Next Article in Journal
Melanin-Concentrating Hormone (MCH): Role in Mediating Reward-Motivated and Emotional Behavior and the Behavioral Disturbances Produced by Repeated Exposure to Reward Substances
Previous Article in Journal
D-Tryptophan Promotes Skin Wound Healing via Extracellular Matrix Remodeling in Normal and Diabetic Models
Previous Article in Special Issue
Updated Meta-Analysis on Vitamin Supplementation for Chronic Pruritus: Expanding Evidence Beyond Vitamin D
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Choline—An Essential Nutrient with Health Benefits and a Signaling Molecule

by
Brianne C. Burns
1,
Jitendra D. Belani
1,
Hailey N. Wittorf
1,
Eugen Brailoiu
2,*,† and
Gabriela C. Brailoiu
1,*
1
Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
2
Department of Neural Sciences and Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
*
Authors to whom correspondence should be addressed.
Current address: Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
Int. J. Mol. Sci. 2025, 26(15), 7159; https://doi.org/10.3390/ijms26157159
Submission received: 12 June 2025 / Revised: 20 July 2025 / Accepted: 22 July 2025 / Published: 24 July 2025

Abstract

Choline has been recognized as an essential nutrient involved in various physiological functions critical to human health. Adequate daily intake of choline has been established by the US National Academy of Medicine in 1998, considering choline requirements for different ages, sex differences and physiological states (e.g., pregnancy). By serving as a precursor for acetylcholine and phospholipids, choline is important for cholinergic transmission and the structural integrity of cell membranes. In addition, choline is involved in lipid and cholesterol transport and serves as a methyl donor after oxidation to betaine. Extracellular choline is transported across the cell membrane via various transport systems (high-affinity and low-affinity choline transporters) with distinct features and roles. An adequate dietary intake of choline during pregnancy supports proper fetal development, and throughout life supports brain, liver, and muscle functions, while choline deficiency is linked to disease states like fatty liver. Choline has important roles in neurodevelopment, cognition, liver function, lipid metabolism, and cardiovascular health. While its signaling role has been considered mostly indirect via acetylcholine and phosphatidylcholine which are synthesized from choline, emerging evidence supports a role for choline as an intracellular messenger acting on Sigma-1R, a non-opioid intracellular receptor. These new findings expand the cell signaling repertoire and increase the current understanding of the role of choline while warranting more research to uncover the molecular mechanisms and significance in the context of GPCR signaling, the relevance for physiology and disease states.

1. Introduction

Choline is a dietary component that has been recognized by the National Academy of Medicine (former Institute of Medicine) as an essential nutrient since 1998 [1]. Structurally, choline (C5H14NO+) is a quaternary ammonium compound characterized by a positively charged nitrogen atom bonded to three methyl groups and an ethanol group (Figure 1). Choline and its metabolites play a vital role in the body, from supporting brain function and neurotransmitter biosynthesis to maintaining liver health. Choline serves as a precursor for acetylcholine, a key neurotransmitter, important for functions such as memory, muscle control, and mood regulation [2,3]. Additionally, choline is integral to the synthesis of phosphatidylcholine and sphingomyelin, which are important for maintaining cell membrane structure and function. Choline supports lipid transport and metabolism within the liver; it is involved in the transport of lipids to various tissues by aiding in very-low-density lipoprotein (VLDL) formation, which helps prevent the accumulation of fats in the liver [4]. Furthermore, choline supports methyl group metabolism, serving as a precursor for S-adenosylmethionine, a critical methyl donor in the body. This methylation process helps maintain normal homocysteine levels, required for cardiovascular health [5]. Endogenous de novo synthesis via the sequential methylation of phosphatidylethanolamine produces limited quantities of choline, insufficient to meet physiological needs, therefore making dietary intake essential [2]. Deficiency in choline can lead to poor health outcomes, including cognitive deficit in infants [6] and liver dysfunction and muscle damage in adults [2].

2. Choline Intake Recommendations and Dietary Sources

Adequate daily intake (AI) of choline has been established by the US National Academy of Medicine-Food and Nutrition Board in 1998, considering choline requirements for different ages, sex differences, and physiological state, and prevention of liver damage in adults [1,7]. The choline AI increases with age: from 125 mg/day for infants (0–6 months) to 150 mg/day (6–12 months), 200 mg/day in children (1–3 years), and 250 mg/day (4–8 years). Boys and girls (9–13 years) have the same choline AI of 375 mg/day, while during adolescence (14–18 years), boys have a higher AI of choline (550 mg/day) as compared with girls (400 mg/day). A higher choline AI is needed for men (550 mg/day) as compared to women (425 mg/day) throughout the entire adult life (>19 years). The demand for choline is particularly high during pregnancy (450 mg/day) and lactation (500 mg/day), as it is critical for fetal development, particularly in brain and memory development [1,7].
Choline and its esters are widely present in various foods, with animal products: eggs, chicken, fish, beef, and dairy products serving as particularly rich sources [7,8]. Cruciferous vegetables and soy beans are also a good source of choline, providing up to 10% of the daily requirement per serving [8]. Choline is often taken as a supplement in the form of various salts such as choline chloride, choline bitartrate, and choline citrate (Figure 2). Some multivitamin supplements, including prenatal vitamins, contain small quantities (25–50 mg) of choline [7].
In the diet, choline is present in both the water-soluble form (phosphocholine, glycerophosphocholine, free choline) and the lipid-soluble form (phosphatidylcholine and sphingomyelin) that reach the liver via portal and lymphatic circulation, respectively [9]. While the dietary choline intake varies, overall suboptimal intake of choline in the United States was determined, and only 11% of the adult population achieves the AI for choline [10]. Early evidence indicates that the plasma level of choline is maintained, relatively constant, at 10 µM [11,12]. More recently, plasma choline level evaluated by liquid chromatography-tandem mass spectrometry was identified at 15.9 μM [13]. However, the plasma choline level decreases by 50% in choline-deficient diets or can increase to 20 µM after ingestion of choline-rich foods [12]. Given the wide-ranging physiological roles of choline and the increased demand during specific life stages, ensuring sufficient dietary intake of choline is essential for long-term health and disease prevention.

3. Mechanisms of Transport of Extracellular Choline

The transport of choline across the cellular membranes involves different transport systems, each playing distinct roles [12,14]. The high-affinity choline transporter (CHT1) (Km < 10 µM), predominantly located in cholinergic neurons, facilitates the transport of choline into presynaptic terminals, which is essential for acetylcholine production [14,15]. The choline uptake via CHT1 is sodium-dependent, inhibited by hemicholinium-3, and represents a rate-limiting step in acetylcholine synthesis. CHT1 is found in intracellular vesicles, such as endosomes and synaptic vesicles, and its presence at the plasma membrane is dynamically regulated through mechanisms involving endocytosis and exocytosis [16]. CHT1 availability at the cell surface is modulated based on neuronal activity levels with increased exocytosis of CHT1 to the plasma membrane during periods of intense cholinergic activity to meet the increased choline requirements [16], highlighting the significance of CHT1 in maintaining adequate choline levels in cholinergic neurons [12]. Conversely, dysregulation in choline transport through CHT1 is associated with neurological and psychiatric disorders such as Alzheimer’s disease, Parkinson’s disease, schizophrenia, attention-deficit hyperactive disorder (ADHD), and depression [17].
The choline transporter-like proteins family consists of five members (CTL1-5, encoded by SLC44A1-5) that mediate choline uptake for phospholipid synthesis in various tissues such as muscle, astrocytes, and cerebral cortex neurons [14]. The choline uptake activity of CTL1 has distinct features from CHT1 regarding the affinity for choline (intermediate versus high), sodium dependence (independent versus dependent), and inhibition by hemicholinium-3 (high Ki versus low Ki) [14].
In contrast, low-affinity choline transporters (Km > 30–100 µM), such as the organic cation transporter (OCT) family, are found ubiquitously; they enable the uptake of choline for synthesis of phosphatidylcholine and other phospholipids, which is vital for cell membrane integrity [12,14]. Unlike high-affinity transporters, low-affinity systems are sodium-independent [12]. In addition to the high-affinity and low-affinity choline transporters, unique choline transport mechanisms were identified in some tissues. The blood–brain barrier (BBB) employs specific carrier-mediated and saturable mechanisms to regulate choline passage into the brain, with mixed characteristics of both low and high-affinity transporters–sodium-independent and low Km [12]. Recently, human feline leukemia virus subgroup C receptor-related proteins 1 and 2 (FLVCR1 and FLVCR2) were identified as choline transporters [18], with FLVCR2 (also known as MFSD7C and SLC49A2) being considered a BBB choline transporter, responsible for the majority of choline uptake into the brain [19,20,21]. FLVCR2 is expressed in endothelial cells of the BBB and displays characteristics of choline uniporter or proton/choline co-transporter with high affinity for choline [19]. Overall, the transporters ensure a sufficient supply of choline for both acetylcholine production in neurons and phospholipid synthesis in various tissues, adapting to the dynamic needs of the body.

4. Biological Roles of Choline and Implications in Disease States

4.1. Choline and Fetal Development

During pregnancy, choline supports the neurodevelopment and overall health of the fetus [6,22]. Adequate choline intake during pregnancy positively influences infant brain function and early cognitive development [23] and plays a protective role in preventing neural tube defects [3]. A long-term (7 years) follow-up study found that children born to mothers who took choline supplements during pregnancy exhibited improved sustained attention, suggesting that prenatal choline intake may contribute to a lower risk of attention-related disorders [24]. Choline intake during pregnancy supports brain health by maintaining membrane integrity and reducing inflammation, which may protect against neurodegenerative conditions such as Alzheimer’s disease in the long term [25]. Prenatal choline supplementation can reduce the risk of brain-related developmental disorders in offspring, suggesting its preventive value against cognitive decline and supporting brain plasticity [25]. Insufficient choline intake among childbearing-age women may compromise fetal neurodevelopment and lead to adverse cognitive outcomes [26]. These studies highlight that prenatal choline supplementation not only supports immediate fetal development but also provides long-lasting cognitive advantages in children [6,24].
Beyond brain development, choline significantly impacts fetal liver function and metabolic health. Choline has an essential role in fetal liver maturation; choline deficiency during pregnancy can result in compromised liver function, potentially predisposing the offspring to metabolic disorders later in life [27]. Choline’s support of lipid transport and cellular membrane structure underscores its foundational role in liver development, emphasizing that an adequate supply during pregnancy is vital to reduce the risk of metabolic health issues in the offspring. Given these implications, the integration of choline into prenatal dietary recommendations is essential to protect against liver dysfunction and support long-term metabolic stability [27].
The benefits of choline in pregnancy are not isolated; they often involve complex interactions with other essential nutrients, notably docosahexaenoic acid (DHA) [28,29]. Prenatal choline supplementation in women already consuming DHA improved maternal biomarkers of DHA status, enhancing DHA levels in the blood [28,29]. This elevation in DHA has been associated with favorable neurodevelopmental outcomes, suggesting that choline and DHA together provide a synergistic benefit, amplifying neurodevelopmental support for the fetus [28,29]. Lysophosphatidylcholine facilitates the brain uptake of DHA [30]. Thereby, these findings underline the importance of not only choline but also its combination with DHA in prenatal supplementation, positioning both nutrients as essential components in maternal nutrition for optimal fetal development [6,28].
Moreover, the role of choline as a methyl donor in epigenetic processes is increasingly recognized for its impact on gene expression and neurodevelopmental outcomes; choline supports DNA and histone methylation, influencing genes that are critical for neurogenesis and synaptic plasticity [31]. Through these epigenetic mechanisms, choline supplementation during pregnancy may promote neural resilience and cognitive functions such as memory and learning [31]. In addition, choline modulates the expression of SOX4, a transcription factor crucial for cortical development, through specific epigenetic pathways [32]. This epigenetic influence of choline supports that adequate intake during pregnancy could confer long-lasting neuroprotective benefits, reducing the risk of neurodevelopmental disorders and age-related cognitive decline [31,32].

4.2. Choline and Liver Function

Several studies in various species and humans indicate that choline is essential for normal liver function by multiple mechanisms: phospholipids derived from choline are critical components of hepatic cell membrane and contribute to lipoprotein-mediated transport of triglycerides, formation of very-low-density lipoprotein (VLDL) and secretion of triglycerides from the liver [27]. In addition, choline serves as a methyl donor after oxidation to betaine that provides S-adenosylmethionine, the cofactor for methyltransferases [27]. Choline deficiency can lead to increased oxidative stress, inflammation, and fat accumulation in the liver, which, if untreated, may progress to more severe conditions such as non-alcoholic fatty liver disease (NAFLD), cirrhosis, and liver carcinoma [4]. Choline supplementation has been proposed as a therapeutic strategy for preventing or managing NAFLD by improving lipid metabolism, reducing inflammation, and protecting liver health, particularly in individuals with genetic predispositions to impaired choline metabolism [4].

4.3. Choline and Cardiovascular Health

Animal studies in rodents indicate that choline has cardiovascular protective effects in arrhythmias [33,34], reduces cardiac hypertrophy [35,36], attenuates cardiac fibrosis [37] and hypertension [38] by various mechanisms. Choline reduces cardiac hypertrophy by restoring the muscle-specific microRNA miR-133a expression, an anti-hypertrophic factor, and reducing the calcineurin protein level [36]. In spontaneous hypertensive rats, choline improves cardiac function and attenuates hypertension by increasing the vagal activity and exerting anti-inflammatory effect [38]. Recent population-based studies of 14,289 participants (mean age 48.08 years) [5] and 7341 older adults (mean age 73.39 years) [39] from the National Health and Nutrition Examination Survey (NHANES) indicate that a proper dietary choline intake is correlated with a reduced risk of cardiovascular disease. However, excessive dietary choline is metabolized by intestinal microbiota to trimethylamine, oxidized to trimethylamine N-oxide (TMAO); increased TMAO levels have been involved in atherosclerosis and are associated with a higher risk of major adverse cardiovascular events [40]. Moderate choline consumption is also linked to lower all-cause mortality, suggesting a potential role for choline in promoting longevity and supporting the inclusion of choline in dietary guidelines for heart health, disease prevention, and overall longevity [5].

4.4. Choline in Alzheimer’s Disease and Cognitive Decline

Choline has neuroprotective potential in mitigating age-related cognitive decline, particularly in conditions like Alzheimer’s disease [41,42]. Cholinergic deficit is one of the contributing factors to the pathogenesis of Alzheimer’s disease [43]. Anticholinergic drugs may exacerbate Alzheimer’s symptoms and accelerate the cognitive decline by increasing amyloid-beta levels and reducing phosphatidylcholine [41]. Maintaining adequate choline intake could help protect against cognitive deterioration by supporting acetylcholine synthesis and neuronal health. In the APP/PS1 mouse model of Alzheimer’s disease, lifelong choline supplementation reduced the amyloid-β plaque, microglia activation, and improved the spatial memory deficits [44]. A transgenerational reduction in Alzheimer’s disease pathology was found in APP/PS1 mice offspring from a mother with a choline-enriched diet and linked to the reduction in brain homocysteine level [45]. In the Ts65Dn mouse model of Down syndrome, an increase in choline intake during gestation and lactation improved cognition of the offspring [46,47]. Choline supplementation improved cognitive performance in patients with transient global amnesia [48] and reduced the chemotherapy-induced cognitive deficit in animal models [49]. Choline administered in combination with uridine and DHA enhances synapse formation and improves cognitive function in aging population [42]. Choline and uridine work in synergy with DHA to increase the synthesis of phosphatidylcholine and improve long-term outcomes in randomized controlled trials in patients with various forms of dementia, ranging from mild cognitive impairment to moderate Alzheimer’s disease [50]. Together, these findings suggest that choline, alongside other critical nutrients, could contribute to sustaining cognitive health, reducing Alzheimer’s disease pathology, and promoting brain resilience [41,42].

4.5. Choline and Addiction

Choline has shown promising potential as a therapeutic intervention for children with Fetal Alcohol Spectrum Disorder (FASD), a condition associated with prenatal alcohol exposure leading to cognitive deficits [51]. Choline intake improved cognitive performance, particularly in children diagnosed with FASD who have specific genetic variations in the SLC44A1 gene, which is involved in choline transport. These children exhibited greater cognitive gains in response to choline, suggesting that genetic factors may influence the effectiveness of choline as a therapeutic intervention, and raising the possibility of personalized choline supplementation for children affected by FASD. Overall, these findings highlight the importance of adequate choline intake in managing neurodevelopmental disorders like FASD and support further exploration of choline’s role in mitigating cognitive impairments linked to prenatal alcohol exposure [51]. We recently reported that choline is involved in the potentiation of orexin A signaling by cocaine, a drug of abuse [52]. Orexin A is an endogenous peptide involved in regulating wakefulness, energy metabolism, and reward [53,54].

4.6. Choline and Cancer

The relationship between choline intake and the risk of cancer remains a complex area of study. Choline deficiency can lead to liver dysfunction that may progress to fibrosis, cirrhosis, and liver cancer [4,55]. Population-based case–control studies and a meta-analysis of epidemiologic studies [56] indicate that high intake of choline and betaine reduced the risk of breast cancer [57], esophageal cancer [58], lung cancer [59], nasopharyngeal cancer [60], colon cancer [5], while did not impact the risk of renal cancer [61] or ovarian cancer [62]. A recent systematic review of choline metabolism in oncology [63] highlights two additional points. First, existing literature still focuses primarily on dietary intake rather than plasma choline levels. Second, in the limited studies that do measure circulating choline, plasma concentrations are mostly inversely associated with incident colorectal and pancreatic cancer, although one nested case–control analysis reported a positive colorectal signal. Moreover, studies measuring plasma choline levels were small and subject to selection bias [63]. Other studies found an increase in the risk of prostate cancer [64,65,66] or colorectal cancer [67] with higher choline intake. These findings suggest that while there may be a weak link between choline and cancer risk, it is not strong enough to warrant dietary changes solely for cancer prevention. Larger, prospective studies that integrate both dietary and plasma choline measures across diverse cancer types are needed to clarify any relationship between choline level and cancer risk and to better understand the impact of dietary interventions across different cancer types [5,63,64,67]. A summary of various roles of choline in physiology and disease states is provided in Figure 3.

5. Detection of Choline in Biological Samples

Accurate detection and quantification of choline in plasma and tissue samples are essential to fully understand its roles in metabolism, signaling, and disease pathology. Nuclear magnetic resonance spectroscopy (NMR), mass spectrometry (MS), and high-performance liquid chromatography (HPLC) have all been used to identify and quantify choline and related compounds. One of the original methods of detection used HPLC separation followed by electrochemical detection of hydrogen peroxide released from the reaction of choline with choline oxidase [68]. The method was subsequently used to measure choline in blood plasma with a linear response in the 1–20 μmol/L range [69]. Improvements in high-throughput detection methods, such as LC-MS and LC-MS/MS, have significantly enhanced the sensitivity, specificity, and efficiency of choline analyses [13]. Moreover, isotope dilution [70], enhanced sample preparation methods [71], and optimized chromatographic conditions [72] have helped improve the accuracy of choline quantification. The detection limits have also improved with studies demonstrating detection as low as 5 ng/L of choline, with strong accuracy and precision [13,73]. Although LC-MS remains significantly more sensitive to the detection of choline and its derivatives [73,74], NMR has played an alternate and important role in their detection and quantification. Initially, high-field 1H and 31P-NMR spectroscopy was used to quantify total choline as a cancer biomarker and it enabled differentiation between phosphocholine and glycerophosphocholine in tumor tissues [75]. Quadrupolar 14N NMR has been explored as an alternative detection method, leveraging the higher natural abundance and sensitivity of 14N, despite challenges related to probe compatibility [76]. More recently, advances in NMR hardware and analytical algorithms have improved the clinical and regulatory relevance of NMR methods. A clinical NMR-based assay was developed to quantify choline with good sensitivity and reproducibility using the Vantera® clinical analyzer [77]. This method employs a non-negative deconvolution algorithm to isolate choline’s spectral signal and demonstrates a strong correlation with LC-MS/MS (R = 0.998), with quantification limits of 7.1 μmol/L in serum and 5.9 μmol/L in plasma [77]. In a separate study reporting threshold impurity for pharmaceutical quality control meeting International Council for Harmonisation (ICH) requirements for impurity detection, NMR has demonstrated the ability to detect choline impurities, such as O-(2-hydroxyethyl)choline, at levels as low as 0.01% in choline chloride samples using high-field 1H NMR spectroscopy [78]. These results reject the widespread assumption that NMR lacks sufficient sensitivity for impurity analysis and highlights its utility even at benchtop field strengths under optimized conditions. Furthermore, de Graaf et al. recently described a 2D-1H–14N heteronuclear single-quantum coherence (HSQC) NMR method that enables simultaneous detection of both protonated and deuterated choline metabolites, including choline, phosphocholine, glycerophosphocholine, CDP-choline, and betaine, in excised tissues [79]. The technique improves analytical resolution and facilitates metabolic tracing of exogenous choline sources by utilizing scalar coupling between 14N and CH2 protons and the chemical shift sensitivity of 14N to nearby deuterium, enabling high-resolution discrimination of metabolite species. This capability is especially important in experimental and nutritional studies, and it enhances the analytical resolution beyond traditional 1H or 2H magnetic resonance spectroscopy. It also supports metabolic tracing using deuterated choline (e.g., D9-choline) and enables quantification of isotopic enrichment alongside absolute concentration measurements [79]. These advances have transformed choline detection from general quantification to precise molecular profiling, allowing insights into dynamic metabolism, impurity control, and metabolic imaging. Once viewed as less sensitive than MS, NMR now offers strong, complementary capabilities that are especially valuable in metabolic tracing, clinical diagnostics, and pharmaceutical quality assurance.

6. Second Messenger Role for Choline Acting on Sigma-1R

The signaling role of choline has been considered mostly indirect via acetylcholine and phosphatidylcholine synthesized from choline. Phospholipase D (PLD) hydrolyzes phosphatidylcholine, the most abundant membrane phospholipid in mammalian cells, releasing choline and phosphatidic acid (PA) [80,81]. PA has been considered the main signaling molecule produced from phosphatidylcholine [80]. Relatively recently, we have identified that choline acts as an intracellular messenger that links extracellular stimuli to intracellular calcium signaling pathways by activating Sigma-1 receptors (Sigma-1R) [82], a non-opioid intracellular receptor located on the endoplasmic reticulum [83,84,85,86,87,88]. Sigma-1Rs bind various ligands, most of which are amines, such as antidepressants (e.g., fluoxetine), antipsychotics (e.g., haloperidol), and drugs of abuse (e.g., cocaine and methamphetamine) [88,89,90,91,92]. Choline, but not its metabolites phosphocholine or betaine, binds Sigma-1R and enhances inositol 1,4,5-trisphosphate (IP3)-evoked Ca2+ release from the endoplasmic reticulum [82]. Therefore, G-protein coupled receptors (GPCRs) signal to IP3Rs through two pathways, IP3 and choline, that converge to the stimulation of IP3Rs (Figure 4). IP3 is generated together with diacylglycerol (DAG) from phosphatidylinositol-4,5-bis-phosphate (PIP2) by phospholipase C (PLC). Several GPCR agonists such as bradykinin, angiotensin II, endothelin-1, carbachol, orexin, and thyroid-stimulating hormone activate PLD and PLC [93,94,95,96,97]. Gq-coupled receptors like AT1 receptor or muscarinic M3 receptor activate PLC and PLD via RhoA and PKC-dependent process [96,98], while other GPCRs, like alpha-1 adrenergic receptors activate PLD via a PKC-independent process [97,99].
The basal PLD activity in mammalian cells is low and transiently increases in response to receptor activation [94]. Mammals have six different PLD enzymes, with PLD1 and PLD2 being the best characterized; there are 2 splice variants for PLD1 and 3 splice variants for PLD2 [100]. PLD isoenzymes have a wide tissue distribution; at the cellular level, PLD1 is localized in the endoplasmic reticulum, Golgi and endosomes [101]; PLD2 is present in the plasma membrane [102]; PLD3 and PLD4 are localized to lysosomes [103,104] and PLD5 and PLD6 are localized to mitochondria [104].
Choline meets the five criteria for a second messenger formulated by Sutherland [105,106,107]. The first criterion set out by Sutherland for a second messenger is that antagonism of the action of the messenger blocks the effects of the extracellular messenger. In support of this criterion, we found that the Ca2+ signals evoked by bradykinin, a GPCR agonist that stimulates PLC and IP3R-evoked increase in Ca2+, were attenuated by BD 1047, a Sigma-1R antagonist [52,108] in NG108-15 cells, neuroblastoma-glioma cells that express Sigma-1R [82]. Reduction in Sigma-1R expression by transfection of cells with Sigma-1R shRNA reduced the amplitude of Ca2+ signals produced by bradykinin or ATP [82] another GPCR agonist that stimulates PLC via P2Y6 receptors in NG108-15 cells [109].
Sutherland’s second criterion is that when the molecule is applied intracellularly, it must mimic the effect of an extracellular stimulus. Multiple lines of evidence indicate that in different cell types Sigma-1Rs potentiate the IP3-evoked increase in cytosolic Ca2+ concentration [110,111,112]. To address this criterion, we determined the effect of microinjection of choline alone or in co-injection with IP3 on cytosolic Ca2+ concentration; microinjection of choline potentiated the IP3-evoked Ca2+ signals in cells endogenously expressing Sigma-1R or transfected with the receptor [82] similarly to the potentiation produced by other agonists [112,113].
The third criterion for the second messenger is that it can be synthesized and metabolized. The pathways for synthesis and metabolism of choline are well-characterized and widely accepted: choline is synthesized by PLD from phosphatidylcholine [80] and is metabolized by phosphorylation to phosphocholine, an inactive derivative, or by oxidation to betaine in the kidney, liver, and brain [114].
The fourth criterion of Sutherland is that the second messenger levels change in response to a physiologically relevant stimulus. Stimulation of NG108-15 cells with ATP increased intracellular choline and IP3 levels; knockdown of PLD1 and PLD2 using shRNA prevented the ATP-induced increase in choline, while it did not affect the IP3 level [82]. These results indicate that stimulation with ATP promotes choline synthesis via a PLD-dependent mechanism.
Sutherland’s fifth criterion for a second messenger is the presence of specific intracellular binding sites. To address this criterion, we performed a competitive binding assay in membranes prepared from Neuro-2A cells stably expressing Sigma-1R incubated with [3H](+) pentazocine, a high-affinity selective Sigma-1R ligand [115] and choline. Choline completely displaced the specific binding of [3H](+) pentazocine (Ki = 525 µM) while phosphocholine, the major choline metabolite, did not displace it; betaine and acetylcholine were less effective than choline [82]. These results support that choline binds to greater affinity than its metabolites to the same site as Sigma-1R ligands [116,117].
Sigma-1R has been considered a promising therapeutic target for several neurological conditions such as Alzheimer’s, Huntington’s and Parkinson’s disease, epilepsy, amyotrophic lateral sclerosis [118,119,120,121,122,123,124,125,126,127,128,129], cognitive and affective disorders [130], psychiatric diseases [131], neuropathic pain [132,133,134], cardiovascular diseases [135,136,137], chronic kidney disease [137,138] and cancer [139,140,141].
Choline-Sigma 1R signaling downstream to GPCR activation is an emerging new concept with potential implications for substance use disorders and eating disorders [52,142,143], spatial memory [130], cognition [44], blood–brain barrier permeability [144], cardiac fibrosis [37] and cancer [66,141]. This new signaling mechanism has been mentioned in the context of Ca2+ signaling in oomycetes [145], as a potential mechanism for the antiviral effect of choline in microglial cells [146] and in the endoplasmic reticulum-mitochondrial calcium handling via FLVCR1a (feline leukemia virus subgroup C receptor 1) [147]. Moreover, PLD dysregulation and choline-Sigma1R may play a role in colorectal cancer and glioblastoma via cross-talk with PI3K-Akt/Wnt/β-catenin pathways [148].
The concept of choline as a second messenger downstream to GPCR stimulation enriches the cellular signaling repertoire and supports the need for further studies to investigate the molecular mechanisms through which GPCR agonists generate choline, to understand interactions with other second messengers and to elucidate its significance in health and disease states.

Author Contributions

Conceptualization, B.C.B., E.B., G.C.B.; writing—original draft preparation, B.C.B., J.D.B., H.N.W., E.B., G.C.B.; writing—review and editing, B.C.B., J.D.B., H.N.W., E.B., G.C.B. visualization, B.C.B., J.D.B., E.B., G.C.B.; supervision, J.D.B., E.B., G.C.B.; funding acquisition, E.B., G.C.B. All authors have read and agreed to the published version of the manuscript.

Funding

Research reported in this publication was supported by the National Institute On Drug Abuse of the National Institutes of Health under Award Number R01DA054921.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AIAdequate daily intake
CHT1High-affinity choline transporter
CTLCholine transporter-like proteins
DAGDiacyl glycerol
DHADocosahexaenoic acid
FASDFetal Alcohol Spectrum Disorder
GPCRG protein-coupled receptor
IP3Inositol 1,4,5-trisphosphate
IP3RInositol 1,4,5-trisphosphate (IP3) receptor
PKCProtein kinase C
PLCPhospholipase C
PLDPhospholipase D
VLDLVery-low-density lipoprotein

References

  1. InstituteMedicine. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline; The National Academies Press: Washington, DC, USA, 1998. [Google Scholar]
  2. Zeisel, S.H.; Da Costa, K.A.; Franklin, P.D.; Alexander, E.A.; Lamont, J.T.; Sheard, N.F.; Beiser, A. Choline, an essential nutrient for humans. FASEB J. 1991, 5, 2093–2098. [Google Scholar] [CrossRef] [PubMed]
  3. Zeisel, S.H.; Blusztajn, J.K. Choline and human nutrition. Annu. Rev. Nutr. 1994, 14, 269–296. [Google Scholar] [CrossRef] [PubMed]
  4. Corbin, K.D.; Zeisel, S.H. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr. Opin. Gastroenterol. 2012, 28, 159–165. [Google Scholar] [CrossRef] [PubMed]
  5. Jieru, P.; Zhang, S.; Cai, L.; Long, W.; Wang, Y.; Zhang, L.; Dong, Y.; Zhang, W.; Liao, J.; Yang, C. Dietary choline intake and health outcomes in U.S. adults: Exploring the impact on cardiovascular disease, cancer prevalence, and all-cause mortality. J. Health Popul. Nutr. 2024, 43, 59. [Google Scholar] [CrossRef] [PubMed]
  6. Wallace, T.C.; Blusztajn, J.K.; Caudill, M.A.; Klatt, K.C.; Zeisel, S.H. Choline: The Neurocognitive Essential Nutrient of Interest to Obstetricians and Gynecologists. J. Diet. Suppl. 2020, 17, 733–752. [Google Scholar] [CrossRef] [PubMed]
  7. Zeisel, S.H.; Klatt, K.C.; Caudill, M.A. Choline. Adv. Nutr. 2018, 9, 58–60. [Google Scholar] [CrossRef] [PubMed]
  8. Zeisel, S.H.; Mar, M.H.; Howe, J.C.; Holden, J.M. Concentrations of choline-containing compounds and betaine in common foods. J. Nutr. 2003, 133, 1302–1307. [Google Scholar] [CrossRef] [PubMed]
  9. Wiedeman, A.M.; Barr, S.I.; Green, T.J.; Xu, Z.; Innis, S.M.; Kitts, D.D. Dietary Choline Intake: Current State of Knowledge Across the Life Cycle. Nutrients 2018, 10, 1513. [Google Scholar] [CrossRef] [PubMed]
  10. Wallace, T.C.; Fulgoni, V.L., 3rd. Assessment of Total Choline Intakes in the United States. J. Am. Coll. Nutr. 2016, 35, 108–112. [Google Scholar] [CrossRef] [PubMed]
  11. Bligh, J. The level of free choline in plasma. J. Physiol. 1952, 117, 234–240. [Google Scholar] [CrossRef] [PubMed]
  12. Lockman, P.R.; Allen, D.D. The transport of choline. Drug Dev. Ind. Pharm. 2002, 28, 749–771. [Google Scholar] [CrossRef] [PubMed]
  13. Guerra, G.; Segrado, F.; Pasanisi, P.; Bruno, E.; Lopez, S.; Raspagliesi, F.; Bianchi, M.; Venturelli, E. Circulating choline and phosphocholine measurement by a hydrophilic interaction liquid chromatography-tandem mass spectrometry. Heliyon 2023, 9, e21921. [Google Scholar] [CrossRef] [PubMed]
  14. Haga, T. Molecular properties of the high-affinity choline transporter CHT1. J. Biochem. 2014, 156, 181–194. [Google Scholar] [CrossRef] [PubMed]
  15. Sarter, M.; Parikh, V. Choline transporters, cholinergic transmission and cognition. Nat. Rev. Neurosci. 2005, 6, 48–56. [Google Scholar] [CrossRef] [PubMed]
  16. Ribeiro, F.M.; Black, S.A.; Prado, V.F.; Rylett, R.J.; Ferguson, S.S.; Prado, M.A. The “ins” and “outs” of the high-affinity choline transporter CHT1. J. Neurochem. 2006, 97, 1–12. [Google Scholar] [CrossRef] [PubMed]
  17. Ojiakor, O.A.; Rylett, R.J. Modulation of sodium-coupled choline transporter CHT function in health and disease. Neurochem. Int. 2020, 140, 104810. [Google Scholar] [CrossRef] [PubMed]
  18. Ri, K.; Weng, T.H.; Claveras Cabezudo, A.; Josting, W.; Zhang, Y.; Bazzone, A.; Leong, N.C.P.; Welsch, S.; Doty, R.T.; Gursu, G.; et al. Molecular mechanism of choline and ethanolamine transport in humans. Nature 2024, 630, 501–508. [Google Scholar] [CrossRef] [PubMed]
  19. Cater, R.J.; Mukherjee, D.; Gil-Iturbe, E.; Erramilli, S.K.; Chen, T.; Koo, K.; Santander, N.; Reckers, A.; Kloss, B.; Gawda, T.; et al. Structural and molecular basis of choline uptake into the brain by FLVCR2. Nature 2024, 629, 704–709. [Google Scholar] [CrossRef] [PubMed]
  20. Nguyen, X.T.A.; Le, T.N.U.; Nguyen, T.Q.; Thi Thuy Ha, H.; Artati, A.; Leong, N.C.P.; Nguyen, D.T.; Lim, P.Y.; Susanto, A.V.; Huang, Q.; et al. MFSD7c functions as a transporter of choline at the blood-brain barrier. Cell Res. 2024, 34, 245–257. [Google Scholar] [CrossRef] [PubMed]
  21. Scala, M.; Leong, N.C.P.; Le, T.N.U.; Zhang, Y.; Wu, Y.; Severino, M.; Madia, F.; Nosrati, M.S.S.; Dostmohammadi, A.; Capra, V.; et al. A hypomorphic FLVCR2 variant resulting in moderate transport deficiency causes hydranencephaly syndrome with brain calcifications. Eur. J. Hum. Genet. 2025. [Google Scholar] [CrossRef] [PubMed]
  22. Taesuwan, S.; McDougall, M.Q.; Malysheva, O.V.; Bender, E.; Nevins, J.E.H.; Devapatla, S.; Vidavalur, R.; Caudill, M.A.; Klatt, K.C. Choline metabolome response to prenatal choline supplementation across pregnancy: A randomized controlled trial. FASEB J. 2021, 35, e22063. [Google Scholar] [CrossRef] [PubMed]
  23. Caudill, M.A.; Strupp, B.J.; Muscalu, L.; Nevins, J.E.H.; Canfield, R.L. Maternal choline supplementation during the third trimester of pregnancy improves infant information processing speed: A randomized, double-blind, controlled feeding study. FASEB J. 2018, 32, 2172–2180. [Google Scholar] [CrossRef] [PubMed]
  24. Bahnfleth, C.L.; Strupp, B.J.; Caudill, M.A.; Canfield, R.L. Prenatal choline supplementation improves child sustained attention: A 7-year follow-up of a randomized controlled feeding trial. FASEB J. 2022, 36, e22054. [Google Scholar] [CrossRef] [PubMed]
  25. Blusztajn, J.K.; Slack, B.E.; Mellott, T.J. Neuroprotective Actions of Dietary Choline. Nutrients 2017, 9, 815. [Google Scholar] [CrossRef] [PubMed]
  26. Herrera-Cuenca, M.; Yepez Garcia, M.C.; Cortes Sanabria, L.Y.; Hernandez, P.; Ramirez, G.; Vasquez, M.; Sifontes, Y.; Gomez, G.; Liria-Dominguez, M.R.; Rigotti, A.; et al. Inadequate Intake of Choline and Essential Fatty Acids in Latin American Childbearing-Age Women as a Regional Pre-Conceptional Disadvantage: ELANS Results. Nutrients 2024, 16, 3150. [Google Scholar] [CrossRef] [PubMed]
  27. Obeid, R.; Schon, C.; Derbyshire, E.; Jiang, X.; Mellott, T.J.; Blusztajn, J.K.; Zeisel, S.H. A Narrative Review on Maternal Choline Intake and Liver Function of the Fetus and the Infant; Implications for Research, Policy, and Practice. Nutrients 2024, 16, 260. [Google Scholar] [CrossRef] [PubMed]
  28. Klatt, K.C.; McDougall, M.Q.; Malysheva, O.V.; Taesuwan, S.; Loinard-Gonzalez, A.A.P.; Nevins, J.E.H.; Beckman, K.; Bhawal, R.; Anderson, E.; Zhang, S.; et al. Prenatal choline supplementation improves biomarkers of maternal docosahexaenoic acid (DHA) status among pregnant participants consuming supplemental DHA: A randomized controlled trial. Am. J. Clin. Nutr. 2022, 116, 820–832. [Google Scholar] [CrossRef] [PubMed]
  29. Bernhard, W.; Bockmann, K.; Maas, C.; Mathes, M.; Hovelmann, J.; Shunova, A.; Hund, V.; Schleicher, E.; Poets, C.F.; Franz, A.R. Combined choline and DHA supplementation: A randomized controlled trial. Eur. J. Nutr. 2020, 59, 729–739. [Google Scholar] [CrossRef] [PubMed]
  30. Lo Van, A.; Bernoud-Hubac, N.; Lagarde, M. Esterification of Docosahexaenoic Acid Enhances Its Transport to the Brain and Its Potential Therapeutic Use in Brain Diseases. Nutrients 2022, 14, 4550. [Google Scholar] [CrossRef] [PubMed]
  31. Blusztajn, J.K.; Mellott, T.J. Choline nutrition programs brain development via DNA and histone methylation. Cent. Nerv. Syst. Agents Med. Chem. 2012, 12, 82–94. [Google Scholar] [CrossRef] [PubMed]
  32. Paules, E.M.; Silva-Gomez, J.A.; Friday, W.B.; Zeisel, S.H.; Trujillo-Gonzalez, I. Choline Regulates SOX4 through miR-129-5p and Modifies H3K27me3 in the Developing Cortex. Nutrients 2023, 15, 2774. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, S.; Han, H.M.; Jiang, Y.N.; Wang, C.; Song, H.X.; Pan, Z.Y.; Fan, K.; Du, J.; Fan, Y.H.; Du, Z.M.; et al. Activation of cardiac M3 muscarinic acetylcholine receptors has cardioprotective effects against ischaemia-induced arrhythmias. Clin. Exp. Pharmacol. Physiol. 2012, 39, 343–349. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, Y.; Sun, H.L.; Li, D.L.; Wang, L.Y.; Gao, Y.; Wang, Y.P.; Du, Z.M.; Lu, Y.J.; Yang, B.F. Choline produces antiarrhythmic actions in animal models by cardiac M3 receptors: Improvement of intracellular Ca2+ handling as a common mechanism. Can. J. Physiol. Pharmacol. 2008, 86, 860–865. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, S.; Han, H.M.; Pan, Z.W.; Hang, P.Z.; Sun, L.H.; Jiang, Y.N.; Song, H.X.; Du, Z.M.; Liu, Y. Choline inhibits angiotensin II-induced cardiac hypertrophy by intracellular calcium signal and p38 MAPK pathway. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2012, 385, 823–831. [Google Scholar] [CrossRef] [PubMed]
  36. Zhao, Y.; Wang, C.; Wu, J.; Wang, Y.; Zhu, W.; Zhang, Y.; Du, Z. Choline protects against cardiac hypertrophy induced by increased after-load. Int. J. Biol. Sci. 2013, 9, 295–302. [Google Scholar] [CrossRef] [PubMed]
  37. Zhao, L.; Chen, T.; Hang, P.; Li, W.; Guo, J.; Pan, Y.; Du, J.; Zheng, Y.; Du, Z. Choline Attenuates Cardiac Fibrosis by Inhibiting p38MAPK Signaling Possibly by Acting on M(3) Muscarinic Acetylcholine Receptor. Front. Pharmacol. 2019, 10, 1386. [Google Scholar] [CrossRef] [PubMed]
  38. Liu, L.; Lu, Y.; Bi, X.; Xu, M.; Yu, X.; Xue, R.; He, X.; Zang, W. Choline ameliorates cardiovascular damage by improving vagal activity and inhibiting the inflammatory response in spontaneously hypertensive rats. Sci. Rep. 2017, 7, 42553. [Google Scholar] [CrossRef] [PubMed]
  39. Li, W.; Liu, S.; Meng, X.; Liu, H. A nutrient wide association study of cardiovascular disease prevalence in older adults from NHANES 2007 to 2018. Sci. Rep. 2025, 15, 12710. [Google Scholar] [CrossRef] [PubMed]
  40. Tang, W.H.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [PubMed]
  41. Wurtman, R.J. How Anticholinergic Drugs Might Promote Alzheimer’s Disease: More Amyloid-beta and Less Phosphatidylcholine. J. Alzheimer’s Dis. 2015, 46, 983–987. [Google Scholar] [CrossRef] [PubMed]
  42. Wurtman, R.J. Synapse formation in the brain can be enhanced by co-administering three specific nutrients. Eur. J. Pharmacol. 2017, 817, 20–21. [Google Scholar] [CrossRef] [PubMed]
  43. Hampel, H.; Mesulam, M.M.; Cuello, A.C.; Khachaturian, A.S.; Vergallo, A.; Farlow, M.R.; Snyder, P.J.; Giacobini, E.; Khachaturian, Z.S. Revisiting the Cholinergic Hypothesis in Alzheimer’s Disease: Emerging Evidence from Translational and Clinical Research. J. Prev. Alzheimer’s Dis. 2019, 6, 2–15. [Google Scholar] [CrossRef] [PubMed]
  44. Velazquez, R.; Ferreira, E.; Knowles, S.; Fux, C.; Rodin, A.; Winslow, W.; Oddo, S. Lifelong choline supplementation ameliorates Alzheimer’s disease pathology and associated cognitive deficits by attenuating microglia activation. Aging Cell 2019, 18, e13037. [Google Scholar] [CrossRef] [PubMed]
  45. Velazquez, R.; Ferreira, E.; Winslow, W.; Dave, N.; Piras, I.S.; Naymik, M.; Huentelman, M.J.; Tran, A.; Caccamo, A.; Oddo, S. Maternal choline supplementation ameliorates Alzheimer’s disease pathology by reducing brain homocysteine levels across multiple generations. Mol. Psychiatry 2019, 25, 2620–2629. [Google Scholar] [CrossRef] [PubMed]
  46. Velazquez, R.; Ash, J.A.; Powers, B.E.; Kelley, C.M.; Strawderman, M.; Luscher, Z.I.; Ginsberg, S.D.; Mufson, E.J.; Strupp, B.J. Maternal choline supplementation improves spatial learning and adult hippocampal neurogenesis in the Ts65Dn mouse model of Down syndrome. Neurobiol. Dis. 2013, 58, 92–101. [Google Scholar] [CrossRef] [PubMed]
  47. Powers, B.E.; Kelley, C.M.; Velazquez, R.; Ash, J.A.; Strawderman, M.S.; Alldred, M.J.; Ginsberg, S.D.; Mufson, E.J.; Strupp, B.J. Maternal choline supplementation in a mouse model of Down syndrome: Effects on attention and nucleus basalis/substantia innominata neuron morphology in adult offspring. Neuroscience 2017, 340, 501–514. [Google Scholar] [CrossRef] [PubMed]
  48. Rahmanian, S.; Shapouri, M.; Mohammadian, M.K.; Mahmoudi, Z.; Saeedirad, Z.; Mobarakeh, K.A.; Parhiz, A.; Shekari, S.; Harsini, A.R.; Valisoltani, N.; et al. Does choline have an effect on Transient Global Amnesia (TGA)? BMC Neurosci. 2024, 25, 72. [Google Scholar] [CrossRef] [PubMed]
  49. Johns, B.E.; Ficken, M.; Engberg, M.E.; Wecker, L.; Philpot, R.M. Increasing dietary choline attenuates spatial memory deficits resulting from exposure to the chemotherapeutic agents cyclophosphamide and doxorubicin. J. Psychopharmacol. 2021, 35, 1300–1309. [Google Scholar] [CrossRef] [PubMed]
  50. Baumel, B.S.; Doraiswamy, P.M.; Sabbagh, M.; Wurtman, R. Potential Neuroregenerative and Neuroprotective Effects of Uridine/Choline-Enriched Multinutrient Dietary Intervention for Mild Cognitive Impairment: A Narrative Review. Neurol. Ther. 2021, 10, 43–60. [Google Scholar] [CrossRef] [PubMed]
  51. Smith, S.M.; Virdee, M.S.; Eckerle, J.K.; Sandness, K.E.; Georgieff, M.K.; Boys, C.J.; Zeisel, S.H.; Wozniak, J.R. Polymorphisms in SLC44A1 are associated with cognitive improvement in children diagnosed with fetal alcohol spectrum disorder: An exploratory study of oral choline supplementation. Am. J. Clin. Nutr. 2021, 114, 617–627. [Google Scholar] [CrossRef] [PubMed]
  52. Barr, J.L.; Zhao, P.; Brailoiu, G.C.; Brailoiu, E. Choline-Sigma-1R as an Additional Mechanism for Potentiation of Orexin by Cocaine. Int. J. Mol. Sci. 2021, 22, 5160. [Google Scholar] [CrossRef] [PubMed]
  53. de Lecea, L.; Kilduff, T.S.; Peyron, C.; Gao, X.; Foye, P.E.; Danielson, P.E.; Fukuhara, C.; Battenberg, E.L.; Gautvik, V.T.; Bartlett, F.S., 2nd; et al. The hypocretins: Hypothalamus-specific peptides with neuroexcitatory activity. Proc. Natl. Acad. Sci. USA 1998, 95, 322–327. [Google Scholar] [CrossRef] [PubMed]
  54. Mahler, S.V.; Smith, R.J.; Moorman, D.E.; Sartor, G.C.; Aston-Jones, G. Multiple roles for orexin/hypocretin in addiction. Prog. Brain Res. 2012, 198, 79–121. [Google Scholar] [CrossRef] [PubMed]
  55. Cheung, O.; Sanyal, A.J. Recent advances in nonalcoholic fatty liver disease. Curr. Opin. Gastroenterol. 2010, 26, 202–208. [Google Scholar] [CrossRef] [PubMed]
  56. Sun, S.; Li, X.; Ren, A.; Du, M.; Du, H.; Shu, Y.; Zhu, L.; Wang, W. Choline and betaine consumption lowers cancer risk: A meta-analysis of epidemiologic studies. Sci. Rep. 2016, 6, 35547. [Google Scholar] [CrossRef] [PubMed]
  57. Xu, X.; Gammon, M.D.; Zeisel, S.H.; Bradshaw, P.T.; Wetmur, J.G.; Teitelbaum, S.L.; Neugut, A.I.; Santella, R.M.; Chen, J. High intakes of choline and betaine reduce breast cancer mortality in a population-based study. FASEB J. 2009, 23, 4022–4028. [Google Scholar] [CrossRef] [PubMed]
  58. Ibiebele, T.I.; Hughes, M.C.; Pandeya, N.; Zhao, Z.; Montgomery, G.; Hayward, N.; Green, A.C.; Whiteman, D.C.; Webb, P.M.; Study of Digestive, H.; et al. High intake of folate from food sources is associated with reduced risk of esophageal cancer in an Australian population. J. Nutr. 2011, 141, 274–283. [Google Scholar] [CrossRef] [PubMed]
  59. Ying, J.; Rahbar, M.H.; Hallman, D.M.; Hernandez, L.M.; Spitz, M.R.; Forman, M.R.; Gorlova, O.Y. Associations between dietary intake of choline and betaine and lung cancer risk. PLoS ONE 2013, 8, e54561. [Google Scholar] [CrossRef] [PubMed]
  60. Zeng, F.F.; Xu, C.H.; Liu, Y.T.; Fan, Y.Y.; Lin, X.L.; Lu, Y.K.; Zhang, C.X.; Chen, Y.M. Choline and betaine intakes are associated with reduced risk of nasopharyngeal carcinoma in adults: A case-control study. Br. J. Cancer 2014, 110, 808–816. [Google Scholar] [CrossRef] [PubMed]
  61. Cho, E.; Giovannucci, E.L.; Joh, H.K. Nutrients related to one-carbon metabolism and risk of renal cell cancer. Cancer Causes Control 2013, 24, 373–382. [Google Scholar] [CrossRef] [PubMed]
  62. Kotsopoulos, J.; Hankinson, S.E.; Tworoger, S.S. Dietary betaine and choline intake are not associated with risk of epithelial ovarian cancer. Eur. J. Clin. Nutr. 2010, 64, 111–114. [Google Scholar] [CrossRef] [PubMed]
  63. Yao, N.; Li, W.; Xu, G.; Duan, N.; Yu, G.; Qu, J. Choline metabolism and its implications in cancer. Front. Oncol. 2023, 13, 1234887. [Google Scholar] [CrossRef] [PubMed]
  64. Han, P.; Bidulescu, A.; Barber, J.R.; Zeisel, S.H.; Joshu, C.E.; Prizment, A.E.; Vitolins, M.Z.; Platz, E.A. Dietary choline and betaine intakes and risk of total and lethal prostate cancer in the Atherosclerosis Risk in Communities (ARIC) Study. Cancer Causes Control 2019, 30, 343–354. [Google Scholar] [CrossRef] [PubMed]
  65. Richman, E.L.; Kenfield, S.A.; Stampfer, M.J.; Giovannucci, E.L.; Zeisel, S.H.; Willett, W.C.; Chan, J.M. Choline intake and risk of lethal prostate cancer: Incidence and survival. Am. J. Clin. Nutr. 2012, 96, 855–863. [Google Scholar] [CrossRef] [PubMed]
  66. Oyer, H.M.; Sanders, C.M.; Kim, F.J. Small-Molecule Modulators of Sigma1 and Sigma2/TMEM97 in the Context of Cancer: Foundational Concepts and Emerging Themes. Front. Pharmacol. 2019, 10, 1141. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, A.Y.; Matich, E.K.; Laryea, J.; Hsu, P.C.; Su, L.J. A Case-Control Study of Dietary Choline Intake and Risk of Colorectal Cancer Modified by Dietary B-Vitamin Intake. Nutrients 2024, 16, 4200. [Google Scholar] [CrossRef] [PubMed]
  68. Potter, P.E.; Meek, J.L.; Neff, N.H. Acetylcholine and choline in neuronal tissue measured by HPLC with electrochemical detection. J. Neurochem. 1983, 41, 188–194. [Google Scholar] [CrossRef] [PubMed]
  69. Webb, L.E.; Johnson, R.C. Choline in plasma measured by liquid-chromatography with electrochemical detection. Clin. Biochem. 1986, 19, 212–215. [Google Scholar] [CrossRef] [PubMed]
  70. Koc, H.; Mar, M.H.; Ranasinghe, A.; Swenberg, J.A.; Zeisel, S.H. Quantitation of choline and its metabolites in tissues and foods by liquid chromatography/electrospray ionization-isotope dilution mass spectrometry. Anal. Chem. 2002, 74, 4734–4740. [Google Scholar] [CrossRef] [PubMed]
  71. Mimmi, M.C.; Picotti, P.; Corazza, A.; Betto, E.; Pucillo, C.E.; Cesaratto, L.; Cedolini, C.; Londero, V.; Zuiani, C.; Bazzocchi, M.; et al. High-performance metabolic marker assessment in breast cancer tissue by mass spectrometry. Clin. Chem. Lab. Med. 2011, 49, 317–324. [Google Scholar] [CrossRef] [PubMed]
  72. Xiong, Y.; Zhao, Y.Y.; Goruk, S.; Oilund, K.; Field, C.J.; Jacobs, R.L.; Curtis, J.M. Validation of an LC-MS/MS method for the quantification of choline-related compounds and phospholipids in foods and tissues. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2012, 911, 170–179. [Google Scholar] [CrossRef] [PubMed]
  73. Lamy, E.; Pilyser, L.; Paquet, C.; Bouaziz-Amar, E.; Grassin-Delyle, S. High-sensitivity quantification of acetylcholine and choline in human cerebrospinal fluid with a validated LC-MS/MS method. Talanta 2021, 224, 121881. [Google Scholar] [CrossRef] [PubMed]
  74. Hefni, M.E.; Bergstrom, M.; Lennqvist, T.; Fagerstrom, C.; Witthoft, C.M. Simultaneous quantification of trimethylamine N-oxide, trimethylamine, choline, betaine, creatinine, and propionyl-, acetyl-, and L-carnitine in clinical and food samples using HILIC-LC-MS. Anal. Bioanal. Chem. 2021, 413, 5349–5360. [Google Scholar] [CrossRef] [PubMed]
  75. Loening, N.M.; Chamberlin, A.M.; Zepeda, A.G.; Gonzalez, R.G.; Cheng, L.L. Quantification of phosphocholine and glycerophosphocholine with 31P edited 1H NMR spectroscopy. NMR Biomed. 2005, 18, 413–420. [Google Scholar] [CrossRef] [PubMed]
  76. Ruiz-Muelle, A.B.; Moreno, P.G.; Fernandez, I. Quantitative quadrupolar NMR (qQNMR) using nitrogen-14 for the determination of choline in complex matrixes. Talanta 2021, 230, 122344. [Google Scholar] [CrossRef] [PubMed]
  77. Garcia, E.; Shalaurova, I.; Matyus, S.P.; Wolak-Dinsmore, J.; Oskardmay, D.N.; Connelly, M.A. Quantification of choline in serum and plasma using a clinical nuclear magnetic resonance analyzer. Clin. Chim. Acta 2022, 524, 106–112. [Google Scholar] [CrossRef] [PubMed]
  78. Achanta, P.S.; Niemitz, M.; Friesen, J.B.; Tadjimukhamedov, F.K.; Bzhelyansky, A.; Giancaspro, G.I.; Chen, S.N.; Pauli, G.F. Pharmaceutical analysis by NMR can accommodate strict impurity thresholds: The case of choline. J. Pharm. Biomed. Anal. 2022, 214, 114709. [Google Scholar] [CrossRef] [PubMed]
  79. de Graaf, R.A.; Thomas, M.A.; De Feyter, H.M. Metabolism of Choline and Deuterated Choline Detected by (1)H-(14)N 2D Heteronuclear Single-Quantum Coherence (HSQC) NMR. Anal. Chem. 2025, 97, 6586–6593. [Google Scholar] [CrossRef] [PubMed]
  80. Exton, J.H. Phospholipase D. Ann. N. Y. Acad. Sci. 2000, 905, 61–68. [Google Scholar] [CrossRef] [PubMed]
  81. Onono, F.O.; Morris, A.J. Phospholipase D and Choline Metabolism. Handb. Exp. Pharmacol. 2020, 259, 205–218. [Google Scholar] [CrossRef] [PubMed]
  82. Brailoiu, E.; Chakraborty, S.; Brailoiu, G.C.; Zhao, P.; Barr, J.L.; Ilies, M.A.; Unterwald, E.M.; Abood, M.E.; Taylor, C.W. Choline Is an Intracellular Messenger Linking Extracellular Stimuli to IP3-Evoked Ca2+ Signals through Sigma-1 Receptors. Cell Rep. 2019, 26, 330–337.e4. [Google Scholar] [CrossRef] [PubMed]
  83. Hayashi, T.; Su, T.P. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca2+ signaling and cell survival. Cell 2007, 131, 596–610. [Google Scholar] [CrossRef] [PubMed]
  84. Lachance, V.; Belanger, S.M.; Hay, C.; Le Corvec, V.; Banouvong, V.; Lapalme, M.; Tarmoun, K.; Beaucaire, G.; Lussier, M.P.; Kourrich, S. Overview of Sigma-1R Subcellular Specific Biological Functions and Role in Neuroprotection. Int. J. Mol. Sci. 2023, 24, 1971. [Google Scholar] [CrossRef] [PubMed]
  85. Sharma, N.; Patel, C.; Shenkman, M.; Kessel, A.; Ben-Tal, N.; Lederkremer, G.Z. The Sigma-1 receptor is an ER-localized type II membrane protein. J. Biol. Chem. 2021, 297, 101299. [Google Scholar] [CrossRef] [PubMed]
  86. Zhemkov, V.; Ditlev, J.A.; Lee, W.R.; Wilson, M.; Liou, J.; Rosen, M.K.; Bezprozvanny, I. The role of sigma 1 receptor in organization of endoplasmic reticulum signaling microdomains. eLife 2021, 10, e65192. [Google Scholar] [CrossRef] [PubMed]
  87. Pergolizzi, J.; Varrassi, G.; Coleman, M.; Breve, F.; Christo, D.K.; Christo, P.J.; Moussa, C. The Sigma Enigma: A Narrative Review of Sigma Receptors. Cureus 2023, 15, e35756. [Google Scholar] [CrossRef] [PubMed]
  88. Schmidt, H.R.; Kruse, A.C. The Molecular Function of sigma Receptors: Past, Present, and Future. Trends Pharmacol. Sci. 2019, 40, 636–654. [Google Scholar] [CrossRef] [PubMed]
  89. Walker, J.M.; Bowen, W.D.; Walker, F.O.; Matsumoto, R.R.; De Costa, B.; Rice, K.C. Sigma receptors: Biology and function. Pharmacol. Rev. 1990, 42, 355–402. [Google Scholar] [CrossRef] [PubMed]
  90. Maurice, T.; Su, T.P. The pharmacology of sigma-1 receptors. Pharmacol. Ther. 2009, 124, 195–206. [Google Scholar] [CrossRef] [PubMed]
  91. Lombardo, L.; Mirabile, S.; Gitto, R.; Cosentino, G.; Alcaro, S.; Dichiara, M.; Marrazzo, A.; Amata, E.; Ortuso, F.; De Luca, L. Exploring Structural Requirements for Sigma-1 Receptor Linear Ligands: Experimental and Computational Approaches. J. Chem. Inf. Model. 2024, 64, 5701–5711. [Google Scholar] [CrossRef] [PubMed]
  92. Li, J.; Satyshur, K.A.; Guo, L.W.; Ruoho, A.E. Sphingoid Bases Regulate the Sigma-1 Receptor-Sphingosine and N,N’-Dimethylsphingosine Are Endogenous Agonists. Int. J. Mol. Sci. 2023, 24, 3013. [Google Scholar] [CrossRef] [PubMed]
  93. Schmidt, M.; Fasselt, B.; Rumenapp, U.; Bienek, C.; Wieland, T.; van Koppen, C.J.; Jakobs, K.H. Rapid and persistent desensitization of m3 muscarinic acetylcholine receptor-stimulated phospholipase D. Concomitant sensitization of phospholipase C. J. Biol. Chem. 1995, 270, 19949–19956. [Google Scholar] [CrossRef] [PubMed]
  94. Cockcroft, S. Signalling roles of mammalian phospholipase D1 and D2. Cell Mol. Life Sci. 2001, 58, 1674–1687. [Google Scholar] [CrossRef] [PubMed]
  95. Jantti, M.H.; Putula, J.; Somerharju, P.; Frohman, M.A.; Kukkonen, J.P. OX1 orexin/hypocretin receptor activation of phospholipase D. Br. J. Pharmacol. 2012, 165, 1109–1123. [Google Scholar] [CrossRef] [PubMed]
  96. Meacci, E.; Nuti, F.; Catarzi, S.; Vasta, V.; Donati, C.; Bourgoin, S.; Bruni, P.; Moss, J.; Vaughan, M. Activation of phospholipase D by bradykinin and sphingosine 1-phosphate in A549 human lung adenocarcinoma cells via different GTP-binding proteins and protein kinase C delta signaling pathways. Biochemistry 2003, 42, 284–292. [Google Scholar] [CrossRef] [PubMed]
  97. Oude Weernink, P.A.; Han, L.; Jakobs, K.H.; Schmidt, M. Dynamic phospholipid signaling by G protein-coupled receptors. Biochim. Biophys. Acta 2007, 1768, 888–900. [Google Scholar] [CrossRef] [PubMed]
  98. Du, G.; Altshuller, Y.M.; Kim, Y.; Han, J.M.; Ryu, S.H.; Morris, A.J.; Frohman, M.A. Dual requirement for rho and protein kinase C in direct activation of phospholipase D1 through G protein-coupled receptor signaling. Mol. Biol. Cell 2000, 11, 4359–4368. [Google Scholar] [CrossRef] [PubMed]
  99. Balboa, M.A.; Insel, P.A. Stimulation of phospholipase D via alpha1-adrenergic receptors in Madin-Darby canine kidney cells is independent of PKCalpha and -epsilon activation. Mol. Pharmacol. 1998, 53, 221–227. [Google Scholar] [CrossRef] [PubMed]
  100. Jenkins, G.M.; Frohman, M.A. Phospholipase D: A lipid centric review. Cell Mol. Life Sci. 2005, 62, 2305–2316. [Google Scholar] [CrossRef] [PubMed]
