Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 71
1.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Article En | MEDLINE | ID: mdl-34848541

Despite having similar structures, each member of the heteromeric amino acid transporter (HAT) family shows exquisite preference for the exchange of certain amino acids. Substrate specificity determines the physiological function of each HAT and their role in human diseases. However, HAT transport preference for some amino acids over others is not yet fully understood. Using cryo-electron microscopy of apo human LAT2/CD98hc and a multidisciplinary approach, we elucidate key molecular determinants governing neutral amino acid specificity in HATs. A few residues in the substrate-binding pocket determine substrate preference. Here, we describe mutations that interconvert the substrate profiles of LAT2/CD98hc, LAT1/CD98hc, and Asc1/CD98hc. In addition, a region far from the substrate-binding pocket critically influences the conformation of the substrate-binding site and substrate preference. This region accumulates mutations that alter substrate specificity and cause hearing loss and cataracts. Here, we uncover molecular mechanisms governing substrate specificity within the HAT family of neutral amino acid transporters and provide the structural bases for mutations in LAT2/CD98hc that alter substrate specificity and that are associated with several pathologies.


Amino Acid Transport Systems, Neutral/physiology , Substrate Specificity/physiology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems/physiology , Amino Acid Transport Systems, Neutral/metabolism , Amino Acids/metabolism , Amino Acids, Neutral/metabolism , Biological Transport/physiology , Cryoelectron Microscopy/methods , Fusion Regulatory Protein 1, Heavy Chain/metabolism , HeLa Cells , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Protein Domains , Structure-Activity Relationship
2.
Biochem J ; 478(21): 3957-3976, 2021 11 12.
Article En | MEDLINE | ID: mdl-34704597

Metabolic reprogramming in cancer necessitates increased amino acid uptake, which is accomplished by up-regulation of specific amino acid transporters. However, not all tumors rely on any single amino acid transporter for this purpose. Here, we report on the differential up-regulation of the amino acid transporter SLC38A5 in triple-negative breast cancer (TNBC). The up-regulation is evident in TNBC tumors, conventional and patient-derived xenograft TNBC cell lines, and a mouse model of spontaneous TNBC mammary tumor. The up-regulation is confirmed by functional assays. SLC38A5 is an amino acid-dependent Na+/H+ exchanger which transports Na+ and amino acids into cells coupled with H+ efflux. Since cell-surface Na+/H+ exchanger is an established inducer of macropinocytosis, an endocytic process for cellular uptake of bulk fluid and its components, we examined the impact of SLC38A5 on macropinocytosis in TNBC cells. We found that the transport function of SLC38A5 is coupled to the induction of macropinocytosis. Surprisingly, the transport function of SLC38A5 is inhibited by amilorides, the well-known inhibitors of Na+/H+ exchanger. Down-regulation of SLC38A5 in TNBC cells attenuates serine-induced macropinocytosis and reduces cell proliferation significantly as assessed by multiple methods, but does not induce cell death. The Cancer Genome Atlas database corroborates SLC38A5 up-regulation in TNBC. This represents the first report on the selective expression of SLC38A5 in TNBC and its role as an inducer of macropinocytosis, thus revealing a novel, hitherto unsuspected, function for an amino acid transporter that goes beyond amino acid delivery but is still relevant to cancer cell nutrition and proliferation.


Amino Acid Transport Systems, Neutral/physiology , Triple Negative Breast Neoplasms/metabolism , Animals , Biological Transport , Cell Line, Tumor , Endocytosis , Female , Humans , Mice , Mice, Transgenic
3.
Inflamm Bowel Dis ; 26(12): 1787-1795, 2020 11 19.
Article En | MEDLINE | ID: mdl-33064147

Angiotensin-converting enzyme 2 (ACE2) has been highlighted for its role as a receptor for SARS-CoV-2, responsible for the current COVID-19 pandemic. This review summarizes current knowledge about ACE2 as a multifunctional protein, focusing on its relevance in inflammatory bowel disease (IBD). As an enzyme, ACE2 may be protective in IBD because it favors the counter-regulatory arm of the renin-angiotensin system or deleterious because it metabolizes other anti-inflammatory/repairing elements. Meanwhile, as a receptor for SARS-CoV-2, the impact of ACE2 expression/activity on infection is still under debate because no direct evidence has been reported and, again, both protective and deleterious pathways are possible. Research has shown that ACE2 regulates the expression of the neutral amino acid transporter B0AT1, controlling tryptophan-associated intestinal inflammation and nutritional status. Finally, intact membrane-bound or shed soluble ACE2 can also trigger integrin signaling, modulating the response to anti-integrin biologic drugs used to treat IBD (such as vedolizumab) and fibrosis, a long-term complication of IBD. As such, future studies on ACE2 expression/activity in IBD can improve monitoring of the disease and explore an alternative pharmacological target.


