Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 19(7)2018 Jul 19.
Article in English | MEDLINE | ID: mdl-30029480

ABSTRACT

Amino acid transporters alanine-serine-cysteine transporter 2 (ASCT2) and L-Type Amino Acid Transporter 1 (LAT1) are coordinately enhanced in human cancers where among other roles, they are thought to drive mechanistic target-of-rapamycin (mTOR) growth signaling. To assess ASCT2 and LAT1 as therapeutic targets, nine unique short hairpin RNA (shRNA) vectors were used to stably suppress transporter expression in human epithelial (Hep3B) and mesenchymal (SK-Hep1) hepatocellular carcinoma (HCC) cell lines. In addition, six unique CRISPR-Cas9 vectors were used to edit the ASCT2 (SLC1A5) and LAT1 (SLC7A5) genes in epithelial (HUH7) and mesenchymal (SK-Hep1) HCC cells. Both approaches successfully diminished glutamine (ASCT2) and leucine (LAT1) initial-rate transport proportional to transporter protein suppression. In spite of profoundly reduced glutamine or leucine transport (up to 90%), transporter suppression or knockout failed to substantially affect cellular proliferation or basal and amino acid-stimulated mTORC1 growth signaling in either HCC cell type. Only LAT1 knockout in HUH7 slightly reduced growth rate. However, intracellular accumulation of radiolabeled glutamine and leucine over longer time periods largely recovered to control levels in ASCT2 and LAT1 knockout cells, respectively, which partially explains the lack of an impaired growth phenotype. These data collectively establish that in an in vitro context, human epithelial and mesenchymal HCC cell lines adapt to ASCT2 or LAT1 knockout. These results comport with an emerging model of amino acid exchangers like ASCT2 and LAT1 as "harmonizers", not drivers, of amino acid accumulation and signaling, despite their long-established dominant role in initial-rate amino acid transport.


Subject(s)
Amino Acid Transport System ASC/metabolism , Epithelium/pathology , Gene Knockout Techniques , Large Neutral Amino Acid-Transporter 1/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mesoderm/pathology , Minor Histocompatibility Antigens/metabolism , Amino Acids/metabolism , Biological Transport/drug effects , CRISPR-Cas Systems/genetics , Cell Death/drug effects , Cell Proliferation/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mifepristone/pharmacology , RNA, Antisense/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Sodium/metabolism
2.
Dalton Trans ; 41(29): 8982-8, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22722329

ABSTRACT

A series of carborane-appended 5-thio-D-glucopyranose (5-TDGP) derivatives containing one to two 5-TDGP moieties were synthesized via click cycloaddition reaction as well as following the traditional methods. Among the carboranyl-5-TDGP derivatives, the decapitated nido-carboranyl derivative 18 was found to be highly water-soluble and therefore its preliminary biodistribution study was conducted. A comparative biological evaluation of 18 versus its carboranyl-D-glucopyranose analog 19 with human hepatocellular carcinoma cells (SK-Hep1) indicated 5-TDGP to be a better boron carrier than normal D-glucopyranose. The carboranyl-5-TDGP 18 showed a nearly two fold increase in cellular boron accumulation than carboranyl-D-glucopyranose analog 19 over a period of 2 h. The accumulation of both 18 and 19 was found to occur in a temperature dependent manner. The higher accumulation of 18 suggested excellent promise for it to be a candidate for further evaluation as a future BNCT agent.


Subject(s)
Boranes/chemistry , Glucose/analogs & derivatives , Thioglucosides/chemistry , Cell Line, Tumor , Glucose/chemistry , Humans , Microscopy, Phase-Contrast , Temperature
3.
Pediatr Res ; 66(3): 266-71, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19542910

ABSTRACT

Neonatal PMN (polymorphonuclear neutrophils) exhibit altered inflammatory responsiveness and greater longevity compared with adult PMN; however, the involved mechanisms are incompletely defined. Receptors containing immunoreceptor tyrosine-based inhibitory motif (ITIM) domains promote apoptosis by activating inhibitory phosphatases, such as Src homology domain 2-containing tyrosine phosphatase-1 (SHP-1), that block survival signals. Sialic acid-binding immunoglobulin-like lectin (Siglec)-9, an immune inhibitory receptor with an ITIM domain, has been shown to induce cell death in adult PMN in association with SHP-1. To test our hypothesis that neonatal PMN inflammatory function may be modulated by unique Siglec-9 and SHP-1 interactions, we compared expression of these proteins in adult and neonatal PMN. Neonatal PMN exhibited diminished cellular expression of Siglec-9, which was phosphorylated in the basal state. Granulocyte-macrophage colony-stimulating factor (GM-CSF) treatment decreased Siglec-9 phosphorylation levels in neonatal PMN but promoted its phosphorylation in adult PMN, observations associated with altered survival signaling. Although SHP-1 expression was also diminished in neonatal PMN, GM-CSF treatment had minimal effect on phosphorylation status. Further analysis revealed that Siglec-9 and SHP-1 physically interact, as has been observed in other immune cells. Our data suggest that age-specific interactions between Siglec-9 and SHP-1 may influence the altered inflammatory responsiveness and longevity of neonatal PMN.


Subject(s)
Antigens, CD/metabolism , Lectins/metabolism , Neutrophils/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Adult , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Infant, Newborn , Neutrophils/cytology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins , bcl-Associated Death Protein/metabolism
4.
Am J Physiol Cell Physiol ; 293(1): C55-63, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17329400

ABSTRACT

System ASC amino acid transporter-2 (ASCT2) was previously demonstrated to be essential for human hepatoma cell growth and survival, as its silencing via inducible antisense RNA expression results in complete apoptosis within 48 h by a mechanism that transcends its role in amino acid delivery. To gain mechanistic insights into the reliance of cancerous liver cells on ASCT2, the aim of this study was to determine the early consequences of its silencing on the growth and survival signaling that presage apoptosis. Induced antisense ASCT2 RNA in SK-Hep1 cells led to >90% suppression of ASCT2 mRNA by 6 h and inhibition of mammalian target-of-rapamycin (mTOR)/raptor (mTOR complex-1; mTORC1) signaling by 8 h, as manifested by diminished p70 ribosomal protein S6 kinase-1 and eukaryotic initiation factor-4E (eIF4E) binding protein-1 phosphorylation, while protein synthesis rates declined by nearly 50% despite no measurable decreases in the cap binding protein eIF4G or cellular ribosomal protein content. Depressed mTORC1 signaling occurred before detectable reduction in ASCT2 activity but coincided with a 30% decline in total cellular ASCT2 protein. By 12 h after ASCT2 silencing, further decrements were observed in protein synthesis rates and ASCT2 protein and activity, each by approximately 50%, while signaling from mTOR/rictor (mTOR complex-2; mTORC2) was stimulated as indexed by enhanced phosphorylation of the Akt/PKB kinase on serine-473 and of its proapoptotic substrate Bad on serine-136. These results suggest that ASCT2 silencing inhibits mTORC1 signaling to the translational machinery followed by an mTORC2-initiated survival response, establishing a link between amino acid transporter expression and mTOR function.


Subject(s)
Amino Acid Transport System ASC/metabolism , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Liver Neoplasms/metabolism , Protein Kinases/metabolism , RNA Interference , Signal Transduction , 3-Phosphoinositide-Dependent Protein Kinases , Adaptor Proteins, Signal Transducing , Amino Acid Transport System ASC/genetics , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Minor Histocompatibility Antigens , Phosphorylation , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Messenger/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases , Time Factors , Transfection , bcl-Associated Death Protein/metabolism
5.
J Surg Res ; 131(1): 26-40, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16154153

ABSTRACT

INTRODUCTION: The amino acid glutamine (GLN) has received considerable attention as a potential therapeutic adjuvant in critical illness and in improving postoperative clinical outcomes. Most studies on the role of GLN in cellular physiology have historically focused on its anabolic roles in specific cell types and its contribution to growth in cancer cells. However, an emerging body of work that examines the consequences of GLN deprivation on cellular survival and gene expression has constructed a new paradigm for this amino acid, namely, that limited extracellular GLN supplies modulate stress and apoptotic responses. METHODS: A survey of the scientific literature was conducted on GLN in cell survival signaling and apoptosis. Work from our laboratory in liver cancer cells also was included in this review. RESULTS: Most studies on this topic have used mammalian cell lines derived from the gut, immune system (including hybridomas), and various cancers. GLN limitation, even in the presence of an adequate glucose supply, impacts stress-related gene expression, differentially modulates receptor-mediated apoptosis, and directly elicits apoptosis through signaling mechanisms and caspase cascades that are specific to cell type. To date, GLN transporters, cellular hydration, glutaminyl-tRNA synthetase, ATP levels, mRNA stability, and glutathione economy have been variably implicated in GLN-dependent survival signaling. CONCLUSION: The cell type-specific mechanisms underlying the regulatory role of GLN in cell survival continue to unfold at a steady pace through in vitro studies. These results have collectively provided testable hypotheses for further in vivo studies into their physiological relevance during GLN "nutritional pharmacology."


Subject(s)
Apoptosis/physiology , Cell Survival/physiology , Gene Expression Regulation , Glutamine/physiology , Humans , Liver/cytology , Oxidative Stress , Signal Transduction , Tumor Cells, Cultured
6.
Exp Biol Med (Maywood) ; 230(9): 606-11, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16179728

ABSTRACT

Skeletal muscle serves as the body's major glutamine repository, and releases glutamine at enhanced rates during diabetes, but whether all muscles are equally affected is unknown. System N(m) activity mediates most trans-sarcolemmal glutamine movement, and although two System N (SN) isoforms have been identified (SN1/sodium-coupled neutral amino acid transporter or System N and A transporters [SNAT]-3; and SN2/SNAT5), their expression in skeletal muscle remains controversial. Here, the impact of Type I diabetes on glutamine uptake and System N transporter expression were examined in fast- and slow-twitch skeletal muscle from spontaneously diabetic (BB/Wor-DP) rats. Net glutamine uptake in fast-twitch fibers was decreased 75%-95%, but enhanced more than 2-fold in slow-twitch muscle from diabetic animals relative to nondiabetic controls. Both SNAT3 and SNAT5 mRNA were expressed in both muscle fiber types and their abundance was unaffected by diabetes. This represents the first report of differential fiber-specific effects of diabetes on skeletal muscle glutamine transport and the co-expression of distinct System N transporters in skeletal muscle.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Glutamine/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Amino Acid Transport Systems, Neutral/genetics , Animals , Base Sequence , DNA, Complementary , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Sequence Homology, Nucleic Acid
7.
Semin Cancer Biol ; 15(4): 254-66, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15916903

ABSTRACT

Relative to other neutral amino acid transporters, the expression levels of ASCT2 and LAT1, are coordinately elevated in a wide spectrum of primary human cancers, suggesting that they are frequently co-opted to support the "tumor metabolome". Each has recently been shown to play important roles in the growth and survival of cancer cell lines, making them potential targets for cancer therapy. The properties and putative relationship of these two amino acid exchangers are discussed in the context of their demonstrated utility in cancer biology, including cellular growth and survival signaling and integrated links to the mammalian target-of-rapamycin (mTOR) kinase.


Subject(s)
Amino Acid Transport System ASC/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Neoplasms/metabolism , Biological Transport , Humans , Minor Histocompatibility Antigens , Neoplasms/pathology , Protein Kinases/metabolism , TOR Serine-Threonine Kinases
8.
Am J Physiol Gastrointest Liver Physiol ; 286(3): G467-78, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14563674

ABSTRACT

Amino acid transporter B(0)/ASC transporter 2 (ATB(0)/ASCT2) is responsible for most glutamine uptake in human hepatoma cells. Because this transporter is not expressed in normal hepatocytes, we hypothesized that its expression is necessary for growth of human liver cancer cells. To test this hypothesis, Sloan Kettering hepatoma (SK-Hep) cells were stably transfected with an inducible 1.3-kb ATB(0)/ASCT2 antisense RNA expression plasmid under the transcriptional control of mifepristone, a synthetic steroid. Induced antisense RNA expression in monolayer cultures decreased ATB(0)/ASCT2 mRNA levels by 73% and glutamine transport rates by 65% compared with controls after 24 h, leading to a 98% decrease in cell number after 48 h. Cellular death was attributable to apoptosis based on cellular blebbing, caspase-3 activation, vital dye and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining, and poly-(ADP-ribose) polymerase (PARP) cleavage. Transporter knockdown also markedly increased activities of caspases-2 and -9, marginally enhanced caspase-8 activity, and dramatically increased ASCT1 mRNA levels, presumably as a futile compensatory response. Apoptosis elicited via transporter silencing was not attributable to the double-stranded RNA-dependent protein kinase R (PKR) pathway. For comparison, glutamine deprivation also caused apoptotic cell death but with slower temporal kinetics, stimulated caspases-2 and -3 but not caspases-8 or -9 activities, and led to considerable PARP cleavage. Thus ASCT2 suppression exerts proapoptotic effects transcending those of glutamine starvation alone. We conclude that ATB(0)/ASCT2 expression is necessary for SK-Hep cell growth and viability and suggest that it be further explored as a selective target for human hepatocellular carcinoma.


Subject(s)
Amino Acid Transport System ASC/genetics , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA, Antisense/genetics , RNA, Antisense/pharmacology , Biological Transport, Active , Blotting, Northern , Blotting, Western , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , Cell Division/drug effects , Cell Line, Tumor , DNA Fragmentation , Enzyme Activation/drug effects , Genetic Vectors , Glutamine/metabolism , Humans , In Situ Nick-End Labeling , Kinetics , Liver Neoplasms/pathology , Minor Histocompatibility Antigens , Transfection
9.
Am J Physiol Gastrointest Liver Physiol ; 283(5): G1062-73, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12381519

ABSTRACT

Human hepatoma cells take up glutamine at rates severalfold faster than the system N-mediated transport rates observed in normal human hepatocytes. Amino acid inhibition, kinetic, Northern blotting, RT-PCR, and restriction enzyme analyses collectively identified the transporter responsible in six human hepatoma cell lines as amino acid transporter B(0) (ATB(0)), the human ortholog of rodent ASCT2. The majority of glutamine uptake in liver fibroblasts and an immortalized human liver epithelial cell line (THLE-5B) was also mediated by ATB(0). The 2.9-kb ATB(0) mRNA was equally expressed in all cell lines, whereas expression of the system A transporters ATA2 and ATA3 was variable. In contrast, the system N isoforms (SN1 and SN2) were expressed only in well-differentiated hepatomas. ATB(0) mRNA was also expressed in cirrhotic liver and adult and pediatric liver cancer biopsies but was not detectable in isolated human hepatocytes or fetal liver. Although the growth of all hepatomas was glutamine dependent, competitive inhibition of ATB(0)-mediated glutamine uptake blocked proliferation only in poorly differentiated cells lacking SN1 or SN2 expression and exhibiting low glutamine synthetase mRNA levels.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glutamine/pharmacokinetics , Liver Neoplasms/metabolism , Liver/metabolism , Amino Acid Transport System ASC/physiology , Biological Transport , Blotting, Northern , Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Cell Division/physiology , Cells, Cultured , Fibroblasts/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamine/metabolism , Humans , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Minor Histocompatibility Antigens , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...