Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Microbiologyopen ; 13(3): e1415, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38780167

RESUMEN

The standard method of receptor activation involves the binding of signals or signal-loaded solute binding proteins (SBPs) to sensor domains. Many sensor histidine kinases (SHKs), which are activated by SBP binding, are encoded adjacent to their corresponding sbp gene. We examined three SBPs of Pseudomonas aeruginosa PAO1, encoded near the genes for the AgtS (PA0600) and AruS (PA4982) SHKs, to determine how common this arrangement is. Ligand screening and microcalorimetric studies revealed that the SBPs PA0602 and PA4985 preferentially bind to GABA (KD = 2.3 and 0.58 µM, respectively), followed by 5-aminovalerate (KD = 30 and 1.6 µM, respectively) and ethanoldiamine (KD = 2.3 and 0.58 µM, respectively). In contrast, AgtB (PA0604) exclusively recognizes 5-aminovaleric acid (KD = 2.9 µM). However, microcalorimetric titrations did not show any binding between the AgtS sensor domain and AgtB or PA0602, regardless of the presence of ligands. Similarly, bacterial two-hybrid assays did not demonstrate an interaction between PA4985 and the AruS sensor domain. Therefore, sbp and shk genes located nearby are not always functionally linked. We previously identified PA0222 as a GABA-specific SBP. The presence of three SBPs for GABA may be linked to GABA's role as a trigger for P. aeruginosa virulence.


Asunto(s)
Proteínas Bacterianas , Unión Proteica , Pseudomonas aeruginosa , Ácido gamma-Aminobutírico , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/enzimología , Ácido gamma-Aminobutírico/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Aminoácidos Neutros/metabolismo , Histidina Quinasa/metabolismo , Histidina Quinasa/genética , Calorimetría
2.
BMC Plant Biol ; 24(1): 447, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783192

RESUMEN

BACKGROUND: Amino acids are not only the main form of N in rice, but also are vital for its growth and development. These processes are facilitated by amino acid transporters within the plant. Despite their significance, only a few AAP amino acid transporters have been reported. RESULTS: In this study, we observed that there were differences in the expression of amino acid transporter OsAAP7 among 521 wild cultivated rice varieties, and it directly negatively correlated with tillering and grain yield per plant. We revealed that OsAAP7 protein was localized to the endoplasmic reticulum and had absorption and transport affinity for amino acids such as phenylalanine (Phe), lysine (Lys), leucine (Leu), and arginine (Arg) using subcellular localization, yeast substrate testing, fluorescent amino acid uptake, and amino acid content determination. Further hydroponic studies showed that exogenous application of amino acids Phe, Lys and Arg inhibited the growth of axillary buds in the overexpression lines, and promoted the elongation of axillary buds in the mutant lines. Finally, RNA-seq analysis showed that the expression patterns of genes related to nitrogen, auxin and cytokinin pathways were changed in axillary buds of OsAAP7 transgenic plants. CONCLUSIONS: This study revealed the gene function of OsAAP7, and found that blocking of amino acid transporter OsAAP7 with CRISPR/Cas9 technology promoted tillering and yield by determining basic and neutral amino acids accumulation in rice.


Asunto(s)
Oryza , Proteínas de Plantas , Oryza/genética , Oryza/metabolismo , Oryza/crecimiento & desarrollo , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Plantas Modificadas Genéticamente , Aminoácidos Neutros/metabolismo , Regulación de la Expresión Génica de las Plantas , Aminoácidos/metabolismo
3.
Pharmacol Rev ; 76(1): 142-193, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37940347