Angiotensin-Converting Enzyme 2/physiology , Inflammatory Bowel Diseases/metabolism , SARS-CoV-2/physiology , Amino Acid Transport Systems, Neutral/physiology , COVID-19/virology , Humans , Inflammatory Bowel Diseases/physiopathology , Renin-Angiotensin System/physiology
4.
PLoS One ; 14(12): e0223954, 2019.
Article En | MEDLINE | ID: mdl-31800572

BACKGROUND: Cystinosis is a rare disorder caused by recessive mutations of the CTNS gene. Current therapy decreases cystine accumulation, thus slowing organ deterioration without reversing renal Fanconi syndrome or preventing eventual need for a kidney transplant.15-20% of cystinosis patients harbour at least one nonsense mutation in CTNS, leading to premature end of translation of the transcript. Aminoglycosides have been shown to permit translational read-through but have high toxicity level, especially in the kidney and inner ear. ELX-02, a modified aminoglycoside, retains it read-through ability without the toxicity. METHODS AND FINDINGS: We ascertained the toxicity of ELX-02 in cells and in mice as well as the effect of ELX-02 on translational read-through of nonsense mutations in cystinotic mice and human cells. ELX-02 was not toxic in vitro or in vivo, and permitted read-through of nonsense mutations in cystinotic mice and human cells. CONCLUSIONS: ELX-02 has translational read-through activity and produces a functional CTNS protein, as evidenced by reduced cystine accumulation. This reduction is comparable to cysteamine treatment. ELX-02 accumulates in the kidney but neither cytotoxicity nor nephrotoxicity was observed.


Amino Acid Transport Systems, Neutral/physiology , Aminoglycosides/pharmacology , Cystine/metabolism , Cystinosis/drug therapy , Lysosomes/metabolism , Mutation , Animals , Biological Transport , Cystinosis/metabolism , Cystinosis/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Biosynthesis
5.
Am J Pathol ; 189(5): 1053-1064, 2019 05.
Article En | MEDLINE | ID: mdl-30794806

Cystinosis is a rare lysosomal storage disorder caused by loss-of-function mutations of the CTNS gene, encoding cystinosin, a symporter that mediates cystine efflux from lysosomes. Approximately 95% of patients with cystinosis display renal Fanconi syndrome, short stature, osteopenia, and rickets. In this study, we investigated whether the absence of cystinosin primarily affects bone remodeling activity, apart from the influences of the Fanconi syndrome on bone mineral metabolism. Using micro-computed tomography and histomorphometric and bone serum biomarker analysis, we evaluated the bone phenotype of 1-month-old Ctns-/- knockout (KO) male mice without tubulopathy. An in vitro study was performed to characterize the effects of cystinosin deficiency on osteoblasts and osteoclasts. Micro-computed tomography analysis showed a reduction of trabecular bone volume, bone mineral density, and number and thickness in KO mice compared with wild-type animals; histomorphometric analysis revealed a reduction of osteoblast and osteoclast parameters in tibiae of cystinotic mice. Decreased levels of serum procollagen type 1 amino-terminal propeptide and tartrate-resistant acid phosphatase in KO mice confirmed reduced bone remodeling activity. In vitro experiments showed an impairment of Ctns-/- osteoblasts and osteoclasts. In conclusion, cystinosin deficiency primarily affects bone cells, leading to a bone loss phenotype of KO mice, independent from renal failure.


Amino Acid Transport Systems, Neutral/physiology , Bone Diseases/pathology , Cystinosis/pathology , Osteoblasts/pathology , Osteogenesis , Animals , Bone Diseases/etiology , Bone Diseases/metabolism , Cystinosis/etiology , Cystinosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism
6.
Biochim Biophys Acta Mol Cell Res ; 1866(2): 252-263, 2019 02.
Article En | MEDLINE | ID: mdl-30445147

A plasma membrane amino acid transporter B0,+ (ATB0,+), encoded by the SLC6A14 gene, is specific for neutral and basic amino acids. It is up-regulated in several types of malignant cancers. Neurotransmitter transporters of the SLC6 family interact with specific SEC24 proteins of the COPII complex along their pathway from the endoplasmic reticulum (ER) to Golgi. This study focused on the possible role of SEC24 proteins in ATB0,+ trafficking. Rat ATB0,+ was expressed in HEK293 cells, its localization and trafficking were examined by Western blot, deglycosylation, immunofluorescence (co-localization with ER and trans-Golgi markers) and biotinylation. The expression of ATB0,+ at the plasma membrane was decreased by dominant negative mutants of SAR1, a GTPase, whose activity triggers the formation of the COPII complex. ATB0,+ co-precipitated with SEC24C (but not with the remaining isoforms A, B and D). This interaction was confirmed by immunocytochemistry and the proximity ligation assay. Co-localization of SEC24C with endogenous ATB0,+ was also observed in MCF-7 breast cancer cells. Contrary to the endogenous transporter, part of the overexpressed ATB0,+ is directed to proteolysis, a process significantly reversed by a proteasome inhibitor bortezomib. Co-transfection with a SEC24C dominant negative mutant attenuated ATB0,+ expression at the plasma membrane, due to proteolytic degradation. These results support a hypothesis that lysine at position +2 downstream of the ER export "RI" motif on the cargo protein is crucial for SEC24C binding and for further trafficking to the Golgi. Moreover, there is an equilibrium between ER export and degradation mechanisms in case of overexpressed transporter.


Amino Acid Transport Systems, Neutral/metabolism , Protein Transport/physiology , Vesicular Transport Proteins/physiology , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems, Neutral/physiology , Animals , COP-Coated Vesicles/physiology , Cell Membrane/metabolism , Cell Membrane/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Stress/physiology , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Membrane Proteins/genetics , Protein Isoforms/genetics , Rats , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
7.
AAPS J ; 20(1): 12, 2017 12 04.
Article En | MEDLINE | ID: mdl-29204754

Amino acids are essential building blocks of all mammalian cells, and amino acid transporters play a vital role in transporting them into cells and their further distribution among the various cellular compartments. There are ~ 430 known transporters in the solute-linked carrier (SLC) gene family, divided into 52 distinct families. Eleven of these gene families contain one or more amino acid transporters. These transporters differ significantly from each other in terms of substrate specificity, ion dependence, and energetics. Given the variety of roles they fulfill in human physiology, it is not surprising that a number of diseases are associated with the malfunction of these transporters. In particular, as amino acids are critical for cell growth, survival, and proliferation, the role of amino acid transporters in cancer is gaining increasing attention in recent years. The present review primarily focuses on one particular amino acid transporter, SLC6A14 (also known as ATB0,+), with regard to its relevance to specific diseases, including cancer, and the molecular mechanisms underlying the disease-related alterations in the expression of the transporter. Furthermore, the review highlights the possible utility of this transporter in drug delivery and also its therapeutic potential for the treatment and diagnosis of cancer.


Amino Acid Transport Systems, Neutral/physiology , Amino Acid Transport Systems , Amino Acid Transport Systems, Neutral/genetics , Crohn Disease/metabolism , Cystic Fibrosis/metabolism , Humans , Infertility, Male , Male , Neoplasms/diagnosis , Neoplasms/metabolism , Obesity/metabolism
8.
Best Pract Res Clin Endocrinol Metab ; 31(2): 241-253, 2017 03.
Article En | MEDLINE | ID: mdl-28648511

Transport of thyroid hormone (TH) across the plasma membrane is essential for intracellular TH metabolism and action, and this is mediated by specific transporter proteins. During the last two decades several transporters capable of transporting TH have been identified, including monocarboxylate transporter 8 (MCT8), MCT10 and organic anion transporting polypeptide 1C1 (OATP1C1). In particular MCT8 and OATP1C1 are important for the regulation of local TH activity in the brain and thus for brain development. MCT8 is a protein containing 12 transmembrane domains, and is encoded by the SLC16A2 gene located on the X chromosome. It facilitates both TH uptake and efflux across the cell membrane. Male subjects with hemizygous mutations in MCT8 are afflicted with severe intellectual and motor disability, also known as the Allan-Herndon-Dudley syndrome (AHDS), which goes together with low serum T4 and high T3 levels. This review concerns molecular and clinical aspects of MCT8 function.


Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Mental Retardation, X-Linked/genetics , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Hypotonia/genetics , Muscular Atrophy/genetics , Thyroid Hormones/metabolism , Amino Acid Transport Systems, Neutral/physiology , Biological Transport/genetics , Brain/metabolism , Cell Membrane/metabolism , Humans , Male , Monocarboxylic Acid Transporters/physiology , Mutation , Symporters
9.
J Am Soc Nephrol ; 27(6): 1678-88, 2016 06.
Article En | MEDLINE | ID: mdl-26449607

Cystinosis is a rare autosomal recessive storage disorder characterized by defective lysosomal efflux of cystine due to mutations in the CTNS gene encoding the lysosomal cystine transporter, cystinosin. Lysosomal cystine accumulation leads to crystal formation and functional impairment of multiple organs. Moreover, cystinosis is the most common inherited cause of renal Fanconi syndrome in children. Oral cysteamine therapy delays disease progression by reducing intracellular cystine levels. However, because cysteamine does not correct all complications of cystinosis, including Fanconi syndrome, we hypothesized that cystinosin could have novel roles in addition to transporting cystine out of the lysosome. By coimmunoprecipitation experiments and mass spectrometry, we found cystinosin interacts with almost all components of vacuolar H(+)-ATPase and the Ragulator complex and with the small GTPases Ras-related GTP-binding protein A (RagA) and RagC. Furthermore, the mammalian target of rapamycin complex 1 (mTORC1) pathway was downregulated in proximal tubular cell lines derived from Ctns(-/-) mice. Decrease of lysosomal cystine levels by cysteamine did not rescue mTORC1 activation in these cells, suggesting that the downregulation of mTORC1 is due to the absence of cystinosin rather than to the accumulation of cystine. Our results show a dual role for cystinosin as a cystine transporter and as a component of the mTORC1 pathway, and provide an explanation for the appearance of Fanconi syndrome in cystinosis. Furthermore, this study highlights the need to develop new treatments not dependent on lysosomal cystine depletion alone for this devastating disease.


Amino Acid Transport Systems, Neutral/physiology , Cystinosis/etiology , Multiprotein Complexes/physiology , Signal Transduction , TOR Serine-Threonine Kinases/physiology , Vacuolar Proton-Translocating ATPases/physiology , Animals , Mechanistic Target of Rapamycin Complex 1 , Mice
10.
J Psychiatr Res ; 68: 261-9, 2015 Sep.
Article En | MEDLINE | ID: mdl-26228428

Although mental disorders as major depression are highly prevalent worldwide their underlying causes remain elusive. Despite the high heritability of depression and a clear genetic contribution to the disease, the identification of genetic risk factors for depression has been very difficult. The first published candidate to reach genome-wide significance in depression was SLC6A15, a neuronal amino acid transporter. With a reported 1,42 fold increased risk of suffering from depression associated with a single nucleotide polymorphism (SNP) in a regulatory region of SLC6A15, the polymorphism was also found to affect hippocampal morphology, integrity, and hippocampus-dependent memory. However, the function of SLC6A15 in the brain is so far largely unknown. To address this question, we investigated if alterations in SLC6A15 expression, either using a full knockout or a targeted hippocampal overexpression, affect hippocampal neurochemistry and consequently behavior. We could show that a lack of SLC6A15 reduced hippocampal tissue levels of proline and other neutral amino acids. In parallel, we observed a decreased overall availability of tissue glutamate and glutamine, while at the same time the basal tone of extracellular glutamate in the hippocampus was increased. By contrast, SLC6A15 overexpression increased glutamate/glutamine tissue concentrations. These neurochemical alterations could be linked to behavioral abnormalities in sensorimotor gating, a key translational endophenotype relevant for many psychiatric disorders. Overall, our data supports SLC6A15 as a crucial factor controlling amino acid content in the hippocampus, thereby likely interfering with glutamatergic transmission and behavior. These findings emphasize SLC6A15 as pivotal risk factor for vulnerability to psychiatric diseases.