  101. Brown, F.D.; Thompson, N.; Saqib, K.M.; Clark, J.M.; Powner, D.; Thompson, N.T.; Solari, R.; Wakelam, M.J. Phospholipase D1 localises to secretory granules and lysosomes and is plasma-membrane translocated on cellular stimulation. Curr. Biol. 1998, 8, 835–838. [Google Scholar] [CrossRef] [PubMed]
  102. Du, G.; Huang, P.; Liang, B.T.; Frohman, M.A. Phospholipase D2 localizes to the plasma membrane and regulates angiotensin II receptor endocytosis. Mol. Biol. Cell 2004, 15, 1024–1030. [Google Scholar] [CrossRef] [PubMed]
  103. Gonzalez, A.C.; Schweizer, M.; Jagdmann, S.; Bernreuther, C.; Reinheckel, T.; Saftig, P.; Damme, M. Unconventional Trafficking of Mammalian Phospholipase D3 to Lysosomes. Cell Rep. 2018, 22, 1040–1053. [Google Scholar] [CrossRef] [PubMed]
  104. Singh, S.; Dransfeld, U.E.; Ambaw, Y.A.; Lopez-Scarim, J.; Farese, R.V., Jr.; Walther, T.C. PLD3 and PLD4 synthesize S,S-BMP, a key phospholipid enabling lipid degradation in lysosomes. Cell 2024, 187, 6820–6834.e24. [Google Scholar] [CrossRef] [PubMed]
  105. Sutherland, E.W. On the biological role of cyclic AMP. JAMA 1970, 214, 1281–1288. [Google Scholar] [CrossRef] [PubMed]
  106. Sutherland, E.W.; Robison, G.A. The role of cyclic-3′,5′-AMP in responses to catecholamines and other hormones. Pharmacol. Rev. 1966, 18, 145–161. [Google Scholar] [CrossRef] [PubMed]
  107. Aley, P.K.; Singh, N.; Brailoiu, G.C.; Brailoiu, E.; Churchill, G.C. Nicotinic acid adenine dinucleotide phosphate (NAADP) is a second messenger in muscarinic receptor-induced contraction of guinea pig trachea. J. Biol. Chem. 2013, 288, 10986–10993. [Google Scholar] [CrossRef] [PubMed]
  108. Son, J.S.; Kwon, Y.B. Sigma-1 Receptor Antagonist BD1047 Reduces Allodynia and Spinal ERK Phosphorylation Following Chronic Compression of Dorsal Root Ganglion in Rats. Korean J. Physiol. Pharmacol. 2010, 14, 359–364. [Google Scholar] [CrossRef] [PubMed]
  109. Sak, K.; Samuel, K.; Kelve, M.; Webb, T.E. Pharmacological characterisation of pyrimidinoceptor responses in NG108-15 cells. Eur. J. Pharmacol. 2001, 415, 127–133. [Google Scholar] [CrossRef] [PubMed]
  110. Hayashi, T.; Maurice, T.; Su, T.P. Ca2+ signaling via sigma(1)-receptors: Novel regulatory mechanism affecting intracellular Ca2+ concentration. J. Pharmacol. Exp. Ther. 2000, 293, 788–798. [Google Scholar] [CrossRef] [PubMed]
  111. Hong, W.; Nuwayhid, S.J.; Werling, L.L. Modulation of bradykinin-induced calcium changes in SH-SY5Y cells by neurosteroids and sigma receptor ligands via a shared mechanism. Synapse 2004, 54, 102–110. [Google Scholar] [CrossRef] [PubMed]
  112. Wu, Z.; Bowen, W.D. Role of sigma-1 receptor C-terminal segment in inositol 1,4,5-trisphosphate receptor activation: Constitutive enhancement of calcium signaling in MCF-7 tumor cells. J. Biol. Chem. 2008, 283, 28198–28215. [Google Scholar] [CrossRef] [PubMed]
  113. Stricker, H.M.; Rommerswinkel, N.; Keil, S.; Gnoth, S.A.; Niggemann, B.; Dittmar, T. The phospholipase D inhibitor FIPI potently blocks EGF-induced calcium signaling in human breast cancer cells. Cell Commun. Signal 2021, 19, 43. [Google Scholar] [CrossRef] [PubMed]
  114. Li, Z.; Vance, D.E. Phosphatidylcholine and choline homeostasis. J. Lipid Res. 2008, 49, 1187–1194. [Google Scholar] [CrossRef] [PubMed]
  115. de Costa, B.R.; Bowen, W.D.; Hellewell, S.B.; Walker, J.M.; Thurkauf, A.; Jacobson, A.E.; Rice, K.C. Synthesis and evaluation of optically pure [3H]-(+)-pentazocine, a highly potent and selective radioligand for sigma receptors. FEBS Lett. 1989, 251, 53–58. [Google Scholar] [CrossRef] [PubMed]
  116. Fu, C.; Xiao, Y.; Zhou, X.; Sun, Z. Insight into binding of endogenous neurosteroid ligands to the sigma-1 receptor. Nat. Commun. 2024, 15, 5619. [Google Scholar] [CrossRef] [PubMed]
  117. Morato, X.; Fernandez-Duenas, V.; Perez-Villamor, P.; Valle-Leon, M.; Vela, J.M.; Merlos, M.; Burgueno, J.; Ciruela, F. Development of a Novel sigma(1) Receptor Biosensor Based on Its Heterodimerization with Binding Immunoglobulin Protein in Living Cells. ACS Chem. Neurosci. 2023, 14, 2201–2207. [Google Scholar] [CrossRef] [PubMed]
  118. Goldberg, Y.P.; Navon-Perry, L.; Cruz-Herranz, A.; Chen, K.; Hecker-Barth, G.; Spiegel, K.; Cohen, Y.; Niethammer, M.; Tan, A.M.; Schuring, H.; et al. The Safety Profile of Pridopidine, a Novel Sigma-1 Receptor Agonist for the Treatment of Huntington’s Disease. CNS Drugs 2025, 39, 485–498. [Google Scholar] [CrossRef] [PubMed]
  119. Djebari, S.; Jimenez-Herrera, R.; Iborra-Lazaro, G.; Jimenez-Diaz, L.; Navarro-Lopez, J.D. Social and contextual memory impairments induced by Amyloid-beta oligomers are rescued by Sigma-1 receptor activation. Biomed. Pharmacother. 2025, 184, 117914. [Google Scholar] [CrossRef] [PubMed]
  120. Martinez-Orozco, H.; Bencomo-Martinez, A.; Maya-Arteaga, J.P.; Rubio-De Anda, P.F.; Sanabria-Romero, F.; Casas, Z.G.M.; Rodriguez-Vargas, I.; Hernandez-Puga, A.G.; Sablon-Carrazana, M.; Menendez-Soto Del Valle, R.; et al. CNEURO-201, an Anti-amyloidogenic Agent and sigma1-Receptor Agonist, Improves Cognition in the 3xTg Mouse Model of Alzheimer’s Disease by Multiple Actions in the Pathology. Int. J. Mol. Sci. 2025, 26, 1301. [Google Scholar] [CrossRef] [PubMed]
  121. Sweed, E.; Khodir, S.A.; Motawea, S.M.; El-Haron, H.; Mostafa, B.A.; Elkholy, M.S.; Salim, M.; Shebl, D.Z.M. Targeting the sigma-1 receptor with pridopidine induces functional neurorestoration in spinal cord ischemia-reperfusion injury. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2025, 398, 9307–9321. [Google Scholar] [CrossRef] [PubMed]
  122. Shokr, M.M.; Badawi, G.A.; Elshazly, S.M.; Zaki, H.F.; Mohamed, A.F. Sigma 1 Receptor and Its Pivotal Role in Neurological Disorders. ACS Pharmacol. Transl. Sci. 2025, 8, 47–65. [Google Scholar] [CrossRef] [PubMed]
  123. Ngo, A.; Fattakhov, N.; Toborek, M. Sigma-1 receptor signaling: A potential therapeutic approach for ischemic stroke. J. Cereb. Blood Flow. Metab. 2024, 44, 1430–1440. [Google Scholar] [CrossRef] [PubMed]
  124. Cheng, D.; Lei, Z.G.; Chu, K.; Lam, O.J.H.; Chiang, C.Y.; Zhang, Z.J. N, N-Dimethyltryptamine, a natural hallucinogen, ameliorates Alzheimer’s disease by restoring neuronal Sigma-1 receptor-mediated endoplasmic reticulum-mitochondria crosstalk. Alzheimers Res. Ther. 2024, 16, 95. [Google Scholar] [CrossRef] [PubMed]
  125. Piechal, A.; Jakimiuk, A.; Mirowska-Guzel, D. Sigma receptors and neurological disorders. Pharmacol. Rep. 2021, 73, 1582–1594. [Google Scholar] [CrossRef] [PubMed]
  126. Wang, T.; Jia, H. The Sigma Receptors in Alzheimer’s Disease: New Potential Targets for Diagnosis and Therapy. Int. J. Mol. Sci. 2023, 24, 2025. [Google Scholar] [CrossRef] [PubMed]
  127. Siddiqui, T.; Bhatt, L.K. Targeting Sigma-1 Receptor: A Promising Strategy in the Treatment of Parkinson’s Disease. Neurochem. Res. 2023, 48, 2925–2935. [Google Scholar] [CrossRef] [PubMed]
  128. Wu, N.H.; Ye, Y.; Wan, B.B.; Yu, Y.D.; Liu, C.; Chen, Q.J. Emerging Benefits: Pathophysiological Functions and Target Drugs of the Sigma-1 Receptor in Neurodegenerative Diseases. Mol. Neurobiol. 2021, 58, 5649–5666. [Google Scholar] [CrossRef] [PubMed]
  129. Zhemkov, V.; Geva, M.; Hayden, M.R.; Bezprozvanny, I. Sigma-1 Receptor (S1R) Interaction with Cholesterol: Mechanisms of S1R Activation and Its Role in Neurodegenerative Diseases. Int. J. Mol. Sci. 2021, 22, 4082. [Google Scholar] [CrossRef] [PubMed]
  130. Salaciak, K.; Pytka, K. Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neurosci. Biobehav. Rev. 2022, 132, 1114–1136. [Google Scholar] [CrossRef] [PubMed]
  131. Ren, P.; Wang, J.Y.; Xu, M.J.; Chen, H.L.; Duan, J.Y.; Li, Y.F. Sigma-1 receptor activation produces faster antidepressant-like effect through enhancement of hippocampal neuroplasticity: Focus on sigma-1-5-HT1A heteroreceptor complex. Neurochem. Int. 2025, 184, 105937. [Google Scholar] [CrossRef] [PubMed]
  132. An, Y.; Cao, S.; Shi, L.; Zhang, Y.; Wang, X.; Yuan, S.; Shi, Y.; Wang, B.; Liu, J.; Han, C.J. Pharmacological modulation of Sigma-1 receptor ameliorates pathological neuroinflammation in rats with diabetic neuropathic pain via the AKT/GSK-3beta/NF-kappaB pathway. Brain Res. Bull. 2025, 221, 111226. [Google Scholar] [CrossRef] [PubMed]
  133. Galvez, R.; Mayoral, V.; Cebrecos, J.; Medel, F.J.; Morte, A.; Sust, M.; Vaque, A.; Montes-Perez, A.; Neira-Reina, F.; Canovas, L.; et al. E-52862-A selective sigma-1 receptor antagonist, in peripheral neuropathic pain: Two randomized, double-blind, phase 2 studies in patients with chronic postsurgical pain and painful diabetic neuropathy. Eur. J. Pain. 2025, 29, e4755. [Google Scholar] [CrossRef] [PubMed]
