top of page
Writer's pictureJoseph Phillips

Spermidine The Synchronizer

Updated: Dec 2, 2023


Spermidine synchronizes a vast array of biological processes. Spermidine is found at its highest source from wheat germ. It’s a naturally occurring polyamine, and it’s gained significant attention in recent years due to its potential health benefits. Numerous studies have explored the effects of spermidine on various aspects of health, including neuroprotection, cardiovascular health, inflammation, and longevity.


Because we are essentially electric beings, governed largely in part by the function of our ion channels, Spermidine becomes an important compound in maintaining our health.


Spermidine synchronizes an array of biological processes, such as Ca2+, Na+, K+ -ATPase ion channels thus maintaining membrane potential and controlling intracellular pH and volume. Spermidine is a longevity agent in mammals due to various mechanisms of action, which are just beginning to be understood. Autophagy is the main mechanism at the molecular level, but evidence has been found for other mechanisms, including inflammation reduction, lipid metabolism, and regulation of cell growth, proliferation, and death.


The Role of Spermidine in Modulating Ion Channels


Ion channels play a crucial role in maintaining cellular homeostasis by regulating the flow of ions across cell membranes. Spermidine, a natural polyamine, has been found to modulate the activity of various ion channels.


Spermidine and Ion Channel Modulation


Spermidine has been shown to modulate the activity of ion channels in various biological systems. Harkat et al. (2017) demonstrated that spermidine can permeate through the pore of ATP-gated P2X receptors, suggesting its ability to interact with and influence ion channel function. Additionally, Han et al. (2022) reported that spermidine can inhibit the N-methyl-D-aspartate receptor (NMDAr), a subtype of ion channel involved in neuroprotection. These findings highlight the potential of spermidine as a modulator of ion channel activity.


Mechanisms of Spermidine's Effects on Ion Channels


The effects of spermidine on ion channels can be attributed to its role in cellular processes. Pegg (2009) stated that polyamine levels, including spermidine, can affect ion channels, cell-cell interactions, cytoskeleton dynamics, and signaling pathways. Spermidine has also been implicated in the regulation of protein and nucleic acid synthesis, protection against oxidative damage, and cell proliferation, differentiation, and apoptosis (Pegg, 2016). These cellular processes are closely linked to ion channel function, suggesting that spermidine may exert its effects on ion channels through these mechanisms.


Polyamines and Intrinsic Rectification of Ion Channels


Polyamines, including spermidine, have been implicated in the intrinsic rectification of ion channels. Lopatin et al. (1995) proposed that polyamines, such as spermine and spermidine, contribute to the gating of inward rectifier potassium channels. They suggested that polyamines can "plug" the pore of potassium channels, leading to rectification of ion flow. Suma et al. (2020) further supported this notion by demonstrating that spermidine and spermine can inhibit ion flow through various cation channels, including eukaryotic CNG channels and BK channels. These findings suggest that spermidine may play a role in regulating ion channel rectification.


In conclusion, Spermidine, a natural polyamine, has been shown to modulate the activity of ion channels. Its effects on ion channels can be attributed to its involvement in various cellular processes, including protein and nucleic acid synthesis, cell signaling, and cell proliferation. Furthermore, spermidine has been implicated in the intrinsic rectification of ion channels. Understanding the role of spermidine in modulating ion channels can provide insights into its potential therapeutic applications in various diseases and conditions associated with ion channel dysfunction.


Spermidine & Channelopathies


Spermidine is a natural polyamine that has been extensively studied for its potential health benefits. It has been found to have various effects on different biological processes, including neuroprotection, autophagy induction, anti-inflammation, anti-oxidation, and ion channel regulation (Han et al., 2022). These properties make spermidine a promising candidate for the treatment of various diseases, including channelopathies.


Channelopathies are a group of disorders that are caused by mutations in ion channel genes, leading to abnormal ion channel function (Imbrici et al., 2016). These disorders can affect various organ systems, including the nervous system, cardiovascular system, and skeletal muscle. The discovery that ion channel mutations can modify the sensitivity to drugs has opened up new possibilities for personalized therapy with increased efficacy and reduced side effects (Imbrici et al., 2016).


Spermidine has been shown to have a protective effect on retinal ganglion cells and promote optic nerve regeneration in mice following optic nerve injury (Noro et al., 2015). This suggests that spermidine may have potential therapeutic applications in the treatment of channelopathies affecting the visual system.


In addition, spermidine has been found to induce autophagy, a cellular process that plays a crucial role in maintaining cellular homeostasis and clearing damaged proteins and organelles (Eisenberg et al., 2009). Autophagy dysfunction has been implicated in the pathogenesis of various channelopathies, including neurodegenerative diseases and cardiovascular disorders. Therefore, spermidine's ability to induce autophagy may have therapeutic implications for these conditions.


Furthermore, spermidine has been shown to have anti-inflammatory properties (Madeo et al., 2018). Inflammation is a common feature of many channelopathies and can contribute to disease progression. By reducing inflammation, spermidine may help alleviate symptoms and slow down disease progression in channelopathies.


Overall, spermidine holds promise as a potential therapeutic agent for channelopathies. Its ability to preserve mitochondrial function, induce autophagy, and exhibit anti-inflammatory properties make it a versatile compound with potential applications in the treatment of various diseases. Further research is needed to fully understand the mechanisms underlying spermidine's effects and to explore its therapeutic potential in specific channelopathies.


The Role of Spermidine in Piezoelectricity


Spermidine is a naturally occurring polyamine that has gained attention for its potential health benefits. It has been found to have anti-aging properties and has been shown to extend lifespan in various model organisms, including yeast, worms, flies, and mice (Volkers et al., 2014). Spermidine has also been shown to have protective effects against age-related diseases, such as cardiovascular disease and neurodegenerative disorders (Volkers et al., 2014).


One of the mechanisms through which spermidine exerts its effects is by activating a class of ion channels known as PIEZO channels. PIEZO channels are mechanically activated channels that respond to mechanical stimuli, such as pressure or stretch, and play a crucial role in various physiological processes, including touch sensation, pain perception, and cardiovascular regulation (Volkers et al., 2014).


The PIEZO channel family consists of two members in humans, PIEZO1 and PIEZO2. These channels are widely expressed in various tissues and cell types and are involved in diverse biological processes (Volkers et al., 2014). They are particularly important in the context of mechanotransduction, which is the process by which cells convert mechanical stimuli into electrical signals (Volkers et al., 2014). PIEZO channels are responsible for converting mechanical forces into electrical signals, allowing cells to sense and respond to their mechanical environment (Volkers et al., 2014).


The structure and function of PIEZO channels have been extensively studied. They are large, multi-domain proteins that form trimeric complexes in the cell membrane (Volkers et al., 2014). The activation of PIEZO channels involves conformational changes in the protein structure in response to mechanical stimuli, leading to the opening of the channel pore and the influx of ions into the cell (Volkers et al., 2014). This influx of ions generates electrical signals that can trigger various cellular responses.


Understanding the structure and function of PIEZO channels is important for elucidating their role in physiological processes and for developing potential therapeutic interventions. For example, modulating the activity of PIEZO channels could have implications for the treatment of conditions such as chronic pain or cardiovascular diseases (Volkers et al., 2014).


In conclusion, spermidine is a natural compound that has been shown to have anti-aging properties and protective effects against age-related diseases. One of the mechanisms through which spermidine exerts its effects is by activating PIEZO channels, which are mechanically activated ion channels involved in various physiological processes. Understanding the structure and function of PIEZO channels is important for further exploring their role in health and disease and for developing potential therapeutic interventions.


Neuroprotective Effects


Spermidine has been shown to preserve mitochondrial function, exhibit anti-inflammatory properties, and prevent stem cell senescence (Madeo et al., 2018). These properties make spermidine a potential neuroprotective agent. Animal studies have demonstrated that spermidine supplementation can decrease age-induced arterial stiffness and oxidative damage of endothelial cells (Muñoz-Esparza et al., 2019). Furthermore, spermidine has been found to improve mitochondrial function and increase the degradation of damaged mitochondria through mitophagy, thereby enhancing overall neuronal health (Gabandé-Rodríguez et al., 2019).


Cardiovascular Health


Research suggests that spermidine supplementation may have cardioprotective effects. Studies have shown that oral spermidine supplementation extends the lifespan of mice, reduces systemic blood pressure, and delays the progression to heart failure (Matsumoto, 2020). Spermidine has also been found to improve the mechanical and elastic properties of cardiomyocytes, promote cardiac autophagy and mitophagy, and enhance mitochondrial respiration (Suzuki et al., 2023). These findings indicate that spermidine may be beneficial for maintaining cardiovascular health and preventing age-related cardiac dysfunction.


Inflammation and Anti-Aging


Spermidine has been linked to anti-inflammatory effects and the promotion of longevity. Animal studies have demonstrated that spermidine administration inhibits oxidative stress and improves cardiac diastolic function, left ventricular elasticity, and mitochondrial function in aged mice (Eisenberg et al., 2009; Zou et al., 2022). Additionally, spermidine supplementation has been found to activate autophagy, a cellular process that plays a crucial role in maintaining cellular homeostasis and delaying the aging process (Fei et al., 2017). These findings suggest that spermidine may have potential anti-aging effects and could be a promising intervention for age-related diseases.


The available evidence suggests that spermidine supplementation may offer several health benefits. Its neuroprotective effects, including preserving mitochondrial function and preventing stem cell senescence, make it a potential candidate for neuroprotection. Furthermore, spermidine has shown promise in promoting cardiovascular health by reducing blood pressure, delaying the progression to heart failure, and improving the mechanical properties of cardiomyocytes. Additionally, spermidine has been linked to anti-inflammatory effects and the promotion of longevity through its ability to activate autophagy and inhibit oxidative stress. Further research is needed to fully understand the mechanisms underlying these effects and to determine optimal dosages and long-term safety. Nonetheless, spermidine holds promise as a natural compound with potential applications in promoting overall health and preventing age-related diseases.


Spermidine & Autophagy


Autophagy is a cellular process involved in the degradation and recycling of cellular components, which plays a crucial role in maintaining cellular homeostasis and preventing the accumulation of damaged proteins and organelles (Eisenberg et al., 2009). Spermidine, a natural polyamine, has been found to induce autophagy and has been associated with various health benefits.


Several studies have demonstrated the role of spermidine in promoting autophagy and its potential therapeutic applications. Eisenberg et al. (2009) showed that spermidine induces autophagy and promotes longevity in various model organisms, including Caenorhabditis elegans, Drosophila melanogaster, and Saccharomyces cerevisiae. They found that spermidine treatment led to increased autophagic flux and improved cellular health.


In addition to its anti-aging effects, spermidine has also been implicated in the treatment of various diseases. Madeo et al. (2018) highlighted the potential of spermidine as a neuroprotective agent and its ability to improve the generation and function of memory lymphocytes. They compared spermidine to other autophagy inducers, such as rapamycin and metformin, and found similar effects on autophagy and immune function.


Spermidine has also been shown to improve gut barrier integrity and gut microbiota function in diet-induced obese mice (Ma et al., 2020). This study demonstrated that spermidine treatment enhanced autophagy in the intestinal mucosa, leading to improved gut barrier function and a shift in the gut microbiota composition. These findings suggest that spermidine may have potential therapeutic applications in the treatment of obesity and related metabolic disorders.


Furthermore, spermidine has been investigated in the context of aging-related cardiovascular diseases. Ren & Zhang (2018) discussed the role of autophagy in aging and cardiovascular diseases and highlighted the findings of Eisenberg et al. (2009) that spermidine improves lifespan and health span through autophagy activation. They suggested that targeting autophagy, including spermidine-induced autophagy, may be a promising strategy for the treatment of aging-related cardiovascular diseases.


In summary, spermidine has emerged as a potent inducer of autophagy with various health benefits. It has been shown to promote longevity, improve immune function, enhance gut barrier integrity, and potentially mitigate aging-related cardiovascular diseases. These findings highlight the therapeutic potential of spermidine and its role in regulating autophagy for maintaining cellular homeostasis and promoting overall health.


The Health Benefits of Autophagy


Autophagy is a cellular process that plays a crucial role in maintaining cellular homeostasis and promoting overall health. It involves the degradation and recycling of damaged organelles, proteins, and intracellular aggregates, thereby preventing the accumulation of harmful substances within cells. Autophagy has been implicated in various physiological and pathological processes, including cancer, neurodegeneration, cardiomyopathy, diabetes, liver disease, autoimmune diseases, and infections (Glick et al., 2010).


Prevention of Diseases


Autophagy has been shown to protect against a wide range of diseases. It promotes cellular senescence, which limits the proliferation of damaged cells and prevents the development of cancer (Glick et al., 2010). Additionally, autophagy plays a crucial role in neurodegenerative diseases, as its upregulation in the brain after exercise has been linked to the prevention of neurodegenerative diseases (Brimson et al., 2021). Furthermore, autophagy is involved in the regulation of cellular metabolism and affects glucagon and insulin levels, making it relevant to the development of type 2 diabetes mellitus (Bozack et al., 2021). Overall, autophagy's ability to eliminate harmful substances and maintain cellular homeostasis contributes to the prevention of various diseases.


Improvement of Cellular Function


Autophagy is essential for maintaining cellular function and overall health. It helps to eliminate damaged organelles and proteins, preventing the accumulation of toxic substances within cells (Glick et al., 2010). Defective autophagy has been associated with chronic NLRP3 inflammasome activity, which contributes to inflammaging, a chronic low-grade inflammation associated with aging (Meyers & Zhu, 2020). Enhancement of autophagy has been shown to improve health outcomes by reducing inflammasome activity (Meyers & Zhu, 2020). Moreover, autophagy plays a role in mitochondrial function and biogenesis, which are crucial for cellular energy production and overall health (Roberts & Markby, 2021). By promoting mitochondrial health, autophagy contributes to the improvement of cellular function.


Longevity and Aging


Autophagy has been implicated in the regulation of lifespan and aging. It counteracts the age-associated accumulation of damaged organelles and proteins, improving the metabolic fitness of cells (Madeo et al., 2015). Autophagy is induced by both starvation and the genetic inactivation of nutrient signaling, suggesting its role in promoting longevity (Madeo et al., 2015). Furthermore, autophagy modulation by plant polyphenols has been proposed as a potential strategy for promoting healthy aging and preventing age-associated diseases (Brimson et al., 2021). The activation of autophagy through exercise has also been linked to the extension of lifespan and the prevention of age-related cognitive decline (He et al., 2012). These findings highlight the potential of autophagy as a therapeutic target for promoting healthy aging.


In conclusion, Autophagy plays a crucial role in maintaining cellular homeostasis and promoting overall health. It contributes to the prevention of various diseases, improves cellular function, and has implications for longevity and aging. The activation of autophagy through exercise, plant polyphenols, and other interventions holds promise for promoting healthy aging and preventing age-associated diseases. Further research is needed to fully understand the mechanisms underlying autophagy and its potential therapeutic applications.


The Role of Spermidine in Immune Function


Spermidine, a naturally occurring polyamine, has gained attention for its potential role in modulating immune function. Several studies have explored the effects of spermidine on various aspects of the immune system, including immune cell differentiation, inflammation, and immune aging.


Spermidine and Immune Cell Differentiation


One study by Carriche et al. (2021) investigated the role of spermidine in T-cell differentiation and function. The researchers found that spermidine can modulate T-cell differentiation, suggesting its potential as a regulator of immune responses. Additionally, Wang et al. (2022) demonstrated that spermidine promotes the generation of lymphocytes, which play a crucial role in immune response. These findings suggest that spermidine may have a positive impact on immune cell differentiation and function.


Spermidine and Inflammation


Spermidine has also been shown to exhibit anti-inflammatory properties. Madeo et al. (2018) reported that spermidine preserves mitochondrial function and exhibits anti-inflammatory effects. This suggests that spermidine may help regulate immune responses by reducing inflammation. Furthermore, Partridge et al. (2020) highlighted the potential of spermidine to enhance immunity, further supporting its role in modulating immune function.


Spermidine and Immune Aging


Immune aging, characterized by a decline in immune function with age, is a significant concern. Partridge et al. (2020) discussed the potential of spermidine to slow down aging through drug discovery. They highlighted spermidine's ability to enhance immunity, suggesting its potential as an anti-aging intervention. These studies suggest that spermidine may have a role in mitigating immune aging.


In conclusion, the available evidence suggests that spermidine may play a significant role in immune function. It has been shown to modulate immune cell differentiation, exhibit anti-inflammatory properties, and potentially counteract immune aging. However, further research is needed to fully elucidate the mechanisms underlying spermidine's effects on the immune system. Understanding the relationship between spermidine and immune function could have implications for the development of novel interventions targeting immune-related diseases and age-related immune decline.


The Role of Spermidine in Autoimmunity


Spermidine is a polyamine that has been extensively studied for its potential role in autoimmune diseases. Several studies have investigated the effects of spermidine on autoimmune conditions and have found promising results.


One study by Madeo et al. (2018) found that spermidine can indirectly prevent the activation of autoimmune-reactive T cells by promoting the polarization of circulatory macrophages to T cell inhibitory M2 cells. This suggests that spermidine may have immunomodulatory effects that can help regulate the immune response in autoimmune diseases.


Another study by Li et al. (2022) demonstrated that spermidine can alleviate inflammatory bowel disease in mice by eliciting an anti-inflammatory phenotype in macrophages through the mtROS-AMPK-Hif-1α axis and autophagy induction. This indicates that spermidine may have anti-inflammatory properties that can be beneficial in autoimmune conditions characterized by excessive inflammation.


Furthermore, Li et al. (2020) identified spermidine as a potential therapeutic agent for autoimmune diseases by suppressing inflammatory dendritic cell (DC) function through the activation of the FOXO3 pathway. This suggests that spermidine may have the ability to modulate specific immune responses and counteract autoimmunity.


In a study by (Yang et al., 2016), spermidine was found to alleviate experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, by inducing inhibitory macrophages. This indicates that spermidine may have a protective effect on the central nervous system in autoimmune conditions.


Moreover, Noro et al. (2015) reported that oral administration of spermidine ameliorated the severity of EAE by suppressing oxidative stress. This suggests that spermidine may have antioxidant properties that can protect against oxidative damage associated with autoimmune diseases.


Overall, these studies suggest that spermidine has potential as a therapeutic agent for autoimmune diseases. Its ability to modulate the immune response, suppress inflammation, induce inhibitory macrophages, and protect against oxidative stress make it a promising candidate for further research and development of treatments for autoimmune conditions.


The Role of Spermidine in Nervous System Health


Spermidine has been found to have various beneficial effects on nervous system health. Several studies have demonstrated the neuroprotective properties of spermidine. For example, exogenously applied spermidine has been shown to have neuroprotective effects in neurodegenerative motor disorders and dementias (Madeo et al., 2018). Additionally, spermidine has been found to promote retinal ganglion cell survival and optic nerve regeneration, suggesting its potential use in the treatment of neurodegenerative diseases such as glaucoma (Noro et al., 2015).


Furthermore, spermidine has been associated with brain health in older adults. A study found that a higher spermidine intake was positively associated with several structural brain parameters, including hippocampal volume and cortical thickness, suggesting that spermidine intake may be beneficial for brain health (Zou et al., 2022). Another study found that spermidine supplementation had beneficial effects on brain and cognitive health in individuals with subjective cognitive decline (Schwarz et al., 2022).


In addition to its neuroprotective effects, spermidine has also been linked to cardiovascular health, which is closely related to nervous system health. High levels of dietary spermidine have been associated with reduced blood pressure and a lower incidence of cardiovascular disease (Eisenberg et al., 2016). This suggests that spermidine may have a broader impact on overall health, including the cardiovascular system, which in turn can influence nervous system health.


The mechanisms underlying the beneficial effects of spermidine on nervous system health are still being investigated. One possible mechanism is through the induction of autophagy, a cellular process that helps remove damaged proteins and organelles. Spermidine has been shown to induce autophagy, which can help protect neurons from degeneration and promote their survival (Madeo et al., 2018). Additionally, spermidine has been found to modulate mitochondrial respiration, which is important for cellular energy production and overall neuronal function (Li et al., 2022).


In conclusion, spermidine has been shown to have various beneficial effects on nervous system health. It has been found to have neuroprotective properties, promote retinal ganglion cell survival and optic nerve regeneration, and be associated with brain health in older adults. Furthermore, spermidine has been linked to cardiovascular health, which can indirectly influence nervous system health. The mechanisms underlying these effects are still being elucidated, but autophagy induction and modulation of mitochondrial respiration are potential mechanisms through which spermidine exerts its beneficial effects on nervous system health.


The Role of Spermidine in Myelination


Spermidine is a polyamine that has been implicated in various biological processes, including myelination. Myelination is the process by which oligodendrocytes in the central nervous system produce the myelin sheath, a protective covering around nerve fibers (Bradl & Lassmann, 2009). Several studies have explored the relationship between spermidine and myelin.


One study by Eichberg et al. (1981) found that spermidine concentrations were highest in myelin-rich areas, suggesting an association between polyamines and membranes (Eichberg et al., 1981). This finding supports the idea that spermidine may play a role in myelination.


Another study by Duncan & Radcliff (2016) investigated the underlying pathology of myelin disorders and the potential for remyelination strategies. They emphasized the importance of understanding whether myelin damage results from a primary attack on myelin or the oligodendrocyte, as well as the role of myelin breakdown and demyelination (Duncan & Radcliff, 2016). This information is relevant because it provides context for the potential involvement of spermidine in myelin-related disorders.


Furthermore, a study by Verden & Macklin (2016) highlighted the link between myelin and axon health and the interest in remyelination as a therapeutic target for neuroprotection. They discussed various approaches to promote de novo myelin generation in injury and disease models (Verden & Macklin, 2016). This information is important because it suggests that enhancing myelination could have neuroprotective effects.


In addition, a study by (2014) explored mitochondrial dysfunction in central nervous system white matter disorders. They found that knockout animals for PGC-1a, a regulator of mitochondrial biogenesis, had deficient postnatal myelination (Morató et al., 2014). This finding suggests that mitochondrial function is crucial for proper myelination and further supports the potential involvement of spermidine, as it is known to modulate mitochondrial respiration (Puleston et al., 2019).


Overall, these studies provide evidence for the potential role of spermidine in myelination. Spermidine concentrations are highest in myelin-rich areas, and its involvement in mitochondrial function suggests a link to myelin production. Further research is needed to fully understand the mechanisms by which spermidine influences myelination and to explore its potential therapeutic applications.


The Role of Spermidine in Pineal Gland Function


The pineal gland is a small endocrine organ located in the brain that plays a crucial role in regulating circadian rhythms and producing the hormone melatonin (Sabry & Matsuzaki, 1991). Several studies have investigated the molecular mechanisms and factors involved in pineal gland development and function. This paper aims to explore the relationship between spermidine and the pineal gland, focusing on its role in development, melatonin production, and potential therapeutic applications.


Role of Spermidine in Pineal Gland Development


Homeobox genes, such as Crx and Otx2, have been identified as key regulators of pineal gland development (Rath et al., 2012; Rath et al., 2006). While Crx is essential for retinal development, it is not required for pineal gland development (Rath et al., 2012). On the other hand, Otx2 expression and protein presence in the pineal gland are consistent with its requirement for pineal development (Rath et al., 2006). These findings suggest that different homeobox genes are involved in the development of specific organs. Further research is needed to elucidate the specific role of spermidine in pineal gland development.


Spermidine and Melatonin Production


The pineal gland is responsible for the synthesis and secretion of melatonin, a hormone that regulates sleep-wake cycles and has antioxidant properties (Song, 2019). Spermidine has been shown to regulate gene expression and enzyme activity related to melatonin production in other organs (Cen et al., 2020). However, the direct effect of spermidine on melatonin production in the pineal gland remains to be investigated.


Therapeutic Potential of Spermidine in Pineal Dysfunction


Pineal dysfunction has been implicated in neurodegenerative diseases, such as Alzheimer's disease (AD) (Song, 2019). Pineal dysfunction in AD may contribute to the progression of AD neuropathology (Song, 2019). Additionally, studies have shown that melatonin administration or pineal grafting can enhance survival and delay aging in mice (Pierpaoli & Regelson, 1994). These findings suggest that spermidine, as a regulator of melatonin production, may have therapeutic potential in the treatment of pineal dysfunction and age-related disorders.


In conclusion, spermidine may play a role in pineal gland development and melatonin production. Homeobox genes, such as Crx and Otx2, are involved in pineal gland development, with Otx2 being essential for pineal development (Rath et al., 2012; Rath et al., 2006). Spermidine has been shown to regulate gene expression and enzyme activity related to melatonin production in other organs (Cen et al., 2020). Pineal dysfunction has been implicated in neurodegenerative diseases, and spermidine may have therapeutic potential in the treatment of pineal dysfunction and age-related disorders (Song, 2019; Pierpaoli & Regelson, 1994). Further research is needed to fully understand the molecular mechanisms underlying the relationship between spermidine and the pineal gland.


Spermidine and the Writers, Readers, and Erasers of DNA


Spermidine is a polyamine compound that plays a crucial role in various biological processes, including DNA synthesis, gene expression, and chromatin remodeling (Natesan et al., 2022). The regulation of DNA structure and function involves a complex interplay between different proteins known as "writers," "readers," and "erasers" (Boo & Kim, 2020). These proteins are responsible for introducing, recognizing, and removing chemical modifications on DNA and histone proteins. Understanding the roles of these proteins is essential for unraveling the mechanisms underlying DNA regulation and its impact on cellular processes.


Writers of DNA Modifications


The process of introducing chemical modifications to DNA and histone tails is carried out by writer proteins (Boo & Kim, 2020). These writer proteins, also known as DNA-modifying enzymes, transfer specific chemical groups to target positions on DNA molecules (Natesan et al., 2022). They play a crucial role in establishing and maintaining epigenetic marks, such as DNA methylation, histone acetylation, and histone methylation (Fahrner & Bjornsson, 2014). These modifications can influence gene expression and chromatin structure, thereby regulating various cellular processes (Grady et al., 2021).


Readers of DNA Modifications


Readers, also referred to as reader proteins or RNA-binding proteins (RBPs), specifically recognize the modified nucleotides on DNA or RNA molecules (Boo & Kim, 2020). These proteins have specialized domains that bind to the modified nucleotides, allowing them to interpret the epigenetic marks and regulate downstream processes (Natesan et al., 2022). For example, readers of DNA methylation, such as methyl-CpG binding domain proteins, can recruit other proteins to modulate gene expression (Bayraktar & Kreutz, 2017). Similarly, readers of RNA modifications, such as m6A, play a role in RNA metabolism and the regulation of mRNA stability (Yang et al., 2018).


Erasers of DNA Modifications


Erasers, as the name suggests, are responsible for removing specific chemical groups from modified nucleotides, converting them back into unmodified nucleotides (Boo & Kim, 2020). These eraser proteins play a critical role in maintaining the dynamic nature of DNA modifications and ensuring proper regulation of gene expression. For example, DNA demethylases, such as Ten-Eleven Translocation (TET) enzymes, are involved in the active removal of DNA methylation marks (Fahrner & Bjornsson, 2014). Similarly, histone demethylases are responsible for removing methyl groups from histone proteins, thereby modulating chromatin structure and gene expression (Filippakopoulos et al., 2010).


The regulation of DNA structure and function involves a complex interplay between writers, readers, and erasers of DNA modifications. These proteins play crucial roles in establishing, interpreting, and removing chemical modifications on DNA and histone proteins. Spermidine, as a polyamine compound, is involved in various biological processes and may also influence the activity of these proteins. Further research is needed to elucidate the specific mechanisms by which spermidine interacts with the writers, readers, and erasers of DNA and its impact on cellular processes.


The Role of Spermidine and the Guardian of the Genome


The tumor suppressor protein p53 plays a crucial role in maintaining genomic stability and preventing the development of cancer (Kang et al., 2019). It is often referred to as the "guardian of the genome" due to its involvement in controlling cell survival and division under various stresses (Kang et al., 2019). One of the mechanisms through which p53 exerts its tumor suppressive function is by regulating ferroptosis, a form of regulated cell death characterized by the accumulation of lipid peroxides (Ou et al., 2016).


Activation of the SAT1 (spermidine/spermine N1-acetyltransferase 1) gene, which is a transcription target of p53, has been shown to engage polyamine metabolism and promote ferroptotic cell death (Ou et al., 2016). SAT1 encodes a rate-limiting enzyme in polyamine catabolism and its upregulation leads to increased levels of ALOX15, a key enzyme involved in lipid peroxidation and ferroptosis (Ou et al., 2016). This process is dependent on the transcriptional inhibition of SLC7A11, a component of the antioxidant system, or the transcriptional induction of SAT1 and glutaminase 2 (Xie et al., 2017).


Furthermore, p53 can induce the expression of SAT1 to upregulate ALOX15, thereby promoting ferroptosis in response to reactive oxygen species (ROS) stress (Lei et al., 2021). The regulation of ferroptosis by p53 is not limited to its role in cell cycle arrest, senescence, and apoptosis, but also extends to other cellular processes such as metabolism and antioxidant defense (Liu et al., 2020). The downregulation of p53 has been shown to limit ferroptosis, while its stabilization enhances the sensitivity of cancer cells to ferroptosis-inducing agents (Liu et al., 2020).


In addition to its role in regulating ferroptosis, p53 also interacts with other pathways involved in cell death and survival, such as autophagy (Sargazi et al., 2021). However, the focus of this paper is on the relationship between p53 and ferroptosis.


In conclusion, p53 acts as a key regulator of ferroptosis, a form of regulated cell death characterized by lipid peroxidation. Activation of the SAT1 gene, a transcription target of p53, promotes ferroptosis by increasing lipid peroxidation through the upregulation of ALOX15. This highlights the multifaceted role of p53 in maintaining genomic stability and preventing the development of cancer.


The Benefits of Spermidine in Eukaryotic Translation


Eukaryotic translation is a complex process that involves the synthesis of proteins from messenger RNA (mRNA) molecules. It is tightly regulated and plays a crucial role in cellular functions. Spermidine, a polyamine compound, has been found to have various benefits in eukaryotic translation.


Spermidine and Translation Initiation


Spermidine has been shown to play a role in translation initiation, the process by which ribosomes assemble on mRNA molecules to initiate protein synthesis (Jackson et al., 2010). It has been found that spermidine is essential for the modification and activation of the eukaryotic translation initiation factor eIF5A (Burger et al., 2007). This modification of eIF5A is important for the proper initiation of translation and subsequent protein synthesis (Miller-Fleming et al., 2015). Additionally, spermidine has been shown to be involved in the regulation of eukaryotic initiation factors, such as eIF2, which is crucial for the initiation of translation (Hinnebusch et al., 2016).


Spermidine and Translation Elongation


In addition to its role in translation initiation, spermidine has also been implicated in translation elongation, the process by which ribosomes synthesize proteins (Landau et al., 2010). Spermidine is required for the hypusination of eukaryotic initiation factor 5A (eIF5A), a modification that is crucial for translation elongation (Landau et al., 2010). The hypusination of eIF5A is necessary for the proper elongation of the peptide chain during translation (Chattopadhyay et al., 2008). Inhibition of eIF5A hypusination by spermidine analogues has been shown to result in the inhibition of translation initiation (Landau et al., 2010).


Benefits of Spermidine in Eukaryotic Translation


The presence of spermidine has been associated with several benefits in eukaryotic translation. Firstly, spermidine has been found to preserve mitochondrial function, which is essential for efficient translation (Madeo et al., 2018). Mitochondria are the powerhouses of the cell and provide the necessary energy for translation. Spermidine's ability to preserve mitochondrial function ensures the availability of energy for translation.


Furthermore, spermidine exhibits anti-inflammatory properties, which can indirectly benefit eukaryotic translation. Inflammation has been shown to impair translation efficiency, and spermidine's anti-inflammatory properties can help maintain optimal translation rates (Madeo et al., 2018).


Moreover, spermidine has been found to prevent stem cell senescence, which can have a positive impact on translation. Senescent cells have reduced translation rates, and by preventing senescence, spermidine can help maintain efficient translation in stem cells (Madeo et al., 2018).


In conclusion, Spermidine plays a crucial role in eukaryotic translation, particularly in translation initiation and elongation. Its involvement in the modification and activation of translation initiation factors and the hypusination of eIF5A highlights its importance in the regulation of translation. The benefits of spermidine in eukaryotic translation, such as preserving mitochondrial function, exhibiting anti-inflammatory properties, and preventing stem cell senescence, further emphasize its significance in maintaining optimal translation rates. Further research is needed to fully understand the mechanisms underlying spermidine's effects on eukaryotic translation and to explore its potential therapeutic applications.


The Benefits of Spermidine on Stem Cells


Recent studies have shown that spermidine exhibits various beneficial effects, including preserving mitochondrial function, preventing stem cell senescence, and promoting cell survival and regeneration.


Preservation of Stem Cell Function


Spermidine has been found to preserve the function of stem cells in various tissues. In a study by (Madeo et al., 2018), spermidine was shown to prevent stem cell senescence and improve muscle regeneration in aging mice. Similarly, Yan et al. (2019) demonstrated that spermidine-enhanced autophagy improved cardiac dysfunction following myocardial infarction by targeting the AMPK/mTOR signaling pathway. These findings suggest that spermidine may have the potential to induce the regeneration of different types of stem cells, including cardiac stem cells.


Cardioprotection and Stem Cells


Eisenberg et al. (2016) investigated the cardioprotective effects of spermidine and its potential impact on stem cells. They found that spermidine extended the lifespan of mice and improved cardiac function. Furthermore, they suggested that spermidine might have a similar effect on other adult stem cells, including cardiac stem cells that can produce new and functional cardiomyocytes. This indicates that spermidine could potentially enhance the regenerative capacity of cardiac stem cells, leading to improved cardiac function.


Hair Follicle Stem Cells


Spermidine has also been studied in the context of hair follicle stem cells. Ramot et al. (2011) examined the effects of spermidine on human hair follicles and epithelial stem cells. They found that spermidine promoted hair growth and modulated the functions of hair follicle stem cells. This suggests that spermidine may have a role in promoting the proliferation and maintenance of stem cells in the hair follicle.


Neural Stem Cells


In addition to its effects on muscle and cardiac stem cells, spermidine has been shown to promote the survival and regeneration of neural stem cells. Noro et al. (2015) demonstrated that spermidine promoted retinal ganglion cell survival and optic nerve regeneration in adult mice following optic nerve injury. They proposed that spermidine facilitated the functional communication between astrocytes, enhancing the survival and regenerative capacity of neural cells.


In conclusion, Spermidine exhibits various beneficial effects on stem cells, including the preservation of stem cell function, promotion of cell survival and regeneration, and modulation of stem cell functions in different tissues. These findings suggest that spermidine has the potential to enhance the regenerative capacity of stem cells, making it a promising candidate for regenerative medicine. Further research is needed to fully understand the mechanisms underlying the effects of spermidine on stem cells and to explore its therapeutic potential.


The Role of Spermidine and NRF2 Activation


Spermidine and NRF2 are both important factors in cellular processes related to oxidative stress and disease prevention. Spermidine has been shown to activate the NRF2 signaling pathway, leading to increased antioxidant production and protection against oxidative damage (Liu et al., 2020; Yang et al., 2022; Aihara et al., 2023; Jiang et al., 2023). This activation of NRF2 by spermidine has been observed in various tissues, including the liver, lung, and kidney (Liu et al., 2020; Yang et al., 2022; Aihara et al., 2023). In the liver, spermidine has been found to suppress liver fibrosis by activating NRF2 and increasing autophagy mediated by MAP1S (Liu et al., 2020; This protective effect was partially reversed in mice lacking NRF2 or p62, and completely inhibited in mice lacking both NRF2 and p62 (Liu et al., 2020). In the lung, spermidine-induced activation of the NRF2 signaling pathway has been shown to mitigate lung inflammatory injury (Yang et al., 2022). Similarly, in the kidney, spermidine has been found to inhibit kidney fibrosis by activating NRF2 (Aihara et al., 2023).


NRF2 itself plays a critical role in mitigating oxidative stress and preventing diseases associated with increased lipid peroxidation and ferroptosis (Dodson et al., 2019). Aberrant NRF2 signaling has been implicated in diseases such as cancer and neurodegenerative disorders (Dodson et al., 2019; Niture et al., 2014). The antioxidant and cell protective properties of NRF2 make it an important target for drug discovery in the prevention and treatment of oxidative stress-related diseases (Sykiotis & Bohmann, 2008). In addition, NRF2 has been shown to have a protective role against diabetic nephropathy, as evidenced by elevated NRF2 levels in the glomeruli of diabetic nephropathy patients (Jiang et al., 2010).


The interaction between spermidine and NRF2 is complex and involves multiple pathways. Spermidine can induce the NRF2 signaling pathway through MAP1S, leading to p62-dependent Keap1 degradation via the autophagy pathway (Yang et al., 2022). Additionally, spermidine can activate the Keap1-NRF2-ARE antioxidant signaling pathway, mediating the expression of antioxidant enzymes such as heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (Jiang et al., 2023). This activation of NRF2 by spermidine contributes to the antioxidant effect of spermidine in various tissues.


Overall, the interaction between spermidine and NRF2 is important for cellular protection against oxidative stress and the prevention of oxidative stress-related diseases. Spermidine activates the NRF2 signaling pathway, leading to increased antioxidant production and protection against oxidative damage. Understanding the mechanisms underlying this interaction may provide insights into the development of therapeutic strategies for oxidative stress-related diseases.


Summary


Spermidine has been shown to preserve mitochondrial function, exhibit anti-inflammatory properties, and prevent stem cell senescence (Madeo et al., 2018). Spermidine-induced autophagy is required for several of its beneficial health effects, including lifespan extension, prevention of cardiac aging, improvement in neuronal function, and preservation of myocyte stemness (Levine & Kroemer, 2019). Additionally, spermidine has been found to exert a potent anti-aging influence on arteries by increasing nitric oxide bioavailability, reducing oxidative stress, modifying structural factors, and enhancing autophagy (Matsumoto, 2020).


In terms of cardiovascular health, spermidine has been shown to reduce systemic blood pressure, prevent cardiac hypertrophy, and delay the progression to heart failure (Eisenberg et al., 2016). It has also been found to decrease age-induced arterial stiffness and oxidative damage of endothelial cells (Muñoz-Esparza et al., 2019). Furthermore, spermidine supplementation has been found to extend lifespan and reverse aging-associated cardiac dysfunction in mice through the induction of autophagy (Tong & Hill, 2017).


Spermidine also displays antioxidant and anti-inflammatory properties, making it beneficial for liver health. It has been shown to prevent ethanol and lipopolysaccharide-induced hepatic injury in mice (Adhikari et al., 2021). In the field of cancer research, spermidine has shown promise as an anticancer agent. It interferes with the tumor cell cycle, inhibiting tumor cell proliferation and suppressing tumor growth (Prasher et al., 2023).


Other health benefits of spermidine include its anti-aging effects, anti-inflammatory effects, cardiovascular protection, and neuromodulation (Zou et al., 2022). It has also been studied for its potential applications in ocular diseases, such as glaucoma, due to its anti-aging, autophagy induction, anti-inflammation, and anti-oxidation properties (Han et al., 2022).


In conclusion, spermidine is a natural polyamine that offers a wide range of health benefits. It has been shown to preserve mitochondrial function, exhibit anti-inflammatory properties, prevent stem cell senescence, and induce autophagy. It has beneficial effects on cardiovascular health, liver health, and has shown promise as an anticancer agent. Additionally, spermidine has anti-aging, anti-inflammatory, cardiovascular protective, and neuromodulatory effects. Its diverse range of health benefits makes it a promising compound for further research and potential therapeutic applications.





References:


Han, W., Li, H., & Chen, B. (2022). Research progress and potential applications of spermidine in ocular diseases. Pharmaceutics, 14(7), 1500. https://doi.org/10.3390/pharmaceutics14071500


Harkat, M., Peverini, L., Cerdan, A., Dunning, K., Beudez, J., Martz, A., … & Grutter, T. (2017). On the permeation of large organic cations through the pore of atp-gated p2x receptors. Proceedings of the National Academy of Sciences, 114(19). https://doi.org/10.1073/pnas.1701379114


Lopatin, A., Makhina, E., & Nichols, C. (1995). The mechanism of inward rectification of potassium channels: "long-pore plugging" by cytoplasmic polyamines.. The Journal of General Physiology, 106(5), 923-955. https://doi.org/10.1085/jgp.106.5.923


Pegg, A. (2009). Mammalian polyamine metabolism and function. Iubmb Life, 61(9), 880-894. https://doi.org/10.1002/iub.230


Pegg, A. (2016). Functions of polyamines in mammals. Journal of Biological Chemistry, 291(29), 14904-14912. https://doi.org/10.1074/jbc.r116.731661


Suma, A., Granata, D., Thomson, A., & Rothberg, B. (2020). Polyamine blockade and binding energetics in the mthk potassium channel. The Journal of General Physiology, 152(7). https://doi.org/10.1085/jgp.201912527


Eisenberg, T., Knauer, H., Schauer, A., Büttner, S., Ruckenstuhl, C., Carmona-Gutiérrez, D., … & Madeo, F. (2009). Induction of autophagy by spermidine promotes longevity. Nature Cell Biology, 11(11), 1305-1314. https://doi.org/10.1038/ncb1975


