. Drug Metab Dispos 35: 1209‐1216, 2007.
 172. Shen H , Smith DE , Yang T , Huang YG , Schnermann JB , Brosius FC, III . Localization of PEPT1 and PEPT2 proton‐coupled oligopeptide transporter mRNA and protein in rat kidney. Am J Physiol 276: F658‐665, 1999.
 173. Shih V . Amino acid analysis. In: Blau N , Duran M , Blaskovics ME , editors. Physician's Guide to the Laboratory Diagnosis of Metabolic Disease. London: Chapman Hall Medical, 1996, pp. 13‐30.
 174. Silbernagl S . Renal transport of amino acids. Klin Wochenschr 57: 1009‐1019, 1979.
 175. Silbernagl S . Tubular reabsorption of L‐glutamine studied by free‐flow micropuncture and microperfusion of rat kidney. Int J Biochem 12: 9‐16, 1980.
 176. Silbernagl S . Renal handling of amino acids and oligopeptides. Contrib Nephrol 24: 18‐29, 1981.
 177. Silbernagl S , Deetjen P . Molecular specificity of the L‐arginine reabsorption mechanism. Microperfusion studies in the proximal tubule of rat kidney. Pflugers Arch 340: 325‐334, 1973.
 178. Silbernagl S , Volker K , Dantzler WH . Cationic amino acid fluxes beyond the proximal convoluted tubule of rat kidney. Pflugers Arch 429: 210‐215, 1994.
 179. Singer D , Camargo SM . Collectrin and ACE2 in renal and intestinal amino acid transport. Channels 5: 410‐423, 2011.
 180. Singer D , Camargo SM , Huggel K , Romeo E , Danilczyk U , Kuba K , Chesnov S , Caron MG , Penninger JM , Verrey F . Orphan transporter SLC6A18 is renal neutral amino acid transporter B0AT3. J Biol Chem 284: 19953‐19960, 2009.
 181. Singer D , Camargo SM , Ramadan T , Schafer M , Mariotta L , Herzog B , Huggel K , Wolfer D , Werner S , Penninger JM , Verrey F . Defective intestinal amino acid absorption in Ace2 null mice. Am J Physiol‐Gastr L 303: G686‐695, 2012.
 182. Skelton MR , Schaefer TL , Graham DL , Degrauw TJ , Clark JF , Williams MT , Vorhees CV . Creatine transporter (CrT; Slc6a8) knockout mice as a model of human CrT deficiency. PloS one 6: e16187, 2011.
 183. Smith CP , Weremowicz S , Kanai Y , Stelzner M , Morton CC , Hediger MA . Assignment of the gene coding for the human high‐affinity glutamate transporter EAAC1 to 9p24: Potential role in dicarboxylic aminoaciduria and neurodegenerative disorders. Genomics 20: 335‐336, 1994.
 184. Smith DE , Pavlova A , Berger UV , Hediger MA , Yang T , Huang YG , Schnermann JB . Tubular localization and tissue distribution of peptide transporters in rat kidney. Pharm Res 15: 1244‐1249, 1998.
 185. Smith KE , Borden LA , Wang CH , Hartig PR , Branchek TA , Weinshank RL . Cloning and expression of a high affinity taurine transporter from rat brain. Mol Pharmacol 42: 563‐569, 1992.
 186. Sogaard R , Novak I , MacAulay N . Elevated ammonium levels: Differential acute effects on three glutamate transporter isoforms. Am J Physiol Cell Physiol 302: C880‐891, 2012.
 187. Sorensen SS , Lauridsen IN , Thomsen K , Pedersen EB . Effect of two regimens of intravenous amino acid infusion on renal haemodynamics, renal tubular function and sodium and water homeostatic hormones in healthy humans. Nephrol Dial Transplant 6: 410‐419, 1991.
 188. Sperandeo MP , Andria G , Sebastio G . Lysinuric protein intolerance: Update and extended mutation analysis of the SLC7A7 gene. Hum Mutat 29: 14‐21, 2008.
