Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Artículo en Zh | MEDLINE | ID: mdl-38677986

RESUMEN

Objective: To explore the mechanism of noise-induced hidden hearing loss by proteomics. Methods: In October 2022, 64 SPF male C57BL/6J mice were divided into control group and noise exposure group with 32 mice in each group according to random sampling method. The noise exposure group was exposed to 100 dB sound pressure level, 2000-16000 Hz broadband noise for 2 h, and the mouse hidden hearing loss model was established. Auditory brainstem response (ABR) was used to test the change of hearing threshold of mice on the 7th day after noise exposure, the damage of basal membrane hair cells was observed by immunofluorescence, and the differentially expressed proteins in the inner ear of mice in each group were identified and analyzed by 4D-Label-free quantitative proteomics, and verified by Western blotting. The results were statistically analyzed by ANOVA and t test. Results: On the 7th day after noise exposure, there was no significant difference in hearing threshold between the control group and the noise exposure group at click and 8000 Hz acoustic stimulation (P>0.05) . The hearing threshold in the noise exposure group was significantly higher than that in the control group under 16000 Hz acoustic stimulation (P<0.05) . Confocal immunofluorescence showed that the basal membrane hair cells of cochlear tissue in noise exposure group were arranged neatly, but the relative expression of C-terminal binding protein 2 antibody of presynaptic membrane in middle gyrus and basal gyrus was significantly lower than that in control group (P<0.05) . GO enrichment analysis showed that the functions of differentially expressed proteins were mainly concentrated in membrane potential regulation, ligand-gated channel activity, and ligand-gated ion channel activity. KEGG pathway enrichment analysis showed that differentially expressed proteins were significantly enriched in phosphatidylinositol 3 kinase-protein kinase B (PI3K-Akt) signaling pathway, NOD-like receptor signaling pathway, calcium signaling pathway, etc. Western blotting showed that the expression of inositol 1, 4, 5-trisphosphate receptor 3 (Itpr3) was increased and the expression of solute carrier family 38 member 2 (Slc38a2) was decreased in the noise exposure group (P<0.05) . Conclusion: Through proteomic analysis, screening and verification of the differential expression proteins Itpr3 and Slc38a2 in the constructed mouse noise-induced hidden hearing loss model, the glutaminergic synaptic related pathways represented by Itpr3 and Slc38a2 may be involved in the occurrence of hidden hearing loss.


Asunto(s)
Potenciales Evocados Auditivos del Tronco Encefálico , Pérdida Auditiva Provocada por Ruido , Ratones Endogámicos C57BL , Ruido , Proteómica , Animales , Ratones , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Provocada por Ruido/fisiopatología , Masculino , Ruido/efectos adversos , Modelos Animales de Enfermedad , Umbral Auditivo , Oído Interno/metabolismo , Pérdida de Audición Oculta
2.
Int J Mol Sci ; 23(21)2022 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-36361683

RESUMEN

More research is required to understand how melatonin protects neurons. The study aimed to find out if and how long non-coding RNA (lncRNA) contributes to melatonin's ability to defend the hippocampus from H2O2-induced oxidative injury. LncRNAs related to oxidative injury were predicted by bioinformatics methods. Mouse hippocampus-derived neuronal HT22 cells were treated with H2O2 with or without melatonin. Viability and apoptosis were detected by Cell Counting Kit-8 and Hoechst33258. RNA and protein levels were measured by quantitative real-time PCR, Western blot, and immunofluorescence. Bioinformatics predicted that 38 lncRNAs were associated with oxidative injury in mouse neurons. LncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) was related to H2O2-induced oxidative injury and up-regulated by melatonin in HT22 cells. The knockdown of NEAT1 exacerbated H2O2-induced oxidative injury, weakened the moderating effect of melatonin, and abolished the increasing effect of melatonin on the mRNA and protein level of Slc38a2. Taken together, melatonin attenuates H2O2-induced oxidative injury by upregulating lncRNA NEAT1, which is essential for melatonin stabilizing the mRNA and protein level of Slc38a2 for the survival of HT22 cells. The research may assist in the treatment of oxidative injury-induced hippocampal degeneration associated with aging using melatonin and its target lncRNA NEAT1.


Asunto(s)
Melatonina , MicroARNs , ARN Largo no Codificante , Ratones , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Melatonina/farmacología , Peróxido de Hidrógeno/toxicidad , Hipocampo/metabolismo , Apoptosis/genética , Estrés Oxidativo , ARN Mensajero/metabolismo , MicroARNs/genética
3.
Int J Mol Sci ; 24(1)2022 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-36613847

RESUMEN

Intrauterine growth restriction (IUGR), predominantly caused by placental insufficiency, affects partitioning of nutrients to the fetus. The system A sodium-coupled transporters (SNAT or SLC38), of types A1, A2, and A4, control non-essential amino acid uptake and supply. Here, we aimed to investigate the expression of these transporters across different placental disease cohorts and cells. To determine disease impact, transporter expressions at the gene (qPCR) and protein (western blots) level were assessed in gestationally matched placental tissues. Early (<34 weeks), and late (34−36 weeks) onset IUGR cases with/out preeclampsia were compared to preterm controls. We also investigated level of transporter expression in primary trophoblasts under glucose deprivation (n = 6) and hypoxia conditions (n = 7). SLC38A4 protein was significantly downregulated in early preterm pregnancies complicated with IUGR with/out preeclampsia. There were no differences in late preterm IUGR cohorts. Furthermore, we demonstrate for the first time in primary trophoblast cells, that gene expression of the transporters was sensitive to and induced by glucose starvation. SLC38A4 mRNA expression was also significantly upregulated in response to hypoxia. Thus, SLC38A4 expression was persistently low in early preterm IUGR pregnancies, regardless of disease aetiology. This suggests that gestational age at delivery, and consequently IUGR severity, may influence loss of its expression.


Asunto(s)
Placenta , Preeclampsia , Recién Nacido , Embarazo , Femenino , Humanos , Placenta/metabolismo , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Trofoblastos/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Sistema de Transporte de Aminoácidos A/genética , Sistema de Transporte de Aminoácidos A/metabolismo
4.
Exp Ther Med ; 27(5): 218, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38590564

RESUMEN

Adenomyosis is a benign uterine disorder that is associated with female infertility, a reduced clinical pregnancy rate and a high risk of miscarriage. Solute carrier family 38 member a2 (SLC38A2) is a glutamine (Gln) transporter that serves roles in various medical conditions. The present study aimed to reveal the role of SLC38A2 in adenomyosis. The mRNA expression levels of SLC38A2 in eutopic endometrial (EU) and ectopic endometrial (EC) tissues from adenomyotic patients were examined by reverse transcription-quantitative PCR. EU and EC cell proliferation and invasion were analyzed by Cell Counting Kit-8 and Transwell assays. Changes in the oxygen consumption rate (OCR) were determined to indicate the mitochondrial respiratory function and observed using a Seahorse analyzer. SLC38A2 expression in EC tissues was upregulated compared with that in normal endometrial tissues. SLC38A2 knockdown repressed EC cell proliferation and invasion. In addition, the Gln content and OCR were decreased in EC cells transfected with SLC38A2-knockdown lentivirus, whereas SLC38A2 overexpression had the opposite effect in EU cells. Furthermore, the increased proliferation and invasion rates and Gln level induced by SLC38A2 overexpression in EU cells were alleviated by CB-839, a glutaminase inhibitor. SLC38A2 overexpression promoted Gln metabolism and oxygen consumption rate, resulting in an increase in cell proliferation and invasion in the adenomyosis context. The present study indicated that reduction of SLC38A2 expression could be a novel target for adenomyosis therapy, and SLC38A2 may be a valuable clinical diagnostic molecule for adenomyosis.

5.
Cancer Biol Ther ; 25(1): 2315651, 2024 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-38390840

RESUMEN

Metabolic reprogramming plays a critical role in hepatocarcinogenesis. However, the mechanisms regulating metabolic reprogramming in primary liver cancer (PLC) are unknown. Differentially expressed miRNAs between PLC and normal tissues were identified using bioinformatic analysis. RT-qPCR was used to determine miR-10b-5p and SCL38A2 expression levels. IHC, WB, and TUNEL assays were used to assess the proliferation and apoptosis of the tissues. The proliferation, migration, invasion, and apoptosis of PLC cells were determined using the CCK-8 assay, Transwell assay, and flow cytometry. The interaction between miR-10b-5p and SLC38A2 was determined using dual-luciferase reporter assay. A PLC xenograft model in BALB/c nude mice was established, and tumorigenicity and SLC38A2 expression were estimated. Finally, liquid chromatography - mass spectrometry (LC-MS) untargeted metabolomics was used to analyze the metabolic profiles of xenograft PLC tissues in nude mice. miR-10b-5p was a key molecule in the regulation of PLC. Compared with para-carcinoma tissues, miR-10b-5p expression was increased in tumor tissues. miR-10b-5p facilitated proliferation, migration, and invasion of PLC cells. Mechanistically, miR-10b-5p targeted SLC38A2 to promote PLC tumor growth. Additionally, miR-10b-5p altered the metabolic features of PLC in vivo. Overexpression of miR-10b-5p resulted in remarkably higher amounts of lumichrome, folic acid, octanoylcarnitine, and Beta-Nicotinamide adenine dinucleotide, but lower levels of 2-methylpropanal, glycyl-leucine, and 2-hydroxycaproic acid. miR-10b-5p facilitates the metabolic reprogramming of PLC by targeting SLC38A2, which ultimately boosts the proliferation, migration, and invasion of PLC cells. Therefore, miR-10b-5p and SLC38A2 are potential targets for PLC diagnosis and treatment.


Asunto(s)
Neoplasias Hepáticas , MicroARNs , Animales , Ratones , Humanos , Ratones Desnudos , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Carcinogénesis , Proliferación Celular , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Apoptosis , Sistema de Transporte de Aminoácidos A/metabolismo
6.
Cancer Biomark ; 37(1): 39-52, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37005877

RESUMEN

OBJECTIVE: This study attempts to investigate whether hsa_circRNA_001859 (circ_001859) could regulate the proliferation and invasion of pancreatic cancer through the miR-21-5p/SLC38A2 pathway. METHODS: GSE79634 microarray was analyzed with R package. The expression of circ_001859 in pancreatic cancer tissues and cells was verified by qRT-PCR. After the overexpression of circ_001859, cell proliferation, cell migration and invasion were verified by colony formation and transwell assay. The targeting relationship between miR-21-5p and circ_001859 was predicted by TargetScan and was verified by dual luciferase reporter assay, RNA pull down and qRT-PCR. The effect of miR-21-5p on cell proliferation, migration and invasion were investigated by colony formation and transwell assay respectively. Similarly, the targeting relationship between miR-21-5p and SLC38A2 was predicted by TargetScan and was verified by dual luciferase reporter assay, western blot and qRT-PCR. The effect of SLC38A2 on cell proliferation was investigated by colony formation. RESULTS: Circ_001859 was lowly expressed in pancreatic cancer tissues and cells. In vitro assays showed that overexpression of circ_001859 could inhibit the proliferation, migration and invasion of pancreatic cancer. In addition, this effect was also confirmed in xenograft transplantation model. Circ_001859 could be bind to miR-21-5p and sponge its expression in pancreatic cancer cells. Overexpression of miR-21-5p enhanced the proliferation, migration and invasion ability of pancreatic cancer cells, while the inhibition of miR-21-5p expression suppressed these abilities. Moreover, miR-21-5p directly targeted at SLC38A2 and inhibited SLC38A2 expression levels while circ_001859 up-regulated SLC38A2 levels. SLC38A2 expression knockdown enhanced cell proliferation but SLC38A2 overexpression resulted in decreased proliferation, and effects of SLC38A2 could be rescued by miR-21-5p and circ_001859. In addition, both QRT-PCR and immunofluorescence confirmed that circ_001859 could regulate tumor epithelial-mesenchymal transition (EMT) through the miR-21-5p/SLC38A2 pathway. CONCLUSIONS: This study suggests that circ_001859 may inhibit the proliferation, invasion and EMT of pancreatic cancer through the miR-21-5p/SLC38A2 pathway.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Humanos , ARN Circular/genética , Transición Epitelial-Mesenquimal/genética , Neoplasias Pancreáticas/genética , Proliferación Celular/genética , MicroARNs/genética , Línea Celular Tumoral , Sistema de Transporte de Aminoácidos A , Neoplasias Pancreáticas
7.
Elife ; 122023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36722887

RESUMEN

Hyperosmolarity of the renal medulla is essential for urine concentration and water homeostasis. However, how renal medullary collecting duct (MCD) cells survive and function under harsh hyperosmotic stress remains unclear. Using RNA-Seq, we identified SLC38A2 as a novel osmoresponsive neutral amino acid transporter in MCD cells. Hyperosmotic stress-induced cell death in MCD cells occurred mainly via ferroptosis, and it was significantly attenuated by SLC38A2 overexpression but worsened by Slc38a2-gene deletion or silencing. Mechanistic studies revealed that the osmoprotective effect of SLC38A2 is dependent on the activation of mTORC1. Moreover, an in vivo study demonstrated that Slc38a2-knockout mice exhibited significantly increased medullary ferroptosis following water restriction. Collectively, these findings reveal that Slc38a2 is an important osmoresponsive gene in the renal medulla and provide novel insights into the critical role of SLC38A2 in protecting MCD cells from hyperosmolarity-induced ferroptosis via the mTORC1 signalling pathway.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Ferroptosis , Animales , Ratones , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Riñón/metabolismo , Médula Renal/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
8.
Cell Rep ; 42(7): 112749, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37405912

RESUMEN

Nutrient starvation drives yeast meiosis, whereas retinoic acid (RA) is required for mammalian meiosis through its germline target Stra8. Here, by using single-cell transcriptomic analysis of wild-type and Stra8-deficient juvenile mouse germ cells, our data show that the expression of nutrient transporter genes, including Slc7a5, Slc38a2, and Slc2a1, is downregulated in germ cells during meiotic initiation, and this process requires Stra8, which binds to these genes and induces their H3K27 deacetylation. Consequently, Stra8-deficient germ cells sustain glutamine and glucose uptake in response to RA and exhibit hyperactive mTORC1/protein kinase A (PKA) activities. Importantly, expression of Slc38a2, a glutamine importer, is negatively correlated with meiotic genes in the GTEx dataset, and Slc38a2 knockdown downregulates mTORC1/PKA activities and induces meiotic gene expression. Thus, our study indicates that RA via Stra8, a chordate morphogen pathway, induces meiosis partially by generating a conserved nutrient restriction signal in mammalian germ cells by downregulating their nutrient transporter expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Glutamina , Ratones , Animales , Glutamina/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Germinativas/metabolismo , Tretinoina/farmacología , Meiosis , Mamíferos/metabolismo
9.
Biomed Pharmacother ; 153: 113269, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35728354

RESUMEN

In a rotenone-induced Parkinson's disease (PD) rat model, behavioral investigation, pathological examination, inflammatory factor analysis, and mitochondrial structure and function investigation verified the anti-PD efficacy of nardosinone. A combined transcriptome and proteome analysis proposed that the anti-PD target of nardosinone is the slc38a2 gene and may involve the GABAergic synaptic pathway and cAMP-signaling pathway. Analysis of targeted slc38a2 knockout cells and expression of key enzyme-encoding genes in both pathways verified the target and pathways proposed by the 'omics analysis. This further confirms that nardosinone can regulate the slc38a2 gene, a potential new target for the treatment of Parkinson's disease, and plays an anti-PD role through the GABAergic synaptic and cAMP pathways.


Asunto(s)
Enfermedad de Parkinson Secundaria , Enfermedad de Parkinson , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Enfermedad de Parkinson Secundaria/inducido químicamente , Sesquiterpenos Policíclicos , Ratas , Rotenona/farmacología
10.
Elife ; 112022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35261338

RESUMEN

Cellular differentiation is associated with the acquisition of a unique protein signature that is essential to attain the ultimate cellular function and activity of the differentiated cell. This is predicted to result in unique biosynthetic demands that arise during differentiation. Using a bioinformatic approach, we discovered that osteoblast differentiation is associated with increased demand for the amino acid proline. When compared to other differentiated cells, osteoblast-associated proteins, including RUNX2, OSX, OCN, and COL1A1, are significantly enriched in proline. Using a genetic and metabolomic approach, we demonstrate that the neutral amino acid transporter SLC38A2 acts cell-autonomously to provide proline to facilitate the efficient synthesis of proline-rich osteoblast proteins. Genetic ablation of SLC38A2 in osteoblasts limits both osteoblast differentiation and bone formation in mice. Mechanistically, proline is primarily incorporated into nascent protein with little metabolism observed. Collectively, these data highlight a requirement for proline in fulfilling the unique biosynthetic requirements that arise during osteoblast differentiation and bone formation.


Bones have diverse roles in the body, such as supporting weight, allowing movement and protecting internal organs. Regardless of their location, all bones in the body are formed and maintained by specialized cells called osteoblasts. To produce the different components of bone, osteoblasts need a constant supply of amino acids, the building blocks of proteins. If these nutrients are limited, this may lead to weak and fragile bones that can fracture more easily. Naïve cells in the bone, which are yet to have a defined role, also require large amounts of amino acids to develop into fully functioning osteoblasts. Previous studies have found that specific amino acids (like glutamine and asparagine) are particularly important for forming the proteins in bone. However, it was unclear which amino acids are critical for the development of osteoblasts. To investigate, Shen et al. studied naïve cells that had been extracted from the embryos of mice and developed into osteoblasts in the laboratory. They found that the developing osteoblasts produced proteins enriched in proline, and that naïve cells required large amounts of this amino acid as they turned into osteoblasts. Genetic analysis revealed that osteoblasts carry the gene for a protein called SLC38A2, which has been shown to transport proline into other types of cells. Shen et al. then used gene editing tools to delete this transporter from the osteoblasts of mice. The mutated mice could not efficiently produce proline-rich proteins during embryonic development and formed less bone. These findings highlight that proline is important for developing osteoblasts and synthesizing the products of bone. Further research is needed, but it is possible that dietary supplements of proline may be beneficial for maintaining or promoting bone formation in adulthood. This could help individuals that have more fragile bones, such as the elderly or patients with bone diseases, like osteoporosis.


Asunto(s)
Osteogénesis , Prolina , Animales , Diferenciación Celular , Ratones , Osteoblastos/metabolismo , Prolina/metabolismo , Proteínas/metabolismo
11.
FASEB Bioadv ; 4(2): 138-152, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35141477

RESUMEN

Exercise is known to create a transient, but potent increase in skeletal muscle expression of potentially anti-inflammatory myokine interleukin-6 (IL-6). This effect may be clinically important in managing chronic inflammatory states. It has previously been proposed that lactic acidosis following exercise promotes this IL-6 up-regulation, but the mechanism of this acidosis effect is unknown. Rat skeletal muscle cell line L6-G8C5 has been used previously to model metabolic effects of acidosis, sensing low pH through the resulting inhibition of amino acid transporter SNAT2(SLC38A2). Use of ionophore ionomycin to model the rise in intracellular Ca2+ concentration occurring in contracting muscle strongly up-regulates IL-6 mRNA in L6-G8C5 myotubes. This study used this model to test the hypothesis that low extracellular pH (7.1) enhances ionomycin-induced IL-6 mRNA up-regulation by inhibiting SNAT2. Incubation of L6-G8C5 myotubes for 6 h with 0.5 µM ionomycin at control pH (7.4) resulted in a 15-fold increase in IL-6 mRNA which was further enhanced (1.74-fold) at pH 7.1. In contrast low pH had no significant effect on IL-6 mRNA without ionomycin, nor on the IL-6 mRNA increase that was induced by cyclic stretch. Even though pH 7.1 halved the transport activity of SNAT2, alternative methods of SNAT2 inhibition (JNK inhibitor SP600125; SNAT2 antagonist MeAIB; or SNAT2 silencing with siRNA) did not mimic the enhancing effect of low pH on IL-6 mRNA. On the contrary, JNK inhibition blunted the effect of pH 7.1 with ionomycin, but had no effect at pH 7.4. It is concluded that low pH promotes Ca2+/ionomycin-induced up-regulation of IL-6 mRNA through a novel SNAT2-independent JNK-dependent pH-sensing pathway not previously described in this skeletal muscle model.

12.
Animals (Basel) ; 12(23)2022 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-36496929

RESUMEN

In recent years, ensuring food security has been an important challenge for the world. It is important to make good use of China's domestic local feed resources to provide safe, stable, efficient, and high-quality rabbit meat products for China and the world. Lysine and methionine are the two most limiting essential amino acids in the rabbit diet. However, little is known about the rational composition of lysine and methionine in rabbit diets and the mechanisms that affect growth and development. Accordingly, in this study, we sought to address this knowledge gap by examining the effects of different compositions of lysine and methionine in rabbit diets. Subsequently, the growth status, nitrogen metabolism, blood biochemical indexes, muscle development, muscle quality, and the growth of satellite cells were evaluated in the animals. The results showed that diets containing 0.80% Lys and 0.40% Met improved average daily weight gain, feed conversion, nitrogen use efficiency, and muscle quality in the rabbits (p < 0.05). Additionally, it altered the amino acid transport potential in muscle by upregulating the expression of the SLC7A10 gene (p < 0.05). Meanwhile, the cell viability and the rate of division and migration of SCs in the 0.80% Lys/0.40 % Met composition group were increased (p < 0.05). SLC38A2 and P−mTOR protein expression was upregulated in the 0.80% lysine/0.40% methionine composition group (p < 0.05). In conclusion, 0.80% Lys/0.40% Met was the most suitable lysine and methionine composition in all tested diets. SLC38A2 acted as an amino acid sensor upstream of mTOR and was involved in the 0.80% Lys/0.40% Met regulation of muscle growth and development, thus implicating the mTOR signaling pathway in these processes.

13.
FASEB Bioadv ; 3(1): 36-48, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33490882

RESUMEN

Chronic metabolic acidosis plays a role in cachexia by enhancing total proteolysis in skeletal muscle. Glucocorticoid also triggers proteolysis and plays a permissive role in the effect of acidosis. The System A amino acid transporter SNAT2/SLC38A2 is ubiquitously expressed in mammalian cells including muscle, performing Na+-dependent active import of neutral amino acids, and is strongly inhibited by low pH. Exposure of rat skeletal muscle cell line L6-G8C5 to low pH rapidly inhibits SNAT2 transport activity and enhances total proteolysis rate. Pharmacological inhibition or silencing of SNAT2 also enhances proteolysis. This study tests the hypothesis that the glucocorticoid dexamethasone (DEX), like low pH, inhibits SNAT2 activity in L6-G8C5 myotubes, thus contributing to total proteolysis. Incubation with 500 nM DEX for 4 h reduced the System A amino acid transport rate to half the rate in control cultures. This inhibition depended on glucocorticoid receptor-mediated gene transcription, but SNAT2 mRNA levels were unaffected by DEX. In contrast, the SNAT2 protein assessed by immunoblotting was significantly depleted. The co-inhibitory effects of DEX and low pH on System A transport activity were additive in stimulating total proteolysis. In keeping with this mechanism, DEX's inhibitory effect on SNAT2 transport activity was significantly blunted by the proteasome inhibitor MG132. Proof of principle was achieved in similar experiments using recombinant expression of a GFP-tagged SNAT2 fusion protein in HEK293A cells. It is concluded that DEX acutely depletes the SNAT2 transporter protein, at least partly through proteasome-dependent degradation of this functionally important transporter.

14.
Front Mol Biosci ; 8: 646574, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33928121

RESUMEN

Amino acid transporters play a vital role in metabolism and nutrient signaling pathways. Typically, transport activity is investigated using single substrates and competing amounts of other amino acids. We used GC-MS and LC-MS for metabolic screening of Xenopus laevis oocytes expressing various human amino acid transporters incubated in complex media to establish their comprehensive substrate profiles. For most transporters, amino acid selectivity matched reported substrate profiles. However, we could not detect substantial accumulation of cationic amino acids by SNAT4 and ATB0,+ in contrast to previous reports. In addition, comparative substrate profiles of two related sodium neutral amino acid transporters known as SNAT1 and SNAT2, revealed the latter as a significant leucine accumulator. As a consequence, SNAT2, but not SNAT1, was shown to be an effective activator of the eukaryotic cellular growth regulator mTORC1. We propose, that metabolomic profiling of membrane transporters in Xe nopus laevis oocytes can be used to test their substrate specificity and role in intracellular signaling pathways.

15.
Mol Oncol ; 15(10): 2782-2800, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34003553

RESUMEN

Oncogenic KRAS mutations develop unique metabolic dependencies on nutrients to support tumor metabolism and cell proliferation. In particular, KRAS mutant cancer cells exploit amino acids (AAs) such as glutamine and leucine, to accelerate energy metabolism, redox balance through glutathione synthesis and macromolecule biosynthesis. However, the identities of the amino acid transporters (AATs) that are prominently upregulated in KRAS mutant cancer cells, and the mechanism regulating their expression have not yet been systematically investigated. Here, we report that the majority of the KRAS mutant colorectal cancer (CRC) cells upregulate selected AATs (SLC7A5/LAT1, SLC38A2/SNAT2, and SLC1A5/ASCT2), which correlates with enhanced uptake of AAs such as glutamine and leucine. Consistently, knockdown of oncogenic KRAS downregulated the expression of AATs, thereby decreasing the levels of amino acids taken up by CRC cells. Moreover, overexpression of mutant KRAS upregulated the expression of AATs (SLC7A5/LAT1, SLC38A2/SNAT2, and SLC1A5/ASCT2) in KRAS wild-type CRC cells and mouse embryonic fibroblasts. In addition, we show that the YAP1 (Yes-associated protein 1) transcriptional coactivator accounts for increased expression of AATs and mTOR activation in KRAS mutant CRC cells. Specific knockdown of AATs by shRNAs or pharmacological blockage of AATs effectively inhibited AA uptake, mTOR activation, and cell proliferation. Collectively, we conclude that oncogenic KRAS mutations enhance the expression of AATs via the hippo effector YAP1, leading to mTOR activation and CRC cell proliferation.


Asunto(s)
Neoplasias Colorrectales , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Aminoácidos/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Fibroblastos/metabolismo , Vía de Señalización Hippo , Humanos , Ratones , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Señalizadoras YAP
16.
Cell Chem Biol ; 27(6): 728-739.e9, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32386596

RESUMEN

With more than 450 members, the solute carrier (SLC) group of proteins represents the largest class of transporters encoded in the human genome. Their several-pass transmembrane domain structure and hydrophobicity contribute to the orphan status of many SLCs, devoid of known cargos or chemical inhibitors. We report that SLC proteins belonging to different families and subcellular compartments are amenable to induced degradation by heterobifunctional ligands. Engineering endogenous alleles via the degradation tag (dTAG) technology enabled chemical control of abundance of the transporter protein, SLC38A2. Moreover, we report the design of d9A-2, a chimeric compound engaging several members of the SLC9 family and leading to their degradation. d9A-2 impairs cellular pH homeostasis and promotes cell death in a range of cancer cell lines. These findings open the era of SLC-targeting chimeric degraders and demonstrate potential access of multi-pass transmembrane proteins of different subcellular localizations to the chemically exploitable degradation machinery.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Línea Celular , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ligandos , Masculino , Proteínas de Transporte de Membrana/química , Dominios Proteicos , Proteolisis
17.
Neuropharmacology ; 161: 107789, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31574264

RESUMEN

Amino acids are essential for cellular protein synthesis, growth, metabolism, signaling and in stress responses. Cell plasma membranes harbor specialized transporters accumulating amino acids to support a variety of cellular biochemical pathways. Several transporters for neutral amino acids have been characterized. However, Slc38a2 (also known as SA1, SAT2, ATA2, SNAT2) representing the classical transport system A activity stands in a unique position: Being a secondarily active transporter energized by the electrochemical gradient of Na+, it creates steep concentration gradients for amino acids such as glutamine: this may subsequently drive the accumulation of additional neutral amino acids through exchange via transport systems ASC and L. Slc38a2 is ubiquitously expressed, yet in a cell-specific manner. In this review, we show that Slc38a2 is regulated at the transcriptional and translational levels as well as by ions and proteins through direct interactions. We describe how Slc38a2 senses amino acid availability and passes this onto intracellular signaling pathways and how it regulates protein synthesis, cellular proliferation and apoptosis through the mechanistic (mammalian) target of rapamycin (mTOR) and general control nonderepressible 2 (GCN2) pathways. Furthermore, we review how this extensively regulated transporter contributes to cellular osmoadaptation and how it is regulated by endoplasmic reticulum stress and various hormonal stimuli to promote cellular metabolism, cellular signaling and cell survival. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Asunto(s)
Sistema de Transporte de Aminoácidos A/genética , Sistema de Transporte de Aminoácidos A/metabolismo , Aminoácidos/metabolismo , Transducción de Señal/genética , Animales , Regulación de la Expresión Génica , Humanos , Biosíntesis de Proteínas
18.
Front Pharmacol ; 9: 785, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30072900

RESUMEN

The glutamine transporter ASCT2 (SLC1A5) is actively investigated as an oncological target, but the field lacks efficient ASCT2 inhibitors. A new group of ASCT2 inhibitors, 2-amino-4-bis(aryloxybenzyl)aminobutanoic acids (AABA), were developed recently and shown to suppress tumor growth in preclinical in vivo models. To test its specificity, we deleted ASCT2 in two human cancer cell lines. Surprisingly, growth of parental and ASCT2-knockout cells was equally sensitive to AABA compounds. AABA compounds inhibited glutamine transport in cells lacking ASCT2, but not in parental cells. Deletion of ASCT2 and amino acid (AA) depletion induced expression of SNAT2 (SLC38A2), the activity of which was inhibited by AABA compounds. They also potently inhibited isoleucine uptake via LAT1 (SLC7A5), a transporter that is upregulated in cancer cells together with ASCT2. Inhibition of SNAT2 and LAT1 was confirmed by recombinant expression in Xenopus laevis oocytes. The reported reduction of tumor growth in pre-clinical models may be explained by a significant disruption of AA homeostasis.

19.
Front Endocrinol (Lausanne) ; 4: 199, 2013 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-24427154

RESUMEN

Intercellular communication is pivotal in optimizing and synchronizing cellular responses to keep homeostasis and to respond adequately to external stimuli. In the central nervous system (CNS), glutamatergic and GABAergic signals are postulated to be dependent on the glutamate/GABA-glutamine cycle for vesicular loading of neurotransmitters, for inactivating the signal and for the replenishment of the neurotransmitters. Islets of Langerhans release the hormones insulin and glucagon, but share similarities with CNS cells in for example transcriptional control of development and differentiation, and chromatin methylation. Interestingly, CNS proteins involved in secretion of the neurotransmitters and emitting their responses as well as the regulation of these processes, are also found in islet cells. Moreover, high levels of glutamate, GABA, and glutamine and their respective vesicular and plasma membrane transporters have been shown in the islet cells and there is emerging support for these amino acids and their transporters playing important roles in the maturation and secretion of insulin and glucagon. In this review, we will discuss the feasibility of recent data in the field in relation to the biophysical properties of the transporters (Slc1, Slc17, Slc32, and Slc38) and physiology of hormone secretion in islets of Langerhans.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA