Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
Biochem Biophys Res Commun ; 716: 150039, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38701556

ABSTRACT

The objective of this study was to better characterize the role of the glutamine transporter SLC38A1 in cervical cancer and explore the underlying mechanisms. Data from public databases and clinical cervical cancer tissue samples were used to assess the expression of SLC38A1 and its prognostic significance. Immunohistochemical staining, qRT-PCR, and Western blotting were used to evaluate the expression of relevant genes and proteins. Cell viability, cell cycle, apoptosis, and intracellular glutamine content were measured using CCK-8, flow cytometry, and biochemical assays. Additionally, the RNA immunoprecipitation (RIP) assay was used to examine the impact of METTL3/IGF2BP3 on the m6A modification of the SLC38A1 3'UTR. Both cervical cancer specimens and cells showed significantly increased expression of SLC38A1 and its expression correlated with an unfavorable prognosis. Knockdown of SLC38A1 inhibited cell viability and cell cycle progression, induced apoptosis, and suppressed tumor growth in vivo. Glutaminase-1 inhibitor CB-839 reversed the effects of SLC38A1 overexpression. METTL3 promoted m6A modification of SLC38A1 and enhanced its mRNA stability through IGF2BP3 recruitment. Moreover, METTL3 silencing inhibited cell viability, cell cycle progression, intracellular glutamine content, and induced apoptosis, but these effects were reversed by SLC38A1 overexpression. In conclusion, METTL3-mediated m6A methylation of SLC38A1 stimulates cervical cancer progression. SLC38A1 inhibition is a potential therapeutic strategy for cervical cancer.


Subject(s)
Adenosine , Methyltransferases , Uterine Cervical Neoplasms , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Humans , Female , Methyltransferases/metabolism , Methyltransferases/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/genetics , Methylation , Cell Line, Tumor , Cell Proliferation/genetics , Animals , Amino Acid Transport System A/metabolism , Amino Acid Transport System A/genetics , Apoptosis/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Mice , Prognosis , Cell Survival/genetics
2.
PLoS One ; 19(4): e0301356, 2024.
Article in English | MEDLINE | ID: mdl-38635778

ABSTRACT

BACKGROUND: CircTADA2A has been demonstrated to play critical roles in the occurrence and development of human cancer. However, the expression pattern and biological mechanisms of circTADA2A in melanoma remains largely unknown. METHODS: CircTADA2A were detected by quantitative real-time RT-PCR (qRT-PCR) and validated by Sanger sequencing. Function of circTADA2A and its protein partner in melanoma cells was investigated using RNA interference and overexpression assays. Interaction of circTADA2A, CCHC-type zinc finger nucleic acid binding protein (CNBP) and solute carrier family 38 member 1 (SLC38A1) was confirmed by RNA immunoprecipitation, RNA pull-down, and dual-luciferase reporter assay. The expression of genes and proteins were detected by qRT-PCR and western blot assays. RESULTS: Data from the investigation showed that a novel circRNA (circTADA2A, hsa_circ_0043278) was markedly downregulated in melanoma cells. Functionally, circTADA2A repressed cell proliferation, migration, invasion in melanoma cells. Mechanistically, circTADA2A interacted with CNBP, acting to suppress the binding of CNBP to the SLC38A1 promoter and subsequently restrained SLC38A1 transcription, which resulting in repression of melanoma progression. CONCLUSIONS: CircTADA2A suppresses melanoma progression by regulating CNBP/SLC38A1 axis, indicating a potential therapeutic target in melanoma.


Subject(s)
Melanoma , MicroRNAs , Humans , Melanoma/genetics , Melanoma/metabolism , RNA/genetics , RNA Interference , RNA, Circular/genetics , MicroRNAs/genetics , Cell Proliferation/genetics , Cell Movement , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism
3.
Cell Signal ; 117: 111110, 2024 05.
Article in English | MEDLINE | ID: mdl-38382691

ABSTRACT

Glutamine addiction is a significant hallmark of metabolic reprogramming in tumors and is crucial to the progression of cancer. Nevertheless, the regulatory mechanisms of glutamine metabolism in endometrial cancer (EC) remains elusive. In this research, we found that elevated expression of CENPA and solute carrier family 38 member 1 (SLC38A1) were firmly associated with worse clinical stage and unfavorable outcomes in EC patients. In addition, ectopic overexpression or silencing of CENPA could either enhance or diminish glutamine metabolism and tumor progression in EC. Mechanistically, CENPA directly regulated the transcriptional activity of the target gene, SLC38A1, leading to enhanced glutamine uptake and metabolism, thereby promoting EC progression. Notably, a prognostic model utilizing the expression levels of CENPA and SLC38A1 genes independently emerged as a prognostic factor for EC. More importantly, CENPA and SLC38A1 were significantly elevated and positively correlated, as well as indicative of poor prognosis in multiple cancers. In brief, our study confirmed that CENPA is a critical transcription factor involved in glutamine metabolism and tumor progression through modulating SLC38A1. This revelation suggests that targeting CENPA could be an appealing therapeutic approach to address pan-cancer glutamine addiction.


Subject(s)
Amino Acid Transport System A , Centromere Protein A , Endometrial Neoplasms , Glutamine , Female , Humans , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Glutamine/metabolism , Histones , Transcription Factors/metabolism , Centromere Protein A/metabolism
4.
Cancer Biol Ther ; 25(1): 2315651, 2024 12 31.
Article in English | MEDLINE | ID: mdl-38390840

ABSTRACT

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.


Subject(s)
Liver Neoplasms , MicroRNAs , Animals , Mice , Humans , Mice, Nude , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Carcinogenesis , Cell Proliferation , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Apoptosis , Amino Acid Transport System A/metabolism
5.
Nat Commun ; 14(1): 8158, 2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38071217

ABSTRACT

Insulin secretion from pancreatic ß cells is regulated by multiple stimuli, including nutrients, hormones, neuronal inputs, and local signalling. Amino acids modulate insulin secretion via amino acid transporters expressed on ß cells. The granin protein VGF has dual roles in ß cells: regulating secretory granule formation and functioning as a multiple peptide precursor. A VGF-derived peptide, neuroendocrine regulatory peptide-4 (NERP-4), increases Ca2+ influx in the pancreata of transgenic mice expressing apoaequorin, a Ca2+-induced bioluminescent protein complex. NERP-4 enhances glucose-stimulated insulin secretion from isolated human and mouse islets and ß-cell-derived MIN6-K8 cells. NERP-4 administration reverses the impairment of ß-cell maintenance and function in db/db mice by enhancing mitochondrial function and reducing metabolic stress. NERP-4 acts on sodium-coupled neutral amino acid transporter 2 (SNAT2), thereby increasing glutamine, alanine, and proline uptake into ß cells and stimulating insulin secretion. SNAT2 deletion and inhibition abolish the protective effects of NERP-4 on ß-cell maintenance. These findings demonstrate a novel autocrine mechanism of ß-cell maintenance and function that is mediated by the peptide-amino acid transporter axis.


Subject(s)
Amino Acid Transport System A , Insulin-Secreting Cells , Nerve Tissue Proteins , Animals , Humans , Mice , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Neurosecretory Systems/metabolism , Peptides/metabolism , Amino Acid Transport System A/metabolism
6.
J Cancer Res Clin Oncol ; 149(17): 15879-15898, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37673823

ABSTRACT

Although hepatocellular carcinoma (HCC) is rather frequent, little is known about the molecular pathways underlying its development, progression, and prognosis. In the current study, we comprehensively analyzed the deferentially expressed metabolism-related genes (MRGs) in HCC based on TCGA datasets attempting to discover the potentially prognostic genes in HCC. The up-regulated MRGs were further subjected to analyze their prognostic values and protein expressions. Twenty-seven genes were identified because their high expressions were significant in OS, PFS, DFS, DSS, and HCC tumor samples. They were then used for GO, KEGG, methylation, genetics changes, immune infiltration analyses. Moreover, we established a prognostic model in HCC using univariate assays and LASSO regression based on these MRGs. Additionally, we also found that SLC38A1, an amino acid metabolism closely related transporter, was a potential prognostic gene in HCC, and its function in HCC was further studied using experiments. We found that the knockdown of SLC38A1 notably suppressed the growth and migration of HCC cells. Further studies revealed that SLC38A1 modulated the development of HCC cells by regulating PI3K/AKT/mTOR signaling via glutamine mediated energy metabolism. In conclusion, this study identified the potentially prognostic MRGs in HCC and uncovered that SLC38A1 regulated HCC development and progression by regulating PI3K/AKT/mTOR signaling via glutamine mediated energy metabolism, which might provide a novel marker and potential therapeutic target in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Glutamine/metabolism , Liver Neoplasms/pathology , Cell Proliferation/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Energy Metabolism , Cell Line, Tumor , Amino Acid Transport System A/metabolism
7.
Nature ; 620(7972): 200-208, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37407815

ABSTRACT

Cancer cells evade T cell-mediated killing through tumour-immune interactions whose mechanisms are not well understood1,2. Dendritic cells (DCs), especially type-1 conventional DCs (cDC1s), mediate T cell priming and therapeutic efficacy against tumours3. DC functions are orchestrated by pattern recognition receptors3-5, although other signals involved remain incompletely defined. Nutrients are emerging mediators of adaptive immunity6-8, but whether nutrients affect DC function or communication between innate and adaptive immune cells is largely unresolved. Here we establish glutamine as an intercellular metabolic checkpoint that dictates tumour-cDC1 crosstalk and licenses cDC1 function in activating cytotoxic T cells. Intratumoral glutamine supplementation inhibits tumour growth by augmenting cDC1-mediated CD8+ T cell immunity, and overcomes therapeutic resistance to checkpoint blockade and T cell-mediated immunotherapies. Mechanistically, tumour cells and cDC1s compete for glutamine uptake via the transporter SLC38A2 to tune anti-tumour immunity. Nutrient screening and integrative analyses show that glutamine is the dominant amino acid in promoting cDC1 function. Further, glutamine signalling via FLCN impinges on TFEB function. Loss of FLCN in DCs selectively impairs cDC1 function in vivo in a TFEB-dependent manner and phenocopies SLC38A2 deficiency by eliminating the anti-tumour therapeutic effect of glutamine supplementation. Our findings establish glutamine-mediated intercellular metabolic crosstalk between tumour cells and cDC1s that underpins tumour immune evasion, and reveal glutamine acquisition and signalling in cDC1s as limiting events for DC activation and putative targets for cancer treatment.


Subject(s)
Amino Acid Transport System A , Dendritic Cells , Glutamine , Neoplasms , Signal Transduction , Amino Acid Transport System A/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glutamine/metabolism , Neoplasms/immunology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism
8.
BMC Gastroenterol ; 23(1): 74, 2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36918802

ABSTRACT

BACKGROUND: Solute carrier family 38 member 2 (SLC38A2) has previously been reported to participate in carcinogenesis. However, its expression and function in gastric cancer (GC) remain unclear. The present study aimed to investigate the role of SLC38A2 in GC. METHODS: The prognostic value and expression of SLC38A2 in GC was analyzed by combining bioinformatics and experimental analyses. Colony formation, Cell Counting Kit-8, wound healing, Transwell and tumor formation assays were performed to assess the biological function of SLC38A2. The cBioPortal, GeneMANIA and LinkedOmics databases were mined to determine the underlying regulatory mechanisms of SLC38A2. The role of SLC38A2 in tumor immune infiltration was explored using the TIMER database. RESULTS: Our results demonstrated that SLC38A2 was upregulated and was correlated with a poor prognosis in GC patients. SLC38A2 downregulation significantly inhibited the proliferation, invasion and migration of GC cells. Abnormal genetic alteration and epigenetic regulation may contribute to the upregulation of SLC38A2 expression levels in GC. The results of enrichment analysis demonstrated that SLC38A2 was associated with 'hippo signaling' and 'ubiquitinyl hydrolase activity'. The results also indicated that SLC38A2 may be a key factor in GC immune infiltration and M2 macrophage polarization. CONCLUSION: Overall, these data identified that SLC38A2 may serve as a potential prognostic biomarker and therapeutic target in GC.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Epigenesis, Genetic , Cell Movement/genetics , Cell Proliferation/genetics , Prognosis , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism
9.
Viruses ; 15(2)2023 01 24.
Article in English | MEDLINE | ID: mdl-36851539

ABSTRACT

Persistent high-risk human papillomavirus infection is the main risk factor for cervical cancer establishment, where the viral oncogenes E6 and E7 promote a cancerous phenotype. Metabolic reprogramming in cancer involves alterations in glutamine metabolism, also named glutaminolysis, to provide energy for supporting cancer processes including migration, proliferation, and production of reactive oxygen species, among others. The aim of this work was to analyze the effect of HPV16 E6 and E7 oncoproteins on the regulation of glutaminolysis and its contribution to cell proliferation. We found that the E6 and E7 oncoproteins exacerbate cell proliferation in a glutamine-dependent manner. Both oncoproteins increased the levels of transporter SNAT1, as well as GLS2 and GS enzymes; E6 also increased LAT1 transporter protein levels, while E7 increased ASCT2 and xCT. Some of these alterations are also regulated at a transcriptional level. Consistently, the amount of SNAT1 protein decreased in Ca Ski cells when E6 and E7 expression was knocked down. In addition, we demonstrated that cell proliferation was partially dependent on SNAT1 in the presence of glutamine. Interestingly, SNAT1 expression was higher in cervical cancer compared with normal cervical cells. The high expression of SNAT1 was associated with poor overall survival of cervical cancer patients. Our results indicate that HPV oncoproteins exacerbate glutaminolysis supporting the malignant phenotype.


Subject(s)
Glutamine , Uterine Cervical Neoplasms , Female , Humans , Cell Proliferation , Human papillomavirus 16/genetics , Papillomavirus E7 Proteins/genetics , Amino Acid Transport System A/metabolism
10.
Anticancer Drugs ; 33(9): 826-839, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36066402

ABSTRACT

Lung cancer is devastating cancer that ranks as the leading cause of cancer-related death. Long noncoding RNA (lncRNA) opioid growth factor receptor pseudogene 1 (OGFRP1) was recognized as an oncogene in many cancers. However, the molecular mechanism of OGFRP1 in lung cancer is still poorly understood. The expression of target RNAs and genes was detected by quantitative real-time PCR and western blot. The interaction between miR-299-3p and OGFRP1 or solute carrier family 38 member 1 (SLC38A1) was predicted by StarbaseV3.0 and verified by dual-luciferase reporter assay and Pearson's correlation coefficient. Besides, a transplantation model of human lung cancer in nude mice was established to evaluate the role of OGFRP1 in lung cancer. OGFRP1 and SLC38A1 were overexpressed, whereas miR-299-3p was lowly expressed in lung cancer tumors and cells. OGFRP1 knockdown suppressed cell proliferation and facilitated ferroptosis by promoting lipid peroxidation and iron accumulation in lung cancer. Besides, Furthermore, miR-299-3p inhibitor or SLC38A1 overexpression attenuated OGFRP1 depletion-induced suppression on cell proliferation and ferroptosis in lung cancer. Animal experiments indicated that OGFRP1 deficiency restrained tumor growth in vivo by regulating the miR-299-3p/SLC38A1 axis. OGFRP1 regulated cell proliferation and ferroptosis in lung cancer by inhibiting miR-299-3p to enhance SLC38A1 expression, providing a novel therapeutic strategy for lung cancer.


Subject(s)
Ferroptosis , Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Ferroptosis/genetics , Gene Expression Regulation, Neoplastic , Humans , Iron/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
11.
Cell Rep ; 40(3): 111092, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858571

ABSTRACT

The integrated stress response (ISR) plays a pivotal role in adaptation of translation machinery to cellular stress. Here, we demonstrate an ISR-independent osmoadaptation mechanism involving reprogramming of translation via coordinated but independent actions of mTOR and plasma membrane amino acid transporter SNAT2. This biphasic response entails reduced global protein synthesis and mTOR signaling followed by translation of SNAT2. Induction of SNAT2 leads to accumulation of amino acids and reactivation of mTOR and global protein synthesis, paralleled by partial reversal of the early-phase, stress-induced translatome. We propose SNAT2 functions as a molecular switch between inhibition of protein synthesis and establishment of an osmoadaptive translation program involving the formation of cytoplasmic condensates of SNAT2-regulated RNA-binding proteins DDX3X and FUS. In summary, we define key roles of SNAT2 in osmotolerance.


Subject(s)
Amino Acid Transport System A , Amino Acids , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Amino Acid Transport Systems/metabolism , Amino Acids/metabolism , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism
12.
Int J Mol Sci ; 24(1)2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36613847

ABSTRACT

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.


Subject(s)
Placenta , Pre-Eclampsia , Infant, Newborn , Pregnancy , Female , Humans , Placenta/metabolism , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Trophoblasts/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism
13.
Dig Dis Sci ; 67(7): 2948-2956, 2022 07.
Article in English | MEDLINE | ID: mdl-34173116

ABSTRACT

BACKGROUND: Glutamine (Gln) is essential for cancer progression, however, few studies have been conducted to investigate the roles of Gln transporters in gastric cancer stem cells (CSCs). AIMS: This work aims to explore the roles of Gln transporters in gastric cancer cell stemness. METHODS: We collected spheres formed by gastric cancer (GC) cells through a 3-dimensional (3D) semisolid culture system which has been shown to hold CSC-like traits. Lentivirus package was used to construct GC cells with SNAT2 overexpression. Analysis of sphere-formation, stemness marker expression, ALDH activity were used to detect the effects of Gln transporters on GC cell stemness. Determination of reactive oxygen species (ROS) and Gln consumption combined with the methods analyzing cell stemness were performed to explore the underlying mechanisms. RESULTS: Gln consumption was upregulated in GC spheres compared to the parental GC cells. The Gln transporter SNAT2 was highly expressed in GC spheres compared to that in the parental GC cells. SNAT2 overexpression significantly increased the Gln consumption in GC cells and increased the expression of stemness markers, sphere-formation ability and ALDH activity. Notably, SNAT2-mediated promoting effects on GC cell stemness were rescued by Gln deprivation. What's more, high expression of SNAT2 was associated with a poor GC patient survival through different online datasets. CONCLUSIONS: SNAT2 can promote the stemness of GC cells in a Gln-dependent manner.


Subject(s)
Amino Acid Transport System A , Stomach Neoplasms , Amino Acid Transport System A/metabolism , Cell Line, Tumor , Glutamine/metabolism , Humans , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
14.
Nutrients ; 13(11)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34836160

ABSTRACT

The influx of essential amino acids into skeletal muscle is primarily mediated by the large neutral amino acid transporter 1 (LAT1), which is dependent on the glutamine gradient generated by the sodium-dependent neutral amino acid transporter 2 (SNAT2). The protein expression and membrane localization of LAT1 may be influenced by amino acid ingestion and/or resistance exercise, although its acute influence on dietary amino acid incorporation into skeletal muscle protein has not been investigated. In a group design, healthy males consumed a mixed carbohydrate (0.75 g·kg-1) crystalline amino acid (0.25 g·kg-1) beverage enriched to 25% and 30% with LAT1 substrates L-[1-13C]leucine (LEU) and L-[ring-2H5]phenylalanine (PHE), respectively, at rest (FED: n = 7, 23 ± 5 y, 77 ± 4 kg) or after a bout of resistance exercise (EXFED: n = 7, 22 ± 2 y, 78 ± 11 kg). Postprandial muscle biopsies were collected at 0, 120, and 300 min to measure transporter protein expression (immunoblot), LAT1 membrane localization (immunofluorescence), and dietary amino acid incorporation into myofibrillar protein (ΔLEU and ΔPHE). Basal LAT1 and SNAT2 protein contents were correlated with each other (r = 0.55, p = 0.04) but their expression did not change across time in FED or EXFED (all, p > 0.05). Membrane localization of LAT1 did not change across time in FED or EXFED whether measured as outer 1.5 µm intensity or membrane-to-fiber ratio (all, p > 0.05). Basal SNAT2 protein expression was not correlated with ΔLEU or ΔPHE (all, p ≥ 0.05) whereas basal LAT1 expression was negatively correlated with ΔPHE in FED (r = -0.76, p = 0.04) and EXFED (r = -0.81, p = 0.03) but not ΔLEU (p > 0.05). Basal LAT1 membrane localization was not correlated with ΔLEU or ΔPHE (all, p > 0.05). Our results suggest that LAT1/SNAT2 protein expression and LAT1 membrane localization are not influenced by acute anabolic stimuli and do not positively influence the incorporation of dietary amino acids for de novo myofibrillar protein synthesis in healthy young males.


Subject(s)
Amino Acid Transport System A/metabolism , Amino Acids/administration & dosage , Amino Acids/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Muscle, Skeletal/metabolism , Resistance Training , Adult , Cell Membrane/metabolism , Diet , Humans , Leucine/metabolism , Male , Muscle Proteins/metabolism , Myofibrils/metabolism , Phenylalanine/metabolism , Postprandial Period , Young Adult
15.
Oncogene ; 40(46): 6443-6455, 2021 11.
Article in English | MEDLINE | ID: mdl-34608265

ABSTRACT

Metastasis is the leading cause of colorectal cancer (CRC)-induced death. However, the underlying molecular mechanisms of CRC metastasis are poorly understood. Metabolic reprogramming is an intrinsic feature of cancer, which have complicated effects on cancer metastasis. Here, we find that a novel metastasis-related protein, cell migration-inducing and hyaluronan-binding protein (CEMIP), can act as a novel adaptor protein of O-GlcNAc transferase (OGT) to promote CRC metastasis through glutamine metabolic reprogramming. Mechanistically, CEMIP interacts with OGT and ß-catenin, which leads to elevated O-GlcNAcylation of ß-catenin and enhanced ß-catenin nuclear translocation from cytomembrane. Furthermore, accumulated ß-catenin in nucleus enhances the transcription of CEMIP to reciprocally regulate ß-catenin and contributes to over-expression of glutaminase 1 and glutamine transporters (SLC1A5 and SLC38A2). Combinational inhibition of CEMIP and glutamine metabolism could dramatically attenuate the metastasis of CRC in vivo. Collectively, this study reveals the importance of glutamine metabolic reprogramming in CEMIP-induced CRC metastasis, indicating the great potential of CEMIP and glutamine metabolism for CRC metastasis prevention.


Subject(s)
Colorectal Neoplasms/pathology , Glutamine/metabolism , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/metabolism , N-Acetylglucosaminyltransferases/metabolism , beta Catenin/metabolism , Amino Acid Transport System A/metabolism , Amino Acid Transport System ASC/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice , Minor Histocompatibility Antigens/metabolism , Neoplasm Metastasis , Neoplasm Transplantation , Transcription, Genetic
16.
Nutrients ; 13(8)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34444747

ABSTRACT

Assisted reproductive technologies (ART) may increase risk for abnormal placental development, preterm delivery and low birthweight. We investigated placental morphology, transporter expression and paired maternal/umbilical fasting blood nutrient levels in human term pregnancies conceived naturally (n = 10) or by intracytoplasmic sperm injection (ICSI; n = 11). Maternal and umbilical vein blood from singleton term (>37 weeks) C-section pregnancies were assessed for levels of free amino acids, glucose, free fatty acids (FFA), cholesterol, high density lipoprotein (HDL), low density lipoprotein (LDL), very low-density lipoprotein (VLDL) and triglycerides. We quantified placental expression of GLUT1 (glucose), SNAT2 (amino acids), P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) (drug) transporters, and placental morphology and pathology. Following ICSI, placental SNAT2 protein expression was downregulated and umbilical cord blood levels of citrulline were increased, while FFA levels were decreased at term (p < 0.05). Placental proliferation and apoptotic rates were increased in ICSI placentae (p < 0.05). No changes in maternal blood nutrient levels, placental GLUT1, P-gp and BCRP expression, or placental histopathology were observed. In term pregnancies, ICSI impairs placental SNAT2 transporter expression and cell turnover, and alters umbilical vein levels of specific nutrients without changing placental morphology. These may represent mechanisms through which ICSI impacts pregnancy outcomes and programs disease risk trajectories in offspring across the life course.


Subject(s)
Fertilization , Fetal Blood/metabolism , Nutrients , Placenta/metabolism , Pregnancy Trimester, Third , Sperm Injections, Intracytoplasmic/adverse effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adult , Amino Acid Transport System A/metabolism , Apoptosis , Cell Proliferation , Female , Glucose Transporter Type 1/metabolism , Humans , Neoplasm Proteins/metabolism , Placenta/pathology , Pregnancy , Pregnancy Outcome , Premature Birth/etiology , Reproductive Techniques, Assisted/adverse effects , Sperm Injections, Intracytoplasmic/methods
17.
Nutrients ; 13(8)2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34445051

ABSTRACT

Intrauterine growth restriction (IUGR) is associated with reduced placental amino acid transport (AAT). However, it remains to be established if changes in AAT contribute to restricted fetal growth. We hypothesized that reduced in vivo placental AAT precedes the development of IUGR in baboons with maternal nutrient restriction (MNR). Baboons were fed either a control (ad libitum) or MNR diet (70% of control diet) from gestational day (GD) 30. At GD 140, in vivo transplacental AA transport was measured by infusing nine (13)C- or (2)H-labeled essential amino acids (EAAs) as a bolus into the maternal circulation at cesarean section. A fetal vein-to-maternal artery mole percent excess ratio for each EAA was measured. Microvillous plasma membrane (MVM) system A and system L transport activity were determined. Fetal and placental weights were not significantly different between MNR and control. In vivo, the fetal vein-to-maternal artery mole percent excess ratio was significantly decreased for tryptophan in MNR. MVM system A and system L activity was markedly reduced in MNR. Reduction of in vivo placental amino acid transport precedes fetal growth restriction in the non-human primate, suggesting that reduced placental amino acid transfer may contribute to IUGR.


Subject(s)
Amino Acid Transport System A/metabolism , Amino Acid Transport System L/metabolism , Amino Acids/metabolism , Diet, Protein-Restricted , Fetal Growth Retardation/etiology , Maternal-Fetal Exchange , Placenta/metabolism , Animals , Biological Transport , Disease Models, Animal , Female , Fetal Development , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/physiopathology , Gestational Age , Maternal Nutritional Physiological Phenomena , Papio , Pregnancy
18.
Br J Cancer ; 125(6): 865-876, 2021 09.
Article in English | MEDLINE | ID: mdl-34274945

ABSTRACT

BACKGROUND: Many molecular alterations are shared by embryonic liver development and hepatocellular carcinoma (HCC). Identifying the common molecular events would provide a novel prognostic biomarker and therapeutic target for HCC. METHODS: Expression levels and clinical relevancies of SLC38A4 and HMGCS2 were investigated by qRT-PCR, western blot, TCGA and GEO datasets. The biological roles of SLC38A4 were investigated by functional assays. The downstream signalling pathway of SLC38A4 was investigated by qRT-PCR, western blot, immunofluorescence, luciferase reporter assay, TCGA and GEO datasets. RESULTS: SLC38A4 silencing was identified as an oncofetal molecular event. DNA hypermethylation contributed to the downregulations of Slc38a4/SLC38A4 in the foetal liver and HCC. Low expression of SLC38A4 was associated with poor prognosis of HCC patients. Functional assays demonstrated that SLC38A4 depletion promoted HCC cellular proliferation, stemness and migration, and inhibited HCC cellular apoptosis in vitro, and further repressed HCC tumorigenesis in vivo. HMGCS2 was identified as a critical downstream target of SLC38A4. SLC38A4 increased HMGCS2 expression via upregulating AXIN1 and repressing Wnt/ß-catenin/MYC axis. Functional rescue assays showed that HMGCS2 overexpression reversed the oncogenic roles of SLC38A4 depletion in HCC. CONCLUSIONS: SLC38A4 downregulation was identified as a novel oncofetal event, and SLC38A4 was identified as a novel tumour suppressor in HCC.


Subject(s)
Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Carcinoma, Hepatocellular/pathology , Down-Regulation , Hydroxymethylglutaryl-CoA Synthase/metabolism , Liver Neoplasms/pathology , Liver/embryology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Neoplasm Transplantation , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , Wnt Signaling Pathway
19.
Mol Nutr Food Res ; 65(18): e2100301, 2021 09.
Article in English | MEDLINE | ID: mdl-34289236

ABSTRACT

INTRODUCTION: As an exogenous food contaminant, dietary oxidized lipid impairs growth and development, and triggers chronic diseases in humans or animals. This study explores the effects of soybean oil with different oxidative degree on the placental injury of gestational rats. METHODS AND RESULTS: Thirty-two female adult rats are randomly assigned to four groups. The control group is fed the purified diet with fresh soybean oil (FSO), and the treatment groups are fed purified diets with lipid content replaced by oxidized soybean oil (OSO) at 200, 400, and 800 mEqO2 kg-1 from conception until delivery. On day 20 of gestation, OSO decreased placental and embryonic weights as the oxidative degree increased linearly and quadratically. The expression of Bax showed a linear increase, and Bcl-2 decreased as the oxidative degree increased. The expression of Fosl1 and Esx1 is linearly and quadratically decreased in OSO-treated groups than FSO group. OSO decreased the level of IL-10 but increased expression of IL-1ß in placenta and plasma. OSO remarkably upregulates levels of Fatp1 and Glut1 and decreases expression of Snat2 and Glut3. CONCLUSION: OSO aggravates placental injury by modulating nutrient transporters and apoptosis-related genes, impedes placental growth and development, and ultimately leads to the decrease of fetal weight.


Subject(s)
Carrier Proteins/metabolism , Maternal Exposure/adverse effects , Placenta/drug effects , Soybean Oil/adverse effects , Soybean Oil/chemistry , Amino Acid Transport System A/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cytokines/blood , Cytokines/genetics , Fatty Acid Transport Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Female , Fetal Weight/drug effects , Gene Expression Regulation, Developmental/drug effects , Glucose Transporter Type 3/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/physiology , Placenta/metabolism , Placenta/pathology , Placentation/drug effects , Pregnancy , Rats, Sprague-Dawley
20.
Neurobiol Learn Mem ; 183: 107480, 2021 09.
Article in English | MEDLINE | ID: mdl-34153453

ABSTRACT

Perturbations in the glutamate-glutamine cycle and glutamate release from presynaptic terminals have been involved in the development of cognitive deficits in Alzheimer's disease (AD) patients and mouse models. Glutamate transporter-1 (GLT-1) removes glutamate from the synaptic cleft and transports it into astrocytes, where it is used as substrate for the glutamate-glutamine cycle. Ceftriaxone has been reported to improve cognitive deficits in AD mice by increasing GLT-1 expression, glutamate transformation to glutamine, and glutamine efflux from astrocytes. However, the impact of ceftriaxone on glutamine metabolism in neurons is unknown. The present study aimed to investigate whether ceftriaxone regulated the production and vesicular assembly of glutamate in the presynaptic terminals of neurons and to determine GLT-1 involvement in this process. We used the amyloid precursor protein (APP)/presenilin-1 (PS1) AD mouse model and GLT-1 knockdown APP/PS1 (GLT-1+/-/APP/PS1) mice. The expression levels of sodium-coupled neutral amino-acid transporter 1 (SNAT1) and vesicular glutamate transporters 1 and 2 (VGLUT1/2) were analyzed by immunofluorescence and immunohistochemistry staining as well as by Western blotting. Glutaminase activity was assayed by fluorometry. Ceftriaxone treatment significantly increased SNAT1 expression and glutaminase activity in neurons in APP/PS1 mice. Similarly, VGLUT1/2 levels were increased in the presynaptic terminals of APP/PS1 mice treated with ceftriaxone. The deletion of one GLT-1 allele in APP/PS1 mice prevented the ceftriaxone-induced upregulation of SNAT1 and VGLUT1/2 expression, indicating that GLT-1 played an important role in ceftriaxone effect. Based on the role of SNAT1, glutaminase, and VGLUT1/2 in the glutamate-glutamine cycle in neurons, the present results suggested that ceftriaxone improved the production and vesicular assembly of glutamate as a neurotransmitter in presynaptic terminals by acting on GLT-1 in APP/PS1 mice.


Subject(s)
Alzheimer Disease/metabolism , Anti-Bacterial Agents/pharmacology , Ceftriaxone/pharmacology , Excitatory Amino Acid Transporter 2/drug effects , Presynaptic Terminals/drug effects , Synaptic Vesicles/drug effects , Alzheimer Disease/genetics , Amino Acid Transport System A/drug effects , Amino Acid Transport System A/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Gene Knockdown Techniques , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Glutaminase/drug effects , Glutaminase/metabolism , Mice , Mice, Transgenic , Presenilin-1/genetics , Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Protein 1/drug effects , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/drug effects , Vesicular Glutamate Transport Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...