 189. Sperandeo MP , Annunziata P , Bozzato A , Piccolo P , Maiuri L , D'Armiento M , Ballabio A , Corso G , Andria G , Borsani G , Sebastio G . Slc7a7 disruption causes fetal growth retardation by downregulating Igf1 in the mouse model of lysinuric protein intolerance. Am J Physiol Cell Physiol 293: C191‐198, 2007.
 190. Stevens BR , Ross HJ , Wright EM . Multiple transport pathways for neutral amino acids in rabbit jejunal brush border vesicles. J Membr Biol 66: 213‐225, 1982.
 191. Suhre K , Wallaschofski H , Raffler J , Friedrich N , Haring R , Michael K , Wasner C , Krebs A , Kronenberg F , Chang D , Meisinger C , Wichmann HE , Hoffmann W , Volzke H , Volker U , Teumer A , Biffar R , Kocher T , Felix SB , Illig T , Kroemer HK , Gieger C , Romisch‐Margl W , Nauck M . A genome‐wide association study of metabolic traits in human urine. Nat Genet 43: 565‐569, 2011.
 192. Swarna M , Rao DN , Reddy PP . Dicarboxylic aminoaciduria associated with mental retardation. Hum Genet 82: 299‐300, 1989.
 193. Takanaga H , Mackenzie B , Suzuki Y , Hediger MA . Identification of mammalian proline transporter SIT1 (SLC6A20) with characteristics of classical system imino. J Biol Chem 280: 8974‐8984, 2005.
 194. Tessari P , Garibotto G , Inchiostro S , Robaudo C , Saffioti S , Vettore M , Zanetti M , Russo R , Deferrari G . Kidney, splanchnic, and leg protein turnover in humans. Insight from leucine and phenylalanine kinetics. J Clin Invest 98: 1481‐1492, 1996.
 195. Thwaites DT , Ford D , Glanville M , Simmons NL . H(+)/solute‐induced intracellular acidification leads to selective activation of apical Na(+)/H(+) exchange in human intestinal epithelial cells. J Clin Invest 104: 629‐635, 1999.
 196. Tietze IN , Sorensen SS , Eiskjaer H , Thomsen K , Pedersen EB . Tubular handling of amino acids after intravenous infusion of amino acids in healthy humans. Nephrol Dial Transplant 7: 493‐500, 1992.
 197. Torrents D , Estevez R , Pineda M , Fernandez E , Lloberas J , Shi YB , Zorzano A , Palacin M . Identification and characterization of a membrane protein (y+L amino acid transporter‐1) that associates with 4F2hc to encode the amino acid transport activity y+L. A candidate gene for lysinuric protein intolerance. J Biol Chem 273: 32437‐32445, 1998.
 198. Treberg JR , Clow KA , Greene KA , Brosnan ME , Brosnan JT . Systemic activation of glutamate dehydrogenase increases renal ammoniagenesis: Implications for the hyperinsulinism/hyperammonemia syndrome. Am J Physiol Endocrinol Metab 298: E1219‐1225, 2010.
 199. Tsumura H , Suzuki N , Saito H , Kawano M , Otake S , Kozuka Y , Komada H , Tsurudome M , Ito Y . The targeted disruption of the CD98 gene results in embryonic lethality. Biochem Biophys Res Commun 308: 847‐851, 2003.
 200. Uchida S , Kwon HM , Yamauchi A , Preston AS , Marumo F , Handler JS . Molecular cloning of the cDNA for an MDCK cell Na(+)‐ and Cl(−)‐dependent taurine transporter that is regulated by hypertonicity. Proc Natl Acad Sci U S A 89: 8230‐8234, 1992.
 201. van de Poll MC , Soeters PB , Deutz NE , Fearon KC , Dejong CH . Renal metabolism of amino acids: Its role in interorgan amino acid exchange. Am J Clin Nutr 79: 185‐197, 2004.
