Skip to main content

Research progress on the microbial metabolism and transport of polyamines and their roles in animal gut homeostasis

Abstract

Polyamines (putrescine, spermidine, and spermine) are aliphatic compounds ubiquitous in prokaryotes and eukaryotes. Positively charged polyamines bind to negatively charged macromolecules, such as nucleic acids and acidic phospholipids, and are involved in physiological activities including cell proliferation, differentiation, apoptosis and gene regulation. Intracellular polyamine levels are regulated by biosynthesis, catabolism and transport. Polyamines in the body originate from two primary sources: dietary intake and intestinal microbial metabolism. These polyamines are then transported into the bloodstream, through which they are distributed to various tissues and organs to exert their biological functions. Polyamines synthesized by intestinal microorganisms serve dual critical roles. First, they are essential for maintaining polyamine concentrations within the digestive tract. Second, through transcriptional and post-transcriptional mechanisms, these microbial-derived polyamines modulate the expression of genes governing key processes in intestinal epithelial cells—including proliferation, migration, apoptosis, and cell–cell interactions. Collectively, these regulatory effects help maintain intestinal epithelial homeostasis and ensure the integrity of the gut barrier. In addition, polyamines interact with the gut microbiota to maintain intestinal homeostasis by promoting microbial growth, biofilm formation, swarming, and endocytosis vesicle production, etc. Supplementation with polyamines has been demonstrated to be important in regulating host intestinal microbial composition, enhancing nutrient absorption, and improving metabolism and immunity. In this review, we will focus on recent advances in the study of polyamine metabolism and transport in intestinal microbes and intestinal epithelial cells. We then summarize the scientific understanding of their roles in intestinal homeostasis, exploring the advances in cellular and molecular mechanisms of polyamines and their potential clinical applications, and providing a rationale for polyamine metabolism as an important target for the treatment of intestinal-based diseases.

Graphical Abstract

Introduction

Polyamines, including putrescine and its downstream metabolites spermidine and spermine (Fig. 1), are widely present in microorganisms and higher plant and animal cells. They primarily exist in a bound state with anionic molecules such as DNA, RNA, specific proteins, ATP, and phospholipids. Polyamines regulate gene expression, maintain the stability of biological membranes, and promote the growth of cells and tissues [1,2,3]. Polyamines in animals usually originate from exogenous dietary supplementation, endogenous cellular metabolism and metabolic production by the intestinal microbiota; polyamines in the lumen of the upper part of the small intestine mainly originate from dietary intake, and putrescine and spermidine, which are metabolized by colonic microorganisms in the lower part of the intestinal tract, are considered to be important sources of polyamines [4, 5]. The intracellular polyamine pool is tightly regulated by various mechanisms, including the synthesis of amino acid precursors, cellular uptake mechanisms for acquiring polyamines from diet and gut microbiota, and degradation and excretion processes [6, 7]. Notably, higher levels of polyamines usually accompany rapidly proliferating and developing tissues, and polyamine levels vary in an age-dependent, tissue- and cell-type-specific manner with an overall decreasing trend during the normal life cycle [8,9,10]. The decrease of polyamine levels caused by aging can be remedied by feeding supplementation or regulation of intestinal microbial composition, thereby promoting colonic epithelial proliferation and macrophage differentiation, alleviating intestinal inflammation, and enhancing intestinal barrier. It can also be transferred to the blood through the intestinal epithelium to protect the kidney and liver or enhance the function of other tissues and organs of the host, such as through the induction of autophagy to enhance cognitive ability of the brain, and to improve the function of the heart [11,12,13,14]. However, when the polyamine metabolic pathway is impaired or abnormal, it is usually accompanied by an imbalance of homeostasis and the occurrence of diseases in the animal organism [15]. Therefore, the relative stability of polyamine levels not only plays an important role in maintaining the normal physiological activities of the organism, but also indirectly reflects the health status of the organism, but little is known about the specific mechanisms of polyamines in playing biological roles.

Fig. 1
figure 1

Putrescine, spermidine and spermine chemical structure

This review primarily summarizes recent advances in the study of polyamine metabolism and transport in gut microbiota and cells. It discusses potential metabolic pathways of spermidine in gut microbiota, providing new strategies to increase polyamine levels in the gut. Additionally, we consolidate information on the mechanisms through which polyamines regulate intestinal epithelial cells, alter gut microbiota composition, and maintain gut homeostasis. We also explore the connections between polyamines and nutrient absorption and metabolism.

Source and metabolism of polyamines

Polyamine biosynthesis

Polyamines are present at certain levels in the animal gut and exhibit significant changes before and after feeding. Following ingestion, the concentration of polyamines in the intestinal lumen can rapidly reach several millimolar. Although the diet is the primary source of polyamines in the digestive tract, most of these compounds are absorbed in the upper sections of the digestive tract and are utilized for the growth and development of the organism [16]. The levels of putrescine and spermidine in the colon primarily depend on the gut microbiota, which metabolize precursor amino acids to release bioactive metabolites that regulate the host's overall balance [17]. In summary, besides dietary and host factors, the microbiota are also considered important determinants of polyamine levels in the digestive tract [18, 19]. The production of polyamines in the organism is a complex process. It involves amino acid precursors and intermediate metabolites. Polyamines can be synthesized from scratch by cells or metabolized by gut microbial catabolism. The process starts with the conversion of arginine to urea and L-ornithine by arginase 1 (ARG1). In mammals and fungi, putrescine is generated by ornithine decarboxylase 1 (ODC1), which relies on pyridoxal phosphate (PLP), a rate-limiting factor. The expression of ODC1 is induced in response to various stimuli. Its activity is regulated at the transcriptional, translational, and post-translational levels [20]. To ensure normal levels of polyamines, ODC1 is controlled by ubiquitin-independent proteasomal degradation mechanisms, mainly consisting of antizyme 1 (AZ1) and ODC antizyme inhibitor 1 (AZIN1), which interact with each other thereby releasing ODCs from the ODC-AZs complex and promoting the biosynthesis of spermidine [21]. S-Adenosylmethionine (SAM) produces decarboxylated S-adenosylmethionine (dcSAM) by the action of Adenosylmethionine decarboxylase 1 (AMD). The dcSAM is used as an aminopropyl donor for aminopropylation by spermidine synthase (SRM) and spermine synthase (SMS) to produce spermidine and spermine, and methylthioadenosine (MTA) [22, 23]. The detailed metabolic pathway is illustrated in Fig. 2. Arginine was found to be the major carbon donor for polyamine biosynthesis and glutamine was found to be a minor carbon donor supporting polyamine biosynthesis in T cells by 13C labeling of arginine, glutamine and proline. Therefore, arginine is now considered to be the major pathway for mammalian polyamine synthesis [24].

Fig. 2
figure 2

Polyamine synthesis and catabolism in mammals. The abbreviations used are as follows: ARG1, arginase 1; ODC1, ornithine decarboxylase; SAM, S-adenosylmethionine; AMD, S-adenosylmethionine decarboxylase; dcSAM, decarboxylated s-adenosylmethionine; SRM, spermidine synthase; MTA, methylthioadenosine; SMS, spermine synthase; SMO, spermine oxidase; 3-AAP, 3-acetylaminopropanal; SSAT/SAT1, spermidine/spermine-N1-acetyltransferase; PAO, polyamine oxidases; AOC1, amine oxidase copper-containing1; H2O2, hydrogen peroxide. Black is the anabolism pathway and orange is the catabolism pathway

Polyamine synthesis in microorganisms

The microbiota in the gut, composed of bacteria, archaea, fungi, protozoa, and viruses, is considered the host's “digestive organ”. It is responsible for metabolizing undigested and unabsorbed nutrients in the gut, providing the host with essential nutrients such as vitamins, amino acids, short-chain fatty acids, and other bioactive compounds [25]. Similarly, polyamines can also be produced by gut microbiota. Unlike host polyamine metabolism, microbial polyamine production is a more complex process involving the uptake and export of intermediate products among different bacterial species [26]. Metabolomics studies of murine gut contents indicate that the concentrations of spermidine and putrescine in the intestinal lumen are dependent on the colonic microbiota, whereas spermine levels remain unaffected [18]. The 74 strains of enterobacteria that are associated with biogenic amine metabolism currently isolated from the human gut belong to several genera, including Bifidobacterium, Clostridium, Enterococcus, and Lactobacillus. These strains mainly utilize amino acid decarboxylases, either in a constitutively or inducible manner, to produce polyamines, with arginine decarboxylation being the main pathway for the production of putrescine [27, 28]. Both ornithine decarboxylase and arginine decarboxylase activities have been detected in bacteria and archaea. The expression and activity of these enzymes are regulated to adapt to the polyamine metabolic processes under various physiological conditions [29, 30]. In Escherichia coli, the constitutive speC and inducible speF are two ornithine decarboxylase genes that, along with the operon potE (ornithine-putrescine antiporter), are expressed under acidic conditions and high levels of ornithine [31]. These enzymes catalyze the decarboxylation of ornithine to produce putrescine. Arginine is first metabolized to agmatine by SpeA and SpeB and then converted to putrescine. The produced putrescine is further condensed with decarboxylated S-adenosylmethionine (AdoMet) under the action of SpeE to form spermidine [32]. In Campylobacter jejuni, the biosynthesis of spermidine relies on L-aspartate-β-semialdehyde (ASA). Putrescine is first converted to carboxyspermidine by carboxyspermidine dehydrogenase (CASDH) and then to spermidine by carboxyspermidine decarboxylase (CASDC) [33]. Many bacterial genomes encode homologs of CASDH and CASDC. For instance, among 56 abundant bacterial species within the genus Bacteroides, 20 species contain homologs of CASDC, which are crucial for spermidine biosynthesis. In bacteria and archaea, an arginine decarboxylase (ADC) pathway for polyamine biosynthesis is also present. For example, in Campylobacter jejuni, arginine is converted to agmatine by ADC. Agmatine is then hydrolyzed by agmatine ureohydrolase (AIH) to produce N-carbamoylputrescine [34]. In Bacteroides thetaiotaomicron, an important species within the gut microbiota, N-carbamoylputrescine aminohydrolase (NCPAH) converts N-carbamoylputrescine into putrescine [35]. The detailed metabolic process is illustrated in Fig. 3.

Fig. 3
figure 3

Polyamine biosynthetic and transport pathways in microorganisms. Polyamine biosynthesis and transport pathways previously described in microorganisms are integrated and elucidated. The abbreviations used are as follows: ADI, arginine desimidase; OTC, ornithine transcarbamylase; SpeA, AdiA, ADC, arginine decarboxylase; SpeB, AUH, agmatine ureohydrolase; SpeC, SpeF, ODC, ornithine decarboxylase; AIH, AgDI, agmatine deiminase; NCPAH, N-carbamoylputrescine amidohydrolase; CASDH, carboxyspermidine dehydrogenase; CASDC, carboxyspermidine decarboxylase; MetK, methionine adenosyltransferase; SpeD, S-adenosylmethionine decarboxylase; SpeE, SPDS, Spermidine synthase; AK, aspartokinase; ASD, aspartate-β-decarboxylase; ASA, aspartate-β-semialdehyde; CAPADH, carboxyaminopropylagmatine dehydrogenase; CAPA, carboxyaminopropylagmatine; CAPADC, carboxyaminopropylagmatine decarboxylase; APA, aminopropylagmatine; APAUH, aminopropylagmatine ureohydrolase. PotABCD, spermidine uptake protein; MdtJI, Spermidine output protein; AguD, PuuP, PlaP, Putrescine uptake protein; PotFGHI, CadB, SapBCDF, Putrescine output protein; PotE, Ornithine-putrescine transporter protein; AdiC, Arginine-agmatine transporter protein; AguD, Agmatine uptake protein

The gut microbiota is a complex mixed system, where the synthesis of metabolic products can be carried out either by single cells or through sequential reactions involving different bacterial species. In certain species such as Enterococcus, Streptococcus, Clostridium, and Lactobacillus, spermidine can be synthesized even in the absence of AdoMet decarboxylase (AdoMet DC) and SRM or their homologs. Therefore, polyamine metabolism and transport pathways are likely to span multiple bacterial species. Nakamura et al. [26] used isotope labeling to show that putrescine in the colon is produced by a collective biosynthetic pathway from different bacteria through complex metabolite exchange, thus proving this possibility. It was found that the concentration of putrescine in the supernatant of mixed culture of eight intestinal bacteria was higher than that of single bacterial culture, and a new putrescine synthesis pathway was found in the process of co-metabolism and transport of polyamines by E. coli and Enterococcus faecalis [36, 37]. When the pH decreases, the acid tolerance system of E. coli becomes active. The arginine-agmatine antiporter (Adi C) facilitates the uptake of environmental arginine, which is then converted to agmatine by arginine decarboxylase (Adi A) [38]. E. faecalis utilizes the agmatine-putrescine antiporter (Agu D) to take up agmatine produced by E. coli. Agmatine is hydrolyzed to N-carbamoylputrescine and ammonia by agmatine deiminase (Agu A), and subsequently converted to putrescine by N-carbamoylputrescine amidohydrolase (Agu B). This process not only generates ammonia but also produces ATP and CO2. Additionally, bifidobacteria, which produce acidic substances in the gut, can enhance this pathway of putrescine formation [39]. Jutta Noack et al. [40] fed indigestible polysaccharides such as pectin to rats, which produced large amounts of short-chain fatty acids and lowered intestinal pH, altered gut microbial composition and metabolism, and promoted the synthesis of polyamines by certain microorganisms such as Bacteroides thetaiotaomicron and Fusobacterium varium, confirming that polyamine formation in some bacterial species in the gut is stimulated by the provision of appropriate metabolic substrates.

Microbes play a crucial role in maintaining the homeostasis of polyamine metabolism, and studies on polyamine metabolism in microorganisms have been ongoing. Recently, a novel spermidine biosynthetic pathway known as the CAPA pathway was discovered in the model cyanobacterium Synechocystis sp. PCC 6803. Compared to the classical spermidine synthase-mediated pathway, the CAPA pathway requires less energy and does not involve putrescine. This pathway is widely distributed across 15 bacterial phyla, including Cyanobacteria, Proteobacteria, Firmicutes, and Bacteroidetes, providing new directions for the biosynthesis of polyamines and their derivatives [41]. However, some microbial polyamine anabolic pathways that have not been extensively studied remain unknown, and it is expected that new metabolic pathways will continue to be discovered in the future [32].

Polyamine catabolism in microorganisms and their hosts

The overall levels of polyamines are regulated on one hand by biosynthesis and uptake mechanisms, and on the other hand by catabolism and efflux mechanisms [42]. Polyamine metabolism in mammalian cells occurs mainly by oxidative deamination in the presence of diamine oxidase and polyamine oxidase (PAO). Spermidine/spermine-N1-acetyltransferase (SSAT) is a key enzyme in the catabolism of polyamines present in the cytoplasm and is involved in catalyzing the first reaction in the polyamine degradation and export pathway. In the degradation pathway, acetyl-coenzyme A (CoA) serves as an acetyl donor. Spermidine and spermine are acetylated by SSAT, resulting in acetyl-spermidine and acetyl-spermine. These acetylated compounds can be excreted from the cytoplasm via transporter proteins in the cytoplasmic membrane, or they can be used as substrates for PAO. PAO oxidizes them to produce putrescine and spermidine by removing the acetamidopropanal group, which is then recycled back into the polyamine pool. Additionally, spermine can be directly oxidized to spermidine by spermine oxidase (SMO) [43, 44]. The whole catabolic process not only generating the corresponding polyamines but also produces potentially toxic by-products, such as H2O2 and aldehydes that cause damage to cells, DNA, etc. Consequently, oxidative damage may be exacerbated by increasing the level of H2O2 when polyamine catabolism is abnormal [45]. However, the effects of polyamines on organism health are not always beneficial. In pathological conditions, polyamine metabolic disorders may further accelerate the development of the disease. This dysregulation is common in cancer. The increase of polyamine biosynthesis and transport and the decrease of catabolism, resulting in elevated polyamine level, which in turn support the rapid proliferation of cancer cells. [15]. In Alzheimer's disease patients, higher concentrations of arginine and increased expression of the polyamine catabolic genes SAT1 and SMOX, and decreased expression of the synthetic genes SRM and ODC1 [46]. Similarly in Parkinson's patients, changes in polyamine levels also have been found, the decreased expression of polyamine metabolic enzyme SAT1 led to an increase in polyamine levels, which in turn reduced the cognitive performance of patients with Parkinson 's disease through the NMDAr pathway [47].

Due to the high concentration of polyamines causing damage to cells, the synthesis and metabolic processes synergistically maintain the dynamic stability of polyamines, which is no exception in microorganisms. They also prevent excessive accumulation of polyamines through the acetylation pathway. In E. coli, spermidine acetyltransferase (SAT), encoded by speG, catalyzes the acetylation and inactivation of spermidine. Silencing this gene leads to spermidine accumulation within the cell [48]. In Bacillus subtilis, the non-membrane-bound SSAT protein PaiA, a member of the N-acetyltransferase superfamily, acts as an N1-acetyltransferase for spermidine and spermine, preventing polyamine accumulation and cellular damage. In E. faecalis and Staphylococcus aureus, the SSAT homologs BltD and SpeG acetylate spermidine and spermine, with a preference for spermine [49, 50]. In E. coli, two pathways metabolize putrescine to succinate via the intermediate γ-aminobutyric acid (GABA). These are the Puu pathway and a pathway involving putrescine transaminase (YgjG) and γ-aminobutyraldehyde dehydrogenase (YdcW) [51,52,53]. In the Puu pathway, putrescine is transported into the cell by PuuP [54], and PuuA uses ATP to conjugate glutamate and putrescine, forming γ-glutamylputrescine [55]. This intermediate is oxidized by PuuB to γ-glutamyl-γ-aminobutyraldehyde, which is further oxidized by PuuC to γ-glutamyl-GABA. PuuD hydrolyzes the γ-glutamyl bond, producing GABA and glutamate [56]. GABA is deaminated by PuuE to form succinate semialdehyde, which is then oxidized to succinate by YneI [57]. Another pathway uses γ-aminobutyraldehyde as an intermediate, and putrescine is metabolized to GABA without undergoing γ-glutamylation [58]. Cultures of three strains of Bifidobacterium species (B. breve, B. catenulatum, B. scardovii) found that all of them could take up putrescine from the medium, but no putrescine was detected in the cells. This may be related to homologs of Gab D, Gab T, Pat A, and Pat D, which are involved in the transketolase pathway for putrescine degradation [59]. However, for some microorganisms, which lack both the proteome required for the transaminase pathway and do not possess the γ-glutamylation pathway, there may be an unknown pathway for polyamine degradation [60].

Translocation of polyamines

Polyamine transport systems play a crucial role in determining polyamine homeostasis and distribution. Exogenous transport allows polyamines to enter the blood circulation through intestinal epithelial cells and transport to other tissues and organs to exert their biological functions. When radioactive-labeled polyamines were orally administered to rats, they were quickly absorbed but unevenly distributed across different tissues. Further studies revealed that polyamines preferentially accumulate in rapidly proliferating tissues [61]. Polyamines produced by gut microbiota are typically present in the colon lumen and the downstream small intestine. They enter the host organism through the colonic epithelial cells and are eventually transported to the proximal intestine via the portal vein circulation and the biliary system. This process is crucial for maintaining the polyamine pool and overall host health [13, 62]. The cellular uptake and efflux of polyamines are attributed to two different transporter systems and involve different carriers. The intensity of uptake is influenced by the cell's own polyamine demand. Cells with high proliferative activity and those experiencing polyamine depletion due to blocked polyamine synthesis pathways have a greater capacity for polyamine uptake [6]. Since the primary and secondary amino groups of polyamines are protonated outside the cell, they cannot cross the cell membrane through passive diffusion [63]. Therefore, there are different hypotheses regarding the transport of polyamines, including plasma transport, vesicle isolation, glycosaminoglycan-mediated endocytosis, and caveolin-mediated endocytosis [64]. Homology analysis of polyamine biosynthesis proteins has shown that some bacteria lack complete polyamine synthesis pathways and must import polyamines from their environment to support growth and adapt to environmental changes. For bacteria capable of synthesizing polyamines, uptake and efflux not only help maintain polyamine homeostasis but also offer convenience. Because polyamines are hydrophilic and positively charged, they cannot cross the hydrophobic cell membrane without specific transporters, making polyamine transport proteins essential for cells [41].

So far, five transport systems have been identified in E. coli. These include two polyamine uptake mechanisms, both belonging to the ATP-binding cassette (ABC) protein family. One system, which preference spermidine, consisting of PotA (ATPase), PotB and PotC (channel proteins), and PotD (substrate-binding protein). This system not only takes up spermidine but also has a low affinity for putrescine [65]. In E. coli, these four proteins are indispensable for the process of spermidine uptake [66]. The spermidine transporter encoded by PotABCD has also been found in E. faecalis and Staphylococcus aureus, compensating for their inability to synthesize polyamines, supporting normal cell growth and biofilm formation. Another system is specific for putrescine uptake and includes PotF (substrate-binding protein), PotG (ATPase), PotH and PotI (channel proteins) [67, 68]. In addition, the proton-dependent putrescine uptake protein PuuP and its homolog PlaP, which plays an important role when E. coli grows with putrescine as the sole carbon or nitrogen source. Unlike PotFGHI, PuuP is not inhibited by feedback inhibition from intracellular polyamines, but is inhibited by glucose [53, 54, 69]. There are also two polyamine-amino acid reverse transporter proteins: PotE (putrescine-ornithine) and CadB (putrescine-lysine). Both are proton-dependent putrescine uptake proteins that function in a neutral environment. SpeF converts ornithine and lysine to putrescine and cadaverine, respectively, by consuming a proton under acidic conditions. PotE exports putrescine through uptake of ornithine, while CadB outputs putrescine through uptake of lysine. This transport system is important for the acclimation of E. coli to acidic environment and for normal growth [70, 71]. Furthermore, the spermidine transporter protein (MdtJI) exports excess spermidine from E. coli cells, maintaining intracellular spermidine levels within a normal range [72]. Lastly, SapBCDF, another ABC transporter, exports putrescine from the cell [73]. A new polyamine transporter may exist during the collaboration of the two acid tolerance mechanisms in the co-culture of E. faecalis and E. coli. This collaboration induces putrescine synthesis, with E. faecalis utilizing the agmatine-putrescine transporter (AguD) to take up agmatine exocytosed by E. coli, and agmatine is converted to putrescine and then excreted extracellularly via AguD [36].

BLAST analysis revealed that proteins similar to PotD and PotF are distributed across several phyla, including Proteobacteria, Firmicutes, Actinobacteria, Fusobacteria, Cyanobacteria, Spirochaetes, Planctomycetes, Chlamydiae and Deinococci. Proteins with high similarity to PotE and CadB are found in Proteobacteria, Bacteroidetes, Firmicutes, and Actinobacteria, while those similar to PuuP and PlaP are limited to Proteobacteria, Firmicutes, Actinobacteria, Acidobacteria, and Sphingobacteria [74]. Apart from E. coli, new transporters have been identified in other bacteria. For instance, Blt is a spermidine exporter in Bacillus subtilis [75]. There are three PotD homologs NspS, PotD1 and PotD2 in Vibrio cholerae, among which NspS acts as a signal sensor to promote polyamine transport [76, 77], and four polyamine transport proteins DUR3, SAM3, GAP1 and AGP2 have been discovered in Saccharomyces cerevisiae, with DUR3 and SAM3 playing major roles in polyamine uptake. Additionally, five polyamine export transporters (TPO1-5) maintain intracellular polyamine stability [78, 79]. Cultivation of 13 human indigenous Bifidobacteria revealed that 10 species possess polyamine transport capabilities. BLAST analysis suggested the potential existence of novel polyamine biosynthesis and transport proteins [60]. Polyamines are endogenous active substances with multi-target regulation characteristics. Identification of these transporters can more comprehensively elucidate their transport processes and physiological mechanisms.

Physiological functions and mechanisms of action of polyamines on microorganisms

The role of polyamines in cell proliferation and differentiation

Putrescine and spermidine are common polyamines in bacteria. They are not only involved in core physiological processes such as gene expression and cell growth, but also affect iron carrier biosynthesis, endocytosed vesicle production, swarming motility, and formation of biofilms [80, 81]. Microbial reverse genetics studies have shown that gene deletion or polyamine depletion in polyamine metabolic pathways has a negative impact on cell survival and proliferation. Intracellular polyamines exist mainly in the form of polyamine-RNA complexes. For instance, in E. coli, 90% of spermidine is present in the cellular RNA complex, which maintains the RNA in a specific conformation and solubility, and is able to stabilise the RNA by interacting with other molecules (such as Mg2+). Polyamines can also bind to the outside of the DNA, allowing for intermolecular interactions to stabilise the double-stranded DNA [82]. Eukaryotic and archaeal use polyamines as aminobutyl donors, which are transferred to the ε-amino group of specific lysine residues of eIF5A under the action of deoxyhypusine synthase (DHS). The resulting deoxyhypusine lysine residues are then catalyzed by deoxyhypusine hydroxylase (DHH) to produce hypusine lysine [83]. All archaea encode DHS homologs, and inhibition of these homologs causes cell cycle arrest, indicating that polyamines or their precursors are essential for the normal growth of archaea. In eIF5A, lysine is modified by hydroxyputrescine, which activates eIF5A. This activation helps eIF5A bind to translationally active ribosomes, increasing the ratio of polyribosomes to monoribosomes. eIF5A plays a role in the elongation phase of translation. The level of polyamine content influences the degree of hydroxyputrescine modification of eIF5A, which in turn affects eIF5A function and cell proliferation [84,85,86]. Polyamine also regulate fungal cell differentiation. In some fungi, polyamines participate in spore germination, forming cell morphologies more resistant to adverse conditions than vegetative cells [87]. In conditionally pathogenic fungi, it was found that the heat-induced dimorphism conversion process of Emergomyces africanus and the conversion of Candida albicans between yeast state and hyphal state required the participation of polyamines [88, 89]. Additionally, spermidine is crucial for the normal growth of Campylobacter jejuni and Pseudomonas aeruginosa [34, 90], and polyamines are essential for the growth of Bacillus subtilis and Streptococcus pneumoniae [27, 91]. Putrescine is also indispensable for the growth of Ralstonia solanacearum [92].

The role of polyamines in physiological stress responses

Physiological stress responses induced by oxidative stress, temperature changes, nitrosative stimuli, or other toxic compounds are present throughout the microbial life cycle. Polyamines provide resistance to both intracellular and environmental stresses. Intracellular polyamine levels changing in response to stress, and depletion of polyamines make cells more sensitive to stress. Reactive oxygen species (ROS) can cause DNA double-strand breaks and structural changes, damaging macromolecules within the cell. ROS levels increase as a byproduct of metabolism with increased metabolic rates. Superoxide dismutase (SOD) protects intracellular nucleic acids from damage caused by superoxide ions and other oxidants. Spermidine has been shown to be effective against alkyl, hydroxyl, and peroxy radicals, acting as a radical scavenger in conjunction with SOD to reduce oxidative damage [87]. In Salmonella Typhimurium, spermidine activates the stress response mechanism by regulating key antioxidant genes to counter ROS-mediated cytotoxicity and improve its survival in macrophages [93]. Polyamines also regulate the expression of stress response genes, mediating E. coli adapt to nitrative stress in the external environment. The cadC is an essential gene for nitrate stress tolerance in E. coli, its deletion leads to significantly reduced intracellular polyamine levels and increased sensitivity to acidified nitrite [94]. In fission yeast Schizosaccharomyces pombe, polyamine transporter Shp2 facilitates phosphate export in an Xpr1-independent manner and contributes to high phosphate tolerance [95]. The CadBA system, activated to counteract acidic stress, is regulated by the pH sensor CadC, which controls the expression of the CadBA operon. Although polyamines were not explicitly mentioned in this process, CadB functions as a lysine-cadaverine antiporter, so polyamines were likely involved in acid tolerance regulation [96, 97]. Engineered Saccharomyces cerevisiae strains with high spermidine levels exhibit resistance to chemicals like acetic acid and furfural, which inhibit microbial growth, metabolism, and ethanol fermentation [98]. Moreover, the survival of gut microbiota under acidic conditions depends on polyamines. In E. coli, polyamines reduce cAMP levels, thereby regulating the expression of the gadA and gadB, which influence the bacterium's sensitivity to acidic environments [99].

The impact of polyamines on microbial biofilms

Polyamines are closely related to the motility and attachment of bacteria, the formation and function of biofilm [100]. Biofilms are aggregates of bacteria or other microbial communities in the extracellular matrix, including polysaccharides, proteins, extracellular nucleic acids and lipids. These membranes can form on living and non-living surfaces in response to specific environmental stimuli, thereby protecting the cell from the extracellular environment [101]. The permeability of the outer membrane of Gram-negative bacteria to hydrophilic compounds mainly depends on porins, which are homotrimeric transmembrane proteins. In E. coli, the porins OmpC and OmpF interact with polyamines in a voltage-dependent manner. Putrescine and spermidine bind to aspartate residues on OmpC and OmpF, altering their charge and pore size, leading to channel closure and reduced outer membrane permeability. Therefore, polyamines regulate bacterial outer membrane permeability [102, 103]. Disruption of the spermidine synthase gene (speE) in E. coli results in severe biofilm formation defects, which is restored by supplementation with spermidine, and biofilm formation is further enhanced by intracellular accumulation of spermidine [104], the absence of potABCD operon also impaired the biofilm formation of Streptococcus pneumoniae, and the addition of more polyamines to the medium stimulated the biofilm formation [105]. In Yersinia pestis, biofilm formation is positively correlated with exogenous putrescine levels but not with spermidine, possibly due to polyamine uptake proteins; putrescine exerts its effects after entering the cell and being converted [106]. Similarly, spermidine promotes biofilm formation in Bacillus subtilis by regulating the transcription of the extracellular polysaccharide and TasA operons through the factor slrR [107]. However, in Vibrio cholerae, spermidine uptake and extracellular spermidine inhibit biofilm formation through the NspS/MbaA signaling pathway [76]. Finally, putrescine mediates biofilm matrix degradation in Shewanella oneidensis, and blocking the putrescine biosynthetic pathway enhances biofilm cohesiveness and performance [108].

The impact of polyamines on gut microbiota

Polyamines are not only metabolically produced by gut microbes but also influence microbial growth and colonization. The animal digestive tract hosts a large microbial community, where polyamines interact with the gut flora to maintain host health and improve metabolic phenotypes in the gut. There are two possible mechanisms for the effect of polyamines on the intestinal microbiota. On the one hand, polyamines serve as growth factors and preferred substrates for gut microbes. On the other hand, polyamines act as bioactive modulators, regulating probiotic colonization patterns and inhibiting harmful bacterial growth. Additionally, polyamines can also change the pH by increasing the volatile fatty acids in the intestine [109]. These findings provide new insights into the regulation of intestinal microbial growth and colonization by polyamines. Feeding neonatal mice with polyamine-rich infant formula showed higher levels of Bifidobacterium, Akkermansia-like bacteria, and Lactobacillus-Enterococcus group, promoting a healthy mucosal status, confirming that polyamines can modulate microbial colonization patterns in the gut [110]. Alistipes and Turicibacter are considered pathogenic bacteria associated with colitis, and spermidine supplementation significantly reduced their abundance in the gut of mice while increasing Lactobacillus levels. However, it also decreased beneficial bacteria such as Odoribacter and Romboutsia [111]. Bacteroides and Parabacteroides increased, while Prevotella and Desulfovibrionaceae decreased in the intestine of abdominal aortic aneurysm (AAA) mouse model. Exogenous supplementation of spermidine alleviated the imbalance of intestinal flora and helped prevent the development of AAA [112]. After piglets were fed a diet containing spermine, the number of Lactobacilli, Bifidobacteria and total bacteria in colon and cecal chyme increased, while the number of E. coli decreased [113]. Similarly, polyamines improve gut microbiota structure and health in poultry [114]. Although many studies suggest that polyamines influence the composition and diversity of gut microbiota (Table 1), the interaction between the two remains unclear.

Table 1 Effects of exogenous polyamines on gut and gut microbiota in different animal models

Polyamines and intestinal homeostasis

Polyamines regulate the proliferation of intestinal epithelial cell layers

The intestinal barrier system consists of the mucus layer, intestinal epithelial cells (IECs), tight junctions (TJs), immune cells, and gut microbiota. The intestinal epithelium forms the largest mucosal surface in the body, composed of a single layer of cells, including crypts and villi [124]. It forms a crucial barrier to protect the body from pathogenic microorganisms, viruses, and various harmful substances [125]. Among these components, IECs are important players in intestinal homeostasis, and multifunctional intestinal epithelial stem cells located in the crypts are constantly proliferating and replicating [126]. The integrity and efficacy of the intestinal barrier depends on the dynamic balance between apoptosis, proliferation, migration, differentiation, and inter-cellular interactions of IECs [127]. The supply of polyamines to dividing cells in the crypt is essential for normal intestinal epithelial renewal and repair of damaged mucosa [128, 129]. Polyamine levels are also biomarkers in the proliferation process of intestinal mucosa, with high levels of spermine and spermidine enhancing intestinal immune barrier function by reducing intestinal mucosal permeability [130]. When cells are stimulated to grow and divide, intracellular polyamine levels rapidly increase. Inhibiting ODC activity reduces polyamine levels in tissues, inducing intestinal mucosal atrophy and hindering intestinal epithelial renewal [131]. Oral administration of polyamines promotes intestinal epithelial cell proliferation and enhances mucosal repair after injury [132].

The regulation of polyamines is a central convergence point for multiple signaling pathways that drive various epithelial cell functions, controlling the expression of genes related to proliferation, arrest, and apoptosis. This includes the regulation of numerous growth-promoting proteins such as c-Myc, c-Fos, and c-Jun, as well as growth-inhibiting factors like p35, nucleophosmin (NPM), JunD, TGF-β, TGF-β receptors, and Smads [133,134,135,136,137]. When the polyamine level in the cells increases, the expression of growth-promoting genes is increased by activating the transcription of genes, while the expression of growth-inhibiting genes is inhibited. ODC overexpression increases the polyamine levels in IECs, stimulates the expression of growth-promoting protein genes, and accelerates the transition from the G1 to the S phase of the cell cycle, contributing to the stimulation of IECs proliferation. In contrast, a decrease in polyamine increases the level of inhibitory factors, leading to growth arrest [138]. In rat intestinal mucosal injury models, the stimulation of mucosal injury repair is accompanied by increased polyamine levels and enhanced expression of c-Myc, c-Fos, and c-Jun genes. By inhibiting ODC with DFMO to reduce polyamine levels, the expression levels of these genes also decrease, delaying the healing of damaged intestinal mucosa [139, 140]. As polyamine levels decrease, the levels of growth-inhibitory factors such as TGF-β and Smads in intestinal epithelium significantly increase [141]. In addition, polyamines can regulate the apoptosis of IECs through Akt kinase, ATF-2, XIAP and NF-κB signaling pathways [142,143,144,145].

Polyamines are not only involved in the process of gene transcription, but also influence the transport, stability, and translation of post-transcriptional mRNA. RNA-binding proteins (RBPs) and miRNAs are crucial in this process. AU-rich element binding proteins (ARE-binding proteins, AUBPs) are significant members of the RBP family. They bind to AREs in the 3'untranslated region (3'UTR) of various mRNAs and influence their stability. They also regulate translation and mRNA export, controlling the expression of multiple important proteins involved in cellular functions [146, 147]. For instance, AUBPs regulate the mRNAs of various inflammatory cytokines to modulate the inflammatory response, these AUBPs include AU-rich element-RNA binding factor 1 (AUF1) and Human antigen R (HuR) [148]. JunD mRNA is the common target of HuR and AUF1, and there is a competitive relationship between them. The decrease of polyamine level in cells can enhance the binding of HuR to JunD mRNA and reduce the transcription level of JunD related to AUF1, thus stabilizing JunD mRNA. Conversely, increased cellular polyamines inhibited the interaction of JunD mRNA with HuR and enhanced its binding to AUF1, resulting in the inhibition of JunD expression [149]. HuR can regulate the stability and translation of multiple mRNAs within cells [150], thus HuR is generally considered a post-transcriptional enhancer of biological processes in intestinal epithelial homeostasis. Knocking out HuR in mouse intestinal epithelial tissue inhibits epithelial cell renewal and delays mucosal repair following injury [151]. Although reduced polyamine levels do not alter the total amount of HuR in cells, they affect its transport between the nucleus and cytoplasm, leading to increased accumulation of HuR in the cytoplasm [135]. Adenosine 5'-monophosphate-activated protein kinase (AMPK) is a protein kinase that regulates metabolism and energy stability. Polyamine-mediated AMPK activation in IECs can regulate the phosphorylation and acetylation of Impα1 to regulate the subcellular localization of HuR. The decrease of polyamine content leads to the inactivation of AMPK-involved Impα1 pathway and the accumulation of HuR in the cytoplasm [152]. MEK-1 is a signal transduction enzyme that plays a role in cell function. Polyamines in IECs reduce the stability of MEK-1 mRNA and inhibit its translation process by inhibiting the binding of HuR to MEK-1 transcripts [153]. Furthermore, studies have shown that polyamines can regulate the translation of c-Myc in IECs through Chk2-mediated phosphorylation of HuR [154].

Polyamines regulate connections between intestinal epithelial cells

Differentiated IECs forms a whole through protein complex connection, establishes a selective permeability barrier, prevents the random diffusion of substances and maintains its own polarity. It is an important part of the intestinal mechanical barrier, and ultimately maintains the integrity of intestinal epithelial cells and reduces the occurrence of intestinal diseases [155]. The connection between IECs include TJs, adhesive junctions (AJs) and desmosomes. Among them, TJs are the most critical type of connection between intestinal cells. TJ proteins are essential for influencing IECs function and determining the defensive capability of the intestinal mucosal barrier, including transmembrane protein Claudin, Occludin and cytoplasmic scaffold protein Zonula Occludens (ZO) family [156, 157]. Research has shown that polyamines regulate the synthesis and stability of these junction proteins. Inhibiting polyamine synthesis by DFMO decreases Occludin protein levels, but does not affect its mRNA expression. The contents of ZO-1, ZO-2, Claudin-2 and Claudin-3 decrease in polyamine-deficient cells [158]. Another study indicated that HuR binds to the 3'UTR of occludin mRNA, enhancing its translation. However, this binding depends on Chk2-dependent phosphorylation of HuR. As polyamine levels decrease, so does Chk2, thereby affecting the translation of occludin mRNA [159]. Below the TJs are AJs, which are rich in cadherins and mediate strong intercellular adhesion, playing a significant role in the formation and regulation of the epithelial barrier. Polyamines regulate E-cadherin expression at the transcriptional level through Ca2+ and the transcription factor c-Myc, promoting the function of the intestinal epithelial barrier and helps maintain the integrity of the intestinal mucosa [160, 161]. Connexin 43 (Cx43), a gap junction protein, is essential for intercellular communication and nutrient diffusion. HuR directly binds to Cx43 mRNA through its 3'UTR in the IECs, stabilizing Cx43 mRNA and enhancing its translation. This promotes the function of Cx43-mediated gap junctions, which play a key role in regulating intestinal epithelial barriers [162]. In IECs, polyamines alter the ratio of stromal interaction molecule 1 to stromal interaction molecule 2 (STIM1/2), controlling transient receptor potential channel 1 (TRPC1)-mediated Ca2+ signaling and influencing cell migration for epithelial remodeling after injury [163]. Additionally, intracellular Ca2+ concentration and membrane potential are affected by the expression of the voltage-gated potassium channel (Kv1.1), which is regulated by polyamines [164]. Wild-type polyamine-producing E. coli and mutant strains missing the polyamine synthesis gene were colonized in the mouse intestine, and polyamines produced by E. coli promoted the proliferation of colonic epithelial cells and macrophages, and reduce the risk of colitis in mice [14]. Given the significant role of polyamines in maintaining intestinal homeostasis, they also show potential in promoting the growth and development of gastrointestinal and colonic mucosa in newborn mammals, thereby reducing intestinal diseases [165].

Polyamines enhance intestinal antioxidant damage

The stability of the intestine also depends on the antioxidant capacity and autophagy induction of polyamines. Polyamines improve the antioxidant defense of the intestine by increasing free radical scavenging capacity and enzymatic and non-enzymatic antioxidant capacity, and alleviate the intestinal oxidative damage caused by weaning stress [121]. Glutathione-S-transferase (GST) is a detoxifying enzyme that protects cells by clearing toxic substances. Glutathione (GSH) is a non-enzymatic antioxidant that can conjugate with H2O2 and lipid hydroperoxides, while one of the primary functions of GST is to promote the reaction between GSH and various endogenous and exogenous electrophilic compounds, producing less toxic or non-toxic substances. Exogenous spermidine intake can increase GST activity and elevate GSH levels in the intestine, mitigating oxidative damage [113]. In an acute colitis mouse model induced by dextran sulfate sodium (DSS), spermidine reduces intestinal inflammation by promoting anti-inflammatory macrophages, maintaining a healthy microbiome, and preserving epithelial barrier integrity in a protein tyrosine phosphatase non-receptor type 2 (PTPN2)-dependent manner [118]. Another study showed that the occurrence of colitis was accompanied by a decrease in SMO content in tissues and was negatively correlated with the severity of the disease. After treatment with spermidine, the symptoms of colitis were significantly improved and the development of tumors was inhibited [119]. In conclusion, current research has revealed that polyamines maintain intestinal homeostasis through various mechanisms, ensuring intestinal health and aiding in the efficient digestion and absorption of nutrients.

Polyamines regulation of nutrient absorption and metabolism

The unique physiological functions and widespread presence of polyamines determine their crucial role in regulating glucose, lipid, and energy homeostasis. Moreover, the acetylation reactions involved in polyamine catabolism require CoA consumption, linking polyamine catabolism with lipid oxidation, energy expenditure, and glucose metabolism. Polyamines have been shown to have an effect on the regulation of metabolic disorders and energy homeostasis in different mouse models [166, 167]. In addition, they are involved in metabolic processes in rat blood, including cell membrane metabolism, lipid metabolism, glucose metabolism, and amino acid metabolism [168].

Polyamines exert significant physiological effects on lipid metabolism, particularly in the differentiation of preadipocytes into adipocytes. The 3T3-L1 cell line is a widely used model for studying adipose metabolism, and spermidine is essential for adipogenic differentiation in 3T3-L1 fibroblasts [169]. Spermidine regulates the fat metabolism process by upregulating fibroblast growth factor 21 (FGF21) signaling and its downstream PI3K/AKT and AMPK pathways [170]. Another study found that exogenous putrescine promoted adipogenic differentiation, increased intracellular lipid accumulation and the expression of adipogenic genes in 3T3-L1 preadipocytes [171]. In contrast, supplementing with spermine inhibited the expression of CCAAT enhancer binding protein α (C/EBPα) mRNA, affecting the adipocyte differentiation process and inhibiting lipid accumulation [172]. Furthermore, spermine regulated the differentiation of adipose tissue-derived mesenchymal stem cells into osteoblasts while inhibiting adipogenesis [173]. Since natural polyamines can interconvert intracellularly, altering the intracellular ratio of spermidine to spermine also affects adipocyte differentiation [174]. When polyamines are depleted in cells, the structure and function of mitochondria are greatly affected, and the decrease of mitochondrial fatty acid oxidation leads to lipid accumulation [175]. Oral administration of spermine to mice fed with high-fat diet found to alleviate the increase in body weight, blood triglycerides and visceral fat caused by high-fat diet [172]. Interestingly, oral administration of spermidine in obese mice also showed a similar effect. The average diameter of visceral fat and adipocytes decreased, and it had higher glucose tolerance and insulin sensitivity. Unlike spermine, spermidine, as an autophagy inducer, plays an important role in alleviating weight gain caused by high-fat diet [176]. Additionally, adding spermidine in an atherosclerotic mouse model inhibited lipid accumulation and necrotic core formation by stimulating cholesterol efflux [177]. Studies on the mechanisms by which polyamines regulate lipid metabolism have shown that the activity of key enzymes in the polyamine metabolic pathway, such as SSAT, ODC, SRM, and SMS, also affects lipid metabolism. Polyamines regulate lipid metabolism through involvement in pathways such as AMPK, mammalian target of rapamycin (mTOR), and C/EBP [178,179,180,181,182].

There is a close relationship between lipid metabolism and glucose metabolism. Reports on the regulation of glucose by polyamines can be traced back to the study of brush border membrane vesicles ( BBMV) at the end of the 20th century. Intestinal luminal polyamines affect the number of glucose carriers in the cell membrane and also increase the affinity of carriers for glucose through direct interactions with vesicular membranes, thereby facilitating glucose uptake and determining the maximal rate of glucose transport [183]. Further investigation has revealed that polyamines enhance glucose absorption in the small intestine by rapidly increasing the glucose transporter protein SGLT1 on the brush border membrane [184]. Oral administration of polyamines to obese mice has shown that spermidine intake is negatively correlated with obesity, significantly improving diet-induced insulin resistance, enhancing glucose utilization, and effectively ameliorating obesity and glycemic status [185, 186]. In the rat small intestine putrescine can also be converted to succinic acid as a source of energy [187]. In addition, polyamines are involved in pancreatic development and regulation of β-cell function, and in human serum spermidine levels are negatively correlated with the triglyceride-glucose index [188, 189]. In polyamine depletion-mediated Warburg-like effects, cells shift from aerobic respiration to glycolysis, leading to an alteration in metabolic pathways. In summary, polyamines play a critical role in maintaining normal energy demands and vital activities by regulating mitochondrial function, glucose and lipid metabolism, and cellular energetics [175].

The impact of polyamines as health modulators on animal production performance and their application prospects

Polyamines, a class of endogenous bioactive molecules with multi-target regulatory properties, play indispensable roles in maintaining cellular homeostasis in mammals. Depletion of polyamines has been shown to disrupt normal cell growth and proliferation. Substantial experimental evidence demonstrates that exogenous polyamine supplementation confers multiple physiological benefits, including improved cardiovascular health, enhanced neuroprotection, immune regulation, tumor suppression, mitigation of inflammatory damage, and increased stress tolerance, while exhibiting potential to delay organismal aging and extend lifespan [9, 190]. Notably, spermidine has been ranked among the top eight most promising anti-aging substances due to its biological efficacy [191].

As the most abundant polyamine in mammals, spermidine promotes ovarian follicular development and oocyte maturation in aged individuals through autophagy activation and mitochondrial function enhancement, thereby improving fertility and litter size [192,193,194]. Furthermore, spermidine and spermine alleviate lipopolysaccharide (LPS)-induced mitochondrial dysfunction and apoptosis in sperm via a CK2-dependent mechanism, significantly improving sperm quality and prolonging the preservation efficiency of porcine semen [195]. In ruminants, polyamines are critical for establishment and maintenance of pregnancy during the peri-implantation period of gestation [196]. These findings suggest that polyamines may serve as novel regulators for reproductive health optimization. Polyamines also enhance intestinal barrier integrity by upregulating tight junction proteins, reducing gut permeability and mucosal atrophy in piglets. Their capacity to stimulate intestinal stem cell proliferation significantly increases the villus height/crypt depth ratio, which correlates with decreased diarrhea incidence and enhanced immune function [115, 116]. From a systems biology perspective, dietary polyamine intake activates systemic cellular metabolic reprogramming pathways, exerting anti-aging effects [197] while improving nutrient absorption and whole-body metabolic efficiency (Fig. 4) [198, 199].

Fig. 4
figure 4

Effects of polyamines on mammalian health

Although polyamines have demonstrated significant biological benefits in foundational studies, their application in livestock production remains underexplored. Future translational opportunities may involve their development as functional feed additives or therapeutic agents to enhance overall health. However, critical challenges must be addressed, including elucidation of dose-response relationships and mechanistic pathways governing polyamine-mediated health regulation.

Conclusion

In this review, we summarize the complex and diverse functions of polyamines in cells. Current evidence indicates that polyamines promote anti-aging processes, enhance stress adaptation, mitigate intestinal damage, and exhibit dual roles in both anti-inflammatory and antioxidant responses. While polyamines negative effects on pathologically relevant diseases such as cancer, Alzheimer's disease, and Parkinson's disease. We don't discuss much about the detrimental effects of polyamines in this article. Dietary intake remains the main source of polyamines and cellular polyamine pools are tightly regulated by synthesis, degradation, uptake and efflux. Notably, gut microbiota emerge as key contributors to maintaining intestinal spermidine levels. Therefore, our discussion has focused on polyamine metabolic pathways and their interplay between microbial communities and the host. However, key microbial components governing polyamine dynamics—including transporters for uptake/efflux, utilization pathways, and biosynthetic modules—remain poorly characterized. Specifically, systematic identification of genes regulating microbial polyamine metabolism and high-throughput screening for high-efficiency probiotic strains capable of polyamine synthesis represent critical research priorities. Further elucidating of how living cells maintain polyamine homeostasis through precise regulation of their biosynthesis, interconversion, catabolism, and conjugation is essential to achieve optimal bioactive concentrations. Such mechanistic insights are critical for understanding how polyamines modulate specific biological processes under both physiological and stress conditions, thereby advancing our comprehensive knowledge of their multifunctional roles across diverse biological systems.

Intestinal health is crucial for the organism and is the basis for the digestion and absorption of nutrients and for ensuring normal life activities of the organism, and its homeostasis is maintained through a variety of mechanisms. Polyamines have a wide range of cellular functions in the intestinal epithelium and are involved in a variety of physiological and pathological processes. Here we emphasize the role of polyamines as active small molecules in maintaining the integrity of the intestinal epithelium as well as in the regulation of lipid, amino acid and glucose metabolism. The growth of the intestinal mucosa is dependent on the efficient supply of polyamines to dividing cells in the crypts. There is growing evidence that polyamines are involved in cell growth, differentiation and apoptosis by regulating gene expression, and show great potential in attenuating oxidative damage, increasing antioxidant status and digestive enzyme activity, and regulating intestinal flora. Although exogenous polyamines are effectively alleviate intestinal inflammation and restore barrier function, their clinical translation remains challenging. With the rapid development of polyamine biology, polyamine metabolism and transport have been used as important targets for the treatment of intestinal disorders and the improvement of body health. However, polyamines are widely used as functional substances, and a lot of work still needs to be done in the future.

Data availability

Not applicable.

Abbreviations

ADC:

Arginine decarboxylase

Adi A:

Acid-induced arginine decarboxylase

Adi C:

Arginine-agmatine antiporter

AdoMet:

S-adenosylmethionine

AdoMet DC:

AdoMet decarboxylase

ARG1:

Arginase 1

Agu A:

Agmatine deiminase

Agu B:

N-carbamoylputrescine amidohydrolase

Agu D:

Agmatine-putrescine transporter

AIH:

Agmatine ureohydrolase

AJs:

Adhesive junctions

AMD:

Adenosylmethionine decarboxylase

AMPK:

Adenosine 5'-monophosphate-activated protein kinase

ASA:

L-aspartate-β-semialdehyde

AUBPs:

AU-rich element binding proteins

AUF1:

AU-rich element-RNA binding factor 1

AZ1:

Antizyme 1

AZIN1:

Antizyme inhibitor 1

CASDC:

Carboxyspermidine decarboxylase

CASDH:

Carboxyspermidine dehydrogenase

CoA:

Acetyl-coenzyme A

Cx43:

Connexin 43

C/EBPα:

CCAAT enhancer binding protein α

dcSAM:

Decarboxylated S-adenosylmethionine

DHH:

Deoxyhypusine hydroxylase

DHS:

Deoxyhypusine synthase

DSS:

Dextran sulfate sodium

FGF21:

Fibroblast growth factor 21

GABA:

γ-Aminobutyric acid

GSH:

Glutathione

GST:

Glutathione-S-transferase

HuR:

Human antigen R

IECs:

Intestinal epithelial cells

LPS:

Lipopolysaccharide

MdtJI:

Spermidine transporter protein

MTA:

Methylthioadenosine

mTOR:

Mammalian target of rapamycin

NCPAH:

N-carbamoylputrescine aminohydrolase

NPM:

Nucleophosmin

ODC1:

Ornithine decarboxylase 1

PAO:

Polyamine oxidase

PLP:

Pyridoxal phosphate

PotABC:

ATP-binding cassette protein

PTPN2:

Protein tyrosine phosphatase non-receptor type 2

RBPs:

RNA-binding proteins

ROS:

Reactive oxygen species

SAM:

S-adenosylmethionine

SAT:

Spermidine acetyltransferase

SMO:

Spermine oxidase

SMS:

Spermine synthase

SOD:

Superoxide dismutase

SRM:

Spermidine synthase

SSAT:

Spermidine/spermine-N1-acetyltransferase

STIM1:

Stromal interaction molecule 1

TJs:

Tight junctions

TRPC1:

Transient receptor potential channel 1

YdcW:

γ-Aminobutyraldehyde dehydrogenase

YgjG:

Putrescine transaminase

ZO:

Zonula Occludens

3'UTR:

3'Untranslated region

References

  1. Tabor CW, Tabor H. Polyamines. Annu Rev Biochem. 1984;53(1):749–90.

    Article  CAS  PubMed  Google Scholar 

  2. Li J, Meng Y, Wu X, Sun Y. Polyamines and related signaling pathways in cancer. Cancer Cell Int. 2020;20(1):539.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Pegg AE. Functions of Polyamines in mammals. J Biol Chem. 2016;291(29):14904–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ramos-Molina B, Queipo-Ortuño MI, Lambertos A, Tinahones FJ, Peñafiel R. Dietary and gut microbiota polyamines in obesity- and age-related diseases. Front Nutr. 2019;6:24.

  5. Matsumoto M, Kakizoe K, Benno Y. Comparison of fecal microbiota and polyamine concentration in adult patients with intractable atopic dermatitis and healthy adults. Microbiol Immunol. 2007;51(1):37–46.

    Article  CAS  PubMed  Google Scholar 

  6. Seiler N, Delcros JG, Moulinoux JP. Polyamine transport in mammalian cells. Int J Biochem Cell Biol. 1996;28(8):843–61.

    Article  CAS  PubMed  Google Scholar 

  7. Thomas T, Thomas TJ. Polyamines in cell growth and cell death: molecular mechanisms and therapeutic applications. Cell Mol Life Sci. 2001;58(2):244–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Minois N, Carmona-Gutierrez D, Madeo F. Polyamines in aging and disease. Aging. 2011;3(8):716–32.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Madeo F, Hofer SJ, Pendl T, Bauer MA, Eisenberg T, Carmona-Gutierrez D, et al. Nutritional aspects of spermidine. Annu Rev Nutr. 2020;40:135–59.

    Article  CAS  PubMed  Google Scholar 

  10. Hofer SJ, Simon AK, Bergmann M, Eisenberg T, Kroemer G, Madeo F, et al. Mechanisms of spermidine-induced autophagy and geroprotection. Nat Aging. 2022;2(12):1112–29.

    Article  PubMed  Google Scholar 

  11. Eisenberg T, Abdellatif M, Schroeder S, Primessnig U, Stekovic S, Pendl T, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22(12):1428–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gupta VK, Scheunemann L, Eisenberg T, Mertel S, Bhukel A, Koemans TS, et al. Restoring polyamines protects from age-induced memory impairment in an autophagy-dependent manner. Nat Neurosci. 2013;16(10):1453–60.

    Article  CAS  PubMed  Google Scholar 

  13. Kibe R, Kurihara S, Sakai Y, Suzuki H, Ooga T, Sawaki E, et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep. 2014;4:4548.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Nakamura A, Kurihara S, Takahashi D, Ohashi W, Nakamura Y, Kimura S, et al. Symbiotic polyamine metabolism regulates epithelial proliferation and macrophage differentiation in the colon. Nat Commun. 2021;12:2105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Casero RA, Murray Stewart T, Pegg AE. Polyamine metabolism and cancer: treatments, challenges and opportunities. Nat Rev Cancer. 2018;18(11):681–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Milovic V. Polyamines in the gut lumen: bioavailability and biodistribution. Eur J Gastroenterol Hepatol. 2001;13(9):1021–5.

    Article  CAS  PubMed  Google Scholar 

  17. Muñoz-Esparza NC, Latorre-Moratalla ML, Comas-Basté O, Toro-Funes N, Veciana-Nogués MT, Vidal-Carou MC. Polyamines in food. Front Nutr. 2019;6:108.

    Article  PubMed  Google Scholar 

  18. Matsumoto M, Kibe R, Ooga T, Aiba Y, Kurihara S, Sawaki E, et al. Impact of intestinal microbiota on intestinal luminal metabolome. Sci Rep. 2012;2:233.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. 2016;16(6):341–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pegg AE. Regulation of ornithine decarboxylase. J Biol Chem. 2006;281(21):14529–32.

    Article  CAS  PubMed  Google Scholar 

  21. Feng Q, Wang H, Shao Y, Xu X. Antizyme inhibitor family: biological and translational research implications. Cell Commun Signal. 2024;22:11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pegg AE, Michael AJ. Spermine synthase. Cell Mol Life Sci. 2010;67(1):113–21.

    Article  CAS  PubMed  Google Scholar 

  23. Wu H, Min J, Ikeguchi Y, Zeng H. Structure and mechanism of spermidine synthases. Biochemistry. 2007;46(28):8331–9.

    Article  CAS  PubMed  Google Scholar 

  24. Wu R, Chen X, Kang S, Wang T, Gnanaprakasam JR, Yao Y, et al. De novo synthesis and salvage pathway coordinately regulate polyamine homeostasis and determine T cell proliferation and function. Sci Adv. 2020;6(51):eabc4275.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bhat MI, Kapila R. Dietary metabolites derived from gut microbiota: critical modulators of epigenetic changes in mammals. Nutr Rev. 2017;75(5):374–89.

    Article  PubMed  Google Scholar 

  26. Nakamura A, Ooga T, Matsumoto M. Intestinal luminal putrescine is produced by collective biosynthetic pathways of the commensal microbiome. Gut Microbes. 2019;10(2):159–71.

    Article  CAS  PubMed  Google Scholar 

  27. Burrell M, Hanfrey CC, Murray EJ, Stanley-Wall NR, Michael AJ. Evolution and multiplicity of arginine decarboxylases in polyamine biosynthesis and essential role in Bacillus subtilis biofilm formation. J Biol Chem. 2010;285(50):39224–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pugin B, Barcik W, Westermann P, Heider A, Wawrzyniak M, Hellings P, et al. A wide diversity of bacteria from the human gut produces and degrades biogenic amines. Microb Ecol Health Dis. 2017;28(1):1353881.

    PubMed  PubMed Central  Google Scholar 

  29. Miller-Fleming L, Olin-Sandoval V, Campbell K, Ralser M. Remaining mysteries of molecular biology: the role of polyamines in the cell. J Mol Biol. 2015;427(21):3389–406.

    Article  CAS  PubMed  Google Scholar 

  30. Michael AJ. Biosynthesis of polyamines and polyamine-containing molecules. Biochem J. 2016;473(15):2315–29.

    Article  CAS  PubMed  Google Scholar 

  31. Kashiwagi K, Watanabe R, Igarashi K. Involvement of ribonuclease III in the enhancement of expression of the speF-potE operon encoding inducible ornithine decarboxylase and polyamine transport protein. Biochem Biophys Res Commun. 1994;200(1):591–7.

    Article  CAS  PubMed  Google Scholar 

  32. Sugiyama Y, Nara M, Sakanaka M, Gotoh A, Kitakata A, Okuda S, et al. Comprehensive analysis of polyamine transport and biosynthesis in the dominant human gut bacteria: potential presence of novel polyamine metabolism and transport genes. Int J Biochem Cell Biol. 2017;93:52–61.

    Article  CAS  PubMed  Google Scholar 

  33. Tait GH. A new Pathway for the biosynthesis of spermidine. Biochem Soc Trans. 1976;4(4):610–2.

    Article  CAS  PubMed  Google Scholar 

  34. Hanfrey CC, Pearson BM, Hazeldine S, Lee J, Gaskin DJ, Woster PM, et al. Alternative spermidine biosynthetic route is critical for growth of Campylobacter jejuni and is the dominant polyamine pathway in human gut microbiota. J Biol Chem. 2011;286(50):43301–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Shimokawa H, Sakanaka M, Fujisawa Y, Ohta H, Sugiyama Y, Kurihara S. N-Carbamoylputrescine amidohydrolase of Bacteroides thetaiotaomicron, a dominant species of the human gut microbiota. Biomedicines. 2023;11(4):1123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kurihara S. Polyamine metabolism and transport in gut microbes. Biosci Biotechnol Biochem. 2022;86(8):957–66.

    PubMed  Google Scholar 

  37. Kitada Y, Muramatsu K, Toju H, Kibe R, Benno Y, Kurihara S, et al. Bioactive polyamine production by a novel hybrid system comprising multiple indigenous gut bacterial strategies. Sci Adv. 2018;4(6):eaat0062.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Gong S, Richard H, Foster JW. YjdE (AdiC) is the arginine:agmatine antiporter essential for arginine-dependent acid resistance in Escherichia coli. J Bacteriol. 2003;185(15):4402–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Suárez C, Espariz M, Blancato VS, Magni C. Expression of the agmatine deiminase pathway in Enterococcus faecalis is activated by the AguR regulator and repressed by CcpA and PTS (Man) systems. PLoS ONE. 2013;8(10):e76170.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Noack J, Dongowski G, Hartmann L, Blaut M. The human gut bacteria Bacteroides thetaiotaomicron and Fusobacterium varium produce putrescine and spermidine in cecum of pectin-fed gnotobiotic rats. J Nutr. 2000;130(5):1225–31.

    Article  CAS  PubMed  Google Scholar 

  41. Xi H, Nie X, Gao F, Liang X, Li H, Zhou H, et al. A bacterial spermidine biosynthetic pathway via carboxyaminopropylagmatine. Sci Adv. 2023;9(43):eadj9075.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Seiler N, Raul F. Polyamines and the intestinal tract. Crit Rev Clin Lab Sci. 2007;44(4):365–411.

  43. Vujcic 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(1):19–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Pegg AE. Spermidine/spermine-N1-acetyltransferase: a key metabolic regulator. Am J Physiol Endocrinol Metab. 2008;294(6):E995-1010.

    Article  CAS  PubMed  Google Scholar 

  45. Pegg AE. Toxicity of polyamines and their metabolic products. Chem Res Toxicol. 2013;26(12):1782–800.

    Article  CAS  PubMed  Google Scholar 

  46. Mahajan UV, Varma VR, Griswold ME, Blackshear CT, An Y, Oommen AM, et al. Dysregulation of multiple metabolic networks related to brain transmethylation and polyamine pathways in Alzheimer disease: a targeted metabolomic and transcriptomic study. PLoS Med. 2020;17(1):e1003012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lewandowski NM, Ju S, Verbitsky M, Ross B, Geddie ML, Rockenstein E, et al. Polyamine pathway contributes to the pathogenesis of Parkinson disease. Proc Natl Acad Sci U S A. 2010;107(39):16970–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Barbagallo M, Martino MLD, Marcocci L, Marcocci L, Pietrangeli P, De Carolis E, et al. A new piece of the Shigella pathogenicity puzzle: spermidine accumulationby silencing of the speG gene. PLoS One. 2011;6(11):e27226.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li B, Maezato Y, Kim SH, Kurihara S, Liang J, Michael AJ. Polyamine-independent growth and biofilm formation, and functional spermidine/spermine N-acetyltransferases in Staphylococcus aureus and Enterococcus faecalis. Mol Microbiol. 2019;111(1):159–75.

    Article  CAS  PubMed  Google Scholar 

  50. Forouhar F, Lee IS, Vujcic J, Vujcic S, Shen J, Vorobiev SM, et al. Structural and functional evidence for Bacillus subtilis PaiA as a novel N1-spermidine/spermine acetyltransferase. J Bio Chem. 2005;280(48):40328–36.

    Article  CAS  Google Scholar 

  51. Schneider BL, Reitzer L. Pathway and enzyme redundancy in putrescine catabolism in Escherichia coli. J Bacteriol. 2012;194(15):4080–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Samsonova NN, Smirnov SV, Altman IB, Ptitsyn LR. Molecular cloning and characterization of Escherichia coli K12 ygjG gene. BMC Microbiol. 2003;3:2.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Kurihara S, Oda S, Kato K, Kim HG, Koyanagi T, Kumagai H, et al. A novel putrescine utilization pathway involves γ-glutamylated intermediates of Escherichia coli K-12. J Biol Chem. 2005;280(6):4602–8.

    Article  CAS  PubMed  Google Scholar 

  54. Kurihara S, Tsuboi Y, Oda S, Kim HG, Kumagai H, Suzuki H. The putrescine importer PuuP of Escherichia coli K-12. J Bacteriol. 2009;191(8):2776–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kurihara S, Oda S, Tsuboi Y, Kim HG, Oshida M, Kumagai H, et al. γ-Glutamylputrescine synthetase in the putrescine utilization pathway of Escherichia coli K-12. J Biol Chem. 2008;283(29):19981–90.

    Article  CAS  PubMed  Google Scholar 

  56. Kurihara S, Oda S, Kumagai H, Suzuki H. Gamma-glutamyl-gamma-aminobutyrate hydrolase in the putrescine utilization pathway of Escherichia coli K-12. FEMS Microbiol Lett. 2006;256(2):318–23.

    Article  CAS  PubMed  Google Scholar 

  57. Kurihara S, Kato K, Asada K, Kumagai H, Suzuki H. A putrescine-inducible pathway comprising PuuE-YneI in which gamma-aminobutyrate is degraded into succinate in Escherichia coli K-12. J Bacteriol. 2010;192(18):4582–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Shaibe E, Metzer E, Halpern YS. Metabolic pathway for the utilization of L-arginine, L-ornithine, agmatine, and putrescine as nitrogen sources in Escherichia coli K-12. J Bacteriol. 1985;163(3):933–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Schneider BL, Hernandez VJ, Reitzer L. Putrescine catabolism is a metabolic response to several stresses in Escherichia coli. Mol Microbiol. 2013;88(3):537–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Sugiyama Y, Nara M, Sakanaka M, Kitakata A, Okuda S, Kurihara S. Analysis of polyamine biosynthetic- and transport ability of human indigenous Bifidobacterium. Biosci Biotechnol Biochem. 2018;82(9):1606–14.

    Article  CAS  PubMed  Google Scholar 

  61. Bardocz S, Grant G, Brown D, Ralph A, Pusztai A. Polyamines in food—implications for growth and health. J Nutr Biochem. 1993;4(2):66–71.

    Article  CAS  Google Scholar 

  62. Osborne DL, Seidel ER. Gastrointestinal luminal polyamines: cellular accumulation and enterohepatic circulation. Am J Physiol. 1990;258(4):G576–84.

    CAS  PubMed  Google Scholar 

  63. 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 U S A. 2013;110(6):2169–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Abdulhussein AA, Wallace HM. Polyamines and membrane transporters. Amino Acids. 2014;46(3):655–60.

    Article  CAS  PubMed  Google Scholar 

  65. Kashiwagi K, Pistocchi R, Shibuya S, Sugiyama S, Morikawa K, Igarashi K. Spermidine-preferential uptake system in Escherichia coli. Identification of amino acids involved in polyamine binding in PotD protein. J Biol Chem. 1996;271(21):12205–8.

  66. Furuchi T, Kashiwagi K, Kobayashi H, Igarashi K. Characteristics of the gene for a spermidine and putrescine transport system that maps at 15 min on the Escherichia coli chromosome. J Biol Chem. 1991;266(31):20928–33.

    Article  CAS  PubMed  Google Scholar 

  67. Kröger P, Shanmugaratnam S, Scheib U, Höcker B. Fine-tuning spermidine binding modes in the putrescine binding protein PotF. J Biol Chem. 2021;297(6):101419.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Igarashi K, Kashiwagi K. Characteristics of cellular polyamine transport in prokaryotes and eukaryotes. Plant Physiol Biochem. 2010;48(7):506–12.

    Article  CAS  PubMed  Google Scholar 

  69. Kurihara S, Suzuki H, Oshida M, Benno Y. A novel putrescine importer required for type 1 pili-driven surface motility induced by extracellular putrescine in Escherichia coli K-12. J Biol Chem. 2011;286(12):10185–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Soksawatmaekhin W, Kuraishi A, Sakata K, Kashiwagi K, Igarashi K. Excretion and uptake of cadaverine by CadB and its physiological functions in Escherichia coli. Mol Microbiol. 2004;51(5):1401–12.

    Article  CAS  PubMed  Google Scholar 

  71. Tomitori H, Kashiwagi K, Igarashi K. Structure and function of polyamine-amino acid antiporters CadB and PotE in Escherichia coli. Amino Acids. 2012;42(2–3):733–40.

    Article  CAS  PubMed  Google Scholar 

  72. Higashi K, Ishigure H, Demizu R, Uemura T, Nishino K, Yamaguchi A, et al. Identification of a spermidine excretion protein complex (MdtJI) in Escherichia coli. J Bacteriol. 2008;190(3):872–8.

    Article  CAS  PubMed  Google Scholar 

  73. Sugiyama Y, Nakamura A, Matsumoto M, Kanbe A, Sakanaka M, Higashi K, et al. A novel putrescine exporter SapBCDF of Escherichia coli. J Biol Chem. 2016;291(51):26343–51.

    Article  CAS  PubMed  Google Scholar 

  74. Kurihara S, Suzuki H. Recent advances in bacterial polyamine transport systems. In: Kusano T, Suzuki H, editors. Polyamines. Tokyo: Springer; 2015. pp. 171–8.

    Chapter  Google Scholar 

  75. Woolridge DP, Vazquez-Laslop N, Markham PN, Chevalier MS, Gerner EW, Neyfakh AA. Efflux of the natural polyamine spermidine facilitated by the Bacillus subtilis multidrug transporter Blt. J Biol Chemi. 1997;272(14):8864–6.

    Article  CAS  Google Scholar 

  76. McGinnis MW, Parker ZM, Walter NE, Rutkovsky AC, Cartaya-Marin C, Karatan E. Spermidine regulates Vibrio cholerae biofilm formation via transport and signaling pathways. Fems Microbiol Lett. 2009;299(2):166–74.

    Article  CAS  PubMed  Google Scholar 

  77. Bridges AA, Bassler BL. Inverse regulation of Vibrio cholerae biofilm dispersal by polyamine signals. Elife. 2021;10:e65487.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kashiwagi K, Igarashi K. Identification and assays of polyamine transport systems in Escherichia coli and Saccharomyces cerevisiae. Methods Mol Biol. 2011;720:295–308.

    Article  CAS  PubMed  Google Scholar 

  79. Uemura T, Kashiwagi K, Igarashi K. Polyamine uptake by DUR3 and SAM3 in Saccharomyces cerevisiae. J Biol Chem. 2007;282(10):7733–41.

    Article  CAS  PubMed  Google Scholar 

  80. Michael AJ. Polyamine function in archaea and bacteria. J Biol Chem. 2018;293(48):18693–701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Lee J, Sperandio V, Frantz DE, Longgood J, Camilli A, Phillips MA, et al. An alternative polyamine biosynthetic pathway is widespread in bacteria and essential for biofilm formation in Vibrio cholerae. J Biol Chem. 2009;284(15):9899–907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Katz AM, Tolokh IS, Pabit SA, Baker N, Onufriev AV, Pollack L. Spermine condenses DNA, but not RNA duplexes. Biophys J. 2017;112(1):22–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Sievert H, Venz S, Platas-Barradas O, Dhople VM, Schaletzky M, Nagel CH, et al. Protein-protein-interaction network organization of the hypusine modification system. Mol Cell Proteomics. 2012;11(11):1289–305.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Saini P, Eyler DE, Green R, Dever TE. Hypusine-containing protein eIF5A promotes translation elongation. Nature. 2009;459(7243):118–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hyvönen MT, Keinänen TA, Khomutov M, Simonian A, Vepsäläinen J, Park JH, et al. Effects of novel C-methylated spermidine analogs on cell growth via hypusination of eukaryotic translation initiation factor 5A. Amino Acids. 2012;42(2–3):685–95.

    Article  PubMed  Google Scholar 

  86. Chattopadhyay MK, Park MH, Tabor H. Hypusine modification for growth is the major function of spermidine in Saccharomyces cerevisiae polyamine auxotrophs grown in limiting spermidine. Proc Natl Acad Sci U S A. 2008;105(18):6554–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Valdés-Santiago L, Ruiz-Herrera J. Stress and polyamine metabolism in fungi. Front Chem. 2013;1:42.

    PubMed  Google Scholar 

  88. Koroleva E, Toplis B, Taylor M, Deventer C, Steffen HC, Heever C, et al. Exploring polyamine metabolism of the yeast-like fungus Emergomyces africanus. FEMS Yeast Res. 2024;24:foae038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Herrero AB, López MC, García S, Schmidt A, Spaltmann F, Ruiz-Herrera J, et al. Control of filament formation in Candida albicans by polyamine levels. Infect Immun. 1999;67(9):4870–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Nakada Y, Itoh Y. Identification of the putrescine biosynthetic genes in Pseudomonas aeruginosa and characterization of agmatine deiminase and N-carbamoylputrescine amidohydrolase of the arginine decarboxylase pathway. Microbiology. 2003;149(3):707–14.

    Article  CAS  PubMed  Google Scholar 

  91. Potter AJ, Paton JC. Spermidine biosynthesis and transport modulate pneumococcal autolysis. J Bacteriol. 2014;196(20):3556–61.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Lowe-Power TM, Hendrich CG, Von Roepenack-Lahaye E, Li B, Wu D, Mitra R, et al. Metabolomics of tomato xylem sap during bacterial wilt reveals Ralstonia solanacearum produces abundant putrescine, a metabolite that accelerates wilt disease. Environ Microbiol. 2018;20(4):1330–49.

    Article  CAS  PubMed  Google Scholar 

  93. Nair AV, Singh A, Rajmani RS, Chakravortty D. Salmonella Typhimurium employs spermidine to exert protection against ROS-mediated cytotoxicity and rewires host polyamine metabolism to ameliorate its survival in macrophages. Redox Biol. 2024;72:103151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Bower JM, Mulvey MA. Polyamine-mediated resistance of uropathogenic Escherichia coli to nitrosative stress. J Bacteriol. 2006;188(3):928–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Komamura T, Nishimura T, Ohta N, Takado M, Matsumoto T, Takeda K. The putative polyamine transporter Shp2 facilitates phosphate export in an Xpr1-independent manner and contributes to high phosphate tolerance. J Biol Chem. 2025;301(1):108056.

    Article  CAS  PubMed  Google Scholar 

  96. Schwarz J, Brameyer S, Hoyer E, Jung K. The interplay of AphB and CadC to activate acid resistance of Vibrio campbellii. J Bacteriol. 2023;205(4):e0045722.

    Article  PubMed  Google Scholar 

  97. Du C, Huo X, Gu H, Wu D, Hu Y. Acid resistance system CadBA is implicated in acid tolerance and biofilm formation and is identified as a new virulence factor of Edwardsiella tarda. Vet Res. 2021;52(1):117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kim SK, Jin YS, Choi IG, Park YC, Seo JH. Enhanced tolerance of Saccharomyces cerevisiae to multiple lignocellulose-derived inhibitors through modulation of spermidine contents. Metab Eng. 2015;29:46–55.

    Article  CAS  PubMed  Google Scholar 

  99. Jung IL, Kim IG. Polyamines and glutamate decarboxylase-based acid resistance in Escherichia coli. J Biol Chem. 2003;278(25):22846–52.

    Article  CAS  PubMed  Google Scholar 

  100. Nair AV, Singh A, Devasurmutt Y, Rahman SA, Tatu US, Chakravortty D. Spermidine constitutes a key determinant of motility and attachment of Salmonella Typhimurium through a novel regulatory mechanism. Microbiol Res. 2024;281:127605.

    Article  CAS  PubMed  Google Scholar 

  101. Barraud N, Kjelleberg S, Rice SA. Dispersal from microbial biofilms. In: Ghannoum M, Parsek M, Whiteley M, Mukherjee PK, editors. Microbial Biofilms, 2nd ed. ASM Press; 2015.

  102. Iyer R, Wu Z, Woster PM, Delcour AH. Molecular basis for the polyamine-ompF porin interactions: inhibitor and mutant studies. J Mol Biol. 2000;297(4):933–45.

    Article  CAS  PubMed  Google Scholar 

  103. Dela Vega AL, Delcour AH. Polyamines decrease Escherichia coli outer membrane permeability. J Bacteriol. 1996;178(13):3715–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Thongbhubate K, Nakafuji Y, Matsuoka R, Kakegawa S, Suzuki H. Effect of spermidine on biofilm formation in Escherichia coli K-12. J Bacteriol. 2021;203(10):e00652-e720.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Vieira B, Alcantara JB, Destro G, Guerra MES, Oliveira S, Lima CA, et al. Role of the polyamine transporter PotABCD during biofilm formation by Streptococcus pneumoniae. PLoS One. 2024;19(8):e0307573.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Wortham BW, Oliveira MA, Fetherston JD, Perry RD. Polyamines are required for the expression of key Hms proteins important for Yersinia pestis biofilm formation. Environ Microbiol. 2010;12(7):2034–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Hobley L, Li B, Wood JL, Kim SH, Naidoo J, Ferreira AS, et al. Spermidine promotes Bacillus subtilis biofilm formation by activating expression of the matrix regulator slrR. J Biol Chem. 2017;292(29):12041–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Ding Y, Peng N, Du Y, Ji L, Cao B. Disruption of putrescine biosynthesis in Shewanella oneidensis enhances biofilm cohesiveness and performance in Cr(VI) immobilization. Appl Environ Microbiol. 2014;80(4):1498–506.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Wen X, Wang L, Zheng C, Yang X, Ma X, Wu Y, et al. Fecal scores and microbial metabolites in weaned piglets fed different protein sources and levels. Anim Nutr. 2018;4(1):31–6.

    Article  PubMed  Google Scholar 

  110. Gómez-Gallego C, Collado MC, Ilo T, Jaakkola UM, Bernal MJ, Periago MJ, et al. Infant formula supplemented with polyamines alters the intestinal microbiota in neonatal BALB/cOlaHsd mice. J Nutr Biochem. 2012;23(11):1508–13.

    Article  PubMed  Google Scholar 

  111. Jiang DM, Wang ZL, Yang JD, Wang X, Niu CY, Ji CW, et al. Effects of spermidine on mouse gut morphology, metabolites, and microbial diversity. Nutrients. 2023;15(3):744.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Liu S, Liu Y, Zhao J, Yang P, Wang W, Liao M. Effects of spermidine on gut microbiota modulation in experimental abdominal aortic aneurysm mice. Nutrients. 2022;14(16):3349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Liu G, Mo W, Cao W, Wu X, Jia G, Zhao H, et al. Effects of spermine on ileal physical barrier, antioxidant capacity, metabolic profile and large intestinal bacteria in piglets. RSC Adv. 2020;10(45):26709–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Wang Z, Jiang D, Wang X, Jiang Y, Sun Q, Ling W, et al. Spermidine improves the antioxidant capacity and morphology of intestinal tissues and regulates intestinal microorganisms in Sichuan white geese. Front Microbiol. 2024;14:1292984.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Liu G, Zheng J, Wu X, Xu X, Jia G, Zhao H, et al. Putrescine enhances intestinal immune function and regulates intestinal bacteria in weaning piglets. Food Funct. 2019;10(7):4134–42.

    Article  CAS  PubMed  Google Scholar 

  116. Liu B, Jiang X, Cai L, Zhao X, Dai Z, Wu G, et al. Putrescine mitigates intestinal atrophy through suppressing inflammatory response in weanling piglets. J Anim Sci Biotechnol. 2019;10:69.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Harrold D, Saunders R, Bailey J. Dietary putrescine supplementation reduces faecal abundance of Clostridium perfringens and markers of inflammation in captive azure-winged magpies. J Zoo Aquar Res. 2020;8(2):114–23.

    Google Scholar 

  118. Niechcial A, Schwarzfischer M, Wawrzyniak M, Atrott K, Laimbacher A, Morsy Y, et al. Spermidine ameliorates colitis via induction of anti-inflammatory macrophages and prevention of intestinal dysbiosis. J Crohns Colitis. 2023;17(9):1489–503.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Gobert AP, Latour YL, Asim M, Barry DP, Allaman MM, Finley JL, et al. Protective role of spermidine in colitis and colon carcinogenesis. Gastroenterology. 2022;162(3):813–27.

    Article  CAS  PubMed  Google Scholar 

  120. Yan B, Mao X, Hu S, Wang S, Liu X, Sun J. Spermidine protects intestinal mucosal barrier function in mice colitis via the AhR/Nrf2 and AhR/STAT3 signaling pathways. Int Immunopharmacology. 2023;119:110166.

    Article  CAS  Google Scholar 

  121. Fang T, Liu G, Cao W, Wu X, Jia G, Zhao H, et al. Spermine: new insights into the intestinal development and serum antioxidant status of suckling piglets. RSC Adv. 2016;6(37):31323–35.

    Article  CAS  Google Scholar 

  122. Cao W, Liu G, Fang T, Wu X, Jia G, Zhao H, et al. Effects of spermine on the morphology, digestive enzyme activities, and antioxidant status of jejunum in suckling rats. RSC Adv. 2015;5(93):76607–14.

    Article  CAS  Google Scholar 

  123. Fang T, Jia G, Zhao H, Chen X, Tang J, Wang J, et al. Effects of spermine supplementation on the morphology, digestive enzyme activities, and antioxidant capacity of intestine in weaning rats. Anim Nutr. 2016;2(4):370–5.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Van Der Flier LG, Clevers H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu Rev Physiol. 2009;71(1):241–60.

    Article  PubMed  Google Scholar 

  125. Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14(3):141–53.

    Article  CAS  PubMed  Google Scholar 

  126. Crosnier C, Stamataki D, Lewis J. Organizing cell renewal in the intestine: stem cells, signals and combinatorial control. Nat Rev Genet. 2006;7(5):349–59.

    Article  CAS  PubMed  Google Scholar 

  127. Wang J, Xiao L, Wang J. Posttranscriptional regulation of intestinal epithelial integrity by noncoding RNAs. Wiley Interdiscip Rev RNA. 2017;8(2):1399.

    Article  Google Scholar 

  128. Wang JY, McCormack SA, Viar MJ, Johnson LR. Stimulation of proximal small intestinal mucosal growth by luminal polyamines. Am J Physiol. 1991;261(3):G504–11.

    CAS  PubMed  Google Scholar 

  129. Wang JY, Johnson LR. Polyamines and ornithine decarboxylase during repair of duodenal mucosa after stress in rats. Gastroenterology. 1991;100(2):333–43.

    Article  CAS  PubMed  Google Scholar 

  130. Dandrifosse G, Peulen O, El Khefif N, Deloyer P, Dandrifosse AC, Grandfils C. Are milk polyamines preventive agents against food allergy? Proc Nutr Soc. 2000;59(1):81–6.

    Article  CAS  PubMed  Google Scholar 

  131. Johnson LR, McCormack SA. Healing of gastrointestinal mucosa: involvement of polyamines. Physiology. 1999;14(1):12–7.

    Article  CAS  Google Scholar 

  132. Wang J, Li GR, Tan BE, Xiong X, Kong XF, Xiao DF, et al. Oral administration of putrescine and proline during the suckling period improves epithelial restitution after early weaning in piglets. J Anim Sci. 2015;93(4):1679–88.

    Article  CAS  PubMed  Google Scholar 

  133. Rao JN, Li L, Bass BL, Wang JY. Expression of the TGF-β receptor gene and sensitivity to growth inhibition following polyamine depletion. Am J Physio. 2023;279(4):C1034–44.

    Article  Google Scholar 

  134. Tabib A, Bachrach U. Activation of the proto-oncogene c-myc and c-fos by c-ras: involvement of polyamines. Biochem Biophys Res Commun. 1994;202(2):720–7.

    Article  CAS  PubMed  Google Scholar 

  135. Zou T, Mazan-Mamczarz K, Rao JN, Liu L, Marasa BS, Zhang AH, 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(28):19387–94.

    Article  CAS  PubMed  Google Scholar 

  136. Li L, Liu L, Rao JN, Esmaili A, Strauch ED, Bass BL, et al. JunD stabilization results in inhibition of normal intestinal epithelial cell growth through P21 after polyamine depletion. Gastroenterology. 2002;123(3):764–79.

    Article  CAS  PubMed  Google Scholar 

  137. Rao JN, Xiao L, Wang JY. Polyamines in gut epithelial renewal and barrier function. Physiology. 2020;35(5):328–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Liu L, Guo X, Rao JN, Zou T, Marasa BS, Chen J, et al. Polyamine-modulated c-Myc expression in normal intestinal epithelial cells regulates p21Cip1 transcription through a proximal promoter region. Biochem J. 2006;398(2):257–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Wang JY, Johnson LR. Expression of protooncogenes c-fos and c-myc in healing of gastric mucosal stress ulcers. Am J Physiol. 1994;266(5):G878–86.

    CAS  PubMed  Google Scholar 

  140. Konturek PCh, 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:S97-104.

    Article  PubMed  Google Scholar 

  141. Liu L, Santora R, Rao JN, Guo X, Zou T, Zhang HM, et al. Activation of TGF-β-Smad signaling pathway following polyamine depletion in intestinal epithelial cells. Am J Physiol Gastrointestinal Liver Physiol. 2003;285(5):G1056–67.

    Article  CAS  Google Scholar 

  142. Li L, Rao JN, Bass BL, Wang JY. NF-κB activation and susceptibility to apoptosis after polyamine depletion in intestinal epithelial cells. Am J Physiol Gastrointestinal Liver Physiol. 2001;280(5):G992-1004.

    Article  CAS  Google Scholar 

  143. Wang C, Ruan P, Zhao Y, Li X, Wang J, Wu X, et al. Spermidine/spermine N1-acetyltransferase regulates cell growth and metastasis via AKT/β-catenin signaling pathways in hepatocellular and colorectal carcinoma cells. Oncotarget. 2016;8(1):1092–109.

    Article  PubMed Central  Google Scholar 

  144. Xiao L, Rao JN, Zou T, Liu L, Marasa BS, Chen J, 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(11):4579–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhang X, Zou T, Rao JN, Liu L, Xiao L, Wang PY, et al. Stabilization of XIAP mRNA through the RNA binding protein HuR regulated by cellular polyamines. Nucleic Acids Res. 2009;37(22):7623–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Barreau C, Paillard L, Osborne HB. AU-rich elements and associated factors: are there unifying principles? Nucleic Acids Res. 2005;33(22):7138–50.

    Article  CAS  PubMed  Google Scholar 

  147. Chen CY, Shyu AB. AU-rich elements: characterization and importance in mRNA degradation. Trends Biochem Sci. 1995;20(11):465–70.

    Article  CAS  PubMed  Google Scholar 

  148. Wang X, Wang T, Zhong Z. The impact of RNA-binding protein AUF1 on the stability of inflammatory cytokines mRNAs. Int Immunol. 2019;42(4):396–401.

    Google Scholar 

  149. Zou T, Rao JN, Liu L, Xiao L, Yu TX, Jiang P, 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(21):5021–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Brennan CM, Steitz JA. HuR and mRNA stability. Cell Mol Life Sci. 2001;58(2):266–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Liu L, Christodoulou-Vafeiadou E, Rao JN, Zou T, Xiao L, Chung HK, et al. RNA-binding protein HuR promotes growth of small intestinal mucosa by activating the Wnt signaling pathway. Mol Biol Cell. 2014;25(21):3308–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Zou T, Liu L, Rao JN, Marasa BS, Chen J, Xiao L, et al. Polyamines modulate the subcellular localization of RNA-binding protein HuR through AMP-activated protein kinase-regulated phosphorylation and acetylation of importin α1. Biochem J. 2007;409(2):389–98.

    Article  Google Scholar 

  153. Wang PY, Rao JN, Zou T, Liu L, Xiao L, Yu TX, et al. Post-transcriptional regulation of MEK-1 by polyamines through the RNA-binding protein HuR modulating intestinal epithelial apoptosis. Biochem J. 2010;426(3):293–306.

    Article  CAS  PubMed  Google Scholar 

  154. Liu L, Rao JN, Zou T, Xiao L, Wang PY, Turner DJ, et al. Polyamines regulate c-Myc translation through Chk2-dependent HuR phosphorylation. Mol Biol Cell. 2009;20(23):4885–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. 2009;9(11):799–809.

    Article  CAS  PubMed  Google Scholar 

  156. Capaldo CT, Nusrat A. Claudin switching: physiological plasticity of the tight junction. Semin Cell Dev Biol. 2015;42:22–9.

    Article  CAS  PubMed  Google Scholar 

  157. Umeda K, Matsui T, Nakayama M, Furuse K, Sasaki H, Furuse M, et al. Establishment and characterization of cultured epithelial cells lacking expression of ZO-1. J Biol Chem. 2004;279(43):44785–94.

    Article  CAS  PubMed  Google Scholar 

  158. Guo X, Rao JN, Liu L, Zou T, Keledjian KM, Boneva D, et al. Polyamines are necessary for synthesis and stability of occludin protein in intestinal epithelial cells. Am J Physiol Gastrointestinal Liver Physiol. 2005;288(6):G1159–69.

    Article  CAS  Google Scholar 

  159. Yu TX, Wang PY, Rao JN, Zou T, Liu L, Xiao L, et al. Chk2-dependent HuR phosphorylation regulates occludin mRNA translation and epithelial barrier function. Nucleic Acids Res. 2011;39(19):8472–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Liu L, Guo X, Rao JN, Zou T, Xiao L, Yu T, et al. Polyamines regulate E-cadherin transcription through c-Myc modulating intestinal epithelial barrier function. Am J Physiol Cell Physiol. 2009;296(4):C801–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Wang JY. Polyamines regulate expression of E-cadherin and play an important role in control of intestinal epithelial barrier function. Inflammopharmacology. 2005;13(1–3):91–101.

    Article  CAS  PubMed  Google Scholar 

  162. Wang SR, Mallard CG, Cairns CA, Chung HK, Yoo D, Jaladanki SK, et al. Stabilization of Cx43 mRNA via RNA-binding protein HuR regulated by polyamines enhances intestinal epithelial barrier function. Am J Physiol Gastrointest Liver Physiol. 2023;325(6):G518–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Rao JN, Rathor N, Zhuang R, Zou T, Liu L, Xiao L, et al. Polyamines regulate intestinal epithelial restitution through TRPC1-mediated Ca2+ signaling by differentially modulating STIM1 and STIM2. Am J Physiol Cell Physiol. 2012;303(3):C308–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. 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(2):C303–14.

    Article  CAS  PubMed  Google Scholar 

  165. Tan B, Xiao D, Wang J, Tan B. The roles of polyamines in intestinal development and function in piglets. Animals. 2024;14(8):1228.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Pankoke S, Pfarrer C, Glage S, Mühlfeld C, Schipke J. Oral supplementation with the polyamine spermidine affects hepatic but not pulmonary lipid metabolism in lean but not obese mice. Nutrients. 2022;14(20):4318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Wang D, Yin J, Zhou Z, Tao Y, Jia Y, Jie H, et al. Oral spermidine targets brown fat and skeletal muscle to mitigate diet-induced obesity and metabolic disorders. Mol Nutr Food Res. 2021;65(19):e2100315.

    Article  PubMed  Google Scholar 

  168. Liu G, Fang T, Yan T, Jia G, Zhao H, Huang Z, et al. Metabolomic strategy for the detection of metabolic effects of spermine supplementation in weaned rats. J Agric Food Chem. 2014;62(36):9035–42.

    Article  CAS  PubMed  Google Scholar 

  169. Vuohelainen S, Pirinen E, Cerrada-Gimenez M, Keinänen TA, Uimari A, Pietilä M, et al. Spermidine is indispensable in differentiation of 3T3-L1 fibroblasts to adipocytes. J Cell Mol Med. 2010;14(6b):1683–92.

    Article  CAS  PubMed  Google Scholar 

  170. Ni Y, Zheng L, Zhang L, Li J, Pan Y, Du H, et al. Spermidine activates adipose tissue thermogenesis through autophagy and fibroblast growth factor 21. J Nutr Biochem. 2024;125:109569.

    Article  CAS  PubMed  Google Scholar 

  171. Eom J, Choi J, Suh SS, Seo JB. SLC3A2 and SLC7A2 mediate the exogenous putrescine-induced adipocyte differentiation. Mol Cells. 2022;45(12):963–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Nakatani S, Horimoto Y, Nakabayashi N, Karasawa M, Wada M, Kobata K. Spermine suppresses adipocyte differentiation and exerts anti-obesity effects in vitro and in vivo. Int J Mol Sci. 2022;23(19):11818.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Tjabringa GS, Zandieh-Doulabi B, Helder MN, Knippenberg M, Wuisman PIJM, Klein-Nulend J. The polymine spermine regulates osteogenic differentiation in adipose stem cells. J Cell Mol Med. 2008;12(5a):1710–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Ishii I, Ikeguchi Y, Mano H, Wada M, Pegg AE, Shirahata A. Polyamine metabolism is involved in adipogenesis of 3T3-L1 cells. Amino Acids. 2012;42(2–3):619–26.

    Article  CAS  PubMed  Google Scholar 

  175. Cruz-Pulido YE, LoMascolo NJ, May D, Hatahet J, Thomas CE, Chu AKW, et al. Polyamines mediate cellular energetics and lipid metabolism through mitochondrial respiration to facilitate virus replication. PLoS Pathog. 2024;20(11):e1012711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Fernández ÁF, Bárcena C, Martínez-García GG, Tamargo-Gómez I, Suárez MF, Pietrocola F, et al. Autophagy couteracts weight gain, lipotoxicity and pancreatic β-cell death upon hypercaloric pro-diabetic regimens. Cell Death Dis. 2017;8(8):e2970.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Michiels CF, Kurdi A, Timmermans JP, De Meyer GRY, Martinet W. Spermidine reduces lipid accumulation and necrotic core formation in atherosclerotic plaques via induction of autophagy. Atherosclerosis. 2016;251:319–27.

    Article  CAS  PubMed  Google Scholar 

  178. Brenner S, Bercovich Z, Feiler Y, Keshet R, Kahana C. Dual regulatory role of polyamines in adipogenesis. J Biol Chem. 2015;290(45):27384–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. 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;282(11):8404–13.

    Article  CAS  PubMed  Google Scholar 

  180. Morales TS, Avis EC, Paskowski EK, Shabar H, Nowotarski SL, DiAngelo JR. The role of spermidine synthase (SpdS) and spermine synthase (Sms) in regulating triglyceride storage in Drosophila. Med Sci. 2021;9(2):27.

    CAS  Google Scholar 

  181. Koponen T, Cerrada-Gimenez M, Pirinen E, Hohtola E, Paananen J, Vuohelainen S, et al. The activation of hepatic and muscle polyamine catabolism improves glucose homeostasis. Amino Acids. 2012;42(2–3):427–40.

    Article  CAS  PubMed  Google Scholar 

  182. Mossmann D, Park S, Hall MN. mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer. 2018;18(12):744–57.

    Article  CAS  PubMed  Google Scholar 

  183. Johnson LR, Brockway PD, Madsen K, Hardin JA, Gall DG. Polyamines alter intestinal glucose transport. Am J Physiol Gastrointestinal Liver Physiol. 1995;268(3):G416–23.

    Article  CAS  Google Scholar 

  184. Uda K, Tsujikawa T, Ihara T, Fujiyama Y, Bamba T. Luminal polyamines upregulate transmural glucose transport in the rat small intestine. J Gastroenterology. 2002;37(6):434–41.

    Article  CAS  Google Scholar 

  185. Ma L, Ni Y, Wang Z, Tu W, Ni L, Zhuge F, et al. Spermidine improves gut barrier integrity and gut microbiota function in diet-induced obese mice. Gut Microbes. 2020;12(1):1–19.

    Article  PubMed  Google Scholar 

  186. Sadasivan SK, Vasamsetti B, Singh J, Marikunte VV, Oommen AM, Jagannath MR, et al. Exogenous administration of spermine improves glucose utilization and decreases bodyweight in mice. Eur J Pharmacol. 2014;729:94–9.

    Article  CAS  PubMed  Google Scholar 

  187. Bardócz S, Grant G, Brown DS, Pusztai A. Putrescine as a source of instant energy in the small intestine of the rat. Gut. 1998;42(1):24–8.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Marselli L, Bosi E, De Luca C, Del Guerra S, Tesi M, Suleiman M, et al. Arginase 2 and polyamines in human pancreatic beta cells: possible role in the pathogenesis of Type 2 diabetes. Int J Mol Sci. 2021;22(22):12099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Zhang R, Xu J, Li R, Yu Z, Yuan W, Gao H, et al. Association between serum spermidine and TyG index: results from a cross-sectional study. Nutrients. 2022;14(18):3847.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Zhang H, Alsaleh G, Feltham J, Sun Y, Napolitano G, Riffelmacher T, et al. Polyamines control eIF5A hypusination, TFEB translation, and autophagy to reverse B cell senescence. Mol Cell. 2019;76(1):110-25.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Guarente L, Sinclair DA, Kroemer G. Human trials exploring anti-aging medicines. Cell Metab. 2024;36(2):354–76.

    Article  CAS  PubMed  Google Scholar 

  192. Zhang Y, Bai J, Cui Z, Li Y, Gao Q, Miao Y, et al. Polyamine metabolite spermidine rejuvenates oocyte quality by enhancing mitophagy during female reproductive aging. Nat Aging. 2023;3(11):1372–86.

    Article  CAS  PubMed  Google Scholar 

  193. Bai J, Zhang Y, Li N, Cui Z, Zhang H, Liu Y, et al. Supplementation of spermidine enhances the quality of postovulatory aged porcine oocytes. Cell Commun Signal. 2024;22(1):499.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Mate NA, Wadhwa G, Taliyan R, Banerjee A. Impact of polyamine supplementation on GnRH expression, folliculogenesis, and puberty onset in young mice. Theriogenology. 2024;229:202–13.

    Article  CAS  PubMed  Google Scholar 

  195. Li R, Wu X, Cheng J, Zhu Z, Guo M, Hou G, et al. Polyamines protect porcine sperm from lipopolysaccharide-induced mitochondrial dysfunction and apoptosis via casein kinase 2 activation. J Anim Sci. 2025;103:skae383.

    Article  PubMed  Google Scholar 

  196. Lenis YY, Johnson GA, Wang X, Tang WW, Dunlap KA, Satterfield MC, et al. Functional roles of ornithine decarboxylase and arginine decarboxylase during the peri-implantation period of pregnancy in sheep. J Anim Sci Biotechnol. 2018;9:10.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Teratani T, Kasahara N, Ijichi T, Fujimoto Y, Sakuma Y, Sata N, et al. Activation of whole body by high levels of polyamine intake in rats. Amino Acids. 2021;53(11):1695–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Kasahara N, Teratani T, Yokota S, Sakuma Y, Sasanuma H, Fujimoto Y, et al. Dietary polyamines promote intestinal adaptation in an experimental model of short bowel syndrome. Sci Rep. 2024;14:4605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Ma Y, Zhong Y, Tang W, Valencak TG, Liu J, Deng Z, et al. Lactobacillus reuteri ZJ617 attenuates metabolic syndrome via microbiota-derived spermidine. Nat Commun. 2025;16:877.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by projects from the Xinjiang Production and Construction Corps Major Science and Technology "Revealing the List and Taking Command" Project (2023AB078), the Ministry of Science and Technology High-end Foreign Expert Project (G2023014066L), the Xinjiang Production and Construction Corps Agricultural Science and Technology Innovation Engineering Special Project (NCG202232).

Author information

Authors and Affiliations

Authors

Contributions

MW conceived and coordinated this work. CZ and YZ wrote the review. YW and YZ collected information. JL and JM refined the grammar and structure of the review. XX checked the manuscript. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Mengzhi Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, C., Zhen, Y., Weng, Y. et al. Research progress on the microbial metabolism and transport of polyamines and their roles in animal gut homeostasis. J Animal Sci Biotechnol 16, 57 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-025-01193-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-025-01193-x

Keywords