  134. Peng, Y.; Zhang, A.H.; Wei, L.; Welsh, W.J. Preclinical Evaluation of Sigma 1 Receptor Antagonists as a Novel Treatment for Painful Diabetic Neuropathy. ACS Pharmacol. Transl. Sci. 2024, 7, 2358–2368. [Google Scholar] [CrossRef] [PubMed]
  135. Almaamari, A.; Sultan, M.; Zhang, T.; Qaed, E.; Wu, S.; Qiao, R.; Duan, Y.; Ding, S.; Liu, G.; Su, S. Sigma-1 Receptor Specific Biological Functions, Protective Role, and Therapeutic Potential in Cardiovascular Diseases. Cardiovasc. Toxicol. 2025, 25, 614–630. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, Y.; Chen, Q.; Yang, J.Z.; Li, X.W.; Chen, L.J.; Zhang, K.K.; Liu, J.L.; Li, J.H.; Hsu, C.; Chen, L.; et al. Multi-Omics Analysis Reveals the Role of Sigma-1 Receptor in a Takotsubo-like Cardiomyopathy Model. Biomedicines 2023, 11, 2766. [Google Scholar] [CrossRef] [PubMed]
  137. Munguia-Galaviz, F.J.; Miranda-Diaz, A.G.; Cardenas-Sosa, M.A.; Echavarria, R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int. J. Mol. Sci. 2023, 24, 1997. [Google Scholar] [CrossRef] [PubMed]
  138. Balogh, D.B.; Hodrea, J.; Saeed, A.; Cserhalmi, M.; Rozsahegyi, A.; Lakat, T.; Lenart, L.; Szabo, A.J.; Wagner, L.J.; Fekete, A. Sigma-1 Receptor as a Novel Therapeutic Target in Diabetic Kidney Disease. Int. J. Mol. Sci. 2024, 25, 3327. [Google Scholar] [CrossRef] [PubMed]
  139. Leotta, C.G.; Barbaraci, C.; Fiorito, J.; Coco, A.; di Giacomo, V.; Amata, E.; Marrazzo, A.; Pitari, G.M. HDAC/sigma1R Dual-Ligand as a Targeted Melanoma Therapeutic. Pharmaceuticals 2025, 18, 179. [Google Scholar] [CrossRef] [PubMed]
  140. Thomas, J.D.; Longen, C.G.; Oyer, H.M.; Chen, N.; Maher, C.M.; Salvino, J.M.; Kania, B.; Anderson, K.N.; Ostrander, W.F.; Knudsen, K.E.; et al. Sigma1 Targeting to Suppress Aberrant Androgen Receptor Signaling in Prostate Cancer. Cancer Res. 2017, 77, 2439–2452. [Google Scholar] [CrossRef] [PubMed]
  141. Robinson, T.S.; Osman, M.A. An Emerging Role for Sigma Receptor 1 in Personalized Treatment of Breast Cancer. Cancers 2023, 15, 3464. [Google Scholar] [CrossRef] [PubMed]
  142. Knowles, L.G.; Armanious, A.J.; Peng, Y.; Welsh, W.J.; James, M.H. Recent advances in drug discovery efforts targeting the sigma 1 receptor system: Implications for novel medications designed to reduce excessive drug and food seeking. Addict. Neurosci. 2023, 8, 100126. [Google Scholar] [CrossRef] [PubMed]
  143. Nakamura, Y.; Dryanovski, D.I.; Kimura, Y.; Jackson, S.N.; Woods, A.S.; Yasui, Y.; Tsai, S.Y.; Patel, S.; Covey, D.P.; Su, T.P.; et al. Cocaine-induced endocannabinoid signaling mediated by sigma-1 receptors and extracellular vesicle secretion. eLife 2019, 8, e47209. [Google Scholar] [CrossRef] [PubMed]
  144. Brailoiu, E.; Barr, J.L.; Wittorf, H.N.; Inan, S.; Unterwald, E.M.; Brailoiu, G.C. Modulation of the Blood-Brain Barrier by Sigma-1R Activation. Int. J. Mol. Sci. 2024, 25, 5147. [Google Scholar] [CrossRef] [PubMed]
  145. Nair, I.M.; Condon, E.; Prestwich, B.D.; Mackrill, J.J. Myo-D-inositol Trisphosphate Signalling in Oomycetes. Microorganisms 2022, 10, 2157. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, S.H.; Damborsky, J.C.; Wilson, B.C.; Fannin, R.D.; Ward, J.M.; Gerrish, K.E.; He, B.; Martin, N.P.; Yakel, J.L. alpha7 nicotinic receptor activation mitigates herpes simplex virus type 1 infection in microglia cells. Antivir. Res. 2024, 228, 105934. [Google Scholar] [CrossRef] [PubMed]
  147. Bertino, F.; Mukherjee, D.; Bonora, M.; Bagowski, C.; Nardelli, J.; Metani, L.; Zanin Venturini, D.I.; Chianese, D.; Santander, N.; Salaroglio, I.C.; et al. Dysregulation of FLVCR1a-dependent mitochondrial calcium handling in neural progenitors causes congenital hydrocephalus. Cell Rep. Med. 2024, 5, 101647. [Google Scholar] [CrossRef] [PubMed]
  148. Lim, S.H.; Lee, H.; Lee, H.J.; Kim, K.; Choi, J.; Han, J.M.; Min, D.S. PLD1 is a key player in cancer stemness and chemoresistance: Therapeutic targeting of cross-talk between the PI3K/Akt and Wnt/beta-catenin pathways. Exp. Mol. Med. 2024, 56, 1479–1487. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of choline and its related metabolites. The molecular structures of choline, phosphocholine, and glycerophosphocholine are illustrated. Phosphocholine and glycerophosphocholine are key compounds involved in choline metabolism.
Figure 1. Chemical structures of choline and its related metabolites. The molecular structures of choline, phosphocholine, and glycerophosphocholine are illustrated. Phosphocholine and glycerophosphocholine are key compounds involved in choline metabolism.
Ijms 26 07159 g001
Figure 2. Structures of choline salts. Shown are the structures of three choline salts commonly used in dietary supplements and clinical formulations: choline chloride, choline bitartrate, and choline citrate. These salts differ in their counterions, affecting their solubility, stability, and bioavailability.
Figure 2. Structures of choline salts. Shown are the structures of three choline salts commonly used in dietary supplements and clinical formulations: choline chloride, choline bitartrate, and choline citrate. These salts differ in their counterions, affecting their solubility, stability, and bioavailability.
Ijms 26 07159 g002
Figure 3. Diagram summarizing the roles of choline in health and disease states. Choline, an essential nutrient, can be transported intracellularly via various transport mechanisms. It serves as a precursor for acetylcholine, a critical neurotransmitter, and contributes to fetal development, liver function, and cardiovascular health. Choline deficit has been involved in cardiovascular diseases, liver dysfunction, Alzheimer’s disease, and addiction, while both deficit and excess of choline were associated with cancer.
Figure 3. Diagram summarizing the roles of choline in health and disease states. Choline, an essential nutrient, can be transported intracellularly via various transport mechanisms. It serves as a precursor for acetylcholine, a critical neurotransmitter, and contributes to fetal development, liver function, and cardiovascular health. Choline deficit has been involved in cardiovascular diseases, liver dysfunction, Alzheimer’s disease, and addiction, while both deficit and excess of choline were associated with cancer.
Ijms 26 07159 g003
Figure 4. Diagram illustrating the second messenger role of choline. G protein-coupled receptor (GPCR) agonists that stimulate phospholipase C (PLC) and phospholipase D (PLD) lead to consequent formation of inositol 1,4,5-trisphosphate (IP3) and choline. IP3 stimulates IP3 receptor (IP3R), while choline binds to Sigma-1 receptors (Sigma-1R), and potentiates IP3R activity.
Figure 4. Diagram illustrating the second messenger role of choline. G protein-coupled receptor (GPCR) agonists that stimulate phospholipase C (PLC) and phospholipase D (PLD) lead to consequent formation of inositol 1,4,5-trisphosphate (IP3) and choline. IP3 stimulates IP3 receptor (IP3R), while choline binds to Sigma-1 receptors (Sigma-1R), and potentiates IP3R activity.
Ijms 26 07159 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Burns, B.C.; Belani, J.D.; Wittorf, H.N.; Brailoiu, E.; Brailoiu, G.C. Choline—An Essential Nutrient with Health Benefits and a Signaling Molecule. Int. J. Mol. Sci. 2025, 26, 7159. https://doi.org/10.3390/ijms26157159

AMA Style

Burns BC, Belani JD, Wittorf HN, Brailoiu E, Brailoiu GC. Choline—An Essential Nutrient with Health Benefits and a Signaling Molecule. International Journal of Molecular Sciences. 2025; 26(15):7159. https://doi.org/10.3390/ijms26157159

Chicago/Turabian Style

Burns, Brianne C., Jitendra D. Belani, Hailey N. Wittorf, Eugen Brailoiu, and Gabriela C. Brailoiu. 2025. "Choline—An Essential Nutrient with Health Benefits and a Signaling Molecule" International Journal of Molecular Sciences 26, no. 15: 7159. https://doi.org/10.3390/ijms26157159

APA Style

Burns, B. C., Belani, J. D., Wittorf, H. N., Brailoiu, E., & Brailoiu, G. C. (2025). Choline—An Essential Nutrient with Health Benefits and a Signaling Molecule. International Journal of Molecular Sciences, 26(15), 7159. https://doi.org/10.3390/ijms26157159

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop