Abstract
Polyamines are small organic molecules ubiquitously present in all living organisms and function as crucial regulators of biological processes ranging from fundamental cellular metabolism to immune regulation. Dysregulation of polyamine metabolism has been implicated in numerous diseases, including neurodegenerative disorders, inflammatory conditions, autoimmune diseases, and cancer. This review provides an overview of pathophysiology of these conditions, highlighting polyamines’ role in immunometabolic alterations in the context of immune regulation. Exploring the intricate mechanisms of polyamine metabolism holds promise for advancing our understanding of disease processes and developing potential innovative therapeutic interventions.
Keywords
Polyamines, HIV, Immune regulation, Immunometabolism, Tumorigenesis, Cancer
Introduction
Polyamines are polycationic molecules containing variable hydrocarbon chains with multiple primary amino groups (-NH2) [1-4]. These occur either as diamines (putrescine, cadaverine, agmatine, 1,3-diaminopropane), polyamines (spermidine, spermine, thermospermine, caldine, thermine), or polyamine conjugates (hypusine, glutathionylspermidine) [5,6]. Polyamine synthesis essentially occurs in almost all cell types, with higher activity of the pathway in rapidly proliferating cells such as epithelial cells of the gastrointestinal tract, various cancers, neurons, glial cells (synthesized in neurons but preferentially stored in astrocytes), immune cells and reproductive cells [7-11]. Because of their cationic properties at the physiological pH, they bind to acidic sites of the cellular components including nucleic acids, membranes, and proteins [4]. They engage in a myriad of cellular functions such as modulating enzyme activities, transcription, nucleic acid synthesis/stability, RNA modifications, regulating the gene expression both at transcriptional and translational levels, protein synthesis, regulation of ion channels, kinase activities, membrane/structure functions, cellular growth, and function [4,5,12-14]. Though eukaryotic cells proficiently synthesize putrescine, spermidine, and spermine, the three major polyamines, ingestion through the diet, and production by resident microbes also serve as physiologically relevant sources [13]. Variations in their levels or obstruction in the metabolism of polyamines undoubtedly contribute to diseases including cancer, inflammation, diabetes, atherosclerosis, Parkinson’s disease (PD), Alzheimer’s disease (AD), osteoporosis, osteoarthritis, sarcopenia, renal failure, and stroke [15,16]. This review provides an overview of polyamines underscoring their roles in the context of immune cell regulation in various settings.
Polyamine Metabolism
Polyamine homeostasis is regulated by a balance between anabolism, catabolism, and the shuttling of polyamines in uni- and multi-cellular organisms [17]. The concentration of intracellular polyamines is precisely regulated via its de novo synthesis, transport, and salvage pathways that coordinate polyamine homeostasis under physiological conditions. Polyamines are largely derived from standard amino acids, arginine, and methionine in two different stages in mammals [12,16]. The first critical step involves the synthesis of ornithine and agmatine from arginine through the activities of mitochondrial arginase and arginine decarboxylase (ADC) respectively. ADC is an alternate enzyme found in nervous tissue, liver, ovary, uterus, and placenta [18]. Polyamine synthesis normally occurs through decarboxylation of its precursor ornithine by a key and a rate-limiting enzyme ornithine decarboxylase (ODC), a pyridoxal-5′-phosphate forming putrescine, which is then converted into spermidine and spermine through respective activities of spermidine synthase and spermine synthase [2,19] (Figure 1A). Hypusination is a post-translational modification that occurs in eukaryotic translation initiation factor 5A (eIF5A) protein in a conserved lysine residue, a process that requires spermidine and enzymes deoxyhypusine synthase (DHS), and deoxyhypusine hydroxylase (DOHH) [20,21]. Inactivation of DHS pathway is detrimental to eukaryotic cell growth and limits tumor progression [22-27] (Figure 1B). Synthesis of spermidine and spermine also occurs through a methionine-mediated pathway (Figure 1A) [28]. Polyamine transportation differs across the species and cell types. In bacteria, it typically involves the ATP binding cassette transporters [29]. Polyamine transport occurs via heparin sulfate and glypican 1 (GPC1) system or through SLC3A2, a solute carrier family of type II transmembrane protein [30-33]. In the gastrointestinal tract, putrescine uptake is mediated through nitric oxide synthase (NOS2)-dependent caveolar endocytosis in the colon and small intestine [32]. Spermidine and spermine are additionally transported by a heterospecific cation transporter known as ornithine transcarbamylase 1-3 (OCT1-3) [34] (Figure 1). The backward conversion of spermine to spermidine, and subsequently into putrescine is catalyzed by spermine oxidase (SMOX) and polyamine oxidase (PAO) respectively [1]. PAO transforms N1-acetylspermine to spermidine by the catalytic activity of spermidine/spermine-N1-acetyltransferase (SSAT) [35,36]. Under normal physiological conditions, SSAT is at negligible concentrations but could increase with elevated levels of free polyamines, regulating negative feedback [37]. Abnormal synthesis or excessive degradation of polyamines leads to extremely toxic metabolite production such as aldehydes, ammonia, peroxides and acrolein (Figure 1C). These by-products are associated with pathophysiology in mammalian systems and are more toxic than reactive oxygen species (ROS) [38-40]. This catabolic conversion involves spontaneous deamination of 3-aminopropanal through the serum PAO and spermidine oxidases to generate the toxic byproduct acrolein [41-43]. In summary, these processes underscore how excessive synthesis or degradation of polyamines could trigger abnormal cellular viability and growth.
Figure 1. The polyamine metabolic pathways, transport, and eIF5A hypusination. Polyamine uptake by cells occurs through the cell surface proteoglycans heparin sulfate and glypican 1 (GPC1) system while putrescine transports through solute carrier SLC3A2. Spermidine and spermine efflux through the cells by ornithine transcarbamylase 1-3 (OCT1-3). The uptake could be inhibited by polyamine transport inhibitors (PTIs). (A) Polyamine synthesis involves two synthetic pathways either involving conversion of L-Arginine or methionine to polyamines. The first pathway involves the conversion of L-Arginine to L-Ornithine by arginase 1 with the release of urea or it is converted to nitric oxide and L-Citrulline by nitric oxide synthase (NOS). The excessive L-Citrulline acts as an allosteric inhibitor of arginase 1. Further, L-Ornithine is sequentially converted to the 3 major polyamines through a rate-limiting enzyme ornithine decarboxylase (ODC) and spermidine/spermine synthases. The ODC activity can be inhibited by difluoromethylornithine (DFMO). The proto-oncogene protein MYC functions as a trans-activator of ODC to enhance the proliferation of cancer cells and to induce tumorigenesis. The second pathway involves the conversion of methionine to polyamines through an ATP-dependent methionine adenosyl transferase 2 (MAT2) to form S-adenosylmethionine (SAMe) which is further decarboxylated to decarboxy SAMe through SAMe decarboxyalse and could be inhibited by methyl glyoxal-bis-guanidylhydrazone (MGBG) and SAM486A (CGP48664 or Sardomozide). (B) Multifaceted role of spermidine and hypusination of eIF5A. Hypusination occurs at a conserved lysine residue of eIF5A and is mediated by spermidine through the action of deoxyhypusine synthase (DHS) forming an inactive intermediate deoxyhypusine eIF5A which is further converted to an active hypusinated eIF5A by deoxyhypusine hydroxylase (DOHH). The active hypusinated eIF5A can be converted to an inactive form through acetylation at lysine 47 through P300/CBP-associated factor (PCAF) or by spermidine/spermine N’- acetyltransferase (SSAT) or back to its active form through deacetylation by histidine deacetylase 6 (HDAC6) or sirtuin-2 (SIRT2). (C) In vitro oxidative deamination of polyamines. The oxidative deamination causes release of toxic by-products such as aldehydes, ammonia, hydrogen peroxide and acrolein. Figure was generated using Microsoft® PowerPoint.
Methodologies Employed to Assess Polyamine Homeostasis
Current methodologies for assessing polyamine homeostasis involve HPLC (high-performance liquid chromatography), a highly reliable and sensitive method allowing for the detection of low concentrations of polyamines [44]. Techniques like LC-MS/MS (liquid chromatography-tandem mass spectrometry) provide accurate quantification and structural characterization of polyamines [45,46]. LC-MS is sensitive and specific, capable of detecting multiple polyamines simultaneously but requires specialized equipment and expertise. Also, a combination of HPLC-MS could be employed [47]. Other commonly used techniques are Western blotting and flow cytometry measuring the expression of enzymes involved in polyamine metabolism, such as ODC [48,49]. The caveats remain to be the inability to measure polyamine concentrations and variations based on antibody specificity and experimental conditions. Although RT-PCR (reverse transcription-polymerase chain reaction) is reliable for assessing gene expression, it does not provide direct measurements of polyamine levels. Metabolomics is also employed for comprehensive analysis of metabolic profiles using techniques like NMR or MS to assess polyamine levels among other metabolites which offers a holistic view of cellular metabolism but may require complex data analysis and interpretation. Each methodology has its advantages and limitations. The choice of method often depends on the specific research question and required sensitivity. Combining multiple approaches can enhance the reliability and comprehensiveness of polyamine homeostasis assessments within immune cells. Measuring polyamine levels in clinical contexts presents several challenges including sample variability arising due to diet, age, gender, health status, etc., also there are no standardized established reference values. Another important challenge would be the stability of polyamines in samples and proper sample handling and storage, which are critical for preventing degradation. Addressing these challenges is essential for the reliable measurement of polyamines in clinical practice, which could enhance their utility as biomarkers for various diseases.
Polyamines Impact Immunometabolism
Polyamines exhibit multifaceted roles in immune cells and can influence both innate and adaptive immune responses that include modulation of (1) immune cell proliferation and differentiation, (2) macrophage polarization, (3) immune senescence and autophagy, (4) cancer immunosuppression [10,49-52]. Although polyamines were initially considered to have immunosuppressive effects, it has now become apparent that their immunobiology is nuanced and context-dependent, based on the cell types and pathological conditions. Early research highlighted the potential role of polyamines in promoting autoimmune conditions. DFMO, an irreversible inhibitor of ODC, inhibits polyamine synthesis, reduce T cell proliferation, and alleviate lupus-like disease symptoms in mice [53]. More recent work by Wagner and colleagues demonstrates that polyamines can regulate the balance between Th17 cells and regulatory T cells (Tregs) [52]. Pathogenic Th17 cells exhibit upregulated polyamine metabolism, and pharmacological inhibition of ODC by DFMO, as well as genetic deficiency of Odc in mice (Odc-/-), lead to a reduction in these cells and disease severity [52]. Stimuli such as TCR activation, hypoxic conditions, stress inducers, and cytokines like IL-2, IL-17, and IL-15 promote the polyamine biosynthetic pathway in T cells. These stimuli regulate critical enzymes like ODC, SAMe decarboxylase, spermidine synthase, and spermine synthase by modulating transcription factors including c-Myc and HIF-1α. This transcriptional regulation ensures that the polyamine pathway supports T cell proliferation, differentiation, and functions differ slightly based on T cell differentiation states [47,48,54-57]. For example, polyamine synthesis can block Treg fate decisions and promote autoimmunity, while spermidine alone could induce anti-inflammatory responses by promoting Treg development [47]. The apparent contradictions in the results of these studies could be attributed to differences in Treg subsets investigated or combined effects of polyamines evaluated in one study versus just one of the polyamines in another. They could also be due to the origin of Tregs from different tissues, or the concentrations of cytokines and TCR stimulants that were engaged to induce naïve cell differentiation into Tregs, as the levels and effects of polyamines are likely different for naïve cells and differentiated Tregs. A recent study demonstrated that deficiency in polyamine synthesis and eIF5A hypusination led to a failure of CD4+ T cells to adopt the correct subset specification associated with perturbations in post-translational modification of eIF5A [48]. Loss of Odc during the development disrupted the fidelity of Th lineage differentiation, ectopic expression of lineage-defining transcription factors, and cytokines in differentiating T cells and caused intestinal inflammation. This study shows that polyamines have another layer of mechanistic regulation of naïve T cell differentiation into various T cell subsets and may be unique to the disease-specific milieu that drives T cell polarization. They indicate a complex role for polyamines in immune regulation, with the potential for both pro-inflammatory and anti-inflammatory effects on sterile inflammation and infections.
In an infection setting, an increase in ODC and polyamine synthesis in T cells parallel with heightened ratios of PD-1+FOXP3+ IFN-γ+ T cells, known as Treg-like Th1 cells that are known to be elevated at the sites of inflammation [49, 58, 59]. HIV-infected T cells display augmented polyamine synthesis, which is dependent on ODC, caspase-1, and IL-1β activity. Blocking the caspase-1 and ODC as well as hypusination reverses T cell aberrations caused by HIV infection. Corroborating these data, salivary putrescine from people living with HIV shows a positive correlation with dysregulated Treg/Th17 ratio and hyperactivation of mucosal T cells [49]. Treg-like Th1 cells are also enriched during Candida infection and are physiologically relevant to infection-caused immunopathology [60]. While polyamines can favor Candida proliferation and altering aminopropyl group acetylation levels and autophagic induction thereby causing host cellular dysfunction, their role in T cell functions remains to be investigated in Candida setting [61]. These studies suggest that local polyamines in tissues have an impact on T cell subtypes and their functions in mucosal infections. Polyamines have been implicated in promoting autoimmunity, partially by regulating Tregs [49,60]. While polyamines can also aggravate oxidative stress and impair immune tolerance and are implicated in IBD, celiac disease, peptic ulcers, the role of T cell-polyamines in directly triggering inflammation or autoimmunity in these contexts is unclear. Mechanistically, more evidence suggest that polyamines are associated with modulating inflammation, cellular proliferation, differentiation, and mucosal healing processes in the context of immune cells. However, the direct role of T cell polyamines remains to be studied [10,62,63].
Polyamines are also important regulators of several other immune cells (Figure 2A). B cell receptor activation increases the levels of polyamines and the expression of enzymes involved in the polyamine pathway. The ability of spermine to regulate activation-associated apoptosis suggests a protective role for polyamines in B-cell clonal deletion processes [64]. In dendritic cells (DCs), arginase-mediated polyamine synthesis induces indoleamine 2,3-dioxygenase (IDO) expression resulting in immunosuppression. IDO signaling in DCs is dependent on arginase 1, which induces IDO phosphorylation and its activation through the activation of Src kinase. Thus, polyamines released by IDO+ arginase 1+ bystander myeloid-derived suppressor cells (MDSCs) or DCs polarize DCs towards immunosuppressive phenotype in a paracrine manner [65] (Figure 2B). Also, macrophages synthesize polyamines both through ODC and arginase pathways. The classically activated M1 macrophages which typically produce nitric oxide via iNOS using arginine as the substrate have reduced arginine availability for polyamine synthesis [66]. However, IL-4 and IL-13-activated M2 macrophages with upregulated arginase 1 have increased polyamine synthesis. They also show ODC-dependent polyamine synthesis mediated by ERK, PI3K, and PKA pathways independent of arginase 1 activity (Figure 2B). Polyamines appear to have diverse roles in regulating macrophage functions depending on the context. Intrinsically synthesized polyamines suppress the pro-inflammatory cytokine expression in LPS-stimulated macrophages (M1 macrophages) [67]. For example, putrescine downregulates the transcription of M1 genes (Nos2 and Il-1β) through the formation of euchromatin. It also inhibits the activation of M1 macrophages by inhibiting NF-κB p65 activation and downregulating TNF-α and IL-8 expression [68-70]. However, Rac1 and actin-dependent import of spermine and spermidine, which is known to increase following efferocytosis stimulate IL-1β and IL-6 expression in LPS-induced macrophages, showing pro-inflammatory effects of intracellularly imported polyamines [68]. In the context of tumor environment, spermidine either intrinsically or in a paracrine manner favors M1 polarization through Nos2 transcription thereby inhibiting tumor growth, while spermine inhibits M1 but promotes M2 polarization, through enhancing autophagy by ATG5 upregulation [66]. Together, these studies underscore the complex and context-dependent role of polyamine metabolism in regulating T cell-mediated and T cell-independent immune responses.
Apart from the dietary arginine, gut microbiota serves as an alternative source of polyamines [13]. The gut microbiome and polyamines are intricately linked, with polyamines playing critical roles in both microbial and host physiology. Certain gut bacteria, including members of the genera Lactobacillus, Bifidobacterium, and Enterococcus are known to produce polyamines [71,72]. Germ-free mice exhibit significantly lower polyamine levels compared to wild type emphasizing microbiome’s role in polyamine synthesis [73]. The gut microbiota converts the dietary and host-derived amino acids to polyamines, thereby increasing local and systemic polyamine levels [74]. Resected colorectal cancer tissues from patients treated with antibiotics display decreased levels of polyamine metabolites indicating the role of gut bacteria in contributing to the human polyamine metabolite pool [75]. Therefore, disruptions in microbiome composition can alter polyamine levels, potentially affecting intestinal integrity and immune responses. Polyamines can enhance bacterial survival in the face of antibiotic treatment for an infection [76]. They bind to macromolecules targeted by antibiotics and reduce their availability for drugs [77]. They modulate outer membrane permeability in bacteria through extensive inter-lipid hydrogen bonding and membrane stabilization, and permeability of porin channels, impacting bacterial adhesion and colonization rates [76]. Polyamines are also shown to increase bacterial susceptibility to various antibiotics [78]. While these studies are of considerable interest, more investigations are needed to further learn how antibiotics causing dysbiosis can alter host responses by affecting microbial polyamine metabolism and how host polyamines directly affect host-microbe interactions. Gut polyamines promote the integrity of intestinal epithelium and lessen the macrophage pro-inflammatory cytokine production [79]. Several studies have demonstrated that polyamine administration improves the health of intestinal resident mucosal immune cells [13,47,80]. However, there is still much to uncover in terms of the biological mechanisms of mucosal polyamines and the full extent of their actions in the context of the microbiome. There could also be intrinsic differences in polyamine effects on mature or pathological Th subsets, and newly activated naïve cells in normal versus infection settings. These contextual changes may also be attributed to the levels of intracellular polyamines versus extracellularly available polyamines and the balance between synthesis enzymes and polyamine oxidases. Because polyamines interact with multiple cellular targets, it is challenging to narrow down their specific roles in different biological contexts. However, unraveling key mechanisms could lead to a deeper understanding of how polyamines contribute to intestinal health, immune regulation, and disease pathogenesis.
Figure 2. Schematic representation of pleiotropic effects of polyamine-induced immunometabolic changes and associated signaling pathways. (A) Polyamine immunometabolism pathways impact various cellular processes and could be manipulated by either blocking the pathway with inhibitors or uptake by polyamine transport inhibitors. Upward and Downward red arrows indicate enhanced or decreased protein expression respectively. (B) Modulation of polyamine-induced signaling pathways in immune cells. TCR: T cell Receptor; HIF- 1α: Hypoxia-inducible Factor-1α; ODC: Ornithine Decarboxylase; ATG5: Autophagy-related Gene 5; mTOR: Mammalian Target of Rapamycin; Nos2: Nitric Oxide Synthase 2; eIF5A: Eukaryotic Translation Initiation Factor 5A; DC: Dendritic Cells; IDO1: Indoleamine 2,3-Ddioxygenase 1; Arg: Arginase); MDSCs: Myeloid-Derived Suppressor Cells.
Polyamines in Immune Regulation and Neuroinflammation
Immune dysregulation may lead to inflammation in the central nervous system and is implicated in the pathogenesis of neurodegenerative diseases such as AD, PD, and age-related cognitive decline [81,82]. Immune cells in the brain, such as microglia, play a key role in neuroinflammation by producing inflammatory mediators in response to various stimuli, including misfolded proteins and oxidative stress [83].
Spermidine can effectively act as a free-radical scavenger and curb the excessive generation of ROS including superoxide, hydroxyl, peroxyl radicals, nitric oxide, peroxynitrite, etc [84]. It also reduces the malondialdehyde formation, an indicator of lipid peroxidation [85,86]. Spermidine is also known to activate mTOR and AMPK pathways and delay brain aging through autophagy induction. Along with the enhanced expression of neurotrophic factors, it also enhances the antioxidant enzyme (activating Keap1-Nrf2-ARE antioxidant signaling pathway), maintains energy of neurons (through mitochondrial ATP production), inhibits apoptosis (by activating autophagy) and limits inflammation (by inducing M2 macrophages) [87,88] (Figure 2B). Optimal polyamine levels are crucial for neuronal replication and maintenance and are synthesized in neurons and shuttled by glial cells [89]. Agmatine produced in the brain, acts as a neurotransmitter and suppresses excessive nitric oxide production, mitigating hypoxic-ischemic brain injury in neonatal rats [90-92]. Spermidine and spermine enhance glutamate and glycine binding to NMDA receptors. The binding of spermidine and spermine acts as a positive allosteric modulator of NMDA receptors thereby enhancing glutamate and glycine binding while putrescine inhibits the process [93,94]. Since these are ionotropic receptors crucial for synaptic transmission and plasticity in the central nervous system, the exogenous addition of putrescine induces potent convulsion and neuropathological lesions in rodents. Therefore, systemic injection of a high dose putrescine (more than the physiological dose of 200 mg/kg) is known to induce a characteristic toxic response in rats [95]. Spermidine inhibits cellular senescence in neuronal cell lines through modulating mitochondrial functions and is known to improve the cognitive functions in mice and Drosophila [96-99]. However, excessive production of putrescine in the brain is known to be involved in seizures [100]. Also, AD is associated with significantly heightened spermidine and spermine levels [101-103]. Cellular polyamines are capable of binding to beta-amyloid (Aβ) plaque peptides and further promote their aggregation and successive memory loss [104]. Moreover, arginine deprivation due to hyperactivation of arginase can also contribute to neurodegeneration [28,105]. Arginine deprivation promotes oxidative and chronic maladaptive polyamine stress response (PSR), thereby accelerating the conversion of ornithine to putrescine, elevating polyamine levels and igniting the neurodegeneration cycle. Additionally, the expression levels of SSAT and PAO are high in the AD brain, suggesting a predominant role for polyamine oxidation in the neurodegenerative process. Thus, glutamate receptors, calcium dynamics, PSR, and polyamine oxidation play crucial roles in and AD pathogenesis [105-109]. Polyamine pathway alteration also leads to the aggregation of α-synuclein in intraneuronal inclusions and cellular dysfunction in PD [110]. Mechanistically, loss of function in the lysosomal polyamine exporter ATP13A2 is responsible for the accumulation of lysosomal polyamines and reducing their availability to mitochondria thereby impacting the mitochondrial functionality [110]. Cells with ATP13A2 deficiency have a higher pH and compromised degradative capacity and the addition of spermine leads to the rupture of lysosomes, release of cathepsin B, and neuronal cell death [111]. While higher acetylated polyamines are reported in the serum, putrescine levels are increased in the cerebrospinal fluid of PD patients, with those exhibiting worse phenotypes showing drastically reduced spermidine levels [112,113]. However, the functional significance of these studies is unclear. Given the significant role of T cells in neuroinflammation [114,115] (reviewed elsewhere) it is tempting to speculate how aforementioned polyamine-dependent T cell dysfunction could contribute to neuroinflammation, although the studies on exact interactions between T cells and neuronal cells in the context of polyamine metabolism remain to be done. However, modulating polyamine metabolism presents a potential therapeutic strategy for managing neuroinflammation.
Polyamines as Immunomodulators and Molecular Drivers of Tumorigenesis
Polyamines, also function as oncometabolites, promote immune suppression, and are often correlated with tumor growth and progression [69,116]. Also, as reviewed elsewhere, tumor-derived polyamines favor M2 macrophages and other immunosuppressive cells favoring further tumor growth [117]. Paracrine effects of M2 macrophage-derived polyamines suppress the activity of T cells and DC and modulate tissue repair and remodeling by promoting the proliferation and migration of fibroblasts and endothelial cells in the tumor microenvironment [67]. Increased tumor polyamine synthesis also decreases IL-12 and IFN-γ levels in immune cells ultimately inhibiting NKT cell’s cytotoxic functions. Moreover, polyamines also contribute to tumor progression by directly enhancing cell adhesion, ECM remodeling, and angiogenesis. A direct relationship between ODC, a transcriptional target and a trans-activator of the MYC is well established [57,118] (Figure 1A). Therefore, polyamine-associated cancer cell growth may involve MYC signaling pathway in a wide range of cancers. Elevated ODC activity, polyamine biosynthesis, and high uptake of polyamines in tumor cells, particularly in rapidly growing tumors are not only considered prognostic markers for cancers but also pro-tumorigenic [116,119]. Studies have reported that tumor cells have elevated polyamine levels due to increased synthesis and uptake/transport with reduced catabolism. Additionally, cancer cells dependent on constant, higher intracellular levels of polyamine pools to provide persistent proliferation [1]. Several oncogenes including MYC, KRAS, JUN, BRAF and FOS are associated with polyamine dependent cellular dysregulation [120-122]. Furthermore, genetic polymorphisms in ODC are often associated with some neuroblastomas, colorectal, gastric, prostate, and breast cancers [123, 124]. Blockade of ODC activity in intestinal epithelial cells inhibits TGF-β1 mediated-SMAD signaling pathway and reduces tumor growth and vascularization [125,126]. Also, DFMO blocks the activation of p38, ERK, and AKT/mTOR/p70S6K induced by N-nitrosomethylbenzylamine in a rat model of esophageal squamous cell cancer [119]. Owing to its low toxicity and oral administration, DFMO has been used in several clinical trials to treat various cancers such as lung, pancreatic, neuroblastoma, endometrial, gastric, and osteosarcoma [127,128]. Because polyamine oxidative catabolic products and oxidative stress can enhance ODC activity and tumor cell growth, these byproducts have also been implicated in various cancers, including head and neck, gastric, lung, colorectal, breast, and prostate, as well as in several other cancer cell lines [116,119,129]. Hypusinated eIF5A is required for the malignant transformation into lymphoma in MYC-overexpressed B cells [130]. In the context of immune cells in the tumor microenvironment, polyamines enhance Ca2+ accumulation in mitochondria thereby promoting T cell activation [131,132]. A recent study by Al-Habsi and co-workers demonstrated that reduced levels of spermidine in CD8+ T cells from aged mice correlated with their non-responsiveness to PD-1 antibody therapy [133]. Spermidine supplementation, likely by its ability to bind to the mitochondrial trifunctional protein, a β–oxidation enzyme, and increase ATP production in CD8+ T cells, enhanced the anti-tumor immunity. Polyamine/hypusine axis, however, downmodulates tissue-resident memory T cell (Trm) differentiation [134]. Inhibition of this axis enhances IFN-γ and TNF-α production upon activation of both mouse and human CD8+ T cells and increases TGF-β induced differentiation of CD69+CD103+ Trm cells. Hypusination of eIF5A induces transcription of mitochondrial genes and when disrupted, greatly reduces oxygen consumption by modulating oxidative phosphorylation and mitochondrial functions in the macrophages [131]. The reasons for these disparate effects of polyamines in cancer in relevance to their direct effects on tumors versus indirect effects on immune cells and immunosurveillance versus regulation are to be explored in the future.
Several clinical trials are underway targeting polyamine pathways. The inclusion of DFMO reduced the tumor size in a mouse xenograft model of neuroblastoma [31]. A recent phase II clinical trial concluded that high-risk neuroblastoma patients receiving DFMO showed a substantial increase in overall survival compared to subjects without DFMO treatment. Earlier addition of DFMO in the maintenance therapy in amalgamation with immunotherapy and cis-retinoic acid could potentiate the therapeutic effect [135]. This has led to the initiation of a number of clinical trials NCT02395666, NCT04301843, NCT02679144, NCT02395666, NCT05717153 [135,136]. Furthermore, blockade of polyamines primes an increase in infiltration of CD8+ T cell and an inflammatory phenotype in tumors [137-141].
Synthetic polyamine analogs are being created to disrupt polyamine function, offering potential treatments for autoimmune conditions [1,142]. However, challenges such as systemic toxicity, the complex role of polyamines in immune regulation, and compensatory cellular mechanisms need to be addressed. More research is required to develop targeted therapies that selectively modulate polyamine metabolism in various diseases without affecting healthy cells and normal physiological functions.
Concluding Remarks
Recent research underscores the pivotal role of polyamines in immune regulation, revealing that they enhance the activation and proliferation of T cells while influencing the function of macrophages and dendritic cells [47-49,52,56,59,117]. Overall, dysregulation of polyamine metabolism can influence disease progression through several mechanisms. Polyamines, particularly spermidine and spermine, are crucial for cellular growth and differentiation. Elevated levels can promote cell proliferation and inhibit apoptosis, contributing to cancer progression by allowing malignant cells to thrive [143,144]. Polyamines stabilize nucleic acids and modulate transcription factors, affecting the expression of genes involved in cell cycle regulation, survival, and inflammation [145,146]. The resulting immune cell changes and heightened recruitment of immune cells that suppress anti-tumor responses by supporting MDSCs, macrophages and Tregs could affect the tumor microenvironment by promoting angiogenesis and immune evasion [69]. Additionally, polyamines interact with gut microbiota, impacting systemic immune responses and speculating connections between gut health and autoimmune conditions [13,147]. Moreover, targeting polyamine metabolism with inhibitors such as DFMO is emerging as a promising therapeutic approach, particularly for cancer treatment and immune modulation. Recent research suggests that polyamines may also be involved in immunosenescence, affecting immune cell functionality in older adults through autophagy [96,148,149]. Collectively, these findings emphasize the importance of polyamines as critical regulators of immune responses and potential therapeutic targets in various diseases.
Future Research Directions and Unanswered Questions
The field of polyamine immunobiology continues to evolve, with ongoing discoveries shedding light on new aspects of their involvement in regulation of immunometabolism and leading to therapeutic interventions. Several questions remain unaddressed: 1) How can therapies be designed to selectively target polyamine metabolism in immune cells in the context of above diseases and immunometabolism? 2) How do polyamines precisely regulate immunometabolism, and how do metabolic shifts in immune cells influence outcomes in different disease environments? 3) How do polyamines produced by gut microbiota interact with host immune regulation and contribute to mucosal diseases? 4) What are reliable biomarkers to monitor polyamine activity and guide therapeutic interventions? 5) How do cells develop resistance to therapies targeting polyamine metabolism, and what compensatory pathways are activated? 6) What is the optimal timing and dosage for polyamine-targeted therapies in various diseases? Addressing these queries will further advance our understanding of polyamine-mediated immune processes and unlock new therapeutic strategies for treating cancers, autoimmune, and inflammatory diseases.
Conflict of Interest
The authors declare that they have no conflict of interest.
Funding
This work was supported by an NIH/NIDCR grant R01 DE026923 to P.P.
Author Contributions
P.P. drafted the framework for the review. S.S.M. wrote the preliminary versions, revised the drafts and created illustrations. P.P. revised and edited the final version of the manuscript. The authors have read and agreed to the published version of the manuscript.
References
2. Sagar NA, Tarafdar S, Agarwal S, Tarafdar A, Sharma S. Polyamines: functions, metabolism, and role in human disease management. Medical Sciences. 2021 Jun 9;9(2):44.
3. Holbert CE, Cullen MT, Casero RA Jr, Stewart TM. Polyamines in cancer: integrating organismal metabolism and antitumour immunity. Nat Rev Cancer. 2022 Aug;22(8):467-80.
4. Pegg AE. Mammalian polyamine metabolism and function. IUBMB Life. 2009 Sep;61(9):880-94.
5. Xuan M, Gu X, Li J, Huang D, Xue C, He Y. Polyamines: their significance for maintaining health and contributing to diseases. Cell Commun Signal. 2023 Dec 4;21(1):348.
6. Michael AJ. Biosynthesis of polyamines and polyamine-containing molecules. Biochem J. 2016 Aug 1;473(15):2315-29.
7. Soda K. The mechanisms by which polyamines accelerate tumor spread. J Exp Clin Cancer Res. 2011 Oct 11;30(1):95.
8. Takano K, Ogura M, Nakamura Y, Yoneda Y. Neuronal and glial responses to polyamines in the ischemic brain. Curr Neurovasc Res. 2005 Jul;2(3):213-23.
9. Skatchkov SN, Woodbury-Fariña MA, Eaton M. The role of glia in stress: polyamines and brain disorders. Psychiatr Clin North Am. 2014 Dec;37(4):653-78.
10. Hesterberg RS, Cleveland JL, Epling-Burnette PK. Role of Polyamines in Immune Cell Functions. Med Sci (Basel). 2018 Mar 8;6(1):22.
11. Lefèvre PL, Palin MF, Murphy BD. Polyamines on the reproductive landscape. Endocr Rev. 2011 Oct;32(5):694-712.
12. Li J, Meng Y, Wu X, Sun Y. Polyamines and related signaling pathways in cancer. Cancer Cell Int. 2020 Nov 5;20(1):539.
13. Tofalo R, Cocchi S, Suzzi G. Polyamines and Gut Microbiota. Front Nutr. 2019 Feb 25;6:16.
14. Pegg AE. Functions of Polyamines in Mammals. J Biol Chem. 2016 Jul 15;291(29):14904-12.
15. Minois N, Carmona-Gutierrez D, Madeo F. Polyamines in aging and disease. Aging (Albany NY). 2011 Aug;3(8):716-32.
16. Jimenez Gutierrez GE, Borbolla Jiménez FV, Muñoz LG, Tapia Guerrero YS, Murillo Melo NM, Cristóbal-Luna JM, et al. The Molecular Role of Polyamines in Age-Related Diseases: An Update. Int J Mol Sci. 2023 Nov 17;24(22):16469.
17. 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 Oct 23;427(21):3389-406.
18. Wang X, Ying W, Dunlap KA, Lin G, Satterfield MC, Burghardt RC, et al. Arginine decarboxylase and agmatinase: an alternative pathway for de novo biosynthesis of polyamines for development of mammalian conceptuses. Biol Reprod. 2014 Apr 25;90(4):84.
19. Kapfhamer D, McKenna J, Yoon CJ, Murray-Stewart T, Casero RA, Gambello MJ. Ornithine decarboxylase, the rate-limiting enzyme of polyamine synthesis, modifies brain pathology in a mouse model of tuberous sclerosis complex. Hum Mol Genet. 2020 Aug 11;29(14):2395-407.
20. Wątor E, Wilk P, Biela A, Rawski M, Zak KM, Steinchen W, et al. Cryo-EM structure of human eIF5A-DHS complex reveals the molecular basis of hypusination-associated neurodegenerative disorders. Nat Commun. 2023 Mar 27;14(1):1698.
21. Ziegler A, Steindl K, Hanner AS, Kar RK, Prouteau C, Boland A, et al. Bi-allelic variants in DOHH, catalyzing the last step of hypusine biosynthesis, are associated with a neurodevelopmental disorder. Am J Hum Genet. 2022 Aug 4;109(8):1549-58.
22. Park MH, Wolff EC. Hypusine, a polyamine-derived amino acid critical for eukaryotic translation. J Biol Chem. 2018 Nov 30;293(48):18710-8.
23. McKenna S. The first step of hypusination. Nat Chem Biol. 2023 Jun;19(6):664.
24. Tauc M, Cougnon M, Carcy R, Melis N, Hauet T, Pellerin L, et al. The eukaryotic initiation factor 5A (eIF5A1), the molecule, mechanisms and recent insights into the pathophysiological roles. Cell Biosci. 2021 Dec 24;11(1):219.
25. Gobert AP, Smith TM, Latour YL, Asim M, Barry DP, Allaman MM, et al. Hypusination Maintains Intestinal Homeostasis and Prevents Colitis and Carcinogenesis by Enhancing Aldehyde Detoxification. Gastroenterology. 2023 Sep;165(3):656-69.e8.
26. Guo JS, Liu KL, Qin YX, Hou L, Jian LY, Yang YH, et al. Hypusination-induced DHPS/eIF5A pathway as a new therapeutic strategy for human diseases: A mechanistic review and structural classification of DHPS inhibitors. Biomed Pharmacother. 2023 Nov;167:115440.
27. Coni S, Serrao SM, Yurtsever ZN, Di Magno L, Bordone R, Bertani C, et al. Blockade of EIF5A hypusination limits colorectal cancer growth by inhibiting MYC elongation. Cell Death Dis. 2020 Dec 10;11(12):1045.
28. Malpica-Nieves CJ, Rivera-Aponte DE, Tejeda-Bayron FA, Mayor AM, Phanstiel O, Veh RW, et al. The involvement of polyamine uptake and synthesis pathways in the proliferation of neonatal astrocytes. Amino Acids. 2020 Aug;52(8):1169-80.
29. Potter AJ, Paton JC. Spermidine biosynthesis and transport modulate pneumococcal autolysis. J Bacteriol. 2014 Oct;196(20):3556-61.
30. Uemura T, Yerushalmi HF, Tsaprailis G, Stringer DE, Pastorian KE, Hawel L 3rd, et al. Identification and characterization of a diamine exporter in colon epithelial cells. J Biol Chem. 2008 Sep 26;283(39):26428-35.
31. Gamble LD, Purgato S, Murray J, Xiao L, Yu DMT, Hanssen KM, et al. Inhibition of polyamine synthesis and uptake reduces tumor progression and prolongs survival in mouse models of neuroblastoma. Sci Transl Med. 2019 Jan 30;11(477):eaau1099.
32. Uemura T, Stringer DE, Blohm-Mangone KA, Gerner EW. Polyamine transport is mediated by both endocytic and solute carrier transport mechanisms in the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol. 2010 Aug;299(2):G517-22.
33. Uemura T, Gerner EW. Polyamine transport systems in mammalian cells and tissues. Methods Mol Biol. 2011;720:339-48.
34. Sala-Rabanal M, Li DC, Dake GR, Kurata HT, Inyushin M, Skatchkov SN, et al. Polyamine transport by the polyspecific organic cation transporters OCT1, OCT2, and OCT3. Mol Pharm. 2013 Apr 1;10(4):1450-8.
35. Henderson Pozzi M, Gawandi V, Fitzpatrick PF. pH dependence of a mammalian polyamine oxidase: insights into substrate specificity and the role of lysine 315. Biochemistry. 2009 Feb 24;48(7):1508-16.
36. Coleman CS, Stanley BA, Jones AD, Pegg AE. Spermidine/spermine-N1-acetyltransferase-2 (SSAT2) acetylates thialysine and is not involved in polyamine metabolism. Biochem J. 2004 Nov 15;384(Pt 1):139-48.
37. Casero RA, Pegg AE. Polyamine catabolism and disease. Biochem J. 2009 Jul 15;421(3):323-38.
38. Igarashi K, Uemura T, Kashiwagi K. Acrolein toxicity at advanced age: present and future. Amino Acids. 2018 Feb;50(2):217-28.
39. Sánchez-Jiménez F, Medina MÁ, Villalobos-Rueda L, Urdiales JL. Polyamines in mammalian pathophysiology. Cellular and Molecular Life Sciences. 2019 Oct;76(20):3987-4008.
40. Igarashi K, Uemura T, Kashiwagi K. Assessing acrolein for determination of the severity of brain stroke, dementia, renal failure, and Sjögren's syndrome. Amino Acids. 2020 Feb;52(2):119-27.
41. Sakamoto A, Sahara J, Kawai G, Yamamoto K, Ishihama A, Uemura T, et al. Cytotoxic Mechanism of Excess Polyamines Functions through Translational Repression of Specific Proteins Encoded by Polyamine Modulon. Int J Mol Sci. 2020 Mar 31;21(7):2406.
42. Pegg AE. Toxicity of polyamines and their metabolic products. Chem Res Toxicol. 2013 Dec 16;26(12):1782-800.
43. Stewart TM, Dunston TT, Woster PM, Casero RA. Polyamine catabolism and oxidative damage. Journal of Biological Chemistry. 2018 Nov 30;293(48):18736-45.
44. Sah P, Zenewicz LA. The Polyamine Putrescine Is a Positive Regulator of Group 3 Innate Lymphocyte Activation. Immunohorizons. 2023 Jan 1;7(1):41-8.
45. Samarra I, Ramos-Molina B, Queipo-Ortuño MI, Tinahones FJ, Arola L, Delpino-Rius A, et al. Gender-Related Differences on Polyamine Metabolome in Liquid Biopsies by a Simple and Sensitive Two-Step Liquid-Liquid Extraction and LC-MS/MS. Biomolecules. 2019 Nov 26;9(12):779.
46. Coradduzza D, Azara E, Medici S, Arru C, Solinas T, Madonia M, et al. A preliminary study procedure for detection of polyamines in plasma samples as a potential diagnostic tool in prostate cancer. J Chromatogr B Analyt Technol Biomed Life Sci. 2021 Jan 1;1162:122468.
47. Carriche GM, Almeida L, Stüve P, Velasquez L, Dhillon-LaBrooy A, Roy U, et al. Regulating T-cell differentiation through the polyamine spermidine. J Allergy Clin Immunol. 2021 Jan;147(1):335-48.e11.
48. Puleston DJ, Baixauli F, Sanin DE, Edwards-Hicks J, Villa M, Kabat AM, et al. Polyamine metabolism is a central determinant of helper T cell lineage fidelity. Cell. 2021 Aug 5;184(16):4186-202.e20.
49. Mahalingam SS, Jayaraman S, Bhaskaran N, Schneider E, Faddoul F, Paes da Silva A, et al. Polyamine metabolism impacts T cell dysfunction in the oral mucosa of people living with HIV. Nat Commun. 2023 Jan 25;14(1):399.
50. Chia TY, Zolp A, Miska J. Polyamine Immunometabolism: Central Regulators of Inflammation, Cancer and Autoimmunity. Cells. 2022 Mar 5;11(5):896.
51. Chamoto K, Zhang B, Tajima M, Honjo T, Fagarasan S. Spermidine - an old molecule with a new age-defying immune function. Trends Cell Biol. 2024 May;34(5):363-70.
52. Wagner A, Wang C, Fessler J, DeTomaso D, Avila-Pacheco J, Kaminski J, et al. Metabolic modeling of single Th17 cells reveals regulators of autoimmunity. Cell. 2021 Aug 5;184(16):4168-85.e21.
53. Claverie N, Pasquali JL, Mamont PS, Danzin C, Weil-Bousson M, Siat M. Immunosuppressive effects of (2R,5R)-6-heptyne-2,5-diamine, an inhibitor of polyamine synthesis: II. Beneficial effects on the development of a lupus-like disease in MRL-lpr/lpr mice. Clin Exp Immunol. 1988 May;72(2):293-8.
54. Mandal S, Mandal A, Park MH. Depletion of the polyamines spermidine and spermine by overexpression of spermidine/spermine N¹-acetyltransferase 1 (SAT1) leads to mitochondria-mediated apoptosis in mammalian cells. Biochem J. 2015 Jun 15;468(3):435-47.
55. Stewart TM, Holbert CE, Casero RA Jr. Helping the helpers: polyamines help maintain helper T-cell lineage fidelity. Immunometabolism (Cobham). 2022 Aug 5;4(3):e00002.
56. 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 Dec 16;6(51):eabc4275.
57. Dong Y, Tu R, Liu H, Qing G. Regulation of cancer cell metabolism: oncogenic MYC in the driver's seat. Signal Transduct Target Ther. 2020 Jul 10;5(1):124.
58. Bhaskaran N, Schneider E, Faddoul F, Paes da Silva A, Asaad R, Talla A, et al. Oral immune dysfunction is associated with the expansion of FOXP3+PD-1+Amphiregulin+ T cells during HIV infection. Nat Commun. 2021 Aug 26;12(1):5143.
59. Liu X, Zhang W, Han Y, Cheng H, Liu Q, Ke S, et al. FOXP3+ regulatory T cell perturbation mediated by the IFNγ-STAT1-IFITM3 feedback loop is essential for anti-tumor immunity. Nat Commun. 2024 Jan 2;15(1):122.
60. Bhaskaran N, Faddoul F, Paes da Silva A, Jayaraman S, Schneider E, Mamileti P, et al. IL-1β-MyD88-mTOR Axis Promotes Immune-Protective IL-17A+Foxp3+ Cells During Mucosal Infection and Is Dysregulated With Aging. Front Immunol. 2020 Nov 6;11:595936.
61. Begum N, Lee S, Portlock TJ, Pellon A, Nasab SDS, Nielsen J, et al. Integrative functional analysis uncovers metabolic differences between Candida species. Commun Biol. 2022 Sep 26;5(1):1013.
62. Park MH, Igarashi K. Polyamines and their metabolites as diagnostic markers of human diseases. Biomol Ther (Seoul). 2013 Jan;21(1):1-9.
63. McNamara KM, Gobert AP, Wilson KT. The role of polyamines in gastric cancer. Oncogene. 2021 Jul;40(26):4399-412.
64. Nitta T, Igarashi K, Yamashita A, Yamamoto M, Yamamoto N. Involvement of polyamines in B cell receptor-mediated apoptosis: spermine functions as a negative modulator. Exp Cell Res. 2001 Apr 15;265(1):174-83.
65. Mondanelli G, Bianchi R, Pallotta MT, Orabona C, Albini E, Iacono A, et al. A Relay Pathway between Arginine and Tryptophan Metabolism Confers Immunosuppressive Properties on Dendritic Cells. Immunity. 2017 Feb 21;46(2):233-44.
66. Latour YL, Gobert AP, Wilson KT. The role of polyamines in the regulation of macrophage polarization and function. Amino Acids. 2020 Feb;52(2):151-60.
67. Van den Bossche J, Lamers WH, Koehler ES, Geuns JM, Alhonen L, Uimari A, et al. Pivotal Advance: Arginase-1-independent polyamine production stimulates the expression of IL-4-induced alternatively activated macrophage markers while inhibiting LPS-induced expression of inflammatory genes. J Leukoc Biol. 2012 May;91(5):685-99.
68. McCubbrey AL, McManus SA, McClendon JD, Thomas SM, Chatwin HB, Reisz JA, et al. Polyamine import and accumulation causes immunomodulation in macrophages engulfing apoptotic cells. Cell Rep. 2022 Jan 11;38(2):110222.
69. Lian J, Liang Y, Zhang H, Lan M, Ye Z, Lin B, et al. The role of polyamine metabolism in remodeling immune responses and blocking therapy within the tumor immune microenvironment. Front Immunol. 2022 Sep 2;13:912279.
70. 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 Sep 10;10:69.
71. 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.
72. 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 Mar 14;6:24.
73. 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.
74. Seiler N, Raul F. Polyamines and the intestinal tract. Crit Rev Clin Lab Sci. 2007;44(4):365-411.
75. Johnson CH, Dejea CM, Edler D, Hoang LT, Santidrian AF, Felding BH, et al. Metabolism links bacterial biofilms and colon carcinogenesis. Cell Metab. 2015 Jun 2;21(6):891-7.
76. Bhagwat AC, Saroj SD. Polyamine as a microenvironment factor in resistance to antibiotics. Critical Reviews in Microbiology. 2023 Jun 20:1-10.
77. Akhova AV, Tkachenko AG. Multifaceted role of polyamines in bacterial adaptation to antibiotic-mediated oxidative stress. The Microbiological Society of Korea. 2020 Jun 30;56(2):103-10.
78. Kwon DH, Lu CD. Polyamines increase antibiotic susceptibility in Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2006 May;50(5):1623-7.
79. Postler TS, Ghosh S. Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System. Cell Metab. 2017 Jul 5;26(1):110-30.
80. 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 Apr 8;12(1):2105.
81. Di Benedetto S, Müller L, Wenger E, Düzel S, Pawelec G. Contribution of neuroinflammation and immunity to brain aging and the mitigating effects of physical and cognitive interventions. Neurosci Biobehav Rev. 2017 Apr;75:114-128.
82. Andronie-Cioara FL, Ardelean AI, Nistor-Cseppento CD, Jurcau A, Jurcau MC, Pascalau N, et al. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer's Disease Progression. Int J Mol Sci. 2023 Jan 18;24(3):1869.
83. d'Avila JC, Siqueira LD, Mazeraud A, Azevedo EP, Foguel D, Castro-Faria-Neto HC, et al. Age-related cognitive impairment is associated with long-term neuroinflammation and oxidative stress in a mouse model of episodic systemic inflammation. J Neuroinflammation. 2018 Jan 30;15(1):28.
84. M.P. Murphy, H. Bayir, V. Belousov, C.J. Chang, K.J.A. Davies, M.J. Davies, et al. Guidelines for 642
85. 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 Jan 16;14:1292984.
86. Jiang D, Sun Q, Jiang Y, Zhou X, Kang L, Wang Z, et al. Effects of exogenous spermidine on autophagy and antioxidant capacity in ovaries and granulosa cells of Sichuan white geese. J Anim Sci. 2023 Jan 3;101:skad301.
87. Xu TT, Li H, Dai Z, Lau GK, Li BY, Zhu WL, et al. Spermidine and spermine delay brain aging by inducing autophagy in SAMP8 mice. Aging (Albany NY). 2020 Apr 8;12(7):6401-14.
88. Jiang D, Guo Y, Niu C, Long S, Jiang Y, Wang Z, et al. Exploration of the Antioxidant Effect of Spermidine on the Ovary and Screening and Identification of Differentially Expressed Proteins. Int J Mol Sci. 2023 Mar 17;24(6):5793.
89. Cervelli M, Averna M, Vergani L, Pedrazzi M, Amato S, Fiorucci C, et al. The Involvement of Polyamines Catabolism in the Crosstalk between Neurons and Astrocytes in Neurodegeneration. Biomedicines. 2022 Jul 21;10(7):1756.
90. Feng Y, Piletz JE, Leblanc MH. Agmatine suppresses nitric oxide production and attenuates hypoxic-ischemic brain injury in neonatal rats. Pediatr Res. 2002 Oct;52(4):606-11.
91. Regunathan S, Reis DJ. Characterization of arginine decarboxylase in rat brain and liver: distinction from ornithine decarboxylase. J Neurochem. 2000 May;74(5):2201-8.
92. Iyo AH, Zhu MY, Ordway GA, Regunathan S. Expression of arginine decarboxylase in brain regions and neuronal cells. J Neurochem. 2006 Feb;96(4):1042-50.
93. Pritchard GA, Fahey JM, Minocha SC, Conaty C, Miller LG. Polyamine potentiation and inhibition of NMDA-mediated increases of intracellular free Ca2+ in cultured chick cortical neurons. Eur J Pharmacol. 1994 Jan 15;266(2):107-15.
94. Mony L, Zhu S, Carvalho S, Paoletti P. Molecular basis of positive allosteric modulation of GluN2B NMDA receptors by polyamines. EMBO J. 2011 Jun 17;30(15):3134-46.
95. de Vera N, Serratosa J, Artigas F, Martínez E. Toxic effects of putrescine in rat brain: Polyamines can be involved in the action of excitotoxins. Amino Acids. 1992 Oct;3(3):261-9.
96. Hofer SJ, Simon AK, Bergmann M, Eisenberg T, Kroemer G, Madeo F. Mechanisms of spermidine-induced autophagy and geroprotection. Nat Aging. 2022 Dec;2(12):1112-29.
97. Jing YH, Yan JL, Wang QJ, Chen HC, Ma XZ, Yin J, et al. Spermidine ameliorates the neuronal aging by improving the mitochondrial function in vitro. Exp Gerontol. 2018 Jul 15;108:77-86.
98. S. Schroeder, S.J. Hofer, A. Zimmermann, R. Pechlaner, C. Dammbrueck, T. Pendl, et al. Dietary spermidine improves cognitive function. Cell Rep. 2021 Apr 13;35(2):108985.
99. S.J. Hofer, Y. Liang, A. Zimmermann, S. Schroeder, J. Dengjel, G. Kroemer, T et al. Spermidine-induced hypusination preserves mitochondrial and 689 cognitive function during aging. Autophagy 17 (2021) 2037-39.
100. Halonen T, Sivenius J, Miettinen R, Halmekytö M, Kauppinen R, Sinervirta R, et al. Elevated seizure threshold and impaired spatial learning in transgenic mice with putrescine overproduction in the brain. Eur J Neurosci. 1993 Sep 1;5(9):1233-9.
101. Luo J, Yu CH, Yu H, Borstnar R, Kamerlin SC, Gräslund A, et al. Cellular polyamines promote amyloid-beta (Aβ) peptide fibrillation and modulate the aggregation pathways. ACS Chem Neurosci. 2013 Mar 20;4(3):454-62.
102. noue K, Tsutsui H, Akatsu H, Hashizume Y, Matsukawa N, Yamamoto T, et al. Metabolic profiling of Alzheimer's disease brains. Sci Rep. 2013;3:2364.
103. Morrison LD, Kish SJ. Brain polyamine levels are altered in Alzheimer's disease. Neurosci Lett. 1995 Sep 1;197(1):5-8.
104. Hampel H, Hardy J, Blennow K, Chen C, Perry G, Kim SH, et al. The Amyloid-β Pathway in Alzheimer's Disease. Mol Psychiatry. 2021 Oct;26(10):5481-503.
105. Polis B, Karasik D, Samson AO. Alzheimer's disease as a chronic maladaptive polyamine stress response. Aging (Albany NY). 2021 Apr 3;13(7):10770-95.
106. Sandusky-Beltran LA, Kovalenko A, Placides DS, Ratnasamy K, Ma C, Hunt JB Jr, et al. Aberrant AZIN2 and polyamine metabolism precipitates tau neuropathology. J Clin Invest. 2021 Feb 15;131(4):e126299.
107. Morrison LD, Cao XC, Kish SJ. Ornithine decarboxylase in human brain: influence of aging, regional distribution, and Alzheimer's disease. J Neurochem. 1998 Jul;71(1):288-94.
108. Kindy MS, Hu Y, Dempsey RJ. Blockade of ornithine decarboxylase enzyme protects against ischemic brain damage. J Cereb Blood Flow Metab. 1994 Nov;14(6):1040-5.
109. 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 Jan 24;17(1):e1003012.
110. Vrijsen S, Houdou M, Cascalho A, Eggermont J, Vangheluwe P. Polyamines in Parkinson's Disease: Balancing Between Neurotoxicity and Neuroprotection. Annu Rev Biochem. 2023 Jun 20;92:435-64.
111. van Veen S, Martin S, Van den Haute C, Benoy V, Lyons J, Vanhoutte R, et al. ATP13A2 deficiency disrupts lysosomal polyamine export. Nature. 2020 Feb;578(7795):419-24.
112. Paik MJ, Ahn YH, Lee PH, Kang H, Park CB, Choi S, et al. Polyamine patterns in the cerebrospinal fluid of patients with Parkinson's disease and multiple system atrophy. Clin Chim Acta. 2010 Oct 9;411(19-20):1532-5.
113. Saiki S, Sasazawa Y, Fujimaki M, Kamagata K, Kaga N, Taka H, et al. A metabolic profile of polyamines in parkinson disease: A promising biomarker. Ann Neurol. 2019 Aug;86(2):251-63.
114. Chen X, Firulyova M, Manis M, Herz J, Smirnov I, Aladyeva E, et al. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature. 2023 Mar;615(7953):668-77.
115. Dai L, Shen Y. Insights into T-cell dysfunction in Alzheimer's disease. Aging Cell. 2021 Dec;20(12):e13511.
116. Novita Sari I, Setiawan T, Seock Kim K, Toni Wijaya Y, Won Cho K, Young Kwon H. Metabolism and function of polyamines in cancer progression. Cancer Lett. 2021 Oct 28;519:91-104.
117. Holbert CE, Casero RA Jr, Stewart TM. Polyamines: the pivotal amines in influencing the tumor microenvironment. Discov Oncol. 2024 May 18;15(1):173.
118. Bello-Fernandez C, Packham G, Cleveland JL. The ornithine decarboxylase gene is a transcriptional target of c-Myc. Proc Natl Acad Sci U S A. 1993 Aug 15;90(16):7804-8.
119. Xie Y, Dong CD, Wu Q, Jiang Y, Yao K, Zhang J, et al. Ornithine decarboxylase inhibition downregulates multiple pathways involved in the formation of precancerous lesions of esophageal squamous cell cancer. Mol Carcinog. 2020 Feb;59(2):215-26.
120. Peters MC, Minton A, Phanstiel Iv O, Gilmour SK. A Novel Polyamine-Targeted Therapy for BRAF Mutant Melanoma Tumors. Med Sci (Basel). 2018 Jan 5;6(1):3.
121. Alexander ET, El Naggar O, Fahey E, Mariner K, Donnelly J, Wolfgang K, et al. Harnessing the polyamine transport system to treat BRAF inhibitor-resistant melanoma. Cancer Biol Ther. 2021 Mar 4;22(3):225-237.
122. Liu X, Ren B, Ren J, Gu M, You L, Zhao Y. The significant role of amino acid metabolic reprogramming in cancer. Cell Commun Signal. 2024 Jul 29;22(1):380.
123. Gamble LD, Purgato S, Henderson MJ, Di Giacomo S, Russell AJ, Pigini P, et al. A G316A Polymorphism in the Ornithine Decarboxylase Gene Promoter Modulates MYCN-Driven Childhood Neuroblastoma. Cancers (Basel). 2021 Apr 9;13(8):1807.
124. Hogarty MD, Ziegler DS, Franson A, Chi YY, Tsao-Wei D, Liu K, et al. Phase 1 study of high-dose DFMO, celecoxib, cyclophosphamide and topotecan for patients with relapsed neuroblastoma: a New Approaches to Neuroblastoma Therapy trial. Br J Cancer. 2024 Mar;130(5):788-97.
125. Kaminski BM, Loitsch SM, Ochs MJ, Reuter KC, Steinhilber D, Stein J, et al. Isothiocyanate sulforaphane inhibits protooncogenic ornithine decarboxylase activity in colorectal cancer cells via induction of the TGF-β/Smad signaling pathway. Mol Nutr Food Res. 2010 Oct;54(10):1486-96.
126. Liu L, Santora R, Rao JN, Guo X, Zou T, Zhang HM, et al. Activation of TGF-beta-Smad signaling pathway following polyamine depletion in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol. 2003 Nov;285(5):G1056-67.
127. Li QZ, Zuo ZW, Zhou ZR, Ji Y. Polyamine homeostasis-based strategies for cancer: The role of combination regimens. Eur J Pharmacol. 2021 Nov 5;910:174456.
128. Alexiou GA, Lianos GD, Ragos V, Galani V, Kyritsis AP. Difluoromethylornithine in cancer: new advances. Future Oncol. 2017 Apr;13(9):809-19.
129. DeFelice BC, Fiehn O, Belafsky P, Ditterich C, Moore M, Abouyared M, et al. Polyamine Metabolites as Biomarkers in Head and Neck Cancer Biofluids. Diagnostics (Basel). 2022 Mar 24;12(4):797.
130. Nakanishi S, Li J, Berglund AE, Kim Y, Zhang Y, Zhang L, et al. The Polyamine-Hypusine Circuit Controls an Oncogenic Translational Program Essential for Malignant Conversion in MYC-Driven Lymphoma. Blood Cancer Discov. 2023 Jul 5;4(4):294-317.
131. Puleston DJ, Buck MD, Klein Geltink RI, Kyle RL, Caputa G, O'Sullivan D, et al. Polyamines and eIF5A Hypusination Modulate Mitochondrial Respiration and Macrophage Activation. Cell Metab. 2019 Aug 6;30(2):352-63.e8.
132. Salvi M, Toninello A. Effects of polyamines on mitochondrial Ca(2+) transport. Biochim Biophys Acta. 2004 Mar 9;1661(2):113-24.
133. Al-Habsi M, Chamoto K, Matsumoto K, Nomura N, Zhang B, Sugiura Y, et al. Spermidine activates mitochondrial trifunctional protein and improves antitumor immunity in mice. Science. 2022 Oct 28;378(6618):eabj3510.
134. Elmarsafawi AG, Hesterberg RS, Fernandez MR, Yang C, Darville LN, Liu M, et al. Modulating the polyamine/hypusine axis controls generation of CD8+ tissue-resident memory T cells. JCI Insight. 2023 Sep 22;8(18):e169308.
135. Lewis EC, Kraveka JM, Ferguson W, Eslin D, Brown VI, Bergendahl G, et al. A subset analysis of a phase II trial evaluating the use of DFMO as maintenance therapy for high-risk neuroblastoma. Int J Cancer. 2020 Dec 1;147(11):3152-9.
136. Sholler GLS, Ferguson W, Bergendahl G, Bond JP, Neville K, Eslin D, et al. Maintenance DFMO Increases Survival in High Risk Neuroblastoma. Sci Rep. 2018 Sep 27;8(1):14445.
137. Miska J, Rashidi A, Lee-Chang C, Gao P, Lopez-Rosas A, Zhang P, et al. Polyamines drive myeloid cell survival by buffering intracellular pH to promote immunosuppression in glioblastoma. Sci Adv. 2021 Feb 17;7(8):eabc8929.
138. Hayes CS, Shicora AC, Keough MP, Snook AE, Burns MR, Gilmour SK. Polyamine-blocking therapy reverses immunosuppression in the tumor microenvironment. Cancer Immunol Res. 2014 Mar;2(3):274-85.
139. Alexander ET, Minton A, Peters MC, Phanstiel O 4th, Gilmour SK. A novel polyamine blockade therapy activates an anti-tumor immune response. Oncotarget. 2017 Aug 24;8(48):84140-84152.
140. Alexander ET, Fahey E, Phanstiel O 4th, Gilmour SK. Loss of Anti-Tumor Efficacy by Polyamine Blocking Therapy in GCN2 Null Mice. Biomedicines. 2023 Oct 5;11(10):2703.
141. Alexander ET, Mariner K, Donnelly J, Phanstiel O 4th, Gilmour SK. Polyamine Blocking Therapy Decreases Survival of Tumor-Infiltrating Immunosuppressive Myeloid Cells and Enhances the Antitumor Efficacy of PD-1 Blockade. Mol Cancer Ther. 2020 Oct;19(10):2012-22.
142. Urban-Wójciuk Z, Graham A, Barker K, Kwok C, Sbirkov Y, Howell L, et al. The biguanide polyamine analog verlindamycin promotes differentiation in neuroblastoma via induction of antizyme. Cancer Gene Ther. 2022 Jul;29(7):940-50.
143. Shi HX, Liang C, Yao CY, Gao ZX, Qin J, Cao JL, et al. Elevation of spermine remodels immunosuppressive microenvironment through driving the modification of PD-L1 in hepatocellular carcinoma. Cell Commun Signal. 2022 Nov 8;20(1):175.
144. Fan J, Feng Z, Chen N. Spermidine as a target for cancer therapy. Pharmacol Res. 2020 Sep;159:104943.
145. Emmons-Bell M, Forsyth G, Sundquist A, Oldeman S, Gardikioti A, de Souza R, et al. Polyamines regulate cell fate by altering the activity of histone-modifying enzymes. bioRxiv [Preprint]. 2024 Jul 2:2024.07.02.600738.
146. Chang L, Li Z, Guo H, Zhang W, Lan W, Wang J, et al. Function of Polyamines in Regulating Cell Cycle Progression of Cultured Silkworm Cells. Insects. 2021 Jul 8;12(7):624.
147. Christovich A, Luo XM. Gut Microbiota, Leaky Gut, and Autoimmune Diseases. Front Immunol. 2022 Jun 27;13:946248.
148. Zhang H, Simon AK. Polyamines reverse immune senescence via the translational control of autophagy. Autophagy. 2020 Jan;16(1):181-2.
149. Arthur R, Jamwal S, Kumar P. A review on polyamines as promising next-generation neuroprotective and anti-aging therapy. Eur J Pharmacol. 2024 Sep 5;978:176804.