Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.050
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 567(7747): 253-256, 2019 03.
Article in English | MEDLINE | ID: mdl-30842655

ABSTRACT

Cancer cells exhibit altered and usually increased metabolic processes to meet their high biogenetic demands1,2. Under these conditions, ammonia is concomitantly produced by the increased metabolic processing. However, it is unclear how tumour cells dispose of excess ammonia and what outcomes might be caused by the accumulation of ammonia. Here we report that the tumour suppressor p53, the most frequently mutated gene in human tumours, regulates ammonia metabolism by repressing the urea cycle. Through transcriptional downregulation of CPS1, OTC and ARG1, p53 suppresses ureagenesis and elimination of ammonia in vitro and in vivo, leading to the inhibition of tumour growth. Conversely, downregulation of these genes reciprocally activates p53 by MDM2-mediated mechanism(s). Furthermore, the accumulation of ammonia causes a significant decline in mRNA translation of the polyamine biosynthetic rate-limiting enzyme ODC, thereby inhibiting the biosynthesis of polyamine and cell proliferation. Together, these findings link p53 to ureagenesis and ammonia metabolism, and further reveal a role for ammonia in controlling polyamine biosynthesis and cell proliferation.


Subject(s)
Ammonia/metabolism , Gene Expression Regulation/genetics , Polyamines/metabolism , Tumor Suppressor Protein p53/metabolism , Urea/metabolism , Arginase/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Cell Proliferation , Humans , Neoplasms/genetics , Neoplasms/pathology , Ornithine Carbamoyltransferase/genetics , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/genetics
2.
Anticancer Drugs ; 28(4): 457-464, 2017 04.
Article in English | MEDLINE | ID: mdl-28157137

ABSTRACT

High levels of polyamines were observed and were related to a poor prognosis in cancer patients. However, the mechanism is not obvious. The aim of this study is to mimic the extracellular polyamines in a tumor microenviroment and to explore the role of extracellular polyamines in the proliferation and migration of cancer cells. Three different concentrations of polyamines composed of putrescine, spermidine, and spermine were used. Colony formation assay, wound healing assay, and transwell migration assay were performed. Akt1-overexpression cells were constructed. The related protein expression was examined using a western blot. In this study, polyamines promoted colony formation and cell migration in a concentration-dependent and time-dependent manner. Polyamines upregulated the expression of ornithine decarboxylase (ODC), SSAT, Akt1, Akt, hypoxia-inducible factors-1α, vascular endothelial growth factor, and matrix metalloproteinases, and downregulated p27 expression. The effects of combination of polyamines and Akt1 overexpression on colony formation and migration were more obvious than the effects of Akt1 overexpression alone. In Akt1-overexpression cells, polyamines also upregulated the expression of ODC, SSAT, hypoxia-inducible factors-1α, vascular endothelial growth factor, and matrix metalloproteinases and downregulated p27 expression. In conclusion, extracellular polyamines induced proliferation and cancer cell migration by inducing ODC and SSAT expression, and the Akt1-mediated pathway.


Subject(s)
Acetyltransferases/metabolism , Carcinoma, Hepatocellular/metabolism , Colorectal Neoplasms/metabolism , Liver Neoplasms/metabolism , Ornithine Decarboxylase/metabolism , Polyamines/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Acetyltransferases/biosynthesis , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , HCT116 Cells , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Ornithine Decarboxylase/biosynthesis , Polyamines/pharmacology , Proto-Oncogene Proteins c-akt/biosynthesis , Tumor Microenvironment
3.
Amino Acids ; 48(10): 2303-11, 2016 10.
Article in English | MEDLINE | ID: mdl-27193233

ABSTRACT

Ornithine decarboxylase (ODC) is the first and usually rate-limiting enzyme in the polyamine biosynthetic pathway. In a normal physiological state, ODC is tightly regulated. However, during neoplastic transformation, ODC expression becomes upregulated. The studies described here show that the ODC mRNA transcript is destabilized by the RNA-binding protein tristetraprolin (TTP). We show that TTP is able to bind to the ODC mRNA transcript in both non-transformed RIE-1 cells and transformed Ras12V cells. Moreover, using mouse embryonic fibroblast cell lines that are devoid of a functional TTP protein, we demonstrate that in the absence of TTP both ODC mRNA stability and ODC enzyme activity increase when compared to wild-type cells. Finally, we show that the ODC 3' untranslated region contains cis acting destabilizing elements that are affected by, but not solely dependent on, TTP expression. Together, these data support the hypothesis that TTP plays a role in the post-transcriptional regulation of the ODC mRNA transcript.


Subject(s)
3' Untranslated Regions/physiology , Ornithine Decarboxylase/biosynthesis , RNA Stability/physiology , Tristetraprolin/metabolism , Animals , Cell Line, Transformed , Mice , Mice, Knockout , Ornithine Decarboxylase/genetics , Tristetraprolin/genetics
4.
J Immunol ; 193(6): 2941-51, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25114102

ABSTRACT

Chlamydia trachomatis is considered the most common agent of sexually transmitted disease worldwide. As an obligate intracellular bacterium, it relies on the host for survival. Production of NO is an effective antimicrobial defense mechanism of the innate immune system. However, whether NO is able to arrest chlamydial growth remains unclear. Similarly, little is known about the mechanisms underlying subversion of cellular innate immunity by C. trachomatis. By analyzing protein and mRNA expression in infected human mesenchymal stem cells, combined with RNA interference and biochemical assays, we observed that infection with C. trachomatis led to downregulated expression of inducible NO synthase (iNOS) in human mesenchymal stem cells in vitro. Furthermore, infection upregulated the expression of the rate-limiting enzyme in the polyamine biosynthetic pathway, ornithine decarboxylase, diverting the iNOS substrate l-arginine toward the synthesis of polyamines. Inhibition of ornithine decarboxylase activity using small interfering RNA or the competitive inhibitor difluoromethylornithine restored iNOS protein expression and activity in infected cells and inhibited chlamydial growth. This inhibition was mediated through tyrosine nitration of chlamydial protein by peroxynitrite, an NO metabolite. Thus, Chlamydia evades innate immunity by inhibiting NO production through induction of the alternative polyamine pathway.


Subject(s)
Chlamydia trachomatis/metabolism , Mesenchymal Stem Cells/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Ornithine Decarboxylase/biosynthesis , Polyamines/metabolism , Arginine/chemistry , Arginine/metabolism , Cells, Cultured , Chlamydia Infections/immunology , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/immunology , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/microbiology , Nitric Oxide Synthase Type II/biosynthesis , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase Inhibitors , Peroxynitrous Acid/chemistry , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Up-Regulation
5.
Antimicrob Agents Chemother ; 58(7): 4162-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24820075

ABSTRACT

Communication of antibiotic resistance among bacteria via small molecules is implicated in transient reduction of bacterial susceptibility to antibiotics, which could lead to therapeutic failures aggravating the problem of antibiotic resistance. Released putrescine from the extremely antibiotic-resistant bacterium Burkholderia cenocepacia protects less-resistant cells from different species against the antimicrobial peptide polymyxin B (PmB). Exposure of B. cenocepacia to sublethal concentrations of PmB and other bactericidal antibiotics induces reactive oxygen species (ROS) production and expression of the oxidative stress response regulator OxyR. We evaluated whether putrescine alleviates antibiotic-induced oxidative stress. The accumulation of intracellular ROS, such as superoxide ion and hydrogen peroxide, was assessed fluorometrically with dichlorofluorescein diacetate, while the expression of OxyR and putrescine synthesis enzymes was determined in luciferase assays using chromosomal promoter-lux reporter system fusions. We evaluated wild-type and isogenic deletion mutant strains with defects in putrescine biosynthesis after exposure to sublethal concentrations of PmB and other bactericidal antibiotics. Exogenous putrescine protected against oxidative stress induced by PmB and other antibiotics, whereas reduced putrescine synthesis resulted in increased ROS generation and a parallel increased sensitivity to PmB. Of the 3 B. cenocepacia putrescine-synthesizing enzymes, PmB induced only BCAL2641, an ornithine decarboxylase. This study reveals BCAL2641 as a critical component of the putrescine-mediated communication of antibiotic resistance and as a plausible target for designing inhibitors that would block the communication of such resistance among different bacteria, ultimately reducing the window of therapeutic failure in treating bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burkholderia cenocepacia/drug effects , Carboxy-Lyases/metabolism , Polymyxin B/pharmacology , Putrescine/metabolism , Burkholderia cenocepacia/genetics , Burkholderia cenocepacia/metabolism , Carboxy-Lyases/biosynthesis , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Bacterial , Hydrogen Peroxide/metabolism , Microbial Sensitivity Tests , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/metabolism , Oxidative Stress/drug effects , Putrescine/biosynthesis
6.
Biochem Biophys Res Commun ; 451(3): 394-401, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25094045

ABSTRACT

Over-expression of ornithine decarboxylase (ODC) is known to be involved in the epidermal carcinogenesis. However, the mechanism by which it enhances skin carcinogenesis remains undefined. Recently, role of stem cells localized in various epidermal compartments has been shown in the pathogenesis of skin cancer. To direct ODC expression in distinct epidermal compartments, we have developed keratin 6 (K6)-ODC/SKH-1 and keratin 14 (K14)-ODC/SKH-1 mice and employed them to investigate the role of ODC directed to these epidermal compartments on UVB-induced carcinogenesis. K6-driven ODC over-expression directed to outer root sheath (ORS) of hair follicle was more effective in augmenting tumorigenesis as compared to mice where K14-driven ODC expression was directed to inter-follicular epidermal keratinocytes. Chronically UVB-irradiated K6-ODC/SKH-1 developed 15±2.5 tumors/mouse whereas K14-ODC/SKH-1 developed only 6.8±1.5 tumors/mouse. K6-ODC/SKH-1 showed augmented UVB-induced proliferation and much higher pro-inflammatory responses than K14-ODC/SKH-1 mice. Tumors induced in K6-ODC/SKH-1 were rapidly growing, invasive and ulcerative squamous cell carcinoma (SCC) showing decreased expression of epidermal polarity marker E-cadherin and enhanced mesenchymal marker, fibronectin. Interestingly, the number of CD34/CK15/p63 positive stem-like cells was significantly higher in chronically UVB-irradiated K6-ODC/SKH-1 as compared to K14-ODC/SKH-1 mice. Reduced Notch1 expression was correlated with the expansion of stem cell compartment in these animals. However, other signaling pathways such as DNA damage response or mTOR signaling pathways were not significantly different in tumors induced in these two murine models suggesting the specificity of Notch pathway in this regard. These data provide a novel role of ODC in augmenting tumorigenesis via negatively regulated Notch-mediated expansion of stem cell compartment.


Subject(s)
Keratin-6/pharmacology , Ornithine Decarboxylase/biosynthesis , Receptors, Notch/biosynthesis , Animals , Carcinogenesis , Carcinoma, Squamous Cell/metabolism , Female , Hair Follicle/metabolism , Keratin-14/metabolism , Keratinocytes/metabolism , Mice , Skin Neoplasms/etiology , Skin Neoplasms/pathology
7.
Transgenic Res ; 23(1): 153-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24174210

ABSTRACT

Polyamines, spermidine, spermine and their precursor putrescine, are ubiquitous cell components essential for normal cell growth. Increased polyamine levels and enhanced biosynthesis have been associated with malignant transformation and tumor formation, and thus, the polyamines have been considered to be a meaningful target to cancer therapies. However, clinical cancer treatment trials using inhibitors of polyamine synthesis have been unsuccessful probably due to compensatory uptake of polyamines from extracellular sources. The antizyme proteins regulate both polyamine biosynthesis and transport, and thus, the antizymes could provide an efficient approach to control cellular proliferation compared to the mere inhibition of biosynthesis. To define the role of antizymes in proliferative processes associated with the whole animal, we have generated transgenic mice overexpressing mouse antizyme 1 gene under its own regulatory sequences. Antizyme 1 protein was abundantly expressed in various organs and the expressed antizyme protein was functional as ornithine decarboxylase activity was significantly reduced in all tissues analyzed. However, antizyme 1 overexpression caused only minor changes in tissue polyamine levels demonstrating the challenges in using the "antizyme approach" to deplete polyamines in a living animal. Neither were there any changes in cellular proliferation in the proliferative tissues of transgenic animals. Interestingly though, there was occurrence of abnormally high level of apoptosis in the non-proliferating part of the colon epithelia. Otherwise, the transgenic founder mice appeared healthy and out of seven founders six were fertile. However, none of the founders could transmit the transgene suggesting that the antizyme 1 overexpression may be deleterious to transgenic gametes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Ornithine Decarboxylase/biosynthesis , Polyamines/metabolism , Proteins/genetics , Animals , Biological Transport/genetics , Gene Expression Regulation , Homeostasis , Mice , Mice, Transgenic , Ornithine Decarboxylase/genetics , Proteins/metabolism , Tissue Distribution
8.
Mol Cell Biochem ; 390(1-2): 133-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24464033

ABSTRACT

Ornithine decarboxylase (ODC) is the first rate-limiting enzyme in polyamine biosynthesis, which is essential for cell survival. We hypothesized that the ODC/polyamine system is involved in ischemic preconditioning (IPC)-mediated cardioprotection through the activation of Erk1/2 and Akt and through the inhibition of the mitochondrial permeability transition (mPT). Isolated rat hearts were subjected to 40 min of ischemia either with or without IPC (3 cycles of 5-min global ischemia), and ODC protein expression, polyamine content, and Akt and Erk1/2 phosphorylation were evaluated after 30 min of reperfusion. IPC significantly upregulated the ODC/polyamine pathway, promoted Erk1/2 and Akt phosphorylation, and reduced the infarct size and heart dysfunction after reperfusion. An inhibitor of ODC, α-difluoromethylornithine (DFMO), abolished the IPC-induced cardioprotection. Moreover, the inhibition of the IPC-induced activation of Erk1/2 and Akt using PD98059 or wortmannin downregulated the ODC/polyamine system. In separate studies, the Ca(2+) load required to open the mPT pore was significantly lower in DFMO-treated cardiac mitochondria than in mitochondria from IPC hearts. Furthermore, spermine or spermidine significantly inhibited the mPT induced by CaCl2. These results suggest that IPC upregulates the ODC/polyamine system and mediates preconditioning cardioprotection, which may depend on the phosphorylation/activation of Erk1/2 and Akt and on the inhibition of the mPT during reperfusion.


Subject(s)
MAP Kinase Signaling System/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Ornithine Decarboxylase/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Animals , Ischemic Preconditioning, Myocardial , Male , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/metabolism , Ornithine Decarboxylase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats
9.
Carcinogenesis ; 34(6): 1315-22, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23385063

ABSTRACT

During the process of skin tumor promotion, expression of the cutaneous cancer stem cell (CSC) marker CD34(+) is required for stem cell activation and tumor formation. A previous study has shown that activation of protein kinase D1 (PKD1) is involved in epidermal tumor promotion; however, the signals that regulate CSCs in skin carcinogenesis have not been characterized. This study was designed to investigate the chemopreventive potential of peracetylated (-)-epigallocatechin-3-gallate (AcEGCG) on 7,12-dimethylbenz[a]-anthracene (DMBA)-initiated and 12-O-tetradecanoylphorbol-13-acetate (TPA)-promoted skin tumorigenesis in ICR mice and to elucidate the possible mechanisms involved in the inhibitory action of PKD1 on CSCs. We demonstrated that topical application of AcEGCG before TPA treatment can be more effective than EGCG in reducing DMBA/TPA-induced tumor incidence and multiplicity. Notably, AcEGCG not only inhibited the expression of p53, p21, c-Myc, cyclin B, p-CDK1 and Cdc25A but also restored the activation of extracellular signal-regulated kinase 1/2 (ERK1/2), which decreased DMBA/TPA-induced increases in tumor proliferation and mitotic index. To clarify the role of PKD1 in cell proliferation and tumorigenesis, we studied the expression and activation of PKD1 in CD34(+) skin stem cells and skin tumors. We found that PKD1 was strongly expressed in CD34(+) cells and that pretreatment with AcEGCG markedly inhibited PKD1 activation and CD34(+) expression. More importantly, pretreatment with AcEGCG remarkably suppressed nuclear factor-kappaB, cyclic adenosine 3',5'-monophosphate-responsive element-binding protein (CREB) and CCAAT-enhancer-binding protein (C/EBPs) activation by inhibiting the phosphorylation of c-Jun-N-terminal kinase 1/2, p38 and phosphatidylinositol 3-kinase (PI3K)/Akt and by attenuating downstream target gene expression, including inducible nitric oxide synthase, cyclooxygenase-2, ornithine decarboxylase and vascular endothelial growth factor. Moreover, this is the first study to demonstrate that AcEGCG is a CD34(+) and PKD1 inhibitor in the multistage mouse skin carcinogenesis model. Overall, our results powerfully suggest that AcEGCG could be developed into a novel chemopreventive agent and that PKD1 may be a preventive and therapeutic target for skin cancer in clinical settings.


Subject(s)
Acetates/pharmacology , Antigens, CD34/metabolism , Catechin/analogs & derivatives , Cell Transformation, Neoplastic/drug effects , Protein Kinase C/antagonists & inhibitors , Skin Neoplasms/prevention & control , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antigens, CD34/biosynthesis , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/metabolism , Catechin/pharmacology , Cell Proliferation/drug effects , Chemoprevention , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 2/biosynthesis , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Mitotic Index , Nitric Oxide Synthase Type II/biosynthesis , Ornithine Decarboxylase/biosynthesis , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/drug therapy , Stem Cells/drug effects , Stem Cells/metabolism , Tetradecanoylphorbol Acetate , Vascular Endothelial Growth Factor A/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Biol Chem ; 287(6): 3674-90, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22157018

ABSTRACT

In a glucose-salt solution (Earle's balanced salt solution), asparagine (Asn) stimulates ornithine decarboxylase (ODC) activity in a dose-dependent manner, and the addition of epidermal growth factor (EGF) potentiates the effect of Asn. However, EGF alone fails to activate ODC. Thus, the mechanism by which Asn activates ODC is important for understanding the regulation of ODC activity. Asn reduced antizyme-1 (AZ1) mRNA and protein. Among the amino acids tested, Asn and glutamine (Gln) effectively inhibited AZ1 expression, suggesting a differential role for amino acids in the regulation of ODC activity. Asn decreased the putrescine-induced AZ1 translation. The absence of amino acids increased the binding of eukaryotic initiation factor 4E-binding protein (4EBP1) to 5'-mRNA cap and thereby inhibited global protein synthesis. Asn failed to prevent the binding of 4EBP1 to mRNA, and the bound 4EBP1 was unphosphorylated, suggesting the involvement of the mammalian target of rapamycin (mTOR) in the regulation of AZ1 synthesis. Rapamycin treatment (4 h) failed to alter the expression of AZ1. However, extending the treatment (24 h) allowed expression in the presence of amino acids, indicating that AZ1 is expressed when TORC1 signaling is decreased. This suggests the involvement of cap-independent translation. However, transient inhibition of mTORC2 by PP242 completely abolished the phosphorylation of 4EBP1 and decreased basal as well as putrescine-induced AZ1 expression. Asn decreased the phosphorylation of mTOR-Ser(2448) and AKT-Ser(473), suggesting the inhibition of mTORC2. In the absence of amino acids, mTORC1 is inhibited, whereas mTORC2 is activated, leading to the inhibition of global protein synthesis and increased AZ1 synthesis via a cap-independent mechanism.


Subject(s)
Asparagine/pharmacology , Gene Expression Regulation/drug effects , Protein Biosynthesis/drug effects , Proteins/metabolism , RNA Caps/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins , Cell Line , Gene Expression Regulation/physiology , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Biosynthesis/physiology , Proteins/genetics , RNA Caps/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Br J Nutr ; 110(11): 1968-77, 2013 Dec 14.
Article in English | MEDLINE | ID: mdl-23656796

ABSTRACT

In the present study, quadruplicate groups of juvenile Atlantic salmon (Salmo salar) were fed plant protein-based diets with increasing arginine inclusions (range 28·8-37·4 g/kg DM) to investigate whether arginine supplementation affects growth and lipid accumulation through an elevated polyamine turnover. Dietary lysine was held at a constant concentration, just below the requirement. All other amino acids were balanced and equal in the diets. Arginine supplementation increased protein and fat accretion, without affecting the hepatosomatic or visceralsomatic indices. Dietary arginine correlated with putrescine in the liver (R 0·78, P= 0·01) and with ornithine in the muscle, liver and plasma (P= 0·0002, 0·003 and 0·0002, respectively). The mRNA of ornithine decarboxylase, the enzyme producing putrescine, was up-regulated in the white adipose tissue of fish fed the high-arginine inclusion compared with those fed the low-arginine diet. Concomitantly, spermidine/spermine-(N1)-acetyltransferase, the rate-limiting enzyme for polyamine turnover that consumes acetyl-CoA, showed an increased activity in the liver of fish fed the arginine-supplemented diets. In addition, lower acetyl-CoA concentrations were observed in the liver of fish fed the high-arginine diet, while ATP, which is used in the process of synthesising spermidine and spermine, did not show a similar trend. Gene expression of the rate-limiting enzyme for ß-oxidation of long-chain fatty acids, carnitine palmitoyl transferase-1, was up-regulated in the liver of fish fed the high-arginine diet. Taken together, the data support that increased dietary arginine activates polyamine turnover and ß-oxidation in the liver of juvenile Atlantic salmon and may act to improve the metabolic status of the fish.


Subject(s)
Arginine/metabolism , Diet/veterinary , Dietary Supplements , Energy Metabolism , Polyamines/metabolism , Salmo salar/metabolism , Acetyltransferases/biosynthesis , Acetyltransferases/genetics , Acetyltransferases/metabolism , Adipose Tissue, White/enzymology , Adipose Tissue, White/growth & development , Adipose Tissue, White/metabolism , Animals , Aquaculture , Arginine/administration & dosage , Carnitine O-Palmitoyltransferase/biosynthesis , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Diet/adverse effects , Dietary Proteins/adverse effects , Dietary Proteins/metabolism , Enzyme Induction , Fish Proteins/biosynthesis , Fish Proteins/genetics , Fish Proteins/metabolism , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Lipid Metabolism , Liver/enzymology , Liver/growth & development , Liver/metabolism , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Ornithine/blood , Ornithine/metabolism , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Plant Proteins/adverse effects , Plant Proteins/metabolism , Putrescine/metabolism , Salmo salar/blood , Salmo salar/growth & development
12.
Biochem J ; 442(1): 199-207, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22070140

ABSTRACT

Upon Ras activation, ODC (ornithine decarboxylase) is markedly induced, and numerous studies suggest that ODC expression is controlled by Ras effector pathways. ODC is therefore a potential target in the treatment and prevention of Ras-driven tumours. In the present study we compared ODC mRNA translation profiles and stability in normal and Ras12V-transformed RIE-1 (rat intestinal epithelial) cells. While translation initiation of ODC increased modestly in Ras12V cells, ODC mRNA was stabilized 8-fold. Treatment with the specific mTORC1 [mTOR (mammalian target of rapamycin) complex 1] inhibitor rapamycin or siRNA (small interfering RNA) knockdown of mTOR destabilized the ODC mRNA, but rapamycin had only a minor effect on ODC translation initiation. Inhibition of mTORC1 also reduced the association of the mRNA-binding protein HuR with the ODC transcript. We have shown previously that HuR binding to the ODC 3'UTR (untranslated region) results in significant stabilization of the ODC mRNA, which contains several AU-rich regions within its 3'UTR that may act as regulatory sequences. Analysis of ODC 3'UTR deletion constructs suggests that cis-acting elements between base 1969 and base 2141 of the ODC mRNA act to stabilize the ODC transcript. These experiments thus define a novel mechanism of ODC synthesis control. Regulation of ODC mRNA decay could be an important means of limiting polyamine accumulation and subsequent tumour development.


Subject(s)
Cell Transformation, Neoplastic/genetics , Intracellular Signaling Peptides and Proteins/physiology , Ornithine Decarboxylase/genetics , RNA Stability/drug effects , RNA, Messenger/metabolism , 3' Untranslated Regions/physiology , Animals , Cell Line, Transformed , ELAV Proteins/metabolism , Genes, ras , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Ornithine Decarboxylase/biosynthesis , Protein Biosynthesis , RNA, Small Interfering/pharmacology , Rats
13.
Am J Physiol Cell Physiol ; 303(3): C308-17, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22592407

ABSTRACT

Early epithelial restitution occurs as a consequence of intestinal epithelial cell (IEC) migration after wounding, and its defective regulation is implicated in various critical pathological conditions. Polyamines stimulate intestinal epithelial restitution, but their exact mechanism remains unclear. Canonical transient receptor potential-1 (TRPC1)-mediated Ca(2+) signaling is crucial for stimulation of IEC migration after wounding, and induced translocation of stromal interaction molecule 1 (STIM1) to the plasma membrane activates TRPC1-mediated Ca(2+) influx and thus enhanced restitution. Here, we show that polyamines regulate intestinal epithelial restitution through TRPC1-mediated Ca(2+) signaling by altering the ratio of STIM1 to STIM2. Increasing cellular polyamines by ectopic overexpression of the ornithine decarboxylase (ODC) gene stimulated STIM1 but inhibited STIM2 expression, whereas depletion of cellular polyamines by inhibiting ODC activity decreased STIM1 but increased STIM2 levels. Induced STIM1/TRPC1 association by increasing polyamines enhanced Ca(2+) influx and stimulated epithelial restitution, while decreased formation of the STIM1/TRPC1 complex by polyamine depletion decreased Ca(2+) influx and repressed cell migration. Induced STIM1/STIM2 heteromers by polyamine depletion or STIM2 overexpression suppressed STIM1 membrane translocation and inhibited Ca(2+) influx and epithelial restitution. These results indicate that polyamines differentially modulate cellular STIM1 and STIM2 levels in IECs, in turn controlling TRPC1-mediated Ca(2+) signaling and influencing cell migration after wounding.


Subject(s)
Calcium Signaling , Cell Adhesion Molecules/metabolism , Intestinal Mucosa/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Polyamines/metabolism , TRPC Cation Channels/metabolism , Caco-2 Cells , Cell Movement/physiology , Humans , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase Inhibitors , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2 , Wound Healing/physiology
14.
Am J Physiol Cell Physiol ; 303(1): C102-11, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22555848

ABSTRACT

Polyamines regulate multiple signaling pathways and are implicated in many aspects of cellular functions, but the exact molecular processes governed by polyamines remain largely unknown. In response to environmental stress, repression of translation is associated with the assembly of stress granules (SGs) that contain a fraction of arrested mRNAs and are thought to function as mRNA storage. Here we show that polyamines modulate the assembly of SGs in normal intestinal epithelial cells (IECs) and that induced SGs following polyamine depletion are implicated in the protection of IECs against apoptosis. Increasing the levels of cellular polyamines by ectopic overexpression of the ornithine decarboxylase gene decreased cytoplasmic levels of SG-signature constituent proteins eukaryotic initiation factor 3b and T-cell intracellular antigen-1 (TIA-1)-related protein and repressed the assembly of SGs induced by exposure to arsenite-induced oxidative stress. In contrast, depletion of cellular polyamines by inhibiting ornithine decarboxylase with α-difluoromethylornithine increased cytoplasmic eukaryotic initiation factor 3b and TIA-1 related protein abundance and enhanced arsenite-induced SG assembly. Polyamine-deficient cells also exhibited an increase in resistance to tumor necrosis factor-α/cycloheximide-induced apoptosis, which was prevented by inhibiting SG formation with silencing SG resident proteins Sort1 and TIA-1. These results indicate that the elevation of cellular polyamines represses the assembly of SGs in normal IECs and that increased SGs in polyamine-deficient cells are crucial for increased resistance to apoptosis.


Subject(s)
Apoptosis , Cytoplasmic Granules/metabolism , Heat-Shock Proteins/biosynthesis , Intestinal Mucosa/metabolism , Polyamines/metabolism , Adaptor Proteins, Vesicular Transport/biosynthesis , Adaptor Proteins, Vesicular Transport/genetics , Animals , Apoptosis/drug effects , Arsenites/pharmacology , Cell Line , Cycloheximide/pharmacology , Cytoplasmic Granules/ultrastructure , Eflornithine/pharmacology , Epithelial Cells/metabolism , Eukaryotic Initiation Factor-3/biosynthesis , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase Inhibitors , Oxidative Stress , Poly(A)-Binding Proteins/biosynthesis , Poly(A)-Binding Proteins/genetics , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/metabolism , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
15.
Carcinogenesis ; 33(3): 637-43, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198215

ABSTRACT

Surveillance colonoscopy is an important strategy for prevention of colorectal cancer. 5-aminosalicylate (ASA) (mesalazine) is discussed as a chemopreventive agent as it reduces the cancer risk in ulcerative colitis patients. The current study analyses the effect of 5-ASA on Wnt/ß-catenin signaling in vitro and in vivo in colon epithelial cells. The effect of 5-ASA was determined using a ß-catenin/T-cell factor (TCF)-reporter assay and by western blotting in cultured colon cancer cells. Formalin fixed paraffin embedded material from 227 polyps removed from a subgroup of 56 patients, who participated in a randomized placebo-controlled 3-year prevention trial with 5-ASA was evaluated according to histomorphological characteristics and expression of ß-catenin and target genes Cox2, cyclin D1 and E-cadherin as well as ornithine decarboxylase (ODC). Patients were grouped into a low-risk and a high-risk group according to the number of adenomas at initial colonoscopy. ß-catenin/TCF signaling activity was significantly reduced by 5-ASA treatment possibly through a reduction in ß-catenin levels. Moreover, 5-ASA significantly reduced ß-catenin levels and nuclear localization in patients' adenomas. In addition, 5-ASA also significantly changed expression of the downstream targets Cox2, cyclin D1 and E-cadherin, correlating with ß-catenin status. Moreover, 5-ASA significantly reduced levels of ODC in vivo. Expression of p53 was unaltered by the 5-ASA treatment. Our study shows a significant in vitro and long-term in vivo effect of 5-ASA on ß-catenin signaling as a key signaling pathway in the development of colorectal adenoma. Therefore, we suggest the use of 5-ASA as a promising drug for prevention of sporadic colorectal carcinoma.


Subject(s)
Adenoma/drug therapy , Colorectal Neoplasms/drug therapy , Mesalamine/pharmacology , beta Catenin/metabolism , Adenoma/prevention & control , Cadherins/biosynthesis , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Colitis, Ulcerative/drug therapy , Colorectal Neoplasms/prevention & control , Cyclin D1/biosynthesis , Cyclooxygenase 2/biosynthesis , Disease Progression , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Male , Mesalamine/therapeutic use , Ornithine Decarboxylase/biosynthesis , Signal Transduction/drug effects , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Tumor Suppressor Protein p53/biosynthesis , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/biosynthesis
16.
J Biol Chem ; 285(41): 31885-94, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20685649

ABSTRACT

Ornithine decarboxylase (ODC) is the first and usually rate-limiting enzyme in the polyamine biosynthetic pathway. Under normal physiological conditions, polyamine content and ODC enzyme activity are highly regulated. However, the induction of ODC activity is an early step in neoplastic transformation. The studies described here use normal mouse keratinocytes (C5N cells), and spindle carcinoma cells (A5 cells) to explore the regulation of ODC in nonmelanoma skin cancer development. Previous results have shown that induction of ODC activity is both necessary and sufficient for the promotion of skin tumors. We see a marked increase in ODC enzyme activity in A5 cells compared with C5N keratinocytes, which correlates with a 4-fold stabilization of ODC mRNA. These data suggest that ODC is post-transcriptionally regulated in skin tumor development. Thus, we sought to investigate whether the ODC transcript interacts with the RNA-binding protein HuR, which is known to bind to and stabilize its target mRNAs. We show that HuR is able to bind to the ODC 3'-UTR in A5 cells but not in C5N cells. Immunofluorescence results reveal that HuR is present in both the nucleus and cytoplasm of A5 cells, whereas C5N cells exhibit strictly nuclear localization of HuR. Knockdown experiments in A5 cells showed that when HuR is depleted, ODC RNA becomes less stable, and ODC enzyme activity decreases. Together, these data support the hypothesis that HuR plays a causative role in ODC up-regulation during nonmelanoma skin cancer development by binding to and stabilizing the ODC transcript.


Subject(s)
3' Untranslated Regions , Antigens, Surface/metabolism , Neoplasm Proteins/metabolism , Ornithine Decarboxylase/biosynthesis , RNA Stability , RNA, Neoplasm/metabolism , RNA-Binding Proteins/metabolism , Sarcoma/metabolism , Skin Neoplasms/metabolism , Animals , Antigens, Surface/genetics , Disease Models, Animal , ELAV Proteins , ELAV-Like Protein 1 , Gene Knockdown Techniques , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Neoplasm Proteins/genetics , Ornithine Decarboxylase/genetics , RNA, Neoplasm/genetics , RNA-Binding Proteins/genetics , Sarcoma/genetics , Sarcoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription, Genetic
17.
Gastroenterology ; 139(5): 1686-98, 1698.e1-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20600019

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori-induced immune responses fail to eradicate the bacterium. Nitric oxide (NO) can kill H pylori. However, translation of inducible NO synthase (iNOS) and NO generation by H pylori-stimulated macrophages is inhibited by the polyamine spermine derived from ornithine decarboxylase (ODC), and is dependent on availability of the iNOS substrate L-arginine (L-Arg). We determined if spermine inhibits iNOS-mediated immunity by reducing L-Arg uptake into macrophages. METHODS: Levels of the inducible cationic amino acid transporter (CAT)2, ODC, and iNOS were measured in macrophages and H pylori gastritis tissues. L-Arg uptake, iNOS expression, and NO levels were assessed in cells with small interfering RNA knockdown of CAT2 or ODC, and in gastric macrophages. The ODC inhibitor, α-difluoromethylornithine, was administered to H pylori-infected mice for 4 months after inoculation. RESULTS: H pylori induced CAT2 and uptake of L-Arg in RAW 264.7 or primary macrophages. Addition of spermine or knockdown of CAT2 inhibited L-Arg uptake, NO production, and iNOS protein levels, whereas knockdown of ODC had the opposite effect. CAT2 and ODC were increased in mouse and human H pylori gastritis tissues and localized to macrophages. Gastric macrophages from H pylori-infected mice showed increased ODC expression, and attenuated iNOS and NO levels upon ex vivo H pylori stimulation versus cells from uninfected mice. α-Difluoromethylornithine treatment of infected mice restored L-Arg uptake, iNOS protein expression, and NO production in gastric macrophages, and significantly reduced both H pylori colonization levels and gastritis severity. CONCLUSIONS: Up-regulation of ODC in gastric macrophages impairs host defense against H pylori by suppressing iNOS-derived NO production.


Subject(s)
Arginine/antagonists & inhibitors , Gastric Mucosa/metabolism , Helicobacter Infections/immunology , Helicobacter pylori/pathogenicity , Immunity, Cellular/physiology , Nitric Oxide/biosynthesis , Spermine/pharmacology , Animals , Arginine/metabolism , Cationic Amino Acid Transporter 2/biosynthesis , Cationic Amino Acid Transporter 2/genetics , Cells, Cultured , Disease Models, Animal , Gastric Mucosa/microbiology , Gastritis/metabolism , Gastritis/microbiology , Gastritis/pathology , Gene Expression Regulation , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/genetics , Polyamines/pharmacology , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Biol Rep ; 38(2): 949-55, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20533091

ABSTRACT

Ornithine decarboxylase (ODC), the first rate-limiting enzyme of polyamine biosynthesis, was found to be associated with cell growth, proliferation and transformation. ODC gene expression in gastric cancer was increased and its level was positively correlated with the degree of malignity of gastric mucosa and development of gastric lesions. In order to evaluate the therapeutic effects of antisense RNA of ODC on gastric cancer, an antisense RNA of ODC expressing plasmid pcDNA-ODCr which delivered a 120 bp fragment complementary to the initiation codon of ODC gene was constructed and transfected to gastric cancer cells SGC7901 and MGC803. Expression of ODC in gastric cancer cells was determined by western blot. Cell proliferation was assessed by MTS assay. Cell cycle was analyzed by flow cytometry and Matrigel assay was performed to assess the ability of gastric cancer cell invasiveness. The results showed that the ODC gene expression in gastric cancer cells transfected with the pcDNA-ODCr was downregulated efficiently. Tumor cell proliferation was suppressed significantly, and cell cycle was arrested at G1 phase. Gastric cancer cells had reduced invasiveness after gene transfer. Our study suggested that antisense RNA of ODC expressing plasmid pcDNA-ODCr had antitumor activity by inhibiting the expression of ODC, and downregulation of ODC expression using a gene therapy approach might be a novel therapeutic strategy for gastric cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Ornithine Decarboxylase/biosynthesis , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Collagen/chemistry , Drug Combinations , Flow Cytometry/methods , Gene Expression Profiling , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Laminin/chemistry , Oligonucleotides, Antisense/genetics , Plasmids/metabolism , Proteoglycans/chemistry , RNA/metabolism
19.
J Pharmacol Exp Ther ; 335(1): 23-31, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20627998

ABSTRACT

E6201 [(3S,4R,5Z,8S,9S,11E)-14-(ethylamino)-8,9,16-trihydroxy-3,4-dimethyl-3,4,9,10-tetrahydro-1H-2-benzoxacyclotetradecine-1,7(8H)-dione)] is a novel anti-inflammatory agent that has potent inhibitory effects on the production of proinflammatory cytokines from leukocytes and antiproliferative activity on keratinocytes. To characterize the in vivo pharmacological activity of E6201, topically administered E6201 was evaluated in several different animal models of dermatitis. E6201 formulated as an ointment or cream showed dose-dependent inhibition of croton oil-induced acute edema formation and neutrophil infiltration into mouse skin. In addition, E6201 cream inhibited the 1-fluoro-2,4-dinitrobenzene-induced contact hypersensitivity reaction mediated by T cells in mice. In this model, E6201 cream also suppressed the migration of neutrophils and lymphocytes into the inflammatory site. Pretreatment with E6201 cream attenuated phorbol-12 myristate 13-acetate-induced ornithine decarboxylase activity, a marker of proliferation in epidermis. Furthermore, E6201 ointment showed inhibitory effects on both mezerein-induced and interleukin (IL)-23-induced epidermal hyperplasia. E6201 also suppressed T cell receptor-stimulated IL-17 production from human T cells. These results indicate that topically administered E6201 may be a useful agent for the prevention and treatment of cutaneous inflammatory and hyperproliferative diseases such as psoriasis.


Subject(s)
Drug Eruptions/pathology , Lactones/pharmacology , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Administration, Topical , Animals , Antineoplastic Agents, Phytogenic , Croton Oil , Dinitrofluorobenzene , Diterpenes , Hyperplasia/chemically induced , Hyperplasia/pathology , Indicators and Reagents , Interleukin-17/biosynthesis , Interleukin-23/toxicity , Lactones/administration & dosage , Male , Mice , Mice, Inbred BALB C , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/metabolism , Peroxidase/metabolism , Protein Kinase Inhibitors/administration & dosage , Psoriasis/chemically induced , Psoriasis/pathology , Skin/pathology , T-Lymphocytes/physiology , Tetradecanoylphorbol Acetate/pharmacology
20.
Curr Microbiol ; 61(6): 567-73, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20440620

ABSTRACT

Interactions between Bacillus anthracis (B. anthracis) and host cells are of particular interest given the implications of anthrax as a biological weapon. Inhaled B. anthracis endospores encounter alveolar macrophages as the first line of defense in the innate immune response. Yet, the consequences of this interaction remain unclear. We have demonstrated that B. anthracis uses arginase, inherent in the endospores, to reduce the ability of macrophages to produce nitric oxide ((•)NO) from inducible nitric oxide synthase (NOS2) by competing for L-arginine, producing L-ornithine at the expense of (•)NO. In the current study, we used genetically engineered B. anthracis endospores to evaluate the contribution of germination and the lethal toxin (LT) in mediating signaling pathways responsible for the induction of NOS2 and ornithine decarboxylase (ODC), which is the rate-limiting enzyme in the conversion of L-ornithine into polyamines. We found that induction of NOS2 and ODC expression in macrophages exposed to B. anthracis occurs through the activation of p38 and ERK1/2 MAP kinases, respectively. Optimal induction of NOS2 was observed following exposure to germination-competent endospores, whereas ODC induction occurred irrespective of the endospores' germination capabilities and was more prominent in macrophages exposed to endospores lacking LT. Our findings suggest that activation of kinase signaling cascades that determine macrophage defense responses against B. anthracis infection occurs through distinct mechanisms.


Subject(s)
Bacillus anthracis/pathogenicity , Macrophages/enzymology , Macrophages/microbiology , Nitric Oxide Synthase Type II/biosynthesis , Ornithine Decarboxylase/biosynthesis , Spores, Bacterial/pathogenicity , Gene Expression Regulation , Host-Pathogen Interactions , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL