Review article

J.-H. GAO1,2, L.-J. GUO2, Z.-Y. HUANG2, J.N. RAO3, C.-W. TANG1,2

ROLES OF CELLULAR POLYAMINES IN MUCOSAL HEALING IN THE GASTROINTESTINAL TRACT

1Division of Peptides Related with Human Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; 2Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China; 3Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
The mammalian gastrointestinal (GI) mucosa is a rapidly self-renewing tissue in the body, and its integrity is preserved through the strict regulation of epithelial cell proliferation, growth arrest, and apoptosis. Polyamines are shown to play an important role in the regulation of gastrointestinal mucosal growth and healing after injury under physiological and various pathological conditions. In this review, we highlight the importance of cellular polyamines in the control GI mucosal proliferation, migration, apoptosis, angiogenesis and GI barrier function during mucosal repair after injury.
Key words:
angiogenesis, apoptosis, barrier function, ornithine decarboxylase, polyamines, proliferation, stem cells, restitution

INTRODUCTION

The gastrointestinal (GI) tract is lined by continuous epithelial cells which maintain the physical and functional barrier to undesirable luminal antigens (1, 2). Epithelial cells of mammalian GI mucosa rapidly proliferate and turnover approximately every three days under biological conditions (2, 3). This process involves the balance among cell proliferation, differentiation, migration, and apoptosis. The GI mucosa is commonly injured by gastric acid, toxic luminal substances, mechanical strain, inflammation, interactions with microbes, oxidative stress and pharmaceuticals (4). After injury, the mucosa undergoes a rapid healing process, which involves multiple mechanisms, including early epithelial restitution, cell proliferation, apoptosis, angiogenesis, and mucosal remodeling (5, 6). The healing of damaged mucosa in the GI tract is highly regulated by multiple signal pathways and is critically controlled by numerous factors including cellular polyamines.

Natural polyamines (putrescine, spermidine and spermine) are low molecular weight and highly charged aliphatic polycations which intimately involved in many distinct cellular functions. For example, polyamines interact with polyanionic DNA molecules, thereby modulating chromatin structure, gene transcription, translation, and DNA stabilization (7). Several studies showed that polyamines are also required for signal transduction, receptor-ligand interactions, cell proliferation, migration, RNA stabilization and function of ion channels (8). An increasing body of evidence has advanced our understanding of the cellular and molecular functions of polyamines, therefore, this review will briefly highlight the roles and mechanisms of cellular polyamines in GI mucosal healing and also point out their potential clinical applications in patients with mucosal injury-associated disorders.

POLYAMINE METABOLISM IN THE GASTROINTESTINAL TRACT

Polyamine levels and subcellular distribution in the gastrointestinal tract

Polyamines are mainly located in the nucleus and cytoplasm (9). The concentration of polyamines is higher in rapidly proliferating tissues and increases rapidly when growth or proliferation is induced (10). In the GI tract, there is a polyamine gradient in the villi-crypt cell axis and along the digestive tube. The greatest luminal polyamines content is observed in the jejunum and colon (11).

Although the intracellular polyamines are in the millimolar range, free polyamines are considerably less abundant (only 7–10% of the total cellular polyamines pool), as polyamines are mostly ionically bound to different polyanionic in the cells (mainly DNA, RNA, proteins and phospholipids) (8). Only the free intracellular polyamines are available for immediate cellular needs. The major sources of exogenous polyamines come from diet and luminal bacteria in GI tract. A rat consumes daily about 160 µmoles/kg body weight (b.w.) polyamines of dietary origin, 30–40 µmoles/kg b.w. come from gut bacteria and 140 µmoles/kg b.w. from endogenous biosynthesis (12). The average daily polyamines intake of adolescents was 316±170 mmol/day (13). The concentration of intracellular polyamines is actively maintained and regulated by dynamic balance among exogenous transport, endogenous biosynthesis, interconversion, and degradation (14).

Transport of polyamines in the gastrointestinal tract

Transiently after meal, the concentration of polyamines in the duodenal and jejunal lumen reaches its maximum level. However, as early as 2 hours after the meal, the concentration of polyamines in the luminal side gradually reduces and returns to the fasting level, which indicates that the polyamines are rapidly absorbed by epithelial cells (15). Five models are currently applied to explain mammalian polyamines transport from lumen of the intestine. Firstly, transepithelial transport of polyamines occurs by passive diffusion (16). Secondly, mammalian polyamine transport proceeds in two tightly connected steps. Polyamines are first transported into the cell by a membrane transporter/carrier which is powered by electronegative membrane potential, then polyamines immediately accumulate into polyamines-sequestering vesicles which involve proton exchange and are driven by a vacuolar-ATPase pH gradient (17). Thirdly, polyamine uptake is a process involving the cell surface-associated heparan sulfate proteoglycans (HSPGs) and glypican 1 (Gpc1) (18, 19). Fourthly, absorption of polyamines is mediated by caveolin-1 dependent endocytosis and NOS2-dependent mechanism (20, 21). In addition, putrescine and acetylated polyamines are exported by an amino acid transporter SLC3A2 via a diamine/arginine exchange activity (21, 22), and other membrane transporters, such as SLC22A1, SLC22A2, SLC22A3, SLC47A1, SLC7A1, SLC12A8A, and SLC22A16, might also be involved in polyamine transport (23).

Biosynthesis and degradation of polyamines in the gastrointestinal tract

Polyamines are synthesized from methionine and ornithine. The first rate-limiting step in polyamine biosynthesis is the production of putrescine from ornithine. Ornithine is decarboxylated to putrescine by ornithine decarboxylase (ODC), which is a key enzyme of polyamine biosynthesis (Fig. 1). Putrescine is subsequently converted into spermidine through the catalysis of S-adenosyl-methionine decarboxylase (AdoMetDC) and spermidine synthase (SPDS). Spermidine is then converted into spermine by AdoMetDC and spermine synthase (SPMS) (Fig. 1). Methionine is the precursor for S-adenosyl-methionine (AdoMet) (8, 24). The decarboxylation product of AdoMet (DCAdoMet) is the precursor of the aminopropyl moieties of spermidine and spermine. Spermine, spermidine and putrescine are interconverted, depending on the physiological needs (14). Polyamines are degraded by spermine oxidases (SPMO), spermidine oxidases (SPDO) and spermine/spermidine-N-acetyltransferase (SSAT) (Fig. 1) (3, 25). In regard to regulating ODC, the ODC-antizyme binds ODC and facilitates proteasomal ODC degradation, thus inhibiting the endogenous biosynthesis of polyamines from ornithine (14, 26). The activity of ODC could also be specifically inhibited by the chemical compound DL-α-difluoromethylornithine (DFMO).

Figure 1 Fig. 1. Polyamine metabolism pathway.
Abbreviations: AdoMet, S-adenosyl-methionine; AdoMetDC, S-adenosyl-methionine decarboxylase; DCAdoMet, decarboxylation product of AdoMet; DFMO, DL-α-difluoromethylornithine; ODC, ornithine decarboxylase; SPDO, spermidine oxidases; SPDS, spermidine synthase; SPMO, spermine oxidases; SPMS, spermine synthase; SSAT, spermine/spermidine-N-acetyltransferase.

THE ROLE OF POLYAMINES IN MUCOSAL HEALING AFTER INJURY

Restitution

Minor injuries in GI epithelia are routinely caused by mechanical strain. After injury, rapid repair of the epithelial barrier is accomplished by a complex process termed epithelial restitution. Epithelial restitution refers to resealing of superficial wounds occurring as a consequence of epithelial cell migration into the defect rather than cell proliferation (4, 27). The process of epithelial restitution is that polarized epithelial cells adjacent to the defect, flatten, migrate, and ultimately repolarize over the exposed basement membrane (27). Restitution is stimulated by a wide range of highly divergent factors including prostaglandins, trefoil proteins, mucins, growth factors and polyamines. In contrast, the blockage of these factors significantly delays restitution (28-31).

Esophageal epithelia

Esophageal mucosa consists of keratinized stratified squamous epithelium. Mucosal injuries in the oesophagus could be induced by exposure to gastric acid. In vitro, the restitution and proliferation of esophageal epithelial cell are affected by several growth factors, by which hepatocyte growth factor (HGF), insulin-like growth factor 1 (IGF-1) and epidermal growth factor (EGF) stimulate the restitution, whereas, transforming growth factor beta (TGFb) inhibits restitution (32, 33). However, epithelial restitution may not involve in the healing of acid-induced injury, because restitution of esophageal epithelial cells are gradually inhibited at pH from 3.0 to 6.5 (34). Alternatively, cell proliferation is stimulated by acid in the normal esophageal tissues (35, 36). Thus, it could be estimated that cell proliferation but not restitution appears to be the major reparative defense for acid-induced esophageal epithelium injury.

Gastric epithelia

Restitution is an important process in the repair of gastric mucosal injury. Growth factors such as HGF and EGF seem to be involved in gastric restitution (37-39). EGF facilitated gastric restitution via basolateral EGF receptors by stimulation of basolateral Na+/H+ exchangers and phosphatidylinositol 3-kinases (PI3K) activation (37, 40). In addition, HGF-mediated epithelial restitution involves cyclooxygenase-2 and polyamines (41, 42). Polyamines facilitated the distribution, diffusion and assembly of microtubules and actin cytoskeleton in normal and damaged cells in vivo, which demonstrate that polyamines are important for gastric mucosal restitution (43-46). Additionally, the reduction of mucous secreting layer thickness is accompanied with suppression of gastric mucosal ODC activity in rats with gastric ulcers and spermidine supplementation reversed the delay in healing (47).

Intestinal epithelia

Restitution is a precisely regulated process involving epithelial cells to polarize and migrate to reseal wounded area. Polyamines play an important role in intestinal epithelia cell (IEC) restitution. Several studies manifested that migration of IEC was inhibited by DFMO and the decreased migration was reversed by exogenous addition of polyamines (48-50). However, the exact molecular mechanisms remain unclear. It has been demonstrated that K+ channel activity, cytoplasmic free Ca2+ ([Ca2+]cyt) and membrane potential (Em) were involved in polyamine-depended IEC migration (49-51). Polyamines could increase [Ca2+]cyt not only by enhancing K+ channel expression which cause membrane hyperpolarization and [Ca2+] influx, but also by canonical transient receptor potential 1 (TRPC1)-mediated [Ca2+] release from internal stores though altering the ratio of stromal interaction molecule 1 (STIM1) to STIM2 (Fig. 2) (51-55). The increased [Ca2+]cyt stimulates IEC migration by regulation of specific proteins during restitution, including up-regulation of phospholipase C-γ1 (PLC-γ1), small GTP binding proteins such as RhoA, Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division control protein 42 (Cdc42) (Fig. 2) (49, 50, 56, 57). Additionally, activation of RhoA by cellular polyamines results in increasing polymerization of myosin regulatory light chain (MRLC) which leading to the enhanced formation of myosin stress fiber and lamellipodia during cell migration after wounding (Fig. 2) (58-60).

Figure 2
Fig. 2. The Integrated polyamine signaling pathway related to restitution, proliferation, apoptosis, angiogenesis and GI barrier function.
Abbreviations: ATF2, activating transcription factor 2; AUF, AU-binding factor 1; Bax, Bcl-2-associated X protein; Bcl2, B cell lymphoma gene 2; [Ca2+]cyt, cytoplasmic free Ca2+; cdc42, cell division control protein 42; CDK4, cyclin-depen-dent kinase 4; Chk2, checkpoint kinase 2; CREB, cAMP response element-binding protein; CUGBP1, CUG-binding protein 1; HuR, human antigen R; H2O2, hydrogen peroxide; IκBα, inhibitor of NF-κB α; MCL1, myeloid cell leukemia sequence 1; MEK1, MAPK mitogen-activated protein kinase kinase-1; MLC, myosin light chain; MRLC, myosin regulatory light chain; NDRG1, N-Myc downregulated gene 1; NFκB, nuclear factor-κB; NPM, nucleophosmin; PDK1, 3-phosphoinositide-dependent protein kinase-1; PI3K, phosphatidylinositol 3-kinases; PLC-γ1, phospholipase C-γ1; Rac1, Ras-related C3 botulinum toxin substrate 1; RhoA, Ras homolog gene family, member A; Rock1, Rho-associated coiled-coil containing protein kinase 1; SGs, stress granules; SPMO, spermine oxidases; STIM, stromal interaction molecule; TGFβ, transforming growth factor beta; TGFβR, TGFβ receptor; TIA1, T-cell intracellular antigen 1; TIAR, TIA1-related protein; TRPC1, canonical transient receptor potential 1; XIAP, X chromosome-linked inhibitor of apoptosis protein; ZO-1, zona occludens-1.

PROLIFERATION

Esophageal epithelia

Epithelial cell proliferation is subsequent to restitution in order to restore normal mucosal architecture if the defect is large. Several studies indicated that polyamines and ODC levels were increased after esophageal epithelial injury (Barrett’s esophagus (BE), BE-associated dysplasia and carcinoma) (61). However, DFMO inhibits the growth of Barrett’s epithelium in vitro and in vivo probably by down-regulation of Kruppel like factor 5 (KLF5) and suppression of replication factor C subunit 5 (RFC5) (62).

Gastric epithelia

Several studies have shown that the cellular polyamines are necessary for cell proliferation in gastric epithelium. Polyamines are associated with increased 3H-thymidine incorporation in gastric ulcer model of rats (63). Reduction of polyamines by inhibiting ODC activity significantly suppressed gastric epithelial cell proliferation (64). Furthermore, lipopolysaccharides (LPS) stimulate gastric enterochromaffin-like cell (ECL) DNA synthesis by activation of the intracellular polyamine pathway and ODC activity via a cluster of differentiation 14 (CD14) receptor on the ECL cells (65). Additionally, polyamines and ODC regulate cell proliferation by altering cytoskeletal reorganization via RhoA (66). In the restraint-stress-induced gastric lesions, pretreatment with DFMO resulted into a remarkable decrease in mucosal DNA synthesis and thereby significantly delayed the mucosal healing. In contrast, treatment with polyamines remarkably accelerated ulcer healing (67, 68).

Intestinal epithelia

IEC proliferation is stimulated by polyamines and repressed by polyamine depletion in vivo and in vitro, which manifested that polyamines are absolutely essential for IEC growth and division (69, 70). Although the precise molecular mechanisms have not yet fully elucidated, increasing evidence reveals that polyamines enhances IEC proliferation by regulating large number of growth related genes. Recent studies revealed that polyamines up-regulate the transcription of growth-promoting genes such as c-Fos, c-Jun, and c-Myc (71, 72) and down-regulate growth-inhibiting genes such as p53, N-Myc downregulated gene 1 (NDRG1), nucleophosmin (NPM), JunD, and TGFβ/TGFβ receptor at the posttranscriptional level (Fig. 2) (69, 73-78). Additionally, polyamines promote translation of cyclin-dependent kinase 4 (CDK4) through CUG-binding protein 1 (CUGBP1), JunD and microRNA-222, thereby induction of IEC cell proliferation (Fig. 2) (79, 80). Polyamines regulate gene expression by binding to negatively charged macromolecules, such as DNA, RNA and proteins. In transcriptional level, polyamines could condense and aggregate DNA and induce B-Z and B-A conformational transitions in vitro (81, 82). In posttranscriptional level, polyamines could regulate the subcellular localization of the RNA-binding proteins human antigen R (HuR) and CUGBP1 which stabilizes its target transcripts such as JunD, CDK4, c-Myc, p53 and NPM mRNA (Fig. 2) (70, 79, 83-85).

APOPTOSIS

Esophageal and gastric epithelia

The GI epithelial homeostasis depends on a balance between cell proliferation and apoptosis. Studies showed that polyamine depletion by DFMO induces esophageal epithelial apoptosis by altering both Bcl-2 and Bax expression (61). Helicobacter pylori (H. pylori) induced oxidative stress could induce apoptosis in host gastric epithelial cells and macrophages by a mitochondrial-dependent cell death pathway (86). The oxidative stress mediated by hydrogen peroxide (H2O2) is generated by the polyamines via the inducible SPMO in H. pylori infected cells (86, 87). Increased expression of polyamines, ODC and SPMO were observed in both H. pylori infected human and mouse gastritis tissues (86, 88, 89). Inhibition or knockdown of SPMO or ODC remarkably attenuates apoptosis and DNA damage in gastric epithelial cells (86). Macrophage apoptosis was stimulated by the polyamines and was abolished by inhibition of ODC or SPMO activity (87, 90). H. pylori infected mice treated with DFMO significantly reduced both H. pylori colonization levels and gastritis severity (88). The phosphorylated extracellular regulated protein kinases (p-ERK) › phosphorylated c-Fos/c-Jun › c-Myc › ODC › SPMO › H2O2 › cytochrome C › caspase-3 pathway might involve in the polyamine-mediated apoptosis in H. pylori induced gastric diseases (Fig. 2) (87, 89, 91).

Intestinal epithelia

Apoptosis occurs in the crypt area or the position of epithelial stem cells (92). It has been shown that polyamine depletion promotes the resistance of IECs to apoptosis by stabilization of activating transcription factor 2 (ATF-2), X chromosome-linked inhibitor of apoptosis protein (XIAP) and MAPK mitogen-activated protein kinase kinase-1 (MEK1) mRNA and increases translation though enhancing HuR levels (Fig. 2). Elevation of cytoplasmic HuR levels by inhibition of cellular polyamines increased ATF-2, XIAP and MEK1 mRNA and protein levels, whereas HuR silencing make these mRNAs unstable and suppressed ATF-2, XIAP and MEK1 expression (92-94). Additionally, depletion of cellular polyamines by DFMO protects normal IEC-6 cells from tumor necrosis factor-α (TNF-α) induced apoptosis. Elevation of nuclear factor-κB (NF-κB) transcriptional activity by reduction of inhibitor of NF-κB a (IκBα) and decreased TRPC1-mediated [Ca2+]cyt were observed after polyamine depletion, indicating that these pathways might also involve in the regulation of polyamine-dependent IEC apoptosis (Fig. 2) (95, 96). Furthermore, polyamine depletion could increase phosphorylation of Akt by enhancing 3-phosphoinositide-dependent protein kinase-1 (PDK1) activity, thereby protecting IECs against TNF-α/cycloheximide (CHX) induced apoptosis (Fig. 2) (97). The stress granules (SGs) also implicate the protection of IECs from apoptosis (98). Following polyamine depletion, SGs formation was activated by silencing SGs resident proteins Sort1 and T-cell intracellular antigen 1 (TIA1), which in turn increase resistance to TNF-α/CHX induced IEC-6 apoptosis (Fig. 2) (99).

Gastrointestinal stem cells

In adult, stem cells are characterized by their self-renewal capacity to generate multiple differentiated cell types (100). The GI epithelium represents one of the most rapidly self-renewing tissues in adult mammals, which continuously self-renews for every 2–5 days (101). It has been suggested that GI stem cells are important for the maintenance of GI mucosal homeostasis (102).

Esophageal and gastric stem cells

Recent studies reveal that esophageal epithelium is maintained and repaired by stem cells (103, 104). Esophageal stem cells are thought to reside within the basal layer of the stratified squamous epithelium (102). Esophageal stem cells are a reversible switch between homeostatic and regenerative in response to wound healing (104).

Each gastric unit of mammalian adult stomach self-renews continually by a tiny population of stem cells (105). Gastric stem cells are important for gastric epithelial repair after injury (106). Although gastric stem cells have been investigated for more than two decades, it has not yet been identified for lack of specific markers (102). Putative gastric stem cells are supposed to locate to the isthmus, the middle portion of the tubule (107). Recently, a subpopulation of gastric progenitors with multiple potential has been identified (108). By using genetic and chemical inhibition approaches, Notch signal pathway is demonstrated to be required for the maintenance of gastric stem cell homeostasis (109).

Intestinal stem cells

Two types of stem cells have been currently defined in the intestine: leucine-rich-repeat-containing G-protein-coupled receptor 5 (Lgr5) positive crypt base columnar (CBC) cells and Bmi1 positive quiescent ‘+4’ cells. CBC cells are predominantly located at the crypt base and cycle rapidly, while the ‘+4’ cells are situated above the crypt base and are largely reside (110-112). Additionally, these two stem cell populations could dynamically interconvert in distinct niches (101). The population of intestinal stem cells are descending gradient from duodenum to colon (113). In a recent study, Lgr5+ stem cells were grown in vitro to produce organoids of epithelial tissue, and the damaged epithelium was rescued by which the organoids were engraft onto the damaged colon within the dextran sulfate sodium (DSS)-induced colitis in mice (114). This study provides proof of principle that stem-cell-based therapies can be applied for repairing damaged epithelium.

The exact role of polyamines in the differentiation of GI stem cells has not been characterized. The polyamine pathway was recently identified to be essential for self-renewal and differentiation of embryonic stem cells, adipose-derived stem cells and mesenchymal stem cell (115-118), which indicated that polyamines might be important for self-renewal and differentiation of GI stem cells.

ANGIOGENESIS

Restitution and subsequent proliferation are effective processes to repair superficial lesions but not deeper lesions in the GI tract. Angiogenesis and proliferation of fibroblasts are involved in the healing of mucosal lesions in deeper damages into muscularis mucosae and lamina propria (119).

Angiogenesis, the development of new blood vessels from the existing vasculature, brings new capillaries and larger blood vessels which are essential for clearance of necrotic tissue and stimulates fibroblasts proliferation as it delivers oxygen and nutrients to injury sites (5, 120). Thus, angiogenesis might be a fundamental process in the wound healing of the GI tract. Angiogenesis in the damaged GI tract is stimulated by various factors, including vascular endothelial growth factor (VEGF), play a pivotal role in the GI mucosa (121).

Angiogenesis in healing of esophagus and stomach

In an eosinophilic esophagitis mice model, mice challenged with egg ovalbumin developed a significant increase in the number of small vessels within the esophageal and remarkably elevated the number of VEGF-positive cells (122), which reveals that angiogenesis might involve in the healing of esophageal injuries.

Previous studies have demonstrated that angiogenesis is commonly associated with gastritis, peptic ulcer, and gastric carcinoma (123). The angiogenesis which is stimulated to repair the gastric mucosa is subsequently increased in patients with H. pylori infection (124). Additionally, there are an increased concentration of angiogenic factors and the formation of new blood vessels in H. pylori-associated diseases. Compared with H. pylori negative patients, the expression of VEGF mRNA and protein was increased in the H. pylori positive dyspeptic patients (125). In a water-immersion and restraint stress induced gastric ulcer rat model and thermal cauterization induced gastric ulcer mice model, VEGF expression and angiogenesis were remarkably increased compared with normal controls (126, 127). The increased VEGF and angiogenesis is important for protection and healing of gastric ulcers (128-130). In contrast, inhibition of VEGF delayed ulcer healing (131). Thus, angiogenesis might facilitate the healing of gastric lesions.

Angiogenesis in healing of intestine

Angiogenesis is accompanied with chronic inflammation in many diseases (132). Stimulation of angiogenesis by treatment with genes or peptides of basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF) or VEGF are sufficient to accelerate duodenal ulcer healing in the GI tract respectively, while blockade of these angiogenic factors resulted in delaying ulcer healing (119, 133, 134). Furthermore, it has been illustrated that angiogenesis might be implicated in the pathogenesis of ulcerative colitis (UC) and Crohn’s disease (CD), the two major forms of inflammatory bowel diseases (IBD) (135, 136). Increased mucosal vascular density and pro-angiogenic molecules content (e.g. VEGF, placental growth factor (PIGF), PDGF and bFGF) were associated with progression of human IBD and animal models of UC (137, 138). Promoting angiogenesis with these pro-angiogenic molecules in experimental models of colitis increased mucosal angiogenesis (139, 140). Blockade of angiogenesis by specific antibodies to neutralize VEGF, integrin avb3 and bFGF is effective to ameliorate experimental colitis (135, 139, 141). Thus, pharmacological inhibition of angiogenesis may represent a new and promising therapeutic target in experimental models of IBD.

However, given the beneficial effect of anti-angiogenesis treatment in IBD, additional studies indicated that stimulation of angiogenesis by PIGF, PDGF or bFGF remarkably accelerated healing of experimental models of UC (137, 138, 142). Blockade of angiogenesis by specific inhibition of PIGF leads an increased disease activity during DSS-induced colitis, which was subsequently prevented by exogenous administration of recombinant PIGF (143). In the early stages of IBD, increased angiogenesis could accelerate the influx of inflammatory cells. However, reduced vessel diameter, decreased vascular density and diminished blood flow were observed in advanced IBD lesions which resulting in sustained tissue hypo-perfusion and ischemia in the colon (132, 144). Thus, anti-angiogenesis therapy might be beneficial in the early stage of IBD but harmful in the advanced stage of IBD.

The effect of polyamines on angiogenesis is mainly explored in cancer cells. The angiogenesis in tumor was completely abrogated with DFMO, and the anti-angiogenesis effect of DFMO could be reversed by the exogenous addition of polyamines (145). Although polyamines are widely applied in the wound healing of the GI tract, mechanisms of polyamines on angiogenesis are not fully understood. Polyamines significantly increased gastric blood flow and accelerated ulcer healing. Inhibition of ODC activity with DFMO remarkably reduced gastric blood flow and delayed ulcer healing (67). Thus, the beneficial effect of polyamines on the wound healing is also facilitated by angiogenesis.

GASTROINTESTINAL BARRIER FUNCTION

Most GI mucosa are covered by mucus gel barrier which is secreted by specialized epithelial cells and created the first barrier to protect large particles from directly contacting epithelial cells (146). Additionally, the surface of GI mucosa is lined by epithelial cells which establish the second barrier between hostile external environments and the underlying tissue. The effectiveness and stability of these epithelial barriers are maintained by tight junctions, adherents, junctions, desmosomes, and gap junctions (2, 146). Defective of epithelium barriers have been demonstrated to be crucial in the pathogenesis of many GI disorders, such as gastroesophageal reflux, H. pylori related gastritis and IBD (147-150).

Esophageal barrier function

Mucus gel barrier is absent in the normal esophagus (151), thus, the stratified squamous epithelium lined within esophagus establish the first barrier to protect the underlying tissue from mechanical and chemical damage. Reflux of acid and bile acids is the most important agent damaging esophageal epithelial barrier which could lead to gastroesophageal reflux diseases (148). Although the influence of refluxed agents on esophageal epithelial barrier function remains unclear, decreased expression of tight junction proteins, e.g. claudin and occludin, might contribute to refluxed agents induced esophageal epithelial barrier impairments (152).

Gastric barrier function

Mucus-bicarbonate-phospholipid barrier and epithelial barrier of the stomach maintain an effective defense against toxins, H. pylori, hydrochloric acid, pepsin, bile acids and other noxious factors. A large number of studies have manifested that H. pylori impairs the gastric barriers. In a chronic H. pylori infection mice model, the firmly adherent mucus layer was significantly reduced in infected mice compared within control mice. And the ability of the gastric mucosa to maintain a neutral pH at the epithelial cell surface was also altered after chronic H. pylori infection (153). Furthermore, several studies have demonstrated that H. pylori could disrupt localization of occludin, claudin and zona occludens-1 (ZO-1) at the tight junction and reduce expression of E-cadherin at adherens junctions, which ultimately resulting alteration of epithelial barrier (154-156). Spermidine, but not putrescine or spermine, not only reversed reduction of mucus synthesis but also significantly promoted ulcer healing in gastric ulcer bearing rats (47).

Intestinal barrier function

Epithelial barrier (forming by monolayer columnar epithelium) and pre-epithelial barrier (containing a thick mucus layer, trefoil peptides and immunoglobulin A) are physical and functional barriers to separate million of bacteria from intestinal lumen to the underlying host tissues (157). Disruption of these barriers is associated with a variety of intestinal diseases, including diarrhea, IBD and cancer (157-159). Increasing evidences have demonstrated that polyamines are required for maintenance of intestinal barrier function. Decreased polyamine level was associated with dysfunction of the epithelial barrier, which was ameliorated by exogenous polyamines. The epithelial barrier function was regulated by polyamines though modulating the expression of ZO-1, occludin and E-cadherin (Fig. 2). Occludin, claudins, ZO-1 and ZO-2 are the major transmembrane and cytosolic tight junction proteins. Suppression of polyamines result in increased JunD expression which regulate ZO-1 expression through both cAMP response element-binding protein-binding (CREB) site within ZO-1 proximal promoter region and enhancing the interaction of the ZO-1 3’-untranslated region (3’UTR) with RNA-binding protein TIA1-related protein (TIAR) (Fig. 2) (160). Reduction of checkpoint kinase 2 (Chk2) by polyamine depletion reduces HuR phosphorylation and substantially inhibits occludin translation (161). Alternatively, the translation of occludin might also modulate through competitive binding of CUGBP1 and HuR to occludin 3’UTR (Fig. 2) (162). E-cadherin is mainly located at the adherens junctions and plays a critical role in maintenance of GI epithelial barrier function. Depletion of cellular polyamine decrease E-cadherin expression though c-Myc mediated reduction of E-cadherin promoter activity and its mRNA expression (Fig. 2) (163, 164).

ROLE OF POLYAMINES IN COMMON GASTROINTESTINAL DISEASES

Polyamine metabolism is involved with a wide variety of GI diseases. Numerous factors may influence polyamine homeostasis, however the changes seem to be tissue-specific. Chronic biliopancreatic reflux which causes severe esophagitis increases ODC activity and polyamine levels (165). The altered polyamine metabolism contributes to epithelial cell proliferation and esophageal carcinogenesis in experimental animals (165, 166). ODC activity is upregulated in BE with a premalignant lesion of adenocarcinoma and correlated with the degree of dysplasia (167). DFMO, an irreversible inhibitor of ODC, is considered as a promising chemoprevention agent for esophageal adenocarcinoma by suppressing polyamine content in BE mucosa (166).

Polyamine metabolism is also involved in gastric mucosal injury. H. pylori, a major pathological factor of gastric tissue, induces immune dysregulation by increasing ODC activity and polyamine content in macrophages (88, 89, 168). Interestingly, H. pylori can not induce ODC in gastric epithelium (89), but up-regulates polyamine catabolism and aggravates polyamine-mediated oxidative stress (89). On the other hand, polyamines seem to provide a protective role against other detrimental factors of gastric tissue such as aspirin, smoking and stress (47, 68, 169). During stress ulcers, both ODC activity and polyamine level are up-regulated and ultimately improved mucosal healing (170). In an animal experiment, oral administration of polyamines, putrescine, cadaverine, spermidine or spermine, immediately after stress accelerate the normal rate of healing (68). DFMO inhibits the early phase of ulcer healing, however, it does not influence ulcer recovery eventually, which may indicate an alternate source of polyamines besides ODC pathway are required in these experimental models (171).

The relationship between polyamines and IBD is extensively studied. Conflicting results are reported in different studies regarding the ODC activity and polyamine content in IBD patients. It has been found that ODC activity was decreased in IBD patients both in involved and uninvolved mucosal tissues (172). Furthermore, the decrease of ODC was related to the severity of disease (172). In contrast, ODC was found to be elevated both in human and animal studies (173-175). The discrepancy may be due to the content of tissue sample as previously indicated in gastric tissue (171, 176). A few studies indicate that increased levels of spermidine and polyamine catabolism in IBD patients and experimental models which may relate to the accelerated proliferation of injured tissues (175, 177). Spermine exerts an inhibitory role on the inflammatory reaction and is down-regulated in severe ulcerative colitis patients and in chronic colitis experimental models (176, 178). It has been noticed that the decrease of spermine content may further aggravate the disease (176). The role of polyamine metabolism in IBD is further supported by the fact that L-arginine improves colitis by enhancing the formation of polyamines in animal models while DFMO worsen the disease (175).

Pathological conditions outside the GI tract also closely relate to polyamine metabolism and interfere with internal polyamine pool. In experimental pancreatitis, polyamine catabolism correlates with the degree of pancreatic necrosis with depletion of blood spermidine and spermine levels and elevation of putrescine level (179). Furthermore, increase of putrescine level is also found in remote organs such as liver, lung and kidney which may relate to remote organ injury during pancreatitis (180). However, in humans only the increase of blood putrescine level is noticed in necrotic pancreatitis (181). During cirrhosis and primary hepatic tumor, erythrocyte polyamines (spermine and spermidine) are elevated which may be due to hepatic cell regeneration (182). Other factors such as surgery, trauma and radiation also influence blood polyamine levels (183, 184). However, the exact mechanisms on the role of internal polyamine pool affecting GI mucosal homeostasis are not yet clearly understood.

MALNUTRITION AND POLYAMINES

Polyamine metabolism is closely related to the whole-body energy and protein metabolism (185). In animal experiments, ODC activity and polyamine content in liver drop significantly after starvation for three days, and re-feeding can rapidly increase these parameters (186). Protein-deprivation diet, on the other hand, induces ODC activity and polyamine content in the early phases in liver and colon tissues (187, 188). It has been reported that the ODC activity and polyamine content were not decreased in liver tissue until the fifth week of protein restriction (188). Feeding with polyamine-related amino acids like methionine, arginine or ornithine does not increase muscle or liver concentrations of polyamines in rats (189). Also, in humans, correction of nutritional deficits does not contribute significantly to the red blood cells (RBC) polyamine pool in patients with nonmalignant diseases (190). Studies indicate that the dietary polyamines are essential for intestinal polyamine pool and significantly contribute to the integrity of GI mucosa (176, 191). Recent studies reveal the relationship between dietary polyamine levels and risk of colorectal adenoma, and low polyamine diet may contribute to the control of certain tumors (192-194). Future studies are warranted to clarify polyamine metabolism in the GI tract under critical situation like fasting and malnutrition, and explore the possible therapeutic use of dietary polyamines.

CONCLUSIONS

An increasing body of evidence indicating that polyamines are necessary for normal GI mucosal growth and that decreasing cellular polyamines inhibits cell proliferation and disrupts epithelial mucosal integrity. Polyamines are shown to regulate intestinal epithelial cell renewal by virtue of their ability to modulate expression of various growth promoting genes. Increasing the levels of cellular polyamines accelerate GI growth and cell proliferation by enhancement of several gene transcriptional and translational abilities, whereas growth inhibition following polyamine depletion results primarily from the activation of growth-inhibiting genes. Moreover, polyamines also regulate apoptosis and barrier function of intestinal epithelial cells through multiple signaling pathways. However, there are still many critical issues that remain to be addressed regarding the roles of polyamines in maintenance of gut epithelial integrity.

Acknowledgements: This study are supported by National Natural Science Fund of China (Grant NO. 81170413, to Cheng-Wei Tang).

Conflict of interests: None declared.

REFERENCES

  1. Jankowski JA, Goodlad RA, Wright NA. Maintenance of normal intestinal mucosa: function, structure, and adaptation. Gut 1994; 35(Suppl. 1): S1-S4.
  2. Catalioto RM, Maggi CA, Giuliani S. Intestinal epithelial barrier dysfunction in disease and possible therapeutical interventions. Curr Med Chem 2011; 18: 398-426.
  3. Timmons J, Chang ET, Wang J-Y, Rao JN. Polyamines and gut mucosal homeostasis. J Gastrointest Digest Sys 2012; S7: 001: 1-9.
  4. Iizuka M, Konno S. Wound healing of intestinal epithelial cells. World J Gastroenterol 2011; 17: 2161-2171.
  5. Mammen JM, Matthews JB. Mucosal repair in the gastrointestinal tract. Crit Care Med 2003; 31(Suppl. 8): S532-S537.
  6. Dignass AU. Mechanisms and modulation of intestinal epithelial repair. Inflamm Bowel Dis 2001; 7: 68-77.
  7. Li SP, Tang TS, Yiu KS, Lo KK. Cyclometalated iridium(III)-polyamine complexes with intense and long-lived multicolor phosphorescence: synthesis, crystal structure, photophysical behavior, cellular uptake, and transfection properties. Chemistry 2012; 18: 13342-13354.
  8. Casero RA, Jr., Marton LJ. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov 2007; 6: 373-390.
  9. Tanabe T, Shin M, Fujiwara K. Immunoelectron microscopy study of polyamines using a newly prepared monoclonal antibody against spermidine: use of a mixture of glutaraldehyde and paraformaldehyde as a cross-linking agent in the preparation of the antigen. J Biochem 2004; 135: 501-507.
  10. Fujiwara K, Masuyama Y, Kitagawa T. Immunocytochemical localization of polyamines in the gastrointestinal tracts of rats and mice. Histochem Cell Biol 1996; 106: 465-471.
  11. Farriol M, Segovia-Silvestre T, Venereo Y, Orta X. Polyamines in the gastrointestinal tract. Nutr Hosp 2000; 15: 85-91.
  12. Bardocz S, White A. Estimation of the polyamine body pool: contribution by de novo biosynthesis, diet and luminal bacteria. In: Polyamines in Health and Nutrition. S. Bardocz, A. White (eds.). Boston, Kluwer Academic Publishers, 1999, pp. 117-127.
  13. Ali MA, Poortvliet E, Stromberg R, Yngve A. Polyamines: total daily intake in adolescents compared to the intake estimated from the Swedish Nutrition Recommendations Objectified (SNO). Food Nutr Res 2011; 55. Doi: 10.3402/fnr.v55i0.5455.
  14. Seiler N, Raul F. Polyamines and the intestinal tract. Crit Rev Clin Lab Sci 2007; 44: 365-411.
  15. Milovic V. Polyamines in the gut lumen: bioavailability and biodistribution. Eur J Gastroenterol Hepatol 2001; 13: 1021-1025.
  16. Milovic V, Faust D, Turchanowa L, Stein J, Caspary WF. Permeability characteristics of polyamines across intestinal epithelium using the Caco-2 monolayer system: comparison between transepithelial flux and mitogen-stimulated uptake into epithelial cells. Nutrition 2001; 17: 462-466.
  17. Soulet D, Gagnon B, Rivest S, Audette M, Poulin R. A fluorescent probe of polyamine transport accumulates into intracellular acidic vesicles via a two-step mechanism. J Biol Chem 2004; 279: 49355-49366.
  18. Belting M, Persson S, Fransson LA. Proteoglycan involvement in polyamine uptake. Biochem J 1999; 338: 317-323.
  19. Belting M, Mani K, Jonsson M, et al. Glypican-1 is a vehicle for polyamine uptake in mammalian cells: a pivital role for nitrosothiol-derived nitric oxide. J Biol Chem 2003; 278: 47181-47189.
  20. Roy UK, Rial NS, Kachel KL, Gerner EW. Activated K-RAS increases polyamine uptake in human colon cancer cells through modulation of caveolar endocytosis. Mol Carcinog 2008; 47: 538-553.
  21. Uemura T, Stringer DE, Blohm-Mangone KA, Gerner EW. Polyamine transport is mediated by both endocytic and solute carrier transport mechanisms in the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2010; 299: G517-G522.
  22. Uemura T, Yerushalmi HF, Tsaprailis G, et al. Identification and characterization of a diamine exporter in colon epithelial cells. J Biol Chem 2008; 283: 26428-26435.
  23. Abdulhussein AA, Wallace HM. Polyamines and membrane transporters. Amino Acids 2013; Jul 13. [epub ahead of print]
  24. Parveen N, Cornell KA. Methylthioadenosine/S-adenosylhomocysteine nucleosidase, a critical enzyme for bacterial metabolism. Mol Microbiol 2011; 79: 7-20.
  25. Persson L. Polyamine homoeostasis. Essays Biochem 2009; 46: 11-24.
  26. Palanimurugan R, Scheel H, Hofmann K, Dohmen RJ. Polyamines regulate their synthesis by inducing expression and blocking degradation of ODC antizyme. EMBO J 2004; 23: 4857-4867.
  27. Nusrat A, Delp C, Madara JL. Intestinal epithelial restitution. Characterization of a cell culture model and mapping of cytoskeletal elements in migrating cells. J Clin Invest 1992; 89: 1501-1511.
  28. Palileo C, Kaunitz JD. Gastrointestinal defense mechanisms. Curr Opin Gastroenterol 2011; 27: 543-548.
  29. Hoffmann W. Trefoil factors TFF (trefoil factor family) peptide-triggered signals promoting mucosal restitution. Cell Mol Life Sci 2005; 62: 2932-2938.
  30. Taupin D, Podolsky DK. Trefoil factors: initiators of mucosal healing. Nat Rev Mol Cell Biol 2003; 4: 721-732.
  31. Dignass AU, Podolsky DK. Cytokine modulation of intestinal epithelial cell restitution: central role of transforming growth factor beta. Gastroenterology 1993; 105: 1323-1332.
  32. Takahashi M, Ota S, Ogura K, Nakamura T, Omata M. Hepatocyte growth factor stimulates wound repair of the rabbit esophageal epithelial cells in primary culture. Biochem Biophys Res Commun 1995; 216: 298-305.
  33. Jimenez P, Lanas A, Piazuelo E, Esteva F. Effect of growth factors and prostaglandin E2 on restitution and proliferation of rabbit esophageal epithelial cells. Dig Dis Sci 1998; 43: 2309-2316.
  34. Jimenez P, Lanas A, Piazuelo E, Esteva F. Effects of extracellular pH on restitution and proliferation of rabbit oesophageal epithelial cells. Aliment Pharmacol Ther 1999; 13: 545-552.
  35. De Backer A, Haentjens P, Willems G. Hydrochloric acid. A trigger of cell proliferation in the esophagus of dogs. Dig Dis Sci 1985; 30: 884-890.
  36. Livstone EM, Sheahan DG, Behar J. Studies of esophageal epithelial cell proliferation in patients with reflux esophagitis. Gastroenterology 1977; 73: 1315-1319.
  37. Yanaka A, Suzuki H, Shibahara T, Matsui H, Nakahara A, Tanaka N. EGF promotes gastric mucosal restitution by activating Na(+)/H(+) exchange of epithelial cells. Am J Physiol Gastrointest Liver Physiol 2002; 282: G866-G876.
  38. Takahashi M, Ota S, Shimada T, et al. Hepatocyte growth factor is the most potent endogenous stimulant of rabbit gastric epithelial cell proliferation and migration in primary culture. J Clin Invest 1995; 95: 1994-2003.
  39. Yoshizawa T, Watanabe S, Hirose M, et al. Effects of growth factors on aspirin-induced inhibition of wound repair in a rabbit gastric epithelial cell model. Aliment Pharmacol Ther 2000; 14(Suppl. 1): 176-182.
  40. Tetreault MP, Chailler P, Beaulieu JF, Rivard N, Menard D. Epidermal growth factor receptor-dependent PI3K-activation promotes restitution of wounded human gastric epithelial monolayers. J Cell Physiol 2008; 214: 545-557.
  41. Horie-Sakata K, Shimada T, Hiraishi H, Terano A. Role of cyclooxygenase 2 in hepatocyte growth factor-mediated gastric epithelial restitution. J Clin Gastroenterol 1998; 27(Suppl. 1): S40-S46.
  42. Nagoshi S, Ota S, Fujiwara K. Contribution of spermidine to stimulation by hepatocyte growth factor in repair after damage of rabbit gastric mucosal cells in primary culture. Biochim Biophys Acta 2003; 1619: 187-192.
  43. Banan A, McCormack SA, Johnson LR. Polyamines are required for microtubule formation during gastric mucosal healing. Am J Physiol 1998; 274: G879-G885.
  44. Mechulam A, Chernov KG, Mucher E, Hamon L, Curmi PA, Pastre D. Polyamine sharing between tubulin dimers favours microtubule nucleation and elongation via facilitated diffusion. PLoS Comput Biol 2009; 5: e1000255.
  45. Hamon L, Savarin P, Curmi PA, Pastre D. Rapid assembly and collective behavior of microtubule bundles in the presence of polyamines. Biophys J 2011; 101: 205-216.
  46. Banan A, Wang JY, McCormack SA, Johnson LR. Relationship between polyamines, actin distribution, and gastric healing in rats. Am J Physiol 1996; 271: G893-G903.
  47. Ma L, Wang WP, Chow JY, Lam SK, Cho CH. The role of polyamines in gastric mucus synthesis inhibited by cigarette smoke or its extract. Gut 2000; 47: 170-177.
  48. Rao JN, Li J, Li L, Bass BL, Wang JY. Differentiated intestinal epithelial cells exhibit increased migration through polyamines and myosin II. Am J Physiol 1999; 277: G1149-G1158.
  49. Rao JN, Liu L, Zou T, et al. Polyamines are required for phospholipase C-gamma1 expression promoting intestinal epithelial restitution after wounding. Am J Physiol Gastrointest Liver Physiol 2007; 292: G335-G343.
  50. Rao JN, Li L, Golovina VA, et al. Ca2+-RhoA signaling pathway required for polyamine-dependent intestinal epithelial cell migration. Am J Physiol Cell Physiol 2001; 280: C993-C1007.
  51. Wang JY, Wang J, Golovina VA, Li L, Platoshyn O, Yuan JX. Role of K(+) channel expression in polyamine-dependent intestinal epithelial cell migration. Am J Physiol Cell Physiol 2000; 278: C303-C314.
  52. Timmons JA, Rao JN, Turner DJ, et al. Induced expression of STIM1 sensitizes intestinal epithelial cells to apoptosis by modulating store-operated Ca2+ influx. J Gastrointest Surg 2012; 16: 1397-1405.
  53. Rao JN, Rathor N, Zou T, et al. STIM1 translocation to the plasma membrane enhances intestinal epithelial restitution by inducing TRPC1-mediated Ca2+ signaling after wounding. Am J Physiol Cell Physiol 2010; 299: C579-C588.
  54. Rao JN, Platoshyn O, Golovina VA, et al. TRPC1 functions as a store-operated Ca2+ channel in intestinal epithelial cells and regulates early mucosal restitution after wounding. Am J Physiol Gastrointest Liver Physiol 2006; 290: G782-G792.
  55. Rao JN, Rathor N, Zhuang R, et al. Polyamines regulate intestinal epithelial restitution through TRPC1-mediated Ca(2)+ signaling by differentially modulating STIM1 and STIM2. Am J Physiol Cell Physiol 2012; 303: C308-C317.
  56. Guo X, Rao JN, Liu L, Rizvi M, Turner DJ, Wang JY. Polyamines regulate beta-catenin tyrosine phosphorylation via Ca(2+) during intestinal epithelial cell migration. Am J Physiol Cell Physiol 2002; 283: C722-C734.
  57. Rao JN, Liu SV, Zou T, et al. Rac1 promotes intestinal epithelial restitution by increasing Ca2+ influx through interaction with phospholipase C-(gamma)1 after wounding. Am J Physiol Cell Physiol 2008; 295: C1499-C1509.
  58. Rao JN, Guo X, Liu L, et al. Polyamines regulate Rho-kinase and myosin phosphorylation during intestinal epithelial restitution. Am J Physiol Cell Physiol 2003; 284: C848-C859.
  59. Bavaria MN, Ray RM, Johnson LR. The phosphorylation state of MRLC is polyamine dependent in intestinal epithelial cells. Am J Physiol Cell Physiol 2011; 300: C164-C175.
  60. Ray RM, Guo H, Patel M, Jin S, Bhattacharya S, Johnson LR. Role of myosin regulatory light chain and Rac1 in the migration of polyamine-depleted intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2007; 292: G983-G995.
  61. Fong LY, Pegg AE, Magee PN. Alpha-difluoromethylornithine inhibits N-nitrosomethylbenzylamine-induced esophageal carcinogenesis in zinc-deficient rats: effects on esophageal cell proliferation and apoptosis. Cancer Res 1998; 58: 5380-5388.
  62. Sinicrope FA, Broaddus R, Joshi N, et al. Evaluation of difluoromethylornithine for the chemoprevention of Barrett’s esophagus and mucosal dysplasia. Cancer Prev Res (Phila) 2011; 4: 829-839.
  63. Jainu M, Vijaimohan K, Kannan K. Cissus quadrangularis L. extract attenuates chronic ulcer by possible involvement of polyamines and proliferating cell nuclear antigen. Pharmacogn Mag 2010; 6: 225-233.
  64. Majumdar AP, Luk GD. Gastric epithelial cell proliferation after injury. J Clin Gastroenterol 1992; 14(Suppl. 1): S25-S28.
  65. Kidd M, Tang LH, Schmid S, Lauffer J, Louw JA, Modlin IM. Helicobacter pylori lipopolysaccharide alters ECL cell DNA synthesis via a CD14 receptor and polyamine pathway in mastomys. Digestion 2000; 62: 217-224.
  66. Makitie LT, Kanerva K, Andersson LC. Ornithine decarboxylase regulates the activity and localization of rhoA via polyamination. Exp Cell Res 2009; 315: 1008-1014.
  67. Konturek PC, Brzozowski T, Konturek SJ, Szlachcic A, Hahn EG. Polyamines and epidermal growth factor in the recovery of gastric mucosa from stress-induced gastric lesions. J Clin Gastroenterol 1998; 27(Suppl. 1): S97-S104.
  68. Wang JY, Johnson LR. Luminal polyamines stimulate repair of gastric mucosal stress ulcers. Am J Physiol 1990; 259: G584-G592.
  69. Rathor N, Wang SR, Chang ET, Rao JN. Differentiated intestinal epithelial cells express high levels of TGF-beta receptors and exhibit increased sensitivity to growth inhibition. Int J Clin Exp Med 2011; 4: 299-308.
  70. Wang JY. Polyamines and mRNA stability in regulation of intestinal mucosal growth. Amino Acids 2007; 33: 241-252.
  71. Patel AR, Wang JY. Polyamines modulate transcription but not posttranscription of c-myc and c-jun in IEC-6 cells. Am J Physiol 1997; 273: C1020-C1029.
  72. Wang JY, McCormack SA, viar MJ, et al. Decreased expression of protooncogenes c-fos, c-myc, and c-jun following polyamine depletion in IEC-6 cells. Am J Physiol 1993; 265: G331-G338.
  73. Zhang AH, Rao JN, Zou T, et al. p53-dependent NDRG1 expression induces inhibition of intestinal epithelial cell proliferation but not apoptosis after polyamine depletion. Am J Physiol Cell Physiol 2007; 293: C379-C389.
  74. Rao JN, Li L, Bass BL, Wang JY. Expression of the TGF-beta receptor gene and sensitivity to growth inhibition following polyamine depletion. Am J Physiol Cell Physiol 2000; 279: C1034-C1044.
  75. Li L, Rao JN, Guo X, et al. Polyamine depletion stabilizes p53 resulting in inhibition of normal intestinal epithelial cell proliferation. Am J Physiol Cell Physiol 2001; 281: C941-C953.
  76. Zou T, Rao JN, Liu L, et al. Polyamine depletion induces nucleophosmin modulating stability and transcriptional activity of p53 in intestinal epithelial cells. Am J Physiol Cell Physiol 2005; 289: C686-C696.
  77. Patel AR, Wang JY. Polyamine depletion is associated with an increase in JunD/AP-1 activity in small intestinal crypt cells. Am J Physiol 1999; 276: G441-G450.
  78. Zou T, Mazan-Mamczarz K, Rao JN, et al. Polyamine depletion increases cytoplasmic levels of RNA-binding protein HuR leading to stabilization of nucleophosmin and p53 mRNAs. J Biol Chem 2006; 281: 19387-19394.
  79. Xiao L, Cui YH, Rao JN, et al. Regulation of cyclin-dependent kinase 4 translation through CUG-binding protein 1 and microRNA-222 by polyamines. Mol Biol Cell 2011; 22: 3055-3069.
  80. Xiao L, Rao JN, Zou T, et al. Induced JunD in intestinal epithelial cells represses CDK4 transcription through its proximal promoter region following polyamine depletion. Biochem J 2007; 403: 573-581.
  81. Feuerstein BG, Williams LD, Basu HS, Marton LJ. Implications and concepts of polyamine-nucleic acid interactions. J Cell Biochem 1991; 46: 37-47.
  82. Yang L, Wang S, Tian T, Zhou X. Advancements in Z-DNA: development of inducers and stabilizers for B to Z transition. Curr Med Chem 2012; 19: 557-568.
  83. Zou T, Rao JN, Liu L, et al. Polyamines regulate the stability of JunD mRNA by modulating the competitive binding of its 3’ untranslated region to HuR and AUF1. Mol Cell Biol 2010; 30: 5021-5032.
  84. Liu L, Rao JN, Zou T, et al. Polyamines regulate c-Myc translation through Chk2-dependent HuR phosphorylation. Mol Biol Cell 2009; 20: 4885-4898.
  85. Xiao L, Wang JY. Posttranscriptional regulation of gene expression in epithelial cells by polyamines. Methods Mol Biol 2011; 720: 67-79.
  86. Xu H, Chaturvedi R, Cheng Y, et al. Spermine oxidation induced by Helicobacter pylori results in apoptosis and DNA damage: implications for gastric carcinogenesis. Cancer Res 2004; 64: 8521-8525.
  87. Chaturvedi R, Cheng Y, Asim M, et al. Induction of polyamine oxidase 1 by Helicobacter pylori causes macrophage apoptosis by hydrogen peroxide release and mitochondrial membrane depolarization. J Biol Chem 2004; 279: 40161-40173.
  88. Chaturvedi R, Asim M, Hoge S, et al. Polyamines impair immunity to Helicobacter pylori by inhibiting L-arginine uptake required for nitric oxide production. Gastroenterology 2010; 139: 1686-1698, 1698.
  89. Chaturvedi R, de Sablet T, Coburn LA, Gobert AP, Wilson KT. Arginine and polyamines in Helicobacter pylori-induced immune dysregulation and gastric carcinogenesis. Amino Acids 2012; 42: 627-640.
  90. Gobert AP, Cheng Y, Wang JY, et al. Helicobacter pylori induces macrophage apoptosis by activation of arginase II. J Immunol 2002; 168: 4692-4700.
  91. Asim M, Chaturvedi R, Hoge S, et al. Helicobacter pylori induces ERK-dependent formation of a phospho-c-Fos c-Jun activator protein-1 complex that causes apoptosis in macrophages. J Biol Chem 2010; 285: 20343-20357.
  92. Xiao L, Rao JN, Zou T, et al. Polyamines regulate the stability of activating transcription factor-2 mRNA through RNA-binding protein HuR in intestinal epithelial cells. Mol Biol Cell 2007; 18: 4579-4590.
  93. Zhang X, Zou T, Rao JN, et al. Stabilization of XIAP mRNA through the RNA binding protein HuR regulated by cellular polyamines. Nucleic Acids Res 2009; 37: 7623-7637.
  94. Wang PY, Rao JN, Zou T, et al. Post-transcriptional regulation of MEK-1 by polyamines through the RNA-binding protein HuR modulating intestinal epithelial apoptosis. Biochem J 2010; 426: 293-306.
  95. Li L, Rao JN, Bass BL, Wang JY. NF-kappaB activation and susceptibility to apoptosis after polyamine depletion in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2001; 280: G992-G1004.
  96. Marasa BS, Rao JN, Zou T, et al. Induced TRPC1 expression sensitizes intestinal epithelial cells to apoptosis by inhibiting NF-kappaB activation through Ca2+ influx. Biochem J 2006; 397: 77-87.
  97. Keledjian KM, Marasa BS, Wang JY, Rao JN. Induced PDK1 kinase activity suppresses apoptosis in intestinal epithelial cells by activating Akt signaling following polyamine depletion. Int J Clin Exp Med 2012; 5: 221-228.
  98. Arimoto K, Fukuda H, Imajoh-Ohmi S, Saito H, Takekawa M. Formation of stress granules inhibits apoptosis by suppressing stress-responsive MAPK pathways. Nat Cell Biol 2008; 10: 1324-1332.
  99. Zou T, Rao JN, Liu L, et al. Polyamines inhibit the assembly of stress granules in normal intestinal epithelial cells regulating apoptosis. Am J Physiol Cell Physiol 2012; 303: C102-C111.
  100. Simons BD, Clevers H. Stem cell self-renewal in intestinal crypt. Exp Cell Res 2011; 317: 2719-2724.
  101. Takeda N, Jain R, LeBoeuf MR, Wang Q, Lu MM, Epstein JA. Interconversion between intestinal stem cell populations in distinct niches. Science 2011; 334: 1420-1424.
  102. Zuante M, Wang TC. Stem cells in gastroenterology and hepatology. Nat Rev Gastroenterol Hepatol 2009; 6: 724-737.
  103. Kushner JA. Development. Esophageal stem cells, where art thou? Science 2012; 337: 1051-1052.
  104. Doupe DP, Alcolea MP, Roshan A, et al. A single progenitor population switches behavior to maintain and repair esophageal epithelium. Science 2012; 337: 1091-1093.
  105. Hoffmann W. Stem cells, self-renewal and cancer of the gastric epithelium. Curr Med Chem 2012; 19: 5975-5983.
  106. Mills JC, Shivdasani RA. Gastric epithelial stem cells. Gastroenterology 2011; 140: 412-424.
  107. Han ME, Oh SO. Gastric stem cells and gastric cancer stem cells. Anat Cell Biol 2013; 46: 8-18.
  108. Qiao XT, Ziel JW, McKimpson W, et al. Prospective identification of a multilineage progenitor in murine stomach epithelium. Gastroenterology 2007; 133: 1989-1998.
  109. Kim TH, Shivdasani RA. Notch signaling in stomach epithelial stem cell homeostasis. J Exp Med 2011; 208: 677-688.
  110. Yan KS, Chia LA, Li X, et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc Natl Acad Sci USA 2012; 109: 466-471.
  111. Barker N, van Es JH, Kuipers J, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007; 449: 1003-1007.
  112. Tian H, Biehs B, Warming S, et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 2011; 478: 255-259.
  113. Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet 2008; 40: 915-920.
  114. Yui S, Nakamura T, Sato T, et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5(+) stem cell. Nat Med 2012; 18: 618-623.
  115. Zhao T, Goh KJ, Ng HH, Vardy LA. A role for polyamine regulators in ESC self-renewal. Cell Cycle 2012; 11: 4517-4523.
  116. Zhang D, Zhao T, Ang HS, et al. AMD1 is essential for ESC self-renewal and is translationally down-regulated on differentiation to neural precursor cells. Genes Dev 2012; 26: 461-473.
  117. Muscari C, Bonafe F, Carboni M, et al. Difluoromethylornithine stimulates early cardiac commitment of mesenchymal stem cells in a model of mixed culture with cardiomyocytes. J Cell Biochem 2008; 103: 1046-1052.
  118. Guidotti S, Facchini A, Platano D, et al. Enhanced osteoblastogenesis of adipose-derived stem cells on spermine delivery via beta-catenin activation. Stem Cells Dev 2013; 22: 1588-1601.
  119. Szabo S, Deng X, Tolstanova G, et al. Angiogenic and anti-angiogenic therapy for gastrointestinal ulcers: new challenges for rational therapeutic predictions and drug design. Curr Pharm Des 2011; 17: 1633-1642.
  120. Ahluwalia A, Tarnawski AS. Activation of the metabolic sensor-AMP activated protein kinase reverses impairment of angiogenesis in aging myocardial microvascular endothelial cells. Implications for the aging heart. J Physiol Pharmacol 2011; 62: 583-587.
  121. Amagase K, Inaba A, Senta T, et al. Gastric ulcerogenic and healing impairment effects of risedronate, a nitrogen-containing bisphosphonate in rats. Comparison with alendronate and minodronate. J Physiol Pharmacol 2011; 62: 609-618.
  122. Rubinstein E, Cho JY, Rosenthal P, et al. Siglec-F inhibition reduces esophageal eosinophilia and angiogenesis in a mouse model of eosinophilic esophagitis. J Pediatr Gastroenterol Nutr 2011; 53: 409-416.
  123. Pousa ID, Gisbert JP. Gastric angiogenesis and Helicobacter pylori infection. Rev Esp Enferm Dig 2006; 98: 527-541.
  124. Yeo M, Kim DK, Han SU, et al. Novel action of gastric proton pump inhibitor on suppression of Helicobacter pylori induced angiogenesis. Gut 2006; 55: 26-33.
  125. Tuccillo C, Cuomo A, Rocco A, et al. Vascular endothelial growth factor and neo-angiogenesis in H. pylori gastritis in humans. J Pathol 2005; 207: 277-284.
  126. Malara B, Josko J, Tyrpien M, Malara P, Steplewska K. Dynamics of changes in vascular endothelial growth factor (VEGF) expression and angiogenesis in stress-induced gastric ulceration in rats. J Physiol Pharmacol 2005; 56: 259-271.
  127. Kawahara Y, Nakase Y, Isomoto Y, et al. Role of macrophage colony-stimulating factor (M-CSF)-dependent macrophages in gastric ulcer healing in mice. J Physiol Pharmacol 2011; 62: 441-448.
  128. Ganguly K, Sharma AV, Reiter RJ, Swarnakar S. Melatonin promotes angiogenesis during protection and healing of indomethacin-induced gastric ulcer: role of matrix metaloproteinase-2. J Pineal Res 2010; 49: 130-140.
  129. Kang JM, Kim N, Kim B, et al. Enhancement of gastric ulcer healing and angiogenesis by Cochinchina Momordica seed extract in rats. J Korean Med Sci 2010; 25: 875-881.
  130. Dudar GK, D’Andrea LD, Di Stasi R, Pedone C, Wallace JL. A vascular endothelial growth factor mimetic accelerates gastric ulcer healing in an iNOS-dependent manner. Am J Physiol Gastrointest Liver Physiol 2008; 295: G374-G381.
  131. Luo JC, Shin VY, Liu ES, et al. Dexamethasone delays ulcer healing by inhibition of angiogenesis in rat stomachs. Eur J Pharmacol 2004; 485: 275-281.
  132. Deban L, Correale C, Vetrano S, Malesci A, Danese S. Multiple pathogenic roles of microvasculature in inflammatory bowel disease: a Jack of all trades. Am J Pathol 2008; 172: 1457-1466.
  133. Szabo S, Folkman J, Vattay P, Morales RE, Pinkus GS, Kato K. Accelerated healing of duodenal ulcers by oral administration of a mutein of basic fibroblast growth factor in rats. Gastroenterology 1994; 106: 1106-1111.
  134. Szabo S, Vincze A, Sandor Z, et al. Vascular approach to gastroduodenal ulceration: new studies with endothelins and VEGF. Dig Dis Sci 1998; 43: 40S-45S.
  135. Koutroubakis IE, Tsiolakidou G, Karmiris K, Kouroumalis EA. Role of angiogenesis in inflammatory bowel disease. Inflamm Bowel Dis 2006; 12: 515-523.
  136. Zwolinska-Wcislo M, Krzysiek-Maczka G, Ptak-Belowska A, et al. Antibiotic treatment with ampicillin accelerates the healing of colonic damage impaired by aspirin and coxib in the experimental colitis. Importance of intestinal bacteria, colonic microcirculation and proinflammatory cytokines. J Physiol Pharmacol 2011; 62: 357-368.
  137. Paunovic B, Deng X, Khomenko T, et al. Molecular mechanisms of basic fibroblast growth factor effect on healing of ulcerative colitis in rats. J Pharmacol Exp Ther 2011; 339: 430-437.
  138. Deng X, Szabo S, Khomenko T, et al. Novel pharmacologic approaches to the prevention and treatment of ulcerative colitis. Curr Pharm Des 2013; 19: 17-28.
  139. Scaldaferri F, Vetrano S, Sans M, et al. VEGF-A links angiogenesis and inflammation in inflammatory bowel disease pathogenesis. Gastroenterology 2009; 136: 585-595. e5.
  140. Tolstanova G, Khomenko T, Deng X, et al. Neutralizing anti-vascular endothelial growth factor (VEGF) antibody reduces severity of experimental ulcerative colitis in rats: direct evidence for the pathogenic role of VEGF. J Pharmacol Exp Ther 2009; 328: 749-757.
  141. Chidlow JH, Jr., Langston W, Greer JJ, et al. Differential angiogenic regulation of experimental colitis. Am J Pathol 2006; 169: 2014-2030.
  142. Matsuura M, Okazaki K, Nishio A, et al. Therapeutic effects of rectal administration of basic fibroblast growth factor on experimental murine colitis. Gastroenterology 2005; 128: 975-986.
  143. Hindryckx P, Waeytens A, Laukens D, et al. Absence of placental growth factor blocks dextran sodium sulfate-induced colonic mucosal angiogenesis, increases mucosal hypoxia and aggravates acute colonic injury. Lab Invest 2010; 90: 566-576.
  144. Erikson U, Fagerberg S, Krause U, Olding L. Angiographic studies in Crohn’s disease and ulcerative colitis. Am J Roentgenol Radium Ther Nucl Med 1970; 110: 385-392.
  145. Jasnis MA, Klein S, Monte M, Davel L, Sacerdote de Lustig E, Algranati ID. Polyamines prevent DFMO-mediated inhibition of angiogenesis. Cancer Lett 1994; 79: 39-43.
  146. Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 2009; 9: 799-809.
  147. Heazlewood CK, Cook MC, Eri R, et al. Aberrant mucin assembly in mice causes endoplasmic reticulum stress and spontaneous inflammation resembling ulcerative colitis. PLoS Med 2008; 5: e54.
  148. Carlsson R, Fandriks L, Jonsson C, Lundell L, Orlando RC. Is the esophageal squamous epithelial barrier function impaired in patients with gastroesophageal reflux disease? Scand J Gastroenterol 1999; 34: 454-458.
  149. Wroblewski LE, Peek RM, Jr. Targeted disruption of the epithelial-barrier by Helicobacter pylori. Cell Commun Signal 2011; 9: 29-39.
  150. Jager S, Stange EF, Wehkamp J. Inflammatory bowel disease: an impaired barrier disease. Langenbecks Arch Surg 2013; 398: 1-12.
  151. Dixon J, Strugala V, Griffin SM, et al. Esophageal mucin: an adherent mucus gel barrier is absent in the normal esophagus but present in columnar-lined Barrett’s esophagus. Am J Gastroenterol 2001; 96: 2575-2583.
  152. Chen X, Oshima T, Tomita T, et al. Acidic bile salts modulate the squamous epithelial barrier function by modulating tight junction proteins. Am J Physiol Gastrointest Liver Physiol 2011; 301: G203-G209.
  153. Henriksnas J, Phillipson M, Storm M, Engstrand L, Soleimani M, Holm L. Impaired mucus-bicarbonate barrier in Helicobacter pylori-infected mice. Am J Physiol Gastrointest Liver Physiol 2006; 291: G396-G403.
  154. Wroblewski LE, Shen L, Ogden S, et al. Helicobacter pylori dysregulation of gastric epithelial tight junctions by urease-mediated myosin II activation. Gastroenterology 2009; 136: 236-246.
  155. Miyazaki T, Murayama Y, Shinomura Y, et al. E-cadherin gene promoter hypermethylation in H. pylori-induced enlarged fold gastritis. Helicobacter 2007; 12: 523-531.
  156. Amieva MR, Vogelmann R, Covacci A, Tompkins LS, Nelson WJ, Falkow S. Disruption of the epithelial apical-junctional complex by Helicobacter pylori CagA. Science 2003; 300: 1430-1434.
  157. McGuckin MA, Eri R, Simms LA, Florin TH, Radford-Smith G. Intestinal barrier dysfunction in inflammatory bowel diseases. Inflamm Bowel Dis 2009; 15: 100-113.
  158. Clayburgh DR, Barrett TA, Tang Y, et al. Epithelial myosin light chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo. J Clin Invest 2005; 115: 2702-2715.
  159. Lin JE, Snook AE, Li P, et al. GUCY2C opposes systemic genotoxic tumorigenesis by regulating AKT-dependent intestinal barrier integrity. PLoS One 2012; 7: e31686.
  160. Chen J, Xiao L, Rao JN, et al. JunD represses transcription and translation of the tight junction protein zona occludens-1 modulating intestinal epithelial barrier function. Mol Biol Cell 2008; 19: 3701-3712.
  161. Yu TX, Wang PY, Rao JN, et al. Chk2-dependent HuR phosphorylation regulates occludin mRNA translation and epithelial barrier function. Nucleic Acids Res 2011; 39: 8472-8487.
  162. Yu TX, Rao JN, Zou T, et al. Competitive binding of CUGBP1 and HuR to occludin mRNA controls its translation and modulates epithelial barrier function. Mol Biol Cell 2013; 24: 85-99.
  163. Guo X, Rao JN, Liu L, et al. Regulation of adherens junctions and epithelial paracellular permeability: a novel function for polyamines. Am J Physiol Cell Physiol 2003; 285: C1174-C1187.
  164. Liu L, Guo X, Rao JN, et al. Polyamines regulate E-cadherin transcription through c-Myc modulating intestinal epithelial barrier function. Am J Physiol Cell Physiol 2009; 296: C801-C810.
  165. Pera M, Grande L, Gelabert M, et al. Epithelial cell hyperproliferation after biliopancreatic reflux into the esophagus of rats. Ann Thorac Surg 1998; 65: 779-786.
  166. Gerner EW, Garewal HS, Emerson SS, Sampliner RE. Gastrointestinal tissue polyamine contents of patients with Barrett’s esophagus treated with alpha-difluoromethylornithine. Cancer Epidemiol Biomarkers Prev 1994; 3: 325-330.
  167. Garewal HS, Sampliner R. Barrett’s esophagus: a model premalignant lesion for adenocarcinoma. Prev Med 1989; 18: 749-756.
  168. Chaturvedi R, Asim M, Barry DP, Frye JW, Casero RA Jr, Wilson KT. Spermine oxidase is a regulator of macrophage host response to Helicobacter pylori: enhancement of antimicrobial nitric oxide generation by depletion of spermine. Amino Acids 2013; Jul 13 [epub ahead of print]
  169. Al-shabanah OA, Raza M. Effect of ulceration on rat gastric tissue polyamine contents in response to different procedures; inhibition of these effects by cimetidine. Pharmacol Res 1999; 40: 75-82.
  170. Brzozowski T, Konturek PC, Konturek SJ, Brzozowska I, Kwiecien S, Hahn EG. Involvement of ornithine decarboxylase and polyamines in epidermal growth factor-induced recovery of gastric mucosa from gastric lesions provoked by stress. Regul Pept 1998; 74: 73-84.
  171. Marcuard SP, Silverman JF, Finley JL, Seidel ER. Ornithine decarboxylase activity during gastric ulcer healing in dogs. Dig Dis Sci 1992; 37: 1015-1019.
  172. Ricci G, Stabellini G, Bersani G, et al. Ornithine decarboxylase in colonic mucosa from patients with moderate or severe Crohn’s disease and ulcerative colitis. Eur J Gastroenterol Hepatol 1999; 11: 903-904.
  173. Pillai RB, Tolia V, Rabah R, Simpson PM, Vijesurier R, Lin CH. Increased colonic ornithine decarboxylase activity in inflammatory bowel disease in children. Dig Dis Sci 1999; 44: 1565-1570.
  174. Pegg AE, Wechter R, Pakala R, Bergeron RJ. Effect of N1,N12-bis(ethyl)spermine and related compounds on growth and polyamine acetylation, content, and excretion in human colon tumor cells. J Biol Chem 1989; 264: 11744-11749.
  175. Gobert AP, Cheng Y, Akhtar M, et al. Protective role of arginase in a mouse model of colitis. J Immunol 2004; 173: 2109-2117.
  176. Weiss TS, Herfarth H, Obermeier F, et al. Intracellular polyamine levels of intestinal epithelial cells in inflammatory bowel disease. Inflamm Bowel Dis 2004; 10: 529-535.
  177. Obayashi M, Matsui-Yuasa I, Matsumoto T, Kitano A, Kobayashi K, Otani S. Polyamine metabolism in colonic mucosa from patients with ulcerative colitis. Am J Gastroenterol 1992; 87: 736-740.
  178. Mossner J, Hammermann R, Racke K. Concomitant down-regulation of L-arginine transport and nitric oxide (NO) synthesis in rat alveolar macrophages by the polyamine spermine. Pulm Pharmacol Ther 2001; 14: 297-305.
  179. Jin HT, Lamsa T, Merentie M, et al. Polyamine levels in the pancreas and the blood change according to the severity of pancreatitis. Pancreatology 2008; 8: 15-24.
  180. Jin HT, Lamsa T, Nordback PH, et al. Association between remote organ injury and tissue polyamine homeostasis in acute experimental pancreatitis - treatment with a polyamine analogue bismethylspermine. Pharmacol Rep 2011; 63: 999-1008.
  181. Jin HT, Raty S, Minkkinen M, et al. Changes in blood polyamine levels in human acute pancreatitis. Scand J Gastroenterol 2009; 44: 1004-1011.
  182. Moulinoux JP, Delamaire D, Beau B, et al. Diagnostic value of erythrocyte-free polyamines and histaminemia in malignant hepatic tumors and in liver cirrhosis. Clin Chim Acta 1985; 145: 77-87.
  183. Stabellini G, Calastrini C, Gagliano N, et al. Polyamine levels and ornithine decarboxylase activity in blood and erythrocytes in human diseases. Int J Clin Pharmacol Res 2003; 23: 17-22.
  184. Porciani S, Lanini A, Balzi M, Faraoni P, Becciolini A. Polyamines as biochemical indicators of radiation injury. Phys Med 2001; 17(Suppl. 1): 187-188.
  185. Poyhonen MJ, Takala JA, Pitkanen O, et al. Urinary excretion of polyamines in patients with surgical and accidental trauma: effect of total parenteral nutrition. Metabolism 1993; 42: 44-51.
  186. Domschke S, Soling HD. Polyamine metabolism in rat liver: effect of starvation and refeeding. Horm Metab Res 1973; 5: 97-101.
  187. Siegler JM, Kazarinoff MN. Effects of acute and chronic protein deprivation on ornithine decarboxylase levels in rat liver and colon. Nutr Cancer 1983; 4: 176-185.
  188. Rozovski SJ, Rosso P, Winick M. Effect of malnutrition and rehabilitation on the metabolism of polyamines in rat liver. J Nutr 1978; 108: 1680-1690.
  189. Smith TK, Hyvonen T, Pajula RL, Eloranta TO. Effect of dietary methionine, arginine and ornithine on the metabolism and accumulation of polyamines, S-adenosylmethionine and macromolecules in rat liver and skeletal muscle. Ann Nutr Metab 1987; 31: 133-145.
  190. Ota DM, Nishioka K, Foulkes M, Grossie VB. Nutritional parameters affecting erythrocyte polyamine levels in cancer patients. J Clin Oncol 1984; 2: 1157-1164.
  191. Loser C, Eisel A, Harms D, Folsch UR. Dietary polyamines are essential luminal growth factors for small intestinal and colonic mucosal growth and development. Gut 1999; 44: 12-16.
  192. Sarhan S, Knodgen B, Seiler N. Polyamine deprivation, malnutrition and tumor growth. Anticancer Res 1992; 12: 457-466.
  193. 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: 133-141.
  194. Raj KP, Zell JA, Rock CL, et al. Role of dietary polyamines in a phase III clinical trial of difluoromethylornithine (DFMO) and sulindac for prevention of sporadic colorectal adenomas. Br J Cancer 2013; 108: 512-518.
R e c e i v e d : October 9, 2013
A c c e p t e d : November 5, 2013
Author’s address: Prof. Cheng-Wei Tang. Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China; e-mail: shcqcdmed@163.com;
Dr. Jaladanki N. Rao, Department of Surgery, University of Maryland School of Medicine, Baltimore VA Medical Center, 10 North Greene Street, Baltimore, Maryland 21201, USA; e-mail: jrao@umaryland.edu