 202. Vanslambrouck JM , Broer A , Thavyogarajah T , Holst J , Bailey CG , Broer S , Rasko JE . Renal imino acid and glycine transport system ontogeny and involvement in developmental iminoglycinuria. Biochem J 428: 397‐407, 2010.
 203. Venta R . Year‐long validation study and reference values for urinary amino acids using a reversed‐phase HPLC method. Clin Chem 47: 575‐583, 2001.
 204. Verrey F , Closs EI , Wagner CA , Palacin M , Endou H , Kanai Y . CATs and HATs: The SLC7 family of amino acid transporters. Pflugers Arch 447: 532‐542, 2004.
 205. Verrey F , Jack DL , Paulsen IT , Saier MH, Jr , Pfeiffer R . New glycoprotein‐associated amino acid transporters. J Membr Biol 172: 181‐192, 1999.
 206. Verrey F , Singer D , Ramadan T , Vuille‐dit‐Bille RN , Mariotta L , Camargo SM . Kidney amino acid transport. Pflugers Arch 458: 53‐60, 2009.
 207. Wagner CA . Metabolic acidosis: New insights from mouse models. Curr Opin Nephrol Hypertens 16: 471‐476, 2007.
 208. Weiner ID , Verlander JW . Role of NH3 and NH4+ transporters in renal acid‐base transport. Am J Physiol Renal Physiol 300: F11‐23, 2011.
 209. Wright EM . Renal Na(+)‐glucose cotransporters. Am J Physiol Renal Physiol 280: F10‐18, 2001.
 210. Xie X , Dumas T , Tang L , Brennan T , Reeder T , Thomas W , Klein RD , Flores J , O'Hara BF , Heller HC , Franken P . Lack of the alanine‐serine‐cysteine transporter 1 causes tremors, seizures, and early postnatal death in mice. Brain Res 1052: 212‐221, 2005.
 211. Yamashita A , Singh SK , Kawate T , Jin Y , Gouaux E . Crystal structure of a bacterial homologue of Na+/Cl−‐dependent neurotransmitter transporters. Nature 437: 215‐223, 2005.
 212. Ye M , Wysocki J , William J , Soler MJ , Cokic I , Batlle D . Glomerular localization and expression of Angiotensin‐converting enzyme 2 and Angiotensin‐converting enzyme: Implications for albuminuria in diabetes. J Am Soc Nephrol 17: 3067‐3075, 2006.
 213. Yernool D , Boudker O , Jin Y , Gouaux E . Structure of a glutamate transporter homologue from Pyrococcus horikoshii. Nature 431: 811‐818, 2004.
 214. Yu MJ , Pisitkun T , Wang G , Shen RF , Knepper MA . LC‐MS/MS analysis of apical and basolateral plasma membranes of rat renal collecting duct cells. Mol Cell Proteomics 5: 2131‐2145, 2006.
 215. Zhang H , Wada J , Hida K , Tsuchiyama Y , Hiragushi K , Shikata K , Wang H , Lin S , Kanwar YS , Makino H . Collectrin, a collecting duct‐specific transmembrane glycoprotein, is a novel homolog of ACE2 and is developmentally regulated in embryonic kidneys. J Biol Chem 276: 17132‐17139, 2001.
 216. Zheng Y , Zhou C , Huang Y , Bu D , Zhu X , Jiang W . A novel missense mutation in the SLC6A19 gene in a Chinese family with Hartnup disorder. Int J Dermatol 48: 388‐392, 2009.
 217. Zhu T , Chen XZ , Steel A , Hediger MA , Smith DE . Differential recognition of ACE inhibitors in Xenopus laevis oocytes expressing rat PEPT1 and PEPT2. Pharm Res 17: 526‐532, 2000.

Related Articles:

Epithelial Transport
Basic Principles of Transport
Glomerular Filtration
Comparative Physiology of the Kidney
Renal Metabolism: Integrated Responses

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Victoria Makrides, Simone M.R. Camargo, François Verrey. Transport of Amino Acids in the Kidney. Compr Physiol 2014, 4: 367-403. doi: 10.1002/cphy.c130028