Next Article in Journal
Key Epigenetic Players in Etiology and Novel Combinatorial Therapies for Treatment of Hepatocellular Carcinoma
Previous Article in Journal
Abscopal Effect with Liver-Directed Therapy: A Review of the Current Literature and Future Directions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring Endogenous Tryptamines: Overlooked Agents Against Fibrosis in Chronic Disease? A Narrative Review

Independent Researcher, New York, NY 11101, USA
Livers 2024, 4(4), 615-637; https://doi.org/10.3390/livers4040043
Submission received: 7 October 2024 / Revised: 21 November 2024 / Accepted: 26 November 2024 / Published: 28 November 2024

Abstract

:
Long regarded as illicit substances with no clinical value, N-dimethylated tryptamines—such as N,N-dimethyltryptamine, 5-methoxy-N,N-dimethyltryptamine, and bufotenine—have been found to produce naturally in a wide variety of species, including humans. Known for their psychoactive effects through serotonin receptors (5-HTRs), N-dimethylated tryptamines are currently being reinvestigated clinically for their long-term benefits in mental disorders. Endogenous tryptamine is methylated by indolethylamine-N-methyltransferase (INMT), which can then serve as an agonist to pro-survival pathways, such as sigma non-opioid intracellular receptor 1 (SIGMAR1) signaling. Fibrogenic diseases, like metabolic-associated fatty liver disease (MAFLD), steatohepatitis (MASH), and chronic kidney disease (CKD) have shown changes in INMT and SIGMAR1 activity in the progression of disease pathogenesis. At the cellular level, endothelial cells and fibroblasts have been found to express INMT in various tissues; however, little is known about tryptamines in endothelial injury and fibrosis. In this review, I will give an overview of the biochemistry, molecular biology, and current evidence of INMT’s role in hepatic fibrogenesis. I will also discuss current pre-clinical and clinical findings of N-methylated tryptamines and highlight new and upcoming therapeutic strategies that may be adapted for mitigating fibrogenic diseases. Finally, I will mention recent findings for mutualistic gut bacteria influencing endogenous tryptamine signaling and metabolism.

1. Introduction

The enactment of the U.S. Controlled Substances Act of 1971 marked a pivotal shift in international drug regulation and domestic healthcare oversight, influencing not only the pharmaceutical landscape but also hindering therapeutic research in psychedelics and disproportionately affecting marginalized communities through its enforcement [1,2,3,4,5,6]. Among some of the first substances deemed to have no medical utility, to be unsafe for treatments under medical supervision, and to have a high potential for abuse are the potent, short-lived psychedelic N,N-methylated tryptamines: N,N-dimethyltryptamine (DMT) as well as bufotenine (5-HO-DMT) and later, 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) in 2011 [1,7,8]. N-methylated tryptamines have since been found to be a component of indigenous South American ethnobotanical ceremonies pre-dating Western colonization [9]. Today, N-methylated tryptamines are often misused outside of medical and scientific research, particularly by younger people and gender and sexual minorities [10,11,12]. However, their clinical application in treating depression, anxiety, substance dependency, and post-traumatic stress disorder (PTSD) has been gaining traction [13,14,15,16].
In the modern era, DMT was first synthesized in 1931 by Richard Manske and characterized in nature in 1946 by Oswaldo Gonçalves de Lima from the root bark of Mimosa hostilis [17,18]. The psychoactive properties and metabolism of DMT were made akin to lysergic acid diethylamide (LSD-25) and mescaline when characterized in humans in 1956 by Stephen Szára [19]. Since then, N,N-methylated tryptamines have had research focus on their effects on the brain, substance misuse, and psychotherapeutics [20,21,22]. Recent robust in vitro and in vivo experiments have found that these substances can induce cellular proliferation, repair and redox mechanisms, as well as anti-inflammatory and antifibrogenic responses [15,23,24,25,26]. The benefits of N,N-methylated tryptamines are continuing to be reassessed and uncovered; however, their endogenous role remains in controversy [27].
In 1961, Axelrod discovered the enzyme indolethylamine-N-methyltransferase (INMT) in rabbit lung [28]. In 1965, concentrations of DMT (blood: 8.0 to 55 ng/mL; urine: 43 μg/day), 5-HO-DMT (blood: 1.0 to 40 ng/mL; urine: 63 μg/day), and 5-MeO-tryptamine (blood: 20 to 80 ng/mL; urine: 37 μg/day) were discovered in normal human blood and urine [29]. In 1972, Lewis Mandel et al. discovered human INMT in lung tissue, and the enzyme was found to monomethylate and dimethylate tryptamine with the universal one-carbon methyl donor S-adenosylmethionine (SAM) [30]. In 1988, an enzyme Thioether S-Methyltransferase (Temt) was discovered in mouse lungs and livers, which was determined to be chiefly responsible for methylating dimethylselenide (DMSe) with SAM to form trimethylselenonium (TMSe+) for selenium metabolite detoxification and elimination [31,32,33]. Although it would not be discovered until 2015 in a study investigating poor selenium status, single nucleotide polymorphism (SNP) influences on TMSe+ production in rural Argentinian and Bangladeshi women revealed TEMT activity by INMT [34,35]. INMT has recently been confirmed to be consequential in the methylation of volatile DMSe to form the urine-excreted trimethylselenonium (TMSe+) through site-directed mutagenic experiments [36]. In 1995, Strassman initiated the investigation of endogenous DMT and Warner et al. cloned and characterized mouse lung TEMT [37]. In 1998, mouse Inmt—identified as Temt—was determined to be a marker for renal proximal tubules, which are key drivers in renal fibrosis [38,39]. In 1999, Thompson et al. cloned and characterized the human INMT gene from skeletal and placental tissues [40]. In 2005, the crystal structure of human INMT in complex with S-adenosylhomocysteine (SAH) was solved (PDB: 2A14; rcsb.org/structure/2a14). In 2014, a selective noncompetitive inhibitor (N,N-Dimethylaminopropyltryptamine) to rabbit INMT, and predicted by in silico modeling to inhibit human INMT, was synthesized [41]. In 2023, E. coli were engineered to produce DMT, 5-HO-DMT, and 5-MeO-DMT in high quantities by expressing human INMT and supplemented media with tryptophan or glucose [42]. It is clear that illicit N-methylated tryptamines are endogenously made, but current research endeavors continue to stress the potential role for INMT in regulating neurochemistry, mood, consciousness, and sensory perception [3,43,44,45,46,47].
Recently, variations in INMT expression and activity have been identified in acute and chronic diseases in the liver, kidneys, and heart, particularly in early fibrogenic states [48,49,50,51]. Fibrosis is a severe feature of both chronic diseases and acute injuries, often originating from or reinforced by endothelial injury and dysfunction at the onset of pathogenesis [52,53,54,55,56]. Endothelial injury in chronic conditions like MAFLD, MASH, and CKD, as well as in acute injuries such as ischemia, disrupt the delicate balance of vascular function and act as major contributors in tissue fibrogenesis [57,58,59,60,61]. In MAFLD and MASH, for example, lipotoxic stress initiates disruption in hepatocyte homeostasis, causing oxidative distress and damage which then can trigger immune infiltration and significant tissue damage [62,63]. The major mediators in liver injury response are liver sinusoidal endothelial cells (LSECs), which are considered early initiators and reinforcers for hepatic stellate cell (HSC) activation [62,63,64]. HSCs are retinoid storing cells, responsible for tissue fibrosis following their transdifferentiation to myofibroblasts, which interestingly have recent evidence of also playing a role in renal fibrosis—known as renal stellate cells (RSCs) in the cortex—in a similar manner [62,65,66,67]. Reactive oxygen species (ROS) as well as lipid peroxidation byproducts from damaged hepatocytes provoke endothelial dysfunction and HSC activation, creating a pro-inflammatory and pro-fibrotic environment [65,68,69]. Similarly, in CKD, endothelial cells in the renal microvasculature are exposed to hemodynamic stress, hyperglycemia in diabetes, circulatory-derived oxidative stress and uremic toxins [70,71,72,73]. Comprising the glomerular filtration barrier—a vital cellular architecture in regulating renal filtration function—are endothelial cells, a basement membrane, and podocytes [74,75]. The recent feature of RSCs in renal fibrosis may also pose a significant avenue in mitigating cortical fibrogenesis [67]. Exposure to these insults increases the risk for endothelial cell activation and podocyte injury, leading to a compromised vascular barrier, increased permeability, recruitment of immune cells, and tissue injury [76]. The resulting transforming growth factor beta (TGFβ)-mediated endothelial and podocyte injury contributes to significant renal fibrosis [53,77,78,79].
Ischemic injury, whether in the liver or kidneys, causes rapid endothelial cell damage due to the lack of oxygen and nutrient supply [80,81]. The resulting hypoxia induces hypoxia inducible factor-1alpha (HIF-1α) stabilization, which, often in feedback synergy with TGFβ, and/or endothelin-1 (ET-1), promotes extracellular collagen deposition, fibroblast, HSC activation, vascular remodeling, and inflammation [82,83,84,85,86]. Notably, hypoxia has also been shown to be responsible for a shift toward N-methylated tryptamines biosynthesis, and in vitro studies found DMT to be protective against hypoxia [87,88]. Hypoxia also impairs endothelial nitric oxide production, leading to increased vascular tone and further endothelial dysfunction [89]. In the liver, fibrosis is often irreversible following severe or chronic insults like ischemia or MASH, and the lack of any current therapeutics available for liver fibrosis leaves patients with a greater risk for developing organ failure and cancer [62]. Collectively, these diseases create a cycle where endothelial injury fuels inflammation, TGFβ signaling, and hypoxia-driven fibroblast and stellate cell activation, leading to sustained fibrosis [63,89]. This endothelial-initiated inflammation and fibrogenic outcome is critical in the pathogenesis of chronic liver and kidney diseases, underscoring the need to target endothelial dysfunction in therapeutic strategies to mitigate fibrosis and prevent disease acceleration [62,89,90].
A key role for endogenous N-methylated tryptamines was identified in 2009 as the agonist to the sigma non-opioid intracellular receptor 1 (SIGMAR1) signaling pathway [91,92,93,94]. SIGMAR1 is a ubiquitous, versatile intracellular receptor involved with inter-organelle crosstalk, specifically the mitochondria and endoplasmic reticulum (ER), and plays a role vascular function [95,96,97,98,99]. Pre-clinical research on SIGMAR1 is currently receiving attention across a spectrum of liver, kidney, and heart diseases for its pro-survival effects against insults [96,100,101,102,103]. In neurobiology, colocalization of SIGMAR1 and INMT in motoneurons is suspected, suggesting that there may be subcellular spatial regulation in favor of agonizing SIGMAR1 via INMT [104]. Indeed, some researchers have suggested the benefits of N-methylated tryptamines for multiple diseases [105]. Advances in our understanding of the role of INMT in human biology will undoubtedly improve our understating of tryptamine and selenium metabolism, thus prompting new therapeutic strategies for diseases.

2. Tryptamine Metabolism and Biochemistry

Serving as the skeleton to a major class of psychedelics, a tryptamine is an indole heterocycle with an ethylamine group at the C3 position (Figure 1A). Tryptamine is also the metabolite formed from the decarboxylation of tryptophan via aromatic L-amino acid decarboxylase (AADC; an alternatively expressed enzyme encoded by the dopa decarboxylase (DDC) gene), which is primarily expressed in liver, kidneys, spleen, brain, and heart and is regulated by hepatocyte nuclear factor 1 (HNF1α) for nonneuronal tissue expression [106,107,108]. Using SAM, INMT methylates tryptamine to form N-methyltryptamine (NMT) and a second methylation can occur by exchanging the spent SAH for new SAM to form DMT (Figure 1B) [43,109]. Chu et al. 2014 reported the allosteric activity of DMT on INMT, which may be a result of end-product inhibition [41]. Other reports have suggested the production of N,N,N-trimethyltryptamine (TMT) and 5-hydroxy-TMT (5-HO-TMT) by INMT, but there is not much evidence exploring their biological role [42,110]. Interestingly, plants and fungi have been shown to produce N,N,N-trimethyltryptamine and the psilocin (4-HO-DMT) derivative aeruginascin (4-HO-TMT), respectively [111,112].
INMT may also methylate serotonin (5-HO-tryptamine; 5-HT) to form the mono- and dimethylated ethylamine derivatives 5-HO-NMT and bufotenine. 5-HT arises from the C5 hydroxylation of tryptophan by tryptophan hydroxylase-1 (TPH-1) prior to decarboxylation by AADC [109,113]. Although not well established, it is suggested that melatonin—a 5-HT derivative canonically produced in a stepwise manner of N-acetylation of the ethylamine by serotonin N-acetyltransferase (SNAT) and then O-methylation of the C5 hydroxyl by N-acetylserotonin O-methyltransferase (ASMT)—is produced in an alternative manner with ASMT preceding SNAT N-acetylation [114]. Localized in the retina and pineal gland, ASMT production of 5-MeO-tryptamine may be possible (Figure 1B), but it is not yet known if 5-MeO-tryptamine may be trafficked for methylation in tissues or cells expressing high levels of INMT [114,115]. The presence of 5-MeO-tryptamine in blood, however, might serve as a source for 5-MeO-DMT via INMT [29,116]. It is worth mentioning here that DMT, 5-HO-DMT, and 5-MeO-DMT have a slight lipophilic character (logP ~2) and have been found in vesicles, suggesting some feasibility in being trafficked or localized to lipid bilayers [43,110,117]. A 1974 study in 5-MeO-DMT biosynthesis suggests that INMT may have the ability to methylate the C5 hydroxyl, but no recent studies have confirmed this [109,118]. Alternatively, a recent study using rat KOs of INMT have shown that the N-methylated tryptamines NMT and DMT are still biosynthesized, which suggests that there could be an unknown redundant or compensatory enzyme for the endogenous production of N-dimethylated tryptamines [119]. Nevertheless, genetic limitations between other mammals could be another reason for the differences in methylation capacity and biological function, since INMT in humans share ~57% sequence homology and are oppositely oriented in the genome compared to mice, along with possible alternative splicing (Gene [INMT]. Bethesda (MD): National Library of Medicine (USA), National Center for Biotechnology Information; 2004 (accessed on 3 October 2024). Available online: https://www.ncbi.nlm.nih.gov/gene/11185) [120]. Recent evidence in prostate cancer has also uncovered SMYD3 in the promoter region of INMT and Inmt, but DNA methylation patterns in INMT may influence variations in expression too, as seen in head and neck squamous cell carcinoma [121,122]. Most interestingly, the knockdown of INMT in vitro amplifies the anticancer activity of selenium compounds, further suggesting INMT’s role in selenium metabolism [121].
In silico experiments exploring DMT in metabolism have provided thermodynamic insight to methylation by INMT as well as catabolic pathways, such as via monoamine oxidases A and B (MAO-A, and -B) [123,124]. A recent study has illustrated the N-methylation(s) of the C3 ethylamine in tryptamine and has determined that the second methylation to form DMT (δG = 142.42 kJ/mol) has a much higher activation barrier than the formation of NMT (δG = 81.42 kJ/mol), suggesting that DMT formation is thermodynamically disfavored, potentially explaining the low physiological abundance in the brain [123]. Another recent in silico study noted the preferential oxidation of DMT by the MAO-A flavoenzyme with FADH (δG = 70.3 kJ/mol), demonstrating a lower activation barrier compared to tryptamine (δG = 78.2 kJ/mol) by δG = 8 kJ/mol, suggesting a more thermodynamically favorable oxidation and again highlighting a rationale for the low abundance of DMT [124]. Indeed, early rat experiments have shown that tryptamine exists for matter of seconds in vivo [125]. Nonetheless, a recent study implementing in silico techniques to investigate INMT methylation of DMSe to TMSe (Figure 1C) reinforced previous evidence for the preferential binding of DMSe over tryptamines in INMT [34,35,36]. While INMT may show versatile activity between both tryptamine and selenium metabolism, the argument can be made that INMT’s contribution to selenium turnover is important for maintaining cellular viability, such as that conducted by the selenoprotein GPX4, in staving off ferroptosis by quenching lipid peroxides arising from antioxidant plasmalogens [126,127,128,129,130]. Conversely, one could also suggest that tryptamine derivatives in hypoxic or fibrotic environments may serve as short-lived antioxidants where the lipophilic character supports localized quenching of ROS, but this has yet to be shown [24,117,131].
The physiological relevance of N-methylated tryptamines has been difficult to determine for many reasons. Its social perception has narrowly associated N-methylated tryptamines with neurobiology [132]. N-methylated tryptamine synthesis may be inconsequential to health and disease since they arise from a minor pathway compared to the classical kynurenine, serotonin/melatonin, and indole metabolic pathways [133]. Improvement in the robustness of high-throughput experiments and the public availability of data have brought tryptophan metabolism, INMT, and endogenous N-methylated tryptamines into the spotlight again [15,26,51,87,134].
Human tryptophan metabolism predominantly occurs when the essential amino acid is liberated by digestive peptidases and absorbed by the intestinal lumen via SLC6A14 and SLC6A19 neutral amino acid transporters and shuttled to portal circulation via SLC16A10, SLC7A5-SLC3A2, and SLC7A8-SLC3A2 [135,136]. Next, entering the blood, tryptophan is then trafficked to the liver by albumin (75–90%) where 90–95% of diet-derived hepatic tryptophan is directed to the kynurenine pathway and remaining tryptophan is directed to minor metabolic pathways: serotonin/melatonin (1–2%) and indole (5%), which primarily occur in the gut (Figure 1A,B) [137,138]. The kynurenine pathway supplies the crucial bioenergetic metabolite nicotinamide adenine dinucleotide (NAD), as well as kynurenine, kynurenic acid, xanthurenic acid, and cinnabarinic acid which play significant roles in immunoregulation and the cellular redox state [139]. In fact, gut microbiota generating indole acetic acid, like tryptamine, dampen hepatic inflammation in MAFLD [140].
Albeit physiologically low in abundance, N-methylated tryptamines also have an impact on other aspects of metabolism like serotonin, such as lipid metabolism [93,141,142]. Mediated by metabolites from the kynurenine pathway and gut microbiota, agonism of the aryl-hydrocarbon receptor (AhR) has been shown to inhibit de novo lipogenesis [139,140]. Mice given 5-HO-DMT demonstrated reduced cyclooxygenase lipid products and particularly reduced 20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid (20-HETE) levels which play an important role in endothelial dysfunction, as seen in lipidomics performed on their paws [93]. Looking to bypass untoward side effects of 5-HO-DMT, a recent study in mice was able to demonstrate liposomal encapsulation of bufotenine for delivery while maintaining the efficacy of 5-HO-DMT in dampening lipid metabolism and inflammation [142]. The untoward psychotoxic effects of methylated tryptamines are a significant challenge to effectively target SIGMAR1. Strategies to deliver methylated tryptamines are growing as mentioned, but many would require a lipophilic scaffold or delivery system that bypasses 5-HTR signaling at the plasma membrane and maintains intracellular efficacy. Further exploration in drug design and delivery is required to optimize the targeting efficiency of these SIGMAR1 agonists. Consistent with N-methylated tryptamines’ influence on lipid metabolism, studies investigating alterations to SIGMAR1 activity have demonstrated changes to lipid metabolism [143,144]. While further studies are required to explore and deepen our understanding of the impact of N-methylated tryptamines, the evidence of protective effects could be investigated further for therapeutic exploitation.

2.1. Redox Biochemistry

N-dimethylated tryptamines have been described for their effect on 5-HTRs (Table 1) [1,22,43,145,146,147,148,149]. 5-HTRs have been long been implicated in peripheral tissues, where serotonin signaling can mediate whole-body lipid metabolism, inflammation, and play a role in fibrogenic diseases [150,151,152,153,154]. N-methylated tryptamines may also owe their positive outcomes to 5-HTR signaling. Particularly of therapeutic interest are 5-HTRs 1B, 2A, and 2B in HSCs, which have been associated with pro-fibrotic outcomes [155,156,157]. Indeed, 5-HTR signaling is vital in immunoregulation, reviewed elsewhere [158,159]. It is not yet established if endogenous N-methylated tryptamines may compensate or interfere with 5-HTR signaling.
Studies on tryptophan derivatives and serotonin have suggested potent radical scavenging properties as a result of favorable free radical attacking substituents and the tryptamine skeleton [131,160]. Consistent with other reports, serotonin also interacts with lipids to protect lipids from oxidation, suggesting that N-methylated tryptamines may have the same function [117,161]. Since serotonin has shown radical scavenging activity at the C5 hydroxyl, substituents to the tryptamine skeleton may drastically influence radical scavenging properties as well as stability of the radical from further propagation. The C2 position of the tryptamine backbone is susceptible to oxidation, suggesting that tryptamine derivatives could also act as antioxidants [131]. Given the lipophilic character, N-methylated tryptamines could also be short lived due to their rapid spontaneous reaction with hydroperoxides, lipoperoxides, superoxide, and other ROS. Tryptophan derivatives have been shown to spontaneously degrade as a result of oxidation and UV exposure into products such as 2-indolone, supporting further investigations of the antioxidant capacity of N-methylated tryptamines in biological systems [131]. Future electron paramagnetic resonance experiments could help in elucidating this.

2.2. Molecular Biology

Besides their known role in 5-HTR signaling, some N-methylated tryptamines have been investigated for their potential as endogenous ligands to trace amine-associated receptors (TAARs) (Table 1) [45,162,163,164]. While mostly studied in the context of the brain and olfaction, TAAR1 has been shown to be agonized by DMT and it is predicted to also agonize TAAR6. In humans, there are nine TAAR genes, with six functional genes and three pseudogenes [163]. TAARs exist outside of the brain, but no conclusive function in peripheral tissues has been determined as of yet [163]. More research is necessary to determine the likelihood of N-methylated tryptamines in TAAR signaling. In the periphery, TAAR1 mRNA is found in the liver, kidney, and heart, suggesting that there may be roles in N-methylated tryptamine signaling through TAAR1 [162,165]. However, recent evidence in the tryptamine perfused rat kidney suggests that vascular responses are likely mediated through 5-HTR1A [166].
Recent evidence in hepatic portal vein hypertension has shown that 5-HTR1A agonism confers reduction in hypertension, suggesting an influence of 5-HTR1A agonism on endothelial dysfunction [167]. 5-HTR1A has not yet been described extensively in endothelial cells, so further research is needed to confirm if endothelial 5-HTR1A is responsible for these observed benefits, similar to the effects of endothelin signaling via endothelin receptor A (ETAR) [68,168,169,170,171]. Canonical endothelial dysfunction, however, is well known to be mediated by ET-1 and has also been implicated in MAFLD, MASH, CKD, and CVD [52,53,54,56,77,172,173,174,175]. ET-1 is a potent vasoconstrictor peptide primarily produced by endothelial cells. ET-1 exerts its effects by binding to endothelin receptor A or -B (ETAR or ETBR, respectively), which are G-protein coupled receptors expressed on endothelial cells, smooth muscle cells, HSCs, podocytes, fibroblasts, and other various cell types [53,54,56,170,173,176]. Upon ET-1 binding, ETAR, for example, activates G proteins, leading to the stimulation of phospholipase C and phosphatidylinositol hydrolysis to form IP3 and subsequently promote the release of calcium, triggering vasoconstriction [176,177]. Endothelial dysfunction arises from insults and pathogenic crosstalk through ET-1/ETAR or ETBR between endothelial and other cells in their local milieu or from circulation [176,178]. In the kidney, this has been documented in models of focal segmental glomerulosclerosis and DKD, demonstrating a promise for ameliorating oxidative stress and fibrosis when targeted [77,179,180,181,182]. Similarly in the liver, endothelin signaling is a contributor to HSC activation for fibrosis, and ETAR blocking has been shown to reduce hepatic fibrosis in rats and mice [173,178,183,184]. As a consequence to endothelin signaling, TGFβ release can be upregulated and commit to a viscous positive feedback loop by TGFβ subsequently upregulating transcription of endothelin pathway proteins [177,182,185,186]. The ET-1/ETAR is an attractive target in mitigating endothelial dysfunction and resulting fibrosis but because of its ubiquity and systemic consequences in human health, there remain therapeutic challenges.
TGFβ signaling is a well-studied fibrogenic pathway that is ubiquitously present throughout the body and exists among most cell types. Briefly, TGFβ is a secreted latent protein at the cell membrane that, upon mechanobiological changes or extracellular sources of TGFβ, stimulate release and signaling via TGFβ receptors, induce SMAD phosphorylation, nuclear translocation, and transcription of extracellular matrix (ECM) proteins, like collagen [187,188]. Endothelial dysfunction promotes inflammation and TGFβ signaling in the local milieu and can contribute to circulating TGFβ [189,190,191,192,193]. ECs, HSCs, and likely RSCs, are susceptible to a complex system of pro-fibrogenic, pro-inflammatory signals that drive responses in acute insults chronic disease. Other overlapping disease factors may instigate endothelial dysfunction, such as xanthine oxidoreductase, as seen in fatty liver, CKD, and CVD [70]. Indeed, pioneering research in ET-1 HSC activation identified in vitro superoxide generated by xanthine oxidoreductase significantly impacted endothelin receptor expression [170]. Now, it has been reported that antagonizing ETAR ameliorates liver fibrosis, as seen in a mouse model using nanoparticle delivery systems [194]. However, while great strides have been made in targeting serotonin, TAAR, endothelin, and TGFβ receptors, new interest in SIGMAR1 agonism may provide benefits without serious influence on mood or undesired effects.
SIGMAR1 was first identified as an opioid receptor but was soon considered an orphan receptor due to low specificity with opioid peptides [91]. It was not until Fontanilla et al. discovered DMT as a ligand for SIGMAR1 in 2009 with a Kd = 14.75 μM [91]. Since then, it has been suggested that 5-HO-DMT and 5-MeO-DMT act on SIGMAR1 in a similar manner; however, it has not yet been fully elucidated if 5-HO-DMT and 5-MeO-DMT have greater effect on SIGMAR1 agonism in reducing fibrogenic and pro-inflammatory genes (Table 1) [15,25,88,93,195]. Moreover, SIGMAR1 can induce antioxidant and anti-fibrogenic effects by prompting antioxidant pathways, anti-autophagic, and endocytic recycling. SIGMAR1 has been implicated in activating antioxidant response elements to upregulate NAD(P)H quinone oxidoreductase 1 (NQO1) and superoxide dismutase 1 (SOD1) [196]. Autophagy can be downregulated through SIGMAR1-inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) interactions that inhibit the endonuclease activity of IRE1α [197,198]. Especially important is the upregulation of endocytic receptors to reduce ECM deposition, like LRP1, which has been recently suggested to have enhanced expression by SIGMAR1 [199,200]. Interestingly, the N-methylated ethylamine moieties of a tryptamine resemble other endogenous agonists to SIGMAR1 signaling, such as the essential nutrient choline [201]. Choline is a one-carbon and lipid metabolite partly relied on from diet that contributes to, among many pathways, one-carbon metabolism as a methyl donor to form SAM [202]. Choline has recently been found to agonize SIGMAR1, deepening its role beyond metabolism and in maintaining mitochondrial and ER dynamics [95,99,201,203]. Choline metabolism, like tryptamine, is partly contributed to by microbiota, like Enterobacteriaceae or Ruminococcaceae, which impacts health and disease [137,138,140,204,205,206,207,208].

2.3. Tryptamine Metabolism and the Gut Microbiome

Gut bacteria deeply impact human health and metabolism, especially in contributing to immunoregulatory mechanisms via kynurenine metabolites [137,140,204,209,210]. Microbiota-derived tryptamine arises from decarboxylation of tryptophan (Figure 1A) by bacteria like Ruminococcus gnavus and has been associated with liver fibrosis in MAFLD [209,211,212,213,214]. Like choline, gut-derived tryptophan metabolites have conflicting evidence to their benefit or harm [212,215,216]. A recent article found R. gnavus-derived tryptamine to impair insulin sensitivity in mice and monkeys via TAAR1 [217]. In contrast, the enrichment of R. gnavus also has been shown to be lower in CVD when complicated with NAFLD, suggesting that comorbidities may influence the microbiome composition [218]. Additionally, Enterobacteriaceae have a positive association with MAFLD severity and progression where shifts in tryptophan and choline metabolism can impact disease pathogenesis [211,216]. Similarly, Enterobacteriaceae has been implicated in CKD, enriching as disease progresses and correlating with low eGFR [219,220]. Targeting Enterobacteriaceae through tryptophan metabolism by using D-tryptophan, however, has shown effectiveness in reducing colitis and may be a potential therapeutic [221,222].
Intriguingly, microbial-derived short-chain fatty acids were recently shown to impact SIGMAR1 [102]. Indeed, a SIGMAR1 KO also showed evidence for gut dysbiosis, suggesting that there may be a microbial relationship to SIGMAR1 activity [223]. It remains to be seen if microbiota-derived tryptamines have any contribution to endogenous N-methylated tryptamine levels, but further experiments may help to determine if SIGMAR1 agonism is mediated through host-microbe circumstances.

3. INMT in Models of Human Diseases

In vivo murine studies, particularly ones in fibrogenic diseases, have demonstrated that Inmt is expressed in many healthy tissues and have found an alteration of Inmt mRNA expression as a result of insult or disease [48,50,224]. Both male and female mouse kidneys have shown a high expression of Inmt in cortical segments of healthy proximal tubule cells, which can be further utilized in in situ RNA-sequencing to identify spatial cellular changes in whole-tissue models of acute kidney injury (AKI) and repair [225]. Indeed, proximal tubules are early contributors to fibrogenic signaling in the injured kidney [39].
In utero experiments have also shown a change in Inmt expression. In 2012, a time-course mouse study using a hyperhomocysteinemia model with cystathionine β-synthase (Cbs)-deficient mice found both wild-type and Cbs-deficient in uterus interimplantation sites had increased Inmt mRNA but several folds more in Cbs-deficient mice in early pregnancy [226]. The study did not further investigate if Inmt activity is consequential for implantation and in utero embryonic development; however, the generation of the Inmt knockout (KO) rat and mouse (mousephenotype.org/data/genes/MGI:102963) has not reported embryonic lethality, suggesting that Inmt may have functionality beyond implantation, pregnancy, and embryonic viability [119,227].

3.1. INMT in Renal Diseases

Originally found as a marker for proximal tubule cells in the kidney, INMT has been implicated in many forms of renal diseases, such as nephrolithiasis, diabetic kidney disease (DKD), AKI, and renal fibrosis [38,50,228,229,230]. In search of a genetic susceptibility to nephrolithiasis, a Japanese 2012 genome-wide association study with 5892 participants identified a risk SNP, rs1000597, in INMT [228]. A model of DKD, a prominent subgroup of CKD, downregulates proximal tubule Inmt expression in db/db mice but is rescued by treatments of the angiotensin receptor blocker irbesartan and the sodium-glucose cotransporter 2 inhibitor (SGLT2i) dapagliflozin, suggesting Inmt may be important in injury or disease resolution [50]. In line with this evidence, publicly available transcriptomic data from DKD-induced mouse models identified the downregulation of Inmt [231].
In a model of AKI, Inmt expression in male C57BL/6 mice subjected to ischemia–reperfusion injury (IRI) was downregulated in pro-fibrotic, maladaptive proximal tubule cells [232]. In another IRI experiment aiming to characterize the transition of AKI-to-CKD, C57BL/6CN male mice were found to have Inmt expression downregulated in proximal tubules as a result of IRI [233]. Interestingly, studies conducted in 2018 and 2019 demonstrated that administering DMT to prevent renal injury in a rat model of clamp-induced IRI significantly mitigated IRI with an intramuscular dose of 2.95 mL/kg body weight of a 2.45 mg/mL DMT solution, administered once 15 min before ischemia and once 15 min before reperfusion [229,230]. Inmt was not assessed in this study, but the study may suggest that the role of Inmt may be in mitigating cell and tissue insults via agonizing SIGMAR [91,229,230]. Ultimately, other studies in rats have found that Inmt is expressed in healthy brain, lung, liver, and kidney tissues [92,234].
Public databases of transcriptomic data suggest human INMT has been found to be expressed in liver, lung, heart, and kidney tissues (Human Protein Atlas: proteinatlas.org/ENSG00000241644-INMT/tissue+cell+type, and The Kidney Precision Medicine Project: https://www.kpmp.org (accessed on 27 September 2024) [235,236]. When breaking down INMT expression by single-cell RNAseq in these organs: fibroblast subpopulations, endothelial, smooth muscle, mesangial cells, macrophages, and HSCs are expressors [235,236]. The KPMP database includes single-nucleus RNAseq which associated INMT expression with repairing fibroblasts and degenerative endothelial cells, and although consistent with a protective effect, more research is necessary to conclude a role for INMT in fibrosis [236].
Recent studies in fibrogenic MAFLD, MASH, CVD, and CKD have implicated changes in SIGMAR1 activity as well as INMT [48,100]. Interestingly INMT variants, which impact the methylation ability of INMT, have been associated with a risk nephrolithiasis, but a recent study has shown amelioration through SIGMAR1 agonism [96]. Fukumoto et al. has also shown the preferential activity of INMT in TMSe production, which has recently been found to be compensated by thiopurine S-methyltransferase [36,237]. Ultimately, INMT may be important as a redundant enzyme between tryptamine and selenium metabolism where insults may determine the preferential activity of INMT.

3.2. INMT in Hepatic Diseases

Identified in hepatic endothelial cells in 2018, INMT became a fibrotic-gene candidate in human MAFLD in 2021 [48,238]. A recent article investigating a mouse model of liver fibrogenesis and regression using thioacetamide found downregulated INMT which rapidly increased to normal expression levels in the regressive period [239]. A 2016 study demonstrated hepatic Inmt is downregulated in mice treated with 30 mg/kg diclofenac, a common analgesic [240]. A recent rat model of primary sclerosing cholangitis identified hepatic tryptophan and monoamine metabolism in regulating liver fibrosis, suggesting tryptamines may play a role in liver fibrosis [241]. In Gata4 KO mice, a transcription factor regulating liver fibrosis, hepatocyte Inmt was found to be upregulated but present in low protein abundance; however, recent evidence by Arroyo et al. demonstrated Gata4 as responsible for HSC deactivation [242,243]. Future studies in the Gata4 KO, with a focus on INMT, will help in delineating the activity of INMT in HSCs activation. HSCs activation and subsequent maladaptive ECM deposition is induced by a myriad of insults; however, ET-1 is implicated as a major driver of fibrosis in MASH [52,172,177,183]. Opposing HSC activation and fibrogenic signaling has been shown by LRP1 mitigating rise in TGFβ in vitro [244]. Interestingly, studies have shown that activated HSCs have ER-localized SIGMAR1 translocated to the plasma membrane, which are suggested to be more susceptible to agonization from agents in the local milieu [245]. Studies have shown in mice that the absence of functional LRP1 accelerates the progression of MAFLD and MASH, further underscoring the benefits to LRP1 endocytic signaling against disease [246]. While it has yet to be shown, the sum of evidence here indicates that the SIGMAR1 agonizing products of INMT may promote endocytic recycling via increased LRP1 levels to mitigate ECM deposition.
Hepatic autophagy through ER-stress induced IRE1α activation, on the other hand, has been implicated with pro-fibrotic effects [247]. While not yet investigated, methylated tryptamines could potentiate the SIGMAR1-dependent inhibition of IRE1α endonuclease activity, thus promoting antifibrogenic effects in the liver. Further direct investigations are necessary to confirm.
A rat model investigating 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) AhR agonism and toxicity found that hepatic Inmt was downregulated following TCDD administration, irrespective of TCDD susceptibility [248]. Responsible for pro-inflammatory gene expression as a transcription factor, AhR is agonized by several tryptophan metabolites from the Kynurenine pathway. The decrease in hepatic Inmt may reflect upstream regulation by pro-inflammatory signals, since methylated tryptamines have been shown to induce anti-inflammatory effects [25,195]. An obese Zucker rat model investigating the anti-steatotic effect of soy protein found Inmt to be upregulated compared to a casein diet by mRNA RNAseq and verified by qPCR [249]. Metformin, a diabetic drug improving liver health, has been used in MAFLD to identify INMT expression as a target [250]. While INMT has not been directly explored in the liver, it is expressed across disease profiles and largely associated with positive outcomes. It has yet to be demonstrated but experiments specifically incorporating the Inmt KO into mouse models of liver and kidney injury may uncover the role of INMT N-methylated tryptamines.
A 24h in vitro FACS flow cytometry experiment investigating the influence of DMT on LX-2 HSCs demonstrated an increase in MHC-II expression (4.6% with 100 μM DMT alone and 11.6% at 100 μM DMT with 500IU interferon gamma (IFNγ)), which is consistent with other reports using IFNγ to downregulate collagens involved in fibrosis (Unpublished data) [251,252]. INMT in HSCs has yet to be fully described, but SIGMAR1 in activated HSCs has been shown to be highly expressed and can be blocked by novel targeted delivery of silencing RNA (siRNA) nanoparticles [253]. While the Han et al. study demonstrated antagonism of SIGMAR1 leads to a reduction in collagen proteins, it remains to be determined if agonism may also influence fibrogenesis. Considering that ferroptosis is a large contributor to MASH and fibrosis, the role INMT in mediating selenium metabolism and SIGMAR1-dependent expression of antioxidant genes may be a significant factor in dampening ferroptotic fate [254,255]. Selenoproteins, like GPX4, are expressed in all forms of life and play a role in quenching ROS and other free-radical substances. The intake of dietary selenium is required for selenoprotein synthesis at very low concentrations at roughly 55 μg/day, thus underscoring the vital importance of sufficient selenium levels to maintain homeostasis. INMT balances selenium levels through the production of the major urine metabolite TMSe; however, there are no studies investigating the flux of TMSe produced by INMT in fibrotic diseases. Given the lipophilic character and susceptibility to oxidation at the indolic C-2 position, methylated tryptamines may also serve to spontaneously quench propagative lipoperoxides that otherwise promote ferroptosis [126]. Indeed, ferroptosis contributes to HSC activation and liver fibrogenesis [256]. More thorough experiments implementing 76Se, 13C-SAM and 13C-tryptophan could help in determining the role of INMT in fibrogenic MAFLD by investigating the contribution of INMT to selenium and N-methylated tryptamine metabolism. This will be vital in understanding INMT activity in HSCs. Expanding an isotope tracing experiment to incorporate human subjects with MAFLD would confirm the preferential flux of selenium or N-methylated tryptamine metabolites in disease.
It is noteworthy to mention that Alzheimer’s disease shares many molecular characteristics of chronic liver and renal pathologies, such as fibrosis, and a recent study found that DMT ameliorates Alzheimer’s disease through SIGMAR1 [257]. Associated with fibrosis, and often with comorbidities, Alzheimer mouse models have similarly linked benefits from choline to reduced pathology [258,259,260,261,262,263]. Indeed, pathologies such as diabetes and Alzheimer’s are relevant to liver and kidney diseases due to the presence of underlying endothelial dysfunction, which in turn promote fibrogenic and ROS-stimulating pathways in these tissues. There could be hope yet for revisiting a clinical application of N-methylated tryptamine in chronic, fibrotic diseases [261].
Altogether, INMT in diseases, such as liver, kidney, and heart fibrosis, Alzheimer’s disease and diabetes, may play a critical role as an antifibrogenic mediator through the production of its metabolites. Future studies must delineate the exact metabolic contribution of INMT to either methylation of tryptamines or selenium metabolism, with a focus on validated models of fibrosis.

3.3. INMT in Cancer

Cancer is notorious in modifying metabolic pathways to support proliferation, evade immune defensive measures, alter the local microenvironment and syphon metabolites from neighboring cells. Across most cancers, INMT is downregulated [122,224]. INMT downregulation in hepatocellular carcinoma (HCC) is a prognostic indicator for poor outcomes and is interestingly met with an increase in SIGMAR1 [224,264]. Indeed, SIGMAR1 has been shown to be protective against ferroptosis in HCC, perhaps as a hijacked mechanism to promote cancer success [254,264]. INMT has been used to develop an HCC-specific profile of selenium metabolism [265]. Selenoproteomic and metabolomic patient studies implementing isotope-tracing 76Se could further our understanding of INMT in progressive-to-terminal liver disease [266]. INMT has also been downregulated in prostate cancer, having an antiproliferative, pro-apoptotic effect when overexpressed [267]. Head and neck squamous cell carcinoma also demonstrates downregulation of INMT expression, influenced by hypomethylation changes in the promoter [122]. Lung adenocarcinoma has also demonstrated a low expression of INMT and is associated with poor prognosis; however, INMT improves immunotherapy response [268]. Perhaps higher expressing INMT in cancer promotes a resistance to cell death via SIGMAR1 in this context [264]. More research is required to determine the metabolic function of INMT across cancer as it often induces global shifts in metabolism.

4. Conclusions

Evidence presented here suggests that endogenously N-dimethylated tryptamines DMT, 5-HO-DMT, and 5-MeO-DMT likely have a biological function outside of neuroregulatory mechanisms. Studies have highlighted the versatile roles of INMT in mediating both tryptamine and selenium metabolic pathways. INMT’s contribution to either tryptamine or selenium metabolism remains to be delineated, but recent data in INMT KO rats suggest that N-dimethylated tryptamines may have an alternative metabolic route [119]. Selenoproteomic and metabolomic patient studies implementing isotope-tracing 76Se, 13C-SAM and 13C-tryptophan could help in determining the role of INMT in MAFLD, MASH, and CKD. Likewise, these studies could define a role for N-methylated tryptamines outside of neuroscience and usher advances in drug targeting and repurposing.
The changes in hepatic, renal, and cardiac INMT expression are consistent with upregulation in injury resolution and downregulation in maladaptive and degenerative tissues. Further studies are required to confirm these phenomena across tissues and cell types.
Benefits of SIGMAR1 agonism in fibrotic, chronic diseases have received growing attention; however, intracellular localization increases the complexity of accurate drug targeting with N-dimethylated tryptamines due to partial 5-HTR agonism at the cell membrane. Studies have highlighted the ubiquity and role of SIGMAR1 in regulating mitochondrial integrity, bioenergetics, and endoplasmic reticulum stress [95,98,99]. SIGMAR1 is an attractive target in mitigating renal injury and fibrosis, growing in popularity. Endothelial agonism of SIGMAR1 may aid in reducing both local and systemic insults (Figure 2). Nonetheless, future research in agonizing SIGMAR1 in liver diseases may be therapeutically promising through means of N-methylated tryptamines or their derivatives.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Nichols, D.E. Psychedelics. Pharmacol. Rev. 2016, 68, 264. [Google Scholar] [CrossRef] [PubMed]
  2. Ortiz, N.R.; Preuss, C.V. Controlled Substance Act. In StatPearls; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar] [PubMed]
  3. Carhart-Harris, R.L.; Goodwin, G.M. The Therapeutic Potential of Psychedelic Drugs: Past, Present, and Future. Neuropsychopharmacology 2017, 42, 2105. [Google Scholar] [CrossRef] [PubMed]
  4. Cohen, A.; Vakharia, S.P.; Netherland, J.; Frederique, K. How the War on Drugs Impacts Social Determinants of Health beyond the Criminal Legal System. Ann. Med. 2022, 54, 2024. [Google Scholar] [CrossRef]
  5. Strassman, R.J. Human Hallucinogenic Drug Research in the United States: A Present-Day Case History and Review of the Process. J. Psychoact. Drugs 1991, 23, 29–38. [Google Scholar] [CrossRef] [PubMed]
  6. McKenna, D.J. Clinical Investigations of the Therapeutic Potential of Ayahuasca: Rationale and Regulatory Challenges. Pharmacol. Ther. 2004, 102, 111–129. [Google Scholar] [CrossRef] [PubMed]
  7. Drug Enforcement Administration. Schedules of Controlled Substances: Placement of 5-Methoxy-N,N-Dimethyltryptamine into Schedule I of the Controlled Substances Act. Final Rule Fed. Fed. Regist. 2010, 75, 79296–79300. [Google Scholar]
  8. McKenna, D.J.; Towers, G.H.N. Biochemistry and Pharmacology of Tryptamines and Beta-Carbolines a Minireview. J. Psychoact. Drugs 1984, 16, 347–358. [Google Scholar] [CrossRef]
  9. Miller, M.J.; Albarracin-Jordan, J.; Moore, C.; Capriles, J.M. Chemical Evidence for the Use of Multiple Psychotropic Plants in a 1,000-Year-Old Ritual Bundle from South America. Proc. Natl. Acad. Sci. USA 2019, 166, 11207–11212. [Google Scholar] [CrossRef]
  10. Palamar, J.J.; Le, A. Trends in DMT and Other Tryptamine Use Among Young Adults in the United States. Am. J. Addict. 2018, 27, 578. [Google Scholar] [CrossRef]
  11. Griffin, M.; Callander, D.; Duncan, D.T.; Palamar, J.J. Differential Risk for Drug Use by Sexual Minority Status among Electronic Dance Music Party Attendees in New York City. Subst. Use Misuse 2020, 55, 230. [Google Scholar] [CrossRef]
  12. Han, B.H.; Duncan, D.T.; Arcila-Mesa, M.; Palamar, J.J. Co-Occurring Mental Illness, Drug Use, and Medical Multimorbidity among Lesbian, Gay, and Bisexual Middle-Aged and Older Adults in the United States: A Nationally Representative Study. BMC Public. Health 2020, 20, 1123. [Google Scholar] [CrossRef] [PubMed]
  13. Timmermann, C.; Zeifman, R.J.; Erritzoe, D.; Nutt, D.J.; Carhart-Harris, R.L. Effects of DMT on Mental Health Outcomes in Healthy Volunteers. Sci. Rep. 2024, 14, 3097. [Google Scholar] [CrossRef] [PubMed]
  14. Davis, A.K.; Averill, L.A.; Sepeda, N.D.; Barsuglia, J.P.; Amoroso, T. Psychedelic Treatment for Trauma-Related Psychological and Cognitive Impairment Among US Special Operations Forces Veterans. Chronic Stress 2020, 4, 2470547020939564. [Google Scholar] [CrossRef] [PubMed]
  15. Kelley, D.P.; Venable, K.; Destouni, A.; Billac, G.; Ebenezer, P.; Stadler, K.; Nichols, C.; Barker, S.; Francis, J. Pharmahuasca and DMT Rescue ROS Production and Differentially Expressed Genes Observed after Predator and Psychosocial Stress: Relevance to Human PTSD. ACS Chem. Neurosci. 2022, 13, 257–274. [Google Scholar] [CrossRef]
  16. Vorobyeva, N.; Kozlova, A.A. Three Naturally-Occurring Psychedelics and Their Significance in the Treatment of Mental Health Disorders. Front. Pharmacol. 2022, 13, 927984. [Google Scholar] [CrossRef]
  17. Manske, R.H.F. A SYNTHESIS OF THE METHYLTRYPTAMINES AND SOME DERIVATIVES. Can. J. Res. 1931, 5, 592–600. [Google Scholar] [CrossRef]
  18. Gonçalves de Lima, O. Observações sobre o “vinho da Jurema” utilizado pelos índios Pancarú de Tacaratú (Pernambuco) [Observations on the “vinho de Jurema” used by the Pancaru’ Indians of Tacaratu’ (Pernambuco)]. Ariquivos Inst. Pesqui. Agron. 1946, 4, 45–80. [Google Scholar]
  19. Szára, S. Dimethyltryptamin: Its Metabolism in Man; the Relation to Its Psychotic Effect to the Serotonin Metabolism. Experientia 1956, 12, 441–442. [Google Scholar] [CrossRef]
  20. Shinozuka, K.; Tabaac, B.J.; Arenas, A.; Beutler, B.D.; Cherian, K.; Evans, V.D.; Fasano, C.; Muir, O.S. Psychedelic Therapy: A Primer for Primary Care Clinicians—N,N-Dimethyltryptamine and Ayahuasca. Am. J. Ther. 2024, 31, E112–E120. [Google Scholar] [CrossRef]
  21. Timmermann, C.; Roseman, L.; Schartner, M.; Milliere, R.; Williams, L.T.J.; Erritzoe, D.; Muthukumaraswamy, S.; Ashton, M.; Bendrioua, A.; Kaur, O.; et al. Neural Correlates of the DMT Experience Assessed with Multivariate EEG. Sci. Rep. 2019, 9, 16324. [Google Scholar] [CrossRef]
  22. Carbonaro, T.M.; Gatch, M.B. Neuropharmacology of N,N-Dimethyltryptamine. Brain Res. Bull. 2016, 126, 74–88. [Google Scholar] [CrossRef]
  23. Morales-Garcia, J.A.; Calleja-Conde, J.; Lopez-Moreno, J.A.; Alonso-Gil, S.; Sanz-SanCristobal, M.; Riba, J.; Perez-Castillo, A. N,N-Dimethyltryptamine Compound Found in the Hallucinogenic Tea Ayahuasca, Regulates Adult Neurogenesis in Vitro and in Vivo. Transl. Psychiatry 2020, 10, 331. [Google Scholar] [CrossRef] [PubMed]
  24. Bentz, E.N.; Lobayan, R.M.; Martínez, H.; Redondo, P.; Largo, A. Intrinsic Antioxidant Potential of the Aminoindole Structure: A Computational Kinetics Study of Tryptamine. J. Phys. Chem. B 2018, 122, 6386–6395. [Google Scholar] [CrossRef] [PubMed]
  25. Szabo, A.; Kovacs, A.; Frecska, E.; Rajnavolgyi, E. Psychedelic N,N-Dimethyltryptamine and 5-Methoxy-N,N-Dimethyltryptamine Modulate Innate and Adaptive Inflammatory Responses through the Sigma-1 Receptor of Human Monocyte-Derived Dendritic Cells. PLoS ONE 2014, 9, e106533. [Google Scholar] [CrossRef] [PubMed]
  26. Dakic, V.; Minardi Nascimento, J.; Costa Sartore, R.; MacIel, R.D.M.; De Araujo, D.B.; Ribeiro, S.; Martins-De-Souza, D.; Rehen, S.K. Short Term Changes in the Proteome of Human Cerebral Organoids Induced by 5-MeO-DMT. Sci. Rep. 2017, 7, 12863. [Google Scholar] [CrossRef] [PubMed]
  27. Jiménez, J.H.; Bouso, J.C. Significance of Mammalian N,N-Dimethyltryptamine (DMT): A 60-Year-Old Debate. J. Psychopharmacol. 2022, 36, 905–919. [Google Scholar] [CrossRef]
  28. Axelrod, J. Enzymatic Formation of Psychotomimetic Metabolites from Normally Occurring Compounds. Science 1961, 134, 343. [Google Scholar] [CrossRef]
  29. Franzen, F.; Gross, H. Tryptamine, N,N-Dimethyltryptamine, N,N-Dimethyl-5-Hydroxytryptamine and 5-Methoxytryptamine in Human Blood and Urine. Nature 1965, 206, 1052. [Google Scholar] [CrossRef]
  30. Mandel, L.R.; Ahn, H.S.; VandenHeuvel, W.J.A.; Walker, R.W. Indoleamine-N-Methyl Transferase in Human Lung. Biochem. Pharmacol. 1972, 21, 1197–1200. [Google Scholar] [CrossRef]
  31. Mozier, N.M.; Mcconnell, K.P.; Hoffman, J.L. S-Adenosyl-L-Methionine:Thioether S-Methyltransferase, a New Enzyme in Sulfur and Selenium Metabolism. J. Biol. Chem. 1988, 263, 4527–4531. [Google Scholar] [CrossRef] [PubMed]
  32. Palmer, I.S.; Gunsalus, R.P.; Halverson, A.W.; Olson, O.E. Trimethylselenonium Ion as a General Excretory Product from Selenium Metabolism in the Rat. Biochim. Biophys. Acta (BBA)—Gen. Subj. 1970, 208, 260–266. [Google Scholar] [CrossRef]
  33. Ganther, H.E. Pathways of Selenium Metabolism Including Respiratory Excretory Products. J. Am. Coll. Toxicol. 1986, 5, 1–5. [Google Scholar] [CrossRef]
  34. Kuehnelt, D.; Engström, K.; Skröder, H.; Kokarnig, S.; Schlebusch, C.; Kippler, M.; Alhamdow, A.; Nermell, B.; Francesconi, K.; Broberg, K.; et al. Selenium Metabolism to the Trimethylselenonium Ion (TMSe) Varies Markedly Because of Polymorphisms in the Indolethylamine N-Methyltransferase Gene. Am. J. Clin. Nutr. 2015, 102, 1406–1415. [Google Scholar] [CrossRef] [PubMed]
  35. Torres, B.; Tyler, J.S.; Satyshur, K.A.; Ruoho, A.E. Human Indole(Ethyl)Amine-N-Methyltransferase (HINMT) Catalyzed Methylation of Tryptamine, Dimethylsulfide and Dimethylselenide Is Enhanced under Reducing Conditions—A Comparison between 254C and 254F, Two Common HINMT Variants. PLoS ONE 2019, 14, e0223546. [Google Scholar] [CrossRef] [PubMed]
  36. Fukumoto, Y.; Kyono, R.; Shibukawa, Y.; Tanaka, Y.K.; Suzuki, N.; Ogra, Y. Differential Molecular Mechanisms of Substrate Recognition by Selenium Methyltransferases, INMT and TPMT, in Selenium Detoxification and Excretion. J. Biol. Chem. 2024, 300, 105599. [Google Scholar] [CrossRef] [PubMed]
  37. Strassman, R.J. Human Psychopharmacology of N,N-Dimethyltryptamine. Behav. Brain Res. 1995, 73, 121–124. [Google Scholar] [CrossRef]
  38. Takenaka, M.; Imai, E.; Kaneko, T.; Ito, T.; Moriyama, T.; Yamauchi, A.; Hori, M.; Kawamoto, S.; Okubo, K. Isolation of Genes Identified in Mouse Renal Proximal Tubule by Comparing Different Gene Expression Profiles. Kidney Int. 1998, 53, 562–572. [Google Scholar] [CrossRef]
  39. Gewin, L.S. Renal Fibrosis: Primacy of the Proximal Tubule. Matrix Biol. 2018, 68–69, 248. [Google Scholar] [CrossRef]
  40. Thompson, M.A.; Moon, E.; Kim, U.J.; Xu, J.; Siciliano, M.J.; Weinshilboum, R.M. Human Indolethylamine N-Methyltransferase: CDNA Cloning and Expression, Gene Cloning, and Chromosomal Localization. Genomics 1999, 61, 285–297. [Google Scholar] [CrossRef]
  41. Chu, U.B.; Vorperian, S.K.; Satyshur, K.; Eickstaedt, K.; Cozzi, N.V.; Mavlyutov, T.; Hajipour, A.R.; Ruoho, A.E. Noncompetitive Inhibition of Indolethylamine-N-Methyltransferase by N,N-Dimethyltryptamine and N,N-Dimethylaminopropyltryptamine. Biochemistry 2014, 53, 2956–2965. [Google Scholar] [CrossRef]
  42. Friedberg, L.M.; Sen, A.K.; Nguyen, Q.; Tonucci, G.P.; Hellwarth, E.B.; Gibbons, W.J.; Jones, J.A. In Vivo Biosynthesis of N,N-Dimethyltryptamine, 5-MeO-N,N-Dimethyltryptamine, and Bufotenine in E. Coli. Metab. Eng. 2023, 78, 61–71. [Google Scholar] [CrossRef]
  43. Barker, S.A. N,N-Dimethyltryptamine (DMT), an Endogenous Hallucinogen: Past, Present, and Future Research to Determine Its Role and Function. Front. Neurosci. 2018, 12, 536. [Google Scholar] [CrossRef]
  44. Cameron, L.P.; Olson, D.E. Dark Classics in Chemical Neuroscience: N,N-Dimethyltryptamine (DMT). ACS Chem. Neurosci. 2018, 9, 2344–2357. [Google Scholar] [CrossRef]
  45. Wallach, J.V. Endogenous Hallucinogens as Ligands of the Trace Amine Receptors: A Possible Role in Sensory Perception. Med. Hypotheses 2009, 72, 91–94. [Google Scholar] [CrossRef]
  46. Hua, H.; Fu, X.; Wang, W.; Wang, S.; Wang, D.; Wu, Z.; Zhang, Q.; He, T.; Yang, C. A Bibliometric Analysis of Research on Psychedelics for Depression Treatment. Heliyon 2024, 10, e36886. [Google Scholar] [CrossRef]
  47. Wyatt, R.J.; Mandel, L.R.; Ahn, H.S.; Walker, R.W.; Vanden Heuvel, W.J.A. Gas Chromatographic-Mass Spectrometric Isotope Dilution Determination of N,N-Dimethyltryptamine Concentrations in Normals and Psychiatric Patients. Psychopharmacologia 1973, 31, 265–270. [Google Scholar] [CrossRef] [PubMed]
  48. Pantano, L.; Agyapong, G.; Shen, Y.; Zhuo, Z.; Fernandez-Albert, F.; Rust, W.; Knebel, D.; Hill, J.; Boustany-Kari, C.M.; Doerner, J.F.; et al. Molecular Characterization and Cell Type Composition Deconvolution of Fibrosis in NAFLD. Sci. Rep. 2021, 11, 18045. [Google Scholar] [CrossRef]
  49. Steinhauser, S.; Estoppey, D.; Buehler, D.P.; Xiong, Y.; Pizzato, N.; Rietsch, A.; Wu, F.; Leroy, N.; Wunderlin, T.; Claerr, I.; et al. The Transcription Factor ZNF469 Regulates Collagen Production in Liver Fibrosis [PREPRINT]. bioRxiv 2024. [Google Scholar] [CrossRef]
  50. Wu, J.; Sun, Z.; Yang, S.; Fu, J.; Fan, Y.; Wang, N.; Hu, J.; Ma, L.; Peng, C.; Wang, Z.; et al. Kidney Single-Cell Transcriptome Profile Reveals Distinct Response of Proximal Tubule Cells to SGLT2i and ARB Treatment in Diabetic Mice. Mol. Ther. 2022, 30, 1741. [Google Scholar] [CrossRef] [PubMed]
  51. Shah, H.; Hacker, A.; Langburt, D.; Dewar, M.; McFadden, M.J.; Zhang, H.; Kuzmanov, U.; Zhou, Y.Q.; Hussain, B.; Ehsan, F.; et al. Myocardial Infarction Induces Cardiac Fibroblast Transformation within Injured and Noninjured Regions of the Mouse Heart. J. Proteome Res. 2021, 20, 2867–2881. [Google Scholar] [CrossRef] [PubMed]
  52. Degertekin, B.; Ozenirler, S.; Elbeg, S.; Akyol, G. The Serum Endothelin-1 Level in Steatosis and NASH, and Its Relation with Severity of Liver Fibrosis. Dig. Dis. Sci. 2007, 52, 2622–2628. [Google Scholar] [CrossRef] [PubMed]
  53. van de Lest, N.A.; Bakker, A.E.; Dijkstra, K.L.; Zandbergen, M.; Heemskerk, S.A.C.; Wolterbeek, R.; Bruijn, J.A.; Scharpfenecker, M. Endothelial Endothelin Receptor A Expression Is Associated With Podocyte Injury and Oxidative Stress in Patients With Focal Segmental Glomerulosclerosis. Kidney Int. Rep. 2021, 6, 1939–1948. [Google Scholar] [CrossRef] [PubMed]
  54. Smeijer, J.D.; Kohan, D.E.; Rossing, P.; Correa-Rotter, R.; Liew, A.; Tang, S.C.W.; de Zeeuw, D.; Gansevoort, R.T.; Ju, W.; Lambers Heerspink, H.J. Insulin Resistance, Kidney Outcomes and Effects of the Endothelin Receptor Antagonist Atrasentan in Patients with Type 2 Diabetes and Chronic Kidney Disease. Cardiovasc. Diabetol. 2023, 22, 251. [Google Scholar] [CrossRef] [PubMed]
  55. Lerman, A.; Holmes, D.R.; Bell, M.R.; Garratt, K.N.; Nishimura, R.A.; Burnett, J.C. Endothelin in Coronary Endothelial Dysfunction and Early Atherosclerosis in Humans. Circulation 1995, 92, 2426–2431. [Google Scholar] [CrossRef] [PubMed]
  56. Abraham, D.; Distler, O. How Does Endothelial Cell Injury Start? The Role of Endothelin in Systemic Sclerosis. Arthritis Res. Ther. 2007, 9, S2. [Google Scholar] [CrossRef] [PubMed]
  57. Villanova, N.; Moscatiello, S.; Ramilli, S.; Bugianesi, E.; Magalotti, D.; Vanni, E.; Zoli, M.; Marchesini, G. Endothelial Dysfunction and Cardiovascular Risk Profile in Nonalcoholic Fatty Liver Disease. Hepatology 2005, 42, 473–480. [Google Scholar] [CrossRef]
  58. Hu, S.; Hang, X.; Wei, Y.; Wang, H.; Zhang, L.; Zhao, L. Crosstalk among Podocytes, Glomerular Endothelial Cells and Mesangial Cells in Diabetic Kidney Disease: An Updated Review. Cell Commun. Signal. 2024, 22, 136. [Google Scholar] [CrossRef]
  59. Xie, G.; Wang, X.; Wang, L.; Wang, L.; Atkinson, R.D.; Kanel, G.C.; Gaarde, W.A.; DeLeve, L.D. Role of Differentiation of Liver Sinusoidal Endothelial Cells in Progression and Regression of Hepatic Fibrosis in Rats. Gastroenterology 2012, 142, 918–927.e6. [Google Scholar] [CrossRef]
  60. Miyao, M.; Kotani, H.; Ishida, T.; Kawai, C.; Manabe, S.; Abiru, H.; Tamaki, K. Pivotal Role of Liver Sinusoidal Endothelial Cells in NAFLD/NASH Progression. Lab. Invest. 2015, 95, 1130–1144. [Google Scholar] [CrossRef]
  61. Marcuccilli, M.; Chonchol, M. NAFLD and Chronic Kidney Disease. Int. J. Mol. Sci. 2016, 17, 562. [Google Scholar] [CrossRef] [PubMed]
  62. Roehlen, N.; Crouchet, E.; Baumert, T.F. Liver Fibrosis: Mechanistic Concepts and Therapeutic Perspectives. Cells 2020, 9, 875. [Google Scholar] [CrossRef] [PubMed]
  63. Lafoz, E.; Ruart, M.; Anton, A.; Oncins, A.; Hernández-Gea, V. The Endothelium as a Driver of Liver Fibrosis and Regeneration. Cells 2020, 9, 929. [Google Scholar] [CrossRef] [PubMed]
  64. Koch, P.S.; Lee, K.H.; Goerdt, S.; Augustin, H.G. Angiodiversity and Organotypic Functions of Sinusoidal Endothelial Cells. Angiogenesis 2021, 24, 289. [Google Scholar] [CrossRef]
  65. Fujita, T.; Soontrapa, K.; Ito, Y.; Iwaisako, K.; Moniaga, C.S.; Asagiri, M.; Majima, M.; Narumiya, S. Hepatic Stellate Cells Relay Inflammation Signaling from Sinusoids to Parenchyma in Mouse Models of Immune-Mediated Hepatitis. Hepatology 2016, 63, 1325–1339. [Google Scholar] [CrossRef]
  66. Nagy, N.E.; Holven, K.B.; Roos, N.; Senoo, H.; Kojima, N.; Norum, K.R.; Blomhoff, R. Storage of Vitamin A in Extrahepatic Stellate Cells in Normal Rats. J. Lipid Res. 1997, 38, 645–658. [Google Scholar] [CrossRef] [PubMed]
  67. Cha, J.J.; Mandal, C.; Ghee, J.Y.; Yoo, J.A.; Lee, M.J.; Kang, Y.S.; Hyun, Y.Y.; Lee, J.E.; Kim, H.W.; Han, S.Y.; et al. Inhibition of Renal Stellate Cell Activation Reduces Renal Fibrosis. Biomedicines 2020, 8, 431. [Google Scholar] [CrossRef]
  68. Ezhilarasan, D. Endothelin-1 in Portal Hypertension: The Intricate Role of Hepatic Stellate Cells. Exp. Biol. Med. 2020, 245, 1504. [Google Scholar] [CrossRef]
  69. Pinzani, M.; Milani, S.; De Franco, R.; Grappone, C.; Caligiuri, A.; Gentilini, A.; Tosti-Guerra, C.; Maggi, M.; Failli, P.; Ruocco, C.; et al. Endothelin 1 Is Overexpressed in Human Cirrhotic Liver and Exerts Multiple Effects on Activated Hepatic Stellate Cells. Gastroenterology 1996, 110, 534–548. [Google Scholar] [CrossRef] [PubMed]
  70. Giulia Battelli, M.; Bolognesi, A.; Korsmo, H.W.; Ekperikpe, U.S.; Daehn, I.S. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants 2024, 13, 712. [Google Scholar] [CrossRef]
  71. Chao, H.H.; Liu, J.C.; Lin, J.W.; Chen, C.H.; Wu, C.H.; Cheng, T.H. Uric Acid Stimulates Endothelin-1 Gene Expression Associated with NADPH Oxidase in Human Aortic Smooth Muscle Cells. Acta Pharmacol. Sin. 2008, 29, 1301–1312. [Google Scholar] [CrossRef]
  72. Khosla, U.M.; Zharikov, S.; Finch, J.L.; Nakagawa, T.; Roncal, C.; Mu, W.; Krotova, K.; Block, E.R.; Prabhakar, S.; Johnson, R.J. Hyperuricemia Induces Endothelial Dysfunction. Kidney Int. 2005, 67, 1739–1742. [Google Scholar] [CrossRef] [PubMed]
  73. Roumeliotis, S.; Mallamaci, F.; Zoccali, C. Endothelial Dysfunction in Chronic Kidney Disease, from Biology to Clinical Outcomes: A 2020 Update. J. Clin. Med. 2020, 9, 2359. [Google Scholar] [CrossRef] [PubMed]
  74. Daehn, I.S.; Duffield, J.S. The Glomerular Filtration Barrier: A Structural Target for Novel Kidney Therapies. Nat. Rev. Drug Discov. 2021, 20, 770–788. [Google Scholar] [CrossRef] [PubMed]
  75. Meliambro, K.; He, J.C.; Campbell, K.N. Podocyte-Targeted Therapies—Progress and Future Directions. Nat. Rev. Nephrol. 2024, 20, 643–658. [Google Scholar] [CrossRef] [PubMed]
  76. Kadatane, S.P.; Satariano, M.; Massey, M.; Mongan, K.; Raina, R. The Role of Inflammation in CKD. Cells 2023, 12, 1581. [Google Scholar] [CrossRef] [PubMed]
  77. Ebefors, K.; Wiener, R.J.; Yu, L.; Azeloglu, E.U.; Yi, Z.; Jia, F.; Zhang, W.; Baron, M.H.; He, J.C.; Haraldsson, B.; et al. Endothelin Receptor-A Mediates Degradation of the Glomerular Endothelial Surface Layer via Pathologic Crosstalk between Activated Podocytes and Glomerular Endothelial Cells. Kidney Int. 2019, 96, 957–970. [Google Scholar] [CrossRef] [PubMed]
  78. Abbate, M.; Zoja, C.; Morigi, M.; Rottoli, D.; Angioletti, S.; Tomasoni, S.; Zanchi, C.; Longaretti, L.; Donadelli, R.; Remuzzi, G. Transforming Growth Factor-Β1 Is Up-Regulated by Podocytes in Response to Excess Intraglomerular Passage of Proteins: A Central Pathway in Progressive Glomerulosclerosis. Am. J. Pathol. 2002, 161, 2179. [Google Scholar] [CrossRef] [PubMed]
  79. Lee, H.S. Mechanisms and Consequences of TGF-β Overexpression by Podocytes in Progressive Podocyte Disease. Cell Tissue Res. 2012, 347, 129–140. [Google Scholar] [CrossRef]
  80. Dar, W.A.; Sullivan, E.; Bynon, J.S.; Eltzschig, H.; Ju, C. Ischemia Reperfusion Injury in Liver Transplantation: Cellular and Molecular Mechanisms. Liver Int. 2019, 39, 788. [Google Scholar] [CrossRef]
  81. Bonventre, J.V.; Yang, L. Cellular Pathophysiology of Ischemic Acute Kidney Injury. J. Clin. Investig. 2011, 121, 4210. [Google Scholar] [CrossRef]
  82. Mallikarjuna, P.; Zhou, Y.; Landström, M. The Synergistic Cooperation between TGF-β and Hypoxia in Cancer and Fibrosis. Biomolecules 2022, 12, 635. [Google Scholar] [CrossRef] [PubMed]
  83. Goto, M.; Takei, Y.; Kawano, S.; Nagano, K.; Tsuji, S.; Masuda, E.; Nishimura, Y.; Okumura, S.; Kashiwagi, T.; Fusamoto, H.; et al. Endothelin-1 Is Involved in the Pathogenesis of Ischemia/Reperfusion Liver Injury by Hepatic Microcirculatory Disturbances. Hepatology 1994, 19, 675–681. [Google Scholar] [CrossRef] [PubMed]
  84. Baumann, B.; Hayashida, T.; Liang, X.; Schnaper, H.W. Hypoxia-Inducible Factor-1α Promotes Glomerulosclerosis and Regulates COL1A2 Expression through Interactions with Smad3. Kidney Int. 2016, 90, 797–808. [Google Scholar] [CrossRef] [PubMed]
  85. Clambey, E.T.; McNamee, E.N.; Westrich, J.A.; Glover, L.E.; Campbell, E.L.; Jedlicka, P.; De Zoeten, E.F.; Cambier, J.C.; Stenmark, K.R.; Colgan, S.P.; et al. Hypoxia-Inducible Factor-1 Alpha-Dependent Induction of FoxP3 Drives Regulatory T-Cell Abundance and Function during Inflammatory Hypoxia of the Mucosa. Proc. Natl. Acad. Sci. USA 2012, 109, E2784–E2793. [Google Scholar] [CrossRef] [PubMed]
  86. Stow, L.R.; Jacobs, M.E.; Wingo, C.S.; Cain, B.D. Endothelin-1 Gene Regulation. FASEB J. 2011, 25, 16. [Google Scholar] [CrossRef] [PubMed]
  87. Mohapatra, S.R.; Sadik, A.; Sharma, S.; Poschet, G.; Gegner, H.M.; Lanz, T.V.; Lucarelli, P.; Klingmüller, U.; Platten, M.; Heiland, I.; et al. Hypoxia Routes Tryptophan Homeostasis Towards Increased Tryptamine Production. Front. Immunol. 2021, 12, 1. [Google Scholar] [CrossRef]
  88. Szabo, A.; Kovacs, A.; Riba, J.; Djurovic, S.; Rajnavolgyi, E.; Frecska, E. The Endogenous Hallucinogen and Trace Amine N,N-Dimethyltryptamine (DMT) Displays Potent Protective Effects against Hypoxia via Sigma-1 Receptor Activation in Human Primary IPSC-Derived Cortical Neurons and Microglia-like Immune Cells. Front. Neurosci. 2016, 10, 207848. [Google Scholar] [CrossRef]
  89. Nasiri-Ansari, N.; Androutsakos, T.; Flessa, C.M.; Kyrou, I.; Siasos, G.; Randeva, H.S.; Kassi, E.; Papavassiliou, A.G. Endothelial Cell Dysfunction and Nonalcoholic Fatty Liver Disease (NAFLD): A Concise Review. Cells 2022, 11, 2511. [Google Scholar] [CrossRef] [PubMed]
  90. Daehn, I.; Bottinger, E.P. Microvascular Endothelial Cells Poised to Take Center Stage in Experimental Renal Fibrosis. J. Am. Soc. Nephrol. 2015, 26, 767. [Google Scholar] [CrossRef]
  91. Fontanilla, D.; Johannessen, M.; Hajipour, A.R.; Cozzi, N.V.; Jackson, M.B.; Ruoho, A.E. The Hallucinogen N,N-Dimethyltryptamine (DMT) Is an Endogenous Sigma-1 Receptor Regulator. Science 2009, 323, 934. [Google Scholar] [CrossRef]
  92. Kärkkäinen, J.; Forsström, T.; Tornaeus, J.; Wähälä, K.; Kiuru, P.; Honkanen, A.; Stenman, U.H.; Turpeinen, U.; Hesso, A. Potentially Hallucinogenic 5-Hydroxytryptamine Receptor Ligands Bufotenine and Dimethyltryptamine in Blood and Tissues. Scand. J. Clin. Lab. Investig. 2005, 65, 189–199. [Google Scholar] [CrossRef]
  93. Wang, J.; Xu, D.; Shen, L.; Zhou, J.; Lv, X.; Ma, H.; Li, N.; Wu, Q.; Duan, J. Anti-Inflammatory and Analgesic Actions of Bufotenine through Inhibiting Lipid Metabolism Pathway. Biomed. Pharmacother. 2021, 140, 111749. [Google Scholar] [CrossRef] [PubMed]
  94. Batalla, M. All-Natural 5-MeO-DMT Sigma Receptor 1 Agonist and Its Therapeutic Impact in Mental and Neurodegener-Ative Diseases through Mitochondrial Activation. Sci. Rev. Biol. 2023, 2, 1–20. [Google Scholar] [CrossRef]
  95. Sawyer, E.M.; Jensen, L.E.; Meehl, J.B.; Larsen, K.P.; Petito, D.A.; Hurley, J.H.; Voeltz, G.K. SigmaR1 Shapes Rough Endoplasmic Reticulum Membrane Sheets. Dev. Cell 2024, 59, 2566–2577. [Google Scholar] [CrossRef] [PubMed]
  96. Ke, H.; Su, X.; Dong, C.; He, Z.; Song, Q.; Song, C.; Zhou, J.; Liao, W.; Wang, C.; Yang, S.; et al. Sigma-1 Receptor Exerts Protective Effects on Ameliorating Nephrolithiasis by Modulating Endoplasmic Reticulum-Mitochondrion Association and Inhibiting Endoplasmic Reticulum Stress-Induced Apoptosis in Renal Tubular Epithelial Cells. Redox Rep. 2024, 29, 2391139. [Google Scholar] [CrossRef]
  97. Bernard-Marissal, N.; Médard, J.J.; Azzedine, H.; Chrast, R. Dysfunction in Endoplasmic Reticulum-Mitochondria Crosstalk Underlies SIGMAR1 Loss of Function Mediated Motor Neuron Degeneration. Brain 2015, 138, 875–890. [Google Scholar] [CrossRef] [PubMed]
  98. Abdullah, C.S.; Aishwarya, R.; Alam, S.; Remex, N.S.; Morshed, M.; Nitu, S.; Miriyala, S.; Panchatcharam, M.; Hartman, B.; King, J.; et al. The Molecular Role of Sigmar1 in Regulating Mitochondrial Function through Mitochondrial Localization in Cardiomyocytes. Mitochondrion 2022, 62, 159–175. [Google Scholar] [CrossRef] [PubMed]
  99. Aishwarya, R.; Abdullah, C.S.; Morshed, M.; Remex, N.S.; Bhuiyan, M.S. Sigmar1’s Molecular, Cellular, and Biological Functions in Regulating Cellular Pathophysiology. Front. Physiol. 2021, 12, 705575. [Google Scholar] [CrossRef]
  100. Saleh, S.R.; Younis, F.A.; Abdelrahman, S.S.; Attia, A.A.; El-Demellawy, M.A.; Newairy, A.A.; Ghareeb, D.A. Attenuation of High-Fat High-Sucrose Diet and CCl4-Induced Non-Alcoholic Steatohepatitis in Rats by Activating Autophagy and SIGMAR1/GRP78/ITPR1 Signaling Using Berberine-Loaded Albumin Nanoparticles: In Vivo Prediction and in-Silico Molecular Modeling. J. Pharm. Investig. 2024, 54, 1–24. [Google Scholar] [CrossRef]
  101. 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]
  102. Zhang, K.K.; Yang, J.Z.; Cheng, C.H.; Wan, J.Y.; Chen, Y.C.; Zhou, H.Q.; Zheng, D.K.; Lan, Z.X.; You, Q.H.; Wang, Q.; et al. Short-Chain Fatty Acids Mitigate Methamphetamine-Induced Hepatic Injuries in a Sigma-1 Receptor-Dependent Manner. Ecotoxicol. Environ. Saf. 2024, 280, 116538. [Google Scholar] [CrossRef] [PubMed]
  103. Remex, N.S.; Abdullah, C.S.; Aishwarya, R.; Kolluru, G.K.; Traylor, J.; Bhuiyan, M.A.N.; Kevil, C.G.; Orr, A.W.; Rom, O.; Pattillo, C.B.; et al. Deletion of Sigmar1 Leads to Increased Arterial Stiffness and Altered Mitochondrial Respiration Resulting in Vascular Dysfunction. Front. Physiol. 2024, 15, 1386296. [Google Scholar] [CrossRef] [PubMed]
  104. Mavlyutov, T.A.; Epstein, M.L.; Liu, P.; Verbny, Y.I.; Ziskind-Conhaim, L.; Ruoho, A.E. Development of the Sigma-1 Receptor in C-Terminals of Motoneurons and Colocalization with the N,N′-Dimethyltryptamine Forming Enzyme, Indole-N-Methyl Transferase. Neuroscience 2012, 206, 60–68. [Google Scholar] [CrossRef] [PubMed]
  105. Frecska, E.; Bokor, P.; Winkelman, M. The Therapeutic Potentials of Ayahuasca: Possible Effects against Various Diseases of Civilization. Front. Pharmacol. 2016, 7, 174229. [Google Scholar] [CrossRef]
  106. Aguanno, A.; Afar, R.; Albert, V.R. Tissue-Specific Expression of the Nonneuronal Promoter of the Aromatic L-Amino Acid Decarboxylase Gene Is Regulated by Hepatocyte Nuclear Factor. J. Biol. Chem. 1996, 271, 4528–4538. [Google Scholar] [CrossRef]
  107. Párrizas, M.; Maestro, M.A.; Boj, S.F.; Paniagua, A.; Casamitjana, R.; Gomis, R.; Rivera, F.; Ferrer, J. Hepatic Nuclear Factor 1-α Directs Nucleosomal Hyperacetylation to Its Tissue-Specific Transcriptional Targets. Mol. Cell Biol. 2001, 21, 3234–3243. [Google Scholar] [CrossRef]
  108. Rebouissou, S.; Imbeaud, S.; Balabaud, C.; Boulanger, V.; Bertrand-Michel, J.; Tercé, F.; Auffray, C.; Bioulac-Sage, P.; Zucman-Rossi, J.; Bertrand, J. HNF1alpha Inactivation Promotes Lipogenesis in Human Hepatocellular Adenoma Independently of SREBP-1 and Carbohydrate-Response Element-Binding Protein (ChREBP) Activation. J. Biol. Chem. 2007, 282, 14437–14446. [Google Scholar] [CrossRef]
  109. Dean, J.G. Indolethylamine- N-Methyltransferase Polymorphisms: Genetic and Biochemical Approaches for Study of Endogenous N,N,-Dimethyltryptamine. Front. Neurosci. 2018, 12, 232. [Google Scholar] [CrossRef]
  110. Vargas, M.V.; Dunlap, L.E.; Dong, C.; Carter, S.J.; Tombari, R.J.; Jami, S.A.; Cameron, L.P.; Patel, S.D.; Hennessey, J.J.; Saeger, H.N.; et al. Psychedelics Promote Neuroplasticity Through Activation of Intracellular 5-HT2A Receptors. Science 2023, 379, 700. [Google Scholar] [CrossRef]
  111. Servillo, L.; Giovane, A.; Balestrieri, M.L.; Cautela, D.; Castaldo, D. N-Methylated Tryptamine Derivatives in Citrus Genus Plants: Identification of N,N,N-Trimethyltryptamine in Bergamot. J. Agric. Food Chem. 2012, 60, 9512–9518. [Google Scholar] [CrossRef] [PubMed]
  112. Chadeayne, A.R.; Manke, D.R.; Pham, D.N.K.; Reid, B.G.; Golen, J.A. Active Metabolite of Aeruginascin (4-Hydroxy-N,N,N-Trimethyltryptamine): Synthesis, Structure, and Serotonergic Binding Affinity. ACS Omega 2020, 5, 16940–16943. [Google Scholar] [CrossRef] [PubMed]
  113. Roberts, K.M.; Fitzpatrick, P.F. Mechanisms of Tryptophan and Tyrosine Hydroxylase. IUBMB Life 2013, 65, 350. [Google Scholar] [CrossRef] [PubMed]
  114. Tan, D.X.; Hardeland, R.; Back, K.; Manchester, L.C.; Alatorre-Jimenez, M.A.; Reiter, R.J. On the Significance of an Alternate Pathway of Melatonin Synthesis via 5-Methoxytryptamine: Comparisons across Species. J. Pineal Res. 2016, 61, 27–40. [Google Scholar] [CrossRef]
  115. Wiechmann, A.F.; Bok, D.; Horwitzj, J. Localization of Hydroxyindole-O-Methyltransferase in the Mammalian Pineal Gland and Retina. Invest. Ophthalmol. Vis. Sci. 1985, 26, 253–265. [Google Scholar] [PubMed]
  116. Wyatt, R.J.; Saavedra, J.M.; Axelrod, J. A Dimethyltryptamine-Forming Enzyme in Human Blood. Am. J. Psychiatry 1973, 130, 754–760. [Google Scholar] [CrossRef] [PubMed]
  117. Zohairi, F.; Khandelia, H.; Hakami Zanjani, A.A. Interaction of Psychedelic Tryptamine Derivatives with a Lipid Bilayer. Chem. Phys. Lipids 2023, 251, 105279. [Google Scholar] [CrossRef]
  118. Mandel, L.R.; Walker, R.W. The Biosynthesis of 5-Methoxy-N,N-Dimethyltryptamine Invitro. Life Sci. 1974, 15, 1457–1463. [Google Scholar] [CrossRef] [PubMed]
  119. Glynos, N.G.; Carter, L.; Lee, S.J.; Kim, Y.; Kennedy, R.T.; Mashour, G.A.; Wang, M.M.; Borjigin, J. Indolethylamine N-Methyltransferase (INMT) Is Not Essential for Endogenous Tryptamine-Dependent Methylation Activity in Rats. Sci. Rep. 2023, 13, 280. [Google Scholar] [CrossRef]
  120. Sayers, E.W.; Bolton, E.E.; Brister, J.R.; Canese, K.; Chan, J.; Comeau, D.C.; Connor, R.; Funk, K.; Kelly, C.; Kim, S.; et al. Database Resources of the National Center for Biotechnology Information. Nucleic Acids Res. 2022, 50, D20–D26. [Google Scholar] [CrossRef]
  121. Zhong, S.; Jeong, J.H.; Huang, C.; Chen, X.; Dickinson, S.I.; Dhillon, J.; Yang, L.; Luo, J.L. Targeting INMT and Interrupting Its Methylation Pathway for the Treatment of Castration Resistant Prostate Cancer. J. Exp. Clin. Cancer Res. 2021, 40, 307. [Google Scholar] [CrossRef]
  122. Cui, K.; Yao, X.; Wei, Z.; Yang, Y.; Liu, X.; Huang, Z.; Huo, H.; Tang, J.; Xie, Y. Poor Prognosis, Hypomethylation, and Immune Infiltrates Are Associated with Downregulation of INMT in Head and Neck Squamous Cell Carcinoma. Front. Genet. 2022, 13, 917344. [Google Scholar] [CrossRef] [PubMed]
  123. Coutinho, L.P.; Silva, S.R.B.; de Lima-Neto, P.; Monteiro, N.d.K.V. A Mechanistic Insight for the Biosynthesis of N,N-Dimethyltryptamine: An ONIOM Theoretical Approach. Biochem. Biophys. Res. Commun. 2023, 678, 148–157. [Google Scholar] [CrossRef] [PubMed]
  124. Kubicskó, K.; Farkas, Ö. Quantum Chemical (QM:MM) Investigation of the Mechanism of Enzymatic Reaction of Tryptamine and: N,N-Dimethyltryptamine with Monoamine Oxidase A. Org. Biomol. Chem. 2020, 18, 9660–9674. [Google Scholar] [CrossRef] [PubMed]
  125. Burden, D.A.; Philips, S.R. Kinetic Measurements of the Turnover Rates of Phenylethylamine and Tryptamine in Vivo in the Rat Brain. J. Neurochem. 1980, 34, 1725–1732. [Google Scholar] [CrossRef] [PubMed]
  126. Yang, W.S.; Kim, K.J.; Gaschler, M.M.; Patel, M.; Shchepinov, M.S.; Stockwell, B.R. Peroxidation of Polyunsaturated Fatty Acids by Lipoxygenases Drives Ferroptosis. Proc. Natl. Acad. Sci. USA 2016, 113, E4966–E4975. [Google Scholar] [CrossRef]
  127. Ingold, I.; Berndt, C.; Schmitt, S.; Doll, S.; Poschmann, G.; Buday, K.; Roveri, A.; Peng, X.; Porto Freitas, F.; Seibt, T.; et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell 2018, 172, 409–422.e21. [Google Scholar] [CrossRef] [PubMed]
  128. Zou, Y.; Henry, W.S.; Ricq, E.L.; Graham, E.T.; Phadnis, V.V.; Maretich, P.; Paradkar, S.; Boehnke, N.; Deik, A.A.; Reinhardt, F.; et al. Plasticity of Ether Lipids Promotes Ferroptosis Susceptibility and Evasion. Nature 2020, 585, 603. [Google Scholar] [CrossRef] [PubMed]
  129. Perez, M.A.; Clostio, A.J.; Houston, I.R.; Ruiz, J.; Magtanong, L.; Dixon, S.J.; Watts, J.L. Ether Lipid Deficiency Disrupts Lipid Homeostasis Leading to Ferroptosis Sensitivity. PLoS Genet. 2022, 18, e1010436. [Google Scholar] [CrossRef]
  130. Braverman, N.E.; Moser, A.B. Functions of Plasmalogen Lipids in Health and Disease. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2012, 1822, 1442–1452. [Google Scholar] [CrossRef]
  131. Bellmaine, S.; Schnellbaecher, A.; Zimmer, A. Reactivity and Degradation Products of Tryptophan in Solution and Proteins. Free Radic. Biol. Med. 2020, 160, 696–718. [Google Scholar] [CrossRef]
  132. Banushi, B.; Polito, V. A Comprehensive Review of the Current Status of the Cellular Neurobiology of Psychedelics. Biology 2023, 12, 1380. [Google Scholar] [CrossRef] [PubMed]
  133. Gomes, M.M.; Coimbra, J.B.; Clara, R.O.; Dörr, F.A.; Moreno, A.C.R.; Chagas, J.R.; Tufik, S.; Pinto, E.; Catalani, L.H.; Campa, A. Biosynthesis of N,N-Dimethyltryptamine (DMT) in a Melanoma Cell Line and Its Metabolization by Peroxidases. Biochem. Pharmacol. 2014, 88, 393–401. [Google Scholar] [CrossRef]
  134. Madrid-Gambin, F.; Fabregat-Safont, D.; Gomez-Gomez, A.; Olesti, E.; Mason, N.L.; Ramaekers, J.G.; Pozo, O.J. Present and Future of Metabolic and Metabolomics Studies Focused on Classical Psychedelics in Humans. Biomed. Pharmacother. 2023, 169, 115775. [Google Scholar] [CrossRef] [PubMed]
  135. Bröer, S.; Bröer, A. Amino Acid Homeostasis and Signalling in Mammalian Cells and Organisms. Biochem. J. 2017, 474, 1935–1963. [Google Scholar] [CrossRef] [PubMed]
  136. Ramadan, T.; Camargo, S.M.R.; Herzog, B.; Bordin, M.; Pos, K.M.; Verrey, F. Recycling of Aromatic Amino Acids via TAT1 Allows Efflux of Neutral Amino Acids via LAT2-4F2hc Exchanger. Pflug. Arch. 2007, 454, 507–516. [Google Scholar] [CrossRef] [PubMed]
  137. Teunis, C.; Nieuwdorp, M.; Hanssen, N. Interactions between Tryptophan Metabolism, the Gut Microbiome and the Immune System as Potential Drivers of Non-Alcoholic Fatty Liver Disease (NAFLD) and Metabolic Diseases. Metabolites 2022, 12, 514. [Google Scholar] [CrossRef]
  138. Salminen, A. Activation of Aryl Hydrocarbon Receptor (AhR) in Alzheimer’s Disease: Role of Tryptophan Metabolites Generated by Gut Host-Microbiota. J. Mol. Med. 2023, 101, 201–222. [Google Scholar] [CrossRef]
  139. Marszalek-Grabska, M.; Walczak, K.; Gawel, K.; Wicha-Komsta, K.; Wnorowska, S.; Wnorowski, A.; Turski, W.A. Kynurenine Emerges from the Shadows—Current Knowledge on Its Fate and Function. Pharmacol. Ther. 2021, 225, 107845. [Google Scholar] [CrossRef]
  140. Krishnan, S.; Ding, Y.; Saedi, N.; Choi, M.; Sridharan, G.V.; Sherr, D.H.; Yarmush, M.L.; Alaniz, R.C.; Jayaraman, A.; Lee, K. Gut Microbiota-Derived Tryptophan Metabolites Modulate Inflammatory Response in Hepatocytes and Macrophages. Cell Rep. 2018, 23, 1099–1111. [Google Scholar] [CrossRef]
  141. Choi, W.; Moon, J.H.; Kim, H. Serotonergic Regulation of Energy Metabolism in Peripheral Tissues. J. Endocrinol. 2020, 245, R1–R10. [Google Scholar] [CrossRef]
  142. Shen, L.; Lv, X.; Yang, X.; Deng, S.; Liu, L.; Zhou, J.; Zhu, Y.; Ma, H. Bufotenines-Loaded Liposome Exerts Anti-Inflammatory, Analgesic Effects and Reduce Gastrointestinal Toxicity through Altering Lipid and Bufotenines Metabolism. Biomed. Pharmacother. 2022, 153, 113492. [Google Scholar] [CrossRef] [PubMed]
  143. Li, J.; Félix-Soriano, E.; Wright, K.R.; Shen, H.; Baer, L.A.; Stanford, K.I.; Guo, L.-W. Differential Responses to Sigma-1 or Sigma-2 Receptor Ablation in Adiposity, Fat Oxidation, and Sexual Dimorphism. Int. J. Mol. Sci. 2022, 2022, 10846. [Google Scholar] [CrossRef]
  144. Guo, M.; Liu, R.; Zhang, F.; Qu, J.; Yang, Y.; Li, X. A New Perspective on Liver Diseases: Focusing on the Mitochondria-Associated Endoplasmic Reticulum Membranes. Pharmacol. Res. 2024, 208, 107409. [Google Scholar] [CrossRef] [PubMed]
  145. Ray, T.S. Psychedelics and the Human Receptorome. PLoS ONE 2010, 5, 9019. [Google Scholar] [CrossRef]
  146. Halberstadt, A.L.; Geyer, M.A. Multiple Receptors Contribute to the Behavioral Effects of Indoleamine Hallucinogens. Neuropharmacology 2011, 61, 364. [Google Scholar] [CrossRef]
  147. McKenna, D.; Riba, J. New World Tryptamine Hallucinogens and the Neuroscience of Ayahuasca. Curr. Top. Behav. Neurosci. 2018, 36, 283–311. [Google Scholar]
  148. Jacob, M.S.; Presti, D.E. Endogenous Psychoactive Tryptamines Reconsidered: An Anxiolytic Role for Dimethyltryptamine. Med. Hypotheses 2005, 64, 930–937. [Google Scholar] [CrossRef]
  149. Kozell, L.B.; Eshleman, A.J.; Swanson, T.L.; Bloom, S.H.; Wolfrum, K.M.; Schmachtenberg, J.L.; Olson, R.J.; Janowsky, A.; Abbas, A.I. Pharmacologic Activity of Substituted Tryptamines at 5-Hydroxytryptamine (5-HT)2A Receptor (5-HT2AR), 5-HT2CR, 5-HT1AR, and Serotonin Transporter. J. Pharmacol. Exp. Ther. 2023, 385, 62. [Google Scholar] [CrossRef]
  150. Namkung, J.; Shong, K.E.; Kim, H.; Oh, C.M.; Park, S.; Kim, H. Inhibition of Serotonin Synthesis Induces Negative Hepatic Lipid Balance. Diabetes Metab. J. 2018, 42, 233–243. [Google Scholar] [CrossRef]
  151. Yabut, J.M.; Crane, J.D.; Green, A.E.; Keating, D.J.; Khan, W.I.; Steinberg, G.R. Emerging Roles for Serotonin in Regulating Metabolism: New Implications for an Ancient Molecule. Endocr. Rev. 2019, 40, 1092. [Google Scholar] [CrossRef]
  152. Watanabe, H.; Akasaka, D.; Ogasawara, H.; Sato, K.; Miyake, M.; Saito, K.; Takahashi, Y.; Kanaya, T.; Takakura, I.; Hondo, T.; et al. Peripheral Serotonin Enhances Lipid Metabolism by Accelerating Bile Acid Turnover. Endocrinology 2010, 151, 4776–4786. [Google Scholar] [CrossRef] [PubMed]
  153. Moon, J.H.; Oh, C.M.; Kim, H. Serotonin in the Regulation of Systemic Energy Metabolism. J. Diabetes Investig. 2022, 13, 1639. [Google Scholar] [CrossRef] [PubMed]
  154. El-Merahbi, R.; Löffler, M.; Mayer, A.; Sumara, G. The Roles of Peripheral Serotonin in Metabolic Homeostasis. FEBS Lett. 2015, 589, 1728–1734. [Google Scholar] [CrossRef] [PubMed]
  155. Ruddell, R.G.; Oakley, F.; Hussain, Z.; Yeung, I.; Bryan-Lluka, L.J.; Ramm, G.A.; Mann, D.A. A Role for Serotonin (5-HT) in Hepatic Stellate Cell Function and Liver Fibrosis. Am. J. Pathol. 2006, 169, 861–876. [Google Scholar] [CrossRef] [PubMed]
  156. Redenšek Trampuž, S.; van Riet, S.; Nordling, Å.; Ingelman-Sundberg, M. Mechanisms of 5-HT Receptor Antagonists in the Regulation of Fibrosis in a 3D Human Liver Spheroid Model. Sci. Rep. 2024, 14, 1396. [Google Scholar] [CrossRef] [PubMed]
  157. Park, J.; Jeong, W.; Yun, C.; Kim, H.; Oh, C.M. Serotonergic Regulation of Hepatic Energy Metabolism. Endocrinol. Metab. 2021, 36, 1151. [Google Scholar] [CrossRef]
  158. Shajib, M.S.; Khan, W.I. The Role of Serotonin and Its Receptors in Activation of Immune Responses and Inflammation. Acta Physiol. 2015, 213, 561–574. [Google Scholar] [CrossRef]
  159. Herr, N.; Bode, C.; Duerschmied, D. The Effects of Serotonin in Immune Cells. Front. Cardiovasc. Med. 2017, 4, 48. [Google Scholar] [CrossRef]
  160. Gülçin, I. Measurement of Antioxidant Ability of Melatonin and Serotonin by the DMPD and CUPRAC Methods as Trolox Equivalent. J. Enzym. Inhib. Med. Chem. 2008, 23, 871–876. [Google Scholar] [CrossRef]
  161. Azouzi, S.; Santuz, H.; Morandat, S.; Pereira, C.; Côté, F.; Hermine, O.; El Kirat, K.; Colin, Y.; Le Van Kim, C.; Etchebest, C.; et al. Antioxidant and Membrane Binding Properties of Serotonin Protect Lipids from Oxidation. Biophys. J. 2017, 112, 1863. [Google Scholar] [CrossRef]
  162. Bunzow, J.R.; Sonders, M.S.; Arttamangkul, S.; Harrison, L.M.; Zhang, G.; Quigley, D.I.; Darland, T.; Suchland, K.L.; Pasumamula, S.; Kennedy, J.L.; et al. Amphetamine, 3,4-Methylenedioxymethamphetamine, Lysergic Acid Diethylamide, and Metabolites of the Catecholamine Neurotransmitters Are Agonists of a Rat Trace Amine Receptor. Mol. Pharmacol. 2001, 60, 1181–1188. [Google Scholar] [CrossRef] [PubMed]
  163. Berry, M.D.; Gainetdinov, R.R.; Hoener, M.C.; Shahid, M. Pharmacology of Human Trace Amine-Associated Receptors: Therapeutic Opportunities and Challenges. Pharmacol. Ther. 2017, 180, 161–180. [Google Scholar] [CrossRef] [PubMed]
  164. Vaganova, A.N.; Katolikova, N.V.; Murtazina, R.Z.; Kuvarzin, S.R.; Gainetdinov, R.R. Public Transcriptomic Data Meta-Analysis Demonstrates TAAR6 Expression in the Mental Disorder-Related Brain Areas in Human and Mouse Brain. Biomolecules 2022, 12, 1259. [Google Scholar] [CrossRef] [PubMed]
  165. Sotnikova, T.D.; Caron, M.G.; Gainetdinov, R.R. Trace Amine-Associated Receptors as Emerging Therapeutic Targets. Mol. Pharmacol. 2009, 76, 229. [Google Scholar] [CrossRef] [PubMed]
  166. Jragh, D.; Chandrasekhar, B.; Yousif, M.H.M.; Oriowo, M.A. Vasodilator Effect of Trace Amines, 3-Iodothyronamine, and RO5263397 in the Rat Perfused Kidney: Comparison with Tryptamine. Pharmacology 2023, 108, 368–378. [Google Scholar] [CrossRef]
  167. Zhu, C.P.; Liu, S.Q.; Wang, K.Q.; Xiong, H.L.; Aristu-Zabalza, P.; Boyer-Díaz, Z.; Feng, J.F.; Song, S.H.; Luo, C.; Chen, W.S.; et al. Targeting 5-Hydroxytryptamine Receptor 1A in the Portal Vein to Decrease Portal Hypertension. Gastroenterology 2024, 167, 993–1007. [Google Scholar] [CrossRef]
  168. Housset, C.; Rockey, D.C.; Bissell, D.M. Endothelin Receptors in Rat Liver: Lipocytes as a Contractile Target for Endothelin 1. Proc. Natl. Acad. Sci. USA 1993, 90, 9266–9270. [Google Scholar] [CrossRef]
  169. Rockey, D.C.; Weisiger, R.A. Endothelin Induced Contractility of Stellate Cells from Normal and Cirrhotic Rat Liver: Implications for Regulation of Portal Pressure and Resistance. Hepatology 1996, 24, 233–240. [Google Scholar] [CrossRef]
  170. Gabriel, A.; Kuddus, R.H.; Abdul, S.R.; Watkins, W.D.; Gandhi, C.R. Superoxide-Induced Changes in Endothelin (ET) Receptors in Hepatic Stellate Cells. J. Hepatol. 1998, 29, 614–627. [Google Scholar] [CrossRef]
  171. Rockey, D.C. Stellate Cells and Portal Hypertension. In Stellate Cells in Health and Disease; Academic Press: Cambridge, MA, USA, 2015; pp. 125–144. ISBN 9780128005446. [Google Scholar]
  172. Albadawy, R.; Agwa, S.H.A.; Khairy, E.; Saad, M.; El Touchy, N.; Othman, M.; El Kassas, M.; Matboli, M. Circulatory Endothelin 1-Regulating Rnas Panel: Promising Biomarkers for Non-Invasive Nafld/Nash Diagnosis and Stratification: Clinical and Molecular Pilot Study. Genes 2021, 12, 1813. [Google Scholar] [CrossRef]
  173. Okamoto, T.; Koda, M.; Miyoshi, K.; Onoyama, T.; Kishina, M.; Matono, T.; Sugihara, T.; Hosho, K.; Okano, J.; Isomoto, H.; et al. Antifibrotic Effects of Ambrisentan, an Endothelin-A Receptor Antagonist, in a Non-Alcoholic Steatohepatitis Mouse Model. World J. Hepatol. 2016, 8, 933. [Google Scholar] [CrossRef] [PubMed]
  174. Kimura, T.; Singh, S.; Tanaka, N.; Umemura, T. Role of G Protein-Coupled Receptors in Hepatic Stellate Cells and Approaches to Anti-Fibrotic Treatment of Non-Alcoholic Fatty Liver Disease. Front. Endocrinol. 2021, 12, 773432. [Google Scholar] [CrossRef] [PubMed]
  175. Böhm, F.; Pernow, J. The Importance of Endothelin-1 for Vascular Dysfunction in Cardiovascular Disease. Cardiovasc. Res. 2007, 76, 8–18. [Google Scholar] [CrossRef] [PubMed]
  176. Barton, M.; Yanagisawa, M. Endothelin: 30 Years from Discovery to Therapy. Hypertension 2019, 74, 1232–1265. [Google Scholar] [CrossRef]
  177. Shi-Wen, X.; Chen, Y.; Denton, C.P.; Eastwood, M.; Renzoni, E.A.; Bou-Gharios, G.; Pearson, J.D.; Dashwood, M.; Du Bois, R.M.; Black, C.M.; et al. Endothelin-1 Promotes Myofibroblast Induction through the ETA Receptor via a Rac/Phosphoinositide 3-Kinase/Akt-Dependent Pathway and Is Essential for the Enhanced Contractile Phenotype of Fibrotic Fibroblasts. Mol. Biol. Cell 2004, 15, 2707–2719. [Google Scholar] [CrossRef]
  178. Dupuis, J.; Stewart, D.J.; Cernacek, P.; Gosselin, G. Human Pulmonary Circulation Is an Important Site for Both Clearance and Production of Endothelin-1. Circulation 1996, 94, 1578–1584. [Google Scholar] [CrossRef]
  179. Lassén, E.; Daehn, I.S. Clues to Glomerular Cell Chatter in Focal Segmental Glomerulosclerosis: Via Endothelin-1/ET A R. Kidney Int. Rep. 2021, 6, 1758–1760. [Google Scholar] [CrossRef]
  180. Lassén, E.; Daehn, I.S. Molecular Mechanisms in Early Diabetic Kidney Disease: Glomerular Endothelial Cell Dysfunction. Int. J. Mol. Sci. 2020, 21, 9456. [Google Scholar] [CrossRef]
  181. Qi, H.; Casalena, G.; Shi, S.; Yu, L.; Ebefors, K.; Sun, Y.; Zhang, W.; D’Agati, V.; Schlondorff, D.; Haraldsson, B.; et al. Glomerular Endothelial Mitochondrial Dysfunction Is Essential and Characteristic of Diabetic Kidney Disease Susceptibility. Diabetes 2017, 66, 763–778. [Google Scholar] [CrossRef]
  182. Daehn, I.; Casalena, G.; Zhang, T.; Shi, S.; Fenninger, F.; Barasch, N.; Yu, L.; D’Agati, V.; Schlondorff, D.; Kriz, W.; et al. Endothelial Mitochondrial Oxidative Stress Determines Podocyte Depletion in Segmental Glomerulosclerosis. J. Clin. Investig. 2014, 124, 1608. [Google Scholar] [CrossRef]
  183. Cho, J.-J.; Hocher, B.; Herbst, H.; Jia, J.-D.; Ruehl, M.; Hahn, E.G.; Otto Riecken, E.; Schuppan, D. An Oral Endothelin-A Receptor Antagonist Blocks Collagen Synthesis and Deposition in Advanced Rat Liver Fibrosis. Gastroenerology 2000, 118, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
  184. Örmeci, N. Endothelins and Liver Cirrhosis. Portal Hypertens. Cirrhosis 2022, 1, 66–72. [Google Scholar] [CrossRef]
  185. Lambers, C.; Roth, M.; Zhong, J.; Campregher, C.; Binder, P.; Burian, B.; Petkov, V.; Block, L.H. The Interaction of Endothelin-1 and TGF-Β1 Mediates Vascular Cell Remodeling. PLoS ONE 2013, 8, e73399. [Google Scholar] [CrossRef] [PubMed]
  186. Jain, R.; Shaul, P.W.; Borok, Z.; Willis, B.C. Endothelin-1 Induces Alveolar Epithelial–Mesenchymal Transition through Endothelin Type A Receptor–Mediated Production of TGF-Β1. Am. J. Respir. Cell Mol. Biol. 2007, 37, 38. [Google Scholar] [CrossRef] [PubMed]
  187. Shi, M.; Zhu, J.; Wang, R.; Chen, X.; Mi, L.; Walz, T.; Springer, T.A. Latent TGF-β Structure and Activation. Nature 2011, 474, 343. [Google Scholar] [CrossRef] [PubMed]
  188. Hiepen, C.; Mendez, P.L.; Knaus, P. It Takes Two to Tango: Endothelial TGFβ/BMP Signaling Crosstalk with Mechanobiology. Cells 2020, 9, 1965. [Google Scholar] [CrossRef]
  189. Goumans, M.J.; Liu, Z.; Ten Dijke, P. TGF-β Signaling in Vascular Biology and Dysfunction. Cell Res. 2008, 19, 116–127. [Google Scholar] [CrossRef]
  190. Chen, P.Y.; Qin, L.; Li, G.; Wang, Z.; Dahlman, J.E.; Malagon-Lopez, J.; Gujja, S.; Cilfone, N.A.; Kauffman, K.J.; Sun, L.; et al. Endothelial TGF-β Signalling Drives Vascular Inflammation and Atherosclerosis. Nat. Metab. 2019, 1, 912. [Google Scholar] [CrossRef]
  191. Blann, A.D.; Wang, J.M.; Wilson, P.B.; Kumar, S. Serum Levels of the TGF-Beta Receptor Are Increased in Atherosclerosis. Atherosclerosis 1996, 120, 221–226. [Google Scholar] [CrossRef]
  192. Ashcroft, G.S. Bidirectional Regulation of Macrophage Function by TGF-β. Microbes Infect. 1999, 1, 1275–1282. [Google Scholar] [CrossRef]
  193. Ueshima, E.; Fujimori, M.; Kodama, H.; Felsen, D.; Chen, J.; Durack, J.C.; Solomon, S.B.; Coleman, J.A.; Srimathveeravalli, G. Macrophage-Secreted TGF-Β1 Contributes to Fibroblast Activation and Ureteral Stricture after Ablation Injury. Am. J. Physiol. Ren. Physiol. 2019, 317, F52. [Google Scholar] [CrossRef]
  194. Ten Hove, M.; Smyris, A.; Booijink, R.; Wachsmuth, L.; Hansen, U.; Alic, L.; Faber, C.; Höltke, C.; Bansal, R. Engineered SPIONs Functionalized with Endothelin a Receptor Antagonist Ameliorate Liver Fibrosis by Inhibiting Hepatic Stellate Cell Activation. Bioact. Mater. 2024, 39, 406. [Google Scholar] [CrossRef] [PubMed]
  195. Szabó, Í.; Varga, V.; Dvorácskó, S.; Farkas, A.E.; Körmöczi, T.; Berkecz, R.; Kecskés, S.; Menyhárt, Á.; Frank, R.; Hantosi, D.; et al. N,N-Dimethyltryptamine Attenuates Spreading Depolarization and Restrains Neurodegeneration by Sigma-1 Receptor Activation in the Ischemic Rat Brain. Neuropharmacology 2021, 192, 108612. [Google Scholar] [CrossRef] [PubMed]
  196. Pal, A.; Fontanilla, D.; Gopalakrishnan, A.; Chae, Y.K.; Markley, J.L.; Ruoho, A.E. The Sigma-1 Receptor Protects against Cellular Oxidative Stress and Activates Antioxidant Response Elements. Eur. J. Pharmacol. 2012, 682, 12. [Google Scholar] [CrossRef] [PubMed]
  197. Rosen, D.A.; Seki, S.M.; Fernández-Castañeda, A.; Beiter, R.M.; Eccles, J.D.; Woodfolk, J.A.; Gaultier, A. Modulation of the Sigma-1 Receptor-IRE1 Pathway Is Beneficial in Preclinical Models of Inflammation and Sepsis. Sci. Transl. Med. 2019, 11, eaau5266. [Google Scholar] [CrossRef]
  198. Das, E.; Sahu, K.K.; Roy, I. The Functional Role of Ire1 in Regulating Autophagy and Proteasomal Degradation under Prolonged Proteotoxic Stress. FEBS J. 2023, 290, 3270–3289. [Google Scholar] [CrossRef]
  199. An, Y.; Qi, Y.; Li, Y.; Li, Z.; Yang, C.; Jia, D. Activation of the Sigma-1 Receptor Attenuates Blood-Brain Barrier Disruption by Inhibiting Amyloid Deposition in Alzheimer’s Disease Mice. Neurosci. Lett. 2022, 774, 136528. [Google Scholar] [CrossRef]
  200. Gaultier, A.; Hollister, M.; Reynolds, I.; Hsieh, E.H.; Gonias, S.L. LRP1 Regulates Remodeling of the Extracellular Matrix by Fibroblasts. Matrix Biol. 2010, 29, 22–30. [Google Scholar] [CrossRef]
  201. 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. [Google Scholar] [CrossRef]
  202. Korsmo, H.W.; Jiang, X. One Carbon Metabolism and Early Development: A Diet-Dependent Destiny. Trends Endocrinol. Metab. 2021, 32, 579–593. [Google Scholar] [CrossRef]
  203. Shioda, N.; Ishikawa, K.; Tagashira, H.; Ishizuka, T.; Yawo, H.; Fukunaga, K. Expression of a Truncated Form of the Endoplasmic Reticulum Chaperone Protein, Σ1 Receptor, Promotes Mitochondrial Energy Depletion and Apoptosis. J. Biol. Chem. 2012, 287, 23318–23331. [Google Scholar] [CrossRef]
  204. Oliphant, K.; Allen-Vercoe, E. Macronutrient Metabolism by the Human Gut Microbiome: Major Fermentation by-Products and Their Impact on Host Health. Microbiome 2019, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
  205. Korsmo, H.W.; Jiang, X.; Caudill, M.A. Choline: Exploring the Growing Science on Its Benefits for Moms and Babies. Nutrients 2019, 11, 1823. [Google Scholar] [CrossRef] [PubMed]
  206. Chen, J.; Vitetta, L. Gut Microbiota Metabolites in NAFLD Pathogenesis and Therapeutic Implications. Int. J. Mol. Sci. 2020, 21, 5214. [Google Scholar] [CrossRef]
  207. Andrikopoulos, P.; Aron-Wisnewsky, J.; Chakaroun, R.; Myridakis, A.; Forslund, S.K.; Nielsen, T.; Adriouch, S.; Holmes, B.; Chilloux, J.; Vieira-Silva, S.; et al. Evidence of a Causal and Modifiable Relationship between Kidney Function and Circulating Trimethylamine N-Oxide. Nat. Commun. 2023, 14, 5843. [Google Scholar] [CrossRef] [PubMed]
  208. Zhang, Y.; Wang, Y.; Ke, B.; Du, J. TMAO: How Gut Microbiota Contributes to Heart Failure. Transl. Res. 2021, 228, 109–125. [Google Scholar] [CrossRef] [PubMed]
  209. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The Severity of Nonalcoholic Fatty Liver Disease Is Associated with Gut Dysbiosis and Shift in the Metabolic Function of the Gut Microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef] [PubMed]
  210. Kearns, R. The Kynurenine Pathway in Gut Permeability and Inflammation. Inflammation 2024, 47, 1–15. [Google Scholar] [CrossRef] [PubMed]
  211. Lee, G.; You, H.J.; Bajaj, J.S.; Joo, S.K.; Yu, J.; Park, S.; Kang, H.; Park, J.H.; Kim, J.H.; Lee, D.H.; et al. Distinct Signatures of Gut Microbiome and Metabolites Associated with Significant Fibrosis in Non-Obese NAFLD. Nat. Commun. 2020, 11, 4982. [Google Scholar] [CrossRef]
  212. Su, X.; Gao, Y.; Yang, R. Gut Microbiota-Derived Tryptophan Metabolites Maintain Gut and Systemic Homeostasis. Cells 2022, 11, 2296. [Google Scholar] [CrossRef]
  213. Long, Q.; Luo, F.; Li, B.; Li, Z.; Guo, Z.; Chen, Z.; Wu, W.; Hu, M. Gut Microbiota and Metabolic Biomarkers in Metabolic Dysfunction–Associated Steatotic Liver Disease. Hepatol. Commun. 2024, 8, e0310. [Google Scholar] [CrossRef] [PubMed]
  214. Oh, T.G.; Kim, S.M.; Caussy, C.; Fu, T.; Guo, J.; Bassirian, S.; Singh, S.; Madamba, E.V.; Bettencourt, R.; Richards, L.; et al. A Universal Gut-Microbiome-Derived Signature Predicts Cirrhosis. Cell Metab. 2020, 32, 878–888.e6. [Google Scholar] [CrossRef] [PubMed]
  215. Crost, E.H.; Coletto, E.; Bell, A.; Juge, N. Ruminococcus Gnavus: Friend or Foe for Human Health. FEMS Microbiol. Rev. 2023, 47, fuad014. [Google Scholar] [CrossRef] [PubMed]
  216. Boopathi, S.; Kumar, R.M.S.; Priya, P.S.; Haridevamuthu, B.; Nayak, S.P.R.R.; Chulenbayeva, L.; Almagul, K.; Arockiaraj, J. Gut Enterobacteriaceae and Uraemic Toxins—Perpetrators for Ageing. Exp. Gerontol. 2023, 173, 112088. [Google Scholar] [CrossRef] [PubMed]
  217. Zhai, L.; Xiao, H.; Lin, C.; Wong, H.L.X.; Lam, Y.Y.; Gong, M.; Wu, G.; Ning, Z.; Huang, C.; Zhang, Y.; et al. Gut Microbiota-Derived Tryptamine and Phenethylamine Impair Insulin Sensitivity in Metabolic Syndrome and Irritable Bowel Syndrome. Nat. Commun. 2023, 14, 4986. [Google Scholar] [CrossRef] [PubMed]
  218. Zhang, Y.; Xu, J.; Wang, X.; Ren, X.; Liu, Y. Changes of Intestinal Bacterial Microbiota in Coronary Heart Disease Complicated with Nonalcoholic Fatty Liver Disease. BMC Genom. 2019, 20, 862. [Google Scholar] [CrossRef]
  219. Vaziri, N.D.; Wong, J.; Pahl, M.; Piceno, Y.M.; Yuan, J.; Desantis, T.Z.; Ni, Z.; Nguyen, T.H.; Andersen, G.L. Chronic Kidney Disease Alters Intestinal Microbial Flora. Kidney Int. 2013, 83, 308–315. [Google Scholar] [CrossRef]
  220. Shah, N.B.; Allegretti, A.S.; Nigwekar, S.U.; Kalim, S.; Zhao, S.; Lelouvier, B.; Servant, F.; Serena, G.; Thadhani, R.I.; Raj, D.S.; et al. Blood Microbiome Profile in CKD: A Pilot Study. Clin. J. Am. Soc. Nephrol. 2019, 14, 692–701. [Google Scholar] [CrossRef]
  221. Seki, N.; Kimizuka, T.; Gondo, M.; Yamaguchi, G.; Sugiura, Y.; Akiyama, M.; Yakabe, K.; Uchiyama, J.; Higashi, S.; Haneda, T.; et al. D-Tryptophan Suppresses Enteric Pathogen and Pathobionts and Prevents Colitis by Modulating Microbial Tryptophan Metabolism. iScience 2022, 25, 104838. [Google Scholar] [CrossRef]
  222. Tang, Z.; Yu, S.; Pan, Y. The Gut Microbiome Tango in the Progression of Chronic Kidney Disease and Potential Therapeutic Strategies. J. Transl. Med. 2023, 21, 689. [Google Scholar] [CrossRef]
  223. Yang, J.Z.; Zhang, K.K.; Shen, H.W.; Liu, Y.; Li, X.W.; Chen, L.J.; Liu, J.L.; Li, J.H.; Zhao, D.; Wang, Q.; et al. Sigma-1 Receptor Knockout Disturbs Gut Microbiota, Remodels Serum Metabolome, and Exacerbates Isoprenaline-Induced Heart Failure. Front. Microbiol. 2023, 14, 1255971. [Google Scholar] [CrossRef]
  224. López-Torres, C.D.; Torres-Mena, J.E.; Castro-Gil, M.P.; Villa-Treviño, S.; Arellanes-Robledo, J.; del Pozo-Yauner, L.; Pérez-Carreón, J.I. Downregulation of Indolethylamine N-Methyltransferase Is an Early Event in the Rat Hepatocarcinogenesis and Is Associated with Poor Prognosis in Hepatocellular Carcinoma Patients. J. Gene Med. 2022, 24, e3439. [Google Scholar] [CrossRef] [PubMed]
  225. Wu, H.; Dixon, E.E.; Xuanyuan, Q.; Guo, J.; Yoshimura, Y.; Debashish, C.; Niesnerova, A.; Xu, H.; Rouault, M.; Humphreys, B.D. High Resolution Spatial Profiling of Kidney Injury and Repair Using RNA Hybridization-Based in Situ Sequencing. Nat. Commun. 2024, 15, 1396. [Google Scholar] [CrossRef] [PubMed]
  226. Nuño-Ayala, M.; Guillén, N.; Arnal, C.; Lou-Bonafonte, J.M.; de Martino, A.; García-de-Jalón, J.A.; Gascón, S.; Osaba, L.; Osada, J.; Navarro, M.A. Cystathionine β-Synthase Deficiency Causes Infertility by Impairing Decidualization and Gene Expression Networks in Uterus Implantation Sites. Physiol. Genom. 2012, 44, 702–716. [Google Scholar] [CrossRef] [PubMed]
  227. Groza, T.; Gomez Lopez, F.L.; Mashhadi, H.H.; Muñoz-Fuentes, V.; Gunes, O.; Wilson, R.; Cacheiro, P.; Frost, A.; Keskivali-Bond, P.; Vardal, B.; et al. The International Mouse Phenotyping Consortium: Comprehensive Knockout Phenotyping Underpinning the Study of Human Disease. Nucleic Acids Res. 2023, 51, D1038–D1045. [Google Scholar] [CrossRef] [PubMed]
  228. Urabe, Y.; Tanikawa, C.; Takahashi, A.; Okada, Y.; Morizono, T.; Tsunoda, T.; Kamatani, N.; Kohri, K.; Chayama, K.; Kubo, M.; et al. A Genome-Wide Association Study of Nephrolithiasis in the Japanese Population Identifies Novel Susceptible Loci at 5q35.3, 7p14.3, and 13q14.1. PLoS Genet. 2012, 8, e1002541. [Google Scholar] [CrossRef]
  229. Peto, K.; Nemeth, N.; Mester, A.; Magyar, Z.; Ghanem, S.; Somogyi, V.; Tanczos, B.; Deak, A.; Bidiga, L.; Frecska, E.; et al. Hemorheological and Metabolic Consequences of Renal Ischemia-Reperfusion and Their Modulation by N,N-Dimethyl-Tryptamine on a Rat Model. Clin. Hemorheol. Microcirc. 2018, 70, 107–117. [Google Scholar] [CrossRef]
  230. Nemes, B.; Pető, K.; Németh, N.; Mester, A.; Magyar, Z.; Ghanem, S.; Sógor, V.; Tánczos, B.; Deák, Á.; Kállay, M.; et al. N,N-Dimethyltryptamine Prevents Renal Ischemia-Reperfusion Injury in a Rat Model. Transplant. Proc. 2019, 51, 1268–1275. [Google Scholar] [CrossRef]
  231. Xu, Y.; Li, L.; Tang, P.; Zhang, J.; Zhong, R.; Luo, J.; Lin, J.; Zhang, L. Identifying Key Genes for Diabetic Kidney Disease by Bioinformatics Analysis. BMC Nephrol. 2023, 24, 305. [Google Scholar] [CrossRef] [PubMed]
  232. Balzer, M.S.; Doke, T.; Yang, Y.W.; Aldridge, D.L.; Hu, H.; Mai, H.; Mukhi, D.; Ma, Z.; Shrestha, R.; Palmer, M.B.; et al. Single-Cell Analysis Highlights Differences in Druggable Pathways Underlying Adaptive or Fibrotic Kidney Regeneration. Nat. Commun. 2022, 13, 4018. [Google Scholar] [CrossRef]
  233. Liu, J.; Kumar, S.; Dolzhenko, E.; Alvarado, G.F.; Guo, J.; Lu, C.; Chen, Y.; Li, M.; Dessing, M.C.; Parvez, R.K.; et al. Molecular Characterization of the Transition from Acute to Chronic Kidney Injury Following Ischemia/Reperfusion. JCI Insight 2017, 2, e94716. [Google Scholar] [CrossRef] [PubMed]
  234. Dean, J.G.; Liu, T.; Huff, S.; Sheler, B.; Barker, S.A.; Strassman, R.J.; Wang, M.M.; Borjigin, J. Biosynthesis and Extracellular Concentrations of N,N-Dimethyltryptamine (DMT) in Mammalian Brain. Sci. Rep. 2019, 9, 9333. [Google Scholar] [CrossRef] [PubMed]
  235. Karlsson, M.; Zhang, C.; Méar, L.; Zhong, W.; Digre, A.; Katona, B.; Sjöstedt, E.; Butler, L.; Odeberg, J.; Dusart, P.; et al. A Single–Cell Type Transcriptomics Map of Human Tissues. Sci. Adv. 2021, 7, eabh2169. [Google Scholar] [CrossRef] [PubMed]
  236. Lake, B.B.; Menon, R.; Winfree, S.; Hu, Q.; Ferreira, R.M.; Kalhor, K.; Barwinska, D.; Otto, E.A.; Ferkowicz, M.; Diep, D.; et al. An Atlas of Healthy and Injured Cell States and Niches in the Human Kidney. Nature 2023, 619, 585–594. [Google Scholar] [CrossRef]
  237. Fukumoto, Y.; Yamada, H.; Matsuhashi, K.; Okada, W.; Tanaka, Y.K.; Suzuki, N.; Ogra, Y. Production of a Urinary Selenium Metabolite, Trimethylselenonium, by Thiopurine S-Methyltransferase and Indolethylamine N-Methyltransferase. Chem. Res. Toxicol. 2020, 33, 2467–2474. [Google Scholar] [CrossRef] [PubMed]
  238. MacParland, S.A.; Liu, J.C.; Ma, X.Z.; Innes, B.T.; Bartczak, A.M.; Gage, B.K.; Manuel, J.; Khuu, N.; Echeverri, J.; Linares, I.; et al. Single Cell RNA Sequencing of Human Liver Reveals Distinct Intrahepatic Macrophage Populations. Nat. Commun. 2018, 9, 4383. [Google Scholar] [CrossRef] [PubMed]
  239. Petrenko, O.; Königshofer, P.; Brusilovskaya, K.; Hofer, B.S.; Bareiner, K.; Simbrunner, B.; Jühling, F.; Baumert, T.F.; Lupberger, J.; Trauner, M.; et al. Transcriptomic Signatures of Progressive and Regressive Liver Fibrosis and Portal Hypertension. iScience 2024, 27, 109301. [Google Scholar] [CrossRef] [PubMed]
  240. Lee, E.-H.; Oh, J.-H.; Selvaraj, S.; Park, S.-M.; Choi, M.-S.; Spanel, R.; Yoon, S.; Borlak, J.; Lee, E.-H.; Oh, J.-H.; et al. Immunogenomics Reveal Molecular Circuits of Diclofenac Induced Liver Injury in Mice. Oncotarget 2016, 7, 14983–15017. [Google Scholar] [CrossRef] [PubMed]
  241. Kyritsi, K.; Chen, L.; O’Brien, A.; Francis, H.; Hein, T.W.; Venter, J.; Wu, N.; Ceci, L.; Zhou, T.; Zawieja, D.; et al. Modulation of the Tryptophan Hydroxylase 1/Monoamine Oxidase-A/5-Hydroxytryptamine/5-Hydroxytryptamine Receptor 2A/2B/2C Axis Regulates Biliary Proliferation and Liver Fibrosis During Cholestasis. Hepatology 2020, 71, 990–1008. [Google Scholar] [CrossRef]
  242. Arroyo, N.; Villamayor, L.; Díaz, I.; Carmona, R.; Ramos-Rodríguez, M.; Muñoz-Chápuli, R.; Pasquali, L.; Toscano, M.G.; Martín, F.; Cano, D.A.; et al. GATA4 Induces Liver Fibrosis Regression by Deactivating Hepatic Stellate Cells. JCI Insight 2021, 6, e150059. [Google Scholar] [CrossRef] [PubMed]
  243. Zheng, R.; Rebolledo-Jaramillo, B.; Zong, Y.; Wang, L.; Russo, P.; Hancock, W.; Stanger, B.Z.; Hardison, R.C.; Blobel, G.A. Function of GATA Factors in the Adult Mouse Liver. PLoS ONE 2013, 8, e83723. [Google Scholar] [CrossRef] [PubMed]
  244. Llorente-Cortes, V.; Barbarigo, V.; Badimon, L. Low Density Lipoprotein Receptor-Related Protein 1 Modulates the Proliferation and Migration of Human Hepatic Stellate Cells. J. Cell Physiol. 2012, 227, 3528–3533. [Google Scholar] [CrossRef]
  245. Chen, L.; Wang, Y. Interdisciplinary Advances Reshape the Delivery Tools for Effective NASH Treatment. Mol. Metab. 2023, 73, 101730. [Google Scholar] [CrossRef] [PubMed]
  246. Hamlin, A.N.; Chinnarasu, S.; Ding, Y.; Xian, X.; Herz, J.; Jaeschke, A.; Hui, D.Y. Low-Density Lipoprotein Receptor–Related Protein-1 Dysfunction Synergizes with Dietary Cholesterol to Accelerate Steatohepatitis Progression. J. Biol. Chem. 2018, 293, 9674. [Google Scholar] [CrossRef] [PubMed]
  247. Jiang, T.; Wang, L.; Li, X.; Song, J.; Wu, X.; Zhou, S. Inositol-Requiring Enzyme 1-Mediated Endoplasmic Reticulum Stress Triggers Apoptosis and Fibrosis Formation in Liver Cirrhosis Rat Models. Mol. Med. Rep. 2015, 11, 2941–2946. [Google Scholar] [CrossRef]
  248. Watson, J.D.; Prokopec, S.D.; Smith, A.B.; Okey, A.B.; Pohjanvirta, R.; Boutros, P.C. TCDD Dysregulation of 13 AHR-Target Genes in Rat Liver. Toxicol. Appl. Pharmacol. 2014, 274, 445–454. [Google Scholar] [CrossRef]
  249. Kozaczek, M.; Bottje, W.; Greene, E.; Lassiter, K.; Kong, B.; Dridi, S.; Korourian, S.; Hakkak, R. Comparison of Liver Gene Expression by RNAseq and PCR Analysis after 8 Weeks of Feeding Soy Protein Isolate- or Casein-Based Diets in an Obese Liver Steatosis Rat Model. Food Funct. 2019, 10, 8218–8229. [Google Scholar] [CrossRef]
  250. Huang, Y.; Wang, X.; Yan, C.; Li, C.; Zhang, L.; Zhang, L.; Liang, E.; Liu, T.; Mao, J. Effect of Metformin on Nonalcoholic Fatty Liver Based on Meta-Analysis and Network Pharmacology. Medicine 2022, 101, E31437. [Google Scholar] [CrossRef]
  251. Xu, Y.; Wang, L.; Butticè, G.; Sengupta, P.K.; Smith, B.D. Major Histocompatibility Class II Transactivator (CIITA) Mediates Repression of Collagen (COL1A2) Transcription by Interferon γ (IFN-γ). J. Biol. Chem. 2004, 279, 41319–41332. [Google Scholar] [CrossRef]
  252. Winau, F.; Hegasy, G.; Weiskirchen, R.; Weber, S.; Cassan, C.; Sieling, P.A.; Modlin, R.L.; Liblau, R.S.; Gressner, A.M.; Kaufmann, S.H.E. Ito Cells Are Liver-Resident Antigen-Presenting Cells for Activating T Cell Responses. Immunity 2007, 26, 117–129. [Google Scholar] [CrossRef] [PubMed]
  253. Han, X.; Gong, N.; Xue, L.; Billingsley, M.M.; El-Mayta, R.; Shepherd, S.J.; Alameh, M.G.; Weissman, D.; Mitchell, M.J. Ligand-Tethered Lipid Nanoparticles for Targeted RNA Delivery to Treat Liver Fibrosis. Nat. Commun. 2023, 14, 75. [Google Scholar] [CrossRef] [PubMed]
  254. Tsurusaki, S.; Tsuchiya, Y.; Koumura, T.; Nakasone, M.; Sakamoto, T.; Matsuoka, M.; Imai, H.; Yuet-Yin Kok, C.; Okochi, H.; Nakano, H.; et al. Hepatic Ferroptosis Plays an Important Role as the Trigger for Initiating Inflammation in Nonalcoholic Steatohepatitis. Cell Death Dis. 2019, 10, 449. [Google Scholar] [CrossRef] [PubMed]
  255. Lai, W.; Wang, B.; Huang, R.; Zhang, C.; Fu, P.; Ma, L. Ferroptosis in Organ Fibrosis: From Mechanisms to Therapeutic Medicines. J. Transl. Int. Med. 2024, 12, 22. [Google Scholar] [CrossRef]
  256. Cho, S.S.; Yang, J.H.; Lee, J.H.; Baek, J.S.; Ku, S.K.; Cho, I.J.; Kim, K.M.; Ki, S.H. Ferroptosis Contribute to Hepatic Stellate Cell Activation and Liver Fibrogenesis. Free Radic. Biol. Med. 2022, 193, 620–637. [Google Scholar] [CrossRef] [PubMed]
  257. 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. Alzheim. Res. Ther. 2024, 16, 95. [Google Scholar] [CrossRef] [PubMed]
  258. D’Ambrosi, N.; Apolloni, S. Fibrotic Scar in Neurodegenerative Diseases. Front. Immunol. 2020, 11, 1394. [Google Scholar] [CrossRef] [PubMed]
  259. Judd, J.M.; Jasbi, P.; Winslow, W.; Serrano, G.E.; Beach, T.G.; Klein-Seetharaman, J.; Velazquez, R. Inflammation and the Pathological Progression of Alzheimer’s Disease Are Associated with Low Circulating Choline Levels. Acta Neuropathol. 2023, 146, 565. [Google Scholar] [CrossRef]
  260. Szegeczki, V.; Perényi, H.; Horváth, G.; Hinnah, B.; Tamás, A.; Radák, Z.; Ábrahám, D.; Zákány, R.; Reglodi, D.; Juhász, T. Physical Training Inhibits the Fibrosis Formation in Alzheimer’s Disease Kidney Influencing the TGFβ Signaling Pathways. J. Alzheimers Dis. 2021, 81, 1195. [Google Scholar] [CrossRef]
  261. Vallianou, N.G.; Kounatidis, D.; Psallida, S.; Panagopoulos, F.; Stratigou, T.; Geladari, E.; Karampela, I.; Tsilingiris, D.; Dalamaga, M. The Interplay Between Dietary Choline and Cardiometabolic Disorders: A Review of Current Evidence. Curr. Nutr. Rep. 2024, 13, 152–165. [Google Scholar] [CrossRef]
  262. Strupp, B.J.; Powers, B.E.; Velazquez, R.; Ash, J.A.; Kelley, C.M.; Alldred, M.J.; Strawderman, M.; Caudill, M.A.; Mufson, E.J.; Ginsberg, S.D. Maternal Choline Supplementation: A Potential Prenatal Treatment for Down Syndrome and Alzheimer’s Disease. Curr. Alzheimer Res. 2015, 13, 97–106. [Google Scholar] [CrossRef]
  263. 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]
  264. Bai, T.; Lei, P.; Zhou, H.; Liang, R.; Zhu, R.; Wang, W.; Zhou, L.; Sun, Y. Sigma-1 Receptor Protects against Ferroptosis in Hepatocellular Carcinoma Cells. J. Cell Mol. Med. 2019, 23, 7349–7359. [Google Scholar] [CrossRef] [PubMed]
  265. Sun, H.; Long, J.; Zuo, B.; Li, Y.; Song, Y.; Yu, M.; Xun, Z.; Wang, Y.; Wang, X.; Sang, X.; et al. Development and Validation of a Selenium Metabolism Regulators Associated Prognostic Model for Hepatocellular Carcinoma. BMC Cancer 2023, 23, 451. [Google Scholar] [CrossRef] [PubMed]
  266. Sonet, J.; Bulteau, A.L.; Touat-hamici, Z.; Mosca, M.; Bierla, K.; Mounicou, S.; Lobinski, R.; Chavatte, L. Selenoproteome Expression Studied by Non-Radioactive Isotopic Selenium-Labeling in Human Cell Lines. Int. J. Mol. Sci. 2021, 22, 7308. [Google Scholar] [CrossRef]
  267. Jianfeng, W.; Yutao, W.; Jianbin, B. Indolethylamine-N-Methyltransferase Inhibits Proliferation and Promotes Apoptosis of Human Prostate Cancer Cells: A Mechanistic Exploration. Front. Cell Dev. Biol. 2022, 10, 805402. [Google Scholar] [CrossRef]
  268. Zhou, X.; Zou, B.; Wang, J.; Wu, L.; Tan, Q.; Ji, C. Low Expression of INMT Is Associated with Poor Prognosis but Favorable Immunotherapy Response in Lung Adenocarcinoma. Front. Genet. 2022, 13, 946848. [Google Scholar] [CrossRef]
Figure 1. (A) Tryptamine Structure. Tryptamine is a heterocyclic indole derivative with an ethylamine at the C3 position. (A,B) Tryptamine Metabolism. Tryptophan (i) is decarboxylated by AADC to form (ii), the precursor to DMT. Serotonin (iii) may be methylated by INMT as well as 5-MeO-tryptamine. R = H-, HO- or MeO- groups. (C) Trimethylselenonium production. Abbreviations: aromatic L-amine decarboxylase (AADC); aldehyde dehydrogenase, (ALDH); N-acetylserotonin O-methyltransferase, (ASMT); indoleamine 2,3-dioxygenase, (IDO); indolethylamine-N-methyltransferase, (INMT); monoamine oxidase A, (MAO-A); S-adenosylhomocysteine, (SAH); S-adenosylmethionine, (SAM); tryptophan 2,3-dioxygenase, (TDO); tryptophan hydroxylase, (TPH); (i) = tryptophan; (ii) = tryptamine; (iii) = serotonin; (iv) = 5-methoxy-tryptamine; (v′) = N-methyltryptamine or derivative; (vi′) = N,N-methyltryptamine or derivative; (vii′) N,N,N-trimethyltryptamine or derivative. (gray) = possible metabolite. Figure were generated using ChemDraw v22.2.0.
Figure 1. (A) Tryptamine Structure. Tryptamine is a heterocyclic indole derivative with an ethylamine at the C3 position. (A,B) Tryptamine Metabolism. Tryptophan (i) is decarboxylated by AADC to form (ii), the precursor to DMT. Serotonin (iii) may be methylated by INMT as well as 5-MeO-tryptamine. R = H-, HO- or MeO- groups. (C) Trimethylselenonium production. Abbreviations: aromatic L-amine decarboxylase (AADC); aldehyde dehydrogenase, (ALDH); N-acetylserotonin O-methyltransferase, (ASMT); indoleamine 2,3-dioxygenase, (IDO); indolethylamine-N-methyltransferase, (INMT); monoamine oxidase A, (MAO-A); S-adenosylhomocysteine, (SAH); S-adenosylmethionine, (SAM); tryptophan 2,3-dioxygenase, (TDO); tryptophan hydroxylase, (TPH); (i) = tryptophan; (ii) = tryptamine; (iii) = serotonin; (iv) = 5-methoxy-tryptamine; (v′) = N-methyltryptamine or derivative; (vi′) = N,N-methyltryptamine or derivative; (vii′) N,N,N-trimethyltryptamine or derivative. (gray) = possible metabolite. Figure were generated using ChemDraw v22.2.0.
Livers 04 00043 g001
Figure 2. INMT’s potential in mediating fibrogenic diseases through N-methylated tryptamines and selenium metabolism. Red arrows denote insults that promote fibrosis. Green arrows denote resolving of fibrosis through SIGMAR1. PDB: 2A14. Abbreviations: endothelin-1, (ET-1); hypoxia-inducible factor 1-alpha, (HIF1α); indolethylamine-N-methyltransferase, (INMT); reactive oxygen species, (ROS); sigma non-opioid intracellular receptor 1, (SIGMAR1); transforming growth factor beta, (TGFβ). Figure were generated using BioRender.
Figure 2. INMT’s potential in mediating fibrogenic diseases through N-methylated tryptamines and selenium metabolism. Red arrows denote insults that promote fibrosis. Green arrows denote resolving of fibrosis through SIGMAR1. PDB: 2A14. Abbreviations: endothelin-1, (ET-1); hypoxia-inducible factor 1-alpha, (HIF1α); indolethylamine-N-methyltransferase, (INMT); reactive oxygen species, (ROS); sigma non-opioid intracellular receptor 1, (SIGMAR1); transforming growth factor beta, (TGFβ). Figure were generated using BioRender.
Livers 04 00043 g002
Table 1. Receptor interactions among N-dimethylated tryptamines.
Table 1. Receptor interactions among N-dimethylated tryptamines.
Receptor DMT5-HO-DMT5-MeO-DMT
5-HTR1AAgonistAgonistAgonist
5-HTR1BPartial AgonistPartial AgonistPartial Agonist
5-HTR1DWeak AgonistUnknownWeak Agonist
5-HTR2AAgonistAgonistAgonist
5-HTR2BAgonistAgonistAgonist
5-HTR2CAgonistAgonistAgonist
5-HTR7AgonistUnknownAgonist
SIGMAR1AgonistAgonistAgonist
TAAR1AgonistUnknownAgonist
TAAR6Predicted AgonistUnknownPredicted Agonist
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

Korsmo, H.W. Exploring Endogenous Tryptamines: Overlooked Agents Against Fibrosis in Chronic Disease? A Narrative Review. Livers 2024, 4, 615-637. https://doi.org/10.3390/livers4040043

AMA Style

Korsmo HW. Exploring Endogenous Tryptamines: Overlooked Agents Against Fibrosis in Chronic Disease? A Narrative Review. Livers. 2024; 4(4):615-637. https://doi.org/10.3390/livers4040043

Chicago/Turabian Style

Korsmo, Hunter W. 2024. "Exploring Endogenous Tryptamines: Overlooked Agents Against Fibrosis in Chronic Disease? A Narrative Review" Livers 4, no. 4: 615-637. https://doi.org/10.3390/livers4040043

APA Style

Korsmo, H. W. (2024). Exploring Endogenous Tryptamines: Overlooked Agents Against Fibrosis in Chronic Disease? A Narrative Review. Livers, 4(4), 615-637. https://doi.org/10.3390/livers4040043

Article Metrics

Back to TopTop