Han, W., Li, H., & Chen, B. (2022). Research progress and potential applications of spermidine in ocular diseases. Pharmaceutics, 14(7), 1500. https://doi.org/10.3390/pharmaceutics14071500


Imbrici, P., Liantonio, A., Camerino, G., Bellis, M., Camerino, C., Mele, A., … & Conte, D. (2016). Therapeutic approaches to genetic ion channelopathies and perspectives in drug discovery. Frontiers in Pharmacology, 7. https://doi.org/10.3389/fphar.2016.00121


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Noro, T., Namekata, K., Kimura, A., Guo, X., Azuchi, Y., Harada, C., … & Harada, T. (2015). Spermidine promotes retinal ganglion cell survival and optic nerve regeneration in adult mice following optic nerve injury. Cell Death and Disease, 6(4), e1720-e1720. https://doi.org/10.1038/cddis.2015.93


Eisenberg, T., Knauer, H., Schauer, A., Büttner, S., Ruckenstuhl, C., Carmona-Gutiérrez, D., … & Madeo, F. (2009). Induction of autophagy by spermidine promotes longevity. Nature Cell Biology, 11(11), 1305-1314. https://doi.org/10.1038/ncb1975


Fei, Y., Li, W., Zou, J., Xu, G., Huang, H., & Liu, L. (2017). Spermidine prolongs lifespan and prevents liver fibrosis and hepatocellular carcinoma by activating map1s-mediated autophagy. Cancer Research, 77(11), 2938-2951. https://doi.org/10.1158/0008-5472.can-16-3462


Gabandé-Rodríguez, E., Heras, M., & Mittelbrunn, M. (2019). Control of inflammation by calorie restriction mimetics: on the crossroad of autophagy and mitochondria. Cells, 9(1), 82. https://doi.org/10.3390/cells9010082


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Matsumoto, M. (2020). Prevention of atherosclerosis by the induction of microbial polyamine production in the intestinal lumen. Biological and Pharmaceutical Bulletin, 43(2), 221-229. https://doi.org/10.1248/bpb.b19-00855


Muñoz-Esparza, N., Latorre-Moratalla, M., Comas-Basté, O., Toro-Funes, N., Veciana-Nogués, M., & Vidal-Carou, M. (2019). Polyamines in food. Frontiers in Nutrition, 6. https://doi.org/10.3389/fnut.2019.00108


Suzuki, H., Fujiwara, Y., Thongbhubate, K., & Kanaori, K. (2023). Spore-forming lactic acid-producing bacterium bacillus coagulans synthesizes and excretes spermidine into the extracellular space. Journal of Agricultural and Food Chemistry, 71(25), 9868-9876. https://doi.org/10.1021/acs.jafc.3c02184


Zou, D., Zhao, Z., Li, L., Yu, M., Zhang, D., Ji, A., … & Wu, X. (2022). A comprehensive review of spermidine: safety, health effects, absorption and metabolism, food materials evaluation, physical and chemical processing, and bioprocessing. Comprehensive Reviews in Food Science and Food Safety, 21(3), 2820-2842. https://doi.org/10.1111/1541-4337.12963


Eisenberg, T., Knauer, H., Schauer, A., Büttner, S., Ruckenstuhl, C., Carmona-Gutiérrez, D., … & Madeo, F. (2009). Induction of autophagy by spermidine promotes longevity. Nature Cell Biology, 11(11), 1305-1314. https://doi.org/10.1038/ncb1975


Ma, L., Ota, T., Wang, Z., Tu, W., Ni, L., Zhuge, F., … & Zheng, L. (2020). Spermidine improves gut barrier integrity and gut microbiota function in diet-induced obese mice. Gut Microbes, 12(1), 1832857. https://doi.org/10.1080/19490976.2020.1832857


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Ren, J. and Zhang, Y. (2018). Targeting autophagy in aging and aging-related cardiovascular diseases. Trends in Pharmacological Sciences, 39(12), 1064-1076. https://doi.org/10.1016/j.tips.2018.10.005


Bozack, A., Boileau, P., Wei, L., Hubbard, A., Sillé, F., Ferreccio, C., … & Gamble, M. (2021). Exposure to arsenic at different life-stages and dna methylation meta-analysis in buccal cells and leukocytes. Environmental Health, 20(1). https://doi.org/10.1186/s12940-021-00754-7


Brimson, J., Prasanth, M., Malar, D., Sharika, R., Sivamaruthi, B., Kesika, P., … & Prasansuklab, A. (2021). Role of herbal teas in regulating cellular homeostasis and autophagy and their implications in regulating overall health. Nutrients, 13(7), 2162. https://doi.org/10.3390/nu13072162


Brimson, J., Prasanth, M., Malar, D., Thitilertdecha, P., Kabra, A., & Prasansuklab, A. (2021). Plant polyphenols for aging health: implication from their autophagy modulating properties in age-associated diseases. Pharmaceuticals, 14(10), 982. https://doi.org/10.3390/ph14100982


Glick, D., Barth, S., & Macleod, K. (2010). Autophagy: cellular and molecular mechanisms. The Journal of Pathology, 221(1), 3-12. https://doi.org/10.1002/path.2697


He, C., Sumpter, R., & Levine, B. (2012). Exercise induces autophagy in peripheral tissues and in the brain. Autophagy, 8(10), 1548-1551. https://doi.org/10.4161/auto.21327


Madeo, F., Zimmermann, A., Maiuri, M., & Kroemer, G. (2015). Essential role for autophagy in life span extension. Journal of Clinical Investigation, 125(1), 85-93. https://doi.org/10.1172/jci73946


Meyers, A. and Zhu, X. (2020). The nlrp3 inflammasome: metabolic regulation and contribution to inflammaging. Cells, 9(8), 1808. https://doi.org/10.3390/cells9081808


Roberts, F. and Markby, G. (2021). New insights into molecular mechanisms mediating adaptation to exercise; a review focusing on mitochondrial biogenesis, mitochondrial function, mitophagy and autophagy. Cells, 10(10), 2639. https://doi.org/10.3390/cells10102639


Carriche, G., Almeida, L., Stüve, P., Velásquez, L., Dhillon-LaBrooy, A., Roy, U., … & Sparwasser, T. (2021). Regulating t-cell differentiation through the polyamine spermidine. Journal of Allergy and Clinical Immunology, 147(1), 335-348.e11. https://doi.org/10.1016/j.jaci.2020.04.037


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Partridge, L., Fuentealba, M., & Kennedy, B. (2020). The quest to slow ageing through drug discovery. Nature Reviews Cancer, 19(8), 513-532. https://doi.org/10.1038/s41573-020-0067-7


Wang, H., Jiang, D., Liu, L., Zhang, Y., Miao, Q., Qu, Y., … & Xu, G. (2022). Spermidine promotes nb car-t mediated cytotoxicity to lymphoma cells through elevating proliferation and memory. Oncotargets and Therapy, Volume 15, 1229-1243. https://doi.org/10.2147/ott.s382540


Zinecker, H. and Simon, A. (2022). Autophagy takes it all – autophagy inducers target immune aging. Disease Models & Mechanisms, 15(1). https://doi.org/10.1242/dmm.049345


Li, G., Ding, H., Yu, X., Meng, Y., Li, J., Guo, Q., … & Zhou, H. (2020). Spermidine suppresses inflammatory dc function by activating the foxo3 pathway and counteracts autoimmunity. Iscience, 23(1), 100807. https://doi.org/10.1016/j.isci.2019.100807


Li, X., Zhou, X., Liu, X., Li, X., Jiang, X., Shi, B., … & Wang, S. (2022). Spermidine protects against acute kidney injury by modulating macrophage nlrp3 inflammasome activation and mitochondrial respiration in an eif5a hypusination-related pathway. Molecular Medicine, 28(1). https://doi.org/10.1186/s10020-022-00533-1


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Noro, T., Namekata, K., Kimura, A., Guo, X., Azuchi, Y., Harada, C., … & Harada, T. (2015). Spermidine promotes retinal ganglion cell survival and optic nerve regeneration in adult mice following optic nerve injury. Cell Death and Disease, 6(4), e1720-e1720. https://doi.org/10.1038/cddis.2015.93


Yang, Q., Zheng, C., Cao, J., Cao, G., Shou, P., Lin, L., … & Shi, Y. (2016). Spermidine alleviates experimental autoimmune encephalomyelitis through inducing inhibitory macrophages. Cell Death and Differentiation, 23(11), 1850-1861. https://doi.org/10.1038/cdd.2016.71


Eisenberg, T., Abdellatif, M., Schroeder, S., Primeßnig, U., Stekovic, S., Pendl, T., … & Madeo, F. (2016). Cardioprotection and lifespan extension by the natural polyamine spermidine. Nature Medicine, 22(12), 1428-1438. https://doi.org/10.1038/nm.4222


Li, X., Zhou, X., Liu, X., Li, X., Jiang, X., Shi, B., … & Wang, S. (2022). Spermidine protects against acute kidney injury by modulating macrophage nlrp3 inflammasome activation and mitochondrial respiration in an eif5a hypusination-related pathway. Molecular Medicine, 28(1). https://doi.org/10.1186/s10020-022-00533-1


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Noro, T., Namekata, K., Kimura, A., Guo, X., Azuchi, Y., Harada, C., … & Harada, T. (2015). Spermidine promotes retinal ganglion cell survival and optic nerve regeneration in adult mice following optic nerve injury. Cell Death and Disease, 6(4), e1720-e1720. https://doi.org/10.1038/cddis.2015.93


Schwarz, C., Benson, G., Horn, N., Wurdack, K., Grittner, U., Schilling, R., … & Flöel, A. (2022). Effects of spermidine supplementation on cognition and biomarkers in older adults with subjective cognitive decline. Jama Network Open, 5(5), e2213875. https://doi.org/10.1001/jamanetworkopen.2022.13875


Zou, D., Zhao, Z., Li, L., Yu, M., Zhang, D., Ji, A., … & Wu, X. (2022). A comprehensive review of spermidine: safety, health effects, absorption and metabolism, food materials evaluation, physical and chemical processing, and bioprocessing. Comprehensive Reviews in Food Science and Food Safety, 21(3), 2820-2842. https://doi.org/10.1111/1541-4337.12963


Bradl, M. and Lassmann, H. (2009). Oligodendrocytes: biology and pathology. Acta Neuropathologica, 119(1), 37-53. https://doi.org/10.1007/s00401-009-0601-5


Duncan, I. and Radcliff, A. (2016). Inherited and acquired disorders of myelin: the underlying myelin pathology. Experimental Neurology, 283, 452-475. https://doi.org/10.1016/j.expneurol.2016.04.002


Eichberg, J., Zetusky, W., Bell, M., & Cavanagh, E. (1981). Effects of polyamines on calcium-dependent rat brain phosphatidylinositol-phosphodiesterase. Journal of Neurochemistry, 36(5), 1868-1871. https://doi.org/10.1111/j.1471-4159.1981.tb00444.x


Morató, L., Bertini, E., Verrigni, D., Ardissone, A., Ruiz, M., Ferrer, I., … & Pujol, A. (2014). Mitochondrial dysfunction in central nervous system white matter disorders. Glia, 62(11), 1878-1894. https://doi.org/10.1002/glia.22670


Puleston, D., Buck, M., Geltink, R., Kyle, R., Caputa, G., O’Sullivan, D., … & Pearce, E. (2019). Polyamines and eif5a hypusination modulate mitochondrial respiration and macrophage activation. Cell Metabolism, 30(2), 352-363.e8. https://doi.org/10.1016/j.cmet.2019.05.003


Verden, D. and Macklin, W. (2016). Neuroprotection by central nervous system remyelination: molecular, cellular, and functional considerations. Journal of Neuroscience Research, 94(12), 1411-1420. https://doi.org/10.1002/jnr.23923


Volkers, L., Mechioukhi, Y., & Coste, B. (2014). Piezo channels: from structure to function. Pflügers Archiv - European Journal of Physiology, 467(1), 95-99. https://doi.org/10.1007/s00424-014-1578-z


Cen, H., Wang, T., Liu, H., Tian, D., & Zhang, Y. (2020). Melatonin application improves salt tolerance of alfalfa (medicago sativa l.) by enhancing antioxidant capacity. Plants, 9(2), 220. https://doi.org/10.3390/plants9020220


Pierpaoli, W. and Regelson, W. (1994). Pineal control of aging: effect of melatonin and pineal grafting on aging mice.. Proceedings of the National Academy of Sciences, 91(2), 787-791. https://doi.org/10.1073/pnas.91.2.787


Rath, M., Muñoz, E., Ganguly, S., Morin, F., Shi, Q., Klein, D., … & Møller, M. (2006). Expression of theotx2homeobox gene in the developing mammalian brain: embryonic and adult expression in the pineal gland. Journal of Neurochemistry, 97(2), 556-566. https://doi.org/10.1111/j.1471-4159.2006.03773.x


Rath, M., Rohde, K., Klein, D., & Møller, M. (2012). Homeobox genes in the rodent pineal gland: roles in development and phenotype maintenance. Neurochemical Research, 38(6), 1100-1112. https://doi.org/10.1007/s11064-012-0906-y


Sabry, I. and Matsuzaki, S. (1991). Daily cycles of putrescine, sperrnidine, and spermine in the liver, pineal gland, harderian gland, anterior pituitary, and testes of rats kept in ld 12:12. Journal of Pineal Research, 11(2), 86-91. https://doi.org/10.1111/j.1600-079x.1991.tb00461.x


Song, J. (2019). Pineal gland dysfunction in alzheimer’s disease: relationship with the immune-pineal axis, sleep disturbance, and neurogenesis. Molecular Neurodegeneration, 14(1). https://doi.org/10.1186/s13024-019-0330-8


Bayraktar, G. and Kreutz, M. (2017). Neuronal dna methyltransferases: epigenetic mediators between synaptic activity and gene expression?. The Neuroscientist, 24(2), 171-185. https://doi.org/10.1177/1073858417707457


Boo, S. and Kim, Y. (2020). The emerging role of rna modifications in the regulation of mrna stability. Experimental & Molecular Medicine, 52(3), 400-408. https://doi.org/10.1038/s12276-020-0407-z


Fahrner, J. and Bjornsson, H. (2014). Mendelian disorders of the epigenetic machinery: tipping the balance of chromatin states. Annual Review of Genomics and Human Genetics, 15(1), 269-293. https://doi.org/10.1146/annurev-genom-090613-094245


Filippakopoulos, P., Qi, J., Picaud, S., Shen, Y., Smith, W., Fedorov, O., … & Bradner, J. (2010). Selective inhibition of bet bromodomains. Nature, 468(7327), 1067-1073. https://doi.org/10.1038/nature09504


Grady, W., Yu, M., & Markowitz, S. (2021). Epigenetic alterations in the gastrointestinal tract: current and emerging use for biomarkers of cancer. Gastroenterology, 160(3), 690-709. https://doi.org/10.1053/j.gastro.2020.09.058


Natesan, M., Kong, M., Shi, M., Ghag, R., & Mollah, S. (2022). Crewdb 1.0: optimizing chromatin readers, erasers, and writers database using machine learning-based approach.. https://doi.org/10.1101/2022.06.02.494594


Yang, Y., Hsu, P., Chen, Y., & Yang, Y. (2018). Dynamic transcriptomic m6a decoration: writers, erasers, readers and functions in rna metabolism. Cell Research, 28(6), 616-624. https://doi.org/10.1038/s41422-018-0040-8


Kang, R., Kroemer, G., & Tang, D. (2019). The tumor suppressor protein p53 and the ferroptosis network. Free Radical Biology and Medicine, 133, 162-168. https://doi.org/10.1016/j.freeradbiomed.2018.05.074


Lei, G., Mao, C., Yan, Y., Li, Z., & Gan, B. (2021). Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein & Cell, 12(11), 836-857. https://doi.org/10.1007/s13238-021-00841-y


Liu, J., Zhang, C., Wang, J., Hu, W., & Feng, Z. (2020). The regulation of ferroptosis by tumor suppressor p53 and its pathway. International Journal of Molecular Sciences, 21(21), 8387. https://doi.org/10.3390/ijms21218387


Ou, Y., Wang, S., Li, D., Chu, B., & Gu, W. (2016). Activation of sat1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proceedings of the National Academy of Sciences, 113(44). https://doi.org/10.1073/pnas.1607152113


Sargazi, S., Sheervalilou, R., Rokni, M., Shirvaliloo, M., Shahraki, O., & Rezaei, N. (2021). The role of autophagy in controlling sars‐cov‐2 infection: an overview on virophagy‐mediated molecular drug targets. Cell Biology International, 45(8), 1599-1612. https://doi.org/10.1002/cbin.11609


Xie, Y., Zhu, S., Song, X., Sun, X., Fan, Y., Liu, J., … & Tang, D. (2017). The tumor suppressor p53 limits ferroptosis by blocking dpp4 activity. Cell Reports, 20(7), 1692-1704. https://doi.org/10.1016/j.celrep.2017.07.055


Burger, P., Birkholtz, L., Joubert, F., Haider, N., Walter, R., & Louw, A. (2007). Structural and mechanistic insights into the action of plasmodium falciparum spermidine synthase. Bioorganic & Medicinal Chemistry, 15(4), 1628-1637. https://doi.org/10.1016/j.bmc.2006.12.017


Chattopadhyay, M., Park, M., & Tabor, H. (2008). Hypusine modification for growth is the major function of spermidine in saccharomyces cerevisiae polyamine auxotrophs grown in limiting spermidine. Proceedings of the National Academy of Sciences, 105(18), 6554-6559. https://doi.org/10.1073/pnas.0710970105


Hinnebusch, A., Ivanov, I., & Sonenberg, N. (2016). Translational control by 5′-untranslated regions of eukaryotic mrnas. Science, 352(6292), 1413-1416. https://doi.org/10.1126/science.aad9868


Jackson, R., Hellen, C., & Pestova, T. (2010). The mechanism of eukaryotic translation initiation and principles of its regulation. Nature Reviews Molecular Cell Biology, 11(2), 113-127. https://doi.org/10.1038/nrm2838


Landau, G., Bercovich, Z., Park, M., & Kahana, C. (2010). The role of polyamines in supporting growth of mammalian cells is mediated through their requirement for translation initiation and elongation. Journal of Biological Chemistry, 285(17), 12474-12481. https://doi.org/10.1074/jbc.m110.106419


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Miller-Fleming, L., Olín-Sandoval, V., Campbell, K., & Ralser, M. (2015). Remaining mysteries of molecular biology: the role of polyamines in the cell. Journal of Molecular Biology, 427(21), 3389-3406. https://doi.org/10.1016/j.jmb.2015.06.020


Eisenberg, T., Abdellatif, M., Schroeder, S., Primeßnig, U., Stekovic, S., Pendl, T., … & Madeo, F. (2016). Cardioprotection and lifespan extension by the natural polyamine spermidine. Nature Medicine, 22(12), 1428-1438. https://doi.org/10.1038/nm.4222


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Noro, T., Namekata, K., Kimura, A., Guo, X., Azuchi, Y., Harada, C., … & Harada, T. (2015). Spermidine promotes retinal ganglion cell survival and optic nerve regeneration in adult mice following optic nerve injury. Cell Death and Disease, 6(4), e1720-e1720. https://doi.org/10.1038/cddis.2015.93


Ramot, Y., Tiede, S., Bíró, T., Bakar, M., Sugawara, K., Philpott, M., … & Paus, R. (2011). Spermidine promotes human hair growth and is a novel modulator of human epithelial stem cell functions. Plos One, 6(7), e22564. https://doi.org/10.1371/journal.pone.0022564


Yan, J., Wang, Y., Ling, Y., Song, X., Wang, S., Liu, H., … & Chen, A. (2019). Spermidine‐enhanced autophagic flux improves cardiac dysfunction following myocardial infarction by targeting the ampk/mtor signalling pathway. British Journal of Pharmacology, 176(17), 3126-3142. https://doi.org/10.1111/bph.1470


Adhikari, R., Shah, R., Reyes-Gordillo, K., Arellanes-Robledo, J., Cheng, Y., Ibrahim, J., … & Tuma, P. (2021). Spermidine prevents ethanol and lipopolysaccharide-induced hepatic injury in mice. Molecules, 26(6), 1786. https://doi.org/10.3390/molecules26061786


Eisenberg, T., Abdellatif, M., Schroeder, S., Primeßnig, U., Stekovic, S., Pendl, T., … & Madeo, F. (2016). Cardioprotection and lifespan extension by the natural polyamine spermidine. Nature Medicine, 22(12), 1428-1438. https://doi.org/10.1038/nm.4222


Han, W., Li, H., & Chen, B. (2022). Research progress and potential applications of spermidine in ocular diseases. Pharmaceutics, 14(7), 1500. https://doi.org/10.3390/pharmaceutics14071500


Levine, B. and Kroemer, G. (2019). Biological functions of autophagy genes: a disease perspective. Cell, 176(1-2), 11-42. https://doi.org/10.1016/j.cell.2018.09.048


Madeo, F., Eisenberg, T., Pietrocola, F., & Kroemer, G. (2018). Spermidine in health and disease. Science, 359(6374). https://doi.org/10.1126/science.aan2788


Matsumoto, M. (2020). Prevention of atherosclerosis by the induction of microbial polyamine production in the intestinal lumen. Biological and Pharmaceutical Bulletin, 43(2), 221-229. https://doi.org/10.1248/bpb.b19-00855


Muñoz-Esparza, N., Latorre-Moratalla, M., Comas-Basté, O., Toro-Funes, N., Veciana-Nogués, M., & Vidal-Carou, M. (2019). Polyamines in food. Frontiers in Nutrition, 6. https://doi.org/10.3389/fnut.2019.00108


Prasher, P., Sharma, M., Singh, S., Gulati, M., Chellappan, D., Rajput, R., … & Dua, K. (2023). Spermidine as a promising anticancer agent: recent advances and newer insights on its molecular mechanisms. Frontiers in Chemistry, 11. https://doi.org/10.3389/fchem.2023.1164477


Tong, D. and Hill, J. (2017). Spermidine promotes cardioprotective autophagy. Circulation Research, 120(8), 1229-1231. https://doi.org/10.1161/circresaha.117.310603


Zou, D., Zhao, Z., Li, L., Yu, M., Zhang, D., Ji, A., … & Wu, X. (2022). A comprehensive review of spermidine: safety, health effects, absorption and metabolism, food materials evaluation, physical and chemical processing, and bioprocessing. Comprehensive Reviews in Food Science and Food Safety, 21(3), 2820-2842. https://doi.org/10.1111/1541-4337.12963

Aihara, S., Torisu, K., Uchida, Y., Imazu, N., Nakano, T., & Kitazono, T. (2023). Spermidine from arginine metabolism activates nrf2 and inhibits kidney fibrosis. Communications Biology, 6(1). https://doi.org/10.1038/s42003-023-05057-w


Dodson, M., Castro-Portuguez, R., & Zhang, D. (2019). Nrf2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biology, 23, 101107. https://doi.org/10.1016/j.redox.2019.101107


Jiang, D., Guo, Y., Niu, C., Long, S., Jiang, Y., Wang, Z., … & Kang, B. (2023). Exploration of the antioxidant effect of spermidine on the ovary and screening and identification of differentially expressed proteins. International Journal of Molecular Sciences, 24(6), 5793. https://doi.org/10.3390/ijms24065793


Jiang, T., Huang, Z., Lin, Y., Zhang, Z., Fang, D., & Zhang, D. (2010). The protective role of nrf2 in streptozotocin-induced diabetic nephropathy. Diabetes, 59(4), 850-860. https://doi.org/10.2337/db09-1342


Liu, R., Wu, J., & Li, X. (2020). Self-eating: friend or foe? the emerging role of autophagy in fibrotic diseases. Theranostics, 10(18), 7993-8017. https://doi.org/10.7150/thno.47826


Niture, S., Khatri, R., & Jaiswal, A. (2014). Regulation of nrf2—an update. Free Radical Biology and Medicine, 66, 36-44. https://doi.org/10.1016/j.freeradbiomed.2013.02.008


Sykiotis, G. and Bohmann, D. (2008). Keap1/nrf2 signaling regulates oxidative stress tolerance and lifespan in drosophila. Developmental Cell, 14(1), 76-85. https://doi.org/10.1016/j.devcel.2007.12.002


Yang, Y., Li, Q., Wei, S., Chu, K., Xue, L., Liu, J., … & Tao, S. (2022). Stat6/vdr axis mitigates lung inflammatory injury by promoting nrf2 signaling pathway. Oxidative Medicine and Cellular Longevity, 2022, 1-15. https://doi.org/10.1155/2022/2485250

1 view0 comments

Yorumlar


bottom of page