Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Nutr ; 147(4): 482-498, 2017 04.
Article in English | MEDLINE | ID: mdl-28250194

ABSTRACT

Background: Previously, we determined that heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) functions as an intracellular physiologic sensor of folate deficiency. In this model, l-homocysteine, which accumulates intracellularly in proportion to the extent of folate deficiency, covalently binds to and thereby activates homocysteinylated hnRNP-E1 to interact with folate receptor-α mRNA; this high-affinity interaction triggers the translational upregulation of cell surface folate receptors, which enables cells to optimize folate uptake from the external milieu. However, integral to this model is the need for ongoing generation of hnRNP-E1 to replenish homocysteinylated hnRNP-E1 that is degraded.Objective: We searched for an interrelated physiologic mechanism that could also maintain the steady-state concentration of hnRNP-E1 during prolonged folate deficiency.Methods: A novel RNA-protein interaction was functionally characterized by using molecular and biochemical approaches in vitro and in vivo.Results: l-homocysteine triggered a dose-dependent high-affinity interaction between hnRNP-E1 and a 25-nucleotide cis element within the 5'-untranslated region of hnRNP-E1 mRNA; this led to a proportionate increase in these RNA-protein complexes, and translation of hnRNP-E1 both in vitro and within placental cells. Targeted perturbation of this RNA-protein interaction either by specific 25-nucleotide antisense oligonucleotides or mutation within this cis element or by small interfering RNA to hnRNP-E1 mRNA significantly reduced cellular biosynthesis of hnRNP-E1. Conversely, transfection of hnRNP-E1 mutant proteins that mimicked homocysteinylated hnRNP-E1 stimulated both cellular hnRNP-E1 and folate receptor biosynthesis. In addition, ferrous sulfate heptahydrate [iron(II)], which also binds hnRNP-E1, significantly perturbed this l-homocysteine-triggered RNA-protein interaction in a dose-dependent manner. Finally, folate deficiency induced dual upregulation of hnRNP-E1 and folate receptors in cultured human cells and tumor xenografts, and more selectively in various fetal tissues of folate-deficient dams.Conclusions: This novel positive feedback loop amplifies hnRNP-E1 during prolonged folate deficiency and thereby maximizes upregulation of folate receptors in order to restore folate homeostasis toward normalcy in placental cells. It will also functionally impact several other mRNAs of the nutrition-sensitive, folate-responsive posttranscriptional RNA operon that is orchestrated by homocysteinylated hnRNP-E1.


Subject(s)
Folate Receptor 2/metabolism , Folic Acid Deficiency/metabolism , Gene Expression Regulation/drug effects , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Placenta/cytology , Up-Regulation/drug effects , Animals , Cell Line , DNA-Binding Proteins , Female , Folate Receptor 2/genetics , Folic Acid/pharmacology , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Uterine Cervical Neoplasms/metabolism
2.
Bioorg Med Chem Lett ; 24(22): 5304-9, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25304896

ABSTRACT

We synthesized a series of serum-stable covalently linked drugs derived from 3'-C-methyladenosine (3'-Me-Ado) and valproic acid (VPA), which are ribonucleotide reductase (RR) and histone deacetylase (HDAC) inhibitors, respectively. While the combination of free VPA and 3'-Me-Ado resulted in a clear synergistic apoptotic effect, the conjugates had lost their HDAC inhibitory effect as well as the corresponding apoptotic activity. Two of the analogs, 2',5'-bis-O-valproyl-3'-C-methyladenosine (A160) and 5'-O-valproyl-3'-C-methyladenosine (A167), showed promising cytotoxic activities against human hematological and solid cancer cell lines. A167 was less potent than A160 but had interesting features as an RR inhibitor. It inhibited RR activity by competing with ATP as an allosteric effector and concomitantly reduced the intracellular deoxyribonucleoside triphosphate (dNTP) pools. A167 represents a novel lead compound, which in contrast to previously used RR nucleoside analogs does not require intracellular kinases for its activity and therefore holds promise against drug resistant tumors with downregulated nucleoside kinases.


Subject(s)
Adenosine/analogs & derivatives , Enzyme Inhibitors/chemical synthesis , Ribonucleotide Reductases/antagonists & inhibitors , Valproic Acid/chemistry , Adenosine/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Esters/chemistry , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Kinetics , Ribonucleotide Reductases/metabolism
3.
J Vasc Interv Radiol ; 24(6): 894-900, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23566523

ABSTRACT

PURPOSE: This pilot trial assesses variability of apoptosis and response 1 day after hepatic intraarterial (IA) benzamide riboside (BR) in rodent hepatomas and its correlation to water apparent diffusion coefficient (ADC) and single-quantum (SQ) and triple-quantum-filtered (TQF) sodium-23 ((23)Na) magnetic resonance (MR) imaging. MATERIALS AND METHODS: Sprague-Dawley rats (n = 8) were inoculated with 10(6) N1-S1 cells. IA BR (20 mg/kg) was infused after 14 days. Animals were killed 1 day (n = 4) or 21 days (n = 4) after therapy. Imaging was performed 1 day before and after treatment. Volume was assessed over 2 weeks. Percentage apoptosis was counted from terminal deoxynucleotidyl transferase dUTP nick-end labeling-stained slides at 400×magnification. Kruskal-Wallis tests were used to compare apoptosis, and Wilcoxon signed-rank tests were used to compare MR signal intensity (SI). RESULTS: Apoptosis was marginally greater in tumor than in nontumor (6.7% vs 1.3%; P = .08), varying from 2% to 10%. Before treatment, MR SI was greater in tumor than in nontumor (ADC, 1.18 vs 0.76 [P = .0078]; SQ, 1.20 vs 1.04 [P = .03]; TQF, 0.55 vs 0.34 [P = .03]). After treatment, tumors increased in volume (0.62 vs 0.33; P = .016) variably over 2 weeks. MR SI remained greater in tumor than in nontumor (ADC, 1.20 vs 0.77 [P = .0078]; SQ, 1.76 vs 1.15 [P = .016]; TQF, 0.84 vs 0.49 [P = .03]). SQ and TQF SI increased by 47% (P = .016) and 53% (P = .016) in tumors, whereas ADC did not change. CONCLUSIONS: Apoptosis was marginal and varied from 2% to 10%. Water ADC, SQ, and TQF MR imaging distinguished tumor from nontumor. Changes in water ADC and sodium MR imaging correlated to apoptosis and volume in select cases, but additional animals are needed to validate this trend against tumor growth.


Subject(s)
Apoptosis/drug effects , Body Water/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Magnetic Resonance Imaging/methods , Nucleosides/therapeutic use , Sodium/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Statistics as Topic , Treatment Outcome
4.
J Biol Chem ; 287(15): 12559-77, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22351779

ABSTRACT

Although HPV16 transforms infected epithelial tissues to cancer in the presence of several co-factors, there is insufficient molecular evidence that poor nutrition has any such role. Because physiological folate deficiency led to the intracellular homocysteinylation of heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) and activated a nutrition-sensitive (homocysteine-responsive) posttranscriptional RNA operon that included interaction with HPV16 L2 mRNA, we investigated the functional consequences of folate deficiency on HPV16 in immortalized HPV16-harboring human (BC-1-Ep/SL) keratinocytes and HPV16-organotypic rafts. Although homocysteinylated hnRNP-E1 interacted with HPV16 L2 mRNA cis-element, it also specifically bound another HPV16 57-nucleotide poly(U)-rich cis-element in the early polyadenylation element (upstream of L2L1 genes) with greater affinity. Together, these interactions led to a profound reduction of both L1 and L2 mRNA and proteins without effects on HPV16 E6 and E7 in vitro, and in cultured keratinocyte monolayers and HPV16-low folate-organotypic rafts developed in physiological low folate medium. In addition, HPV16-low folate-organotypic rafts contained fewer HPV16 viral particles, a similar HPV16 DNA viral load, and a much greater extent of integration of HPV16 DNA into genomic DNA when compared with HPV16-high folate-organotypic rafts. Subcutaneous implantation of 18-day old HPV16-low folate-organotypic rafts into folate-replete immunodeficient mice transformed this benign keratinocyte-derived raft tissue into an aggressive HPV16-induced cancer within 12 weeks. Collectively, these studies establish a likely molecular linkage between poor folate nutrition and HPV16 and predict that nutritional folate and/or vitamin-B(12) deficiency, which are both common worldwide, will alter the natural history of HPV16 infections and also warrant serious consideration as reversible co-factors in oncogenic transformation of HPV16-infected tissues to cancer.


Subject(s)
Cell Transformation, Viral , Folic Acid Deficiency , Human papillomavirus 16/physiology , Keratinocytes/virology , Neoplasms, Experimental/virology , Papillomavirus Infections/virology , Animals , Base Sequence , Capsid Proteins/genetics , Capsid Proteins/metabolism , Catalase/biosynthesis , Catalase/genetics , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins , Female , Folic Acid/metabolism , Genes, Reporter , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Homocysteine/chemistry , Homocysteine/metabolism , Host-Pathogen Interactions , Human papillomavirus 16/genetics , Humans , Keratinocytes/metabolism , Keratinocytes/transplantation , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms, Experimental/pathology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Protein Binding , Proteolysis , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA-Binding Proteins , Sulfhydryl Compounds/metabolism , Tumor Burden , Virus Integration
5.
J Biol Chem ; 286(45): 39100-15, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21930702

ABSTRACT

The mechanism underlying the sensing of varying degrees of physiological folate deficiency, prior to adaptive optimization of cellular folate uptake through the translational up-regulation of folate receptors (FR) is unclear. Because homocysteine, which accumulates intracellularly during folate deficiency, stimulated interactions between heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) and an 18-base FR-α mRNA cis-element that led to increased FR biosynthesis and net up-regulation of FR at cell surfaces, hnRNP-E1 was a plausible candidate sensor of folate deficiency. Accordingly, using purified components, we evaluated the physiological basis whereby L-homocysteine triggered these RNA-protein interactions to stimulate FR biosynthesis. L-homocysteine induced a concentration-dependent increase in RNA-protein binding affinity throughout the range of physiological folate deficiency, which correlated with a proportionate increase in translation of FR in vitro and in cultured human cells. Targeted reduction of newly synthesized hnRNP-E1 proteins by siRNA to hnRNP-E1 mRNA reduced both constitutive and L-homocysteine-induced rates of FR biosynthesis. Furthermore, L-homocysteine covalently bound hnRNP-E1 via multiple protein-cysteine-S-S-homocysteine mixed disulfide bonds within K-homology domains known to interact with mRNA. These data suggest that a concentration-dependent, sequential disruption of critical cysteine-S-S-cysteine bonds by covalently bound L-homocysteine progressively unmasks an underlying RNA-binding pocket in hnRNP-E1 to optimize interaction with FR-α mRNA cis-element preparatory to FR up-regulation. Collectively, such data incriminate hnRNP-E1 as a physiologically relevant, sensitive, cellular sensor of folate deficiency. Because diverse mammalian and viral mRNAs also interact with this RNA-binding domain with functional consequences to their protein expression, homocysteinylated hnRNP-E1 also appears well positioned to orchestrate a novel, nutrition-sensitive (homocysteine-responsive), posttranscriptional RNA operon in folate-deficient cells.


Subject(s)
Folate Receptor 1/biosynthesis , Folic Acid Deficiency/metabolism , Folic Acid/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Homocysteine/metabolism , Protein Binding , DNA-Binding Proteins , Disulfides/metabolism , Folate Receptor 1/genetics , Folic Acid Deficiency/genetics , HeLa Cells , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Homocysteine/genetics , Humans , Protein Structure, Tertiary , RNA, Messenger , RNA-Binding Proteins , Up-Regulation
6.
Anticancer Res ; 31(6): 2045-51, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21737621

ABSTRACT

AIM: To monitor the effects of the apoptotic agent benzamide riboside (BR) on tumor volume and water apparent diffusion coefficient (ADC) in rat hepatocellular carcinoma (HCC). MATERIALS AND METHODS: Water ADC of the tumors and nearby liver tissue was measured using diffusion-weighted 1H MRI (DWI). The two groups of BR-treated animals, which differed in their sensitivity to the treatment, were identified as responsive (RBR) and non-responsive (NRBR). RESULTS: Tumor growth in the RBR group was arrested and the mean tumor volume in this group was 1/6th and 1/16th compared to that of the NRBR group on days 7 and 14 after treatment, respectively. Water ADC of HCC was higher than in nearby normal liver tissue. Before BR treatment, the mean water ADC was significantly higher in the RBR group compared to the NRBR group. BR therapy did not change the water ADC value regardless of tumor sensitivity. CONCLUSION: Although the water ADC did not change after chemotherapy by BR, DWI has great potential for detecting and predicting response to chemotherapy in HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Nucleosides/pharmacology , Water/metabolism , Animals , Cell Line, Tumor , Diffusion , Hepatic Artery , Infusions, Intra-Arterial , Liver Neoplasms, Experimental/pathology , Magnetic Resonance Angiography/methods , Male , Rats , Rats, Sprague-Dawley , Water/analysis
7.
Bioorg Med Chem ; 19(5): 1594-605, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21324702

ABSTRACT

Cofactor-type inhibitors of inosine monophosphate dehydrogenase (IMPDH) that target the nicotinamide adenine dinucleotide (NAD) binding domain of the enzyme are modular in nature. They interact with the three sub-sites of the cofactor binding domain; the nicotinamide monophosphate (NMN) binding sub-site (N sub-site), the adenosine monophosphate (AMP) binding sub-site (A sub-site), and the pyrophosphate binding sub-site (P sub-site or P-groove). Mycophenolic acid (MPA) shows high affinity to the N sub-site of human IMPDH mimicking NMN binding. We found that the attachment of adenosine to the MPA through variety of linkers afforded numerous mycophenolic adenine dinucleotide (MAD) analogues that inhibit the two isoforms of the human enzyme in low nanomolar to low micromolar range. An analogue 4, in which 2-ethyladenosine is attached to the mycophenolic alcohol moiety through the difluoromethylenebis(phosphonate) linker, was found to be a potent inhibitor of hIMPDH1 (K(i)=5 nM), and one of the most potent, sub-micromolar inhibitor of leukemia K562 cells proliferation (IC(50)=0.45 µM). Compound 4 was as potent as Gleevec (IC(50)=0.56 µM) heralded as a 'magic bullet' against chronic myelogenous leukemia (CML). MAD analogues 7 and 8 containing an extended ethylenebis(phosphonate) linkage showed low nanomolar inhibition of IMPDH and low micromolar inhibition of K562 cells proliferation. Some novel MAD analogues described herein containing linkers of different length and geometry were found to inhibit IMPDH with K(i)'s lower than 100 nM. Thus, such linkers can be used for connection of other molecular fragments with high affinity to the N- and A-sub-site of IMPDH.


Subject(s)
Diphosphates/metabolism , Drug Design , Enzyme Inhibitors/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , Binding Sites , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , IMP Dehydrogenase/chemistry , IMP Dehydrogenase/metabolism , Inhibitory Concentration 50 , K562 Cells , Models, Molecular , Molecular Structure
8.
Eur J Med Chem ; 46(5): 1499-504, 2011 May.
Article in English | MEDLINE | ID: mdl-21349610

ABSTRACT

A series of N6-aminopurine-9-ß-D-ribonucleosides and ribose-modified 3'-C-methyl analogues substituted at N6-position with a small group like hydroxy, methoxy or amino group or at C2(N6) position have been synthesized and tested against a panel of human leukemia and carcinoma cell lines. N6-Hydrazino-9-ß-D-ribofuranosyl-purine (5) displayed the best antiproliferative activity in the low micromolar or submicromolar range against all tested tumor cell lines. The activity of this nucleoside is related in part to ribonucleotide reductase inhibition. C2-modification or 3'-C-methylation in N6-substituted adenosine analogues leads to a decrease or loss in activity.


Subject(s)
Adenine/chemistry , Antineoplastic Agents/pharmacology , Ribonucleosides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Caco-2 Cells , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Molecular Structure , Recombinant Proteins/metabolism , Ribonucleosides/chemical synthesis , Ribonucleosides/chemistry , Ribonucleotide Reductases/metabolism , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
9.
Med Chem ; 7(6): 718-26, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22313312

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is synthesized by the action of nicotinamide mononucleotide adenylyltransferase (NMNAT) from NMN and ATP. The mouse homolog of NMNAT-2 (mmNMNAT-2) was cloned, expressed, and subsequently identified using MALDI-TOF in conjunction with the ProFound database. Circular dichroism analyses of recombinant mmNMNAT-2 showed α helical and ß sheet secondary structures, consistent with the known structure of the human isoform. Competition experiments using mouse pancreatic tissue lysates with recombinant mmNMNAT-2 demonstrated that the activity of the expressed protein was similar to the human isoform. Immunohistochemistry of mouse embryonic tissues with hNMNAT-2 also showed a tissue- and cellular-specific expression of this isoform. Therefore, our studies demonstrate for the first time the clear biological evidence for the existence of a mouse isoform of hNMNAT-2. These studies may help in future investigations aimed at understanding the regulation of this gene and its pathway, and in turn, will spur the development of novel therapies for diseases such as cancer and diabetes since mice are the most frequently used experimental system for in vivo studies.


Subject(s)
Cloning, Molecular , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Animals , Circular Dichroism , Humans , Immunohistochemistry , Mice , NAD/biosynthesis , NAD/chemistry , Nicotinamide-Nucleotide Adenylyltransferase/chemistry , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Bioorg Med Chem ; 18(16): 5950-64, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20650640

ABSTRACT

Small molecules that act on multiple biological targets have been proposed to combat the drug resistance commonly observed for cancer chemotherapy. By combining the structural features of known inhibitors of inosine monophosphate dehydrogense (IMPDH) and histone deacetylase (HDAC), dual inhibitors of IMPDH and HDAC based on the scaffold of cinnamic hydroxamic acid (CHA) have been designed, synthesized, and evaluated in biological assays. Key features, including the linker length, linker functionality, substitution position, and interacting groups, have been explored. Their individual contribution to the inhibitory activities against human IMPDH1 and IMPDH2 as well as HDAC has been assessed.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cinnamates/chemical synthesis , Cinnamates/chemistry , Cinnamates/pharmacology , Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , IMP Dehydrogenase/metabolism , Models, Molecular
11.
Arch Biochem Biophys ; 485(1): 10-5, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19236843

ABSTRACT

BACKGROUND: AMP-dependent protein kinase (AMPK) and peroxisome proliferator-activated receptor (PPAR) alpha facilitate fatty acid oxidation. We have shown that treatment of hepatoma cells with ethanol or feeding ethanol-containing diets to mice inhibited both PPARalpha and AMPK activity. Importantly, WY-14,643 reversed the development of fatty liver in alcohol-fed mice. Whether WY-14,643, a PPARalpha agonist, has any effects on AMPK is not known. The aim of this study was to investigate the effect of WY-14,643 on AMPK activity. METHODS: The effect of WY-14,643 on AMPK phosphorylation and activity were examined in rat hepatoma cells (H4IIEC3). The effect of WY-14,643 on upstream kinases of AMPK, PKC-zeta/LKB1, intracellular AMP:ATP ratio, oxidative stress, and AMPK gene expression were studied. RESULTS: Treatment of the H4IIEC3 cells with WY-14,643 for 24h led to 60% increase in the phosphorylation of AMPK. The effect of WY-14,643 on AMPK phosphorylation is PKC-zeta/LKB1 independent. WY-14,643 did not alter the levels of intracellular AMP:ATP ratio and it did not increase the levels of reactive oxygen species at 24-h of treatment. WY-14,643-induced AMPK alpha subunit expression by 2- to 2.5-fold, but there was no change in AMPKalpha subunit protein at 24h. The effect of WY-14,643 on AMPK phosphorylation did not altered by the presence of an NADPH oxidase inhibitor. CONCLUSIONS: WY-14,643 induced AMPKalpha subunit phosphorylation and the activity of the enzyme. This was associated with induction of AMPKalpha1 and alpha2 mRNA, but the mechanism for this activation is uncertain.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Pyrimidines/pharmacology , AMP-Activated Protein Kinases/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Cell Line , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , PPAR alpha/metabolism , Phosphorylation/drug effects , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism
12.
J Vasc Interv Radiol ; 20(3): 410-4, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19167243

ABSTRACT

The present study was undertaken to assess the technical feasibility of transfemoral hepatic artery catheterization in rats and to describe the imaging techniques that can be used on tumors in rats. A total of 106 N1-S1 cells were inoculated into the left lobes of 74 rats. In 17, transfemoral angiography was attempted. Tumor volumes for 2 weeks before angiography were measured with magnetic resonance imaging in 40 animals. Tumors grew in 63 animals. Angiography was successful in 16 rats. Mean tumor volumes were 0.13 mL and 0.9 mL after 1 and 2 weeks, respectively. In conclusion, transfemoral hepatic artery catheterization is feasible in this animal model.


Subject(s)
Angiography/methods , Disease Models, Animal , Embolization, Therapeutic/methods , Hepatic Artery/surgery , Liver Neoplasms/diagnosis , Liver Neoplasms/therapy , Animals , Cell Line, Tumor , Feasibility Studies , Humans , Rats , Treatment Outcome
14.
Am J Physiol Gastrointest Liver Physiol ; 295(6): G1173-81, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18832448

ABSTRACT

AMP-activated protein kinase (AMPK) responds to oxidative stress. Previous work has shown that ethanol treatment of cultured hepatoma cells and of mice inhibited the activity of AMPK and reduced the amount of AMPK protein. Ethanol generates oxidative stress in the liver. Since AMPK is activated by reactive oxygen species, it seems paradoxical that ethanol would inhibit AMPK in the hepatoma cells. In an attempt to understand the mechanism whereby ethanol inhibits AMPK, we studied the effect of ethanol on AMPK activation by exogenous hydrogen peroxide. The effects of ethanol, hydrogen peroxide, and inhibitors of protein phosphatase 2A (PP2A) [either okadaic acid or PP2A small interference RNA (siRNA)] on AMPK phosphorylation and activity were examined in rat hepatoma cells (H4IIEC3) and HeLa cells. In H4IIEC3 cells, hydrogen peroxide (H(2)O(2), 1 mM) transiently increased the level of phospho-AMPK to 1.5-fold over control (P < 0.05). Similar findings were observed in HeLa cells, which do not express the upstream AMPK kinase, LKB1. H(2)O(2) markedly increased the phosphorylation of LKB1 in H4IIEC3 cells. Ethanol significantly inhibited the phosphorylation of PKC-zeta, LKB1, and AMPK caused by exposure to H(2)O(2). This inhibitory effect of ethanol required its metabolism. More importantly, the inhibitory effects of ethanol on H(2)O(2)-induced AMPK phosphorylation were attenuated by the presence of the PP2A inhibitor, okadaic acid, or PP2A siRNA. The inhibitory effect of ethanol on AMPK phosphorylation is exerted through the inhibition of PKC-zeta and LKB1 phosphorylation and the activation of PP2A.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Ethanol/pharmacology , Hydrogen Peroxide/pharmacology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Enzyme Activation/drug effects , HeLa Cells , Humans , Liver Neoplasms, Experimental , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Tumor Cells, Cultured
15.
Bioorg Med Chem ; 16(20): 9340-5, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18809333

ABSTRACT

Mycophenolic acid (MPA), a clinically used immunosuppressant, is extensively metabolized into an inactive C7-glucuronide and removed from circulation. To circumvent the metabolic liability imposed by the C7-hydroxyl group, we have designed a series of hybrid MPA analogs based on the pharmacophores present in MPA and new generations of inosine monophosphate dehydrogenase (IMPDH) inhibitors. The synthesis of MPA analogs has been accomplished by an allylic substitution of a common lactone. Biological evaluations of these analogs and a preliminary structure-activity relationship (SAR) are presented.


Subject(s)
Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/metabolism , Computer Simulation , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Mycophenolic Acid/chemical synthesis , Mycophenolic Acid/chemistry
16.
J Med Chem ; 51(14): 4260-9, 2008 Jul 24.
Article in English | MEDLINE | ID: mdl-18588281

ABSTRACT

A series of cycloalkyl, bicycloalkyl, aryl, and heteroaryl N (6)-substituted derivatives of the antitumor agent 3'- C-methyladenosine (3'-Me-Ado), an inhibitor of the alpha Rnr1 subunit of mammalian ribonucleotide reductase (RR), were synthesized. The cytotoxicity of these compounds was evaluated against a panel of human leukemia and carcinoma cell lines and compared to that of some corresponding N (6)-substituted adenosine analogues. N (6)-cycloalkyl-3'- C-methylribonucleosides 2- 7 and N (6)-phenyl analogue 8 were found to inhibit the proliferation of K562 leukemia cells. N (6)-(+/-)- endo-2-norbornyl-3'- C-methyladenosine ( 7) was found to be the most cytotoxic compound, with GI 50 values slightly higher than that of 3'-Me-Ado against K562 and carcinoma cell lines and 2.7 fold higher cytotoxicity against human promyelocytic leukemia HL-60 cells. The SAR study confirms that an unsubstituted N (6)-amino group is essential for optimal cytotoxicity of 3'-Me-Ado against both K562 and carcinoma cell lines. Computational studies, carried out on the eukaryotic alpha subunit (Rnr1) of RR from Saccharomyces cerevisiae were performed to rationalize the observed structure-activity relationships.


Subject(s)
Adenosine/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Ribonucleotide Reductases/antagonists & inhibitors , Ribose/chemistry , Adenosine/chemical synthesis , Adenosine/chemistry , Adenosine/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Humans , Structure-Activity Relationship
18.
J Med Chem ; 50(26): 6685-91, 2007 Dec 27.
Article in English | MEDLINE | ID: mdl-18038969

ABSTRACT

Mycophenolic acid (MPA), an inhibitor of IMP-dehydrogenase (IMPDH), is used worldwide in transplantation. Recently, numerous studies showed its importance in cancer treatment. Consequently, MPA entered clinical trials in advanced multiple myeloma patients. Suberoylanilide hydroxamic acid (SAHA), a potent differentiation agent acting through inhibition of histone deacetylases (HDACs), was recently approved for treatment of cutaneous T cell lymphoma. We report herein the synthesis of dual inhibitors of IMPDH and HDACs. We found that mycophenolic hydroxamic acid (9, MAHA) inhibits both IMPDH (Ki=30 nM) and HDAC (IC50=5.0 microM). A modification of SAHA with groups known to interact with IMPDH afforded a SAHA analogue 14, which inhibits IMPDH (Ki=1.7 microM) and HDAC (IC50=0.06 microM). Both MAHA (IC50=4.8 microM) and SAHA analogue 14 (IC50=7.7 microM) were more potent than parent compounds as antiproliferation agents. They were also significantly more potent as differentiation inducers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Differentiation , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Histone Deacetylases/chemistry , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , IMP Dehydrogenase/chemistry , K562 Cells , Mycophenolic Acid/chemistry , Mycophenolic Acid/pharmacology , Structure-Activity Relationship , Vorinostat
19.
J Med Chem ; 50(23): 5743-51, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17958343

ABSTRACT

Novel tiazofurin adenine dinucleotide (TAD) analogues 25-33 containing a substituent at C2 of the adenine ring have been synthesized as inhibitors of the two isoforms of human IMP-dehydrogenase. The 2-ethyl TAD analogue 33 [Ki = 1 nM (type I), Ki = 14 nM (type II)] was found to be the most potent. It did not inhibit three other cellular dehydrogenases up to 50 microM. Mycophenolic adenine bis(phosphonate)s containing a 2-phenyl (37) or 2-ethyl group (38), were prepared as metabolically stable compounds, both nanomolar inhibitors. Compound 38 [Ki = 16 nM (type I), Ki = 38 nM (type II)] inhibited proliferation of leukemic K562 cells (IC50 = 1.1 microM) more potently than tiazofurin (IC50 = 12.4 microM) or mycophenolic acid (IC50 = 7.7 microM).


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Antineoplastic Agents/chemical synthesis , Diphosphonates/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , NAD/analogs & derivatives , NAD/chemical synthesis , Adenosine Monophosphate/chemical synthesis , Adenosine Monophosphate/pharmacology , Antineoplastic Agents/pharmacology , Diphosphonates/pharmacology , Drug Screening Assays, Antitumor , Humans , IMP Dehydrogenase/chemistry , IMP Dehydrogenase/metabolism , Isoenzymes/metabolism , K562 Cells , Models, Molecular , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/chemical synthesis , Mycophenolic Acid/pharmacology , NAD/pharmacology , Protein Binding , Ribavirin/analogs & derivatives , Ribavirin/chemical synthesis , Ribavirin/pharmacology
20.
Bioorg Med Chem Lett ; 17(11): 3152-5, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17395461

ABSTRACT

A methylenebis(sulfonamide) linked NAD analogue has been designed to circumvent the metabolically unstable, ionic nature of the natural pyrophosphate linkage. This NAD analogue is assembled through two Mitsunobu reactions of a methylenebis(sulfonamide) linker with two protected nucleosides. A 2,4-dimethoxybenzyl group is used as a sulfonamide protective group, which allows facile removal under mildly acidic conditions. This NAD analogue inhibits IMPDH at low micromolar concentration.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Humans , Molecular Structure , NAD/chemistry , Sulfonamides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...