Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
Annu Rev Nutr ; 43: 73-99, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37285555

RESUMEN

Amino acids derived from protein digestion are important nutrients for the growth and maintenance of organisms. Approximately half of the 20 proteinogenic amino acids can be synthesized by mammalian organisms, while the other half are essential and must be acquired from the nutrition. Absorption of amino acids is mediated by a set of amino acid transporters together with transport of di- and tripeptides. They provide amino acids for systemic needs and for enterocyte metabolism. Absorption is largely complete at the end of the small intestine. The large intestine mediates the uptake of amino acids derived from bacterial metabolism and endogenous sources. Lack of amino acid transporters and peptide transporter delays the absorption of amino acids and changes sensing and usage of amino acids by the intestine. This can affect metabolic health through amino acid restriction, sensing of amino acids, and production of antimicrobial peptides.


Asunto(s)
Antifibrinolíticos , Intestinos , Humanos , Animales , Transporte Biológico , Aminoácidos , Estado Nutricional , Enterocitos , Mamíferos
2.
PLoS Pathog ; 17(2): e1009259, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33600495

RESUMEN

The human malaria parasite Plasmodium falciparum relies on lipids to survive; this makes its lipid metabolism an attractive drug target. The lipid phosphatidylserine (PS) is usually confined to the inner leaflet of the red blood cell membrane (RBC) bilayer; however, some studies suggest that infection with the intracellular parasite results in the presence of this lipid in the RBC membrane outer leaflet, where it could act as a recognition signal to phagocytes. Here, we used fluorescent lipid analogues and probes to investigate the enzymatic reactions responsible for maintaining asymmetry between membrane leaflets, and found that in parasitised RBCs the maintenance of membrane asymmetry was partly disrupted, and PS was increased in the outer leaflet. We examined the underlying causes for the differences between uninfected and infected RBCs using fluorescent dyes and probes, and found that calcium levels increased in the infected RBC cytoplasm, whereas membrane cholesterol was depleted from the erythrocyte plasma membrane. We explored the resulting effect of PS exposure on enhanced phagocytosis by monocytes, and show that infected RBCs must expend energy to limit phagocyte recognition, and provide experimental evidence that PS exposure contributes to phagocytic recognition of P. falciparum-infected RBCs. Together, these findings underscore the pivotal role for PS exposure on the surface of Plasmodium falciparum-infected erythrocytes for in vivo interactions with the host immune system, and provide a rationale for targeted antimalarial drug design.


Asunto(s)
Calcio/metabolismo , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Malaria Falciparum/metabolismo , Monocitos/metabolismo , Fagocitosis , Fosfatidilserinas/metabolismo , Membrana Eritrocítica/parasitología , Eritrocitos/parasitología , Humanos , Malaria Falciparum/parasitología , Monocitos/parasitología , Plasmodium falciparum/aislamiento & purificación
3.
PLoS Pathog ; 17(8): e1009835, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34432856

RESUMEN

Intracellular parasites of the phylum Apicomplexa are dependent on the scavenging of essential amino acids from their hosts. We previously identified a large family of apicomplexan-specific plasma membrane-localized amino acid transporters, the ApiATs, and showed that the Toxoplasma gondii transporter TgApiAT1 functions in the selective uptake of arginine. TgApiAT1 is essential for parasite virulence, but dispensable for parasite growth in medium containing high concentrations of arginine, indicating the presence of at least one other arginine transporter. Here we identify TgApiAT6-1 as the second arginine transporter. Using a combination of parasite assays and heterologous characterisation of TgApiAT6-1 in Xenopus laevis oocytes, we demonstrate that TgApiAT6-1 is a general cationic amino acid transporter that mediates both the high-affinity uptake of lysine and the low-affinity uptake of arginine. TgApiAT6-1 is the primary lysine transporter in the disease-causing tachyzoite stage of T. gondii and is essential for parasite proliferation. We demonstrate that the uptake of cationic amino acids by TgApiAT6-1 is 'trans-stimulated' by cationic and neutral amino acids and is likely promoted by an inwardly negative membrane potential. These findings demonstrate that T. gondii has evolved overlapping transport mechanisms for the uptake of essential cationic amino acids, and we draw together our findings into a comprehensive model that highlights the finely-tuned, regulated processes that mediate cationic amino acid scavenging by these intracellular parasites.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Aminoácidos Esenciales/metabolismo , Fibroblastos/metabolismo , Oocitos/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasmosis/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Animales , Arginina/metabolismo , Transporte Biológico , Fibroblastos/parasitología , Humanos , Lisina/metabolismo , Oocitos/parasitología , Proteínas Protozoarias/genética , Toxoplasma/fisiología , Toxoplasmosis/parasitología , Xenopus laevis
4.
Nat Immunol ; 12(5): 441-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21423173

