Abstract
Clinical practice and experimental studies have shown the necessity of sufficient quantities of folic acid intake for normal embryogenesis and fetal development in the prevention of neural tube defects (NTDs) and neurological malformations. So, women of childbearing age must be sure to have an adequate folate intake periconceptionally, prior to and during pregnancy. Folic acid fortification of all enriched cereal grain product flour has been implemented in many countries. Thus, hundreds of thousands of people have been exposed to an increased intake of folic acid. Folate plays an essential role in the biosynthesis of methionine. Methionine is the principal aminopropyl donor required for polyamine biosynthesis, which is up-regulated in actively growing cells, including cancer cells. Folates are important in RNA and DNA synthesis, DNA stability and integrity. Clinical and epidemiological evidence links folate deficiency to DNA damage and cancer. On the other hand, long-term folate oversupplementation leads to adverse toxic effects, resulting in the appearance of malignancy. Considering the relationship of polyamines and rapidly proliferating tissues (especially cancers), there is a need for better investigation of the relationship between the ingestion of high amounts of folic acid in food supplementation and polyamine metabolism, related to malignant processes in the human body.
Introduction
Polyamine biochemistry and functions
The polyamines (PAs) diamine putrescine [H2N(CH2)4NH2], triamine spermidine (Spd) [H2N(CH2)3NH(CH2)4NH2] and spermine (Sp) [H2N(CH2)3NH(CH2)4NH(CH2)3NH2] (tetramine), are ubiquitous small basic molecules that play multiple essential roles in mammalian cell physiology. Despite the extensive research of their functions in physiology, they are not yet well understood [1], [2], [3], [4], [5], [6], [7], [8], [9], [10], [11].
PAs have been implicated in diverse biological processes that are important for maintaining cell viability, including replication, transcription, translation, posttranslational modification, membrane stability and interactions with ion channels. They regulate cellular proliferation, transformation, differentiation, or cell growth and apoptosis, and especially tumorigenesis [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22]. In cells, PAs, as polycations, interact electrostatically with negatively charged moieties, such as DNA, RNA, phospholipids, mucopolysaccharides or various glycosaminoglycans (GAGs) and proteins. Binding to DNA, PAs play a significant role in stabilizing DNA, especially chromatin structure, during the cell cycle [23], [24], [25], [26], [27]. The affinity for binding to both DNA and RNA is: Sp>spermidine>putrescine. PAs stabilize the double helix of DNA, probably by forming a bridge across the narrow groove, involving electrostatic bonding with the phosphate group [27], [28], [29], [30], [31]. They occur in the free form as cations, but are often conjugated to various macromolecules [32]. Nuclear aggregates of PAs (NAPs) are formed by the formation of ionic bonds between PA ammonium and DNA phosphate groups. These aggregates of PAs with DNA influence, more efficaciously than single PAs, both the conformation and the protection of the DNA [28], [29], [30], [31], [32]. The interaction between NAPs and genomic DNA provides evidence for the decisive role of “natural” NAPs in the regulation of the important aspects of DNA physiology, such as conformation, protection and packaging, thus suggesting a new vision of the functions that PAs accomplish in the cell nucleus [30]. Spd is not concentrated in the nucleus [1], [25], [32], [33], [34], [35], [36].
The interactions of PAs with the components of membranes, such as phospholipids or negatively charged residues of membrane-bound proteins, could affect some properties of biological membranes [14], [15], [16], [17]. Also, the binding of PAs to RNA increases the efficiency of protein synthesis, as a substantial proportion of intracellular PAs is attached to the ribosomes. PAs are involved in the correct assembly of the ribosomal subunits during the process of protein synthesis. However, the common chemical or physical processes that can only operate in the presence of PAs have not yet been presented. Therefore, a better understanding of the role of PAs might reveal its key feature in cellular and molecular biology [12], [37], [38], [39].
Intracellular PA levels are regulated in a very fast, sensitive and precise manner. Cells have developed complex regulatory machinery for the maintenance and regulation of their intracellular homeostasis by their biosynthesis, catabolism and transport, namely the uptake and excretion mechanisms. The enzymes in PA metabolism are controlled at the level of transcription, translation and protein degradation. The complexity of PA metabolism and a multitude of compensatory mechanisms are invoked to maintain PA homeostasis [1], [38], [39], [40], [41], [42].
Transport is one of the main ways by which the intracellular PA content is regulated. The PA uptake is stimulated by the reduction of the intracellular PA content [10], [12], [13], [22], [42], [43], [44], [45]. PA uptake depends, in part, on the expression of cell surface proteoglycans (PGs), in particular those containing glycosaminoglycan heparan sulfate, highly charged polysaccharide, consisting of variably sulfated glucosamine residues and glucuronic or iduronic acid. The negatively charged carboxyl and sulfate groups can interact with the positive charges on the PAs with an affinity equal to or even higher than that for DNA. The inhibition of GAG synthesis or exogenous heparan sulfate reduces the uptake of exogenous PAs [45], [46], [47].
PA metabolism
Biosynthesis
PA biosynthesis is enhanced during growth and under the influence of growth-promoting stimuli, as in many diseases and cancer [1], [3], [4], [9], [10], [11], [12], [13], [14], [21], [22], [23], [32], [33], [34], [35], [36], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53]
PA synthesis occurs in the cytoplasm of cells of all tissues. In eukaryotic cells, three PAs are synthesized from two amino acids: L-methionine and L-ornithine (an amino acid that is a part of the urea cycle) [1], [5], [9], [10], [11], [12], [35], [43], [51], [52], [53].
The precursor of PAs is ornithine, an amino acid intermediate of the urea cycle, which is derived from arginine by the action of arginase (EC 3.5.3.1). Ornithine is then decarboxylated by ornithine decarboxylase (ODC, EC 4.1.1.17), a pyridoxal phosphate-dependent enzyme, the first and the rate-limiting enzyme in PA biosynthesis [1], [11], [54], [55], [56], [57]. ODC is characterized by an extremely short half-life, ranging from 15 to 25 min in eukaryotic cells, in comparison with many mammalian enzymes, the half-lives of which are more often expressed in days [1], [9], [10], [11], [12], [22], [54]. ODC activity is elevated in most human cancers [51]. The high level of ODC in tumor cells may be the cause of cell transformation, as it has been described as a proto-oncogene [1], [9], [10], [11], [12], [13], [21], [52], [54], [57].
The most important factor limiting ODC content appears to be antizyme (Az). This protein binds to ODC and increases its binding to the 26S proteasome. ODC is then degraded and Az is released, resulting in a further degradation of ODC [1], [40], [57], [58], [59], [60], [61], [62], [63].
Az is involved not only in ODC degradation, but also in the negative regulation of PA transport. Degradation is controlled by the pathway involving two proteins termed antizyme (Az) and antizyme inhibitor (AzI). Az can induce cell death associated with a rapid decline of intracellular PA contents [59]. Az inhibits the uptake of PAs and stimulates PA excretion [60]. As a result, Azs are regulated at the translational level by the level of PAs. AzI serves as a regulator; its action is based on its high affinity for Az, which is greater than the affinity Az has for ODC. AzI interferes with the binding of Az to ODC, thus rescuing ODC from degradation. The ability of Azs to degrade ODC, inhibit PA uptake and consequently suppress cellular proliferation suggests that Azs act as tumor suppressors, while the ability of AzIs to neutralize the Az function indicates their growth-promoting and oncogenic potential [57], [60], [61], [62], [63].
Produced putrescine is then converted to higher PAs, Spd and SP, via Spd synthase and Sp synthase, respectively [64]. Another important enzyme in the biosynthesis of PAs is S-adenosylmethionine decarboxylase (AdoMetDC), the key enzyme for Spd and Sp synthesis in mammals. It catalyzes the formation of decarboxylated S-adenosylmethionine (dcAdoMet), which is used as the aminopropyl donor. This is the sole function of the decarboxylated AdoMet. The aminopropyl group, formed by the decarboxylation of AdoMet, is conjugated with putrescine, by the action of Spd synthase, to form PA Spd. Spd is conjugated with another propylamino group, by the action of Sp synthase, to form polyamine Sp. The best known aminopropyltransferases are Spd synthase and Sp synthase [64], [65], [66], [67], [68], [69], [70], [71]. AdoMet is also the methyl source for many transmethylation reactions, including DNA and histone methylation. Thus, DNA methylation and PA synthesis depend on a common substrate, AdoMet [72], [73], [74], [75], [76]. As a consequence, changes in the cellular PA levels may affect the degree of DNA methylation [71], [72], [73], [76]. DcAdoMet is a poor methyl group donor; it starts inhibiting DNA methylation when its concentration exceeds that of AdoMet. At the dcAdoMet/AdoMet ratio of 5:1, there is very little methyl transfer [71], [72], [73], [74]. This is potentially very significant in the biological system. All known AdoMetDCs are members of the more limited class of decarboxylases that use a covalently bound pyruvate as the prosthetic group, which is a small number of pyruvoyl-dependent decarboxylases [64], [65], [66], [67], [68], [69], [70].
PA catabolism
The catabolic pathway of Sp and Spd is a two-step process involving, in the first reaction, Spd/Sp N′-acetyltransferase and in the second reaction, PA oxidase (PAO) [1], [22], [34], [40]. Sp and Spd are first acetylated by Spd/Sp N1-acetyltransferase (SSAT) and subsequently oxidized by PAO to produce Spd and putrescine, respectively (Figure 1) [1], [10], [72], [73], [74], [75], [76]. Acetylated Sp and Spd then move into peroxisomes where they are oxidized by PAO (EC 1.5.3.11), another enzyme of PA degradation [78]. SSAT (EC 2.3.1.57) participates in PA homeostasis by regulating PA catabolism and thus PA concentrations. Acetylation of PAs is a part of the catabolic process that allows the removal of PAs from the cell [76]. This enzyme catalyzes the formation of N1-acetylspermine and N1-acetylspermidine, less charged derivatives, by the transfer of an acetyl group from acetyl-CoA to the N1 position of Spd or Sp [74], [76]. Acetylation, as the means to diminish the number of charges on PA molecules, serves as an ordered mechanism to control DNA replication and transcription in vivo [79], [80]. Acetylation neutralizes the positive charge of the epsilon amino group of lysine residues, loosening the interaction between histones and the negatively charged DNA. The acetylation of PAs reduces their affinity for DNA and nucleosomes; thus, the helical twist of DNA in nucleosomes could be regulated by cells through acetylation. Histone and PA acetylations act synergistically to modulate chromatin structure [80], [81], [82]. The acetylating enzyme, SSAT, participates in PA homeostasis by regulating PA export and catabolism. SSAT may influence cellular metabolism by perturbing the content of acetyl-CoA and ATP, fatty acid precursor, and also the product of β-oxidation of fatty acids and the process of glycolysis [78], [83].
![Figure 1: Polyamine metabolism and methionine salvage pathway (Casero and Pegg [77], modified).MATtr, methionine adenosyltransferase; AdoMetDC, S-adenosylmethionine decarboxylase; ODC, L-ornithine decarboxylase; APAO, acetylpolyamine oxidase; PAO, polyamine oxidase; MTA, methylthioadenosine.](/document/doi/10.1515/pterid-2017-0012/asset/graphic/j_pterid-2017-0012_fig_001.jpg)
Polyamine metabolism and methionine salvage pathway (Casero and Pegg [77], modified).
MATtr, methionine adenosyltransferase; AdoMetDC, S-adenosylmethionine decarboxylase; ODC, L-ornithine decarboxylase; APAO, acetylpolyamine oxidase; PAO, polyamine oxidase; MTA, methylthioadenosine.
Many histone acetylases and deacetylases can be regulated by gene promoters and gene inhibitors, which, in turn, may be controlled by various PAs [82].
Acetylpolyamine oxidase (APAO) catalyzes the conversion of N1-acetylspermine to Spd and N1-acetylspermidine to putrescine. Acetylated PAs are generally the preferred substrates of APAO (EC 1.5.3.13), a flavin containing amine oxidase, the peroxisomal enzyme present in all vertebrate tissues and biological fluids [81]. PAO may also use nonacetylated Sp and Spd in vertebrate tissues [84], [85], [86], [87].
PAO, a flavine-adenine-nucleotide (FAD) containing enzyme, is also found in all vertebrate tissues and biological fluids. PAO represents the important enzyme of peroxisomes in the catabolic pathway of Sp and Spd. PAO is especially active toward Sp and N1-acetylated derivatives (N1-acetylspermine and N1-acetylspermidine), but less toward Spd. PAO catalyzes the oxidative deamination of Sp or Spd, producing Spd or putrescine, respectively, depending on the nature of the substrate. In its enzymatic activity PAO produces ammonia (NH3), corresponding amino aldehydes and hydrogen peroxide (H2O2). Malondialdehyde (MDA) and acrolein (CH2=CHCHO) are spontaneously formed from aminoaldehydes. The products of PAO activity are potentially toxic agents. Thus, PAO plays a role in triggering and/or participating in the progression of apoptosis and tumor appearance. The coupled responses of SSAT and PAO are responsible for the prevention of the over accumulation of PAs [20], [77], [88], [89], [90], [91], [92], [93], [94].
The acetylated PA products of the SSAT reactions, N1,N12-diacetylspermine (DiAcSpm) and N1,N8-diacetylspermidine (DiAcSpd), are excreted in human urine. Thus, DiAcSpm and DiAcSpd are minor components of human urinary polyamines [94].
Diamine oxidase (DAO)
Diamine oxidase (DAO, EC 1.4.3.6) is the enzyme that degrades putrescine, the main product of PA catabolism or interconversion. DAO, a copper-containing enzyme, catalyzes the oxidation of diamine putrescine to gamma-amino-butiric acid (GABA) or MDA [95], [96], [97], [98]. GABA, synthesized from putrescine, is well known to function as an inhibitory neurotransmitter in the central nervous system [99], [100].
Backconversion pathway of PAs
The decarboxylation and propylamine transferase reactions are practically irreversible. So, the entirely distinct system exists to convert higher PAs back to putrescine (Figure 1). This system utilizes two different enzymes, a cytosolic SSAT and a peroxisomal flavoprotein PAO [50].
5′-Methylthioadenosine
5′-Methylthioadenosine (MTA) is produced from AdoMet during the biosynthetic pathway of the PAs Sp and Spd. S-adenosylmethionine decarboxylase (AdoMetDC) catalyzes the formation of decarboxylated AdoMet, which is used as the aminopropyl donor in this reaction in which an amine acceptor (putrescine or Spd) forms PA (Spd or Sp). MTA formed in cellular processes is converted back to methionine in the methionine salvage pathway (Figure 1) [1], [50], [101].
MTA is a sulfur-containing nucleoside present in all mammalian tissues, including the prostate, liver, lung, spleen, kidney and heart. MTA is a potent inhibitor of cell proliferation. This compound contains methylthioribose-1-phosphate (MTR-1P) with adenine. MTA is normally rapidly degraded by MTA phosphorylase (MTAP). The action of MTAP starts the pathway by which the purine and methylthio-moieties of AdoMet, which are not used for PAs, are recycled via salvage pathways. Many tumors lack MTAP due to either the deletion or promoter hypermethylation of the MTAP gene. This lack of MTAP activity causes MTA accumulation. Spd-synthase and Sp-synthase are inhibited by MTA with Sp synthase being more sensitive [64], [65], [66], [67], [68], [69], [70]. MTAP is a well-known tumor suppressor and a regulator of purine and pyrimidine synthesis and metabolism. It influences numerous critical responses of the cell, including the regulation of gene expression, proliferation, differentiation and apoptosis. Several previous studies have shown that MTAP could be a prognostic marker in multiple cancer types [100], [101], [102], [103], [104], [105], [106]. MTAP is a gatekeeper which controls the balance between the de novo synthesis and the purine salvage synthesis, the folate cycle and the regulation of DNA methylation status. The loss of MTAP function leads to tumorigenesis and correlates with the survival of patients [103], [104], [105], [106], [107]. The hypermethylated state of promoter is the major modification of MTAP activity [106].
PA functions
Recent studies have identified that PAs have a number of key area effects on cellular metabolism [13]. But, despite several decades of intensive research work, their exact physiological functions remains obscure [108].
PAs have a multitude of functions affecting cell differentiation, proliferation, or cell growth and development [12]. At physiological pH, PAs carry a positive charge on each nitrogen atom; as polyanions they interact with nucleic acids, RNA and DNA, and proteins, glycoproteins and phospholipids in cell membranes. PA interaction with nucleic acids has been shown to affect the stability of double-stranded DNA; at physiological concentration, PAs can condense DNA and stabilize DNA compact forms [8], [24], [27], [28], [109], [110], [111]. These interactions influence the rate of biochemical reactions. The acetylation of PAs seems to be an important mode of PA-chromatin interaction regulation. Purified histone acetyltransferase (HAT) also possesses PA acetylation activity; thus, histones and PA acetylation may occur in tandem to regulate DNA replication and transcription [83].
Most PAs exist as a PA-RNA complex, and this complex influences protein synthesis. PA-mediated effects on RNA are frequently distinct from those of divalent cations (i.e. Mg2+), confirming their roles as independent molecular entities which help drive RNA-mediated processes [27]. By binding to DNA and RNA, PAs influence protein synthesis by the regulation of gene transcription and translation; they regulate translation both at the initiation and at the elongation steps. The group of genes whose expression is enhanced by PAs at the level of translation has been referred to as a “polyamine modulon” [9], [10], [11], [12], [34], [35], [111], [112], [113], [114], [115], [116].
The primary function of PAs, Spd and Sp, in mammalian cells is in translation [3], [14]. Spd participates in the synthesis of eukaryotic initiation factor 5A (eIF5A), serving as the source of hypusine by the enzyme deoxyhypusine synthase [14], [112], [113], [114], [115], [116], [117]. At the global level, PAs may regulate translation via hypusination of the putative translation factor eIF5A, the only protein that contains the unusual Spd-derived amino acid residue hypusine, which is essential for its biological activity [34], [35], [114], [115], [116], [117]. The requirement of eIF5A, which was originally suggested to function as the translation initiation factor, for cell growth raises the possibility that eIF5A hypusination may represent the major or even the entire requirement of PAs for cellular proliferation [14], [113], [114], [115]. But, Tomitori et al. [91] demonstrated that both eIF5A and PAs influenced cell growth, but in an independent manner. The decrease in either active eIF5A or PAs inhibited cell growth, indicating that eIF5A and PAs were independently involved in cell growth [115], [116], [117].
Also, PAs influence protein synthesis by the correct assembly of the ribosomal subunits [37], [38], [39]. PAs are repressors of transcription in vivo [14], [118]. They influence the formation of compact chromatin [118]. One role of histone hyperacetylation is to antagonize the ability of PAs to stabilize the highly condensed states of chromosomal fibers [33], [119]. They bind stronger AT-rich DNA compared to the GC-rich DNA and the binding varies depending on the charge on the PAs (Sp, Spd and putrescine) [33], [110].
PAs can interact with PGs with equal to or even higher affinity than that of DNA [5]. These highly charged polysaccharides consist of variably sulfated glucosamine residues and glucuronic or iduronic acids. The negatively charged carboxyl and sulfate groups can interact with the positive charges on PAs [5], [45], [46], [47], [48], [110].
PAs, especially Spd, affect lipid metabolism. They are present in mast cell secretory granules and indicate an essential role of these polycations during the biogenesis and homeostasis of these organelles. Mast cell secretory granules contain highly sulfated serglycin PG; PAs interact with PGs [120]. Lipid metabolism is a strong regulator of health and lifespan. Spd is a key factor in the process of adipogenesis. SSAT may influence cellular metabolism by perturbing the content of acetyl-CoA and ATP, fatty acid precursor, and also the product of β-oxidation of fatty acid and the process of glycolysis [74], [78], [83], [88]. The changes in the lipid profile will modulate membrane fluidity and lead to oxidative damage, as well as signaling. PA levels decrease with age in many organisms. This decrease could play a part in the cellular aging phenotype [120], [121], [122], [123].
PAs may play a direct and crucial role in cellular immunity and may be involved not only in inflammatory processes, but also in cytocidal activities. PAs could be considered as natural immunosuppressive factors. The immune function in cancer patients is suppressed; PAs represent one of the causative factors of immune suppression. Immune cells in an environment with increased PA levels lose anti-tumor immune functions. Increased blood PA levels, often observed in cancer patients, have negative impacts on patient prognosis and are associated with tumor progression [1], [3], [9], [13], [124], [125], [126].
PAs are potential anti-inflammatory agents. They have anti-inflammatory activity in acute and chronic inflammation, which can be attributed to their anti-oxidant and/or lysosomal stabilization properties [127], [128], [129], [130], [131]. The endogenous Sp normally inhibits the innate inflammatory response by restraining macrophages [126].
The association between inflammation and cancer has been illustrated by numerous epidemiologic and clinical studies [127], [128], [129], [130], [131]. The increased PA levels in cancer tissues and in blood may be one of the factors that hinder the immune function of cancer patients [127]. The increased blood Sp levels may be an important factor in the suppression of the anti-tumor immune cell function [132].
The PA levels in serum and in urine of healthy human beings are age related, declining progressively with increasing age [123].
PAs and cancer
PAs, putrescine, Spd and Sp, play an essential role in the proliferation and development of cancer cells. There is an enormous body of evidence for the important role of PAs in cancer. Cancer cells are rich in PAs [1], [2], [3], [4], [11], [21], [23], [51], [52], [93], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142], [143], [144]. PA levels are elevated in organs and tissues, as well as in the blood and urine of cancer patients due to the increased synthesis and active intercellular transport in cancer cells. The high levels of intracellular PAs promote HAT activity, resulting in chromatin hyperacetylation and the facilitation of tumor growth. Many histone acetylases and deacetylases can be regulated by gene promoters and gene inhibitors, which, in turn, may be controlled by various PAs [82]. Increased PA concentrations in tissues and organs correlate with diseases of neoplastic origin. The exact role of PAs in cancer is still unclear; it is likely that their high levels in cancer cells can help these cells maintain their proliferation capacity [21], [93], [140], [141], [142], [143], [144].
PAs are important for gene expression due to their ability to bind nucleic acids and proteins [1], [2], [3], [4]. They are gene-specific repressors of transcription in vivo, and one role of Gcn5p HAT is to antagonize PA function. PA and histone acetylations act synergistically to modulate the chromatin structure [118], [119]. In the presence of the increased amount of Sp, DNA transcriptional activity is enhanced [145].
The molecular linkages of PAs to signal transduction and oncogene/cancer suppressor gene expressions in various cell types have been reported. PA depletion and the inhibition of eIF5A hypusination inhibit cellular proliferation [116]. Elevated intracellular PA levels promote histone acetylation in proliferating cells, suggesting a mechanism by which altered PA biosynthesis contributes to the aberrant expression of genes, facilitating tumor growth [77], [81]. The dysregulation of cellular PAs is associated with various pathological conditions, including cancer [1], [2], [6], [146], [147], [148], [149]. A continuous supply of PAs is required for cancer cell proliferation [6], [132], [134], [143], [144], [147], [148], [149], [150]. PAs should be clinically utilized to track tumor evolution and tumor response to therapy in patients at high risk [21], [23], [144]. The elevated levels of PAs have been associated with brain tumors, epithelial, breast, colon, lung, prostate and skin cancers, where the altered levels of rate-limiting enzymes in both biosynthesis and catabolism have been observed [75], [77], [80], [149], [150], [151], [152], [153], [154], [155]. The enhanced levels of PA biosynthetic enzymes, ODC and AdoMetDC, are often associated with hyperproliferation and cancer. The aberrant expression of ODC plays a causal role in tumorigenesis. ODC activity is higher in cancer tissues than in normal surrounding tissues. ODC has long been known as a marker of carcinogenesis and tumor progression [150], [151], [152], [153], [154], [155]. Human breast cancer cells overproducing AdoMetDC (~five-fold) manifest reduced ODC, while SSAT is variably increased [152]. The enhanced proliferation in colon cancer cells is associated with increased AdoMetDC activity. AdoMetDC might be a preferable target for therapeutic attempts to impair growth by reducing intracellular PA pools in colon cancer [153]. The total PA content in breast cancer tissues is higher than in benign breast diseases and correlates well with ODC activity [153], [155]. Thus, the ornithine-derived putrescine plays a key role in tumor proliferation [93], [156].
The PA-hypusine axis is a new tumor suppressor network regulating apoptosis [157], [158]. Among the new tumor suppressors, there are adenosylmethionine decarboxylase 1 (AMD1) and eukaryotic translation initiation factor 5A (eIF5A), two molecules associated with hypusine.
PAs stimulate the expression of a variety of genes, including many implicated in cell proliferation. Tumor necrosis factor-α (TNF-α) can lead to the induction of NF-κB signaling with a concomitant increase in SSAT expression in A549 and H157 non-small cell lung cancer cells [75]. SSAT is increased in tumor tissue, whereas PAO is decreased [11]. PAO activity is significantly lower in the neoplastic tissue than in the surrounding mucosa in human colorectal cancer. The changes in PAO correlate with prognostic factors, and this enzyme activity decreases as tumor histological grade increases [11], [140]. Also, the significant elevation of DAO activity is in positive correlation with neoplastic growth [97], [159].
The evidence indicates that DiAcSpm and DiAcSpd are promising novel tumor markers. The urinary excretion of these PA derivatives is increased in association with every type of cancer [94]. Remission is usually accompanied by the normal level of these compounds. Thus, they deserve more intensive study, including a study of their biochemistry and metabolism [11], [93].
Hypoxia, a common condition in cancer tissues, exerts a strong pressure on cells to separate from the tumor cluster and migrate into circulation [160]. Tsujinaka et al. [147] reported that cancer cells under hypoxia lost the regulation of PA homeostasis and increased PA uptake from surrounding tissues [160], [161], [162]. Increased PA intake accelerates the growth of established tumors [163], [164], [165]. Cells can take up PAs from extracellular sources, such as food and intestinal microbes [163]. Luminal PAs are crucially involved in the normal and neoplastic growth of the gut. The major sources of exogenous PAs come from the diet and luminal bacteria. Dietary PAs are taken up by the intestinal tract and enter the systemic circulation [164], [165], [166]. Either produced by actively proliferating normal or cancer cells, or absorbed from the gastro-intestinal tract (food and colonic microfloral population), circulating PAs could favor, in vivo, malignant cell proliferation [163], [166]. On the other hand, in cancer patients, reduced PA dietary intake has been shown to be beneficial on the quality of life [165].
PAs have been attributed as markers of neoplastic proliferation in the colon. Dietary PAs have a major role in colorectal adenoma risk assessment. These findings would confirm a previously unrecognized, modifiable dietary risk factor for colorectal adenoma [151], [164], [165]. PAs have a preferential effect in controlling nuclear proto-oncogene expression (myc, jun and fos), but the preferential role of each of the PAs on proto-oncogene expression has not been explored. The increased PA availability enhances the capability of cancer cells to invade and metastasize to new tissues while diminishing immune cells – “anti-tumor immune functions” [132], [134], [140], [141].
Biochemistry of folates and their function
Biochemistry
Folic acid, folacin or pteroylglutamic acid, is a water-soluble vitamin. The name folic acid is derived from the Latin word folium, meaning “leaf”, as it is found in many leafy plants. The best folate sources in the diet are green leafy vegetables as well as animal liver and kidney [166], [167], [168], [169]. Mammalian cells are devoid of the enzymatic capacity for folate biosynthesis and are absolutely dependent on folate uptake from exogenous dietary sources. Thus, the intestine plays a central role in regulating body folate homeostasis. The primary deposits of folate are the liver and kidney [168]. Similarly, the kidneys play a pivotal role in regulating body folate homeostasis by reabsorbing the filtered vitamin, thus preventing its loss by urine [167], [168], [169]. Folic acid refers to the oxidized synthetic compound used in dietary supplements and fortified food, while folate occurs naturally in food. It is well established that an adequate folate intake from the consumption of folate-rich foods is essential for health [168].
Folate coenzymes are specifically concerned with biochemical reactions, involving the transfer and utilization of the single carbon (C1) moieties, such as methyl-, methylene-, methenyl-, formyl or formimino-groups to various substrates in a variety of enzymatic reactions that are intimately related to the synthesis of DNA, RNA, proteins and phospholipids (lecithin and sphingomyelin) and to the metabolism of several amino acids, including methionine and others, cell division or red blood cell formation. Increased amounts of folate are required for the growth and reproduction of all body cells during periods of rapid cell regeneration and growth, such as cancer cells [168], [169].
The roles of folates
Folate is important for the prevention of diseases, such as neural tube defects (NTDs) and cardiovascular diseases [170], [171], [172], [173], [174], [175], [176], [177], [178], [179], [180], [181], [182], [183], [184]. During periods of rapid cell regeneration and growth, such as in cancer cells, increased amounts of folate are required. One-carbon metabolism is one of the metabolic pathways which have been successfully targeted in oncology. AdoMet is used for PA biosynthesis by the transfer of the propylamine moiety, and also as a methyl donor for widely biological methylation reactions [49], [71], [185], [186], [187], [188], [189]. Folate is necessary for the synthesis of methionine, for de novo purine synthesis and de novo synthesis of pyrimidine – thymidylate from deoxyuridylate. In mammalian cells, the de novo synthesis of thymidylate from deoxyuridylate is a rate-limiting step for DNA synthesis and requires 5,10-methylene tetrahydrofolate as the coenzyme [168], [169], [171], [172], [173], [174].
The methylation of DNA plays an important role in the control of gene expression and is critical during cell differentiation. Aberrations in DNA methylation have been linked to the development of cancer [190], [191], [192], [193], [194], [195], [196], [197], [198]. However, the intervention trials with high doses of folic acid have not generally shown any benefit on cancer incidence. On the other hand, it is possible that folate depletion manifests through disturbances in normal DNA, and possibly RNA, maybe through PA metabolism, producing its pro-carcinogenic effects [190], [191], [195], [196], [197].
Methionine pathways depend not only on the folate coenzyme (5-methyltetrahydrofolate), but also on coenzyme of vitamin B12 (Figure 2). The folate coenzyme plays a crucial role in methionine biosynthesis [185], [186], [187], [188], [189], [190], [191]. Thus, folate (and/or vitamin B12) deficiency can result in the decreased synthesis of methionine [192].
![Figure 2: Methionine metabolic pathways (Deacon et al. [170], modified). The folate coenzyme plays a crucial role in methionine biosynthesis. Methionine has many functions in the human body.](/document/doi/10.1515/pterid-2017-0012/asset/graphic/j_pterid-2017-0012_fig_002.jpg)
Methionine metabolic pathways (Deacon et al. [170], modified). The folate coenzyme plays a crucial role in methionine biosynthesis. Methionine has many functions in the human body.
PAs and folates are interconnected by methionine
Folates participating in methionine metabolism have a profound function in PA biosynthesis. Together with vitamin B12 and vitamin B6, methionine is the principal precursor of Spd and Sp [1], [49]. The overaccumulation of PAs induces apoptosis and cell malignant transformation, whereas PA depletion has been shown to inhibit cell proliferation [28]. The administration of folic acid to hepatectomized animals (30%) resulted in diminution of PAO activity in the regenerating rat liver tissue after hepatectomy [193]. The supplementation of experimental animals with vitamin B12 alone or together with folic acid increased the Spd and Sp levels in the rat liver. At the same time, the supplementation of experimental animals with vitamin B12 together with folic acid caused the decrease of PAO activity. Our experimental results indicated the importance of folic acid and cobalamin in PA metabolism [194].
Mild folate deficiency influences PA synthesis that occurs in all living organisms. Bistulfi et al. [195] found a significant correlation between PA biosynthesis and the amount of folate required to sustain cell line proliferation. These data demonstrate that PA biosynthesis is the critical factor in determining sensitivity to folate depletion and may be particularly important in the prostate, where the biosynthesis of PAs is characteristically high due to its secretory function. But, the interdependency of the folate and PA pathways has not yet been studied enough [196].
DNA methylation and cancer
The connection between PAs and nucleic acids appears crucial for the cellular functions of PAs. The normal mammalian development is dependent on DNA methylation [173], [174], [185], [190], [191]. The methylation of DNA by S-adenosyl-L-methionine is crucial for the regulation of gene expression and is a potentially mutagenic reaction [71], [81], [199], [200], [201], [202], [203], [204], [205], [206], [207], [208], [209], [210], [211], [212], [213], [214], [215], [216], [217], [218], [219], [220], [221], [222]. Cancer is thought to arise from DNA damage and an inappropriate expression of critical genes [184], [190]. The modulation of DNA methylation in response to folate has been demonstrated [203], [204].
PA synthesis and DNA methylation depend on a common substrate, AdoMet. As a consequence, changes in the cellular PA levels may affect the degree of DNA methylation [173], [193], [223].
DNA and histones are the targets for covalent modifications. The amino-terminal tails of the histone proteins protrude from the nucleosome core and are the subject of the diverse array of post-translational modifications that include acetylation, methylation and others. DNA methylation and histone acetylation are two of the best conserved chromatin modifications throughout eukaryotes and they are interconnected [186], [187], [188], [189]. These modifications induce significant changes in the genetic function in living cells [147], [189], [196], [197], [198], [199], [200], [201], [202], [203], [204], [205], [206], [224], [225].
All DNA aberrations enhance carcinogenesis by altering the expression of critical tumor suppressor genes and proto-oncogenes [186], [189], [206], [207], [208]. DNA methylation is directly involved in the suppression of transcription. Hypermethylation represses the transcription of the promoter sections of tumor suppressor genes, leading to gene silencing [174], [203], [206], [209], [210], [211], [212]. The inactivation of tumor suppressor genes by the hypermethylation of promoter regions is associated with a variety of human neoplasms and tumors. The hypermethylation of CpG islands associated with the tumor suppressor genes is the first manifestation of cancer [206], [207], [208], [209], [210], [211], [212].
DNA modification is one of the epigenetic phenomena, an alteration in gene expression without any change in the nucleotide sequence. DNA methylation is considered to be a stable modification associated with the epigenetic silencing of genomic loci and maintained through cellular division [173]. DNA methylation in eukaryotes involves the addition of a methyl group to the carbon 5position of the cytosine ring. This reaction is catalyzed by DNA methyltransferase, which transfers a methyl group from AdoMet in the direction of the sequence 5′-CG-3′, which is also referred to as a CpG dinucleotide, named CpG islands. DNA methyltransferases (DNMTs) comprise a family of nuclear enzymes that catalyze the methylation of CpG dinucleotides, resulting in an epigenetic methylome distinguished between normal cells and those in a diseased state, such as cancer. Tumorigenic cells contain higher levels of methylating capacity than nontumorigenic cells; the literature data show that methyltransferase activity was significantly higher in tumorigenic than in nontumorigenic cells [204], [205]. PAs, Spd and to a lesser extent Sp, control DNA methylation. Sp and Spd (but not putrescine) selectively inhibit cytosine-DNMT activity [145]. Approximately 4% of the genomic DNA is hypermethylated primarily in the form of 5-methylcytosines in CpG dinucleotides [205]. In normal human cells, the majority of CpGs are methylated. The methylation of DNA at CpG dinucleotides represents some of the most important epigenetic mechanisms involved in the control of gene expression in vertebrate cells. Large stretches of DNA can become abnormally methylated in cancer [211], [212], [213], [214], [215], [216], [217], [218].
The hypermethylation consistently observed in CpG islands, therefore, represents a change in 5-methylcytosine distribution across the genome rather than an overall increase in the total amount of methylation. The hypermethylation of promoter regions of DNA inactivates tumor suppressor genes; this is associated with a variety of human neoplasms and tumors [200], [201], [202], [203], [204], [205], [206], [207], [208], [211], [212], [213], [214], [215], [216], [217], [218], [219].
Chromatin exists either in a transcriptionally active state in which histones are acetylated or in a repressed state in which histones are not acetylated [145], [220]. The structural modifications of DNA and histones, such as methylation and acetylation, induce significant changes in the genetic function in living cells [222], [226], [227], [228].
Hypomethylation has also been identified as a cause of oncogenesis. Folate deficiency causes a shortage in nucleotides and might lead to genome instability and DNA mutations. The hypomethylation of CpG islands often leads to gene reactivation [229], [230]. Folate deficiency causes massive incorporation of uracil into human DNA (4 million per cell) and chromosome breaks [229], [230]. The likely mechanism is the deficient methylation of dUMP to dTMP and the subsequent incorporation of uracil into DNA. Such breaks could contribute to an increased risk of cancer [193], [229], [230].
Cancer-associated DNA hypomethylation is as prevalent as cancer-linked hypermethylation, but these two types of epigenetic abnormalities usually seem to affect different DNA sequences [190], [223], [231], [232]. DNA methylation and demethylation are considerably more dynamic than previously thought and may be involved in the repression and derepression of gene activity during the lifespan of a cell [217], [218]. In general, DNA methylation represses transcription, and the loss of methylation is associated with gene activation [192], [193], [193], [194], [195], [196], [197], [198], [199], [200], [201], [202], [203], [204], [205], [206], [207], [208], [209], [210], [211], [212], [224], [225]. There is considerable evidence that aberrant DNA methylation plays an integral role in oncogenesis [186], [187], [188], [189], [190], [191], [195], [196], [197], [198], [199], [200], [203], [204], [205], [206], [207], [208], [209], [210], [211], [223], [224], [225].
Histone acetylation and methylation are two major modifications that function as a specific transcription regulator and are interconnected [220], [221], [222]. DNA methylation takes part in this process by inducing decreased levels of chromatin acetylation [229]. Also, the methylation of messenger RNAs (mRNAs) at 5′-terminal cap plays an important role in mRNA export from the nucleus, efficient translation and the protection of the integrity of mRNAs [186], [195].
Two classes of enzymes reversibly control the acetylation level of histones: HATs and histone deacetylases (HDACs). In general, transcriptional activators recruit HATs, whereas transcriptional repressors and co-repressors associate with HDACs [215].
The PA and histone acetylations act synergistically to modulate the chromatin structure [12], [80]. The aberrations in chromatin remodeling are associated with cancer formation [21], [80], [233], [234], [235]. PAs enhance the action of HATs either directly or indirectly [145]. HAT also possesses PA acetylation activity; thus, histone and PA acetylations may occur in tandem to alter the structure/function of the nucleosome, thereby regulating DNA replication and transcription. The acetylation of a PA is important for gene expression due to their ability to bind to nucleic acids and proteins [8], [224]. One role of the Gcn5p histone acetyltransferase is to antagonize PA function [118], [119], [196]. Hypoacetylated chromatin domains are often correlated with transcriptional inactivity [81], [82], [119]. Many histone acetylases and deacetylases can be regulated by gene promoters and gene inhibitors, which, in turn, may be controlled by various PAs [93]. The contribution of PAs to transcriptional regulation in vivo may also be controlled by the balance between histone acetylation and deacetylation reactions. PAs have linkages to the actions of epithelial growth factor, transforming tumor growth factor β, TNF-α, and hepatocyte growth factor and to several oncogenes including NF-κB, c-myc, c-jun, and c-fos and cancer suppressor genes, including p53 [93].
The PA amount in the cells, especially Sp, may affect the transcriptional activity of DNA. In the presence of an increase in the Sp concentration, transcriptional activity is enhanced [119]. Elevated intracellular PA levels increase histone acetylation in proliferating cells, suggesting the mechanism by which altered PA biosynthesis contributes to aberrant expression of genes, facilitating tumor growth [93].
Acetylation neutralizes the positive charge of the epsilon amino group of lysine residues, loosening the interaction between histones and the negatively charged DNA. The acetylation of PAs reduces their affinity for DNA and nucleosomes; thus, the helical twist of DNA in nucleosomes could be regulated by cells through acetylation [81], [82], [93], [119].
Acetylation as the means of diminishing the number of charges on PA molecules serves as an ordered mechanism to control DNA replication and transcription in vivo. One of the major functions of histone acetylation is to open chromatin to allow transcription factors to gain access to the regulatory elements in DNA [82].
Sp seems to play important roles in inhibiting age-associated and PA-deficient induced abnormal gene methylation, as well as pathological changes, including tumorigenesis [161].
PA and folates in carcinogenesis
The impact of folate on health and disease, particularly pregnancy complications and congenital malformations, is without doubt [167], [168], [169], [175], [176], [235], [236]. It is well established that adequate folate intake from the consumption of folate-rich foods is essential for health [167], [168], [169]. However, an entirely protective role of folate against carcinogenesis has been questioned. Over the past few years, the world population has been exposed to the significant increase in folate intake, for which no data on safety exist. Increased folate intake may be associated with unexpected adverse effects [237], [238], [239], [240], [241], [242], [243]. Although folic acid is generally regarded as safe, folic acid fortification may have adverse effects. Maintaining adequate folate levels may be important in lowering the risk of cancer. Recent data indicate that an excessive intake of synthetic folic acid (from high-dose supplements or fortified foods) may increase human cancers by accelerating the growth of precancerous lesions. Inadequate folate intake contributes to genome instability and chromosomebreakage that often characterize cancer development [190], [191], [199], [203], [238].
When the intake of folic acid is higher than 400μg/day, unmetabolized folic acid appears in peripheral blood and this form of folic acid may have adverse effects. The presence of unmetabolized folic acid (PteGlu) in blood is one of the etiopathogenetic causes of colorectal cancer [242], [243]. The biochemical and physiologic consequences of the increased exposure to circulating folic acid are unknown, but it is very important to understand the effects of chronic exposure to circulating folic acid [238], [239], [240], [241], [242], [243], [244], [245], [246].
Folate has a dual effect on cancer: protecting against cancer initiation, but facilitating the progression and growth of preneoplastic cells; a low folate status might inhibit colorectal carcinogenesis and a high folate status may promote colorectal carcinogenesis. These initial dual effects of folate may be explained by their function in the nucleotide synthesis. Rapidly proliferating tissues, including tumors, have an increased requirement for nucleotides; many cancers up-regulate folate receptors, and antifolate drugs are efficacious in cancer treatment [240], [241].
Data from the majority of human studies suggest that a high dose of folic acid supplementation may enhance the risk of carcinogenesis in certain circumstances [189], [206], [242], [243], [244], [245], [246], [247], [248], [249], [250]. Folic acid supplementation may enhance the development and progression of already existing, undiagnosed premalignant and malignant lesions. Folate supplementation can be preventive before a neoplastic transformation, but can act as a growth promoter in neoplastic cells [203], [246].
The possibility of the cancer-promoting effect of folic acid supplementation needs to be considered by a careful monitoring of the long-term effects of folic acid fortification on a vast majority of the population, who intake folic acid from fortified products and are not at risk of NTDs [203].
Conclusion
We believe that the understanding of the metabolism of PAs and their roles in cellular metabolism, in physiological and pathological circumstances, could help in solving the problem of the folate supplementation-cancer relationship. Further study is necessary to clarify the interactions between folate and PA metabolism and to determine whether PAs are involved in the damaging effects of folate oversupplementation.
Conflict of interest statement: The authors have declared no conflicts of interest.
Funding: This study was funded by Ministarstvo Prosvete, Nauke i Tehnološkog Razvoja, (Grant/Award Number: III41018).
References
1. Persson L. Polyamine homoeostasis. Essays Biochem 2009;46:11–24.10.1042/bse0460002Search in Google Scholar PubMed
2. Raina A, Janne J. Physiology of the natural polyamines putrescine, spermidine and spermine. Med Biol 1975;53:121–47.Search in Google Scholar PubMed
3. Tabor CW, Tabor H. Polyamines. Ann Rev Biochem 1984;53:749–90.10.1146/annurev.bi.53.070184.003533Search in Google Scholar PubMed
4. Morgan DM. Polyamines. An overview. Mol Biotechnol 1999;11:229–50.10.1007/BF02788682Search in Google Scholar PubMed
5. Pegg AE. Recent advances in the biochemistry of polyamines in eukaryotes. Biochem J 1986;234:249–62.10.1042/bj2340249Search in Google Scholar PubMed PubMed Central
6. Jänne J, Alhonen L, Leinonen P. Polyamines: from molecular biology to clinical applications. Ann Med 1991;23:241–59.10.3109/07853899109148056Search in Google Scholar PubMed
7. Yatin M. Polyamines in living organisms. J Cell Mol Biology 2002;1:57–67.10.2307/4451033Search in Google Scholar
8. Jänne J, Alhonen L, Pietilä M, Keinänen TA. Genetic approaches to the cellular functions of polyamines in mammals. J Biochem 2004;271:877–94.10.1111/j.1432-1033.2004.04009.xSearch in Google Scholar PubMed
9. Tabor H, Tabor CW. Biosynthesis and metabolism of 1,4-diaminobutane, spermidine, spermine, and related amines. Adv Enzymol Relat Areas Mol Biol 2006;36:203–68.10.1002/9780470122815.ch7Search in Google Scholar PubMed
10. Pegg AE. Mammalian polyamine metabolism and function. IUBMB Life 2009;61:880–94.10.1002/iub.230Search in Google Scholar PubMed PubMed Central
11. Wallace HM. The polyamines: past, present and future. Essays Biochem 2009;46:1–9.10.1042/bse0460001Search in Google Scholar PubMed
12. Pegg AE, Casero RA. Current status of the polyamine research field. Methods Mol Biol 2011;720:3–35.10.1007/978-1-61779-034-8_1Search in Google Scholar PubMed PubMed Central
13. Miller-Fleming L, Olin-Sandoval V, Campbel K, Remaining RM. Mysteries of molecular biology: the role of polyamines in the cell. J Mol Biol 2015;427:3389–406.10.1016/j.jmb.2015.06.020Search in Google Scholar PubMed
14. Mandal S, Mandal A, Johansson HE, Orjalo AV, Park MH. Depletion of cellular polyamines, spermidine and spermine, causes a total arrest in translation and growth in mammalian cells. Proc Natl Acad Sci USA 2013;110:2169–74.10.1073/pnas.1219002110Search in Google Scholar PubMed PubMed Central
15. Schuber F. Influence of polyamines on membrane functions. Biochem J 1989;260:1–10.10.1042/bj2600001Search in Google Scholar PubMed PubMed Central
16. Coburn FR. Polyamine effects on cell function: possible central role of plasma membrane PI(4,5)P2. J Cell Physiol 2009;221:544–51.10.1002/jcp.21899Search in Google Scholar PubMed
17. Williams K. Interactions of polyamines with ion channels. Biochem J 1997;325:289–97.10.1042/bj3250289Search in Google Scholar PubMed PubMed Central
18. Desforges B, Curmi PA, Bounedjah O, Nakib S, Hamon L, De Bandt J-P, et al. An intercellular polyamine transfer via gap junctions regulates proliferation and response to stress in epithelial cells. Mol Biol Cell 2013;15:1529–43.10.1091/mbc.e12-10-0729Search in Google Scholar PubMed PubMed Central
19. Seiler N, Raul F. Polyamines and apoptosis. J Cell Mol Med 2005;9:623–42.10.1111/j.1582-4934.2005.tb00493.xSearch in Google Scholar PubMed PubMed Central
20. Moschou PN, Roubelakis-Angelakis KA. Polyamines and programmed cell death. J Exp Bot 2014;65:1285–96.10.1093/jxb/ert373Search in Google Scholar PubMed
21. Pegg AE. Polyamine metabolism and its importance in neoplastic growth and as a target for chemotherapy. Cancer Res 1988;48:759–74.Search in Google Scholar PubMed
22. Moinard C, Cynober L, de Bandt JP. Polyamines: metabolism and implications in human diseases. Clin Nutr 2005;24:184–97.10.1016/j.clnu.2004.11.001Search in Google Scholar PubMed
23. Nowotarski SL, Woster PM, Casero RA Jr. Polyamines and cancer: implications for chemotherapy and chemoprevention. Expert Rev Mol Med 2013;15:e3.10.1017/erm.2013.3Search in Google Scholar PubMed PubMed Central
24. Stevens L. The biochemical role of naturally occurring polyamines in nucleic acid synthesis. Biol Rev 1970;45:1–25.10.1111/j.1469-185X.1970.tb01073.xSearch in Google Scholar PubMed
25. Snyder RD. Polyamine depletion is associated with altered chromatin structure in HeLa cells. Biochem J 1989;260:697–704.10.1042/bj2600697Search in Google Scholar PubMed PubMed Central
26. Oredsson SM. Polyamine dependence of normal cell-cycle progression. Biochem Soc Trans 2003;31:366–70.10.1042/bst0310366Search in Google Scholar PubMed
27. Lightfoot HL, Hall J. Endogenous polyamine function – the RNA perspective. Nucleic Acids Res 2014;42:11275–90.10.1093/nar/gku837Search in Google Scholar PubMed PubMed Central
28. Balasundaram D, Tyagi AK. Polyamine-DNA nexus: structural ramifications and biological implications. Mol Cell Biochem 1991;100:129–40.10.1007/BF00234162Search in Google Scholar PubMed
29. D’Agostino L, Di Luccia A. Polyamines interact with DNA as molecular aggregates. Eur J Biochem 2002;269:4317–25.10.1046/j.1432-1033.2002.03128.xSearch in Google Scholar PubMed
30. D’Agostino L, di Pietro M, Di Luccia A. Nuclear aggregates of polyamines. IUBMB Life 2006;58:75–82.10.1080/15216540600662525Search in Google Scholar PubMed
31. Iacomino G, Picariello G, D’Agostino L. DNA and nuclear aggregates of polyamines. Biochim Biophys Acta (BBA) – Mol Cell Res 2012;1823:1745–55.10.1016/j.bbamcr.2012.05.033Search in Google Scholar PubMed
32. Belting M, Havsmark B, Jonsson M, Persson S, Fransson LA. Heparan sulphate/heparin glycosaminoglycans with strong affinity for the growth-promoter spermine have high antiproliferative activity. Glycobiology 1996;6:121–9.10.1093/glycob/6.2.121Search in Google Scholar PubMed
33. Bachrach U. Naturally occurring polyamines: interaction with macromolecules. Curr Protein Pept Sci 2005;6:559–66.10.2174/138920305774933240Search in Google Scholar PubMed
34. Igarashi K, Kashiwagi K. Polyamines: mysterious modulators of cellular functions. Biochem Biophys Res Commun 2000;271:559–64.10.1006/bbrc.2000.2601Search in Google Scholar PubMed
35. Igarashi K, Kashiwagi K. Modulation of protein synthesis by polyamines. IUBMB Life 2015;67:160–9.10.1002/iub.1363Search in Google Scholar PubMed
36. Park MH, Igarashi K. Polyamines and their metabolites as diagnostic markers of human diseases. Biomol Ther 2013; 21:1–9.10.4062/biomolther.2012.097Search in Google Scholar
37. Ceung YN, Maag D, Mitchell SF, Fekete CA, Algire MA, Takacs JE, et al. Dissociation of eIF1 from the 40S ribosomal subunit is a key step in start codon selection in vivo. Genes Dev 2007;21:1217–30.10.1101/gad.1528307Search in Google Scholar PubMed
38. Moruzzi G, Barbiroli B. Caldarera CM. Polyamines and nucleic acid metabolism in chick embryo. Incorporation of labelled precusors into nucleic acids of subcelllular fractions polyribosomal paterns. Biochem J 1968;107:609.10.1042/bj1070609Search in Google Scholar
39. Shimogori T, Kashiwagi K, Igarashi K. Spermidine regulation of protein synthesis at the level of initiation complex formation of MettRNAi, mRNA and ribosomes. Biochem Biophys Res Commun 1996;223:544–8.10.1006/bbrc.1996.0931Search in Google Scholar PubMed
40. Wallace HM, Duthie J, Evans DM, Lamond S, Nicoll KM, Heys SD. Alterations in polyamine catabolic enzymes in human breast cancer tissue. Clin Cancer Res 2000;6:3657–61.Search in Google Scholar PubMed
41. Thomas T, Thomas TJ. Polyamines in cell growth and cell death: molecular mechanisms and therapeutic applications. Cell Mol Life Sci 2001;58:244–58.10.1007/PL00000852Search in Google Scholar PubMed
42. Heby O, Persson L. Molecular genetics of polyamine synthesis in eukaryotic cells. Trends Biochem Sci 1990;15:153–8.10.1016/0968-0004(90)90216-XSearch in Google Scholar PubMed
43. Wallace HM, Fraser AV, Hughes A. A perspective of polyamine metabolism. Biochem J 2003;376:1–14.10.1042/bj20031327Search in Google Scholar PubMed PubMed Central
44. Uemura T, Gerner EW. Polyamine transport systems in mammalian cells and tissues. Methods Mol Biol 2011;720:339–48.10.1007/978-1-61779-034-8_21Search in Google Scholar PubMed PubMed Central
45. Lander AD, Scott BS. The elusive functions of proteoglycans. In vivo veritas. J Cell Biol 2000;148:227–32.10.1083/jcb.148.2.227Search in Google Scholar PubMed PubMed Central
46. Belting M, Persson S, Fransson LÅ. Proteoglycan involvement in polyamine uptake. Biochem J 1999;338:317–23.10.1042/bj3380317Search in Google Scholar PubMed
47. Welch J, Svensson K, Kucharzewska P, Belting M. Heparan sulfate proteoglycan-mediated polyamine uptake. Methods Mol Biol 2011;720:327–38.10.1007/978-1-61779-034-8_20Search in Google Scholar PubMed
48. Belting M, Borsig L, Fuster MM, Brown Ji R, Persson L, Fransson LÅ, et al. Tumor attenuation by combined heparan sulfate and polyamine depletion. Proc Natl Acad Sci USA 2002;99:371–6.10.1073/pnas.012346499Search in Google Scholar PubMed PubMed Central
49. Loenen WA. S-adenosylmethionine: jack of all trades and master of everything? Biochem Soc Trans 2006;34:330–3.10.1042/BST0340330Search in Google Scholar PubMed
50. Sauter M, Moffatt B, Saechao MC, Wirtz RH. Methionine salvage and S adenosylmethionine: essential links between sulfur, ethylene and polyamine biosynthesis. Biochem J 2013;451: 145–54.10.1042/BJ20121744Search in Google Scholar PubMed
51. Gerner EW, Meyskens FL. Polyamines and cancer: old molecules, new understanding. Nat Rev Cancer 2004;4:781–92.10.1038/nrc1454Search in Google Scholar PubMed
52. Pegg AE, Feith DJ. Polyamines and neoplastic growth. Biochem Soc Trans 2007;35:295–9.10.1042/BST0350295Search in Google Scholar PubMed
53. Battaglia V, De Stefano Shields C, Murray-Stewart T, Casero RA. Polyamine catabolism in carcinogenesis: potential targets for chemotherapy and chemoprevention. Amino Acids 2014;46:511–9.10.1007/s00726-013-1529-6Search in Google Scholar PubMed PubMed Central
54. Auvinen M, Paasinen A, Andersson LC, Holtta E. Ornithine decarboxylase activity is critical for cell transformation. Nature 1992;360:355–8.10.1038/360355a0Search in Google Scholar PubMed
55. Coffino P. Regulation of cellular polyamines by antizyme. Nat Rev Mol Cell Biol 2001;2:188–94.10.1038/35056508Search in Google Scholar PubMed
56. Mangold U. The antizyme family: polyamines and beyond. IUBMB Life 2005;57:671–6.10.1080/15216540500307031Search in Google Scholar PubMed
57. Palanimurugan R, Scheel H, Hofmann K, Jürgen Dohmen R. Polyamines regulate their synthesis by inducing expression and blocking degradation of ODC antizyme. EMBO J 2004;23: 4857–67.10.1038/sj.emboj.7600473Search in Google Scholar PubMed PubMed Central
58. Serganov A. The long and the short of riboswitches. Curr Opin Struct Biol 2009;19:251–9.10.1016/j.sbi.2009.02.002Search in Google Scholar PubMed PubMed Central
59. Iwata S, Sato Y, Asada M, Takagi M, Tsujimoto A, Inaba T, et al. Anti-tumor activity of antizyme which targets the ornithine decarboxylase (ODC) required for cell growth and transformation. Oncogene 1999;18:165–72.10.1038/sj.onc.1202275Search in Google Scholar PubMed
60. Sakata K, Kashiwagi K, Igarashi K. Properties of a polyamine transporter regulated by antizyme. Biochem J 2000;347:297–303.10.1042/bj3470297Search in Google Scholar PubMed
61. Keren-Paz A, Bercovich Z, Kahana C. Antizyme inhibitor: a defective ornithine decarboxylase or a physiological regulator of polyamine biosynthesis and cellular proliferation. Biochem Soc Trans 2007;35:311–3.10.1042/BST0350311Search in Google Scholar PubMed
62. Kahana C. Regulation of cellular polyamine levels and cellular proliferation by antizyme and antizyme inhibitor. Essays Biochem 2009;46:47–61.10.1042/bse0460004Search in Google Scholar PubMed
63. Kahana C. Antizyme and antizyme inhibitor, a regulatory tango. Cell Mol Life Sci 2009;66:2479–88.10.1007/s00018-009-0033-3Search in Google Scholar PubMed
64. Pegg AE, Michael AJ. Spermine synthase. Cell Mol Life Sci 2010;67:113–21.10.1007/s00018-009-0165-5Search in Google Scholar PubMed PubMed Central
65. Pegg EA. The function of spermine. IUBMB Life 2014;66:8–18.10.1002/iub.1237Search in Google Scholar PubMed
66. Ikeguchi Y, Bewley MC, Pegg AE. Aminopropyltransferases: function, structure and genetics. J Biochem 2006;139:1–9.10.1093/jb/mvj019Search in Google Scholar PubMed
67. Wu H, Min J, Ikeguchi Y, Zeng H, Dong A, Loppnau P, et al. Structure and mechanism of spermidine synthases. Biochemistry 2007;46:8331–9.10.1021/bi602498kSearch in Google Scholar PubMed
68. Pegg AE. S-adenosylmethionine decarboxylase. Essays Biochem 2009;46:25–45.10.1042/bse0460003Search in Google Scholar PubMed
69. Pegg AE, Wang X, Schwartz CE, McCloskey DE. Spermine synthase activity affects the content of decarboxylated S-adenosylmethionine. Biochem J 2011;433:139–44.10.1042/BJ20101228Search in Google Scholar PubMed
70. Yerlikaya A. Polyamines and S-adenosylmethionine decarboxylase. Turk J Biochem 2004;29:208–14.Search in Google Scholar
71. Fontecave M, Atta M, Mulliez E. S-adenosylmethionine: nothing goes to waste. Trends Biochem Sci 2004;29:243–9.10.1016/j.tibs.2004.03.007Search in Google Scholar PubMed
72. Pegg AE. Spermidine/spermine-N1-acetyltransferase: a key metabolic regulator. Am J Physiol Endocrinol Metab 2008;294:E995–E1010.10.1152/ajpendo.90217.2008Search in Google Scholar PubMed
73. Casero RA, Pegg AE. Spermidine/spermine N1-acetyltransferase: the turning point in polyamine metabolism. FASEB J 1993;7:653–61.10.1096/fasebj.7.8.8500690Search in Google Scholar PubMed
74. Kramer DL, Diegelman P, Jell J, Vujcic S, Merali S, Porter CW. Polyamine acetylation modulates polyamine metabolic flux, a prelude to broader metabolic consequences. J Biol Chem 2008;283:4241–51.10.1074/jbc.M706806200Search in Google Scholar PubMed
75. Babbar N, Hacker A, Huang Y, Casero RA Jr. Tumor necrosis factor alpha induces spermidine/spermine N1-acetyltransferase through nuclear factor kappaB in non-small cell lung cancer cells. J Biol Chem 2006;281:24182–92.10.1074/jbc.M601871200Search in Google Scholar PubMed
76. Seiler N. Functions of polyamine acetylation. Can J Physiol Pharmacol 1987;65:2024–35.10.1139/y87-317Search in Google Scholar PubMed
77. Casero RA, Pegg AE. Polyamine catabolism and disease. Biochem J 2009;421:323–38.10.1042/BJ20090598Search in Google Scholar PubMed
78. Pirinen E, Kuulasmaa T, Pietila M, Heikkinen S, Tusa M, Itkone Paula, et al. Enhanced polyamine catabolism alters homeostatic control of white adipose tissue mass, energy expenditure, and glucose metabolism. Mol Cell Biol 2007;l27:4953–67.10.1128/MCB.02034-06Search in Google Scholar
79. Seiler N. Catabolism of polyamines. Amino Acids 2004; 26:217–33.10.1007/s00726-004-0070-zSearch in Google Scholar PubMed
80. Wang Y, Casero RA. Mammalian polyamine catabolism: a therapeutic target, a pathological problem, or both? J Biochem 2006;139:17–25.10.1093/jb/mvj021Search in Google Scholar PubMed
81. Matthews HR. Polyamines, chromatin structure and transcription. Bioessays 1993;15:561–6.10.1002/bies.950150811Search in Google Scholar PubMed
82. Hobbs CA, Gilmour SK. High levels of intracellular polyamines promote histone acetyltransferase activity resulting in chromatin hyperacetylation. J Cell Biochem 2000;77:345–60.10.1002/(SICI)1097-4644(20000601)77:3<345::AID-JCB1>3.0.CO;2-PSearch in Google Scholar PubMed
83. Jell J, Merali S, Hensen ML, Mazurchuk R, Spernyak JA, Diegelman P, et al. Genetically altered expression of spermidine/spermine N1-acetyltransferase affects fat metabolism in mice via Acetyl-CoA. J Biol Chem 2007;82:8404–13.10.1074/jbc.M610265200Search in Google Scholar
84. Wang, Y, Devereux W, Woster PM, Stewart TM, Hacker A, Casero RA. Cloning and characterization of a human polyamine oxidase that is inducible by polyamine analogue exposure. Cancer Res 2001;61:5370–3.Search in Google Scholar PubMed
85. Vujcic S, Diegelman P, Bacchi CJ, Kramer DL, Porter CW. Identification and characterization of a novel flavin-containing spermine oxidase of mammalian cell origin. Biochem J 2002;367:665–75.10.1042/bj20020720Search in Google Scholar PubMed
86. Murray-Stewart T, Wang Y, Devereux W, Casero RA. Cloning and characterization of multiple human polyamine oxidase splice variants that code for isoenzymes with different biochemical characteristics. Biochem J 2002;368:673–7.10.1042/bj20021587Search in Google Scholar PubMed
87. Vujic S, Liang P, Diegelman P, Kramer DL, Porter CW. Genomic identification and biochemical characterization of the mammalian polyamine oxidase involved in polyamine back-conversion. Biochem J 2003;370:19–28.10.1042/bj20021779Search in Google Scholar PubMed
88. Holtta E. Oxidation of spermidine and spermine in rat liver: purification and properties of polyamine oxidase. Biochemistry 1977;16:91–100.10.1021/bi00620a015Search in Google Scholar PubMed
89. Seiler N. Polyamine oxidase, properties and functions. Prog Brain Res 1995;106:333–44.10.1016/S0079-6123(08)61229-7Search in Google Scholar PubMed
90. Seiler N. How important is the oxidative degradation of spermine? Mini review article. Amino Acids 2004;26:317–9.10.1007/s00726-004-0090-8Search in Google Scholar PubMed
91. Tomitori H, Usui T, Saeki N, Ueda S, Kase H, Nishimura K, et al. Polyamine oxidase and acrolein as novel biochemical markers for diagnosis of cerebral stroke. Stroke 2005;36:2609–17.10.1161/01.STR.0000190004.36793.2dSearch in Google Scholar PubMed
92. Pegg AE. Toxicity of polyamines and their metabolic products. Chem Res Toxicol 2013;26:1782–800.10.1021/tx400316sSearch in Google Scholar PubMed
93. Criss WE. A Review of polyamines and cancer. Turk J Med Sci 2003;33:195–205.Search in Google Scholar
94. Kawakita M, Hiramatsu K. Diacetylated derivatives of spermine and spermidine as novel promising tumor markers. J Biochem 2006;139:315–22.10.1093/jb/mvj068Search in Google Scholar PubMed
95. Anderson CA, Henningsson S, Persson L, Rosengren E. Aspect on diamine oxidase activity and its determination. Acta Physiol Scand 1978;102:159–66.10.1111/j.1748-1716.1978.tb06059.xSearch in Google Scholar PubMed
96. Wolvekamp MC, de Bruin RW. Diamine oxidase: an overview of historical, biochemical and functional aspects. Dig Dis 1994;12:2–14.10.1159/000171432Search in Google Scholar PubMed
97. Linsalata M, Russo F, Cavallini A, Berloco P, Di Leo A. Polyamines, diamine oxidase, and ornithine decarboxylase activity in colorectal cancer and in normal surrounding mucosa. Dis Colon Rectum 1993;36:662–7.10.1007/BF02238593Search in Google Scholar PubMed
98. Konishi H, Nakajama T, Saano I. Metabolism of putrescine in the central nervous system. J Biochem 1977;81:355–60.10.1093/oxfordjournals.jbchem.a131466Search in Google Scholar PubMed
99. Caron PC, Cote LJ, Kremzner LT. Putrescine, a source of gamma-aminobutyric acid in the adrenal gland of the rat. Biochem J 1988;251:559–62.10.1042/bj2510559Search in Google Scholar PubMed
100. Albers E. Metabolic characteristics and importance of the universal methionine salvage pathway recycling methionine from 5,-Methylthioadenosine. IUBMB Life 2009;61:1132–42.10.1002/iub.278Search in Google Scholar PubMed
101. Avila MA, García-Trevijano ER, Lu SC, Corrales FJ, Mato JM. Methylthioadenosine. Int J Biochem Cell Biol 2004;36:2125–30.10.1016/j.biocel.2003.11.016Search in Google Scholar PubMed
102. Kamatani N, Carson DA. Abnormal regulation of methylthioadenosine and polyamine metabolism in methylthioadenosine phosphorylase-deficient human leukemic cell lines 1. Cancer Res 1980;40:4178–82.Search in Google Scholar
103. Williams-Ashman HG, Seidenfeld J, Galletti P. Trends in the biochemical pharmacology of 5′-deoxy-5′-methylthioadenosine. Biochem Pharmacol 1982;31:277–88.10.1016/0006-2952(82)90171-XSearch in Google Scholar PubMed
104. Nobori T, Szinai I, Amox D, Parker B, Olopade OI, Buchhagen DL, et al. Methylthioadenosine phosphorylase deficiency in human non-small cell lung cancers. Cancer Res 1993;53:1098–101.Search in Google Scholar PubMed
105. Hellerbrand C, Muhlbauer M, Wallner S, Schuierer M, Behrmann I, Bataille F, et al. Promoter-hypermethylation is causing functional relevant downregulation of methylthioadenosine phosphorylase (MTAP) expression in hepatocellular carcinoma. Carcinogenesis 2006;27:64–72.10.1093/carcin/bgi201Search in Google Scholar PubMed
106. Moreno B, Lopez I, Fernandez-Dıez B, Gottlieb M, Matute C, Sánchez-Gómez MV, et al. Differential neuroprotective effects of 5′-deoxy-5′-methylthioadenosine. PloS ONE 2014;9:e90671. 1–9.10.1371/journal.pone.0090671Search in Google Scholar PubMed PubMed Central
107. Chang YC, Su CY, Hsiao M. Therapeutic targeting of methylthioadenosine phosphorylase. Cancer Cell Microenvironment 2016;3:e1322. 1–7.Search in Google Scholar
108. van Dam L, Korolev N, Nordenskiöld L. Polyamine-nucleic acid interactions and the effects on structure in oriented DNA fibers. Nucl Acids Res 2002;30:419–28.10.1093/nar/30.2.419Search in Google Scholar PubMed PubMed Central
109. Xiao L, Swank RA, Matthews HR. Photoaffinity polyamines: sequence-specific interactions with DNA. Nucleic Acids Res 1991;19:3701–8.10.1093/nar/19.13.3701Search in Google Scholar PubMed PubMed Central
110. Kabir A, Kumar SG. Binding of the biogenic polyamines to deoxyribonucleic acids of varying base composition: base specificity and the associated energetics of the interaction. PLoS One 2013;8:e70510.10.1371/journal.pone.0070510Search in Google Scholar PubMed PubMed Central
111. Igarashi K, Kashiwagi K. Modulation of cellular function by polyamines. Int J Biochem Cell Biol 2010;42:39–51.10.1016/j.biocel.2009.07.009Search in Google Scholar PubMed
112. Yoshida M, Kashiwagi K, Shigemasa A, Taniguchi S, Yamamoto K, Makinoshima H, et al. A unifying model for the role of polyamines in bacterial cell growth, the polyamine modulon. J Biol Chem 2004;279:46008–13.10.1074/jbc.M404393200Search in Google Scholar PubMed
113. Landau G, Bercovich Z, Park MH, Kahana C. The role of polyamines in supporting growth of mammalian cells is mediated through their requirement for translation initiation and elongation. J Biol Chem 2010;285:12474–8.10.1074/jbc.M110.106419Search in Google Scholar PubMed
114. Park MH, Lee YB, Joe YA. Hypusine is essential for eukaryotic cell proliferation. Biol Signals 1997;6:115–23.10.1159/000109117Search in Google Scholar PubMed
115. Park MH. The post-translational synthesis of a polyamine-derived amino acid, hypusine, in the eukaryotic translation initiation factor 5A (eIF5A). J Biochem (Tokyo) 2006;139:161–9.10.1093/jb/mvj034Search in Google Scholar
116. Park MH, Nishimura K, Zanelli CF, Valentini SR. Functional significance of eIF5A and its hypusine modification in eukaryotes. Amino Acids 2010;41:2538–45.10.1007/s00726-009-0408-7Search in Google Scholar
117. Algranati ID, Goldemberg SH. Polyamines and their role in protein synthesis. Trends Biochem Sci 1977;2:272–4.10.1016/0968-0004(77)90278-XSearch in Google Scholar
118. Childs AC, Mehta DJ, Gerner EW. Polyamine-dependent gene expression. Cell Mol Life Sci 2003;60:1394–406.10.1007/s00018-003-2332-4Search in Google Scholar PubMed
119. Pollard KJ, Samuels ML, Crowley KA, Hansen JC, Peterson CL. Functional interaction between GCN5 and polyamines: a new role for core histone acetylation. EMBO J 1999;18:5622–33.10.1093/emboj/18.20.5622Search in Google Scholar PubMed PubMed Central
120. García-Faroldi G, Rodríguez CE, Urdiales JL, Pérez-Pomares JM, Dávila JC, Pejler G, et al. Polyamines are present in mast cell secretory granules and are important for granule homeostasis. PLoS One 2010;5:e15071.10.1371/journal.pone.0015071Search in Google Scholar PubMed PubMed Central
121. Minois N. Molecular basis of the ‘anti-aging’ effect of spermidine and other natural polyamines – a mini-review. Gerontology 2014;60:319–32.10.1159/000356748Search in Google Scholar PubMed
122. Minois N, Carmona-Gutierrez D, Madeo F. Polyamines in aging and disease. Aging 2011;3:1–17.10.18632/aging.100361Search in Google Scholar
123. Scalabrino G, Ferioli ME. Polyamines in mammalian ageing: an oncological problem, too? A review. Mech Ageing Dev 1984;26:149–64.10.1016/0047-6374(84)90090-3Search in Google Scholar
124. Seiler N, Atanassov CL. The natural polyamines and immune system. Prog Drug Res 1994;43:87–126.10.1007/978-3-0348-7156-3_4Search in Google Scholar PubMed
125. Lagishetty CV, Naik SR. Polyamines: potential anti-inflammatory agents and their possible mechanism of action. Indian J Pharmacol 2008;40:121–5.10.4103/0253-7613.42305Search in Google Scholar PubMed PubMed Central
126. Zhang M, Borovikova LV, Wang H, Metz C, Tracey KJ. Spermine inhibition of monocyte activation and inflammation. Mol Med 1999;5:595–605.10.1007/BF03402072Search in Google Scholar PubMed
127. Kano Y, Soda K, Nakamura T, Saitoh M, Kawakami M, Konishi F. Increased blood spermine levels decrease the cytotoxic activity of lymphokine-activated killer cells: a novel mechanism of cancer evasion. Cancer Immunol Immunother 2007;56:771–81.10.1007/s00262-006-0229-4Search in Google Scholar PubMed
128. Bjelaković G, Stojanović I, Jevtović Stoimenov T, Pavlović D, Kocić G, Rossi S, et al. Metabolic correlations of glucocorticoids and polyamines in inflammation and apoptosis. Amino Acids 2010;39:29–43.10.1007/s00726-010-0489-3Search in Google Scholar PubMed
129. Chamaillard L, Quemener V, Havouis R, Moulinoux JP. Polyamine deprivation stimulates natural killer cell activity in cancerous mice. Anticancer Res 1993;13:1027–33.Search in Google Scholar PubMed
130. Babbar N, Murray-Stewart T, Casero RA. Inflammation and polyamine catabolism: the good, the bad and the ugly. Biochem Soc Trans 2007;35:301–4.10.1042/BST0350300Search in Google Scholar PubMed
131. Hayes CS, Burns MR, Gilmour SK. Polyamine blockade promotes antitumor Immunity. OncoImmunology 2014:3,e27360.10.4161/onci.27360Search in Google Scholar PubMed PubMed Central
132. Soda K, Kano Y, Nakamura T, Kawakami M, Konishi F. Spermine and spermidine induce some of the imunesuppresion observed in cancer patients. Ann Cancer Res Ther 2003;11:243–53.10.4993/acrt1992.11.243Search in Google Scholar
133. Milano G, Viguier E, Cassuto JP, Schneider M, Namer M, Boublil JL, et al. Polyamines and malignant diseases. Pathol Biol (Paris) 1980;28:328–34.Search in Google Scholar PubMed
134. Soda K. The mechanisms by which polyamines accelerate tumor spread. J Exp Clin Cancer Res 2011;30:95:1–9.10.1186/1756-9966-30-95Search in Google Scholar
135. Russell HD. The roles of the polyamines, putrescine, spermidine and spermine in normal and malignant tissues. Life Sci 1973;13:1635–47.10.1016/0024-3205(73)90111-2Search in Google Scholar PubMed
136. Russell HD. Clinical relevance of polyamines as biochemical markers of tumor kinetics. Clin Chem 1977;23:22–7.10.1093/clinchem/23.1.22Search in Google Scholar PubMed
137. Russell DH. Clinical relevance of polyamines. Crit Rev Clin Lab Sci 1983;18:261–311.10.3109/10408368209085073Search in Google Scholar PubMed
138. Russell DH. Ornithine decarboxylase: a key regulatory enzyme in normal and neoplastic growth. Drug Metab Rev 1985;16:1–88.10.3109/03602538508991430Search in Google Scholar PubMed
139. Tabib A, Bachrach U. Role of polyamines in mediating malignant transformation and oncogene expression. Inter J Biochem Cell Biol 1999;31:1289–95.10.1016/S1357-2725(99)00098-9Search in Google Scholar
140. Thomas T, Thomas TJ. Polyamine metabolism and cancer. J Cell Mol Med 2003;7:113–26.10.1111/j.1582-4934.2003.tb00210.xSearch in Google Scholar PubMed PubMed Central
141. Bachrach U. Polyamines and carcinogenesis. Acta Facultatis Medicae Naissensis 2012;29:165–174.10.2478/v10283-012-0023-5Search in Google Scholar
142. Moulinoux JP, Quemener V, Khan NA. Biological significance of circulating polyamines in oncology. Cell Mol Biol 1991;37:773–83.Search in Google Scholar PubMed
143. Johnson RM. Polyamines, their biochemistry and role in neoplasia. Proc West Pharmacol Soc 2005;48:21–3.Search in Google Scholar PubMed
144. Casero RA, Marton LJ. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov 2007;6:373–90.10.1038/nrd2243Search in Google Scholar PubMed
145. Luckel F, Kubo K, Tsumoto K, Yoshikawa K. Enhancement and inhibition of DNA transcriptional activity by spermine: a marked difference between linear and circular templates. FEBS Letters 2005;579:5119–22.10.1016/j.febslet.2005.07.095Search in Google Scholar PubMed
146. Mounce BC, Cesaro T, Moratorio G, Hooikaas PJ, Yakovleva A, Werneke SW, et al. Inhibition of polyamine biosynthesis is a broad-spectrum strategy against RNA Viruses. J Virol 2016;90:9683.10.1128/JVI.01347-16Search in Google Scholar PubMed
147. Tsujinaka S, Soda K, Kano Y, Konishi F. Spermine accelerates hypoxia-initiated cancer cell migration. Int J Oncol 2011;38:305–12.10.3892/ijo.2010.849Search in Google Scholar PubMed
148. Gugliucci A. Polyamines as clinical laboratory tools. Clin Chim Acta 2004;344:23–35.10.1016/j.cccn.2004.02.022Search in Google Scholar PubMed
149. Ernestus RI, Röhn G, Schröder R, Els T, Klekner Á, Paschen W, et al. Polyamine metabolism in brain tumours: diagnostic relevance of quantitative biochemistry. J Neurol Neurosurg Psychiatry 2001;71:88–92.10.1136/jnnp.71.1.88Search in Google Scholar PubMed
150. Manni A, Badger B, Grove R, Kunselman S, Demers L. Isolation and characterization of human breast cancer cells overexpressing S-adenosylmethionine decarboxylase. Cancer Letters 1995;95:23–2.10.1016/0304-3835(95)03860-YSearch in Google Scholar PubMed
151. Milovic V, Turchanowa L. Polyamines and colon cancer. Biochem Soc Trans 2003;31:381–3.10.1042/bst0310381Search in Google Scholar PubMed
152. Cañizares F, Salinas J, de las Heras M, Diaz J, Tovar I, Martinez P, et al. Prognostic value of ornithine decarboxylase and polyamines in human breast cancer: correlation with clinicopathologic parameters. Clin Cancer Res August 1999;5:2035–41.Search in Google Scholar
153. Luk GD, Baylin SB. Ornithine decarboxylase as a biologic marker in familial colonic polyposis. N Engl J Med 1984; 311:80–3.10.1056/NEJM198407123110202Search in Google Scholar PubMed
154. Patchett SE, Katelaris PH, Zhang ZW, Alstead EM, Domizio P, Farthing MJ. Ornithine decarboxylase activity is a marker of premalignancy in long standing Helicobacter pylori infection. Gut 1996;39:807–10.10.1136/gut.39.6.807Search in Google Scholar
155. Medina MA, Rodrıguez Quesada A, Nunez de Castro I, Sanchez-Jimenez F. Histamine, polyamines, and cancer. Biochem Pharmacol 1999;57:1341–4.10.1016/S0006-2952(99)00005-2Search in Google Scholar PubMed
156. Auvinen M. Cel transformation, invasion, and angiogenesis: a regulatory role for ornithine decarboxylase and polyamines? J Natl Cancer Inst 1997;89:533–7.10.1093/jnci/89.8.533Search in Google Scholar PubMed
157. Bachrach U, Wang YC, Tabib A. Polyamines: new cues in cellular signal transduction. News Physiol Sci 2001;16:106–9.10.1152/physiologyonline.2001.16.3.106Search in Google Scholar PubMed
158. Scuoppo C, Cornelius M, Lindqvist L, Reyes J, Ruse C, Appelmann I. A tumour suppressor network relying on the polyamine-hypusine axis. Nature 2012;487:244–50.10.1038/nature11126Search in Google Scholar PubMed PubMed Central
159. Bandopadhyay M, Ganguly AK. Putrescine, DNA, RNA and protein contents in human uterine, breast and rectal cancer. J Postgrad Med 2000;46:172–5.Search in Google Scholar PubMed
160. Höckel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 2001;93:266–76.10.1093/jnci/93.4.266Search in Google Scholar PubMed
161. Soda K, Kano Y, Chiba F, Koizumi K, Miyaki Y. Increased polyamine intake inhibits age-associated alteration in global DNA methylation and 1,2-dimethylhydrazine-induced tumorigenesis. PLoS One 2013;8:e64357.10.1371/journal.pone.0064357Search in Google Scholar PubMed PubMed Central
162. Svensson K. Mechanistic studies on the role of polyamines and microvesicles in tumor growth and hypoxia-mediated angiogenesis. Doctoral Dissertation 2012 Lund University, Faculty of Medicine, Series 2012;10:1–94.Search in Google Scholar
163. Büyükuslu N. Dietary polyamines and diseases: reducing polyamine intake can be beneficial in cancer treatment. J Nutrients 2015;2:27–38.10.18488/journal.87/2015.2.2/87.2.27.38Search in Google Scholar
164. Milovic V. Polyamines in the gut lumen: bioavailability and biodistribution. Eur J Gastroenterol Hepatol 2001;13:1021–5.10.1097/00042737-200109000-00004Search in Google Scholar PubMed
165. Vargas AJ, Wertheim BC, Gerner EW, Thomson CA, Rock CL, Thompson PA. Dietary polyamine intake and risk of colorectal adenomatous polyps. Am J Clin Nutr 2012;96:133–41.10.3945/ajcn.111.030353Search in Google Scholar PubMed PubMed Central
166. Kalac P, Krausov P. A review of dietary polyamines: formation, implications for growth and health and occurrence in foods. Food Chem 2005;90:219–30.10.1016/j.foodchem.2004.03.044Search in Google Scholar
167. Linus Pouling Institute, Oregon State University. Micronutrient information center. Folate. Copyright 2016.Search in Google Scholar
168. King MW, PhD. Vitamins and Minerals.org, LLC | info@ themedicalbiochemistrypage.org, Last updated 2016.Search in Google Scholar
169. McNulty H, Scott JM. Intake and status of folate and related B-vitamins: considerations and challenges in achieving optimal status. Bri J Nutr 2008;99:S48–S54.10.1017/S0007114508006855Search in Google Scholar PubMed
170. Deacon R, Bottiglieri T, Chanarin I, Lumb M, Perry J. Methylthioadenosine serves as a single carbon source to the folate co-enzyme pool in rat bone marrow cells. Biochim Biophys Acta 1990;1034:342–6.10.1016/0304-4165(90)90062-2Search in Google Scholar PubMed
171. Lavoie A, Cooper BA. Rapid Transfer of folic acid from blood to bile in man, and its conversion into folate coenzymes and into a pteroylglutamate with little biological activity. Clin Sci Mol Med 1974;46:729–41.10.1042/cs0460729Search in Google Scholar PubMed
172. Liew SC. Folic acid and diseases – supplement it or not? Rev Assoc Med Bras 2016;62:90–100.10.1590/1806-9282.62.01.90Search in Google Scholar
173. Heby O. DNA methylation and polyamines in embryonic development and cancer. Int J Dev Biol 1995;39:737–57.Search in Google Scholar PubMed
174. Cedar H. DNA methylation and gene activity. Cell 1988;53:3–4.10.1016/0092-8674(88)90479-5Search in Google Scholar PubMed
175. van der Linden IJ, Afman LA, Heil SG, Blom HJ. Genetic variation in genes of folate metabolism and neural-tube defect risk. Proc Nutr Soc 2006;65:204–15.10.1079/PNS2006495Search in Google Scholar PubMed
176. Blom HJ, Shaw GM, den Heijer M, Richard H, Finnell M. Neural tube defects and folate: case far from closed. Nat Rev Neurosci 2006;7:724–31.10.1038/nrn1986Search in Google Scholar PubMed PubMed Central
177. Bailey LB, Gregory JF. Folate metabolism and requirements. J Nutr 1999;129:779–82.10.1093/jn/129.4.779Search in Google Scholar PubMed
178. Lucock M. Is folic acid the ultimate functional food component for disease prevention? Br Med J 2004;328:211–4.10.1136/bmj.328.7433.211Search in Google Scholar PubMed PubMed Central
179. Zhao R, Diop-Bove N, Visentin M, Goldman ID. Mechanisms of membrane transport of folates into cells and across epithelia. Annu Rev Nutr 2011;21:177–201.10.1146/annurev-nutr-072610-145133Search in Google Scholar PubMed PubMed Central
180. Sirotnak FM, Tolner B. Carrier-mediated membrane transport of folates in mammalian cells. Annu Rev Nutr 1999;19:91–122.10.1146/annurev.nutr.19.1.91Search in Google Scholar PubMed
181. Zhao R, Matherly LH, Goldman ID. Membrane transporters and folate homeostasis: intestinal absorption and transport into systemic compartments and tissues. Expert Rev Mol Med 2009;11:e4.10.1017/S1462399409000969Search in Google Scholar PubMed
182. Milman N. Intestinal absorption of folic acid - new physiologic and molecular aspects. Indian J Med Res 2012;136:725–8.Search in Google Scholar PubMed
183. Matherly LH, Goldman DI. Membrane transport of folates. Vitam Horm 2003;66:403–56.10.1016/S0083-6729(03)01012-4Search in Google Scholar PubMed
184. Steinberg ES. Mechanisms of folate homeostasis. Am J Physiol 1984;246:G319–24.10.1152/ajpgi.1984.246.4.G319Search in Google Scholar PubMed
185. Troen AM, Lutgens E, Smith DE, Rosenberg IH, Selhub J. The atherogenic effect of excess methionine intake. Proc Natl Acad Sci USA 2003;100:15089–94.10.1073/pnas.2436385100Search in Google Scholar PubMed PubMed Central
186. Chiang PK, Gordon RK, Tal J, Zeng GC, Doctor BP, Parthasaradhi GC, et al. S-adenosylmethionine and methylation. FASEB J 1996;10:471–80.10.1096/fasebj.10.4.8647346Search in Google Scholar PubMed
187. Bottiglieri T. S-adenosyl-L-methionine (SAMe): from the bench to the bedside – molecular basis of a pleiotrophic molecule. Am J Clin Nutr 2002;76:1151S–7S.10.1093/ajcn/76.5.1151SSearch in Google Scholar
188. Lieber CS, Packer L. S-adenosylmethionine: molecular, biological, and clinical aspects – an introduction. Am J Clin Nutr 2002;76:1148S–50S.10.1093/ajcn/76.5.1148SSearch in Google Scholar
189. Krebs AH, Hems R, Tyler B. The regulation of folate and methionine metabolism. Biochem J 1976;158:341–53.10.1042/bj1580341Search in Google Scholar PubMed PubMed Central
190. Choi SW, Mason JB. Folate and carcinogenesis: an integrated scheme. J Nutr 2000;130:129–32.10.1093/jn/130.2.129Search in Google Scholar PubMed
191. Crider KS, Yang TP, Berry RJ, Bailey LB. Folate and DNA methylation: a review of molecular mechanisms and the evidence for folate’s role. Adv Nutr 2012;3:21–38.10.3945/an.111.000992Search in Google Scholar PubMed PubMed Central
192. Sun D, Wollin A, Stephen MA. Moderate folate deficiency influences polyamine synthesis in rats. J Nutr 2002;132:2632–7.10.1093/jn/132.9.2632Search in Google Scholar PubMed
193. Bjelakovic G, Kocic G, Pavlovic D, Nikolic J, Stojanovic I, Bjelakovic GB, et al. Effects of folic acid on polyamine concentrations and polyamine oxidase activity in regenerating rat liver. Pteridines 2003;14:109–13.Search in Google Scholar
194. Bjelakovic G, Pavlovic D, Jevtovic T, Stojanovic I, Sokolovic D, Bjelakovic GB, et al. Vitamin B12 and folic acid effects on polyamine metabolism in rat liver. Pteridines 2006;17:90–4.10.1515/pteridines.2006.17.3.90Search in Google Scholar
195. Bistulfi G, Diegelman, P, Foster BA, Kramer DL, Porter CW, Smiraglia DJ. Polyamine biosynthesis impacts cellular folate requirements necessaryto maintain S-adenosylmethionine and nucleotide pools. FASEB J 2009;23:2888–97.10.1096/fj.09-130708Search in Google Scholar PubMed PubMed Central
196. Bistulfi G, VanDette E, Matsui SI, Smiraglia DJ. Mild folate deficiency induces genetic and epigenetic instability and phenotype changes in prostate cancer cells. BMC Biology 2010;8:1–12.10.1186/1741-7007-8-6Search in Google Scholar PubMed PubMed Central
197. Meenhan R, Lewis J, Cross S, Nan X, Jeppesen P, Bird A. Transcriptional repression by méthylation of CpG. J Cell Sci 1992;16:9–14.10.1242/jcs.1992.Supplement_16.2Search in Google Scholar
198. Razin A. CpG methylation, chromatin structure and gene silencing-a three-way connection. EMBO J 1998;17:4905–8.10.1093/emboj/17.17.4905Search in Google Scholar PubMed PubMed Central
199. Jacob RA. Folate, DNA methylation, and gene expression: factors of nature and nurture. Am J Clin Nutr 2000;72:903–4.10.1093/ajcn/72.4.903Search in Google Scholar PubMed
200. Duranton B, Keith G, Gossé F, Bergmann C, Schleiffer R, Raul F. Concomitant changes in polyamine pools and DNA methylation during growth in inhibition of human colonic cancer cells. Exp Cell Res 1998;243:319–25.10.1006/excr.1998.4148Search in Google Scholar PubMed
201. McGlynn AP, Wasson GR, Reilly SL, McNulty H, Downes CS, Chang C, et al. Low colonocyte folate is associated with uracil misincorporation and global DNA hypomethylation in human colorectum. J Nutr 2013;143:27–33.10.3945/jn.112.167148Search in Google Scholar PubMed
202. Blount BC, Mack MM, Wehr CM, MacGregor JT, Hiatt RA, Wang G, et al. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci USA 1997;94:3290–5.10.1073/pnas.94.7.3290Search in Google Scholar PubMed PubMed Central
203. Kim YI. Folate and DNA methylation: A mechanistic link between folate deficiency and colorectal cancer? Cancer Epidemiol Biomarkers Prev 2004;13:511–9.10.1158/1055-9965.511.13.4Search in Google Scholar PubMed
204. McCabe MT, Brandes JC, Vertino PM. Cancer DNA methylation: molecular mechanisms and clinical implications. Clin Cancer Res 2009;15:3927–37.10.1158/1078-0432.CCR-08-2784Search in Google Scholar PubMed
205. Baylin SB. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol 2005;2:S4–11.10.1038/ncponc0354Search in Google Scholar PubMed
206. Akhavan-Niaki H, Samadani AA. DNA methylation and cancer development: molecular mechanism. Cell Bioch Biophys 2013;67:501–13.10.1007/s12013-013-9555-2Search in Google Scholar
207. Kim JK, Samaranayake M, Pradhan S. Epigenetic mechanisms in mammals. Cell Mol Life Sci 2009;66:596–612.10.1007/s00018-008-8432-4Search in Google Scholar PubMed
208. Zhang G, Pradhan S. Critical review mammalian epigenetic Mechanisms. IUBMB Life 2014;66:240–56.10.1002/iub.1264Search in Google Scholar
209. Kautiainen TL, Jones PA. DNA methyltransferase levels in tumorigenic and nontumorigenic cells in culture. J Biol Chem 1986;261:1594–8.10.1016/S0021-9258(17)35981-1Search in Google Scholar PubMed
210. Foulks JM, Parnell KM, Nix RN, Chau S, Swierczek K, Saunders M, et al. Epigenetic drug discovery targeting DNA methyltransferases. J Biomol Screen 2012;17:2–17.10.1177/1087057111421212Search in Google Scholar PubMed
211. Robertson DK, Jones AP. DNA methylation: past, present and future directions. Cancerogenesis 2000;21:461–7.10.1093/carcin/21.3.461Search in Google Scholar PubMed
212. Lee JY, Lee T-H. Effects of histone acetylation and CpG methylation on the structure of nucleosomes. Biochim Biophys Acta 2012;1824:974–82.10.1016/j.bbapap.2012.05.006Search in Google Scholar PubMed PubMed Central
213. Li Y, Tollefsbol TO. Impact on DNA methylation in cancer prevention and therapy by bioactive dietary components. Curr Med Chem 2010;17:2141–51.10.2174/092986710791299966Search in Google Scholar PubMed PubMed Central
214. Collings CK, Waddell PJ, Anderson JN. Effects of DNA methylation on nucleosome stability. Nucleic Acids Res 2013;41:2918–31.10.1093/nar/gks893Search in Google Scholar PubMed PubMed Central
215. An W. Histone acetylation and methylation: combinatorial players for transcriptional regulation. Subcell Biochem 2007;41:351–69.Search in Google Scholar PubMed
216. Fang M, Chen D, Yang CS. Dietary polyphenols may affect DNA methylation. J Nutr 2007;137:223S–8S.10.1093/jn/137.1.223SSearch in Google Scholar PubMed
217. Michael D, Tollefsbol TO. Epigenetic linkage of aging, cancer and nutrition. J Exp Biol 2015;218:59–70.10.1242/jeb.107110Search in Google Scholar PubMed PubMed Central
218. Trewick SC, McLaughlin PJ, Allshire RC. Methylation: lost in hydroxylation? EMBO reports 2005;6:315–20.10.1038/sj.embor.7400379Search in Google Scholar PubMed PubMed Central
219. Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007;128:683–92.10.1016/j.cell.2007.01.029Search in Google Scholar PubMed PubMed Central
220. Struhl K. Histone acetylation and transcriptional regulatory mechanisms. Genes Dev 1988;12:599–606.10.1101/gad.12.5.599Search in Google Scholar PubMed
221. Wani NA, Hamid A, Kaur J. Folate status in various pathophysiological conditions. IUBMB Life 2008;6:834–42.10.1002/iub.133Search in Google Scholar PubMed
222. Giovannucci E. Epidemiologic studies of folate and colorectal neoplasia: a review. J Nutr 2002;132:2350S–5S.10.1093/jn/132.8.2350SSearch in Google Scholar PubMed
223. Eloranta TO, Raina AM. S-adenosylmethionine. Metabolism and its relation to polyamine synthesis in rat liver. Biochem J 1977;168:179–85.10.1042/bj1680179Search in Google Scholar PubMed PubMed Central
224. Gibbons RJ. Histone modifying and chromatin remodelling enzymes in cancer and dysplastic syndromes. Hum Mol Genet 2005;14:R85–R92.10.1093/hmg/ddi106Search in Google Scholar PubMed
225. Williams EA. Folate, colorectal cancer and the involvement of DNA methylation. Proc Nutr Soc 2012;71:592–7.10.1017/S0029665112000717Search in Google Scholar PubMed
226. Sharma S, Theresa KK, Jones PA. Epigenetics in cancer. Carcinogenesis 2010;31:27–36.10.1093/carcin/bgp220Search in Google Scholar PubMed
227. Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc Natl Acad Sci 1964;51:786–94.10.1073/pnas.51.5.786Search in Google Scholar
228. Hake SB, Xiao A, Allis CD. Linking the epigenetic ‘language’ of covalent histone modifications to cancer. Br J Cancer 2004;90:761–9.10.1038/sj.bjc.6601575Search in Google Scholar PubMed
229. Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med 2003;349:2042–54.10.1056/NEJMra023075Search in Google Scholar PubMed
230. Baylin SB, Ohm JE. Epigenetic gene silencing in cancer – a mechanism for early oncogenic pathway addiction? Nature Rev Cancer 2006;6:107–15.10.1038/nrc1799Search in Google Scholar
231. Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene 2002;21:5400–13.10.1038/sj.onc.1205651Search in Google Scholar PubMed
232. Kouzmenko A, Ohtake F, Fujiki R, Kato S. Hormonal gene regulation through DNA methylation and demethylation. Epigenomics 2010;2:765–74.10.2217/epi.10.58Search in Google Scholar PubMed
233. Zhang K, Dent SY. Histone modifying enzymes and cancer: going beyond histones. J Cell Biochem 2005;96:1137–48.10.1002/jcb.20615Search in Google Scholar PubMed
234. Rice JC, Allis CD. Histone methylation versus histone acetylation: new insights into epigenetic regulation. Curr Opin Cell Biol 2001;13:263–73.10.1016/S0955-0674(00)00208-8Search in Google Scholar PubMed
235. Raikov Z, Raikova E, Ivanov V. Mutagenesis, cancerogenesis, and prevention of cancer. TJS 2005;3:55–9.Search in Google Scholar
236. Mamede AC, Tavares SD, Abrantes AM, Trindade J, Maia JM, Botelho MF. The role of vitamins in cancer: a review. Nutr Cancer 2011;63:479–94.10.1080/01635581.2011.539315Search in Google Scholar PubMed
237. Wien TN, Pike E, Wisløff T, Staff A, Smeland S, Klemp M. Cancer risk with folic acid supplements: a systematic review and meta-analysis. Bm jopen.bmj.com/, BMJ Open 2012;2: 1–14:e000653.10.1136/bmjopen-2011-000653Search in Google Scholar PubMed PubMed Central
238. Pogribny IP, Tryndyak VP, Muskhelishvili L, Rusyn I, Ross SA. Methyl deficiency, alterations in global histone modifications and carcinogenesis. J Nutr 2007;137:216S–22S.10.1093/jn/137.1.216SSearch in Google Scholar PubMed
239. Wright AJ, Dainty JR, Finglas PM. Folic acid metabolism in human subjects revisited: potential implications for proposed mandatory folic acid fortification. Br J Nutr 2007;98:667–75.10.1017/S0007114507777140Search in Google Scholar PubMed
240. Ulrich CM, Potter JD. Folate and cancer – timing is everything. J Am Med Assoc 2007;297:2408–9.10.1001/jama.297.21.2408Search in Google Scholar PubMed
241. Kim YI. Folic acid fortification and supplementation – good for some but not so good for others. Nutr Rev 2007;65:504–11.10.1301/nr.2007.nov.504-511Search in Google Scholar PubMed
242. Duthie SJ. Folate and cancer: how DNA damage, repair and methylation impact on colon carcinogenesis. J Inherit Metab Dis 2011;34:101–9.10.1007/s10545-010-9128-0Search in Google Scholar PubMed
243. Choi SW. Friso S. Epigenetics: a new bridge between nutrition and health. Adv Nutr 2010;1:8–16.10.3945/an.110.1004Search in Google Scholar PubMed PubMed Central
244. Kalmbach RD, Choumenkovitch SF, Troen AM, D’Agostino R, Jacques PF, Selhub J. Circulating folic acid in plasma: relation to folic acid fortification. Am J Clin Nutr 2008;88:763–8.10.1093/ajcn/88.3.763Search in Google Scholar PubMed PubMed Central
245. Sauer J, Mason JB, Choi SW. Too much folate – a risk factor for cancer and cardiovascular disease? Curr Opin Clin Nutr Metab Care 2009;12:30–6.10.1097/MCO.0b013e32831cec62Search in Google Scholar PubMed PubMed Central
246. Strickland KC, Krupenko NI, Krupenko SA. Molecular mechanisms underlying the potentially adverse effects of folate. Clin Chem Lab Med 2013;51:607–16.10.1515/cclm-2012-0561Search in Google Scholar PubMed PubMed Central
247. Smith AD, Kim YI, Refsum H. Is folic acid good for everyone? Am J Clin Nutr 2008;87:517–33.10.1093/ajcn/87.3.517Search in Google Scholar PubMed
248. Bjelakovic G, Stojanovic I, Jevtovic-Stoimenov T, Bjelakovic L, Kamenov B, Pavlovic D, et al. Is folic acid supplementation to food benefit or risk for human health? Pteridines 2013;24:165–81.10.1515/pterid-2013-0024Search in Google Scholar
249. Kim YI. Folate: a magic bullet or a double edged sword for colorectal cancer prevention? Gut 2006;55:1387–9.10.1136/gut.2006.095463Search in Google Scholar PubMed PubMed Central
250. Jänne J, Alhonen L, Keinänen TA, Pietilä M, Uimari A, Pirinen E, et al. Animal disease models generated by genetic engineering of polyamine metabolism. J Cell Mol Med 2005;9:865–82.10.1111/j.1582-4934.2005.tb00385.xSearch in Google Scholar PubMed PubMed Central
©2017 Walter de Gruyter GmbH, Berlin/Boston
This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Articles in the same Issue
- Frontmatter
- Reviews
- Photosensitization of peptides and proteins by pterin derivatives
- Polyamines, folic acid supplementation and cancerogenesis
- Mini review
- Medical significance of simultaneous application of red blood cell distribution width (RDW) and neopterin as diagnostic/prognostic biomarkers in clinical practice
- Original articles
- Molecular architecture of pterin deaminase from Saccharomyces cerevisiae NCIM 3458
- Quantitative analysis by flow cytometry of green fluorescent protein-tagged human phenylalanine hydroxylase expressed in Dictyostelium
- Age-dependance of pteridines in the malaria vector, Anopheles stephensi
- Seasonality of blood neopterin levels in the Old Order Amish
- Correlation of salivary neopterin and plasma fibrinogen levels in patients with chronic periodontitis and/or type 2 diabetes mellitus
- Positive association between Toxoplasma gondii IgG serointensity and current dysphoria/hopelessness scores in the Old Order Amish: a preliminary study
- Sleep onset insomnia, daytime sleepiness and sleep duration in relationship to Toxoplasma gondii IgG seropositivity and serointensity
- Concentrations of neopterin, kynurenine and tryptophan in wound secretions of patients with breast cancer and malignant melanoma: a pilot study
- Comparison of performance of composite biomarkers of inflammatory response in determining the prognosis of breast cancer patients
- Association of peripheral blood cell count-derived ratios, biomarkers of inflammatory response and tumor growth with outcome in previously treated metastatic colorectal carcinoma patients receiving cetuximab
- Neoadjuvant combination therapy with trastuzumab in a breast cancer patient with synchronous rectal carcinoma: a case report and biomarker study
Articles in the same Issue
- Frontmatter
- Reviews
- Photosensitization of peptides and proteins by pterin derivatives
- Polyamines, folic acid supplementation and cancerogenesis
- Mini review
- Medical significance of simultaneous application of red blood cell distribution width (RDW) and neopterin as diagnostic/prognostic biomarkers in clinical practice
- Original articles
- Molecular architecture of pterin deaminase from Saccharomyces cerevisiae NCIM 3458
- Quantitative analysis by flow cytometry of green fluorescent protein-tagged human phenylalanine hydroxylase expressed in Dictyostelium
- Age-dependance of pteridines in the malaria vector, Anopheles stephensi
- Seasonality of blood neopterin levels in the Old Order Amish
- Correlation of salivary neopterin and plasma fibrinogen levels in patients with chronic periodontitis and/or type 2 diabetes mellitus
- Positive association between Toxoplasma gondii IgG serointensity and current dysphoria/hopelessness scores in the Old Order Amish: a preliminary study
- Sleep onset insomnia, daytime sleepiness and sleep duration in relationship to Toxoplasma gondii IgG seropositivity and serointensity
- Concentrations of neopterin, kynurenine and tryptophan in wound secretions of patients with breast cancer and malignant melanoma: a pilot study
- Comparison of performance of composite biomarkers of inflammatory response in determining the prognosis of breast cancer patients
- Association of peripheral blood cell count-derived ratios, biomarkers of inflammatory response and tumor growth with outcome in previously treated metastatic colorectal carcinoma patients receiving cetuximab
- Neoadjuvant combination therapy with trastuzumab in a breast cancer patient with synchronous rectal carcinoma: a case report and biomarker study