Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 51
1.
Article En | MEDLINE | ID: mdl-33657457

Replacement of fishmeal as the major protein source in feeds is critical for continued growth and sustainability of the aquaculture industry. However, numerous studies have shown suboptimal fish growth performance and reduced protein retention efficiency when carnivorous fish species are fed low fishmeal-high plant protein feeds. A study was conducted using a commercial strain and a genetically improved strain of rainbow trout selected for improved performance when fed an all plant protein diet to identify physiological differences associated with growth performance in the selected trout strain. Fifty individuals per strain (average weight ~ 580 g) were force-fed a plant-protein blend with and without amino acid supplementation (lysine, methionine and threonine) at 0.5% body weight and sampled at intervals over 24 h. Samples from intestine and liver were analyzed for specific gene expression analysis related to amino acid transporters, digestive process control, protein degradation and amino acid metabolism. The results showed that expression levels of various intestinal amino acid transporters (SLC1A1, SLC7A9, SLC15A, SLC1A5 SLC6A19 and SLC36A1) were affected by strain, diet and time. Moreover, significant interactions were found regarding the temporal expression levels of cholecystokinin (CCK-L), Krüppel-like factor 15 (KLF15) and aspartate aminotransferase (GOT) transcripts in the examined tissues. The results provide evidence that improved growth and protein retention of the selected strain fed an all-plant protein diet is a result of nutritional adaptation and an overall change in physiological homeostatic control.


Amino Acid Transport Systems/biosynthesis , Amino Acids/pharmacology , Animal Feed , Fish Proteins/biosynthesis , Gene Expression Regulation/drug effects , Oncorhynchus mykiss/metabolism , Animals
2.
Protein Expr Purif ; 173: 105648, 2020 09.
Article En | MEDLINE | ID: mdl-32335303

The S-adenosylmethionine carrier (SAMC) is a membrane transport protein located on the inner membrane of mitochondria that catalyzes the import of S-adenosylmethionine (SAM) into the mitochondrial matrix. SAMC mutations can cause a series of mitochondrial defects, including those affecting RNA stability, protein modification, mitochondrial translation and biosynthesis. Here, we describe the expression, purification and oligomerization of SAMC. The SAMC genes from three species were cloned into a eukaryotic expression vector with a GFP tag, and confocal microscopy analysis showed that these SAMCs were localized to mitochondria. A BacMam expression system was used for the expression of D. rerio SAMC with a FLAG tag. A size-exclusion chromatography analysis showed that SAMC may form a hexamer. A negative-staining electron microscopy analysis showed that SAMC formed tiny uniform particles and also confirmed the oligomerization of SAMC.


Amino Acid Transport Systems , Gene Expression , Protein Multimerization , Zebrafish Proteins , Zebrafish/genetics , Amino Acid Transport Systems/biosynthesis , Amino Acid Transport Systems/chemistry , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/isolation & purification , Animals , Humans , Male , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Zebrafish/metabolism , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish Proteins/isolation & purification
3.
Int J Mol Sci ; 20(23)2019 Nov 22.
Article En | MEDLINE | ID: mdl-31766598

Watermelon fruit contains a high percentage of amino acid citrulline (Cit) and arginine (Arg). Cit and Arg accumulation in watermelon fruit are most likely mediated by both de novo synthesis from other amino acids within fruits and direct import from source tissues (leaves) through the phloem. The amino acid transporters involved in the import of Cit, Arg, and their precursors into developing fruits of watermelon have not been reported. In this study, we have compiled the list of putative amino acid transporters in watermelon and characterized transporters that are expressed in the early stage of fruit development. Using the yeast complementation study, we characterized ClAAP3 (Cla023187) and ClAAP6 (Cla023090) as functional amino acid transporters belonging to the family of amino acid permease (AAP) genes. The yeast growth and uptake assays of radiolabeled amino acid suggested that ClAAP3 and ClAAP6 can transport a broad spectrum of amino acids. Expression of translational fusion proteins with a GFP reporter in Nicotiana benthamiana leaves confirmed the ER- and plasma membrane-specific localization, suggesting the role of ClAAP proteins in the cellular import of amino acids. Based on the gene expression profiles and functional characterization, ClAAP3 and ClAAP6 are expected to play a major role in regulation of amino acid import into developing watermelon fruits.


Amino Acid Transport Systems/biosynthesis , Citrullus/metabolism , Fruit/metabolism , Plant Proteins/biosynthesis , Amino Acid Transport Systems/genetics , Arginine/genetics , Arginine/metabolism , Citrulline/genetics , Citrulline/metabolism , Citrullus/genetics , Fruit/genetics , Plant Proteins/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Substrate Specificity , Nicotiana/genetics , Nicotiana/metabolism
4.
Biochimie ; 154: 25-34, 2018 Nov.
Article En | MEDLINE | ID: mdl-30076902

The mitochondrial S-adenosylmethionine carrier (SAMC), encoded by the SLC25A26 gene, catalyzes the uptake of S-adenosylmethionine (SAM) from the cytosol into mitochondria in exchange for S-adenosylhomocysteine (SAH), produced inside the mitochondria. In the last years we have been functionally characterizing the promoter of SLC25A26 gene. In this study we show that a silencer activity is present in the region from -756 bp to -504 bp, which specifically binds a protein present in Caski cells nuclear extracts. By in silico analysis, EMSA, ChIP, overexpressing and silencing experiments this protein was identified as FOXD3 which acts as a repressor of SLC25A26 expression. Interestingly, the repressor activity of FOXD3 is completely abolished by treating Caski cells with folate via a mechanism that involves methylation of FOXD3 gene promoter. This finding could have important impact in cancer cells where SLC25A26 is downregulated. Finally, the DPE and INR putative sites were also identified.


Amino Acid Transport Systems/biosynthesis , Calcium-Binding Proteins/biosynthesis , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Mitochondrial Proteins/biosynthesis , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Amino Acid Transport Systems/genetics , Caco-2 Cells , Calcium-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , Hep G2 Cells , Humans , Mitochondrial Proteins/genetics , Neoplasm Proteins/genetics , Repressor Proteins/genetics
5.
Metab Eng ; 44: 273-283, 2017 11.
Article En | MEDLINE | ID: mdl-29111438

L-lysine and other amino acids are commonly produced through fermentation using strains of heterotrophic bacteria such as Corynebacterium glutamicum. Given the large amount of sugar this process consumes, direct photosynthetic production is intriguing alternative. In this study, we report the development of a cyanobacterium, Synechococcus sp. strain PCC 7002, capable of producing L-lysine with CO2 as the sole carbon-source. We found that heterologous expression of a lysine transporter was required to excrete lysine and avoid intracellular accumulation that correlated with poor fitness. Simultaneous expression of a feedback inhibition resistant aspartate kinase and lysine transporter were sufficient for high productivities, but this was also met with a decreased chlorophyll content and reduced growth rates. Increasing the reductant supply by using NH4+, a more reduced nitrogen source relative to NO3-, resulted in a two-fold increase in productivity directing 18% of fixed carbon to lysine. Given this advantage, we demonstrated lysine production from media formulated with a municipal wastewater treatment sidestream as a nutrient source for increased economic and environmental sustainability. Based on our results, we project that Synechococcus sp. strain PCC 7002 could produce lysine at areal productivities approaching that of sugar cane to lysine via fermentation using non-agricultural lands and low-cost feedstocks.


Amino Acid Transport Systems , Aspartate Kinase , Bacterial Proteins , Corynebacterium glutamicum/genetics , Photosynthesis , Synechococcus , Amino Acid Transport Systems/biosynthesis , Amino Acid Transport Systems/genetics , Aspartate Kinase/biosynthesis , Aspartate Kinase/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Corynebacterium glutamicum/metabolism , Lysine , Synechococcus/genetics , Synechococcus/metabolism
6.
Sci Rep ; 7(1): 1051, 2017 04 21.
Article En | MEDLINE | ID: mdl-28432336

The transmembrane DNA-binding protein CadC of E. coli, a representative of the ToxR-like receptor family, combines input and effector domains for signal sensing and transcriptional activation, respectively, in a single protein, thus representing one of the simplest signalling systems. At acidic pH in a lysine-rich environment, CadC activates the transcription of the cadBA operon through recruitment of the RNA polymerase (RNAP) to the two cadBA promoter sites, Cad1 and Cad2, which are directly bound by CadC. However, the molecular details for its interaction with DNA have remained elusive. Here, we present the crystal structure of the CadC DNA-binding domain (DBD) and show that it adopts a winged helix-turn-helix fold. The interaction with the cadBA promoter site Cad1 is studied by using nuclear magnetic resonance (NMR) spectroscopy, biophysical methods and functional assays and reveals a preference for AT-rich regions. By mutational analysis we identify amino acids within the CadC DBD that are crucial for DNA-binding and functional activity. Experimentally derived structural models of the CadC-DNA complex indicate that the CadC DBD employs mainly non-sequence-specific over a few specific contacts. Our data provide molecular insights into the CadC-DNA interaction and suggest how CadC dimerization may provide high-affinity binding to the Cad1 promoter.


DNA, Bacterial/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Gene Expression Regulation, Bacterial , Trans-Activators/chemistry , Trans-Activators/metabolism , Adenosine Triphosphatases/biosynthesis , Amino Acid Transport Systems/biosynthesis , Antiporters/biosynthesis , Crystallography, X-Ray , DNA Mutational Analysis , Escherichia coli Proteins/biosynthesis , Helix-Turn-Helix Motifs , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Models, Molecular , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Transcription, Genetic
7.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1422-1435, 2017 06.
Article En | MEDLINE | ID: mdl-28235644

The mitochondrial aspartate-glutamate carrier isoform 1 (AGC1) catalyzes a Ca2+-stimulated export of aspartate to the cytosol in exchange for glutamate, and is a key component of the malate-aspartate shuttle which transfers NADH reducing equivalents from the cytosol to mitochondria. By sustaining the complete glucose oxidation, AGC1 is thought to be important in providing energy for cells, in particular in the CNS and muscle where this protein is mainly expressed. Defects in the AGC1 gene cause AGC1 deficiency, an infantile encephalopathy with delayed myelination and reduced brain N-acetylaspartate (NAA) levels, the precursor of myelin synthesis in the CNS. Here, we show that undifferentiated Neuro2A cells with down-regulated AGC1 display a significant proliferation deficit associated with reduced mitochondrial respiration, and are unable to synthesize NAA properly. In the presence of high glutamine oxidation, cells with reduced AGC1 restore cell proliferation, although oxidative stress increases and NAA synthesis deficit persists. Our data suggest that the cellular energetic deficit due to AGC1 impairment is associated with inappropriate aspartate levels to support neuronal proliferation when glutamine is not used as metabolic substrate, and we propose that delayed myelination in AGC1 deficiency patients could be attributable, at least in part, to neuronal loss combined with lack of NAA synthesis occurring during the nervous system development.


Amino Acid Transport Systems/biosynthesis , Aspartic Acid/analogs & derivatives , Cell Proliferation , Down-Regulation , Mitochondrial Proteins/biosynthesis , Neurons/metabolism , Amino Acid Transport Systems, Acidic/deficiency , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Antiporters/deficiency , Antiporters/genetics , Antiporters/metabolism , Aspartic Acid/biosynthesis , Cell Line , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Hereditary Central Nervous System Demyelinating Diseases/pathology , Humans , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Neurons/pathology , Psychomotor Disorders/genetics , Psychomotor Disorders/metabolism , Psychomotor Disorders/pathology
8.
FEBS J ; 284(6): 967-984, 2017 03.
Article En | MEDLINE | ID: mdl-28118529

Cancer cells down-regulate different genes to give them a selective advantage in invasiveness and/or metastasis. The SLC25A26 gene encodes the mitochondrial carrier that catalyzes the import of S-adenosylmethionine (SAM) into the mitochondrial matrix, required for mitochondrial methylation processes, and is down-regulated in cervical cancer cells. In this study we show that SLC25A26 is down-regulated due to gene promoter hypermethylation, as a mechanism to promote cell survival and proliferation. Furthermore, overexpression of SLC25A26 in CaSki cells increases mitochondrial SAM availability and promotes hypermethylation of mitochondrial DNA, leading to decreased expression of key respiratory complex subunits, reduction of mitochondrial ATP and release of cytochrome c. In addition, increased SAM transport into mitochondria leads to impairment of the methionine cycle with accumulation of homocysteine at the expense of glutathione, which is strongly reduced. All these events concur to arrest the cell cycle in the S phase, induce apoptosis and enhance chemosensitivity of SAM carrier-overexpressing CaSki cells to cisplatin.


Amino Acid Transport Systems/biosynthesis , Calcium-Binding Proteins/biosynthesis , Cytochromes c/biosynthesis , DNA, Mitochondrial/genetics , Drug Resistance, Neoplasm/genetics , Uterine Cervical Neoplasms/genetics , Adenosine Triphosphate/metabolism , Amino Acid Transport Systems/genetics , Apoptosis/genetics , Calcium-Binding Proteins/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cisplatin/administration & dosage , Cytochromes c/genetics , DNA Methylation/genetics , DNA, Mitochondrial/metabolism , Female , Gene Expression Regulation, Neoplastic , Glutathione/metabolism , Humans , Methionine/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Promoter Regions, Genetic , S-Adenosylmethionine/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
9.
Int J Mol Sci ; 17(12)2016 Dec 01.
Article En | MEDLINE | ID: mdl-27916945

Transporter genes and cytokinins are key targets for crop improvement. These genes are active during the development of the seed and its establishment as a strong sink. However, during germination, the seed transitions to being a source for the developing root and shoot. To determine if the sucrose transporter (SUT), amino acid permease (AAP), Sugar Will Eventually be Exported Transporter (SWEET), cell wall invertase (CWINV), cytokinin biosynthesis (IPT), activation (LOG) and degradation (CKX) gene family members are involved in both the sink and source activities of seeds, we used RT-qPCR to determine the expression of multiple gene family members, and LC-MS/MS to ascertain endogenous cytokinin levels in germinating Pisum sativum L. We show that genes that are actively expressed when the seed is a strong sink during its development, are also expressed when the seed is in the reverse role of being an active source during germination and early seedling growth. Cytokinins were detected in the imbibing seeds and were actively biosynthesised during germination. We conclude that, when the above gene family members are targeted for seed yield improvement, a downstream effect on subsequent seed germination or seedling vigour must be taken into consideration.


Cytokinins/biosynthesis , Germination/genetics , Pisum sativum/genetics , Seeds/genetics , Amino Acid Transport Systems/biosynthesis , Amino Acid Transport Systems/genetics , Cell Wall/enzymology , Cytokinins/genetics , Gene Expression Regulation, Plant , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/genetics , Pisum sativum/growth & development , Plant Growth Regulators/genetics , Seedlings/genetics , Seeds/growth & development , Tandem Mass Spectrometry , beta-Fructofuranosidase/biosynthesis , beta-Fructofuranosidase/genetics
10.
Genome Biol Evol ; 8(3): 753-64, 2016 Feb 15.
Article En | MEDLINE | ID: mdl-26878871

Facilitating the evolution of new gene functions, gene duplication is a major mechanism driving evolutionary innovation. Gene family expansions relevant to host/symbiont interactions are increasingly being discovered in eukaryotes that host endosymbiotic microbes. Such discoveries entice speculation that gene duplication facilitates the evolution of novel, endosymbiotic relationships. Here, using a comparative transcriptomic approach combined with differential gene expression analysis, we investigate the importance of endosymbiosis in retention of amino acid transporter paralogs in aphid genomes. To pinpoint the timing of amino acid transporter duplications we inferred gene phylogenies for five aphid species and three outgroups. We found that while some duplications arose in the aphid common ancestor concurrent with endosymbiont acquisition, others predate aphid divergence from related insects without intracellular symbionts, and still others appeared during aphid diversification. Interestingly, several aphid-specific paralogs have conserved enriched expression in bacteriocytes, the insect cells that host primary symbionts. Conserved bacteriocyte enrichment suggests that the transporters were recruited to the aphid/endosymbiont interface in the aphid common ancestor, consistent with a role for gene duplication in facilitating the evolution of endosymbiosis in aphids. In contrast, the temporal variability of amino acid transporter duplication indicates that endosymbiosis is not the only trait driving selection for retention of amino acid transporter paralogs in sap-feeding insects. This study cautions against simplistic interpretations of the role of gene family expansion in the evolution of novel host/symbiont interactions by further highlighting that multiple complex factors maintain gene family paralogs in the genomes of eukaryotes that host endosymbiotic microbes.


Amino Acid Transport Systems/genetics , Aphids/genetics , Bacteria/genetics , Symbiosis/genetics , Amino Acid Transport Systems/biosynthesis , Animals , Aphids/microbiology , Evolution, Molecular , Gene Expression Regulation , Phylogeny , Transcriptome/genetics
11.
Oncogene ; 35(23): 3004-15, 2016 06 09.
Article En | MEDLINE | ID: mdl-26434594

Tumour cells can use strategies that make them resistant to nutrient deprivation to outcompete their neighbours. A key integrator of the cell's responses to starvation and other stresses is amino-acid-dependent mechanistic target of rapamycin complex 1 (mTORC1). Activation of mTORC1 on late endosomes and lysosomes is facilitated by amino-acid transporters within the solute-linked carrier 36 (SLC36) and SLC38 families. Here, we analyse the functions of SLC36 family member, SLC36A4, otherwise known as proton-assisted amino-acid transporter 4 (PAT4), in colorectal cancer. We show that independent of other major pathological factors, high PAT4 expression is associated with reduced relapse-free survival after colorectal cancer surgery. Consistent with this, PAT4 promotes HCT116 human colorectal cancer cell proliferation in culture and tumour growth in xenograft models. Inducible knockdown in HCT116 cells reveals that PAT4 regulates a form of mTORC1 with two distinct properties: first, it preferentially targets eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), and second, it is resistant to rapamycin treatment. Furthermore, in HCT116 cells two non-essential amino acids, glutamine and serine, which are often rapidly metabolised by tumour cells, regulate rapamycin-resistant mTORC1 in a PAT4-dependent manner. Overexpressed PAT4 is also able to promote rapamycin resistance in human embryonic kidney-293 cells. PAT4 is predominantly associated with the Golgi apparatus in a range of cell types, and in situ proximity ligation analysis shows that PAT4 interacts with both mTORC1 and its regulator Rab1A on the Golgi. These findings, together with other studies, suggest that differentially localised intracellular amino-acid transporters contribute to the activation of alternate forms of mTORC1. Furthermore, our data predict that colorectal cancer cells with high PAT4 expression will be more resistant to depletion of serine and glutamine, allowing them to survive and outgrow neighbouring normal and tumorigenic cells, and potentially providing a new route for pharmacological intervention.


Amino Acid Transport Systems/metabolism , Amino Acids/metabolism , Colorectal Neoplasms/metabolism , Golgi Apparatus/metabolism , Multiprotein Complexes/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Amino Acid Transport Systems/biosynthesis , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm , Female , HCT116 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Signal Transduction , Treatment Outcome
12.
PLoS One ; 10(12): e0144677, 2015.
Article En | MEDLINE | ID: mdl-26689777

In the fission yeast, two Tor isoforms, Tor1 and Tor2, oppositely regulate gene expression of amino acid permeases. To elucidate the transcriptional machinery for these regulations, here we have employed the cap analysis of gene expression (CAGE), a method of analyzing expression profiles and identifying transcriptional start sites (TSSs). The loss of Tor1 decreased, and Tor2 inhibition by its temperature sensitive mutation increased, mRNA expression of isp5+, per1+, put4+ and SPBPB2B2.01. In contrast, the loss of Tor1 increased, and Tor2 inhibition decreased, the expression of cat1+. These changes were confirmed by semi-quantitative RT-PCR. These opposite effects by the loss of Tor1 and Tor2 inhibition appeared to occur evenly across multiple TSSs for the respective genes. The motif discovery analysis based on the CAGE results identified the GATA motifs as a potential cis-regulatory element for Tor-mediated regulation. In the luciferase reporter assay, the loss of Tor1 reduced, and Tor2 inhibition and nitrogen depletion increased, the activity of isp5+ promoter as well as that of a GATAAG reporter. One of the GATAAG motifs in isp5+ promoter was critical for its transcriptional activity, and a GATA transcription factor Gaf1 was critical for the activities of isp5+ promoter and the GATAAG reporter. Furthermore, Tor2 inhibition and nitrogen depletion induced nuclear localization of Gaf1 from the cytosol and its dephosphorylation. These results suggest that Tor2 inhibition, which is known to be induced by nitrogen depletion, promotes nuclear localization of Gaf1, thereby inducing isp5+ transcription through Gaf1 binding to the GATAAG motif in its promoter. Since Gaf1 was also critical for transcription of per1+ and put4+, Tor-Gaf1 signaling may coordinate transcription of multiple amino acid permeases according to nutrient availability.


Amino Acid Transport Systems/biosynthesis , Cell Nucleus/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Transcription, Genetic/physiology , Amino Acid Transport Systems/genetics , Cell Nucleus/genetics , Phosphatidylinositol 3-Kinases/genetics , Protein Kinases/genetics , Response Elements/physiology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Trans-Activators/genetics
13.
PLoS Genet ; 11(10): e1005600, 2015 Oct.
Article En | MEDLINE | ID: mdl-26473603

The global transcriptional regulator, CodY, binds strongly to the regulatory region of the braB gene, which encodes a Bacillus subtilis branched-chain amino acid (BCAA) permease. However, under conditions that maximize CodY activity, braB expression was similar in wild-type and codY null mutant cells. Nonetheless, expression from the braB promoter was significantly elevated in cells containing partially active mutant versions of CodY or in wild-type cells under growth conditions leading to intermediate levels of CodY activity. This novel pattern of regulation was shown to be due to two opposing mechanisms, negative and positive, by which CodY affects braB expression. A strong CodY-binding site located downstream of the transcription start point conferred negative regulation by direct interaction with CodY. Additionally, sequences upstream and downstream of the promoter were required for repression by a second pleiotropic B. subtilis regulator, ScoC, whose own expression is repressed by CodY. ScoC-mediated repression of braB in codY null mutants cells was as efficient as direct, CodY-mediated repression in wild-type cells under conditions of high CodY activity. However, under conditions of reduced CodY activity, CodY-mediated repression was relieved to a greater extent than ScoC-mediated repression was increased, leading to elevated braB expression. We conclude that restricting increased expression of braB to conditions of moderate nutrient limitation is the raison d'être of the feed-forward regulatory loop formed by CodY and ScoC at the braB promoter. The increase in BraB expression only at intermediate activities of CodY may facilitate the uptake of BCAA when they are not in excess but prevent unneeded BraB synthesis when other BCAA transporters are active.


Amino Acid Transport Systems/genetics , Bacillus subtilis/genetics , Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , Promoter Regions, Genetic , Amino Acid Transport Systems/biosynthesis , Amino Acid Transport Systems/metabolism , Amino Acids, Branched-Chain/genetics , Bacillus subtilis/pathogenicity , Bacterial Proteins/metabolism , Binding Sites , Gene Expression Regulation, Bacterial , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Genet Mol Res ; 14(2): 5246-57, 2015 May 18.
Article En | MEDLINE | ID: mdl-26125719

In order to evaluate the effect of high-concentrate diet supplementation on milk protein content, six Holstein dairy cows were assigned into high-concentrate diet (HC) or low-concentrate diet (LC) groups (N = 3/group) for 50 days. With regard to milk protein, HC feeding significantly reduced the percentage of milk protein (P < 0.01), and milk protein yield also reduced. The milk somatic cell count numbers and N-acetyl-D-glucosaminidase activity was significantly higher (P < 0.01) in the HC group than in the LC group. A pre-column derivatization procedure of o-phthalaldehyde was used to analyze the milk amino acid profile, the contents of Asp, Gln, Ala, Ile, Leu, and Lys were significantly lower in milk (P < 0.05), but Arg and Phe were significantly higher (P < 0.05) in the HC group than in the LC group. The mRNA abundance for amino acid transporters SLC7A8, SLC7A10 (P < 0.05), SLC1A3 (P < 0.05), and SLC16A10 (P < 0.05) were decreased in the HC group. These data indicate that expression of amino acid transporters alters regulation of amino acid utilization and decreases milk quality in dairy cows.


Amino Acid Transport Systems/biosynthesis , Diet , Milk Proteins/biosynthesis , Milk , Amino Acid Transport Systems/genetics , Animal Feed , Animals , Cattle , Female , Gene Expression , Lactation/genetics , Milk Proteins/genetics
15.
Mol Neurobiol ; 52(3): 1440-1457, 2015 Dec.
Article En | MEDLINE | ID: mdl-25352445

Modulation of spinal reactive gliosis following peripheral nerve injury (PNI) is a promising strategy to restore synaptic homeostasis. Oxidized ATP (OxATP), a nonselective antagonist of purinergic P2X receptors, was found to recover a neuropathic behavior following PNI. We investigated the role of intraperitoneal (i.p.) OxATP treatment in restoring the expression of neuronal and glial markers in the mouse spinal cord after sciatic spared nerve injury (SNI). Using in vivo two-photon microscopy, we imaged Ca(2+) transients in neurons and astrocytes of the dorsal horn of spinal cord at rest and upon right hind paw electrical stimulation in sham, SNI, and OxATP-treated mice. Neuropathic behavior was investigated by von Frey and thermal plantar test. Glial [glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba1)] and GABAergic [vesicular GABA transporter (vGAT) and glutamic acid decarboxylase 65/76 (GAD65/67)] markers and glial [glutamate transporter (GLT1) and GLAST] and neuronal amino acid [EAAC1, vesicular glutamate transporter 1 (vGLUT1)] transporters have been evaluated. In SNI mice, we found (i) increased glial response, (ii) decreased glial amino acid transporters, and (iii) increased levels of neuronal amino acid transporters, and (iv) in vivo analysis of spinal neurons and astrocytes showed a persistent increase of Ca(2+) levels. OxATP administration reduced glial activation, modulated the expression of glial and neuronal glutamate/GABA transporters, restored neuronal and astrocytic Ca(2+) levels, and prevented neuropathic behavior. In vitro studies validated that OxATP (i) reduced levels of reactive oxygen species (ROS), (ii) reduced astrocytic proliferation, (iii) increase vGLUT expression. All together, these data support the correlation between reactive gliosis and perturbation of the spinal synaptic homeostasis and the role played by the purinergic system in modulating spinal plasticity following PNI.


Gliosis/prevention & control , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Peroneal Nerve/injuries , Receptors, Purinergic P2X/physiology , Spinal Cord/physiopathology , Tibial Nerve/injuries , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Amino Acid Transport Systems/biosynthesis , Amino Acid Transport Systems/genetics , Amino Acids/metabolism , Animals , Astrocytes/drug effects , Astrocytes/physiology , Biomarkers , Calcium/metabolism , Down-Regulation , Gliosis/physiopathology , Homeostasis , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuralgia/etiology , Neuralgia/physiopathology , Neuronal Plasticity/drug effects , Neurons/metabolism , Neurons/pathology , Pain Threshold/drug effects , Pain Threshold/physiology , Purinergic P2X Receptor Antagonists/pharmacology , Reactive Oxygen Species/metabolism , Spinal Cord/ultrastructure
16.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 10): 1362-7, 2014 Oct.
Article En | MEDLINE | ID: mdl-25286940

The prokaryotic lysine-specific permease (LysP) belongs to the amino acid-polyamine-organocation (APC) transporter superfamily. In the cell, members of this family are responsible for the uptake and recycling of nutrients, for the maintenance of a constant internal ion concentration and for cell volume regulation. The detailed mechanism of substrate selectivity and transport of L-lysine by LysP is not understood. A high-resolution crystal structure would enormously facilitate such an understanding. To this end, LysP from Pseudomonas aeruginosa was recombinantly expressed in Escherichia coli and purified to near homogeneity by immobilized metal ion-affinity chromatography (IMAC) and size-exclusion chromatography (SEC). Hexagonal- and rod-shaped crystals were obtained in the presence of L-lysine and the L-lysine analogue L-4-thialysine by vapour diffusion and diffracted to 7.5 Šresolution. The diffraction data were indexed in space group P21, with unit-cell parameters a = 169.53, b = 169.53, c = 290.13 Å, γ = 120°.


Amino Acid Transport Systems/chemistry , Bacterial Proteins/chemistry , Pseudomonas aeruginosa/enzymology , Amino Acid Sequence , Amino Acid Transport Systems/biosynthesis , Amino Acid Transport Systems/isolation & purification , Bacterial Proteins/biosynthesis , Bacterial Proteins/isolation & purification , Chromatography, Affinity , Chromatography, Gel , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Escherichia coli , Gene Expression , Molecular Sequence Data
17.
J Appl Physiol (1985) ; 116(11): 1353-64, 2014 Jun 01.
Article En | MEDLINE | ID: mdl-24699854

Increasing amino acid availability (via infusion or ingestion) at rest or postexercise enhances amino acid transport into human skeletal muscle. It is unknown whether alterations in amino acid availability, from ingesting different dietary proteins, can enhance amino acid transport rates and amino acid transporter (AAT) mRNA expression. We hypothesized that the prolonged hyperaminoacidemia from ingesting a blend of proteins with different digestion rates postexercise would enhance amino acid transport into muscle and AAT expression compared with the ingestion of a rapidly digested protein. In a double-blind, randomized clinical trial, we studied 16 young adults at rest and after acute resistance exercise coupled with postexercise (1 h) ingestion of either a (soy-dairy) protein blend or whey protein. Phenylalanine net balance and transport rate into skeletal muscle were measured using stable isotopic methods in combination with femoral arteriovenous blood sampling and muscle biopsies obtained at rest and 3 and 5 h postexercise. Phenylalanine transport into muscle and mRNA expression of select AATs [system L amino acid transporter 1/solute-linked carrier (SLC) 7A5, CD98/SLC3A2, system A amino acid transporter 2/SLC38A2, proton-assisted amino acid transporter 1/SLC36A1, cationic amino acid transporter 1/SLC7A1] increased to a similar extent in both groups (P < 0.05). However, the ingestion of the protein blend resulted in a prolonged and positive net phenylalanine balance during postexercise recovery compared with whey protein (P < 0.05). Postexercise myofibrillar protein synthesis increased similarly between groups. We conclude that, while both protein sources enhanced postexercise AAT expression, transport into muscle, and myofibrillar protein synthesis, postexercise ingestion of a protein blend results in a slightly prolonged net amino acid balance across the leg compared with whey protein.


Amino Acid Transport Systems/biosynthesis , Amino Acids/metabolism , Dietary Proteins/administration & dosage , Milk Proteins/administration & dosage , Muscle, Skeletal/physiology , Resistance Training/methods , Soybean Proteins/administration & dosage , Administration, Oral , Adult , Amino Acid Transport Systems/drug effects , Amino Acids/drug effects , Dietary Proteins/metabolism , Double-Blind Method , Eating/physiology , Female , Humans , Male , Muscle, Skeletal/drug effects , Soybean Proteins/pharmacokinetics , Up-Regulation/drug effects , Up-Regulation/physiology , Whey Proteins , Young Adult
18.
Am J Physiol Endocrinol Metab ; 306(6): E668-80, 2014 Mar.
Article En | MEDLINE | ID: mdl-24425764

The oligopeptide transporter peptide cotransporter-1 Slc15a1 (PEPT1) plays a major role in the regulation of nitrogen supply, since it is responsible for 70% of the dietary nitrogen absorption. Previous studies demonstrated that PEPT1 expression and function in jejunum are reduced in diabetes and obesity, suggesting a nitrogen malabsorption from the diet. Surprisingly, we reported here a decrease in gut nitrogen excretion in high-fat diet (HFD)-fed mice and further investigated the mechanisms that could explain this apparent contradiction. Upon HFD, mice exhibited an increased concentration of free amino acids (AAs) in the portal vein (60%) along with a selective increase in the expression of two AA transporters (Slc6a20a, Slc36a1), pointing to a specific and adaptive absorption of some AAs. A delayed transit time (+40%) and an increased intestinal permeability (+80%) also contribute to the increase in nitrogen absorption. Besides, HFD mice exhibited a 2.2-fold decrease in fecal DNA resulting from a reduction in nitrogen catabolism from cell desquamation and/or in the intestinal microbiota. Indeed, major quantitative (2.5-fold reduction) and qualitative alterations of intestinal microbiota were observed in feces of HFD mice. Collectively, our results strongly suggest that both increased AA transporters, intestinal permeability and transit time, and changes in gut microbiota are involved in the increased circulating AA levels. Modifications in nitrogen homeostasis provide a new insight in HFD-induced obesity and glucose intolerance; however, whether these modifications are beneficial or detrimental for the HFD-associated metabolic complications remains an open issue.


Amino Acid Transport Systems/biosynthesis , Amino Acids/metabolism , Disease Models, Animal , Glucose Intolerance/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Obesity/metabolism , Symporters/biosynthesis , Allostasis , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Amino Acids/blood , Animals , DNA/analysis , Diet, High-Fat/adverse effects , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Feces/chemistry , Feces/microbiology , Gene Expression Regulation , Glucose Intolerance/etiology , Glucose Intolerance/microbiology , Glucose Intolerance/pathology , Gram-Negative Bacteria/growth & development , Gram-Negative Bacteria/isolation & purification , Gram-Positive Bacteria/growth & development , Gram-Positive Bacteria/isolation & purification , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestines/microbiology , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Nitrogen/analysis , Nitrogen/metabolism , Obesity/etiology , Obesity/microbiology , Obesity/pathology , Peptide Transporter 1 , Symporters/genetics , Symporters/metabolism
19.
Placenta ; 35(2): 132-8, 2014 Feb.
Article En | MEDLINE | ID: mdl-24360522

Previous work in our laboratory demonstrated that over-expression of human insulin-like growth factor-11 (hIGF-1) in the placenta corrects fetal weight deficits in mouse, rat, and rabbit models of intrauterine growth restriction without changes in placental weight. The underlying mechanisms of this effect have not been elucidated. To investigate the effect of intra-placental IGF-1 over-expression on placental function we examined amino acid transporter expression and localization in both a mouse model of placental Insufficiency (PI) and a model of human trophoblast, the BeWo Choriocarcinoma cell line. For in vitro human studies, BeWo Choriocarcinoma cells were maintained in F12 complete medium + 10%FBS. Cells were incubated in serum-free control media ± Ad-IGF-1 or Ad-LacZ for 48 h. MOIs of 10:1 and 100:1 were utilized. In BeWo, transfection efficiency was 100% at an MOI of 100:1 and Ad-IGF-1 significantly increased IGF-1 secretion, proliferation and invasion but reduced apoptosis compared to controls. In vitro, amino acid uptake was increased following Ad-IGF-1 treatment and associated with significantly increased RNA expression of SNAT1, 2, LAT1 and 4F2hc. Only SNAT2 protein expression was increased but LAT1 showed relocalization from a perinuclear location to the cytoplasm and cell membrane. For in vivo studies, timed-pregnant animals were divided into four groups on day 18; sham-operated controls, uterine artery branch ligation (UABL), UABL + Ad-hIGF-1 (10(8) PFU), UABL + Ad-LacZ (10(8) PFU). At gestational day 20, pups and placentas were harvested by C-section. Only LAT1 mRNA expression changed, showing that a reduced expression of the transporter levels in the PI model could be partially rectified with Ad-hIGF1 treatment. At the protein level, System L was reduced in PI but remained at control levels following Ad-hIGF1. The System A isoforms were differentially regulated with SNAT2 expression diminished but SNAT1 increased in PI and Ad-hIGF1 groups. Enhanced amino acid isoform transporter expression and relocalization to the membrane may be an important mechanism contributing to Ad-hIGF-1 mediated correction of placental insufficiency.


Amino Acid Transport Systems/biosynthesis , Insulin-Like Growth Factor I/biosynthesis , Placenta/metabolism , Placental Insufficiency/therapy , Amino Acids/metabolism , Animals , Cell Line, Tumor , Choriocarcinoma , Disease Models, Animal , Female , Genetic Therapy , Humans , Insulin-Like Growth Factor I/therapeutic use , Mice , Pregnancy , Transfection , Trophoblasts/metabolism
20.
Pancreatology ; 13(5): 475-85, 2013.
Article En | MEDLINE | ID: mdl-24075511

Pancreatic acinar cells accumulate amino acids against a marked concentration gradient to synthesize digestive enzymes. Thus, the function of acinar cells depends on amino acid uptake mediated by active transport. Despite the importance of this process, pancreatic amino acid transporter expression and cellular localization is still unclear. We screened mouse pancreas for the expression of genes encoding amino acid transporters. We showed that the most highly expressed transporters, namely sodium dependent SNAT3 (Slc38a3) and SNAT5 (Slc38a5) and sodium independent neutral amino acids transporters LAT1 (Slc7a5) and LAT2 (Slc7a8), are expressed in the basolateral membrane of acinar cells. SNAT3 and SNAT5, LAT1 and LAT2 are expressed in acinar cells. Additional evidence that these transporters are expressed in mature acinar cells was gained using acinar cell culture and acute pancreatitis models. In the acute phase of pancreatic injury, when acinar cell loss occurs, and in an acinar cell culture model, which mimics changes occurring during pancreatitis, SNAT3 and SNAT5 are strongly down-regulated. LAT1 and LAT2 were down-regulated only in the in vitro model. At protein level, SNAT3 and SNAT5 expression was also reduced during pancreatitis. Expression of other amino acid transporters was also modified in both models of pancreatitis. The subset of transporters with differential expression patterns during acute pancreatitis might be involved in the injury/regeneration phases. Further expression, localization and functional studies will follow to better understand changes occurring during acute pancreatitis. These findings provide insight into pancreatic amino acid transport in healthy pancreas and during acute pancreatitis injury.


Acinar Cells/metabolism , Amino Acid Transport Systems/biosynthesis , Pancreas/physiology , Pancreatitis/physiopathology , Acute Disease , Amino Acid Transport System y+/biosynthesis , Amino Acid Transport Systems, Neutral/biosynthesis , Animals , Cells, Cultured , Fusion Regulatory Protein 1, Light Chains/biosynthesis , Large Neutral Amino Acid-Transporter 1/biosynthesis , Male , Mice , Pancreas/physiopathology
...