Amino Acid Transport Systems, Neutral/physiology , Behavior, Animal/physiology , Glutamic Acid/metabolism , Hippocampus/metabolism , Sensory Gating/physiology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Hippocampus/anatomy & histology , Hippocampus/chemistry , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proline/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction
12.
Curr Opin Hematol ; 21(3): 201-9, 2014 May.
Article En | MEDLINE | ID: mdl-24626044

PURPOSE OF REVIEW: This review discusses the mechanisms involved in the generation of thorny red blood cells (RBCs), known as acanthocytes, in patients with neuroacanthocytosis, a heterogenous group of neurodegenerative hereditary disorders that include chorea-acanthocytosis (ChAc) and McLeod syndrome (MLS). RECENT FINDINGS: Although molecular defects associated with neuroacanthocytosis have been identified recently, their pathophysiology and the related RBC abnormalities are largely unknown. Studies in ChAc RBCs have shown an altered association between the cytoskeleton and the integral membrane protein compartment in the absence of major changes in RBC membrane composition. In ChAc RBCs, abnormal Lyn kinase activation in a Syk-independent fashion has been reported recently, resulting in increased band 3 tyrosine phosphorylation and perturbation of the stability of the multiprotein band 3-based complexes bridging the membrane to the spectrin-based membrane skeleton. Similarly, in MLS, the absence of XK-protein, which is associated with the spectrin-actin-4.1 junctional complex, is associated with an abnormal membrane protein phosphorylation state, with destabilization of the membrane skeletal network resulting in generation of acanthocytes. SUMMARY: A novel mechanism in generation of acanthocytes involving abnormal Lyn activation, identified in ChAc, expands the acanthocytosis phenomenon toward protein-protein interactions, controlled by phosphorylation-related abnormal signaling.


Acanthocytes/physiology , Neuroacanthocytosis/physiopathology , Amino Acid Transport Systems, Neutral/physiology , Cytoskeleton/physiology , Humans , Phosphotransferases (Alcohol Group Acceptor)/physiology , Vesicular Transport Proteins/physiology
13.
Am J Physiol Endocrinol Metab ; 306(1): E65-74, 2014 Jan 01.
Article En | MEDLINE | ID: mdl-24222668

The proton-coupled amino acid transporter 1 (PAT1) is a transporter of amino acids in small intestinal enterocytes. PAT1 is, however, also capable of regulating cell growth and sensing the availability of amino acids in other cell types. The aim of the present study was to investigate the localization and function of PAT1 in smooth muscle cells (SMCs). The PAT1 protein was found in smooth muscles from rat intestine and in the embryonic rat aorta cell line A7r5. Immunolocalization and cellular fractionation studies revealed that the majority of the PAT1 protein located within the cell nucleus of A7r5 cells. These results were confirmed in primary SMCs derived from rat aorta and colon. A 3'-untranslated region of the PAT1 transcript directed the nuclear localization. Neither cellular starvation nor cell division altered the nuclear localization. In agreement, uptake studies of l-proline, a PAT1 substrate, in A7r5 cells suggested an alternative role for PAT1 in SMCs than in transport. To shed light on the function of PAT1 in A7r5 cells, experiments with downregulation of the PAT1 level by use of a siRNA approach were conducted. The growth rates of the cells were evaluated, and knockdown of PAT1 led to induced cellular growth, suggesting a role for PAT1 in regulating cellular proliferation of SMCs.


Amino Acid Transport Systems, Neutral/analysis , Amino Acid Transport Systems, Neutral/physiology , Cell Nucleus/chemistry , Cell Proliferation , Myocytes, Smooth Muscle/physiology , Myocytes, Smooth Muscle/ultrastructure , Symporters/analysis , Symporters/physiology , Amino Acid Transport Systems, Neutral/genetics , Animals , Aorta , Caco-2 Cells , Cell Fractionation , Cell Line , Colon , Embryo, Mammalian , Gene Expression , Humans , Male , Proline/metabolism , RNA, Messenger/analysis , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Symporters/genetics , Transfection
14.
Neurochem Int ; 73: 166-71, 2014 Jul.
Article En | MEDLINE | ID: mdl-24333324

Solute neutral amino acid transporter 5 (SNAT5/SN2) is a member of the System N family, expressed in glial cells in the adult brain, able to transport glutamine, histidine or glycine among other substrates. Its tight association with synapses and its electroneutral mode of operation that allows the bidirectional movement of substrates, supports the idea that this transporter participates in the function of the glutamine-glutamate cycle between neurons and glia. Moreover, SNAT5/SN2 might contribute to the regulation of glycine concentration in glutamatergic synapses and, therefore, to the functioning of the N-methyl-d-aspartate (NMDA) subtype of glutamate receptors. Ontogenic maturation of these synapses occurs postnatally through the coordinate expression of a large number of receptors, transporters, structural and regulatory proteins that ensure the correct operation of the excitatory pathways in the central nervous system. Since the temporal pattern of expression of SNAT5/SN2 is unknown, we analyzed it by immunoblot and immunohistochemical techniques. Results indicate that the expression of SNAT5/SN2 is triggered between the second and third postnatal week in the cerebral cortex, in parallel to the expression of the vesicular glutamate transporter vGLUT1 and the glial glutamate transporter GLT1/EAAT2. In the cerebellum, this process occurs about one week later than in the cerebral cortex. Immunohistochemical staining of cortical sections shows that from postnatal day 14 to adulthood the transporter was expressed exclusively in glial cells. Our results are consistent with the idea that SNAT5/SN2 expression is coordinated with that of other proteins necessary for the operation of glutamatergic synapses and reinforce the existence of a regulatory cross-talk between neurons and glia that orchestrates the building up of these synapses.


Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/physiology , Glutamic Acid/physiology , Synaptic Transmission/physiology , Amino Acid Transport System X-AG/metabolism , Animals , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Female , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/genetics , Male , Neuroglia/physiology , Pregnancy , Rats , Rats, Wistar , Synaptic Transmission/genetics
15.
J Neurosci ; 33(44): 17429-34, 2013 Oct 30.
Article En | MEDLINE | ID: mdl-24174676

The mechanisms by which the excitatory neurotransmitter glutamate is recycled at synapses are currently unknown. By examining the functional expression of plasma membrane transporters at presynaptic terminals, we aim to elucidate some of the mechanisms of glutamate recycling. Using whole-cell voltage-clamp recordings from rat calyx of Held presynaptic terminals, our data show, for the first time, that the glutamate precursor glutamine causes the direct activation of an electrogenic, sodium-dependent presynaptic transporter, which supplies glutamine for generation of presynaptic glutamate and helps sustain synaptic transmission. Interestingly, the functional expression of this transporter at the presynaptic plasma membrane is dynamically controlled by electrical activity of the terminal, indicating that uptake of neurotransmitter precursors is controlled by the demand at an individual terminal. Induction of the transporter current is calcium-dependent and inhibited by botulinum neurotoxin C, demonstrating the involvement of SNARE-dependent exocytosis in inserting transporters into the plasma membrane when the terminal is active. Conversely, inactivity of the presynaptic terminal results in removal of transporters via clathrin-mediated endocytosis. To investigate whether the presynaptic glutamine transporter supplies the precursor for generating the synaptically released glutamate, we measured miniature EPSCs to assess vesicular glutamate content. When the presynaptic glutamate pool was turned over by synaptic activity, inhibiting the presynaptic glutamine transporters with MeAIB reduced the miniature EPSC amplitude significantly. This demonstrates that presynaptic glutamine transport is centrally involved in the production of glutamate and assists in maintaining excitatory neurotransmission.


Amino Acid Transport Systems, Neutral/biosynthesis , Brain Stem/physiology , Glutamic Acid/physiology , Glutamine/biosynthesis , Presynaptic Terminals/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Amino Acid Transport Systems, Neutral/physiology , Animals , Biological Transport, Active/physiology , Brain Stem/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Glutamic Acid/metabolism , Glutamine/metabolism , Glutamine/physiology , Male , Organ Culture Techniques , Rats , Rats, Wistar , Synapses/physiology , Synaptic Transmission/drug effects , beta-Alanine/analogs & derivatives , beta-Alanine/pharmacology
16.
Mol Aspects Med ; 34(2-3): 139-58, 2013.
Article En | MEDLINE | ID: mdl-23506863

Amino acids are necessary for all living cells and organisms. Specialized transporters mediate the transfer of amino acids across plasma membranes. Malfunction of these proteins can affect whole-body homoeostasis giving raise to diverse human diseases. Here, we review the main features of the SLC3 and SLC7 families of amino acid transporters. The SLC7 family is divided into two subfamilies, the cationic amino acid transporters (CATs), and the L-type amino acid transporters (LATs). The latter are the light or catalytic subunits of the heteromeric amino acid transporters (HATs), which are associated by a disulfide bridge with the heavy subunits 4F2hc or rBAT. These two subunits are glycoproteins and form the SLC3 family. Most CAT subfamily members were functionally characterized and shown to function as facilitated diffusers mediating the entry and efflux of cationic amino acids. In certain cells, CATs play an important role in the delivery of L-arginine for the synthesis of nitric oxide. HATs are mostly exchangers with a broad spectrum of substrates and are crucial in renal and intestinal re-absorption and cell redox balance. Furthermore, the role of the HAT 4F2hc/LAT1 in tumor growth and the application of LAT1 inhibitors and PET tracers for reduction of tumor progression and imaging of tumors are discussed. Finally, we describe the link between specific mutations in HATs and the primary inherited aminoacidurias, cystinuria and lysinuric protein intolerance.


Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Basic/physiology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/physiology , Models, Biological , Models, Molecular , Multigene Family/genetics , Neoplasms/metabolism , Amino Acid Transport Systems, Basic/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Biological Transport/physiology , Humans , Phylogeny , Protein Conformation
17.
Mol Aspects Med ; 34(2-3): 197-219, 2013.
Article En | MEDLINE | ID: mdl-23506866

The SLC6 family of secondary active transporters are integral membrane solute carrier proteins characterized by the Na(+)-dependent translocation of small amino acid or amino acid-like substrates. SLC6 transporters, which include the serotonin, dopamine, norepinephrine, GABA, taurine, creatine, as well as amino acid transporters, are associated with a number of human diseases and disorders making this family a critical target for therapeutic development. In addition, several members of this family are directly involved in the action of drugs of abuse such as cocaine, amphetamines, and ecstasy. Recent advances providing structural insight into this family have vastly accelerated our ability to study these proteins and their involvement in complex biological processes.


Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/physiology , Models, Molecular , Multigene Family/genetics , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/physiology , Protein Conformation , Sodium/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Biological Transport/physiology , Gene Expression Regulation/physiology , Humans , Lipoylation , Phylogeny , Plasma Membrane Neurotransmitter Transport Proteins/metabolism
18.
Blood Cells Mol Dis ; 50(2): 80-5, 2013 Feb.
Article En | MEDLINE | ID: mdl-23122227

XK is a putative transporter of unknown function that is ubiquitously expressed and linked through disulfide bonds to Kell protein, an endothelin-3 (ET-3)-converting enzyme. We generated three knockout (KO) mice that lacked either Xk, Kell or both proteins and characterized erythrocyte cation levels, transport and hematological parameters. Absence of Xk or Kell was accompanied by changes in erythrocyte K(+), Mg(2+), Na(+) and Ca(2+) transport that were associated with changes in mean cellular volume and corpuscular hemoglobin concentration mean. Baseline Ca(2+)-ATPase activity was undetected in erythrocytes from all three mouse types but was restored upon pre-incubation with ET-3. Consistent with these alterations in Ca(2+) handling, we observed increased Gardos channel activity in Kel and Xk KO mice. In addition Kel deletion was associated with increased Mg(2+) permeability while Xk deletion blocked Na/Mg exchanger activity. Our results provide evidence that cellular divalent cation regulation is functionally coupled to the Kell/XK system in erythrocytes and loss of this complex may contribute to acanthocytosis formation in McLeod syndrome.


Amino Acid Transport Systems, Neutral/physiology , Cations, Divalent/blood , Erythrocytes/metabolism , Kell Blood-Group System/physiology , Abetalipoproteinemia/blood , Abetalipoproteinemia/genetics , Acanthocytes , Amino Acid Transport Systems, Neutral/blood , Amino Acid Transport Systems, Neutral/deficiency , Amino Acid Transport Systems, Neutral/genetics , Animals , Antiporters/blood , Calcium/blood , Calcium-Transporting ATPases/blood , Endothelin-3/pharmacology , Erythrocyte Volume , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/genetics , Hemolysis/genetics , Homeostasis , Intermediate-Conductance Calcium-Activated Potassium Channels/blood , Ion Transport , Kell Blood-Group System/genetics , Magnesium/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes , Neuroacanthocytosis , Potassium/blood , Receptors, Endothelin/blood , Sodium/blood
19.
FASEB J ; 26(9): 3779-89, 2012 Sep.
Article En | MEDLINE | ID: mdl-22649030

Cystinosis is a rare autosomal recessive disease characterized by cystine crystal accumulation leading to multiorgan dysfunctions and caused by mutation in CTNS. CTNS encodes cystinosin, a cystine/H(+) symporter that exports cystine out of the lysosomes. Patients with cystinosis frequently exhibit blond hair and fair complexion, suggesting an alteration in melanogenesis. However, the pigmentation singularities of these patients have not been studied, and the role of cystinosin in melanogenesis has remained unknown. In our study, a clinical evaluation of 27 patients with cystinosis showed that 44% had a cutaneous pigmentation dilution compared to their relatives. Analysis of the hair melanin content in these patients by HPLC demonstrated a 50% decrease in eumelanin (4360 vs. 9360 ng/mg), and a 2-fold increase in pheomelanin (53 vs. 20 ng/mg), the yellow/red pigments. Cystinosin-deficient mice also showed a 4-fold increase in hair pheomelanin content. In vitro studies showed that cystinosin was located at melanosomes. CTNS silencing led to a 75% reduction of melanin synthesis that was caused by a degradation of tyrosinase by lysosomal proteases. Our results objectify the pigmentation defect in patients with cystinosis. We also identify the role of CTNS in melanogenesis and add a new gene to the list of the genes involved in the control of skin and hair pigmentation.


Amino Acid Transport Systems, Neutral/physiology , Melanins/biosynthesis , Melanosomes/metabolism , Adolescent , Adult , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Cell Line, Tumor , Child , Child, Preschool , Cystinosis/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Skin Pigmentation/genetics , Young Adult
20.
Biochim Biophys Acta ; 1818(7): 1737-44, 2012 Jul.
Article En | MEDLINE | ID: mdl-22402268

K+-coupled amino acid transporter 1 (KAAT1) belongs to the NSS family of solute transporters and it is expressed in the midgut and in salivary glands of Manduca sexta larvae. As more than 80% of family members, KAATI shows a stretch of three glycines (G85-G87) that according to the structure of the prototype transporter LeuT, is located close to the access of the permeation pathway. In this work the role of the triplet has been investigated by alanine and cysteine scanning methods in protein heterologously expressed in Xenopus laevis oocytes. All the mutants were functional but the surface expression level was reduced for G85A and G87A mutants and unaffected for G86A mutant. All presented altered amino acid uptake and transport associated currents in the presence of each of the cations (Na+, K+, Li+) that can be exploited by the wt. G87A mutant induced increased uncoupled fluxes in the presence of all the cations. Cross-linking studies, performed by the treatment of cysteine mutants with the oxidative complex Cu(Il)(l,10-phenanthroline)3, showed that limiting the flexibility of the region by covalent blockage of position 87, causes a significant reduction of amino acid uptake. Na+ protected G87C mutant from oxidation, both directly and indirectly. The conserved glycine triplet in KAAT1 plays therefore a complex role that allows initial steps of cation interaction with the transporter.


Amino Acid Transport Systems, Neutral/genetics , Glycine/genetics , Insect Proteins/genetics , Mutation , Alanine/genetics , Alanine/metabolism , Alanine/physiology , Amino Acid Sequence , Amino Acid Substitution , Amino Acid Transport Systems, Neutral/metabolism , Amino Acid Transport Systems, Neutral/physiology , Amino Acids/metabolism , Animals , Biological Transport/drug effects , Conserved Sequence/genetics , Dose-Response Relationship, Drug , Glycine/metabolism , Glycine/physiology , Insect Proteins/metabolism , Insect Proteins/physiology , Lithium/metabolism , Lithium/pharmacology , Manduca/genetics , Manduca/metabolism , Membrane Potentials/drug effects , Oocytes/metabolism , Oocytes/physiology , Potassium/metabolism , Potassium/pharmacology , Sequence Homology, Amino Acid , Sodium/metabolism , Sodium/pharmacology , Xenopus laevis
...