RESUMEN

The neutral amino acid transporter subfamily that consists of six members, consecutively SLC6A15-SLC620, also called orphan transporters, represents membrane, sodium-dependent symporter proteins that belong to the family of solute carrier 6 (SLC6). Primarily, they mediate the transport of neutral amino acids from the extracellular milieu toward cell or storage vesicles utilizing an electric membrane potential as the driving force. Orphan transporters are widely distributed throughout the body, covering many systems; for instance, the central nervous, renal, or intestinal system, supplying cells into molecules used in biochemical, signaling, and building pathways afterward. They are responsible for intestinal absorption and renal reabsorption of amino acids. In the central nervous system, orphan transporters constitute a significant medium for the provision of neurotransmitter precursors. Diseases related with aforementioned transporters highlight their significance; SLC6A19 mutations are associated with metabolic Hartnup disorder, whereas altered expression of SLC6A15 has been associated with a depression/stress-related disorders. Mutations of SLC6A18-SLCA20 cause iminoglycinuria and/or hyperglycinuria. SLC6A18-SLC6A20 to reach the cellular membrane require an ancillary unit ACE2 that is a molecular target for the spike protein of the SARS-CoV-2 virus. SLC6A19 has been proposed as a molecular target for the treatment of metabolic disorders resembling gastric surgery bypass. Inhibition of SLC6A15 appears to have a promising outcome in the treatment of psychiatric disorders. SLC6A19 and SLC6A20 have been suggested as potential targets in the treatment of COVID-19. In this review, we gathered recent advances on orphan transporters, their structure, functions, related disorders, and diseases, and in particular their relevance as therapeutic targets. SIGNIFICANCE STATEMENT: The following review systematizes current knowledge about the SLC6A15-SLCA20 neutral amino acid transporter subfamily and their therapeutic relevance in the treatment of different diseases.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Aminoácidos Neutros , COVID-19 , Humanos , Sistemas de Transporte de Aminoácidos Neutros/química , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Riñón/metabolismo , Aminoácidos/metabolismo , Aminoácidos Neutros/metabolismo , COVID-19/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
4.
Cell ; 186(9): 1950-1967.e25, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36996814

RESUMEN

Little is known about the critical metabolic changes that neural cells have to undergo during development and how temporary shifts in this program can influence brain circuitries and behavior. Inspired by the discovery that mutations in SLC7A5, a transporter of metabolically essential large neutral amino acids (LNAAs), lead to autism, we employed metabolomic profiling to study the metabolic states of the cerebral cortex across different developmental stages. We found that the forebrain undergoes significant metabolic remodeling throughout development, with certain groups of metabolites showing stage-specific changes, but what are the consequences of perturbing this metabolic program? By manipulating Slc7a5 expression in neural cells, we found that the metabolism of LNAAs and lipids are interconnected in the cortex. Deletion of Slc7a5 in neurons affects the postnatal metabolic state, leading to a shift in lipid metabolism. Additionally, it causes stage- and cell-type-specific alterations in neuronal activity patterns, resulting in a long-term circuit dysfunction.


Asunto(s)
Aminoácidos Neutros , Transportador de Aminoácidos Neutros Grandes 1 , Femenino , Humanos , Embarazo , Aminoácidos Neutros/genética , Aminoácidos Neutros/metabolismo , Encéfalo/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Mutación , Neuronas/metabolismo , Animales , Ratones
5.
Life Sci ; 316: 121402, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36669678

RESUMEN

AIMS: Despite its high concentration in pancreatic islets of Langerhans and broad range of antihyperglycemic effects, the route facilitating the import of dietary taurine into pancreatic ß-cell and mechanisms underlying its insulinotropic activity are unclear. We therefore studied the impact of taurine on beta-cell function, alongside that of other small neutral amino acids, L-alanine and L-proline. MAIN METHODS: Pharmacological profiling of insulin secretion was conducted using clonal BRIN BD11 ß-cells, the impact of taurine on the metabolic fate of glucose carbons was assessed using NMR and the findings were verified by real-time imaging of Ca2+ dynamics in the cytosol of primary mouse and human islet beta-cells. KEY FINDINGS: In our hands, taurine, alanine and proline induced secretory responses that were dependent on the plasma membrane depolarisation, import of Ca2+, homeostasis of K+ and Na+ as well as on cell glycolytic and oxidative metabolism. Taurine shifted the balance between the oxidation and anaplerosis towards the latter, in BRIN BD11 beta-cells. Furthermore, the amino acid signalling was significantly attenuated by inhibition of Na+-K+-Cl- symporter (NKCC). SIGNIFICANCE: These data suggest that taurine, like L-alanine and L-proline, acutely induces glucose-dependent insulin-secretory responses by modulating electrogenic Na+ transport, with potential role of intracellular K+ and Cl- in the signal transduction. The acute action delineated would be consistent with antidiabetic potential of dietary taurine supplementation.


Asunto(s)
Aminoácidos Neutros , Islotes Pancreáticos , Ratones , Animales , Humanos , Insulina/metabolismo , Taurina/farmacología , Taurina/metabolismo , Aminoácidos Neutros/metabolismo , Aminoácidos Neutros/farmacología , Línea Celular , Islotes Pancreáticos/metabolismo , Alanina/farmacología , Alanina/metabolismo , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Prolina/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34848541

RESUMEN

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.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/fisiología , Especificidad por Sustrato/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos/fisiología , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos/metabolismo , Aminoácidos Neutros/metabolismo , Transporte Biológico/fisiología , Microscopía por Crioelectrón/métodos , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Células HeLa , Humanos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Dominios Proteicos , Relación Estructura-Actividad
7.
Front Immunol ; 12: 641563, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33841424

RESUMEN

At present, the central role played by arginine in the modulation of the inflammatory cellular responses is well-recognized, and many pro-inflammatory stimuli are known to modulate the expression and activity of its transmembrane transporters. In this regard, we have addressed the effects of bacterial flagellin from Pseudomonas aeruginosa (FLA-PA) on the uptake of the amino acid in human epithelial respiratory cells. Among the arginine transporters, only ATB0,+, y+L, and y+ were operative in bronchial epithelial Calu-3 cells under control conditions; however, only the expression and activity of ATB0,+ were stimulated upon incubation with flagellin, whereas those of systems y+L and y+ were not stimulated. As a result, this induction, in turn, led to an increase in the intracellular content of arginine without making any change to its metabolic pathway. In addition, flagellin upregulated the amount of other amino acids substrates of ATB0,+, in particular, all the essential amino acids, such as valine, isoleucine, and leucine, along with the non-essential glutamine. At the molecular level, these effects were directly referable to the stimulation of a toll-like receptor-5 (TLR5) signaling pathway and to the induction of nuclear factor-κB (NF-κB) transcription factor. An induction of ATB0,+ expression has been observed also in EpiAirway™, a model of primary human normal tracheal-bronchial epithelial cells that mimics the in vitro pseudostratified columnar epithelium of the airways. In this tissue model, the incubation with flagellin is associated with the upregulation of messenger RNAs (mRNAs) for the chemokine IL-8 and for the cytokines IL-6 and interleukin-1ß (IL-1ß); as for the latter, a marked secretion in the extracellular medium was also observed due to the concomitant activation of caspase-1. The overall findings indicate that, in human respiratory epithelium, flagellin promotes cellular responses associating the increase of intracellular amino acids through ATB0,+ with the activation of the inflammasome. Given the role of the ATB0,+ transporter as a delivery system for bronchodilators in human airway epithelial cells, its induction under inflammatory conditions gains particular relevance in the field of respiratory pharmacology.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/inmunología , Arginina/metabolismo , Flagelina/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Sistema de Transporte de Aminoácidos ASC/metabolismo , Aminoácidos Neutros/metabolismo , Antígenos Bacterianos/inmunología , Células Cultivadas , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Pseudomonas aeruginosa
8.
Biochem Biophys Res Commun ; 552: 170-175, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33751934

RESUMEN

Biobased production of 5-aminovalerate (5AVA) from biomass can support a sustainable and economic biorefinery process to produce bio-based nylon 5 for food packaging materials. Cost-competitive production of 5AVA from biomass is a key factor in the successful commercialization of nylon 5. Bioproduction of 5AVA is a promising candidate for the industrial process to the current petrochemical route. In this study, we developed an artificial 2-keto-6-aminocaproate-mediated pathway for cost-competitive and high efficiency production of 5AVA in engineered Escherichia coli. Firstly, the combination of native l-lysine α-oxidase (RaiP) from Scomber japonicas, α-ketoacid decarboxylase (KivD) from Lactococcus lactis and aldehyde dehydrogenase (PadA) from Escherichia coli could efficiently convert l-lysine into 5AVA. Moreover, the engineered strains ML03-PnirB-RKP, ML03-PPL-PR-RKP, ML03-PM1-93-RKP induced by anaerobic condition, temperature-induced, constitutive expression instead of expensive isopropyl ß-D-thiogalactoside were constructed, respectively. The use of nirB promoter induced by anaerobic condition not only could attain a higher titer of 5AVA than PL-PR and M1-93 promoters, but omit cost of expensive exogenous inducers. After the replacement of industrial materials, 5AVA titer successfully reached 33.68 g/L in engineered strain ML03-PnirB-RKP via biotransformation. This biotransformation process conduces to the cosmically industrial 5AVA bioproduction.


Asunto(s)
Aminoácidos Neutros/metabolismo , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Ingeniería Metabólica/métodos , Nitrito Reductasas/genética , Regiones Promotoras Genéticas/genética , Anaerobiosis , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Microbiología Industrial/métodos , Nitrito Reductasas/metabolismo , Reproducibilidad de los Resultados
9.
Int J Mol Sci ; 21(5)2020 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-32164327

RESUMEN

In cultured human fibroblasts, SNAT transporters (System A) account for the accumulation of non-essential neutral amino acids, are adaptively up-regulated upon amino acid deprivation and play a major role in cell volume recovery upon hypertonic stress. No information is instead available on the expression and activity of SNAT transporters in human bone marrow mesenchymal stromal cells (MSC), although they are increasingly investigated for their staminal and immunomodulatory properties and used for several therapeutic applications. The uptake of glutamine and proline, two substrates of SNAT1 and SNAT2 transporters, was measured in primary human MSC and an MSC line. The amino acid analogue MeAIB, a specific substrate of these carriers, has been used to selectively inhibit SNAT-dependent transport of glutamine and, through its sodium-dependent transport, as an indicator of SNAT1/2 activity. SNAT1/2 expression and localization were assessed with RT-PCR and confocal microscopy, respectively. Cell volume was assessed from urea distribution space. In all these experiments, primary human fibroblasts were used as the positive control for SNAT expression and activity. Compared with fibroblasts, MSC have a lower SNAT1 expression and hardly detectable membrane localization of both SNAT1 and SNAT2. Moreover, they exhibit no sodium-dependent MeAIB uptake or MeAIB-inhibitable glutamine transport, and exhibit a lower ability to accumulate glutamine and proline than fibroblasts. MSC exhibited an only marginal increase in MeAIB transport upon amino acid starvation and did not recover cell volume after hypertonic stress. In conclusion, the activity of SNAT transporters is low in human MSC. MSC adaptation to amino acid shortage is expected to rely on intracellular synthesis, given the absence of an effective up-regulation of the SNAT transporters.


Asunto(s)
Sistema de Transporte de Aminoácidos A/metabolismo , Aminoácidos Neutros/metabolismo , Células Madre Mesenquimatosas/citología , Sistema de Transporte de Aminoácidos A/genética , Técnicas de Cultivo de Célula/métodos , Membrana Celular/metabolismo , Células Cultivadas , Medios de Cultivo/química , Fibroblastos/citología , Fibroblastos/metabolismo , Glutamina/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Prolina/metabolismo , Transporte de Proteínas , beta-Alanina/análogos & derivados , beta-Alanina/metabolismo
10.
J Ind Microbiol Biotechnol ; 47(3): 311-318, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32140931

RESUMEN

Glutaric acid is an important organic acid applied widely in different fields. Most previous researches have focused on the production of glutaric acid in various strains using the 5-aminovaleric acid (AMV) or pentenoic acid synthesis pathways. We previously utilized a five-step reversed adipic acid degradation pathway (RADP) in Escherichia coli BL21 (DE3) to construct strain Bgl146. Herein, we found that malonyl-CoA was strictly limited in this strain, and increasing its abundance could improve glutaric acid production. We, therefore, constructed a malonic acid uptake pathway in E. coli using matB (malonic acid synthetase) and matC (malonic acid carrier protein) from Clover rhizobia. The titer of glutaric acid was improved by 2.1-fold and 1.45-fold, respectively, reaching 0.56 g/L and 4.35 g/L in shake flask and batch fermentation following addition of malonic acid. Finally, the highest titer of glutaric acid was 6.3 g/L in fed-batch fermentation at optimized fermentation conditions.


Asunto(s)
Escherichia coli/metabolismo , Glutaratos/metabolismo , Malonatos/metabolismo , Adipatos/metabolismo , Aminoácidos Neutros/metabolismo , Vías Biosintéticas , Escherichia coli/genética , Fermentación , Malonil Coenzima A/metabolismo
11.
Sci Rep ; 9(1): 17152, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31748639

RESUMEN

The transport of nutrients across the placenta involves trophoblast cell specific transporters modulated through the mammalian target of rapamycin (mTOR). The vasoactive intestinal peptide (VIP) has embryotrophic effects in mice and regulates human cytotrophoblast cell migration and invasion. Here we explored the effect of VIP on glucose and System A amino acid uptake by human trophoblast-derived cells (Swan 71 and BeWo cell lines). VIP activated D-glucose specific uptake in single cytotrophoblast cells in a concentration-dependent manner through PKA, MAPK, PI3K and mTOR signalling pathways. Glucose uptake was reduced in VIP-knocked down cytotrophoblast cells. Also, VIP stimulated System A amino acid uptake and the expression of GLUT1 glucose transporter and SNAT1 neutral amino acid transporter. VIP increased mTOR expression and mTOR/S6 phosphorylation whereas VIP silencing reduced mTOR mRNA and protein expression. Inhibition of mTOR signalling with rapamycin reduced the expression of endogenous VIP and of VIP-induced S6 phosphorylation. Our findings support a role of VIP in the transport of glucose and neutral amino acids in cytotrophoblast cells through mTOR-regulated pathways and they are instrumental for understanding the physiological regulation of nutrient sensing by endogenous VIP at the maternal-foetal interface.


Asunto(s)
Aminoácidos Neutros/metabolismo , Glucosa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Trofoblastos/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Transporte Biológico/fisiología , Línea Celular , Femenino , Humanos , Placenta/metabolismo , Embarazo , ARN Mensajero/metabolismo , Transducción de Señal/fisiología
12.
Scand J Clin Lab Invest ; 79(8): 595-600, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31657241

RESUMEN

Hypoxaemia is present in many critically ill patients, and may contribute to encephalopathy. Changes in the passage of large neutral amino acids (LNAAs) across the blood-brain barrier (BBB) with an increased cerebral influx of aromatic amino acids into the brain may concurrently be present and also contribute to encephalopathy, but it has not been established whether hypoxaemia per se may trigger such changes. We measured cerebral blood flow (CBF) in 11 healthy men using the Kety-Schmidt technique and obtained paired arterial and jugular-venous blood samples for the determination of LNAAs by high performance liquid chromatography at baseline and after 9 hours of poikilocapnic normobaric hypoxia (12% O2). Transcerebral net exchange was determined by the Fick principle, and transport of LNAAs across the BBB was determined mathematically. Hypoxia increased both the systemic and corresponding cerebral delivery of the aromatic amino acid phenylalanine, and the branched-chain amino acids leucine and isoleucine. Despite this, the transcerebral net exchange values and mathematically derived brain extracellular concentrations for all LNAAs were unaffected. In conclusion, the observed changes in circulating LNAAs triggered by hypoxaemia do not affect the transcerebral exchange kinetics of LNAAs to such an extent that their brain extracellular concentrations are affected.


Asunto(s)
Aminoácidos Neutros/metabolismo , Encéfalo/metabolismo , Hipoxia/metabolismo , Inhalación , Enfermedad Aguda , Adulto , Arterias/metabolismo , Transporte Biológico , Análisis de los Gases de la Sangre , Barrera Hematoencefálica/metabolismo , Circulación Cerebrovascular , Femenino , Humanos , Cinética , Masculino
13.
Biomaterials ; 221: 119399, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31421314

RESUMEN

Hernia repair outcomes have improved with more robust material options for surgeons and optimized surgical techniques. However, ventral hernia repairs remain challenging with an inherent risk of post-surgical adhesions in the peritoneal space which can occur regardless of interventional material or its surgical placement. Herein, amino acid-based poly(ester urea)s (PEUs) with varied amount of an allyl ether side chains were modified post polymerization modification with the zwitterionic sulfnate group (3-((3-((3-mercaptopropanoyl)oxy)propyl) dimethylammonio)propane-1-sulfonate) to promote anti-adhesive properties. These alloc-PEUs were processed using roll-to-roll fabrication methods to afford films that were amenable to surface functionalization via a zwitterion-thiol. Functional group availability on the surface was confirmed via fluorescence microscopy, x-ray photoelectron spectroscopy (XPS), and quartz crystal microbalance (QCM) measurements. Zwitterionic treated PEUs exhibited reduced fibrinogen adsorption in vitro when compared to unfunctionalized control polymer. A rat intrabdominal cecal abrasion adhesion model was used to assess the extent and tenacity of adhesion formation in the presence of the PEUs. The 10% alloc-PEU zwitterion functionalized material was found to reduce the extent and tenacity of adhesions when compared to adhesion controls and the unfunctionalized PEU controls.


Asunto(s)
Aminoácidos Neutros/metabolismo , Materiales Biocompatibles/química , Materiales Biocompatibles/uso terapéutico , Poliésteres/química , Poliésteres/uso terapéutico , Adherencias Tisulares/prevención & control , Urea/análogos & derivados , Animales , Femenino , Fibrinógeno/metabolismo , Herniorrafia/métodos , Tecnicas de Microbalanza del Cristal de Cuarzo , Ratas , Ratas Sprague-Dawley , Urea/uso terapéutico
14.
Enzyme Microb Technol ; 128: 72-78, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31186113

RESUMEN

Glutaric acid is an attractive C5 dicarboxylic acid with wide applications in the biochemical industry. Glutaric acid can be produced by fermentation and bioconversion, and several of its biosynthesis pathways have been well characterized, especially the simple pathway involving glutaric acid from l-lysine using 5-aminovaleric acid. We previously reported the production of glutaric acid using 5-aminovaleric acid and α-ketoglutaric acid by a whole-cell reaction, resulting in a high conversion yield. In this study, we sought to enhance the stability and reusability of this whole-cell system for realizing the efficient production of glutaric acid under harsh reaction conditions. To this end, various matrices were screened to immobilize Escherichia coli whole-cell overexpressing 4-aminobutyrate aminotransferase (GabT), succinate semi-aldehyde dehydrogenase (GabD), and NAD(P)H oxidase (NOX). We ultimately selected a PVA-PEG gel (LentiKats®) for cell entrapment, and several factors of the reaction were optimized. The optimal temperature and pH were 35 °C and 8.5, respectively. Treatment with Tween 80 as a surfactant, as well as additional NOX, was found to be effective. Under the optimized conditions, an immobilized cell retained 55% of its initial activity even after the eighth cycle, achieving 995.2 mM accumulated glutaric acid, whereas free cell lost most of their activity after only two cycles. This optimized whole-cell system can be used in the large-scale production of glutaric acid.


Asunto(s)
Aminoácidos Neutros/metabolismo , Células Inmovilizadas/metabolismo , Escherichia coli/metabolismo , Glutaratos/metabolismo , Biotransformación , Escherichia coli/enzimología , Geles , Concentración de Iones de Hidrógeno , Polietilenglicoles , Alcohol Polivinílico , Temperatura
15.
Toxins (Basel) ; 11(3)2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30893830

RESUMEN

In vitro experiments have demonstrated that camel foregut-fluid has the capacity to metabolize indospicine, a natural toxin which causes hepatotoxicosis, but such metabolism is in competition with absorption and outflow of indospicine from the different segments of the digestive system. Six young camels were fed Indigofera spicata (337 µg indospicine/kg BW/day) for 32 days, at which time three camels were euthanized. The remaining camels were monitored for a further 100 days after cessation of this indospicine diet. In a retrospective investigation, relative levels of indospicine foregut-metabolism products were examined by UHPLC-MS/MS in plasma, collected during both accumulation and depletion stages of this experiment. The metabolite 2-aminopimelamic acid could be detected at low levels in almost all plasma samples, whereas 2-aminopimelic acid could not be detected. In the euthanized camels, 2-aminopimelamic acid could be found in all tissues except muscle, whereas 2-aminopimelic acid was only found in the kidney, pancreas, and liver tissues. The clearance rate for these metabolites was considerably greater than for indospicine, which was still present in plasma of the remaining camels 100 days after cessation of Indigofera consumption.


Asunto(s)
Sistema Digestivo/metabolismo , Indigofera , Norleucina/análogos & derivados , Aminoácidos Neutros/sangre , Aminoácidos Neutros/metabolismo , Animales , Camelus , Contaminación de Alimentos , Norleucina/sangre , Norleucina/farmacocinética , Ácidos Pimélicos/sangre , Ácidos Pimélicos/metabolismo , Distribución Tisular
16.
Nat Commun ; 9(1): 5071, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30498244

RESUMEN

Lysine degradation has remained elusive in many organisms including Escherichia coli. Here we report catabolism of lysine to succinate in E. coli involving glutarate and L-2-hydroxyglutarate as intermediates. We show that CsiD acts as an α-ketoglutarate-dependent dioxygenase catalysing hydroxylation of glutarate to L-2-hydroxyglutarate. CsiD is found widespread in bacteria. We present crystal structures of CsiD in complex with glutarate, succinate, and the inhibitor N-oxalyl-glycine, demonstrating strong discrimination between the structurally related ligands. We show that L-2-hydroxyglutarate is converted to α-ketoglutarate by LhgO acting as a membrane-bound, ubiquinone-linked dehydrogenase. Lysine enters the pathway via 5-aminovalerate by the promiscuous enzymes GabT and GabD. We demonstrate that repression of the pathway by CsiR is relieved upon glutarate binding. In conclusion, lysine degradation provides an important link in central metabolism. Our results imply the gut microbiome as a potential source of glutarate and L-2-hydroxyglutarate associated with human diseases such as cancer and organic acidurias.


Asunto(s)
Glutaratos/metabolismo , Lisina/metabolismo , Aminoácidos Neutros/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Succionato-Semialdehído Deshidrogenasa/metabolismo
17.
Enzyme Microb Technol ; 118: 57-65, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30143200

RESUMEN

Glutaric acid is one of the promising C5 platform compounds in the biochemical industry. It can be produced chemically, through the ring-opening of butyrolactone followed by hydrolysis. Alternatively, glutaric acid can be produced via lysine degradation pathways by microorganisms. In microorganisms, the overexpression of enzymes involved in this pathway from E. coli and C. glutamicum has resulted in high accumulation of 5-aminovaleric acid. However, the conversion from 5-aminovaleric acid to glutaric acid has resulted in a relatively low conversion yield for unknown reasons. In this study, as a solution to improve the production of glutaric acid, we introduced gabTD genes from B. subtilis to E. coli for a whole cell biocatalytic approach. This approach enabled us to determine the effect of co-factors on reaction and to achieve a high conversion yield from 5-aminovaleric acid at the optimized reaction condition. Optimization of whole cell reaction by different plasmids, pH, temperature, substrate concentration, and cofactor concentration achieved full conversion with 100 mM of 5-aminovaleric acid to glutaric acid. Nicotinamide adenine dinucleotide phosphate (NAD(P)+) and α-ketoglutaric acid were found to be critical factors in the enhancement of conversion in selected conditions. Whole cell reaction with a higher concentration of substrates gave 141 mM of glutaric acid from 300 mM 5-aminovaleric acid, 150 mM α-ketoglutaric acid, and 60 mM NAD+ at 30 °C, with a pH of 8.5 within 24 h (47.1% and 94.2% of conversion based on 5-aminovaleric acid and α-ketoglutaric acid, respectively). The whole cell biocatalyst was recycled 5 times with the addition of substrates; this enabled the accumulation of extra glutaric acid.


Asunto(s)
4-Aminobutirato Transaminasa/metabolismo , Aminoácidos Neutros/metabolismo , Bacillus subtilis/enzimología , Escherichia coli/metabolismo , Glutaratos/metabolismo , Succionato-Semialdehído Deshidrogenasa/metabolismo , 4-Aminobutirato Transaminasa/genética , Bacillus subtilis/genética , Biocatálisis , Escherichia coli/genética , Succionato-Semialdehído Deshidrogenasa/genética
18.
JCI Insight ; 3(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30046012

RESUMEN

The neuropathological effects of phenylketonuria (PKU) stem from the inability of the body to metabolize excess phenylalanine (Phe), resulting in accumulation of Phe in the blood and brain. Since the kidney normally reabsorbs circulating amino acids with high efficiency, we hypothesized that preventing the renal uptake of Phe might provide a disposal pathway that could lower systemic Phe levels. SLC6A19 is a neutral amino acid transporter responsible for absorption of the majority of free Phe in the small intestine and reuptake of Phe by renal proximal tubule cells. Transgenic KO mice lacking SLC6A19 have elevated levels of Phe and other amino acids in their urine but are otherwise healthy. Here, we crossed the Pahenu2 mouse model of PKU with the Slc6a19-KO mouse. These mutant/KO mice exhibited abundant excretion of Phe in the urine and an approximately 70% decrease in plasma Phe levels. Importantly, brain Phe levels were decreased by 50%, and the levels of key neurotransmitters were increased in the mutant/KO mice. In addition, a deficit in spatial working memory and markers of neuropathology were corrected. Finally, treatment of Pahenu2 mice with Slc6a19 antisense oligonucleotides lowered Phe levels. The results suggest that inhibition of SLC6A19 may represent a novel approach for the treatment of PKU and related aminoacidopathies.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/análisis , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos Neutros/metabolismo , Transporte Biológico/efectos de los fármacos , Fenilcetonurias/terapia , Aminas , Sistemas de Transporte de Aminoácidos Neutros/genética , Aminoácidos Neutros/sangre , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Enfermedades Genéticas Congénitas/terapia , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Masculino , Memoria a Corto Plazo , Ratones , Ratones Noqueados , Morfolinos/farmacología , Oligonucleótidos/farmacología , Fenilalanina/sangre , Fenilalanina/metabolismo , Fenilcetonurias/patología , Reabsorción Renal/efectos de los fármacos
19.
J Biol Chem ; 293(26): 9945-9957, 2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29743237

RESUMEN

The isozymes of photosynthetic phosphoenolpyruvate carboxylase from C4 plants (PEPC-C4) play a critical role in their atmospheric CO2 assimilation and productivity. They are allosterically activated by phosphorylated trioses or hexoses, such as d-glucose 6-phosphate, and inhibited by l-malate or l-aspartate. Additionally, PEPC-C4 isozymes from grasses are activated by glycine, serine, or alanine, but the allosteric site for these compounds remains unknown. Here, we report a new crystal structure of the isozyme from Zea mays (ZmPEPC-C4) with glycine bound at the monomer-monomer interfaces of the two dimers of the tetramer, making interactions with residues of both monomers. This binding site is close to, but different from, the one proposed to bind glucose 6-phosphate. Docking experiments indicated that d/l-serine or d/l-alanine could also bind to this site, which does not exist in the PEPC-C4 isozyme from the eudicot plant Flaveria, mainly because of a lysyl residue at the equivalent position of Ser-100 in ZmPEPC-C4 Accordingly, the ZmPEPC-C4 S100K mutant is not activated by glycine, serine, or alanine. Amino acid sequence alignments showed that PEPC-C4 isozymes from the monocot family Poaceae have either serine or glycine at this position, whereas those from Cyperaceae and eudicot families have lysine. The size and charge of the residue equivalent to Ser-100 are not only crucial for the activation of PEPC-C4 isozymes by neutral amino acids but also affect their affinity for the substrate phosphoenolpyruvate and their allosteric regulation by glucose 6-phosphate and malate, accounting for the reported kinetic differences between PEPC-C4 isozymes from monocot and eudicot plants.


Asunto(s)
Sitio Alostérico , Aminoácidos Neutros/metabolismo , Fosfoenolpiruvato Carboxilasa/química , Fosfoenolpiruvato Carboxilasa/metabolismo , Serina/metabolismo , Zea mays/enzimología , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Modelos Moleculares , Conformación Proteica , Especificidad por Sustrato
20.
J Am Soc Nephrol ; 29(6): 1624-1635, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29610403

RESUMEN

Background Reabsorption of amino acids (AAs) across the renal proximal tubule is crucial for intracellular and whole organism AA homeostasis. Although the luminal transport step is well understood, with several diseases caused by dysregulation of this process, the basolateral transport step is not understood. In humans, only cationic aminoaciduria due to malfunction of the basolateral transporter y+LAT1/CD98hc (SLC7A7/SLC3A2), which mediates the export of cationic AAs, has been described. Thus, the physiologic roles of basolateral transporters of neutral AAs, such as the antiporter LAT2/CD98hc (SLC7A8/SLC3A2), a heterodimer that exports most neutral AAs, and the uniporter TAT1 (SLC16A10), which exports only aromatic AAs, remain unclear. Functional cooperation between TAT1 and LAT2/CD98hc has been suggested by in vitro studies but has not been evaluated in vivoMethods To study the functional relationship of TAT1 and LAT2/CD98hc in vivo, we generated a double-knockout mouse model lacking TAT1 and LAT2, the catalytic subunit of LAT2/CD98hc (dKO LAT2-TAT1 mice).Results Compared with mice lacking only TAT1 or LAT2, dKO LAT2-TAT1 mice lost larger amounts of aromatic and other neutral AAs in their urine due to a tubular reabsorption defect. Notably, dKO mice also displayed decreased tubular reabsorption of cationic AAs and increased expression of y+LAT1/CD98hc.Conclusions The LAT2/CD98hc and TAT1 transporters functionally cooperate in vivo, and y+LAT1/CD98hc may compensate for the loss of LAT2/CD98hc and TAT1, functioning as a neutral AA exporter at the expense of some urinary loss of cationic AAs. Cooperative and compensatory mechanisms of AA transporters may explain the lack of basolateral neutral aminoacidurias in humans.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Aminoácidos Neutros/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Reabsorción Renal , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos Neutros/orina , Animales , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Túbulos Renales/fisiología , Masculino , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA