Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Cutan Ocul Toxicol ; 43(1): 87-96, 2024 Mar.
Article En | MEDLINE | ID: mdl-38127818

PURPOSE: Skin exposure to noxious agents leads to cutaneous lesion marked by an increase in inflammation, cellular proliferation, and hyperplasiogenic reactions. Studies have demonstrated that these damages breach the skin integrity resulting in the aetiology of various cutaneous disorders like atopic dermatitis, eczema, psoriasis, and development of non-melanoma skin cancer. Celecoxib, a cyclooxygenase-2 (COX-2) inhibitor, is an effective treatment for a variety of inflammatory diseases. Its importance in the therapy of skin problems, however, remains under appreciated. METHODS: We tested efficacy of topically applied celecoxib in mitigating skin inflammation, cellular proliferation, and hyperplasia induced by the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) in Swiss albino mice. RESULTS: Celecoxib (5 and 10 µmol) markedly reduced TPA (10 nmol) induced prostaglandin E2 (PGE2) production, oedema formation, myeloperoxidase (MPO) activity, and levels of pro-inflammatory cytokines such as tumour necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), and interleukin-6 (IL-6). It also resulted in a considerable decrease in ornithine decarboxylase (ODC) activity and the incorporation of [3H]-thymidine into DNA. In addition, there was a significant reduction in histoarchitectural abnormalities such as epidermal thickness, number of epidermal cell layers, neutrophil infiltration, intercellular oedema, and vasodilation. CONCLUSION: Our results demonstrate that topical celecoxib can reduce the inflammation, hyperproliferation, and hyperplasiogenic events of skin insults suggesting that it may prove to be a valuable management option for cutaneous lesion and associated illnesses such as atopic dermatitis, eczema, and psoriasis, as well as the emergence of non-melanoma cancer.


Dermatitis, Atopic , Eczema , Psoriasis , Skin Diseases , Skin Neoplasms , Mice , Animals , Celecoxib/adverse effects , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/pharmacology , Skin , Tetradecanoylphorbol Acetate/toxicity , Tetradecanoylphorbol Acetate/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Skin Diseases/pathology , Psoriasis/pathology , Edema/metabolism , Acetates/adverse effects , Acetates/metabolism , Eczema/metabolism , Eczema/pathology , Skin Neoplasms/pathology
2.
J Mol Recognit ; 36(7): e3021, 2023 07.
Article En | MEDLINE | ID: mdl-37092713

Visceral leishmaniasis (VL) is caused by Leishmania donovani (Ld), and most cases occur in Brazil, East Africa, and India. The treatment for VL is limited and has many adverse effects. The development of safer and more efficacious drugs is urgently needed. Drug repurposing is one of the best processes to repurpose existing drugs. Ornithine decarboxylase (ODC) is an important target against L. donovani in the polyamine biosynthesis pathway. In this study, we have modeled the 3D structure of ODC and performed high-throughput virtual screening of 8630 ZINC database ligands against Leishmania donovani ornithine decarboxylase (Ld ODC), selecting 45 ligands based on their high binding score. It is further validated through molecular docking simulation and the selection of the top two lead molecules (ceftaroline fosamil and rimegepant) for Molecular Dynamics (MD) simulation, Density functional theory (DFT), and molecular mechanics generalized born surface area (MMGBSA) analysis. The results showed that the binding affinities of ceftaroline fosamil, and rimegepant are, respectively, -10.719 and 10.159 kcal/mol. The docking complexes of the two lead compounds, ceftaroline fosamil, and rimegepant, with the target ODC, were found stable during molecular dynamics simulations. Furthermore, the analysis of MMGBSA revealed that these compounds had a high binding free energy. The DFT analysis showed that the top lead molecules were more reactive than the standard drug (pentamidine). In-silico findings demonstrated that ceftaroline fosamil, and rimegepant might be recognized as potent antagonists against ODC for the treatment of VL.


Leishmania donovani , Leishmaniasis, Visceral , Humans , Ornithine Decarboxylase Inhibitors/chemistry , Ornithine Decarboxylase Inhibitors/pharmacology , Drug Repositioning , Molecular Docking Simulation , Ornithine Decarboxylase/chemistry , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/pharmacology , Ligands , Leishmania donovani/metabolism , Ceftaroline
3.
Cancer Lett ; 559: 216120, 2023 04 10.
Article En | MEDLINE | ID: mdl-36893894

A key mechanism driving colorectal cancer (CRC) development is the upregulation of MYC and its targets, including ornithine decarboxylase (ODC), a master regulator of polyamine metabolism. Elevated polyamines promote tumorigenesis in part by activating DHPS-mediated hypusination of the translation factor eIF5A, thereby inducing MYC biosynthesis. Thus, MYC, ODC and eIF5A orchestrate a positive feedback loop that represents an attractive therapeutic target for CRC therapy. Here we show that combined inhibition of ODC and eIF5A induces a synergistic antitumor response in CRC cells, leading to MYC suppression. We found that genes of the polyamine biosynthesis and hypusination pathways are significantly upregulated in colorectal cancer patients and that inhibition of ODC or DHPS alone limits CRC cell proliferation through a cytostatic mechanism, while combined ODC and DHPS/eIF5A blockade induces a synergistic inhibition, accompanied to apoptotic cell death in vitro and in mouse models of CRC and FAP. Mechanistically, we found that this dual treatment causes complete inhibition of MYC biosynthesis in a bimodal fashion, by preventing translational elongation and initiation. Together, these data illustrate a novel strategy for CRC treatment, based on the combined suppression of ODC and eIF5A, which holds promise for the treatment of CRC.


Colorectal Neoplasms , Peptide Initiation Factors , Polyamines , Proto-Oncogene Proteins c-myc , Animals , Mice , Apoptosis , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/pharmacology , Polyamines/metabolism , Humans , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Eukaryotic Translation Initiation Factor 5A
4.
J Oleo Sci ; 71(10): 1481-1492, 2022 Sep 30.
Article En | MEDLINE | ID: mdl-36089399

Renal tissue plays a crucial function in maintaining homeostasis, making it vulnerable to xenobiotic toxicity. Pueraria montana has more beneficial potential against the various diseases and has long history used as a traditional Chinese medicine. But its effect against the renal cancer not scrutinize. The goal of this study is to see if Pueraria montana can protect rats from developing kidney tumors caused by diethylnitrosamine (DEN) and ferric nitrite (Fe-NTA). Wistar rats was selected for the current study and DEN (use as an inducer) and Fe-NTA (promoter) for induction the renal cancer. For 22 weeks, the rats were given orally Pueraria montana (12.5, 25, and 50 mg/kg) treatment. At regular intervals, the body weight and food intake were calculated. The rats were macroscopically evaluated for identification of cancer in the renal tissue. The renal tumor makers, renal parameters, antioxidant enzymes, phase I and II enzymes, inflammatory cytokines and mediators were estimated at end of the experimental study. Pueraria montana treated rats displayed the suppression of renal tumors, incidence of the tumors along with suppression of tumor percentage. Pueraria montana treated rats significantly (p < 0.001) increased body weight and suppressed the renal weight and food intake. It also reduced the level of renal tumor marker ornithine decarboxylase (ODC) and [3H] thymidine incorporation along with suppression of renal parameter such as uric acid, blood urea nitrogen (BUN), urea and creatinine. Pueraria montana treatment significantly (p < 0.001) altered the level of phase enzymes and antioxidant. Pueraria montana treatment significantly (p < 0.001) repressed the level of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and improved the level of interleukin-10 (IL-10). Pueraria montana treatment suppressed the level of prostaglandin (PGE2), cyclooxygenase-2 (COX-2), nuclear kappa B factor (NF-κB) and transforming growth factor beta 1 (TGF-ß1). Pueraria montana suppressed the inflammatory necrosis, size the bowman capsules in the renal histopathology. Pueraria montana exhibited the chemoprotective effect via dual mechanism such as suppression of inflammatory reaction and oxidative stress.


Kidney Neoplasms , Pueraria , Animals , Antioxidants/pharmacology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/pharmacology , Body Weight , Creatinine/pharmacology , Cyclooxygenase 2/metabolism , Diethylnitrosamine/pharmacology , Ferric Compounds , Inflammation/drug therapy , Interleukin-10 , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Kidney Neoplasms/chemically induced , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , NF-kappa B/metabolism , Nitrilotriacetic Acid/analogs & derivatives , Nitrites/pharmacology , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/pharmacology , Oxidative Stress , Prostaglandins , Prostaglandins E/metabolism , Prostaglandins E/pharmacology , Pueraria/metabolism , Rats , Rats, Wistar , Thymidine/metabolism , Thymidine/pharmacology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Urea , Uric Acid/pharmacology , Xenobiotics/pharmacology
5.
Biol Reprod ; 106(4): 792-801, 2022 04 26.
Article En | MEDLINE | ID: mdl-34935905

In all mammalian species examined thus far, the ovaries produce a burst of ornithine decarboxylase (ODC) and putrescine during ovulation or after application of human chorionic gonadotropin (hCG). Aged mice have significantly reduced levels of this periovulatory ODC and putrescine rise. Putrescine supplementation, in vitro during oocyte maturation or in mouse drinking water during the periovulatory period, reduces egg aneuploidies and embryo resorption, improving fertility of aged mice. These studies suggest that periovulatory putrescine supplementation may be a simple and effective therapy for reproductive aging for women. However, putrescine supplementation is expected to increase widespread tissue putrescine levels, raising concerns of nonspecific and unwanted side effects. Given that ODC is highly expressed in the ovaries during ovulation but otherwise exhibits low activity in most tissues, we hypothesized that periovulatory supplementation of L-ornithine, the substrate of ODC, might be suitable for delivering putrescine specifically to the ovaries. In this study, we have demonstrated that systemic application of L-ornithine via oral gavage or subcutaneous injection increased ovarian putrescine levels; the increase was restricted to animals that had been injected with hCG. Furthermore, L-ornithine specifically increased ovarian putrescine levels without affecting putrescine levels in any other tissues. However, our attempts to improve fertility of aged mice through L-ornithine supplementation in mouse drinking water produced either no effects (1% L-ornithine) or negative impact on fertility (4% ornithine). Our results suggest that it might not be feasible to achieve fertility-enhancing ovarian putrescine levels via L-ornithine supplementation in drinking water without encountering undesired consequences of high dose of exogenous L-ornithine.


Dietary Supplements , Ornithine , Putrescine , Animals , Chorionic Gonadotropin/pharmacology , Drinking Water , Female , Humans , Mice , Ornithine/pharmacology , Ornithine Decarboxylase/pharmacology , Ovary , Ovulation , Putrescine/pharmacology
6.
Nat Commun ; 10(1): 65, 2019 01 08.
Article En | MEDLINE | ID: mdl-30622262

Polyamines, often elevated in cancer cells, have been shown to promote cell growth and proliferation. Whether polyamines regulate other cell functions remains unclear. Here, we explore whether and how polyamines affect genome integrity. When DNA double-strand break (DSB) is induced in hair follicles by ionizing radiation, reduction of cellular polyamines augments dystrophic changes with delayed regeneration. Mechanistically, polyamines facilitate homologous recombination-mediated DSB repair without affecting repair via non-homologous DNA end-joining and single-strand DNA annealing. Biochemical reconstitution and functional analyses demonstrate that polyamines enhance the DNA strand exchange activity of RAD51 recombinase. The effect of polyamines on RAD51 stems from their ability to enhance the capture of homologous duplex DNA and synaptic complex formation by the RAD51-ssDNA nucleoprotein filament. Our work demonstrates a novel function of polyamines in the maintenance of genome integrity via homology-directed DNA repair.


Polyamines/metabolism , Rad51 Recombinase/metabolism , Recombinational DNA Repair/physiology , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/drug effects , DNA End-Joining Repair/physiology , DNA, Single-Stranded/metabolism , Female , Gamma Rays/adverse effects , HEK293 Cells , Hair Follicle/metabolism , Hair Follicle/radiation effects , Humans , Mice , Mice, Inbred C57BL , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase/pharmacology , Ornithine Decarboxylase Inhibitors , Phthalazines/pharmacology , Piperazines/pharmacology , Rad51 Recombinase/genetics , Recombinational DNA Repair/drug effects
7.
Basic Clin Pharmacol Toxicol ; 115(5): 379-88, 2014 Nov.
Article En | MEDLINE | ID: mdl-24666424

The polyamines putrescine, spermidine and spermine play essential roles in cell proliferation and migration, two processes involved in the development of vascular disease. Thus, intervention with polyamine formation may represent a way to inhibit unwanted vascular smooth muscle cell (VSMC) proliferation. The aim of the present study was to assess the importance of polyamines for VSMC proliferation and vascular contractility. The rate-limiting step in polyamine biosynthesis is catalysed by ornithine decarboxylase (ODC). Treatment with α-difluoromethylornithine (DFMO), an irreversible inhibitor of ODC, reduced DNA synthesis in primary rat VSMCs in a concentration-dependent manner with an IC50 value of 100 µM. Moreover, DFMO reduced VSMC migration assessed in a scratch assay. The DFMO-induced attenuation of VSMC proliferation was associated with lowered cellular amount of polyamines. The antiproliferative effect of DFMO was specific because supplementation with polyamines reversed the effect of DFMO on proliferation and normalized cellular polyamine levels. Isometric force recordings in cultured rat tail artery rings showed that DFMO counteracts the decrease in contractility caused by culture with foetal bovine serum as growth stimulant. We conclude that inhibition of polyamine synthesis by DFMO may limit the first wave of cell proliferation and migration, which occurs in the acute phase after vascular injury. Besides its antiproliferative effect, DFMO may prevent loss of the smooth muscle contractile phenotype in vascular injury.


Ornithine Decarboxylase/pharmacology , Putrescine/metabolism , Spermidine/metabolism , Spermine/metabolism , Animals , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Eflornithine/administration & dosage , Eflornithine/pharmacology , Inhibitory Concentration 50 , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Phenotype , Rats , Rats, Sprague-Dawley
8.
Exp Cell Res ; 308(2): 254-64, 2005 Aug 15.
Article En | MEDLINE | ID: mdl-15923003

Polyamines are organic cations, which are considered essential for normal cell cycle progression. This view is based on results from numerous studies using a variety of enzyme inhibitors or polyamine analogues interfering with either the metabolism or the physiological functions of the polyamines. However, the presence of non-specific effects may be hard to rule out in such studies. In the present study, we have for the first time used a transgenic cell system to analyze the importance of polyamines in cell growth. We have earlier shown that expression of trypanosomal ODC in an ODC-deficient variant of CHO cells (C55.7) supported growth of these otherwise polyamine auxotrophic cells. However, one of the transgenic cell lines grew much slower than the others. As shown in the present study, the level of ODC activity was much lower in these cells, and that was reflected in a reduction of cellular polyamine levels. Analysis of cell cycle kinetics revealed that reduction of growth was correlated to prolongation of the G1, S, and G2+M phases in the cells. Providing exogenous putrescine to the cells resulted in a normalization of polyamine levels as well as cell cycle kinetics indicating a causal relationship.


Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Ornithine Decarboxylase/metabolism , Polyamines/metabolism , Animals , CHO Cells , Cell Cycle/drug effects , Cell Division/drug effects , Cell Division/physiology , Cell Proliferation , Cricetinae , DNA/metabolism , G1 Phase/drug effects , G1 Phase/physiology , G2 Phase/drug effects , G2 Phase/physiology , Kinetics , Ornithine Decarboxylase/pharmacology , Polyamines/pharmacology , Putrescine/metabolism , Putrescine/pharmacology , S Phase/drug effects , S Phase/physiology , Time Factors , Transfection , Transgenes/physiology
9.
Eur J Pharmacol ; 444(1-2): 115-21, 2002 May 24.
Article En | MEDLINE | ID: mdl-12191590

Nicotine is one of the most representative components in cigarette smoke leading to gastric ulceration. Both ornithine decarboxylase and potassium ion (K(+)) channels are essential for cell growth and wound repair. The aim of the present study is to elucidate the causative relationship of these two factors during wound healing and the influence of nicotine on this healing process in rat gastric mucosal epithelial cells (RGM-1). Nicotine markedly inhibited cell migration and proliferation in RGM-1 cells. The latter effect was significantly antagonized by a nicotinic receptor blocker, mecamylamine. Nicotine also suppressed ornithine decarboxylase activity significantly. Our data showed that inhibition of cell proliferation and ornithine decarboxylase activity by nicotine was accompanied with a reduction in K(+) channel protein expression, all of which were significantly alleviated by spermidine pretreatment. These results suggested that there was a cause/effect link between ornithine decarboxylase and K(+) channel on wound repair. Nicotine in cigarette smoke inhibited this healing process and delayed wound repair in gastric epithelial cells.


Nicotine/toxicity , Ornithine Decarboxylase/physiology , Polyamines/antagonists & inhibitors , Potassium Channels/drug effects , Wound Healing/drug effects , Animals , Cell Movement/drug effects , Gastric Mucosa/drug effects , Ornithine Decarboxylase/pharmacology , Rats
10.
Biochem Biophys Res Commun ; 250(3): 731-4, 1998 Sep 29.
Article En | MEDLINE | ID: mdl-9784414

Hepatocyte growth factor (HGF) increased both levels of phosphorylated and non-phosphorylated forms of retinoblastoma protein (RB) in primary cultured rat hepatocytes. Combined treatment of HGF and a specific inhibitor of ornithine decarboxylase (ODC), alpha-difluoromethylornithine (DFMO), reduced the levels of hyper-phosphorylated and hypo-phosphorylated forms of RB and increased the levels of the non-phosphorylated form, compared to HGF alone, but did not affect the total level of RB. Polyamines added exogenously overcame the effects of DFMO; they increased hyper- and hypo-phosphorylated forms and decreased non-phosphorylated RB. TGF-beta1 inhibited the increases in ODC activity, RB phosphorylation, and DNA synthesis induced by HGF. However, polyamines added exogenously could not overcome the inhibition by RB phosphorylation and DNA synthesis by TGF-beta1. These results suggest that polyamines are involved in the phosphorylation of RB, but the inhibition of polyamine biosynthesis by TGF-beta1 did not result in the inhibition of RB phosphorylation and DNA synthesis.


Hepatocyte Growth Factor/pharmacology , Liver/metabolism , Ornithine Decarboxylase/metabolism , Retinoblastoma Protein/metabolism , Animals , Cells, Cultured , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Male , Ornithine Decarboxylase/pharmacology , Ornithine Decarboxylase Inhibitors , Phosphorylation , Rats , Rats, Wistar , Transforming Growth Factor beta/pharmacology
11.
Nihon Geka Gakkai Zasshi ; 99(6): 362-7, 1998 Jun.
Article Ja | MEDLINE | ID: mdl-9695073

Chemoprevention of colorectal cancer has been extensively investigated in animal models and in high-risk human populations with inherited or acquired genetic changes, using anticarcinogenic agents from natural and synthetic sources. To understand active agents using a short-term assay, reliable intermediate biomarkers other than cancer are required as end-points. Endoscopically detectable aberrant crypt foci and adenomas are useful biomarkers in human intervention trials. Indomethacin and other nonsteroidal antiinflammatory drugs (NSAIDs) inhibit carcinogen-induced colon cancer development in rats. It was reported that a number of colorectal polyps in familial adenomatous polyposis patients regress after several months of sulindac treatment. Epidemiological studies have shown that regular use of aspirin and other NSAIDs reduces the risk of colorectal cancers and adenomas. In addition, ursodeoxycholic acid and alpha-difluoromethylornithine, a selective inhibitor of ornithine decarboxylase, have been employed in human intervention trials. Vegetable antioxidants such as carotenoids and flavonoids, omega-3 fatty acids, lactic acid bacteria, and indigestible oligosaccharides may also be promising chemopreventive agents.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/prevention & control , Eflornithine/pharmacology , Animals , Carotenoids/pharmacology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Fatty Acids, Omega-3/pharmacology , Humans , Ornithine Decarboxylase/pharmacology , Rats , Ursodeoxycholic Acid/pharmacology
12.
Cancer Res ; 58(8): 1650-3, 1998 Apr 15.
Article En | MEDLINE | ID: mdl-9563477

Ribonucleotide reductase, which is composed of the two protein components R1 and R2, is a highly regulated enzyme activity that is essential for DNA synthesis and repair. Recent studies have shown that elevated expression of the rate-limiting R2 component increases Raf-1 protein activation and mitogen-activated protein kinase activity and acts as a novel malignancy determinant in cooperation with H-ras and rac-1. We show that R2 cooperation in cellular transformation extends to a variety of oncogenes with different functions and cellular locations. Anchorage-independent growth of cells transformed with v-fms, v-src, A-raf, v-fes, c-myc, and ornithine decarboxylase was markedly enhanced when the R2 component of ribonucleotide reductase was overexpressed. In addition, we observed that elevated R2 expression conferred on c-myc-transformed NIH 3T3 cells an increased tumorigenic potential in immunoincompetent mice. Taken together, these observations demonstrate that the R2 protein is not only a rate-limiting component for ribonucleotide reduction but that it is also capable of acting in cooperation with a variety of oncogenes to determine transformation and tumorigenic potential.


Cell Transformation, Neoplastic , Ribonucleotide Reductases/physiology , 3T3 Cells , Animals , Blotting, Western , Cell Line , Fusion Proteins, gag-onc/physiology , Mice , Mice, Nude , Neoplasm Transplantation , Oncogene Protein gp140(v-fms)/physiology , Oncogene Protein pp60(v-src)/physiology , Ornithine Decarboxylase/pharmacology , Proto-Oncogene Proteins c-myc/physiology , Proto-Oncogene Proteins c-raf/physiology , Ribonucleotide Reductases/metabolism , Tumor Stem Cell Assay
14.
Biochemistry ; 26(8): 2349-53, 1987 Apr 21.
Article En | MEDLINE | ID: mdl-2887201

In adrenalectomized rats, the tumor-promoting phorbol ester 12-O-tetradecanoylphorbol 13-acetate (TPA) markedly enhanced the inductions of tyrosine aminotransferase (TAT) and ornithine decarboxylase by glucocorticoids, even with sufficient concentration of glucocorticoids to have a maximal effect, whereas it had no effect on TAT activity and increased ornithine decarboxylase activity only slightly in the absence of glucocorticoids. Phorbol derivatives and components of TPA such as 4 beta-phorbol, phorbol 12-tetradecanoate, phorbol 13-acetate, and 4-O-methylphorbol 12-tetradecanoate 13-acetate, which have no tumor-promoting activity or ability to activate protein kinase C, did not have any effect on TAT induction by glucocorticoid. TPA enhanced the induction of TAT by various glucocorticoids but had no effect on induction of TAT by glucagon or insulin and did not enhance the induction of glucose-6-phosphate dehydrogenase by 17 beta-estradiol. These results suggest that TPA specifically enhances the induction of TAT and ornithine decarboxylase by glucocorticoids. Similar effects of TPA on TAT induction by glucocorticoid were observed in primary cultures of adult rat hepatocytes. Another activator of protein kinase C, rac-1,2-dioctanoylglycerol, was also found to have similar effects on the cells.


Dexamethasone/pharmacology , Ornithine Decarboxylase/pharmacology , Phorbol Esters/pharmacology , Tyrosine Transaminase/biosynthesis , Adrenalectomy , Animals , Drug Synergism , Enzyme Induction , Glucagon/pharmacology , Insulin/pharmacology , Kinetics , Male , Rats , Rats, Inbred Strains , Tetradecanoylphorbol Acetate/pharmacology
15.
Biochem J ; 239(3): 745-50, 1986 Nov 01.
Article En | MEDLINE | ID: mdl-3548704

In this study we tested the hypothesis that stimulation of univalent-cation fluxes which follow the addition of growth factors are required for cell transition through the G1-phase of the cell cycle. The effect of two drugs, amiloride and bumetanide, were tested on exit of BALB/c 3T3 cells from G0/G1-phase and entry into S-phase (DNA synthesis). Amiloride, an inhibitor of the Na+/H+ antiport, only partially inhibited DNA synthesis induced by serum. Bumetanide, an inhibitor of the Na+/K+ co-transport, only slightly suppressed DNA synthesis by itself, but when added together with amiloride completely blocked cell transition through G1 and entry into S-phase. Similar inhibitory effects of the two drugs were found on the induction of ornithine decarboxylase (ODC) (a marker of mid-G1-phase) in synchronized cells stimulated by either partially purified fibroblast growth factor (FGF) or serum. To test this hypothesis further, cells arrested in G0/G1 were stimulated by serum, insulin or FGF. All induced similar elevations of cellular K+ content during the early G1-phase of the cell cycle. However, serum and FGF, but not insulin, released the cells from the G0/G1 arrest, as measured by ODC enzyme induction. This result implies that the increase in cellular K+ content may be necessary but not sufficient for induction of early events during the G1-phase. The synergistic inhibitory effects of amiloride and bumetanide on the two activities stimulated by serum growth factors, namely ODC induction (mid-G1) and thymidine incorporation into DNA (S-phase), suggested that the amiloride-sensitive Na+/H+ antiport system together with the bumetanide-sensitive Na+/K+ transporter play a role in the mitogenic signal.


Amiloride/pharmacology , Bumetanide/pharmacology , Diuretics/pharmacology , Interphase/drug effects , Cell Line , DNA/biosynthesis , Drug Synergism , Fibroblast Growth Factors/pharmacology , Insulin/pharmacology , Ornithine Decarboxylase/pharmacology , Potassium/metabolism , Sodium/metabolism
17.
Biochem Pharmacol ; 31(21): 3373-8, 1982 Nov 01.
Article En | MEDLINE | ID: mdl-7150360

Purified ornithine decarboxylase (EC 4.1.1.17, ODC) transamidated with four putrescine moieties on four glutamine residues through the action of transglutaminase (EC 2.3.2.13, TGase) purified from guinea pig liver, when added to isolated rat liver nuclei, stoichiometrically increased the activity of RNA polymerase I (EC 2.7.7.6). The increase was relative to the pmoles of purified conjugated ODC added to the reaction and could be reinitiated after the reaction had plateaued by the further addition of ODC-putrescine conjugate. The kinetics of the reaction suggest that the ODC-putrescine conjugate was not reused but degraded after each initiation. Otherwise, the rapid plateau would not be observed. The repeated addition of 278 pmoles of purified ODC-putrescine conjugate to rat liver nuclear preparations containing 200 micrograms total protein consistently stimulated the incorporation of 600-700 pmoles UMP/mg protein. We suggest that ODC transamidated by its product putrescine may be the posttranslationally modified 65,000 Mr protein which has been reported by several laboratories to serve as a labile subunit of RNA polymerase I.


Carboxy-Lyases/pharmacology , DNA-Directed RNA Polymerases/metabolism , Liver/enzymology , Ornithine Decarboxylase/pharmacology , Putrescine/pharmacology , RNA Polymerase I/metabolism , Animals , Cell Nucleus/enzymology , Dose-Response Relationship, Drug , Guinea Pigs , Kinetics , Macromolecular Substances , Magnesium/pharmacology , Ornithine/pharmacology , Protein Processing, Post-Translational , Rats
19.
Science ; 195(4277): 505-6, 1977 Feb 04.
Article En | MEDLINE | ID: mdl-835013

Reparts suggest that the activity of RNA polymerase I is modulated by a labile protein with a hlaf-life of 10 to 20 minutes. Ornithine decarboxylase is the only labile protein (half-life, 10 to 20 minutes) that increases in activity prior to increased RNA polymerase I activity. The addition of a small amount of a highly purified ornithine decarboxylase preparation to an RNA polymerase I assay increases the initial rate of the reaction as well as the time for which the assay is linear. The incorporation patterns of 14C-labeled adenosine triphosphate and 32P-labeled adenosine triphosphate into RNA indicate that the addition of ornithine decarboxylase to the RNA polymerase assay increases the rate of initiation. This report demonstrates a novel way to purify ornithine decarboxylase by RNA polymerase I affinity chromatography and presents data in support of the hypothesis that the labile protein which modulates RNA polymerase I activity is ornithine decarboxylase.


Carboxy-Lyases/pharmacology , DNA-Directed RNA Polymerases/metabolism , Ornithine Decarboxylase/pharmacology , RNA Polymerase I/metabolism , Transcription Factors/metabolism , Animals , Enzyme Activation , Kinetics , Liver/enzymology , Male , Ornithine Decarboxylase/isolation & purification , Rats
...