RESUMEN

Subcompartments of the plasma membrane are believed to be critical for lymphocyte responses, but few genetic tools are available to test their function. Here we describe a previously unknown X-linked B cell-deficiency syndrome in mice caused by mutations in Atp11c, which encodes a member of the P4 ATPase family thought to serve as 'flippases' that concentrate aminophospholipids in the cytoplasmic leaflet of cell membranes. Defective ATP11C resulted in a lower rate of phosphatidylserine translocation in pro-B cells and much lower pre-B cell and B cell numbers despite expression of pre-rearranged immunoglobulin transgenes or enforced expression of the prosurvival protein Bcl-2 to prevent apoptosis and abolished pre-B cell population expansion in response to a transgene encoding interleukin 7. The only other abnormalities we noted were anemia, hyperbilirubinemia and hepatocellular carcinoma. Our results identify an intimate connection between phospholipid transport and B lymphocyte function.


Asunto(s)
Adenosina Trifosfatasas/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Endocitosis/inmunología , Fosfoserina/inmunología , Adenosina Trifosfatasas/genética , Animales , Linfocitos B/enzimología , Secuencia de Bases , Femenino , Citometría de Flujo , Genes bcl-2/inmunología , Interleucina-7/genética , Interleucina-7/inmunología , Hígado/citología , Hígado/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Mutagénesis/inmunología , ARN Mensajero/química , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Am J Physiol Renal Physiol ; 323(4): F455-F467, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35979966

RESUMEN

B0AT1 (Slc6a19) mediates absorption of neutral amino acids in the small intestine and in the kidneys, where it is primarily expressed in early proximal tubules (S1-S2). To determine the role of B0AT1 in nephropathy induced by aristolochic acid (AA), which targets the proximal tubule, littermate female B0AT1-deficient (Slc6a19-/-), heterozygous (Slc6a19+/-), and wild-type (WT) mice were administered AA (10 mg/kg ip) or vehicle every 3 days for 3 wk, and analyses were performed after the last injection or 3 wk later. Vehicle-treated mice lacking Slc6a19 showed normal body and kidney weight and plasma creatinine versus WT mice. The urinary glucose-to-creatinine ratio (UGCR) and urinary albumin-to-creatinine ratio (UACR) were two to four times higher in vehicle-treated Slc6a19-/- versus WT mice, associated with lesser expression of early proximal transporters Na+-glucose cotransporter 2 and megalin, respectively. AA caused tubular injury independently of B0AT1, including robust increases in cortical mRNA expression of p53, p21, and hepatitis A virus cellular receptor 1 (Havcr1), downregulation of related proximal tubule amino acid transporters B0AT2 (Slc6a15), B0AT3 (Slc6a18), and Slc7a9, and modest histological tubular damage and a rise in plasma creatinine. Absence of B0AT1, however, attenuated AA-induced cortical upregulation of mRNA markers of senescence (p16), inflammation [lipocalin 2 (Lcn2), C-C motif chemokine ligand 2 (Ccl2), and C-C motif chemokine receptor 2 (Ccr2)], and fibrosis [tissue inhibitor of metallopeptidase 1 (Timp1), transforming growth factor-ß1 (Tgfb1), and collagen type I-α1 (Col1a1)], associated with lesser fibrosis staining, lesser suppression of proximal tubular organic anion transporter 1, restoration of Na+-glucose cotransporter 2 expression, and prevention of the AA-induced fivefold increase in the urinary albumin-to-creatinine ratio observed in WT mice. The data suggest that proximal tubular B0AT1 is important for the physiology of renal glucose and albumin retention but potentially deleterious for the kidney response following AA-induced kidney injury.NEW & NOTEWORTHY Based on insights from studies manipulating glucose transport, the hypothesis has been proposed that inhibiting intestinal uptake or renal reabsorption of energy substrates has unique therapeutic potential to improve metabolic disease and kidney outcome in response to injury. The present study takes this idea to B0AT1, the major transporter for neutral amino acids in the intestine and kidney, and shows that its absence attenuates aristolochic acid-induced nephropathy.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Aminoácidos Neutros , Ácidos Aristolóquicos , Enfermedades Renales , Albúminas/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animales , Ácidos Aristolóquicos/toxicidad , Creatinina , Femenino , Fibrosis , Glucosa , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Ratones , ARN Mensajero
6.
J Nutr ; 152(1): 16-28, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34718668

RESUMEN

Amino acid homeostasis is maintained by import, export, oxidation, and synthesis of nonessential amino acids, and by the synthesis and breakdown of protein. These processes work in conjunction with regulatory elements that sense amino acids or their metabolites. During and after nutrient intake, amino acid homeostasis is dominated by autoregulatory processes such as transport and oxidation of excess amino acids. Amino acid deprivation triggers processes such as autophagy and the execution of broader transcriptional programs to maintain plasma amino acid concentrations. Amino acid transport plays a crucial role in the absorption of amino acids in the intestine, the distribution of amino acids across cells and organs, the recycling of amino acids in the kidney, and the recycling of amino acids after protein breakdown.


Asunto(s)
Aminoácidos , Autofagia , Aminoácidos/metabolismo , Animales , Transporte Biológico , Homeostasis , Oxidación-Reducción
7.
PLoS Pathog ; 15(2): e1007577, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30742695

RESUMEN

Apicomplexan parasites are auxotrophic for a range of amino acids which must be salvaged from their host cells, either through direct uptake or degradation of host proteins. Here, we describe a family of plasma membrane-localized amino acid transporters, termed the Apicomplexan Amino acid Transporters (ApiATs), that are ubiquitous in apicomplexan parasites. Functional characterization of the ApiATs of Toxoplasma gondii indicate that several of these transporters are important for intracellular growth of the tachyzoite stage of the parasite, which is responsible for acute infections. We demonstrate that the ApiAT protein TgApiAT5-3 is an exchanger for aromatic and large neutral amino acids, with particular importance for L-tyrosine scavenging and amino acid homeostasis, and that TgApiAT5-3 is critical for parasite virulence. Our data indicate that T. gondii expresses additional proteins involved in the uptake of aromatic amino acids, and we present a model for the uptake and homeostasis of these amino acids. Our findings identify a family of amino acid transporters in apicomplexans, and highlight the importance of amino acid scavenging for the biology of this important phylum of intracellular parasites.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Toxoplasma/metabolismo , Tirosina/fisiología , Animales , Apicomplexa/metabolismo , Transporte Biológico , Interacciones Huésped-Parásitos , Transporte Iónico , Parásitos , Proteínas Protozoarias , Tirosina/metabolismo
8.
J Biol Chem ; 294(11): 4012-4026, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30635397

RESUMEN

The neutral amino acid transporter solute carrier family 1 member 5 (SLC1A5 or ASCT2) is overexpressed in many cancers. To identify its roles in tumors, we employed 143B osteosarcoma cells and HCC1806 triple-negative breast cancer cells with or without ASCT2 deletion. ASCT2ko 143B cells grew well in standard culture media, but ASCT2 was required for optimal growth at <0.5 mm glutamine, with tumor spheroid growth and monolayer migration of 143B ASCT2ko cells being strongly impaired at lower glutamine concentrations. However, the ASCT2 deletion did not affect matrix-dependent invasion. ASCT2ko 143B xenografts in nude mice exhibited a slower onset of growth and a higher number of small tumors than ASCT2wt 143B xenografts, but did not differ in average tumor size 25 days after xenotransplantation. ASCT2 deficiency was compensated by increased levels of sodium neutral amino acid transporter 1 (SNAT1 or SLC38A1) and SNAT2 (SLC38A2) in ASCT2ko 143B cells, mediated by a GCN2 EIF2α kinase (GCN2)-dependent pathway, but this compensation was not observed in ASCT2ko HCC1806 cells. Combined SNAT1 silencing and GCN2 inhibition significantly inhibited growth of ASCT2ko HCC1806 cells, but not of ASCT2ko 143B cells. Similarly, pharmacological inhibition of l-type amino acid transporter 1 (LAT1) and GCN2 significantly inhibited growth of ASCT2ko HCC1806 cells, but not of ASCT2ko 143B cells. We conclude that cancer cells with reduced transporter plasticity are more vulnerable to disruption of amino acid homeostasis than cells with a full capacity to up-regulate redundant transporters by an integrated stress response.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/genética , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Antígenos de Histocompatibilidad Menor/genética , Osteosarcoma/genética , Osteosarcoma/patología , Sistema de Transporte de Aminoácidos ASC/deficiencia , Sistema de Transporte de Aminoácidos ASC/metabolismo , Animales , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/metabolismo , Mutación , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Osteosarcoma/metabolismo , Células Tumorales Cultivadas
9.
Int J Mol Sci ; 21(17)2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32859034

RESUMEN

Amino acids are indispensable for the growth of cancer cells. This includes essential amino acids, the carbon skeleton of which cannot be synthesized, and conditionally essential amino acids, for which the metabolic demands exceed the capacity to synthesize them. Moreover, amino acids are important signaling molecules regulating metabolic pathways, protein translation, autophagy, defense against reactive oxygen species, and many other functions. Blocking uptake of amino acids into cancer cells is therefore a viable strategy to reduce growth. A number of studies have used genome-wide silencing or knock-out approaches, which cover all known amino acid transporters in a large variety of cancer cell lines. In this review, these studies are interrogated together with other databases to identify vulnerabilities with regard to amino acid transport. Several themes emerge, such as synthetic lethality, reduced redundancy, and selective vulnerability, which can be exploited to stop cancer cell growth.


Asunto(s)
Sistemas de Transporte de Aminoácidos/genética , Antineoplásicos/farmacología , Neoplasias/genética , Sistemas de Transporte de Aminoácidos/efectos de los fármacos , Aminoácidos/metabolismo , Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Silenciador del Gen , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Mutaciones Letales Sintéticas
10.
Biochem J ; 474(12): 1935-1963, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28546457

RESUMEN

Cells have a constant turnover of proteins that recycle most amino acids over time. Net loss is mainly due to amino acid oxidation. Homeostasis is achieved through exchange of essential amino acids with non-essential amino acids and the transfer of amino groups from oxidised amino acids to amino acid biosynthesis. This homeostatic condition is maintained through an active mTORC1 complex. Under amino acid depletion, mTORC1 is inactivated. This increases the breakdown of cellular proteins through autophagy and reduces protein biosynthesis. The general control non-derepressable 2/ATF4 pathway may be activated in addition, resulting in transcription of genes involved in amino acid transport and biosynthesis of non-essential amino acids. Metabolism is autoregulated to minimise oxidation of amino acids. Systemic amino acid levels are also tightly regulated. Food intake briefly increases plasma amino acid levels, which stimulates insulin release and mTOR-dependent protein synthesis in muscle. Excess amino acids are oxidised, resulting in increased urea production. Short-term fasting does not result in depletion of plasma amino acids due to reduced protein synthesis and the onset of autophagy. Owing to the fact that half of all amino acids are essential, reduction in protein synthesis and amino acid oxidation are the only two measures to reduce amino acid demand. Long-term malnutrition causes depletion of plasma amino acids. The CNS appears to generate a protein-specific response upon amino acid depletion, resulting in avoidance of an inadequate diet. High protein levels, in contrast, contribute together with other nutrients to a reduction in food intake.


Asunto(s)
Aminoácidos/metabolismo , Dieta , Homeostasis , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Regulación del Apetito , Autofagia , Transporte Biológico , Sistema Nervioso Central/metabolismo , Dieta/efectos adversos , Ayuno/metabolismo , Regulación de la Expresión Génica , Humanos , Desnutrición/etiología , Desnutrición/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Neuronas/metabolismo , Oxidación-Reducción , Periodo Posprandial , Proteínas Serina-Treonina Quinasas/metabolismo
11.
Int J Mol Sci ; 19(11)2018 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-30441827

RESUMEN

Recent studies have established that dietary protein restriction improves metabolic health and glucose homeostasis. SLC6A19 (B°AT1) is the major neutral amino acid transporter in the intestine and carries out the bulk of amino acid absorption from the diet. Mice lacking SLC6A19 show signs of protein restriction, have improved glucose tolerance, and are protected from diet-induced obesity. Pharmacological blockage of this transporter could be used to induce protein restriction and to treat metabolic diseases such as type 2 diabetes. A few novel inhibitors of SLC6A19 have recently been identified using in vitro compound screening, but it remains unclear whether these compounds block the transporter in vivo. To evaluate the efficacy of SLC6A19 inhibitors biomarkers are required that can reliably detect successful inhibition of the transporter in mice. A gas chromatography mass spectrometry (GC-MS)-based untargeted metabolomics approach was used to discriminate global metabolite profiles in plasma, urine and faecal samples from SLC6A19ko and wt mice. Due to inefficient absorption in the intestine and lack of reabsorption in the kidney, significantly elevated amino acids levels were observed in urine and faecal samples. By contrast, a few neutral amino acids were reduced in the plasma of male SLC6A19ko mice as compared to other biological samples. Metabolites of bacterial protein fermentation such as p-cresol glucuronide and 3-indole-propionic acid were more abundant in SLC6A19ko mice, indicating protein malabsorption of dietary amino acids. Consistently, plasma appearance rates of [14C]-labelled neutral amino acids were delayed in SLC6A19ko mice as compared to wt after intra-gastric administration of a mixture of amino acids. Receiver operating characteristic (ROC) curve analysis was used to validate the potential use of these metabolites as biomarkers. These findings provide putative metabolite biomarkers that can be used to detect protein malabsorption and the inhibition of this transporter in intestine and kidney.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inhibidores , Aminoácidos/sangre , Enfermedades Metabólicas/sangre , Aminoácidos/orina , Animales , Benzotropina/farmacología , Biomarcadores/metabolismo , Biomarcadores/orina , Proteínas en la Dieta/metabolismo , Femenino , Absorción Intestinal , Masculino , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/orina , Metaboloma , Ratones , Ratones Endogámicos C57BL , Reabsorción Renal
12.
Int J Mol Sci ; 19(4)2018 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-29561757

RESUMEN

Astrocytes are glial cells that have an intimate physical and functional association with synapses in the brain. One of their main roles is to recycle the neurotransmitters glutamate and gamma-aminobutyric acid (GABA), as a component of the glutamate/GABA-glutamine cycle. They perform this function by sequestering neurotransmitters and releasing glutamine via the neutral amino acid transporter SNAT3. In this way, astrocytes regulate the availability of neurotransmitters and subsequently influence synaptic function. Since many plasma membrane transporters are regulated by protein kinase C (PKC), the aim of this study was to understand how PKC influences SNAT3 glutamine transport in astrocytes located immediately adjacent to synapses. We studied SNAT3 transport by whole-cell patch-clamping and fluorescence pH imaging of single astrocytes in acutely isolated brainstem slices, adjacent to the calyx of the Held synapse. Activation of SNAT3-mediated glutamine transport in these astrocytes was reduced to 77 ± 6% when PKC was activated with phorbol 12-myristate 13-acetate (PMA). This effect was very rapid (within ~20 min) and eliminated by application of bisindolylmaleimide I (Bis I) or 7-hydroxystaurosporine (UCN-01), suggesting that activation of conventional isoforms of PKC reduces SNAT3 function. In addition, cell surface biotinylation experiments in these brain slices show that the amount of SNAT3 in the plasma membrane is reduced by a comparable amount (to 68 ± 5%) upon activation of PKC. This indicates a role for PKC in dynamically controlling the trafficking of SNAT3 transporters in astrocytes in situ. These data demonstrate that PKC rapidly regulates the astrocytic glutamine release mechanism, which would influence the glutamine availability for adjacent synapses and control levels of neurotransmission.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Astrocitos/metabolismo , Proteína Quinasa C/metabolismo , Sinapsis/metabolismo , Animales , Encéfalo/metabolismo , Endocitosis , Activación Enzimática , Isoenzimas/metabolismo , Ratones Endogámicos C57BL , Ratas Wistar
13.
J Biol Chem ; 291(25): 13194-205, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27129276

RESUMEN

Many cancer cells depend on glutamine as they use the glutaminolysis pathway to generate building blocks and energy for anabolic purposes. As a result, glutamine transporters are essential for cancer growth and are potential targets for cancer chemotherapy with ASCT2 (SLC1A5) being investigated most intensively. Here we show that HeLa epithelial cervical cancer cells and 143B osteosarcoma cells express a set of glutamine transporters including SNAT1 (SLC38A1), SNAT2 (SLC38A2), SNAT4 (SLC38A4), LAT1 (SLC7A5), and ASCT2 (SLC1A5). Net glutamine uptake did not depend on ASCT2 but required expression of SNAT1 and SNAT2. Deletion of ASCT2 did not reduce cell growth but caused an amino acid starvation response and up-regulation of SNAT1 to replace ASCT2 functionally. Silencing of GCN2 in the ASCT2(-/-) background reduced cell growth, showing that a combined targeted approach would inhibit growth of glutamine-dependent cancer cells.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos A/fisiología , Proliferación Celular , Eliminación de Gen , Expresión Génica , Glutamina/metabolismo , Células HeLa , Homeostasis , Humanos , Transporte Iónico , Redes y Vías Metabólicas , Antígenos de Histocompatibilidad Menor
14.
Glia ; 65(6): 900-916, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28272791

RESUMEN

The release of glutamine from astrocytes adjacent to synapses in the central nervous system is thought to play a vital role in the mechanism of glutamate recycling and is therefore important for maintaining excitatory neurotransmission. Here we investigate the nature of astrocytic membrane transport of glutamine in rat brainstem slices, using electrophysiological recording and fluorescent imaging of pHi and Nai+. Glutamine application to perisynaptic astrocytes induced a membrane current, caused by activation of system A (SA) family transporters. A significant electroneutral component was also observed, which was mediated by the system N (SN) family transporters. This response was stimulated by glutamine (KM of 1.57 mM), histidine, and asparagine, but not by leucine or serine, indicating activation of the SNAT3 isoform of SN. We hypothesized that increasing the [Na+ ]i would alter the SNAT3 transporter equilibrium, thereby stimulating glutamine release. In support of this hypothesis, we show that SNAT3 transport can be driven by changing cation concentration and that manipulations to raise [Na+ ]i (activation of excitatory amino acid transporters (EAATs), SA transporters or AMPA receptors) all directly influence SNAT3 transport rate. A kinetic model of glutamine fluxes is presented, which shows that EAAT activation causes the release of glutamine, driven mainly by the increased [Na+ ]i . These data demonstrate that SNAT3 is functionally active in perisynaptic astrocytes in situ. As a result, astrocytic Nai+ signaling, as would be stimulated by neighboring synaptic activity, has the capacity to stimulate astrocytic glutamine release to support glutamate recycling.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Astrocitos/metabolismo , Glutamina/metabolismo , Espacio Intracelular/metabolismo , Sodio/metabolismo , Sinapsis/metabolismo , Sistema de Transporte de Aminoácidos A/metabolismo , Animales , Astrocitos/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Cationes Monovalentes/metabolismo , Femenino , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Concentración de Iones de Hidrógeno , Espacio Intracelular/efectos de los fármacos , Cinética , Litio/metabolismo , Masculino , Modelos Neurológicos , Ratas Wistar , Receptores AMPA/metabolismo , Sinapsis/efectos de los fármacos , Técnicas de Cultivo de Tejidos
15.
J Biol Chem ; 290(40): 24308-25, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26240152

RESUMEN

Many solute carrier 6 (SLC6) family transporters require ancillary subunits to modify their expression and activity. The main apical membrane neutral amino acid transporters in mouse intestine and kidney, B(0)AT1 and B(0)AT3, require the ancillary protein collectrin or ACE2 for plasma membrane expression. Expression and activity of SLC6 neurotransmitter transporters are modulated by interaction with syntaxin 1A. Utilizing monocarboxylate-B(0)AT1/3 fusion constructs, we discovered that collectrin is also necessary for B(0)AT1 and B(0)AT3 catalytic function. Syntaxin 1A and syntaxin 3 inhibit the membrane expression of B(0)AT1 by competing with collectrin for access. A mutagenesis screening approach identified residues on trans-membrane domains 1α, 5, and 7 on one face of B(0)AT3 as a key region involved in interaction with collectrin. Mutant analysis established residues that were involved in collectrin-dependent functions as follows: plasma membrane expression of B(0)AT3, catalytic activation, or both. These results identify a potential binding site for collectrin and other SLC6 ancillary proteins.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos/genética , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animales , Sitios de Unión , Biotinilación , Células CHO , Catálisis , Cricetinae , Cricetulus , Drosophila melanogaster , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Mutagénesis , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Qa-SNARE/metabolismo , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , Sintaxina 1/metabolismo , Xenopus laevis
16.
Pflugers Arch ; 468(2): 213-27, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26490457

RESUMEN

Glutamine, the most abundant amino acid in mammals, is critical for cell and organ functions. Its metabolism depends on the ability of cells to take up or release glutamine by transporters located in the plasma membrane. Several solute carrier (SLC) families transport glutamine, but the SLC38 family has been thought to be mostly responsible for glutamine transport. We demonstrate that despite the large number of glutamine transporters, the loss of Snat3/Slc38a3 glutamine transporter has a major impact on the function of organs expressing it. Snat3 mutant mice were generated by N-ethyl-N-nitrosurea (ENU) mutagenesis and showed stunted growth, altered amino acid levels, hypoglycemia, and died around 20 days after birth. Hepatic concentrations of glutamine, glutamate, leucine, phenylalanine, and tryptophan were highly reduced paralleled by downregulation of the mTOR pathway possibly linking reduced amino acid availability to impaired growth and glucose homeostasis. Snat3-deficient mice had altered urea levels paralleled by dysregulation of the urea cycle, gluconeogenesis, and glutamine synthesis. Mice were ataxic with higher glutamine but reduced glutamate and gamma-aminobutyric acid (GABA) levels in brain consistent with a major role of Snat3 in the glutamine-glutamate cycle. Renal ammonium excretion was lower, and the expression of enzymes and amino acid transporters involved in ammoniagenesis were altered. Thus, SNAT3 is a glutamine transporter required for amino acid homeostasis and determines critical functions in various organs. Despite the large number of glutamine transporters, loss of Snat3 cannot be compensated, suggesting that this transporter is a major route of glutamine transport in the liver, brain, and kidney.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos/metabolismo , Encéfalo/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Mutación , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/química , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Gluconeogénesis , Glucosa/metabolismo , Homeostasis , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Serina-Treonina Quinasas TOR/metabolismo
17.
J Biol Chem ; 289(28): 19531-7, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24898253

RESUMEN

Transmembrane lipid transporters are believed to establish and maintain phospholipid asymmetry in biological membranes; however, little is known about the in vivo function of the specific transporters involved. Here, we report that developing erythrocytes from mice lacking the putative phosphatidylserine flippase ATP11C showed a lower rate of PS translocation in vitro compared with erythrocytes from wild-type littermates. Furthermore, the mutant mice had an elevated percentage of phosphatidylserine-exposing mature erythrocytes in the periphery. Although erythrocyte development in ATP11C-deficient mice was normal, the mature erythrocytes had an abnormal shape (stomatocytosis), and the life span of mature erythrocytes was shortened relative to that in control littermates, resulting in anemia in the mutant mice. Thus, our findings uncover an essential role for ATP11C in erythrocyte morphology and survival and provide a new candidate for the rare inherited blood disorder stomatocytosis with uncompensated anemia.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Membrana Eritrocítica/enzimología , Fosfolípidos/metabolismo , Desequilibrio Ácido-Base/genética , Desequilibrio Ácido-Base/metabolismo , Desequilibrio Ácido-Base/patología , Adenosina Trifosfatasas/genética , Anemia Hemolítica Congénita/genética , Anemia Hemolítica Congénita/metabolismo , Anemia Hemolítica Congénita/patología , Animales , Transporte Biológico Activo , Supervivencia Celular/fisiología , Membrana Eritrocítica/genética , Eritrocitos Anormales/metabolismo , Eritrocitos Anormales/patología , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Errores Innatos del Metabolismo/patología , Ratones , Ratones Mutantes , Fosfolípidos/genética
18.
J Biol Chem ; 288(47): 33813-33823, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24121511

RESUMEN

Enterocytes are specialized to absorb nutrients from the lumen of the small intestine by expressing a select set of genes to maximize the uptake of nutrients. They develop from stem cells in the crypt and differentiate into mature enterocytes while moving along the crypt-villus axis. Using the Slc6a19 gene as an example, encoding the neutral amino acid transporter B(0)AT1, we studied regulation of the gene by transcription factors and epigenetic factors in the intestine. To investigate this question, we used a fractionation method to separate mature enterocytes from crypt cells and analyzed gene expression. Transcription factors HNF1a and HNF4a activate transcription of the Slc6a19 gene in villus enterocytes, whereas high levels of SOX9 repress expression in the crypts. CpG dinucleotides in the proximal promoter were highly methylated in the crypt and fully de-methylated in the villus. Furthermore, histone modification H3K27Ac, indicating an active promoter, was prevalent in villus cells but barely detectable in crypt cells. The results suggest that Slc6a19 expression in the intestine is regulated at three different levels involving promoter methylation, histone modification, and opposing transcription factors.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/biosíntesis , Enterocitos/metabolismo , Epigénesis Genética/fisiología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Regiones Promotoras Genéticas/fisiología , Factor de Transcripción SOX9/metabolismo , Transcripción Genética/fisiología , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Células Cultivadas , Metilación de ADN/fisiología , Enterocitos/citología , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Histonas/genética , Histonas/metabolismo , Ratones , Procesamiento Proteico-Postraduccional/fisiología , Factor de Transcripción SOX9/genética
19.
J Biol Chem ; 288(24): 17202-13, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23645676

RESUMEN

Endoplasmic reticulum (ER) stress-induced responses are associated with the loss of insulin-producing ß-cells in type 2 diabetes mellitus. ß-Cell survival during ER stress is believed to depend on decreased protein synthesis rates that are mediated via phosphorylation of the translation initiation factor eIF2α. It is reported here that chronic ER stress correlated with increased islet protein synthesis and apoptosis in ß-cells in vivo. Paradoxically, chronic ER stress in ß-cells induced an anabolic transcription program to overcome translational repression by eIF2α phosphorylation. This program included expression of amino acid transporter and aminoacyl-tRNA synthetase genes downstream of the stress-induced ATF4-mediated transcription program. The anabolic response was associated with increased amino acid flux and charging of tRNAs for branched chain and aromatic amino acids (e.g. leucine and tryptophan), the levels of which are early serum indicators of diabetes. We conclude that regulation of amino acid transport in ß-cells during ER stress involves responses leading to increased protein synthesis, which can be protective during acute stress but can lead to apoptosis during chronic stress. These studies suggest that the increased expression of amino acid transporters in islets can serve as early diagnostic biomarkers for the development of diabetes.


Asunto(s)
Aminoácidos/metabolismo , Apoptosis , Diabetes Mellitus Tipo 2/metabolismo , Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/fisiología , Factor de Transcripción Activador 4/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Animales , Supervivencia Celular , Diabetes Mellitus Tipo 2/patología , Factor 2 Eucariótico de Iniciación/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , ARN de Transferencia/metabolismo , Activación Transcripcional
20.
Pflugers Arch ; 466(1): 155-72, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24193407

RESUMEN

Transporters of the SLC38 family are found in all cell types of the body. They mediate Na(+)-dependent net uptake and efflux of small neutral amino acids. As a result they are particularly expressed in cells that grow actively, or in cells that carry out significant amino acid metabolism, such as liver, kidney and brain. SLC38 transporters occur in membranes that face intercellular space or blood vessels, but do not occur in the apical membrane of absorptive epithelia. In the placenta, they play a significant role in the transfer of amino acids to the foetus. Members of the SLC38 family are highly regulated in response to amino acid depletion, hypertonicity and hormonal stimuli. SLC38 transporters play an important role in amino acid signalling and have been proposed to act as transceptors independent of their transport function. The structure of SLC38 transporters is characterised by the 5 + 5 inverted repeat fold, which is observed in a wide variety of transport proteins.


Asunto(s)
Sistema de Transporte de Aminoácidos A/metabolismo , Aminoácidos/metabolismo , Sodio/metabolismo , Sistema de Transporte de Aminoácidos A/química , Sistema de Transporte de Aminoácidos A/genética , Sistema de Transporte de Aminoácidos A/inmunología , Animales , Femenino , Hepatocitos/metabolismo , Humanos , Músculos/metabolismo , Neuronas/metabolismo , Especificidad de Órganos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda