Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Exp Biol Med (Maywood) ; 246(9): 1121-1138, 2021 05.
Article in English | MEDLINE | ID: mdl-33601913

ABSTRACT

In the present minireview, we intend to provide a brief history of the field of CD9 involvement in oncogenesis and in the metastatic process of cancer, considering its potential value as a tumor-associated antigenic target. Over the years, CD9 has been identified as a favorable prognostic marker or predictor of metastatic potential depending on the cancer type. To understand its implications in cancer beside its use as an antigenic biomarker, it is essential to know its physiological functions, including its molecular partners in a given cell system. Moreover, the discovery that CD9 is one of the most specific and broadly expressed markers of extracellular membrane vesicles, nanometer-sized entities that are released into extracellular space and various physiological body fluids and play a role in intercellular communication under physiological and pathological conditions, notably the establishment of cancer metastases, has added a new dimension to our knowledge of CD9 function in cancer. Here, we will discuss these issues as well as the possible cancer therapeutic implications of CD9, their limitations, and pitfalls.


Subject(s)
Neoplasms , Tetraspanin 29 , Animals , Humans
2.
PLoS One ; 11(1): e0146994, 2016.
Article in English | MEDLINE | ID: mdl-26789264

ABSTRACT

Uridine is a pyrimidine nucleoside that exerts restorative functions in tissues under stress. Short-term co-administration of uridine with multiple unrelated drugs prevents drug-induced liver lipid accumulation. Uridine has the ability to modulate liver metabolism; however, the precise mechanism has not been delineated. In this study, long-term effects of uridine on liver metabolism were examined in both HepG2 cell cultures and C57BL/6J mice. We report that uridine administration was associated with O-GlcNAc modification of FOXO1, increased gluconeogenesis, reduced insulin signaling activity, and reduced expression of a liver-specific fatty acid binding protein FABP1. Long-term uridine feeding induced systemic glucose intolerance and severe liver lipid accumulation in mice. Our findings suggest that the therapeutic potentials of uridine should be designed for short-term acute administration.


Subject(s)
Fatty Liver , Forkhead Transcription Factors/metabolism , Gluconeogenesis/drug effects , Glucose Intolerance , Prediabetic State , Uridine/adverse effects , Animals , Fatty Liver/chemically induced , Fatty Liver/metabolism , Fatty Liver/pathology , Forkhead Box Protein O1 , Glucose Intolerance/chemically induced , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Hep G2 Cells , Humans , Mice , Prediabetic State/chemically induced , Prediabetic State/metabolism , Prediabetic State/pathology , Uridine/pharmacology
3.
BMC Pharmacol Toxicol ; 15: 27, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24887406

ABSTRACT

BACKGROUND: Tamoxifen, an agonist of estrogen receptor, is widely prescribed for the prevention and long-term treatment of breast cancer. A side effect of tamoxifen is fatty liver, which increases the risk for non-alcoholic fatty liver disease. Prevention of tamoxifen-induced fatty liver has the potential to improve the safety of long-term tamoxifen usage. METHODS: Uridine, a pyrimidine nucleoside with reported protective effects against drug-induced fatty liver, was co-administered with tamoxifen in C57BL/6J mice. Liver lipid levels were evaluated with lipid visualization using coherent anti-Stokes Raman scatting (CARS) microscopy, biochemical assay measurement of triacylglyceride (TAG), and liquid chromatography coupled with mass spectrometry (LC-MS) measurement of membrane phospholipid. Blood TAG and cholesterol levels were measured. Mitochondrial respiration of primary hepatocytes in the presence of tamoxifen and/or uridine was evaluated by measuring oxygen consumption rate with an extracellular flux analyzer. Liver protein lysine acetylation profiles were evaluated with 1D and 2D Western blots. In addition, the relationship between endogenous uridine levels, fatty liver, and tamoxifen administration was evaluated in transgenic mice UPase1-/-and UPase1-TG. RESULTS: Uridine co-administration prevented tamoxifen-induced liver lipid droplet accumulation in mice. The most prominent effect of uridine co-administration with tamoxifen was the stimulation of liver membrane phospholipid biosynthesis. Uridine had no protective effect against tamoxifen-induced impairment to mitochondrial respiration of primary hepatocytes or liver TAG and cholesterol export. Uridine had no effect on tamoxifen-induced changes to liver protein acetylation profile. Transgenic mice UPase1-/-with increased pyrimidine salvage activity were protected against tamoxifen-induced liver lipid droplet accumulation. In contrast, UPase1-TG mice with increased pyrimidine catabolism activity had intrinsic liver lipid droplet accumulation, which was aggravated following tamoxifen administration. CONCLUSION: Uridine co-administration was effective at preventing tamoxifen-induced liver lipid droplet accumulation. The ability of uridine to prevent tamoxifen-induced fatty liver appeared to depend on the pyrimidine salvage pathway, which promotes biosynthesis of membrane phospholipid.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Fatty Liver/prevention & control , Protective Agents/therapeutic use , Tamoxifen/adverse effects , Uridine/therapeutic use , Animals , Fatty Liver/chemically induced , Fatty Liver/metabolism , Female , Lipid Metabolism/drug effects , Lipids/blood , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protective Agents/pharmacology , Uridine/pharmacology , Uridine Phosphorylase/deficiency , Uridine Phosphorylase/genetics
4.
PLoS One ; 9(6): e99728, 2014.
Article in English | MEDLINE | ID: mdl-24918436

ABSTRACT

Purines and pyrimidines are complementary bases of the genetic code. The roles of purines and their derivatives in cellular signal transduction and energy metabolism are well-known. In contrast, the roles of pyrimidines and their derivatives in cellular function remain poorly understood. In this study, the roles of uridine, a pyrimidine nucleoside, in liver metabolism are examined in mice. We report that short-term uridine administration in C57BL/6J mice increases liver protein glycosylation profiles, reduces phosphorylation level of insulin signaling proteins, and activates the HRI-eIF-2α-ATF4 heme-deficiency stress response pathway. Short-term uridine administration is also associated with reduced liver hemin level and reduced ability for insulin-stimulated blood glucose removal during an insulin tolerance test. Some of the short-term effects of exogenous uridine in C57BL/6J mice are conserved in transgenic UPase1-/- mice with long-term elevation of endogenous uridine level. UPase1-/- mice exhibit activation of the liver HRI-eIF-2α-ATF4 heme-deficiency stress response pathway. UPase1-/- mice also exhibit impaired ability for insulin-stimulated blood glucose removal. However, other short-term effects of exogenous uridine in C57BL/6J mice are not conserved in UPase1-/- mice. UPase1-/- mice exhibit normal phosphorylation level of liver insulin signaling proteins and increased liver hemin concentration compared to untreated control C57BL/6J mice. Contrasting short-term and long-term consequences of uridine on liver metabolism suggest that uridine exerts transient effects and elicits adaptive responses. Taken together, our data support potential roles of pyrimidines and their derivatives in the regulation of liver metabolism.


Subject(s)
Glycosylation/drug effects , Heme/metabolism , Insulin/metabolism , Liver/drug effects , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , Uridine/administration & dosage , Activating Transcription Factor 4/metabolism , Animals , Blood Glucose/drug effects , Eukaryotic Initiation Factor-2/metabolism , Glucose/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/drug effects
5.
Brain ; 137(Pt 5): 1337-49, 2014 May.
Article in English | MEDLINE | ID: mdl-24727567

ABSTRACT

Balanced pools of deoxyribonucleoside triphosphate precursors are required for DNA replication, and alterations of this balance are relevant to human mitochondrial diseases including mitochondrial neurogastrointestinal encephalopathy. In this disease, autosomal recessive TYMP mutations cause severe reductions of thymidine phosphorylase activity; marked elevations of the pyrimidine nucleosides thymidine and deoxyuridine in plasma and tissues, and somatic multiple deletions, depletion and site-specific point mutations of mitochondrial DNA. Thymidine phosphorylase and uridine phosphorylase double knockout mice recapitulated several features of these patients including thymidine phosphorylase activity deficiency, elevated thymidine and deoxyuridine in tissues, mitochondrial DNA depletion, respiratory chain defects and white matter changes. However, in contrast to patients with this disease, mutant mice showed mitochondrial alterations only in the brain. To test the hypothesis that elevated levels of nucleotides cause unbalanced deoxyribonucleoside triphosphate pools and, in turn, pathogenic mitochondrial DNA instability, we have stressed double knockout mice with exogenous thymidine and deoxyuridine, and assessed clinical, neuroradiological, histological, molecular, and biochemical consequences. Mutant mice treated with exogenous thymidine and deoxyuridine showed reduced survival, body weight, and muscle strength, relative to untreated animals. Moreover, in treated mutants, leukoencephalopathy, a hallmark of the disease, was enhanced and the small intestine showed a reduction of smooth muscle cells and increased fibrosis. Levels of mitochondrial DNA were depleted not only in the brain but also in the small intestine, and deoxyribonucleoside triphosphate imbalance was observed in the brain. The relative proportion, rather than the absolute amount of deoxyribonucleoside triphosphate, was critical for mitochondrial DNA maintenance. Thus, our results demonstrate that stress of exogenous pyrimidine nucleosides enhances the mitochondrial phenotype of our knockout mice. Our mouse studies provide insights into the pathogenic role of thymidine and deoxyuridine imbalance in mitochondrial neurogastrointestinal encephalopathy and an excellent model to study new therapeutic approaches.


Subject(s)
Deoxyribonucleosides/adverse effects , Intestinal Pseudo-Obstruction/chemically induced , Intestinal Pseudo-Obstruction/genetics , Mitochondrial Encephalomyopathies/chemically induced , Mitochondrial Encephalomyopathies/genetics , Age Factors , Animals , Body Weight/drug effects , Body Weight/genetics , Brain/pathology , Deoxyribonucleosides/metabolism , Disease Models, Animal , Intestinal Pseudo-Obstruction/mortality , Intestinal Pseudo-Obstruction/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases/etiology , Mitochondrial Diseases/genetics , Mitochondrial Encephalomyopathies/mortality , Mitochondrial Encephalomyopathies/physiopathology , Motor Activity/drug effects , Muscle Strength/drug effects , Muscle Strength/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Oculopharyngeal , Ophthalmoplegia/congenital , Psychomotor Disorders/etiology , Psychomotor Disorders/genetics , Succinate Dehydrogenase/metabolism , Thymidine/adverse effects , Thymidine/metabolism , Thymidine Phosphorylase/deficiency , Uridine Phosphorylase/deficiency
6.
PLoS One ; 9(1): e87179, 2014.
Article in English | MEDLINE | ID: mdl-24475249

ABSTRACT

Uridine, a pyrimidine nucleoside, can modulate liver lipid metabolism although its specific acting targets have not been identified. Using mice with fenofibrate-induced fatty liver as a model system, the effects of uridine on liver lipid metabolism are examined. At a daily dosage of 400 mg/kg, fenofibrate treatment causes reduction of liver NAD(+)/NADH ratio, induces hyper-acetylation of peroxisomal bifunctional enzyme (ECHD) and acyl-CoA oxidase 1 (ACOX1), and induces excessive accumulation of long chain fatty acids (LCFA) and very long chain fatty acids (VLCFA). Uridine co-administration at a daily dosage of 400 mg/kg raises NAD(+)/NADH ratio, inhibits fenofibrate-induced hyper-acetylation of ECHD, ACOX1, and reduces accumulation of LCFA and VLCFA. Our data indicates a therapeutic potential for uridine co-administration to prevent fenofibrate-induced fatty liver.


Subject(s)
Fatty Liver/prevention & control , Fenofibrate/adverse effects , Hypolipidemic Agents/adverse effects , Protective Agents/pharmacology , Uridine/pharmacology , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Biological Transport/drug effects , Fatty Acids/metabolism , Fatty Liver/chemically induced , Fatty Liver/enzymology , Fatty Liver/pathology , Gene Expression Regulation , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD/metabolism , Oxidation-Reduction/drug effects , Peroxisomal Bifunctional Enzyme/genetics , Peroxisomal Bifunctional Enzyme/metabolism
7.
Mol Ther ; 22(5): 901-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24448160

ABSTRACT

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is an autosomal recessive disorder caused by mutations in TYMP, enconding thymidine phosphorylase (TP). TP deficiency results in systemic accumulation of thymidine and deoxyuridine, which interferes with mitochondrial DNA (mtDNA) replication and leads to mitochondrial dysfunction. To date, the only treatment available for MNGIE patients is allogeneic hematopoietic stem cell transplantation, which is associated with high morbidity and mortality. Here, we report that AAV2/8-mediated transfer of the human TYMP coding sequence (hcTYMP) under the control of a liver-specific promoter prevents the biochemical imbalances in a murine model of MNGIE. hcTYMP expression was restricted to liver, and a dose as low as 2 × 10(11) genome copies/kg led to a permanent reduction in systemic nucleoside levels to normal values in about 50% of treated mice. Higher doses resulted in reductions to normal or slightly below normal levels in virtually all mice treated. The nucleoside reduction achieved by this treatment prevented deoxycytidine triphosphate (dCTP) depletion, which is the limiting factor affecting mtDNA replication in this disease. These results demonstrate that the use of AAV to direct TYMP expression in liver is feasible as a potentially safe gene therapy strategy for MNGIE.


Subject(s)
Genetic Therapy , Intestinal Pseudo-Obstruction/genetics , Intestinal Pseudo-Obstruction/therapy , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/therapy , Thymidine Phosphorylase/genetics , Animals , DNA, Mitochondrial/genetics , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Homeostasis/genetics , Humans , Intestinal Pseudo-Obstruction/pathology , Liver/metabolism , Mice , Mitochondrial Encephalomyopathies/pathology , Muscular Dystrophy, Oculopharyngeal , Mutation , Ophthalmoplegia/congenital , Thymidine/metabolism , Thymidine Phosphorylase/biosynthesis
8.
Thromb Haemost ; 109(6): 1108-19, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23494007

ABSTRACT

Using direct injection mass spectrometry (DIMS) we discovered that deoxyribose-1-phosphate (dRP) is released by platelets upon activation. Interestingly, the addition of exogenous dRP to human platelets significantly increased platelet aggregation and integrin αIIbß3 activation in response to thrombin. In parallel, genetically modified platelets with double genetic deletion of thymidine phosphorylase and uridine phosphorylase were characterised by reduced release of dRP, impaired aggregation and decreased integrin αIIbß3 activation in response to thrombin. In vitro platelet adhesion onto fibrinogen and collagen under physiological flow conditions was potentiated by treatment of human platelets with exogenous dRP and impaired in transgenic platelets with reduced dRP release. Human and mouse platelets responded to dRP treatment with a sizeable increase in reactive oxygen species (ROS) generation and the pre-treament with the antioxidant apocynin abolished the effect of dRP on aggregation and integrin activation. Experiments directly assessing the activation of the small G protein Rap1b and protein kinase C suggested that dRP increases the basal levels of activity of these two pivotal platelet-activating pathways in a redox-dependent manner. Taken together, we present evidence that dRP is a novel autocrine amplifier of platelet activity, which acts on platelet redox levels and modulates integrin αIIbß3.


Subject(s)
Platelet Activation/drug effects , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Ribosemonophosphates/chemistry , Animals , Blood Platelets/metabolism , Flow Cytometry , Humans , Mass Spectrometry , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Protein Kinase C/metabolism , Reactive Oxygen Species , Signal Transduction , Thrombin/metabolism , Thymidine Phosphorylase/metabolism , Uridine Phosphorylase/metabolism , rap GTP-Binding Proteins/metabolism
9.
J Lipid Res ; 54(4): 1044-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23355744

ABSTRACT

We report in this study an intrinsic link between pyrimidine metabolism and liver lipid accumulation utilizing a uridine phosphorylase 1 transgenic mouse model UPase1-TG. Hepatic microvesicular steatosis is induced by disruption of uridine homeostasis through transgenic overexpression of UPase1, an enzyme of the pyrimidine catabolism and salvage pathway. Microvesicular steatosis is also induced by the inhibition of dihydroorotate dehydrogenase (DHODH), an enzyme of the de novo pyrimidine biosynthesis pathway. Interestingly, uridine supplementation completely suppresses microvesicular steatosis in both scenarios. The effective concentration (EC(50)) for uridine to suppress microvesicular steatosis is approximately 20 µM in primary hepatocytes of UPase1-TG mice. We find that uridine does not have any effect on in vitro DHODH enzymatic activity. On the other hand, uridine supplementation alters the liver NAD(+)/NADH and NADP(+)/NADPH ratios and the acetylation profile of metabolic, oxidation-reduction, and antioxidation enzymes. Protein acetylation is emerging as a key regulatory mechanism for cellular metabolism. Therefore, we propose that uridine suppresses fatty liver by modulating the liver protein acetylation profile. Our findings reveal a novel link between uridine homeostasis, pyrimidine metabolism, and liver lipid metabolism.


Subject(s)
Liver/metabolism , Pyrimidines/metabolism , Uridine/metabolism , Animals , Cells, Cultured , Fatty Acids/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Male , Mice , Mice, Transgenic , Triglycerides/metabolism , Uridine Phosphorylase/genetics , Uridine Phosphorylase/metabolism
10.
PLoS One ; 7(11): e51092, 2012.
Article in English | MEDLINE | ID: mdl-23226469

ABSTRACT

Hepatic microvesicular steatosis is a hallmark of drug-induced hepatotoxicity and early-stage fatty liver disease. Current histopathology techniques are inadequate for the clinical evaluation of hepatic microvesicular steatosis. In this paper, we explore the use of multimodal coherent anti-Stokes Raman scattering (CARS) microscopy for the detection and characterization of hepatic microvesicular steatosis. We show that CARS microscopy is more sensitive than Oil Red O histology for the detection of microvesicular steatosis. Computer-assisted analysis of liver lipid level based on CARS signal intensity is consistent with triglyceride measurement using a standard biochemical assay. Most importantly, in a single measurement procedure on unprocessed and unstained liver tissues, multimodal CARS imaging provides a wealth of critical information including the detection of microvesicular steatosis and quantitation of liver lipid content, number and size of lipid droplets, and lipid unsaturation and packing order of lipid droplets. Such information can only be assessed by multiple different methods on processed and stained liver tissues or tissue extracts using current standard analytical techniques. Multimodal CARS microscopy also permits label-free identification of lipid-rich non-parenchymal cells. In addition, label-free and non-perturbative CARS imaging allow rapid screening of mitochondrial toxins-induced microvesicular steatosis in primary hepatocyte cultures. With its sensitivity and versatility, multimodal CARS microscopy should be a powerful tool for the clinical evaluation of hepatic microvesicular steatosis.


Subject(s)
Fatty Liver/pathology , Liver/blood supply , Liver/pathology , Microscopy/methods , Microvessels/pathology , Spectrum Analysis, Raman/methods , Staining and Labeling , Animals , Azo Compounds/metabolism , Cells, Cultured , Hepatocytes/metabolism , Hepatocytes/pathology , Image Processing, Computer-Assisted , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism
11.
Recent Pat Anticancer Drug Discov ; 7(2): 154-67, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22339355

ABSTRACT

ATP citrate lyase (ACL or ACLY) is an extra-mitochondrial enzyme widely distributed in various human and animal tissues. ACL links glucose and lipid metabolism by catalyzing the formation of acetyl-CoA and oxaloacetate from citrate produced by glycolysis in the presence of ATP and CoA. ACL is aberrantly expressed in many immortalized cells and tumors, such as breast, liver, colon, lung and prostate cancers, and is correlated reversely with tumor stage and differentiation, serving as a negative prognostic marker. ACL is an upstream enzyme of the long chain fatty acid synthesis, providing acetyl-CoA as an essential component of the fatty acid synthesis. Therefore, ACL is a key enzyme of cellular lipogenesis and potent target for cancer therapy. As a hypolipidemic strategy of metabolic syndrome and cancer treatment, many small chemicals targeting ACL have been designed and developed. This review article provides an update for the research and development of ACL inhibitors with a focus on their patent status, offering a new insight into their potential application.


Subject(s)
ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Lipogenesis/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Citric Acid/analogs & derivatives , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Mice , Patents as Topic , Protein Processing, Post-Translational
12.
Mol Cancer Ther ; 10(12): 2330-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21954436

ABSTRACT

Abrogation of uridine phosphorylase (UPase) leads to abnormalities in pyrimidine metabolism and host protection against 5-fluorouracil (5-FU) toxicity. We elucidated the effects on the metabolism and antitumor efficacy of 5-FU and capecitabine (N(4)-pentyloxycarbonyl-5'-deoxy-5-fluorocytidine) in our UPase knockout (UPase(-/-)) model. Treatment with 5-FU (85 mg/kg) or capecitabine (1,000 mg/kg) five days a week for four weeks caused severe toxicity and structural damage to the intestines of wild-type (WT) mice, but not in UPase(-/-) animals. Capecitabine treatment resulted in a 70% decrease in blood cell counts of WT animals, with only a marginal effect in UPase(-/-) mice. UPase expressing colon 38 tumors implanted in UPase(-/-) mice revealed an improved therapeutic efficacy when treated with 5-FU and capecitabine because of the higher maximum tolerated dose for fluoropyrimidines achievable in UPase(-/-) mice. (19)F-MRS evaluation of capecitabine metabolism in tumors revealed similar activation of the prodrug in UPase(-/-) mice compared with WT. In WT mice, approximately 60% of capecitabine was transformed over three hours into its active metabolites, whereas 80% was transformed in tumors implanted in UPase(-/-) mice. In UPase(-/-) mice, prolonged retention of 5'dFUR allowed a proportional increase in tumor tissue. The similar presence of fluorinated catabolic species confirms that dihydropyrimidine dehydrogenase activity was not altered in UPase(-/-) mice. Overall, these results indicate the importance of UPase in the activation of fluoropyrimidines, the effect of uridine in protecting normal tissues, and the role for tumor-specific modulation of the phosphorolytic activity in 5-FU or capecitabine-based chemotherapy.


Subject(s)
Fluorouracil/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Uridine Phosphorylase/genetics , Animals , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Capecitabine , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Fluorouracil/analogs & derivatives , Fluorouracil/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/enzymology , Neoplasms/metabolism , Prodrugs/metabolism , Prodrugs/therapeutic use , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Treatment Outcome , Uridine Phosphorylase/metabolism , Uridine Phosphorylase/physiology
13.
J Struct Biol ; 176(2): 229-37, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21855639

ABSTRACT

Uridine phosphorylase (UPP) catalyzes the reversible conversion of uridine to uracil and ribose-1-phosphate and plays an important pharmacological role in activating fluoropyrimidine nucleoside chemotherapeutic agents such as 5-fluorouracil and capecitabine. Most vertebrate animals, including humans, possess two homologs of this enzyme (UPP1 & UPP2), of which UPP1 has been more thoroughly studied and is better characterized. Here, we report two crystallographic structures of human UPP2 (hUPP2) in distinctly active and inactive conformations. These structures reveal that a conditional intramolecular disulfide bridge can form within the protein that dislocates a critical phosphate-coordinating arginine residue (R100) away from the active site, disabling the enzyme. In vitro activity measurements on both recombinant hUPP2 and native mouse UPP2 confirm the redox sensitivity of this enzyme, in contrast to UPP1. Sequence analysis shows that this feature is conserved among UPP2 homologs and lacking in all UPP1 proteins due to the absence of a necessary cysteine residue. The state of the disulfide bridge has further structural consequences for one face of the enzyme that suggest UPP2 may have additional functions in sensing and initiating cellular responses to oxidative stress. The molecular details surrounding these dynamic aspects of hUPP2 structure and regulation provide new insights as to how novel inhibitors of this protein may be developed with improved specificity and affinity. As uridine is emerging as a promising protective compound in neuro-degenerative diseases, including Alzheimer's and Parkinson's, understanding the regulatory mechanisms underlying UPP control of uridine concentration is key to improving clinical outcomes in these illnesses.


Subject(s)
Uridine Phosphorylase/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Cystine/chemistry , Enzyme Assays , Humans , Hydrogen Bonding , Mice , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Uracil/analogs & derivatives , Uracil/chemistry , Uridine Phosphorylase/antagonists & inhibitors
14.
Chemother Res Pract ; 2011: 965826, 2011.
Article in English | MEDLINE | ID: mdl-22295208

ABSTRACT

Myelodysplastic syndromes (MDSs) represent a group of incurable stem-cell malignancies which are predominantly treated by supportive care. Epigenetic silencing through promoter methylation of a number of genes is present in poor-risk subtypes of MDS and often predicts transformation to acute myelogenous leukemia (AML). Azacitidine and decitabine, two FDA-approved DNA methyltransferase (DNMT) inhibitors, are able to improve overall response although their oral bioavailability complicates their clinical use. This study evaluated 2', 3', 5'-triacetyl-5-azacitidine (TAC) as a potential prodrug for azacitidine. The prodrug demonstrated significant pharmacokinetic improvements in bioavailability, solubility, and stability over the parent compound. In vivo analyses indicated a lack of general toxicity coupled with significantly improved survival. Pharmacodynamic analyses confirmed its ability to suppress global methylation in vivo. These data indicate that esterified nucleoside derivatives may be effective prodrugs for azacitidine and encourages further investigation of TAC into its metabolism, activity, and possible clinical evaluation.

15.
Arterioscler Thromb Vasc Biol ; 30(12): 2631-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20884872

ABSTRACT

OBJECTIVE: Micromolar concentrations of the proangiogenic metabolite deoxyribose-1-phosphate (dRP) were detected in platelet supernatants by mass spectrometry. In this study, we assessed whether the release of dRP by platelets stimulates endothelial cell migration and angiogenesis. METHODS AND RESULTS: Protein-free supernatants from thrombin-stimulated platelets increased human umbilical vein endothelial cell migratory activity in transmigration and monolayer repair assays. This phenomenon was ablated by genetic silencing of dRP-generating uridine phosphorylase (UP) and thymidine phosphorylase (TP) or pharmacological inhibition of UP and restored by exogenous dRP. The stimulation of endothelial cell migration by platelet-derived dRP correlated with upregulation of integrin ß(3), which was induced in a reactive oxygen species-dependent manner, and was mediated by the activity of the integrin heterodimer α(v)ß(3). The physiological relevance of dRP release by platelets was confirmed in a chick chorioallantoic membrane assay, where the presence of this metabolite in platelet supernatants strongly induced capillary formation. CONCLUSIONS: Platelet-derived dRP stimulates endothelial cell migration by upregulating integrin ß(3) in a reactive oxygen species-dependent manner. As demonstrated by our in vivo experiments, this novel paracrine regulatory pathway is likely to play an important role in the stimulation of angiogenesis by platelets.


Subject(s)
Blood Platelets/metabolism , Cell Movement , Chorioallantoic Membrane/blood supply , Endothelial Cells/metabolism , Neovascularization, Physiologic , Paracrine Communication , Ribosemonophosphates/metabolism , Animals , Blood Platelets/drug effects , Cell Movement/drug effects , Cells, Cultured , Chick Embryo , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Gas Chromatography-Mass Spectrometry , Gene Silencing , Humans , Integrin alphaV/metabolism , Integrin alphaVbeta3/metabolism , Integrin beta3/metabolism , Neovascularization, Physiologic/drug effects , Paracrine Communication/drug effects , Reactive Oxygen Species/metabolism , Thrombin/metabolism , Thymidine Phosphorylase/antagonists & inhibitors , Thymidine Phosphorylase/genetics , Thymidine Phosphorylase/metabolism , Time Factors , Uridine Phosphorylase/antagonists & inhibitors , Uridine Phosphorylase/genetics , Uridine Phosphorylase/metabolism
16.
Surg Obes Relat Dis ; 6(5): 538-41, 2010.
Article in English | MEDLINE | ID: mdl-20688580

ABSTRACT

BACKGROUND: Obesity is associated with increased tumorigenesis. Previously, we demonstrated that inflammation in obesity caused cancer fighting cells to display greater surface receptor levels, predisposing them to early cell death. We measured the inflammatory tumor growth factor levels to determine whether inflammation in obesity increases expression of these factors, potentially predisposing these patients to greater rates of neoplasia. METHODS: A total of 24 patients undergoing weight loss surgery had samples collected preoperatively and at 6 and 12 months after surgery. The growth factors analyzed included tumor necrosis factor (TNF)-α, granulocyte-macrophage colony-stimulating factor, interferon-γ, interleukin (IL)-1b, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, vascular endothelial growth factor, hepatocyte growth factor, TNF-receptor I (TNF-RI), TNF-RII, death receptor 5, leptin, and adiponectin. Control samples were obtained from 10 healthy, normal weight volunteers. RESULTS: The tumor growth factors TNF-α, TNF-RI, TNF-RII, vascular endothelial growth factor, hepatocyte growth factor, interferon-γ, IL-2, IL-5, and IL-6 all decreased significantly (P <.05) compared with the preoperative values. The IL-4, IL-8, leptin, death receptor 5, adiponectin, and granulocyte-macrophage colony-stimulating factor levels did not change significantly over time. The IL-1b and IL-10 levels were less than the detection limit at all points. When obese patient serum was compared with healthy volunteer pooled serum, we found that the leptin, death receptor 5, hepatocyte growth factor, vascular endothelial growth factor, TNF-RI, TNF-RII, TNF-α, IFN-γ, granulocyte-macrophage colony-stimulating factor, IL-4, IL-5, IL-6, and IL-8 levels were all 2-37 times greater than the levels in the controls at baseline. The concentrations of these same growth factors had decreased levels only 1-3.5 times greater than those of the controls at 12 months postoperatively. CONCLUSION: Many inflammatory tumor growth factors are present in greater concentrations in obese individuals. This could explain the greater prevalence of neoplasia in the morbidly obese population.


Subject(s)
Neoplasms/metabolism , Obesity/metabolism , Tumor Necrosis Factor-alpha/metabolism , Weight Loss , Adiponectin/metabolism , Adult , Disease Susceptibility , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukins/metabolism , Leptin/metabolism , Male , Neoplasms/etiology , Obesity/complications , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Risk Factors , Vascular Endothelial Growth Factor A/metabolism
17.
Nucleosides Nucleotides Nucleic Acids ; 29(4-6): 488-503, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20544543

ABSTRACT

Uridine phosphorylase (UPase) has been shown to be induced in various human and murine tumors and could potentially serve as a specific target for the modulation of tumor-selectivity of fluoropyrimidines. However, the signaling mechanisms underlying the regulation of UPase gene expression have not been determined. In this study, we investigated the effects of IFN-gamma on the regulation of TNF-alpha-induced UPase activity and have uncovered the molecular mechanisms of this potentiation, utilizing murine EMT6 breast cancer cells. Our data has shown that IFN-gamma can significantly increase UPase mRNA expression and the enzymatic activity induced by TNF-alpha in a dose-dependent manner, resulting in an enhanced sensitivity to 5-fluorouracil (5-FU) and 5'-Deoxy-5-fluorouridine (5'DFUR). We have previously shown that TNF-alpha activates NF-kappaB through increased translocation of NF-kappaB p65 from the cytoplasm into the nuclei. Exposure to IFN-gamma mainly affects nuclear IRF-1 and STAT1 in EMT6, but inhibits NF-kappaB p65 activity, indicating that the cooperative stimulation was the result of the independent activation of NF-kappaB, STAT1 and IRF-1 transcriptional factors through binding to their unique sites in the UPase promoter. Notably, the activation of NF-kappaB and STAT1 in human breast tissues is consistent with UPase activity; signifying their role in the up-regulation of the UPase gene expression in human tumors.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferon-gamma/pharmacology , NF-kappa B/metabolism , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Uridine Phosphorylase/metabolism , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Enzyme Activation/drug effects , Female , Gene Expression/drug effects , Humans , In Vitro Techniques , Mice
18.
Mol Cancer Ther ; 8(5): 1037-44, 2009 May.
Article in English | MEDLINE | ID: mdl-19383847

ABSTRACT

5-Fluorouracil (5-FU) continues to be widely used for treatment of gastrointestinal cancers. Because many tumors show primary or acquired resistance, it is important to understand the molecular basis underlying the mechanism of resistance to 5-FU. In addition to its effect on thymidylate synthase inhibition and DNA synthesis, 5-FU may also influence RNA metabolism. Our previous studies revealed that colorectal cancer cells resistant to bolus 5-FU (HCT-8/4hFU) showed significantly decreased incorporation of the drug into RNA. Resistance to bolus 5-FU was associated with lower expression of UMP kinase (UMPK), an enzyme that plays an important role in the activation of 5-FU to 5-FUTP and its incorporation into RNA. Activities of other 5-FU-metabolizing enzymes (e.g., thymidine kinase, uridine phosphorylase, thymidine phosphorylase, and orotate phosphoribosyltransferase) remained unchanged between sensitive and resistant cell lines. Herein, we show that UMPK down-regulation in 5-FU-sensitive cells (HCT-8/P) induces resistance to bolus 5-FU treatment. Moreover, HCT-8/4hFU cells are even more cross-resistant to treatment with 5-fluorouridine, consistent with the current understanding of 5-fluorouridine as a RNA-directed drug. Importantly, colorectal cancer hepatic metastases isolated from patients clinically resistant to weekly bolus 5-FU/leucovorin treatment exhibited decreased mRNA expression of UMPK but not thymidylate synthase or dihydropyrimidine dehydrogenase compared with tumor samples of patients not previously exposed to 5-FU. Our findings provide new insights into the mechanisms of acquired resistance to 5-FU in colorectal cancer and implicate UMPK as an important mechanism of clinical resistance to pulse 5-FU treatment in some patients.


Subject(s)
Colonic Neoplasms/enzymology , Drug Resistance, Neoplasm/genetics , Liver Neoplasms/enzymology , Nucleoside-Phosphate Kinase/metabolism , Uridine/analogs & derivatives , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Liver Neoplasms/drug therapy , Nucleoside-Phosphate Kinase/genetics , RNA Interference , Uridine/pharmacology , Uridine/therapeutic use
19.
BMC Struct Biol ; 9: 14, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19291308

ABSTRACT

BACKGROUND: Uridine phosphorylase (UPP) is a key enzyme of pyrimidine salvage pathways, catalyzing the reversible phosphorolysis of ribosides of uracil to nucleobases and ribose 1-phosphate. It is also a critical enzyme in the activation of pyrimidine-based chemotherapeutic compounds such a 5-fluorouracil (5-FU) and its prodrug capecitabine. Additionally, an elevated level of this enzyme in certain tumours is believed to contribute to the selectivity of such drugs. However, the clinical effectiveness of these fluoropyrimidine antimetabolites is hampered by their toxicity to normal tissue. In response to this limitation, specific inhibitors of UPP, such as 5-benzylacyclouridine (BAU), have been developed and investigated for their ability to modulate the cytotoxic side effects of 5-FU and its derivatives, so as to increase the therapeutic index of these agents. RESULTS: In this report we present the high resolution structures of human uridine phosphorylase 1 (hUPP1) in ligand-free and BAU-inhibited conformations. The structures confirm the unexpected solution observation that the human enzyme is dimeric in contrast to the hexameric assembly present in microbial UPPs. They also reveal in detail the mechanism by which BAU engages the active site of the protein and subsequently disables the enzyme by locking the protein in a closed conformation. The observed inter-domain motion of the dimeric human enzyme is much greater than that seen in previous UPP structures and may result from the simpler oligomeric organization. CONCLUSION: The structural details underlying hUPP1's active site and additional surfaces beyond these catalytic residues, which coordinate binding of BAU and other acyclouridine analogues, suggest avenues for future design of more potent inhibitors of this enzyme. Notably, the loop forming the back wall of the substrate binding pocket is conformationally different and substantially less flexible in hUPP1 than in previously studied microbial homologues. These distinctions can be utilized to discover novel inhibitory compounds specifically optimized for efficacy against the human enzyme as a step toward the development of more effective chemotherapeutic regimens that can selectively protect normal tissues with inherently lower UPP activity.


Subject(s)
Enzyme Inhibitors/metabolism , Uracil/analogs & derivatives , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Crystallization , Dimerization , Drug Design , Enzyme Inhibitors/chemistry , Escherichia coli/genetics , Humans , Protein Binding , Protein Conformation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein , Uracil/chemistry , Uracil/metabolism , Uridine Phosphorylase/antagonists & inhibitors , Uridine Phosphorylase/genetics
20.
Hum Mol Genet ; 18(4): 714-22, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19028666

ABSTRACT

Replication and repair of DNA require equilibrated pools of deoxynucleoside triphosphate precursors. This concept has been proven by in vitro studies over many years, but in vivo models are required to demonstrate its relevance to multicellular organisms and to human diseases. Accordingly, we have generated thymidine phosphorylase (TP) and uridine phosphorylase (UP) double knockout (TP(-/-)UP(-/-)) mice, which show severe TP deficiency, increased thymidine and deoxyuridine in tissues and elevated mitochondrial deoxythymidine triphosphate. As consequences of the nucleotide pool imbalances, brains of mutant mice developed partial depletion of mtDNA, deficiencies of respiratory chain complexes and encephalopathy. These findings largely account for the pathogenesis of mitochondrial neurogastrointestinal encephalopathy (MNGIE), the first inherited human disorder of nucleoside metabolism associated with somatic DNA instability.


Subject(s)
DNA, Mitochondrial/chemistry , Deoxyribonucleotides/metabolism , Genomic Instability , Purine-Pyrimidine Metabolism, Inborn Errors/metabolism , Thymidine Phosphorylase/deficiency , Uridine Phosphorylase/metabolism , Animals , Brain/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Thymidine Phosphorylase/genetics , Uridine Phosphorylase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...