Next Article in Journal
Stress-Activated Protein Kinases in Intervertebral Disc Degeneration: Unraveling the Impact of JNK and p38 MAPK
Previous Article in Journal
DPP Inhibition Enhances the Efficacy of PD-1 Blockade by Remodeling the Tumor Microenvironment in Lewis Lung Carcinoma Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inflammation and Organic Cation Transporters Novel (OCTNs)

by
Lorena Pochini
1,2,*,
Michele Galluccio
1,
Lara Console
1,
Mariafrancesca Scalise
1,
Ivano Eberini
3 and
Cesare Indiveri
1,2,*
1
Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy
2
Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
3
Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
*
Authors to whom correspondence should be addressed.
Biomolecules 2024, 14(4), 392; https://doi.org/10.3390/biom14040392
Submission received: 15 February 2024 / Revised: 20 March 2024 / Accepted: 21 March 2024 / Published: 25 March 2024

Abstract

:
Inflammation is a physiological condition characterized by a complex interplay between different cells handled by metabolites and specific inflammatory-related molecules. In some pathological situations, inflammation persists underlying and worsening the pathological state. Over the years, two membrane transporters namely OCTN1 (SLC22A4) and OCTN2 (SLC22A5) have been shown to play specific roles in inflammation. These transporters form the OCTN subfamily within the larger SLC22 family. The link between these proteins and inflammation has been proposed based on their link to some chronic inflammatory diseases such as asthma, Crohn’s disease (CD), and rheumatoid arthritis (RA). Moreover, the two transporters show the ability to mediate the transport of several compounds including carnitine, carnitine derivatives, acetylcholine, ergothioneine, and gut microbiota by-products, which have been specifically associated with inflammation for their anti- or proinflammatory action. Therefore, the absorption and distribution of these molecules rely on the presence of OCTN1 and OCTN2, whose expression is modulated by inflammatory cytokines and transcription factors typically activated by inflammation. In the present review, we wish to provide a state of the art on OCTN1 and OCTN2 transport function and regulation in relationships with inflammation and inflammatory diseases focusing on the metabolic signature collected in different body districts and gene polymorphisms related to inflammatory diseases.

1. Introduction

Inflammation is a multistep and complex physiological reaction of the host to infections and damage. The purpose of inflammatory responses is to restore body homeostasis even though prolonged and unresolved inflammatory states are responsible for a plethora of pathological conditions with a broad range of severity. In the last few years, an interesting link between inflammation and metabolism has been suggested due to the increased need for nutrients from inflammatory cells [1]. In this scenario, a close relationship between inflammatory processes and membrane transporters has been described, in line with the role of these proteins in allowing for the flux of nutrients across tissues. Another relevant issue is the traffic of compounds that are produced or whose levels change during the inflammatory processes. Therefore, membrane transporters which can interact with or transport these inflammation-related compounds represent interesting targets for therapy in a wide range of diseases directly or indirectly related to inflammation. Among transporters exhibiting the property of interacting with many different compounds, there are the human Organic Cation Transporters Novel OCTN1 (SLC22A4) and OCTN2 (SLC22A5). These two proteins correspond to the A4 and A5 members of the Solute Carrier (SLC) family 22, which includes a large number of cations, zwitterions, and anion transporters. The cation and zwitterion transporter members share significant structural features [2,3]. OCTN1 and OCTN2 constitute a small subgroup sharing more than 76% identity to each other, which would indicate similar, or at least related, cellular roles and/or biochemical functions [4]. Notwithstanding, differences in the function of the two human transporters emerged from many studies performed in the last two decades [5,6]. Ergothioneine and acetylcholine have been identified as the major OCTN1 substrates, whereas carnitine and carnitine derivatives have been identified as the major substrates for OCTN2 [6,7,8,9,10,11,12,13,14]. Interestingly, ergothioneine was also reported to be transported by OCTN2 [15], and carnitine was reported as a low-affinity OCTN1 substrate more than 20 years ago [16]. Very recently, this finding has been confirmed by in vitro experiments showing that OCTN1 catalyzes a sodium-dependent carnitine transport [17]. Moreover, similarly to the OCT members of the SLC22 family [18,19], OCTN1 and 2 share polispecificity towards ligands including molecules related to the inflammatory processes (Figure 1).
Based on metabolomics, it has been found that small molecules that are enriched in an inflamed status in plasma, urine, synovial fluid, feces, or different tissues are typical ligands of SLC22 members and, especially, of OCTNs [18,19,22,23].
In particular, the transport of carnitine, acetylcarnitine, and ergothioneine mediated by OCTN1 occurs mainly by an uptake mode, whereas the transport of acetylcholine occurs by an efflux mode. In the case of the OCTN2 transport of carnitine or its analogs, it occurs by an uptake mode.
Circulating metabolites are conditioned by many factors, such as the diet–microbiome axis as well as by the activity of many enzymes and transporters. Indeed, metabolic and immune signals of the microbiota can enter circulation exploiting the activity of intestinal membrane transporters. In this context, OCTN1 and OCTN2 may also affect the level of these metabolites which are known (or still unknown) substrates of these transporters; moreover, the inflammatory processes may act directly or indirectly on the expression of OCTN1 and OCTN2 [24], thus influencing the level of the metabolites. In good agreement, altered levels of metabolites, which are known or possibly unknown OCTN substrates, have been found in the case of chronic inflammatory diseases, such as Inflammatory Bowel Diseases (IBDs) [23]. The similarity of the AlphaFold structures of the two proteins, shown in Figure 2A, correlates well with the sequence similarity between OCTN1 and OCTN2 (Figure 2B) and also with the Cryo-EM 3D structures of OCT1, 2, and 3 sharing from 31% to 35% identity with the OCTNs [25,26]. The predicted OCTN1-2 3D structures highlight the presence of a large extracellular loop with potential N-glycosylation sites. Moreover, the OCTNs share the presence of an intracellular nucleotide-binding domain which is different from the NBDs of the ABC transporters [27,28,29,30]. This is in line with the described regulation of OCTN1 by intracellular [31] or intraliposomal ATP [9,10,11,32].
The external loops of OCTN1 and OCTN2 contain four cysteines arranged in two couples of vicinal residues that can form disulfides (Figure 2).
This feature makes OCTNs sensitive to thiol reagents. As an example, toxic mercury derivatives can modify the function of these transporters at concentrations close to those found in contaminated environments [33,34]. In the case of OCTN1, the Cys residues responsible for the response to thiol-reactive compounds, such as mercury compounds, cysteine, N-acetylcysteine, and carboxymethyl cysteine, have been identified by site-directed mutagenesis [33,34]. It is then likely that OCTNs may interact with physiological thiol-reacting compounds such as Reactive Oxygen Species (ROS), thus responding to redox signaling [35]. These features correlate well with the above-mentioned involvement of the OCTNs in inflammatory processes [36,37,38,39], characterized, among other factors, by excess ROS formation [40]. OCTN1 and OCTN2 also share localization in tissues such as the lungs, gut epithelia, and immune cells, directly or indirectly involved in inflammatory processes [5,6,21,41,42,43,44,45]. Indeed, the frontier epithelia are exposed to microorganisms, inhaled drugs, cigarette smoke, and other pollutants that may give rise to inflammation [21,44,46]. OCTN1 also regulates the activation of microglia which are macrophages resident in the central nervous system and responsible for initiating innate immune responses to a variety of different pathogens. Microglial cells activate and migrate to the damaged regions, where the production of inflammatory cytokines occurs (IL-1β and TNFα, ROS, and neurotrophic factors). The chronic over-activation of microglia damages neurons as well, resulting in the onset of neurodegenerative disorders, such as Alzheimer’s and Parkinson’s diseases [47].
All the described OCTN features strongly support a role in inflammatory diseases and host defense mechanisms. Indeed, several reports indicate the involvement of OCTN1 and 2 in asthma, CD, and RA [5,6,45,48]. However, the molecular bases of the described link are still poorly defined. OCTNs could exert a role in the intestinal absorption of inflammation-linked metabolites, their distribution to tissues, and kidney excretion, also playing a role in cell communication processes. Interestingly, OCTN2 has been found as novel exosome cargo whose level in these endosomal-derived nano-vesicles is regulated by the proinflammatory cytokine INF-γ [24]. In this review, we will provide an overview of the relationships between inflammatory processes and OCTNs.

2. OCTN Functions and Dysfunctions

2.1. Relationships between Functions and Diseases

OCTN2 has a well-recognized role as a sodium-dependent carnitine transporter in most tissues. This function is fundamental for the accomplishment of the mitochondrial β-oxidation pathway that has an essential requirement for carnitine. In this frame, OCTN2 plays two important roles: (i) mediating carnitine absorption from diet, accounting for more than half of the carnitine body pool to compensate for the limited biosynthesis; (ii) distributing carnitine to most tissues that cannot synthesize it. Besides the well-established role of carnitine in the mitochondrial carnitine shuttle, other functions have also been demonstrated or proposed [49,50,51,52,53]. In the case of OCTN1, the major substrates are ergothioneine and acetylcholine, which are also related to antioxidant and/or anti-inflammatory effects. Ergothioneine is a mushroom metabolite known for its antioxidant activity [54]. Acetylcholine, differently from ergothioneine, is a physiological compound. Interestingly, acetylcholine, besides the well-known neurotransmitter function, also plays an anti-inflammatory role [55,56,57]. Acetylcholine is a ubiquitous signaling molecule produced by numerous non-neuronal cells that possess the same acetylcholine biosynthesis pathway as neurons [9,58,59]. The only difference is that non-neuronal cells do not excrete the neurotransmitter by a quantal (vesicular) mechanism but by a slow efflux mediated by transporters of the SLC22 family, among which is OCTN1 [10,60]. It has been known for 20 years that OCTN1 can transport carnitine as well, although with an affinity about two orders of magnitude lower than that of OCTN2. Very recently, this OCTN1 feature has been better clarified, confirming that OCTN1 mediates carnitine transport with a similar mechanism to OCTN2 [17]. However, the molecular basis of the difference in affinity is still a matter of investigation. Interestingly, some physio-pathological observations correlate well with the described similarities and differences of the two proteins with respect to carnitine. The defects of OCTN2 are causative of Primary Carnitine Deficiency (PCD), a severe syndrome characterized by progressive skeletal muscle weakness and cardiomyopathy. During infancy, it may cause hypoketotic hypoglycemia and Reye syndrome, characterized by encephalopathy, hyperammonia, and sudden infant death [6,61,62]. However, PCD is not lethal, especially if treated with carnitine administration, differently from Secondary Carnitine Deficiency, caused by defects of the mitochondrial carnitine transporter [63]. The reason for the lack of lethality is the presence of alternative low-affinity carnitine absorption pathways. Therefore, owing to the overlapping localization of OCTN1 and OCTN2 and the ability of OCTN1 to transport carnitine, it is not trivial to assume that OCTN1 represents the main alternative pathway. Moreover, the involvement of OCTN2, and partially of OCTN1, in the fatty acid β-oxidation, constitutes a link between the two transporters with ROS. Indeed, it is well known that fatty acid catabolism contributes to ROS formation, and in the case of an excess of these reactive species, such as ischemia–reperfusion, damaged tissues benefit from the inhibition of fatty acid catabolism, as an example, by mildronate, a strong inhibitor of OCTN2 [64]. In line with the apparent accessory role of OCTN1 in fatty acid catabolism, the knockout of OCTN1, at least in mice, does not show any phenotype even though it exhibited greater susceptibility to intestinal inflammation under the ischemia and reperfusion model [65]. Differently from OCTN2, mutations of OCTN1 associated with a loss of function in humans are not known, and we cannot ensure that a hypothetical knocking out in humans would also not show any phenotype. Indeed, the mouse model is not considered a suitable model for humans anymore [6], considering the presence of a further transporter of the subfamily (OCTN3) that is absent in humans [5,16].

2.2. OCTN Polymorphisms and Relationships with Pathologies

So far, some OCTN polymorphisms have been described which are associated with pathologies characterized by inflammation status (Supplementary Table S1). rs1050152 is the most well described OCTN1 polymorphism, leading to L503F amino acid substitution. This is associated with CD, an IBD that causes chronic inflammation of the gastrointestinal tract. Both OCTN genes, SLC22A4 and SLC22A5, are located within the IBD5 locus on chromosome 5. This region is implicated in susceptibility to IBDs, and OCTN variants have been associated with IBDs [48,66] (Supplementary Table S1). An SNP located in a Runt-related transcription factor 1 (RUNX1)-binding sequence in SLC22A4 has been found to affect the expression of OCTN1 by altering RUNX1 binding affinity [37]. The number of variants described for the SLC22A5 gene in the Variant Viewer tool of the UniProt database (which includes data deriving from different genome databases and bioinformatics resources among which are gnomAD, dbSNP, and TOPMed) is remarkably higher, counting 685 missense variations, involving 413 out of 557 amino acids (74%). Among these protein variants, 145 are pathogenic, most of which, 103, are associated with renal carnitine transport defects, and the other 38 are referred to as likely pathogenic. Among the pathogenic variants, 73 protein variants cause PCD with the loss of carnitine transport. The number of variants described in the Variant Viewer tool for the SLC22A4 gene counts 422 missense variations. These variations result in 298 out of the 557 amino acids of the OCTN1 protein sequence (54%) which have been found to be mutated. Supplementary Table S1 reports all the references to the SNPs.

3. OCTNs’ Role in Inflammation

3.1. Involvement of OCTN Substrates in Inflammatory Processes

As already mentioned, a role in the inflammatory process has been reported for the major substrates of OCTNs [8,9,10,11,12,13,67,68].
Indeed, acetylcholine is known to regulate the expression of inflammatory cytokines in microglial cells [69]. Moreover, acetylcholine is involved in controlling inflammation via the non-neuronal cholinergic system by acting on the alpha 7 nicotinic receptors. Interestingly, OCTN1 can mediate a low-rate acetylcholine efflux from cells [11], which may be the basis of this physiological role.
Another OCTN substrate, ergothioneine, may be involved in the suppression of the inflammatory cytokine IL-1β expression. This effect is based on the antioxidant property of ergothioneine [47,70].
The common OCTN substrate, carnitine, has been acknowledged for its involvement in inflammation [71]. This became clear by observing the juvenile visceral steatosis (OCTN2−/−) mouse, which is a model of systemic carnitine deficiency. These mice developed intestinal villous atrophy, inflammation, ulcer formation, and gut perforation [72]. In these mice, the downregulation of the TGF-beta/BMP pathway has been observed [73]. Subsequently, other evidence confirmed the anti-inflammatory, immunosuppressive, and therapeutic properties of carnitine [74]. Carnitine proved to be effective in suppressing lipopolysaccharides (LPS)-induced cytokine production and improving murine survival rates during cachexia and septic shock. This substrate also exerts an inhibitory effect on inducible nitric oxide synthase (iNOS) and, hence, on nitric oxide (NO) production, reducing inflammation and histological damage in murine trinitrobenzene sulphonic acid-induced colitis [52,75]. Carnitine involvement in inflammation could also be correlated to the metabolic reprogramming from glucose to fatty acid oxidation, occurring during inflammatory responses [76,77]. Indeed, carnitine would play a critical role in the activation of M2 macrophages and inflammasome activation in M1 macrophages. However, the role of fatty acid oxidation in macrophage polarization (towards M1 or M2) and the molecular mechanism behind this process are still controversial [77]. Interestingly, the activation of the oxidative program in M2 is mediated by the activation of the Peroxisome proliferator-activated receptor (PPAR) γ, the transcription factor involved in regulating lipid metabolism, energy homeostasis, and OCTN transcription (see Section 4).
Anti-inflammatory effects of the carnitine derivatives are also reported. Acetyl-L-carnitine exerts a role in LPS-induced neuroinflammation in rats, by targeting the TLR4/ nuclear factor kappa-light-chain-enhancer of the activated B cells (NFκB) pathway [78]. In a valproate model of autism, chronic treatment with acetyl-carnitine alleviated behavioral abnormalities through different mechanisms including the recovery of inflammation in the brain [79]. OCTN2 expression in the blood–brain barrier (BBB) [80] would facilitate the permeation of carnitine and acetyl-carnitine to the brain tissue [81]. This makes carnitine and carnitine derivatives therapeutic candidates for all those neurological diseases in which inflammation represents a hallmark, such as Parkinson’s and Alzheimer’s diseases. The effects of carnitine supplementation on inflammatory markers have been reported and investigated in randomized controlled trials as well [82,83]. Experiments performed in atherosclerotic rats revealed a decrease in the level of mRNA and protein of CPR, TNF-a, IL-1b, and iNOS in the aorta and heart tissues as a consequence of acetyl-carnitine treatment [84]. The supplementation of propionyl-carnitine has been found to improve clinical response in ulcerative colitis (UC) [85,86].
The metabolism of choline, another substrate/ligand of OCTN1 [11], has been linked to the control of NLRP3 inflammasome-dependent inflammation [87]. Moreover, it has been observed that in the serum of two rodent models of Alzheimer’s disease, the levels of circulating choline were reduced, while proinflammatory cytokine TNFα was elevated [88].
In methionine-choline deficiency (MCD)-induced Non-Alcoholic Fatty Liver Disease (NAFLD), the OCTN2 substrate betaine [89] might reduce liver inflammation and damage, at least partly, by improving the balance between proinflammatory (TNF, IL-6) and anti-inflammatory (IL-10, TGF-β) cytokines [90].
Taken together, the reported data highlight an overall anti-inflammatory effect mediated by OCTN substrates.

3.2. OCTN Substrates and Gut Microbiota Communication/Interconnections

In a frontier district, i.e., facing the external environment, such as the gut, many of the OCTN2 and OCTN1 substrates may encounter a different fate: indeed, in gut lumen, microbiota may compete with the intestine epithelium for the absorption of the OCTN substrates [91,92]. The ammonium groups of choline, carnitine, ergothioneine, and betaine are converted into trimethylamine (TMA) by the gut microbiota [93]. TMA is absorbed through the intestine and oxidized by the liver enzymes monooxygenases in trimethylamine oxide (TMAO) [94]. Growing experimental evidence in animal models demonstrates the contribution of TMAO to inflammation [95], via the transcription factor NF-κB, which, in turn, triggers cytokine production [96]. Therefore, in this pathway, intestinal carnitine would have a role in TMAO production and inflammation. However, it has to be stressed that the absorption of carnitine by the intestine is a fast and efficient process due to the concentrative mechanism of transport resulting from the cooperation of OCTN2 and OCTN1 above-described in terms of carnitine transport [17]. Altogether, these observations indicate that most of the carnitine derived from a normal diet could be absorbed by the intestine. We then hypothesize that the possible indirect proinflammatory role of carnitine and/or choline could occur only under conditions of over-administration or defects of intestinal absorption, i.e., some pathological states [97]. This could in part explain the TMAO-mediated cardiomyopathy exacerbation in PCD, which is caused by mutations of OCTN2 (see Supplementary Table S1). Furthermore, carnitine would mediate the cross-talk between the microbiota and intestinal epithelium; a microbial metabolite, butyrate, is the primary metabolic fuel of the colonic epithelial cells: its oxidation, to which carnitine contributes, would provide colonocytes with 70% energy [75].
Other OCTN substrates contribute to the cross-talk between microbiota and intestinal cells. The intestinal microbiota adopts cholinergic metabolism as well [98]. In agreement, the release of epithelial acetylcholine, which is a substrate of OCTN1, is stimulated when the short-chain fatty acid (SCFA) receptor GPR41 (FFA3) and/or GPR43 (FFA2) bind propionate, another microbial metabolite produced during the fermentation of carbohydrates in the lumen of the large intestine. Microbes create total luminal concentrations of SCFA in the range of 100 mM by the fermentation of carbohydrates. Besides acetylcholine, the colonic mucosa can produce atypical choline esters such as propionylcholine and butyrylcholine due to the relative unspecificity of the choline acetyltransferase (ChAT) towards the SCFA which is esterified with choline; these choline derivatives may also be substrates of OCTN1, as it is known for choline. These atypical esters act on cholinergic epithelial receptors, too, but with a much lower affinity, so they are thought to modify epithelial cholinergic signaling by the desensitization of cholinergic receptors against acetylcholine. Differences in the SCFA profile should, therefore, affect the efficiency of the non-neuronal system with implications for the communication between intestinal microbiota and the mammalian host, modulating immunity and inflammation. In line with these observations, bacteria fermenting fibers and producing SCFAs are reduced in the mucosa and feces of patients with IBD [99]. Based on the described experimental evidence, it is clear that OCTN1 and OCTN2 are important players in the biochemical communication between microbiota and epithelial cells through the traffic of their substrates.

4. Regulation of OCTN Expression and Inflammation

4.1. Major Players of OCTN Regulation

SLC22A4 and SLC22A5 are target genes of the PPAR family. The members of the PPAR family (PPARα, PPARγ, and PPARβ/δ) are transcription factors involved in the regulation of lipid metabolism and energy homeostasis [100,101]. Among the plethora of functions controlled by these receptors, evidence suggests that all three PPAR subtypes play a significant role in controlling inflammatory responses [102] and in IBD [103]. A ligand-activated PPAR assembles into a complex with retinoid X receptor α (RXRα) and, as such, translocates to the nucleus. The anti-inflammatory efficacy of PPAR ligands is based on the PPAR/RXR-mediated blockade of the nuclear translocation of NF-kB, resulting in the transcriptional blockage of inflammatory cytokines, chemokines, and other stress response elements, such as cyclooxygenase-2 (COX2) and iNOS [103]. PPARs can control gene expression by associating with activator proteins or by binding Peroxisome Proliferator Responsive Elements (PPREs) on DNA. In the case of OCTN2, PPREs have been found within the promoter [104,105]. Moreover, the estrogen receptor is another regulator of OCTN2 expression [14] due to the presence of an estrogen receptor-responsive element in the SLC22A5 first intron [106]. Further transcriptional regulation involves the binding of a heat–shock element to the SLC22A5 promoter [36]. Unlike the above-described mechanisms, the methylation of the SLC22A5 promoter is responsible for its transcriptional downregulation [107]. The same epigenetic regulation has been observed as responsible for reduced OCTN1 expression [108]. The OCTN1 promoter region includes several other consensus recognition sites for ubiquitously expressed transcription factors, such as Sp1, RUNX1, and NF-kB [109]. Sp1 would be involved in the regulation of the tissue-specific expression of OCTN1 and not in its basal transcriptional regulation. RUNX1 has been found to be associated with OCTN1 transcriptional regulation: it functions both to activate and to repress transcription through interactions with cofactors [37] and is not associated with the basal promoter activity of OCTN1 [109]. Human fibroblasts derived from RA patients have been employed to investigate the regulation of SLC22A4 and SLC22A5 expression in the context of inflammation. In these cells, OCTN1 has been identified as a susceptibility gene for RA being regulated by RUNX1 [37]. NF-kB activation was observed when an agonist of the Toll-like receptor 3 (TLR3), polyinosinic–polycytidylic acid (poly I:C), was used to stimulate viral inflammation in rats. In this context, the downregulation of mRNA levels in Octn1 and Octn2 was found [110]. Concerning OCTN2, its mRNA was affected by neither IL-1beta nor TNFα; levels of OCTN1 mRNA were increased by stimulation with TNFα. IL-1beta nor TNFα are associated with OCTN1 transcriptional regulation via the NF-kB signaling cascade: the over-expression of NF-kB would activate the promoter activity of OCTN1. The increased OCTN1 level in inflammatory conditions would explain the increased uptake of the tyrosine kinase inhibitor, saracatinib, considering that hOCTN1 was identified as the main transport system for the accumulation of this TKI in RASF [111]. Concerning OCTN2, the involvement of NF-κB in the TNFα stimulation of the OCTN2 gene expression has been demonstrated in the Madin-Darby bovine kidney (MDBK) cell line [112].

4.2. OCTN Regulation in Immune System

Leukocytes (monocytes), intestinal immune cells, alveolar macrophages, and microglia are some of the immune cell types where higher OCTN expression levels have been found [20,37,47,68]. As a result of stimuli from pathogens, monocytes are recruited from the circulation and differentiate into macrophages (macrophage differentiation). OCTNs emerge as novel markers of this process (Figure 3).
Indeed, undifferentiated monocytes express high protein levels of OCTN1 that dramatically drop along with the differentiation into macrophages; the OCTN2 protein, undetectable in monocytes, became strongly expressed in monocyte-derived macrophages, with a 20-fold increase in the corresponding mRNA [74]. This process involves changes in gene expression driven by multiple transcription factors, among which are those related to the PPAR and STAT (signal transducers and activators of transcription) families. Monocyte/macrophage development is mainly influenced by the monocyte colony-stimulating factor (also known as CSF-1) and by the cytokine Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF). In particular, OCTN2 is induced during the differentiation of monocytes to macrophages by the mTOR-STAT3 pathway after a GM-CSF stimulus and not by PPAR transcription factors: GM-CSF causes the activation of mTOR kinase, leading to the phosphorylation and activation of the transcription factor STAT3, which, in turn, is responsible for OCTN2 transcription. Rapamycin, the specific inhibitor of mTOR, reduced the expression of the transporter at both mRNA and protein levels. Other activators of STAT3, like IL-6 and CSF-1, leading to the phosphorylation of STAT3, induced OCTN2 expression. Concerning CSF-1, a comparable induction of OCTN2 mRNA and protein is also observed upon the differentiation of monocytes. OCTN1, which is highly expressed in CD14+ monocytes, is downregulated by CSF1 [113]. Recently, a study proposed that the dysregulation of monocyte adaptation to the environment of the gastrointestinal mucosa is the key process leading to IBD. In the context of colitis, an increase in OCTN1 gene expression after the differentiation of THP-1 monocytic leukemia cells into macrophages using phorbol-12-myristate-13-acetate has been observed [68].

4.3. OCTN Regulation in Epithelia

Many of the findings concerning OCTN regulation in inflammation have been obtained focusing on the respiratory and gastrointestinal epithelia, which, although distinct, are considered part of a shared mucosal immune system, the “gut–lung axis”.

4.3.1. Airway Epithelium

In the context of the airway epithelium, contradictory data were collected, and some of the most recent examples are reported here. An impressive increase in OCTN2 mRNA levels and OCTN1 and 2 protein expression was observed if an inflammatory reaction (representative of epithelial inflammation in asthma) was induced by exposing to microbe-specific stimulus LPS or HDM the ALI Calu-3 layers, an in vitro model anatomically similar to the native bronchial epithelium [114]. In contrast, a lack of the regulation of the OCTN2 expression levels by LPS has been observed in a further study performed in Calu-3 cells and in the EpiAirway™ system, which consists of a pseudostratified epithelium containing the differentiated cell types found in the respiratory epithelium. Similarly, neither TNFα nor the anti-inflammatory Interleukin 4 (IL-4) affected OCTN2 expression [115]. In another paper, LPS treatment has been found to induce the downregulation of the expression of both OCTN1 and OCTN2 mRNA and proteins with a concomitant increase in inflammation in the alveolar epithelial cell model of the A549 cells [116].

4.3.2. Gut Epithelium

The dextran sodium sulfate (DSS)-induced colitis model is widely used for the study of IBD pathogenesis because this model represents many similarities to the immunological and histopathological features of human IBD. PPARα, PPARγ, and RXRα mRNA levels were downregulated in the colon of DSS-treated mice. In DSS-induced colitis, the agonist of PPARγ, luteolin, stimulating PPARγ reduced the expression levels of IL-1β and IL-6 and increased both the mouse Octn2 mRNA and protein expression. Mouse Octn1 upregulation has been described in the apical membrane of small intestinal epithelial cells after DSS treatment [68] and refs. herein. The severity of intestinal inflammation in DSS-induced colitis was greater in OCTN1−/− mice than in wild-type mice.
PPARγ expression in colon epithelial cells is associated with intestinal microorganisms likely from the involvement of the TLR4. The enteric microbiome is responsible for the strong regulation of PPARγ activation and OCTN2 expression [104]. A computational approach has been employed to analyze the effects of gut microbiome composition on gene expression in intestinal epithelial cells. Then, to screen differentially expressed genes, microbiota-depleted mouse samples were compared to control mouse samples, and OCTN1 resulted as enriched in the samples of IBD [117].
The effect of the proinflammatory cytokines TNF-α, IFN-γ, and IL-1β on OCTN2 expression was investigated in the human colon cell line FHC cells [67]. Mixed proinflammatory cytokines TNF-α, IL-1β, and IFNγ downregulated the expression of OCTN2 and reduced the carnitine content acting by PPARγ/RXRα pathways in FHC cells. Oppositely, in a further study, in the case of intestinal inflammation, IFN-γ was responsible for the stimulation of OCTN2 mRNA and protein expression along with increased total protein expression in the proximal colon and also increased apical OCTN2 abundance. TNFα does not alter colonic OCTN2 expression or activity but increases apical abundance and OCTN2 activity in the small intestine [118]. IFN-γ and TNFα increased the carnitine uptake. A decrease in the mRNA expression of OCTN2 was reported in the sigmoid region of the UC colon [119]. In a further study, the mRNA expression levels of OCTNs were detected in the terminal ileum and the colon of the intestine in IBD and UC patients [120]: in the terminal ileum of IBD patients, the mRNA expression levels of OCTN2 were significantly decreased, whereas changes in OCTN1 were not statistically significant. In the colon of IBD patients, OCTN2 mRNA levels were also significantly decreased. In the case of the mRNA expression of OCTN1, a significant increase was only observed in UC patients. The whole mucosal mRNA expression of OCTN1 was not significantly different between inflamed and non-inflamed mucosa, both in CD and UC patients. In contrast, the whole expression level of OCTN2 mRNA at the inflamed mucosa was significantly reduced compared to non-inflamed areas, both in CD and UC patients [121].

4.3.3. Other Epithelia

In the placenta, the significant endogenous expression of TNFα has been detected during the first trimester of pregnancy; indeed, inflammation accompanies pregnancy for physiological reasons. OCTN1 and OCTN2 expression in placenta have been found [6]. This information would allow us to hypothesize a possible modulation in the mRNA expression of the OCTNs.
The effects of inflammation in the lactating mammary gland have been investigated, as well, by evaluating LPS-induced inflammation effects in the lactating rat mammary gland at different lactation stages [122]. The mRNA expression levels of OCTN1 were markedly higher in mammary glands at lactation day 11 compared to lactation day 4, though no statistical significance was observed. LPS downregulated OCTN2.

5. Relationships of OCTNs with Altered Metabolite Profiles in Inflammation-Based Diseases

In the inflammation-based pathologies, an alteration of OCTNs would correspond to altered protein activity or expression levels, resulting in an alteration of substrate concentration in tissues and body fluids. On this basis, studies that investigated the metabolite profiles of subjects affected by inflammation-based pathologies have been analyzed to identify the possible derangement of OCTN substrates.
A complex endogenous inflammatory cascade accompanies injury and repair processes after stroke. A metabolomic analysis of patient plasma samples collected multiple times after stroke reveals that five carnitine derivatives, and hence potential OCTN substrates, showed a gradually decreased concentration. The finding correlated well with the increase in ischemia’s energy requirement, which may cause a time-dependent depletion of acylcarnitines [123]. Moreover, other studies that compare carnitine and acylcarnitine abundance between patients with stroke and healthy subjects highlighted higher levels of these two metabolites in patient’s samples [124,125].
An alteration of potential OCTN substrates was also found in patients affected by RA, a chronic, inflammatory autoimmune disease characterized by joint inflammation, pain, and swelling, leading to cartilage and bone damage. For example, a recent study finds that erythrocyte ergothioneine concentrations are higher in patients with mild RA disease activity than in healthy individuals [126]. Moreover, in the case of RA, the concentrations of the circulating metabolites are modulated depending, on the one hand, on their release from the inflamed joint, and on the other hand, on their uptake by the synovium [127], thus highlighting the potential involvement of the transporters’ activities in determining the RA metabolomic profile. In particular, macrophages, T cells, and the fibroblast-like synoviocytes (FLSs), key cells involved in the pathogenesis and progression of RA, potentially release metabolites into the bloodstream. However, the evaluation of metabolic profile modifications in RA is not easy; indeed, it is influenced by many factors like food, drugs, the microbiome, etc. An increase in the abundance of bacteria responsible for TMAO production was found in new-onset untreated RA patients, and the metabolites related to the choline pathway were found in several studies in synovial tissue, synovial fluid, and blood (serum/plasma) samples in both animal models and humans. TMAO, as well as choline, was found to be increased in serum samples in the murine K/BxN model of arthritis compared to control mice as well [127]. Conversely, other studies conducted using rats with Collagen-Induced Arthritis (CIA), an autoimmune disease model that shares features with RA, showed reduced levels of choline in CIA rats compared to the control [128]. A decrease in plasma or blood choline levels was also described in studies that recruited human RA patients [129,130]. Choline decrease may be due to an increased choline uptake and consumption by the inflamed synovium [130,131]. Another study focused on the identification of the differentially abundant metabolites between higher and lower RA activity patients showed that among the 31 metabolites that increased in lower disease activity, seven (3-hydroxydecanoylcarnitine, dihomo-linoleoylcarnitine, eicosenoylcarnitine, linoleoylcarnitine, linoleoylcarnitine, stearoylcarnitine, and palmitoylcarnitine) are a part of acylcarnitine metabolism [132]. Moreover, the role of carnitine in the increased level of CCL20 in RA was suggested. Indeed, fatty acid oxidation, as well as glycolysis, has been implicated in the immune regulation and activation of macrophages. It has been hypothesized that the exposure of monocytes to the hypoxic and inflammatory RA environment can impact their metabolic state and it would suggest that the increased carnitine abundance is part of a hypermetabolic state that can drive a CCL20-mediated inflammatory cascade to promote disease pathogenesis [133,134].
Osteoarthritis is another joint disease with chronic inflammation, progressive articular cartilage destruction, and subchondral bone sclerosis. Inflammation drives chondrocytes to express ECM-degrading enzymes, and the interruption of this pathway is a viable target to prevent cartilage degradation. It has been demonstrated that inflammation can alter the intracellular metabolism of chondrocytes, a process known as metabolic reprogramming [135]. Carlson et al. detected 1233 metabolites in the synovial fluid (SF), representative of the most accessible tissue near chondrocytes. In these samples, 35 potential biomarkers of OA, including phosphatidylcholine, lysophosphatidylcholine, and carnitine derivatives, have been identified [136]. Interestingly, a previous study conducted using a targeted metabolomics approach to identify metabolic markers for different class osteoarthritis patients, found that acylcarnitine and free carnitine may be potentially useful in distinguishing the different OA subtypes by analyzing SF samples [137]. Moreover, Mickiewicz et al. found reduced levels of acetylcarnitine, hexanoylcarnitine, N-phenylacetylglycine, and ethanolamine in OA samples compared to the controls [138]. Similarly, Tootsi et al. describe decreased medium- and long-chain acylcarnitines associated with OA severity [139]. The same study suggested that acylcarnitines might be important in the link between OA and cardiovascular comorbidity.
Another set of pathologies characterized by inflammation is IBD. The alteration of OCTN substrates/potential substrates in IBD patients has been found [140]. On the contrary, acylcarnitines appear to be more abundant in the CD patients compared with healthy controls. Interestingly, the abundance of certain types of acylcarnitines can help to classify IBD, IBD with damp-heat syndrome (IBD-DH), and IBD with spleen deficiency syndrome (IBD-SD). Indeed, Wu et al. demonstrated that three specific acylcarnitines (ACar 20:4, ACar 18:1, and ACar 20:3) were significantly increased in IBD plasma samples and that the ACar 8:1 was significantly increased in IBD with damp-heat syndrome when compared with IBD with spleen deficiency syndrome [141]. Another study found that the fecal acylcarnitine content in patients with IBD and mice with experimental colitis tended to increase compared to the control group, and the reason may be related to intestinal inflammation that led to mitochondrial dysfunction in the apical domain of the surface epithelium that may reduce the consumption of fatty acids [86]. Similar results were obtained from a fecal metabolome study on 424 patients with IBD and 255 non-IBD controls [142]. Moreover, the prediction of the disease course is highly desirable. This goal could be achievable using a multi-omics approach on serum samples; indeed, an established model of four metabolites, propionyl-L-carnitine, carnitine sarcosine, and sorbitol, combined with three proteins, IL-10, glial cell line-derived neurotrophic factor, and the T-cell surface CD8 alpha chain, was found to be predictive of relapse within two years [143].
Chronic inflammation has also been shown to play a role in the pathogenesis of frailty, a geriatric syndrome. The age-related significant rise in inflammatory markers, “inflammaging”, could predispose to frailty. The dysregulation of the carnitine shuttle has been found to play a role in the risk of frailty. Indeed, a frailty metabolic phenotype, including a decrease in six carnitines, distinguishes frail from non-frail phenotypes [144]. In the same work, an SNP for OCTN1 has been recently associated with decreased carnitine levels in frailty.

6. Perspectives and Concluding Remarks

The link between OCTNs and inflammatory diseases emerged in the last years. The ubiquitous tissue expression of these transporters together with the increasing numbers of studies on their regulation indicate good perspectives of deeply understanding their role in inflammation. Hence, due to the complexity of metabolic phenomena occurring in inflammation, future studies on OCTNs will furnish an important piece of knowledge in dissecting the molecular aspects of metabolic alterations occurring in inflammatory processes. Moreover, the increasing refinement of the structural aspects of these transporters and their structure/function relationships will give great support for the design of novel drugs as well as for the appropriate repurposing of already-known drugs for controlling deleterious aspects of inflammation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom14040392/s1, Table S1: SLC22A4 and SLC22A5 SNPs and links with human pathologies [70,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188].

Author Contributions

Conceptualization, L.P. and C.I.; visualization, L.P., M.G., L.C., M.S. and I.E.; writing—original draft preparation, L.P., L.C., M.G. and C.I.; writing—review and editing, L.P., M.S. and C.I.; supervision, C.I.; funding acquisition, L.P., I.E. and M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by PRIN (Progetti di Ricerca di Interesse Nazionale), project code 2022JWT5XS, CUP H53D23004820006 to L.P. and I.E. granted by MUR (Ministry of University and Research)—Italy, funded by the European Union—Next Generation EU—PNRR M4.C2.1.1. We acknowledge co-funding from the Next Generation EU, in the context of the National Recovery and Resilience Plan, Investment PE8—Project Age-It: “Ageing Well in an Ageing Society” to M.S. This resource was co-financed by the Next Generation EU [DM 1557 11.10.2022]. The views and opinions expressed are only those of the authors and do not necessarily reflect those of the European Union or the European Commission. Neither the European Union nor the European Commission can be held responsible for them.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Yu, W.; Wang, Z.; Yu, X.; Zhao, Y.; Xie, Z.; Zhang, K.; Chi, Z.; Chen, S.; Xu, T.; Jiang, D.; et al. Kir2.1-mediated membrane potential promotes nutrient acquisition and inflammation through regulation of nutrient transporters. Nat. Commun. 2022, 13, 3544. [Google Scholar] [CrossRef] [PubMed]
  2. Engelhart, D.C.; Granados, J.C.; Shi, D.; Saier, M.H., Jr.; Baker, M.E.; Abagyan, R.; Nigam, S.K. Systems Biology Analysis Reveals Eight SLC22 Transporter Subgroups, Including OATs, OCTs, and OCTNs. Int. J. Mol. Sci. 2020, 21, 1791. [Google Scholar] [CrossRef] [PubMed]
  3. Zhu, C.; Nigam, K.B.; Date, R.C.; Bush, K.T.; Springer, S.A.; Saier, M.H., Jr.; Wu, W.; Nigam, S.K. Evolutionary Analysis and Classification of OATs, OCTs, OCTNs, and Other SLC22 Transporters: Structure-Function Implications and Analysis of Sequence Motifs. PLoS ONE 2015, 10, e0140569. [Google Scholar] [CrossRef]
  4. Eraly, S.A.; Monte, J.C.; Nigam, S.K. Novel slc22 transporter homologs in fly, worm, and human clarify the phylogeny of organic anion and cation transporters. Physiol. Genom. 2004, 18, 12–24. [Google Scholar] [CrossRef] [PubMed]
  5. Pochini, L.; Galluccio, M.; Scalise, M.; Console, L.; Indiveri, C. OCTN: A Small Transporter Subfamily with Great Relevance to Human Pathophysiology, Drug Discovery, and Diagnostics. SLAS Discov. 2019, 24, 89–110. [Google Scholar] [CrossRef] [PubMed]
  6. Koepsell, H. Organic Cation Transporters in Health and Disease. Pharmacol. Rev. 2020, 72, 253–319. [Google Scholar] [CrossRef] [PubMed]
  7. Masuo, Y.; Ohba, Y.; Yamada, K.; Al-Shammari, A.H.; Seba, N.; Nakamichi, N.; Ogihara, T.; Kunishima, M.; Kato, Y. Combination Metabolomics Approach for Identifying Endogenous Substrates of Carnitine/Organic Cation Transporter OCTN1. Pharm. Res. 2018, 35, 224. [Google Scholar] [CrossRef] [PubMed]
  8. Drenberg, C.D.; Gibson, A.A.; Pounds, S.B.; Shi, L.; Rhinehart, D.P.; Li, L.; Hu, S.; Du, G.; Nies, A.T.; Schwab, M.; et al. OCTN1 Is a High-Affinity Carrier of Nucleoside Analogues. Cancer Res. 2017, 77, 2102–2111. [Google Scholar] [CrossRef]
  9. Pochini, L.; Scalise, M.; Galluccio, M.; Pani, G.; Siminovitch, K.A.; Indiveri, C. The human OCTN1 (SLC22A4) reconstituted in liposomes catalyzes acetylcholine transport which is defective in the mutant L503F associated to the Crohn’s disease. Biochim. Biophys. Acta 2012, 1818, 559–565. [Google Scholar] [CrossRef]
  10. Pochini, L.; Scalise, M.; Di Silvestre, S.; Belviso, S.; Pandolfi, A.; Arduini, A.; Bonomini, M.; Indiveri, C. Acetylcholine and acetylcarnitine transport in peritoneum: Role of the SLC22A4 (OCTN1) transporter. Biochim. Biophys. Acta 2016, 1858, 653–660. [Google Scholar] [CrossRef]
  11. Pochini, L.; Scalise, M.; Indiveri, C. Immuno-detection of OCTN1 (SLC22A4) in HeLa cells and characterization of transport function. Int. Immunopharmacol. 2015, 29, 21–26. [Google Scholar] [CrossRef] [PubMed]
  12. Grundemann, D.; Harlfinger, S.; Golz, S.; Geerts, A.; Lazar, A.; Berkels, R.; Jung, N.; Rubbert, A.; Schomig, E. Discovery of the ergothioneine transporter. Proc. Natl. Acad. Sci. USA 2005, 102, 5256–5261. [Google Scholar] [CrossRef] [PubMed]
  13. Nishiyama, M.; Nakamichi, N.; Yoshimura, T.; Masuo, Y.; Komori, T.; Ishimoto, T.; Matsuo, J.I.; Kato, Y. Homostachydrine is a Xenobiotic Substrate of OCTN1/SLC22A4 and Potentially Sensitizes Pentylenetetrazole-Induced Seizures in Mice. Neurochem. Res. 2020, 45, 2664–2678. [Google Scholar] [CrossRef] [PubMed]
  14. Juraszek, B.; Nalecz, K.A. SLC22A5 (OCTN2) Carnitine Transporter-Indispensable for Cell Metabolism, a Jekyll and Hyde of Human Cancer. Molecules 2020, 25, 14. [Google Scholar] [CrossRef]
  15. Ingoglia, F.; Visigalli, R.; Rotoli, B.M.; Barilli, A.; Riccardi, B.; Puccini, P.; Dall’Asta, V. Functional activity of L-carnitine transporters in human airway epithelial cells. Biochim. Biophys. Acta 2016, 1858, 210–219. [Google Scholar] [CrossRef] [PubMed]
  16. Tamai, I.; Ohashi, R.; Nezu, J.I.; Sai, Y.; Kobayashi, D.; Oku, A.; Shimane, M.; Tsuji, A. Molecular and functional characterization of organic cation/carnitine transporter family in mice. J. Biol. Chem. 2000, 275, 40064–40072. [Google Scholar] [CrossRef] [PubMed]
  17. Pochini, L.; Barone, F.; Console, L.; Brunocilla, C.; Galluccio, M.; Scalise, M.; Indiveri, C. OCTN1 (SLC22A4) displays two different transport pathways for organic cations or zwitterions. Biochim. Biophys. Acta Biomembr. 2023, 1866, 184263. [Google Scholar] [CrossRef]
  18. Brockmoller, J.; Tzvetkov, M.V.; Hu, S. (Eds.) Organic Cation Transporter 1 (OCT1): Not Vital for Life, but of Substantial Biomedical Relevance. Front. Pharmacol. 2021, 11, 143. [Google Scholar]
  19. Nigam, S.K. The SLC22 Transporter Family: A Paradigm for the Impact of Drug Transporters on Metabolic Pathways, Signaling, and Disease. Annu. Rev. Pharmacol. Toxicol. 2018, 58, 663–687. [Google Scholar] [CrossRef]
  20. Horvath, G.; Schmid, N.; Fragoso, M.A.; Schmid, A.; Conner, G.E.; Salathe, M.; Wanner, A. Epithelial organic cation transporters ensure pH-dependent drug absorption in the airway. Am. J. Respir. Cell Mol. Biol. 2007, 36, 53–60. [Google Scholar] [CrossRef]
  21. Barilli, A.; Visigalli, R.; Ferrari, F.; Di Lascia, M.; Riccardi, B.; Puccini, P.; Dall’Asta, V.; Rotoli, B.M. Organic cation transporters (OCTs/OCTNs) in human primary alveolar epithelial cells. Biochem. Biophys. Res. Commun. 2021, 576, 27–32. [Google Scholar] [CrossRef] [PubMed]
  22. Chandler, J.D.; Hu, X.; Ko, E.J.; Park, S.; Lee, Y.T.; Orr, M.; Fernandes, J.; Uppal, K.; Kang, S.M.; Jones, D.P.; et al. Metabolic pathways of lung inflammation revealed by high-resolution metabolomics (HRM) of H1N1 influenza virus infection in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 311, R906–R916. [Google Scholar] [CrossRef] [PubMed]
  23. Gallagher, K.; Catesson, A.; Griffin, J.L.; Holmes, E.; Williams, H.R.T. Metabolomic Analysis in Inflammatory Bowel Disease: A Systematic Review. J. Crohn’s Colitis 2021, 15, 813–826. [Google Scholar] [CrossRef] [PubMed]
  24. Console, L.; Scalise, M.; Indiveri, C. Exosomes in inflammation and role as biomarkers. Clin. Chim. Acta 2019, 488, 165–171. [Google Scholar] [CrossRef] [PubMed]
  25. Suo, Y.; Wright, N.J.; Guterres, H.; Fedor, J.G.; Butay, K.J.; Borgnia, M.J.; Im, W.; Lee, S.Y. Molecular basis of polyspecific drug binding and transport by OCT1 and OCT2. bioRxiv 2023. [Google Scholar] [CrossRef]
  26. Khanppnavar, B.; Maier, J.; Herborg, F.; Gradisch, R.; Lazzarin, E.; Luethi, D.; Yang, J.W.; Qi, C.; Holy, M.; Jantsch, K.; et al. Structural basis of organic cation transporter-3 inhibition. Nat. Commun. 2022, 13, 6714. [Google Scholar] [CrossRef] [PubMed]
  27. Pochini, L.; Scalise, M.; Galluccio, M.; Indiveri, C. OCTN cation transporters in health and disease: Role as drug targets and assay development. J. Biomol. Screen. 2013, 18, 851–867. [Google Scholar] [CrossRef] [PubMed]
  28. Tamai, I. Pharmacological and pathophysiological roles of carnitine/organic cation transporters (OCTNs: SLC22A4, SLC22A5 and Slc22a21). Biopharm. Drug Dispos. 2013, 34, 29–44. [Google Scholar] [CrossRef]
  29. Wu, X.; Prasad, P.D.; Leibach, F.H.; Ganapathy, V. cDNA sequence, transport function, and genomic organization of human OCTN2, a new member of the organic cation transporter family. Biochem. Biophys. Res. Commun. 1998, 246, 589–595. [Google Scholar] [CrossRef]
  30. Longo, N.; Frigeni, M.; Pasquali, M. Carnitine transport and fatty acid oxidation. Biochim. Biophys. Acta 2016, 1863, 2422–2435. [Google Scholar] [CrossRef]
  31. Tamai, I.; Yabuuchi, H.; Nezu, J.; Sai, Y.; Oku, A.; Shimane, M.; Tsuji, A. Cloning and characterization of a novel human pH-dependent organic cation transporter, OCTN1. FEBS Lett. 1997, 419, 107–111. [Google Scholar] [CrossRef] [PubMed]
  32. Pochini, L.; Scalise, M.; Galluccio, M.; Amelio, L.; Indiveri, C. Reconstitution in liposomes of the functionally active human OCTN1 (SLC22A4) transporter overexpressed in Escherichia coli. Biochem. J. 2011, 439, 227–233. [Google Scholar] [CrossRef] [PubMed]
  33. Pochini, L.; Peta, V.; Indiveri, C. Inhibition of the OCTN2 carnitine transporter by HgCl2 and methylmercury in the proteoliposome experimental model: Insights in the mechanism of toxicity. Toxicol. Mech. Methods 2013, 23, 68–76. [Google Scholar] [CrossRef] [PubMed]
  34. Galluccio, M.; Pochini, L.; Peta, V.; Ianni, M.; Scalise, M.; Indiveri, C. Functional and molecular effects of mercury compounds on the human OCTN1 cation transporter: C50 and C136 are the targets for potent inhibition. Toxicol. Sci. 2015, 144, 105–113. [Google Scholar] [CrossRef] [PubMed]
  35. Baba, S.P.; Bhatnagar, A. Role of Thiols in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 133–139. [Google Scholar] [CrossRef] [PubMed]
  36. Peltekova, V.D.; Wintle, R.F.; Rubin, L.A.; Amos, C.I.; Huang, Q.; Gu, X.; Newman, B.; Van Oene, M.; Cescon, D.; Greenberg, G.; et al. Functional variants of OCTN cation transporter genes are associated with Crohn disease. Nat. Genet. 2004, 36, 471–475. [Google Scholar] [CrossRef] [PubMed]
  37. Tokuhiro, S.; Yamada, R.; Chang, X.; Suzuki, A.; Kochi, Y.; Sawada, T.; Suzuki, M.; Nagasaki, M.; Ohtsuki, M.; Ono, M.; et al. An intronic SNP in a RUNX1 binding site of SLC22A4, encoding an organic cation transporter, is associated with rheumatoid arthritis. Nat. Genet. 2003, 35, 341–348. [Google Scholar] [CrossRef] [PubMed]
  38. Selo, M.A.; Sake, J.A.; Ehrhardt, C.; Salomon, J.J. Organic Cation Transporters in the Lung-Current and Emerging (Patho)Physiological and Pharmacological Concepts. Int. J. Mol. Sci. 2020, 21, 9168. [Google Scholar] [CrossRef]
  39. Salomon, J.J.; Gausterer, J.C.; Selo, M.A.; Hosoya, K.I.; Huwer, H.; Schneider-Daum, N.; Lehr, C.M.; Ehrhardt, C. OCTN2-Mediated Acetyl-l-Carnitine Transport in Human Pulmonary Epithelial Cells In Vitro. Pharmaceutics 2019, 11, 396. [Google Scholar] [CrossRef]
  40. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  41. Saeterstad, S.; Ostvik, A.E.; Royset, E.S.; Bakke, I.; Sandvik, A.K.; Granlund, A.V.B. Profound gene expression changes in the epithelial monolayer of active ulcerative colitis and Crohn’s disease. PLoS ONE 2022, 17, e0265189. [Google Scholar] [CrossRef] [PubMed]
  42. Gopallawa, I.; Dehinwal, R.; Bhatia, V.; Gujar, V.; Chirmule, N. A four-part guide to lung immunology: Invasion, inflammation, immunity, and intervention. Front. Immunol. 2023, 14, 1119564. [Google Scholar] [CrossRef] [PubMed]
  43. Peterson, L.W.; Artis, D. Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 2014, 14, 141–153. [Google Scholar] [CrossRef]
  44. Keulers, L.; Dehghani, A.; Knippels, L.; Garssen, J.; Papadopoulos, N.; Folkerts, G.; Braber, S.; van Bergenhenegouwen, J. Probiotics, prebiotics, and synbiotics to prevent or combat air pollution consequences: The gut-lung axis. Environ. Pollut. 2022, 302, 119066. [Google Scholar] [CrossRef] [PubMed]
  45. Martinez, A.; Martin, M.C.; Mendoza, J.L.; Taxonera, C.; Diaz-Rubio, M.; de la Concha, E.G.; Urcelay, E. Association of the organic cation transporter OCTN genes with Crohn’s disease in the Spanish population. Eur. J. Hum. Genet. 2006, 14, 222–226. [Google Scholar] [CrossRef]
  46. Defois, C.; Ratel, J.; Garrait, G.; Denis, S.; Le Goff, O.; Talvas, J.; Mosoni, P.; Engel, E.; Peyret, P. Food Chemicals Disrupt Human Gut Microbiota Activity And Impact Intestinal Homeostasis As Revealed By In Vitro Systems. Sci. Rep. 2018, 8, 11006. [Google Scholar] [CrossRef] [PubMed]
  47. Ishimoto, T.; Nakamichi, N.; Nishijima, H.; Masuo, Y.; Kato, Y. Carnitine/Organic Cation Transporter OCTN1 Negatively Regulates Activation in Murine Cultured Microglial Cells. Neurochem. Res. 2018, 43, 116–128. [Google Scholar] [CrossRef]
  48. Waller, S.; Tremelling, M.; Bredin, F.; Godfrey, L.; Howson, J.; Parkes, M. Evidence for association of OCTN genes and IBD5 with ulcerative colitis. Gut 2006, 55, 809–814. [Google Scholar] [CrossRef]
  49. Gulcin, I. Antioxidant and antiradical activities of L-carnitine. Life Sci. 2006, 78, 803–811. [Google Scholar] [CrossRef]
  50. Lee, B.J.; Lin, J.S.; Lin, Y.C.; Lin, P.T. Effects of L-carnitine supplementation on oxidative stress and antioxidant enzymes activities in patients with coronary artery disease: A randomized, placebo-controlled trial. Nutr. J. 2014, 13, 79. [Google Scholar] [CrossRef]
  51. Ribas, G.S.; Vargas, C.R.; Wajner, M. L-carnitine supplementation as a potential antioxidant therapy for inherited neurometabolic disorders. Gene 2014, 533, 469–476. [Google Scholar] [CrossRef] [PubMed]
  52. Fortin, G.; Yurchenko, K.; Collette, C.; Rubio, M.; Villani, A.C.; Bitton, A.; Sarfati, M.; Franchimont, D. L-carnitine, a diet component and organic cation transporter OCTN ligand, displays immunosuppressive properties and abrogates intestinal inflammation. Clin. Exp. Immunol. 2009, 156, 161–171. [Google Scholar] [CrossRef] [PubMed]
  53. Li, X.; Meng, F.; Li, H.; Hua, X.; Wu, L.; Yuan, X. L-carnitine alleviates oxidative stress-related damage via MAPK signaling in human lens epithelial cells exposed to H2O2. Int. J. Mol. Med. 2019, 44, 1515–1522. [Google Scholar] [CrossRef] [PubMed]
  54. Grundemann, D. The ergothioneine transporter controls and indicates ergothioneine activity—A review. Prev. Med. 2012, 54, S71–S74. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, H.; Yu, M.; Ochani, M.; Amella, C.A.; Tanovic, M.; Susarla, S.; Li, J.H.; Wang, H.; Yang, H.; Ulloa, L.; et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 2003, 421, 384–388. [Google Scholar] [CrossRef] [PubMed]
  56. Cox, M.A.; Bassi, C.; Saunders, M.E.; Nechanitzky, R.; Morgado-Palacin, I.; Zheng, C.; Mak, T.W. Beyond neurotransmission: Acetylcholine in immunity and inflammation. J. Intern. Med. 2020, 287, 120–133. [Google Scholar] [CrossRef] [PubMed]
  57. Halder, N.; Lal, G. Cholinergic System and Its Therapeutic Importance in Inflammation and Autoimmunity. Front. Immunol. 2021, 12, 660342. [Google Scholar] [CrossRef]
  58. Wessler, I.; Kirkpatrick, C.J. Acetylcholine beyond neurons: The non-neuronal cholinergic system in humans. Br. J. Pharmacol. 2008, 154, 1558–1571. [Google Scholar] [CrossRef]
  59. Kummer, W.; Krasteva-Christ, G. Non-neuronal cholinergic airway epithelium biology. Curr. Opin. Pharmacol. 2014, 16, 43–49. [Google Scholar] [CrossRef]
  60. Lips, K.S.; Volk, C.; Schmitt, B.M.; Pfeil, U.; Arndt, P.; Miska, D.; Ermert, L.; Kummer, W.; Koepsell, H. Polyspecific cation transporters mediate luminal release of acetylcholine from bronchial epithelium. Am. J. Respir. Cell Mol. Biol. 2005, 33, 79–88. [Google Scholar] [CrossRef]
  61. Nezu, J.; Tamai, I.; Oku, A.; Ohashi, R.; Yabuuchi, H.; Hashimoto, N.; Nikaido, H.; Sai, Y.; Koizumi, A.; Shoji, Y.; et al. Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter. Nat. Genet. 1999, 21, 91–94. [Google Scholar] [CrossRef] [PubMed]
  62. Stanley, C.A.; DeLeeuw, S.; Coates, P.M.; Vianey-Liaud, C.; Divry, P.; Bonnefont, J.P.; Saudubray, J.M.; Haymond, M.; Trefz, F.K.; Breningstall, G.N.; et al. Chronic cardiomyopathy and weakness or acute coma in children with a defect in carnitine uptake. Ann. Neurol. 1991, 30, 709–716. [Google Scholar] [CrossRef] [PubMed]
  63. Indiveri, C.; Iacobazzi, V.; Tonazzi, A.; Giangregorio, N.; Infantino, V.; Convertini, P.; Console, L.; Palmieri, F. The mitochondrial carnitine/acylcarnitine carrier: Function, structure and physiopathology. Mol. Asp. Med. 2011, 32, 223–233. [Google Scholar] [CrossRef] [PubMed]
  64. Grigat, S.; Fork, C.; Bach, M.; Golz, S.; Geerts, A.; Schomig, E.; Grundemann, D. The carnitine transporter SLC22A5 is not a general drug transporter, but it efficiently translocates mildronate. Drug Metab. Dispos. 2009, 37, 330–337. [Google Scholar] [CrossRef] [PubMed]
  65. Kato, Y.; Kubo, Y.; Iwata, D.; Kato, S.; Sudo, T.; Sugiura, T.; Kagaya, T.; Wakayama, T.; Hirayama, A.; Sugimoto, M.; et al. Gene knockout and metabolome analysis of carnitine/organic cation transporter OCTN1. Pharm. Res. 2010, 27, 832–840. [Google Scholar] [CrossRef]
  66. Park, H.J.; Jung, E.S.; Kong, K.A.; Park, E.M.; Cheon, J.H.; Choi, J.H. Identification of OCTN2 variants and their association with phenotypes of Crohn’s disease in a Korean population. Sci. Rep. 2016, 6, 22887. [Google Scholar] [CrossRef] [PubMed]
  67. Li, P.; Wang, Y.; Luo, J.; Zeng, Q.; Wang, M.; Bai, M.; Zhou, H.; Wang, J.; Jiang, H. Downregulation of OCTN2 by cytokines plays an important role in the progression of inflammatory bowel disease. Biochem. Pharmacol. 2020, 178, 114115. [Google Scholar] [CrossRef] [PubMed]
  68. Shimizu, T.; Masuo, Y.; Takahashi, S.; Nakamichi, N.; Kato, Y. Organic cation transporter Octn1-mediated uptake of food-derived antioxidant ergothioneine into infiltrating macrophages during intestinal inflammation in mice. Drug Metab. Pharmacokinet. 2015, 30, 231–239. [Google Scholar] [CrossRef]
  69. Rock, R.B.; Gekker, G.; Aravalli, R.N.; Hu, S.; Sheng, W.S.; Peterson, P.K. Potentiation of HIV-1 expression in microglial cells by nicotine: Involvement of transforming growth factor-beta 1. J. Neuroimmune Pharmacol. 2008, 3, 143–149. [Google Scholar] [CrossRef]
  70. Martini, M.; Ferrara, A.M.; Giachelia, M.; Panieri, E.; Siminovitch, K.; Galeotti, T.; Larocca, L.M.; Pani, G. Association of the OCTN1/1672T variant with increased risk for colorectal cancer in young individuals and ulcerative colitis patients. Inflamm. Bowel Dis. 2012, 18, 439–448. [Google Scholar] [CrossRef]
  71. Haghighatdoost, F.; Jabbari, M.; Hariri, M. The effect of L-carnitine on inflammatory mediators: A systematic review and meta-analysis of randomized clinical trials. Eur. J. Clin. Pharmacol. 2019, 75, 1037–1046. [Google Scholar] [CrossRef] [PubMed]
  72. Shekhawat, P.S.; Srinivas, S.R.; Matern, D.; Bennett, M.J.; Boriack, R.; George, V.; Xu, H.; Prasad, P.D.; Roon, P.; Ganapathy, V. Spontaneous development of intestinal and colonic atrophy and inflammation in the carnitine-deficient jvs (OCTN2(−/−)) mice. Mol. Genet. Metab. 2007, 92, 315–324. [Google Scholar] [CrossRef] [PubMed]
  73. Sonne, S.; Shekhawat, P.S.; Matern, D.; Ganapathy, V.; Ignatowicz, L. Carnitine deficiency in OCTN2−/− newborn mice leads to a severe gut and immune phenotype with widespread atrophy, apoptosis and a pro-inflammatory response. PLoS ONE 2012, 7, e47729. [Google Scholar] [CrossRef] [PubMed]
  74. Ingoglia, F.; Visigalli, R.; Rotoli, B.M.; Barilli, A.; Riccardi, B.; Puccini, P.; Milioli, M.; Di Lascia, M.; Bernuzzi, G.; Dall’Asta, V. Human macrophage differentiation induces OCTN2-mediated L-carnitine transport through stimulation of mTOR-STAT3 axis. J. Leukoc. Biol. 2017, 101, 665–674. [Google Scholar] [CrossRef] [PubMed]
  75. D’Argenio, G.; Calvani, M.; Casamassimi, A.; Petillo, O.; Margarucci, S.; Rienzo, M.; Peluso, I.; Calvani, R.; Ciccodicola, A.; Caporaso, N.; et al. Experimental colitis: Decreased Octn2 and Atb0+ expression in rat colonocytes induces carnitine depletion that is reversible by carnitine-loaded liposomes. FASEB J. 2006, 20, 2544–2546. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, T.F.; Vachharajani, V.T.; Yoza, B.K.; McCall, C.E. NAD+-dependent sirtuin 1 and 6 proteins coordinate a switch from glucose to fatty acid oxidation during the acute inflammatory response. J. Biol. Chem. 2012, 287, 25758–25769. [Google Scholar] [CrossRef] [PubMed]
  77. Batista-Gonzalez, A.; Vidal, R.; Criollo, A.; Carreno, L.J. New Insights on the Role of Lipid Metabolism in the Metabolic Reprogramming of Macrophages. Front. Immunol. 2019, 10, 2993. [Google Scholar] [CrossRef] [PubMed]
  78. Jamali-Raeufy, N.; Alizadeh, F.; Mehrabi, Z.; Mehrabi, S.; Goudarzi, M. Acetyl-L-carnitine confers neuroprotection against lipopolysaccharide (LPS) -induced neuroinflammation by targeting TLR4/NFkappaB, autophagy, inflammation and oxidative stress. Metab. Brain Dis. 2021, 36, 1391–1401. [Google Scholar] [CrossRef]
  79. Zahedi, E.; Sadr, S.S.; Sanaeierad, A.; Roghani, M. Chronic acetyl-L-carnitine treatment alleviates behavioral deficits and neuroinflammation through enhancing microbiota derived-SCFA in valproate model of autism. Biomed. Pharmacother. 2023, 163, 114848. [Google Scholar] [CrossRef]
  80. Miecz, D.; Januszewicz, E.; Czeredys, M.; Hinton, B.T.; Berezowski, V.; Cecchelli, R.; Nalecz, K.A. Localization of organic cation/carnitine transporter (OCTN2) in cells forming the blood-brain barrier. J. Neurochem. 2008, 104, 113–123. [Google Scholar] [CrossRef]
  81. Inano, A.; Sai, Y.; Nikaido, H.; Hasimoto, N.; Asano, M.; Tsuji, A.; Tamai, I. Acetyl-L-carnitine permeability across the blood-brain barrier and involvement of carnitine transporter OCTN2. Biopharm. Drug Dispos. 2003, 24, 357–365. [Google Scholar] [CrossRef] [PubMed]
  82. Keshani, M.; Alikiaii, B.; Askari, G.; Yahyapoor, F.; Ferns, G.A.; Bagherniya, M. The effects of L-carnitine supplementation on inflammatory factors, oxidative stress, and clinical outcomes in patients with sepsis admitted to the intensive care unit (ICU): Study protocol for a double blind, randomized, placebo-controlled clinical trial. Trials 2022, 23, 170. [Google Scholar] [CrossRef] [PubMed]
  83. Yahyapoor, F.; Sedaghat, A.; Feizi, A.; Bagherniya, M.; Pahlavani, N.; Khadem-Rezaiyan, M.; Safarian, M.; Islam, M.S.; Zarifi, S.H.; Arabi, S.M.; et al. The effects of l-Carnitine supplementation on inflammatory markers, clinical status, and 28 days mortality in critically ill patients: A double-blind, randomized, placebo-controlled trial. Clin. Nutr. ESPEN 2022, 49, 61–67. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, S.; Xu, J.; Zheng, J.; Zhang, X.; Shao, J.; Zhao, L.; Hao, J. Anti-Inflammatory and Antioxidant Effects of Acetyl-L-Carnitine on Atherosclerotic Rats. Med. Sci. Monit. 2020, 26, e920250. [Google Scholar] [CrossRef] [PubMed]
  85. Mikhailova, T.L.; Sishkova, E.; Poniewierka, E.; Zhidkov, K.P.; Bakulin, I.G.; Kupcinskas, L.; Lesniakowski, K.; Grinevich, V.B.; Malecka-Panas, E.; Ardizzone, S.; et al. Randomised clinical trial: The efficacy and safety of propionyl-L-carnitine therapy in patients with ulcerative colitis receiving stable oral treatment. Aliment. Pharmacol. Ther. 2011, 34, 1088–1097. [Google Scholar] [CrossRef] [PubMed]
  86. Smith, S.A.; Ogawa, S.A.; Chau, L.; Whelan, K.A.; Hamilton, K.E.; Chen, J.; Tan, L.; Chen, E.Z.; Keilbaugh, S.; Fogt, F.; et al. Mitochondrial dysfunction in inflammatory bowel disease alters intestinal epithelial metabolism of hepatic acylcarnitines. J. Clin. Investig. 2021, 131. [Google Scholar] [CrossRef] [PubMed]
  87. Sanchez-Lopez, E.; Zhong, Z.; Stubelius, A.; Sweeney, S.R.; Booshehri, L.M.; Antonucci, L.; Liu-Bryan, R.; Lodi, A.; Terkeltaub, R.; Lacal, J.C.; et al. Choline Uptake and Metabolism Modulate Macrophage IL-1beta and IL-18 Production. Cell Metab. 2019, 29, 1350–1362. [Google Scholar] [CrossRef] [PubMed]
  88. 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, 565–583. [Google Scholar] [CrossRef]
  89. Wagner, C.A.; Lukewille, U.; Kaltenbach, S.; Moschen, I.; Broer, A.; Risler, T.; Broer, S.; Lang, F. Functional and pharmacological characterization of human Na(+)-carnitine cotransporter hOCTN2. Am. J. Physiol. Renal Physiol. 2000, 279, F584–F591. [Google Scholar] [CrossRef]
  90. Veskovic, M.; Mladenovic, D.; Milenkovic, M.; Tosic, J.; Borozan, S.; Gopcevic, K.; Labudovic-Borovic, M.; Dragutinovic, V.; Vucevic, D.; Jorgacevic, B.; et al. Betaine modulates oxidative stress, inflammation, apoptosis, autophagy, and Akt/mTOR signaling in methionine-choline deficiency-induced fatty liver disease. Eur. J. Pharmacol. 2019, 848, 39–48. [Google Scholar] [CrossRef]
  91. Zhou, S.; Xue, J.; Shan, J.; Hong, Y.; Zhu, W.; Nie, Z.; Zhang, Y.; Ji, N.; Luo, X.; Zhang, T.; et al. Gut-Flora-Dependent Metabolite Trimethylamine-N-Oxide Promotes Atherosclerosis-Associated Inflammation Responses by Indirect ROS Stimulation and Signaling Involving AMPK and SIRT1. Nutrients 2022, 14, 3338. [Google Scholar] [CrossRef] [PubMed]
  92. Fu, B.C.; Hullar, M.A.J.; Randolph, T.W.; Franke, A.A.; Monroe, K.R.; Cheng, I.; Wilkens, L.R.; Shepherd, J.A.; Madeleine, M.M.; Le Marchand, L.; et al. Associations of plasma trimethylamine N-oxide, choline, carnitine, and betaine with inflammatory and cardiometabolic risk biomarkers and the fecal microbiome in the Multiethnic Cohort Adiposity Phenotype Study. Am. J. Clin. Nutr. 2020, 111, 1226–1234. [Google Scholar] [CrossRef] [PubMed]
  93. Tacconi, E.; Palma, G.; De Biase, D.; Luciano, A.; Barbieri, M.; de Nigris, F.; Bruzzese, F. Microbiota Effect on Trimethylamine N-Oxide Production: From Cancer to Fitness-A Practical Preventing Recommendation and Therapies. Nutrients 2023, 15, 563. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, S.; Li, X.; Yang, F.; Zhao, R.; Pan, X.; Liang, J.; Tian, L.; Li, X.; Liu, L.; Xing, Y.; et al. Gut Microbiota-Dependent Marker TMAO in Promoting Cardiovascular Disease: Inflammation Mechanism, Clinical Prognostic, and Potential as a Therapeutic Target. Front. Pharmacol. 2019, 10, 1360. [Google Scholar] [CrossRef] [PubMed]
  95. Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.; Wang, Z.; Hazen, S.L.; Lusis, A.J.; Shih, D.M. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-kappaB. J. Am. Heart Assoc. 2016, 5, e002767. [Google Scholar] [CrossRef] [PubMed]
  96. Constantino-Jonapa, L.A.; Espinoza-Palacios, Y.; Escalona-Montano, A.R.; Hernandez-Ruiz, P.; Amezcua-Guerra, L.M.; Amedei, A.; Aguirre-Garcia, M.M. Contribution of Trimethylamine N-Oxide (TMAO) to Chronic Inflammatory and Degenerative Diseases. Biomedicines 2023, 11, 431. [Google Scholar] [CrossRef] [PubMed]
  97. Vallance, H.D.; Koochin, A.; Branov, J.; Rosen-Heath, A.; Bosdet, T.; Wang, Z.; Hazen, S.L.; Horvath, G. Marked elevation in plasma trimethylamine-N-oxide (TMAO) in patients with mitochondrial disorders treated with oral l-carnitine. Mol. Genet. Metab. Rep. 2018, 15, 130–133. [Google Scholar] [CrossRef]
  98. Bader, S.; Diener, M. Segmental differences in the non-neuronal cholinergic system in rat caecum. Pflug. Arch. 2018, 470, 669–679. [Google Scholar] [CrossRef]
  99. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; Gonzalez, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef]
  100. Luo, H.; Zhang, Y.; Guo, H.; Zhang, L.; Li, X.; Ringseis, R.; Wen, G.; Hui, D.; Liang, A.; Eder, K.; et al. Transcriptional regulation of the human, porcine and bovine OCTN2 gene by PPARalpha via a conserved PPRE located in intron 1. BMC Genet. 2014, 15, 90. [Google Scholar] [CrossRef]
  101. Wada, E.; Koyanagi, S.; Kusunose, N.; Akamine, T.; Masui, H.; Hashimoto, H.; Matsunaga, N.; Ohdo, S. Modulation of peroxisome proliferator-activated receptor-alpha activity by bile acids causes circadian changes in the intestinal expression of Octn1/Slc22a4 in mice. Mol. Pharmacol. 2015, 87, 314–322. [Google Scholar] [CrossRef]
  102. Youssef, J.; Badr, M. Role of Peroxisome Proliferator-Activated Receptors in Inflammation Control. J. Biomed. Biotechnol. 2004, 2004, 156–166. [Google Scholar] [CrossRef] [PubMed]
  103. Decara, J.; Rivera, P.; Lopez-Gambero, A.J.; Serrano, A.; Pavon, F.J.; Baixeras, E.; Rodriguez de Fonseca, F.; Suarez, J. Peroxisome Proliferator-Activated Receptors: Experimental Targeting for the Treatment of Inflammatory Bowel Diseases. Front. Pharmacol. 2020, 11, 730. [Google Scholar] [CrossRef] [PubMed]
  104. D’Argenio, G.; Petillo, O.; Margarucci, S.; Torpedine, A.; Calarco, A.; Koverech, A.; Boccia, A.; Paolella, G.; Peluso, G. Colon OCTN2 gene expression is up-regulated by peroxisome proliferator-activated receptor gamma in humans and mice and contributes to local and systemic carnitine homeostasis. J. Biol. Chem. 2010, 285, 27078–27087. [Google Scholar] [CrossRef] [PubMed]
  105. Wen, G.; Ringseis, R.; Eder, K. Mouse OCTN2 is directly regulated by peroxisome proliferator-activated receptor alpha (PPARalpha) via a PPRE located in the first intron. Biochem. Pharmacol. 2010, 79, 768–776. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, C.; Uray, I.P.; Mazumdar, A.; Mayer, J.A.; Brown, P.H. SLC22A5/OCTN2 expression in breast cancer is induced by estrogen via a novel intronic estrogen-response element (ERE). Breast Cancer Res. Treat. 2012, 134, 101–115. [Google Scholar] [CrossRef] [PubMed]
  107. Qu, Q.; Qu, J.; Zhan, M.; Wu, L.X.; Zhang, Y.W.; Lou, X.Y.; Fu, L.J.; Zhou, H.H. Different involvement of promoter methylation in the expression of organic cation/carnitine transporter 2 (OCTN2) in cancer cell lines. PLoS ONE 2013, 8, e76474. [Google Scholar] [CrossRef] [PubMed]
  108. Buelow, D.R.; Anderson, J.T.; Pounds, S.B.; Shi, L.; Lamba, J.K.; Hu, S.; Gibson, A.A.; Goodwin, E.A.; Sparreboom, A.; Baker, S.D. DNA Methylation-Based Epigenetic Repression of SLC22A4 Promotes Resistance to Cytarabine in Acute Myeloid Leukemia. Clin. Transl. Sci. 2021, 14, 137–142. [Google Scholar] [CrossRef]
  109. Maeda, T.; Hirayama, M.; Kobayashi, D.; Miyazawa, K.; Tamai, I. Mechanism of the regulation of organic cation/carnitine transporter 1 (SLC22A4) by rheumatoid arthritis-associated transcriptional factor RUNX1 and inflammatory cytokines. Drug Metab. Dispos. 2007, 35, 394–401. [Google Scholar] [CrossRef]
  110. Karimian Pour, N.; McColl, E.R.; Piquette-Miller, M. Impact of Viral Inflammation on the Expression of Renal Drug Transporters in Pregnant Rats. Pharmaceutics 2019, 11, 624. [Google Scholar] [CrossRef]
  111. Harrach, S.; Edemir, B.; Schmidt-Lauber, C.; Pap, T.; Bertrand, J.; Ciarimboli, G. Importance of the novel organic cation transporter 1 for tyrosine kinase inhibition by saracatinib in rheumatoid arthritis synovial fibroblasts. Sci. Rep. 2017, 7, 1258. [Google Scholar] [CrossRef] [PubMed]
  112. Zhou, X.; Ringseis, R.; Wen, G.; Eder, K. The pro-inflammatory cytokine tumor necrosis factor alpha stimulates expression of the carnitine transporter OCTN2 (novel organic cation transporter 2) and carnitine uptake via nuclear factor-kappaB in Madin-Darby bovine kidney cells. J. Dairy Sci. 2015, 98, 3840–3848. [Google Scholar] [CrossRef] [PubMed]
  113. Baillie, J.K.; Arner, E.; Daub, C.; De Hoon, M.; Itoh, M.; Kawaji, H.; Lassmann, T.; Carninci, P.; Forrest, A.R.; Hayashizaki, Y.; et al. Analysis of the human monocyte-derived macrophage transcriptome and response to lipopolysaccharide provides new insights into genetic aetiology of inflammatory bowel disease. PLoS Genet. 2017, 13, e1006641. [Google Scholar] [CrossRef] [PubMed]
  114. Mukherjee, M.; Cingolani, E.; Pritchard, D.I.; Bosquillon, C. Enhanced expression of Organic Cation Transporters in bronchial epithelial cell layers following insults associated with asthma—Impact on salbutamol transport. Eur. J. Pharm. Sci. 2017, 106, 62–70. [Google Scholar] [CrossRef]
  115. Rotoli, B.M.; Visigalli, R.; Barilli, A.; Ferrari, F.; Bianchi, M.G.; Di Lascia, M.; Riccardi, B.; Puccini, P.; Dall’Asta, V. Functional analysis of OCTN2 and ATB0,+ in normal human airway epithelial cells. PLoS ONE 2020, 15, e0228568. [Google Scholar] [CrossRef]
  116. Li, D.; Qi, C.; Zhou, J.; Wen, Z.; Zhu, X.; Xia, H.; Song, J. LPS-induced inflammation delays the transportation of ASP(+) due to down-regulation of OCTN1/2 in alveolar epithelial cells. J. Drug Target. 2020, 28, 437–447. [Google Scholar] [CrossRef] [PubMed]
  117. Zhu, W.; Li, J.; Wu, B. Gene expression profiling of the mouse gut: Effect of intestinal flora on intestinal health. Mol. Med. Rep. 2018, 17, 3667–3673. [Google Scholar] [CrossRef]
  118. Fujiya, M.; Inaba, Y.; Musch, M.W.; Hu, S.; Kohgo, Y.; Chang, E.B. Cytokine regulation of OCTN2 expression and activity in small and large intestine. Inflamm. Bowel Dis. 2011, 17, 907–916. [Google Scholar] [CrossRef]
  119. Noble, C.L.; Abbas, A.R.; Cornelius, J.; Lees, C.W.; Ho, G.T.; Toy, K.; Modrusan, Z.; Pal, N.; Zhong, F.; Chalasani, S.; et al. Regional variation in gene expression in the healthy colon is dysregulated in ulcerative colitis. Gut 2008, 57, 1398–1405. [Google Scholar] [CrossRef]
  120. Wojtal, K.A.; Eloranta, J.J.; Hruz, P.; Gutmann, H.; Drewe, J.; Staumann, A.; Beglinger, C.; Fried, M.; Kullak-Ublick, G.A.; Vavricka, S.R. Changes in mRNA expression levels of solute carrier transporters in inflammatory bowel disease patients. Drug Metab. Dispos. 2009, 37, 1871–1877. [Google Scholar] [CrossRef]
  121. Palmieri, O.; Latiano, A.; Scimeca, D.; Bossa, F.; Corritore, G.; Latiano, T.; Andriulli, A.; Annese, V. IL23R, ATG16L1, IRGM, OCTN1, and OCTN2 mRNA expression in inflamed and noninflamed mucosa of IBD patients. Inflamm. Bowel Dis. 2011, 17, 1832–1833. [Google Scholar] [CrossRef] [PubMed]
  122. Ling, B.; Alcorn, J. LPS-induced inflammation downregulates mammary gland glucose, fatty acid, and L-carnitine transporter expression at different lactation stages. Res. Vet. Sci. 2010, 89, 200–202. [Google Scholar] [CrossRef] [PubMed]
  123. Ahmed, W.; White, I.R.; Wilkinson, M.; Johnson, C.F.; Rattray, N.; Kishore, A.K.; Goodacre, R.; Smith, C.J.; Fowler, S.J. Breath and plasma metabolomics to assess inflammation in acute stroke. Sci. Rep. 2021, 11, 21949. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, P.; Li, R.; Antonov, A.A.; Wang, L.; Li, W.; Hua, Y.; Guo, H.; Wang, L.; Liu, P.; Chen, L.; et al. Discovery of Metabolite Biomarkers for Acute Ischemic Stroke Progression. J. Proteome Res. 2017, 16, 773–779. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, M.; Zhou, K.; Li, H.; Dong, X.; Tan, G.; Chai, Y.; Wang, W.; Bi, X. Potential of serum metabolites for diagnosing post-stroke cognitive impairment. Mol. Biosyst. 2015, 11, 3287–3296. [Google Scholar] [CrossRef] [PubMed]
  126. Taubert, D.; Lazar, A.; Grimberg, G.; Jung, N.; Rubbert, A.; Delank, K.S.; Perniok, A.; Erdmann, E.; Schomig, E. Association of rheumatoid arthritis with ergothioneine levels in red blood cells: A case control study. J. Rheumatol. 2006, 33, 2139–2145. [Google Scholar] [PubMed]
  127. Coras, R.; Murillo-Saich, J.D.; Guma, M. Circulating Pro- and Anti-Inflammatory Metabolites and Its Potential Role in Rheumatoid Arthritis Pathogenesis. Cells 2020, 9, 827. [Google Scholar] [CrossRef] [PubMed]
  128. Srivastava, N.K.; Sharma, S.; Sharma, R.; Sinha, N.; Mandal, S.K.; Sharma, D. Metabolic fingerprinting of joint tissue of collagen-induced arthritis (CIA) rat: In vitro, high resolution NMR (nuclear magnetic resonance) spectroscopy based analysis. EXCLI J. 2018, 17, 257–272. [Google Scholar] [CrossRef]
  129. Zhu, J.; Wang, T.; Lin, Y.; Xiong, M.; Chen, J.; Jian, C.; Zhang, J.; Xie, H.; Zeng, F.; Huang, Q.; et al. The change of plasma metabolic profile and gut microbiome dysbiosis in patients with rheumatoid arthritis. Front. Microbiol. 2022, 13, 931431. [Google Scholar] [CrossRef]
  130. Narasimhan, R.; Coras, R.; Rosenthal, S.B.; Sweeney, S.R.; Lodi, A.; Tiziani, S.; Boyle, D.; Kavanaugh, A.; Guma, M. Serum metabolomic profiling predicts synovial gene expression in rheumatoid arthritis. Arthritis Res. Ther. 2018, 20, 164. [Google Scholar] [CrossRef]
  131. Roivainen, A.; Parkkola, R.; Yli-Kerttula, T.; Lehikoinen, P.; Viljanen, T.; Mottonen, T.; Nuutila, P.; Minn, H. Use of positron emission tomography with methyl-11C-choline and 2-18F-fluoro-2-deoxy-D-glucose in comparison with magnetic resonance imaging for the assessment of inflammatory proliferation of synovium. Arthritis Rheum. 2003, 48, 3077–3084. [Google Scholar] [CrossRef] [PubMed]
  132. Hur, B.; Gupta, V.K.; Huang, H.; Wright, K.A.; Warrington, K.J.; Taneja, V.; Davis, J.M., 3rd; Sung, J. Plasma metabolomic profiling in patients with rheumatoid arthritis identifies biochemical features predictive of quantitative disease activity. Arthritis Res. Ther. 2021, 23, 164. [Google Scholar] [CrossRef] [PubMed]
  133. Rodgers, L.C.; Cole, J.; Rattigan, K.M.; Barrett, M.P.; Kurian, N.; McInnes, I.B.; Goodyear, C.S. The rheumatoid synovial environment alters fatty acid metabolism in human monocytes and enhances CCL20 secretion. Rheumatology 2020, 59, 869–878. [Google Scholar] [CrossRef] [PubMed]
  134. Zeisbrich, M.; Yanes, R.E.; Zhang, H.; Watanabe, R.; Li, Y.; Brosig, L.; Hong, J.; Wallis, B.B.; Giacomini, J.C.; Assimes, T.L.; et al. Hypermetabolic macrophages in rheumatoid arthritis and coronary artery disease due to glycogen synthase kinase 3b inactivation. Ann. Rheum. Dis. 2018, 77, 1053–1062. [Google Scholar] [CrossRef] [PubMed]
  135. Arra, M.; Abu-Amer, Y. Cross-talk of inflammation and chondrocyte intracellular metabolism in osteoarthritis. Osteoarthr. Cartil. 2023, 31, 1012–1021. [Google Scholar] [CrossRef] [PubMed]
  136. Carlson, A.K.; Rawle, R.A.; Adams, E.; Greenwood, M.C.; Bothner, B.; June, R.K. Application of global metabolomic profiling of synovial fluid for osteoarthritis biomarkers. Biochem. Biophys. Res. Commun. 2018, 499, 182–188. [Google Scholar] [CrossRef] [PubMed]
  137. Zhang, W.; Likhodii, S.; Zhang, Y.; Aref-Eshghi, E.; Harper, P.E.; Randell, E.; Green, R.; Martin, G.; Furey, A.; Sun, G.; et al. Classification of osteoarthritis phenotypes by metabolomics analysis. BMJ Open 2014, 4, e006286. [Google Scholar] [CrossRef]
  138. Mickiewicz, B.; Kelly, J.J.; Ludwig, T.E.; Weljie, A.M.; Wiley, J.P.; Schmidt, T.A.; Vogel, H.J. Metabolic analysis of knee synovial fluid as a potential diagnostic approach for osteoarthritis. J. Orthop. Res. 2015, 33, 1631–1638. [Google Scholar] [CrossRef]
  139. Tootsi, K.; Kals, J.; Zilmer, M.; Paapstel, K.; Ottas, A.; Martson, A. Medium- and long-chain acylcarnitines are associated with osteoarthritis severity and arterial stiffness in end-stage osteoarthritis patients: A case-control study. Int. J. Rheum. Dis. 2018, 21, 1211–1218. [Google Scholar] [CrossRef]
  140. Li, M.; Yang, L.; Mu, C.; Sun, Y.; Gu, Y.; Chen, D.; Liu, T.; Cao, H. Gut microbial metabolome in inflammatory bowel disease: From association to therapeutic perspectives. Comput. Struct. Biotechnol. J. 2022, 20, 2402–2414. [Google Scholar] [CrossRef]
  141. Wu, X.; Liu, K.; Wu, Q.; Wang, M.; Chen, X.; Li, Y.; Qian, L.; Li, C.; Dai, G.; Zhang, Q.; et al. Biomarkers of Metabolomics in Inflammatory Bowel Disease and Damp-Heat Syndrome: A Preliminary Study. Evid. Based Complement. Altern. Med. 2022, 2022, 3319646. [Google Scholar] [CrossRef]
  142. Vich Vila, A.; Hu, S.; Andreu-Sanchez, S.; Collij, V.; Jansen, B.H.; Augustijn, H.E.; Bolte, L.A.; Ruigrok, R.; Abu-Ali, G.; Giallourakis, C.; et al. Faecal metabolome and its determinants in inflammatory bowel disease. Gut 2023, 72, 1472–1485. [Google Scholar] [CrossRef] [PubMed]
  143. Borren, N.Z.; Plichta, D.; Joshi, A.D.; Bonilla, G.; Sadreyev, R.; Vlamakis, H.; Xavier, R.J.; Ananthakrishnan, A.N. Multi-”-Omics” Profiling in Patients With Quiescent Inflammatory Bowel Disease Identifies Biomarkers Predicting Relapse. Inflamm. Bowel Dis. 2020, 26, 1524–1532. [Google Scholar] [CrossRef] [PubMed]
  144. Rattray, N.J.W.; Trivedi, D.K.; Xu, Y.; Chandola, T.; Johnson, C.H.; Marshall, A.D.; Mekli, K.; Rattray, Z.; Tampubolon, G.; Vanhoutte, B.; et al. Metabolic dysregulation in vitamin E and carnitine shuttle energy mechanisms associate with human frailty. Nat. Commun. 2019, 10, 5027. [Google Scholar] [CrossRef] [PubMed]
  145. Futatsugi, A.; Masuo, Y.; Kawabata, S.; Nakamichi, N.; Kato, Y. L503F variant of carnitine/organic cation transporter 1 efficiently transports metformin and other biguanides. J. Pharm. Pharmacol. 2016, 68, 1160–1169. [Google Scholar] [CrossRef] [PubMed]
  146. Petito, V.; Fidaleo, M.; Pani, G.; Putignani, L.; Gasbarrini, A.; Scaldaferri, F. Tumor necrosis factor-alpha and solute carrier family 22 member 4 gene polymorphisms as potential determinants of intestinal dysbiosis. Dig. Liver. Dis. 2020, 52, 691–693. [Google Scholar] [CrossRef] [PubMed]
  147. Lee, Y.H.; Song, G.G. Pathway analysis of a genome-wide association study of ileal Crohn’s disease. DNA Cell Biol. 2012, 31, 1549–1554. [Google Scholar] [CrossRef]
  148. Xuan, C.; Zhang, B.B.; Yang, T.; Deng, K.F.; Li, M.; Tian, R.J. Association between OCTN1/2 gene polymorphisms (1672C-T, 207G-C) and susceptibility of Crohn’s disease: A meta-analysis. Int. J. Colorectal. Dis. 2012, 27, 11–19. [Google Scholar] [CrossRef]
  149. Lin, Z.; Nelson, L.; Franke, A.; Poritz, L.; Li, T.Y.; Wu, R.; Wang, Y.; MacNeill, C.; Thomas, N.J.; Schreiber, S.; et al. OCTN1 variant L503F is associated with familial and sporadic inflammatory bowel disease. J. Crohns. Colitis. 2010, 4, 132–138. [Google Scholar] [CrossRef]
  150. Repnik, K.; Potocnik, U. Haplotype in the IBD5 region is associated with refractory Crohn’s disease in Slovenian patients and modulates expression of the SLC22A5 gene. J. Gastroenterol. 2011, 46, 1081–1091. [Google Scholar] [CrossRef]
  151. Angelini, S.; Pantaleo, M.A.; Ravegnini, G.; Zenesini, C.; Cavrini, G.; Nannini, M.; Fumagalli, E.; Palassini, E.; Saponara, M.; Di Battista, M.; et al. Polymorphisms in OCTN1 and OCTN2 transporters genes are associated with prolonged time to progression in unresectable gastrointestinal stromal tumours treated with imatinib therapy. Pharmacol. Res. 2013, 68, 1–6. [Google Scholar] [CrossRef]
  152. Ryckman, K.K.; Smith, C.J.; Jelliffe-Pawlowski, L.L.; Momany, A.M.; Berberich, S.L.; Murray, J.C. Metabolic heritability at birth: Implications for chronic disease research. Hum. Genet. 2014, 133, 1049–1057. [Google Scholar] [CrossRef] [PubMed]
  153. Wagner, J.; Sim, W.H.; Ellis, J.A.; Ong, E.K.; Catto-Smith, A.G.; Cameron, D.J.; Bishop, R.F.; Kirkwood, C.D. Interaction of Crohn’s disease susceptibility genes in an Australian paediatric cohort. PLoS ONE 2010, 5, e15376. [Google Scholar] [CrossRef] [PubMed]
  154. Cucchiara, S.; Latiano, A.; Palmieri, O.; Staiano, A.M.; D’Inca, R.; Guariso, G.; Vieni, G.; Rutigliano, V.; Borrelli, O.; Valvano, M.R.; et al. Role of CARD15, DLG5 and OCTN genes polymorphisms in children with inflammatory bowel diseases. World J. Gastroenterol. 2007, 13, 1221–1229. [Google Scholar] [CrossRef] [PubMed]
  155. Torkvist, L.; Noble, C.L.; Lordal, M.; Sjoqvist, U.; Lindforss, U.; Nimmo, E.R.; Lofberg, R.; Russell, R.K.; Satsangi, J. Contribution of the IBD5 locus to Crohn’s disease in the Swedish population. Scand. J. Gastroenterol. 2007, 42, 200–206. [Google Scholar] [CrossRef]
  156. Silverberg, M.S.; Duerr, R.H.; Brant, S.R.; Bromfield, G.; Datta, L.W.; Jani, N.; Kane, S.V.; Rotter, J.I.; Philip Schumm, L.; Hillary Steinhart, A.; et al. Refined genomic localization and ethnic differences observed for the IBD5 association with Crohn’s disease. Eur. J. Hum. Genet. 2007, 15, 328–335. [Google Scholar] [CrossRef] [PubMed]
  157. Dobrowolski, S.F.; McKinney, J.T.; Amat di San Filippo, C.; Giak Sim, K.; Wilcken, B.; Longo, N. Validation of dye-binding/high-resolution thermal denaturation for the identification of mutations in the SLC22A5 gene. Hum. Mutat. 2005, 25, 306–313. [Google Scholar] [CrossRef]
  158. Li, Y.; Chang, M.; Schrodi, S.J.; Callis-Duffin, K.P.; Matsunami, N.; Civello, D.; Bui, N.; Catanese, J.J.; Leppert, M.F.; Krueger, G.G.; et al. The 5q31 variants associated with psoriasis and Crohn’s disease are distinct. Hum. Mol. Genet. 2008, 17, 2978–2985. [Google Scholar] [CrossRef]
  159. Urban, T.J.; Gallagher, R.C.; Brown, C.; Castro, R.A.; Lagpacan, L.L.; Brett, C.M.; Taylor, T.R.; Carlson, E.J.; Ferrin, T.E.; Burchard, E.G.; et al. Functional genetic diversity in the high-affinity carnitine transporter OCTN2 (SLC22A5). Mol. Pharmacol. 2006, 70, 1602–1611. [Google Scholar] [CrossRef]
  160. Li, F.Y.; El-Hattab, A.W.; Bawle, E.V.; Boles, R.G.; Schmitt, E.S.; Scaglia, F.; Wong, L.J. Molecular spectrum of SLC22A5 (OCTN2) gene mutations detected in 143 subjects evaluated for systemic carnitine deficiency. Hum. Mutat. 2010, 31, E1632–E1651. [Google Scholar] [CrossRef]
  161. Lee, N.C.; Tang, N.L.; Chien, Y.H.; Chen, C.A.; Lin, S.J.; Chiu, P.C.; Huang, A.C.; Hwu, W.L. Diagnoses of newborns and mothers with carnitine uptake defects through newborn screening. Mol. Genet. Metab. 2010, 100, 46–50. [Google Scholar] [CrossRef] [PubMed]
  162. Vaz, F.M.; Scholte, H.R.; Ruiter, J.; Hussaarts-Odijk, L.M.; Pereira, R.R.; Schweitzer, S.; de Klerk, J.B.; Waterham, H.R.; Wanders, R.J. Identification of two novel mutations in OCTN2 of three patients with systemic carnitine deficiency. Hum. Genet. 1999, 105, 157–161. [Google Scholar] [CrossRef] [PubMed]
  163. Frigeni, M.; Balakrishnan, B.; Yin, X.; Calderon, F.R.O.; Mao, R.; Pasquali, M.; Longo, N. Functional and molecular studies in primary carnitine deficiency. Hum. Mutat. 2017, 38, 1684–1699. [Google Scholar] [CrossRef] [PubMed]
  164. Burwinkel, B.; Kreuder, J.; Schweitzer, S.; Vorgerd, M.; Gempel, K.; Gerbitz, K.D.; Kilimann, M.W. Carnitine transporter OCTN2 mutations in systemic primary carnitine deficiency: A novel Arg169Gln mutation and a recurrent Arg282ter mutation associated with an unconventional splicing abnormality. Biochem. Biophys. Res. Commun. 1999, 261, 484–487. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, Y.; Taroni, F.; Garavaglia, B.; Longo, N. Functional analysis of mutations in the OCTN2 transporter causing primary carnitine deficiency: Lack of genotype-phenotype correlation. Hum. Mutat. 2000, 16, 401–407. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, Y.; Korman, S.H.; Ye, J.; Gargus, J.J.; Gutman, A.; Taroni, F.; Garavaglia, B.; Longo, N. Phenotype and genotype variation in primary carnitine deficiency. Genet. Med. 2001, 3, 387–392. [Google Scholar] [CrossRef] [PubMed]
  167. El-Hattab, A.W.; Li, F.Y.; Shen, J.; Powell, B.R.; Bawle, E.V.; Adams, D.J.; Wahl, E.; Kobori, J.A.; Graham, B.; Scaglia, F.; et al. Maternal systemic primary carnitine deficiency uncovered by newborn screening: Clinical, biochemical, and molecular aspects. Genet. Med. 2010, 12, 19–24. [Google Scholar] [CrossRef]
  168. Schimmenti, L.A.; Crombez, E.A.; Schwahn, B.C.; Heese, B.A.; Wood, T.C.; Schroer, R.J.; Bentler, K.; Cederbaum, S.; Sarafoglou, K.; McCann, M.; et al. Expanded newborn screening identifies maternal primary carnitine deficiency. Mol. Genet. Metab. 2007, 90, 441–445. [Google Scholar] [CrossRef]
  169. Tang, M.F.; Sy, H.Y.; Kong, A.P.; Ko, F.W.; Wang, S.S.; Liu, T.C.; Chan, W.C.; Wong, G.W.; Hon, K.L.; Chan, J.C.; et al. Genetic effects of multiple asthma loci identified by genomewide association studies on asthma and spirometric indices. Pediatr. Allergy Immunol. 2016, 27, 185–194. [Google Scholar] [CrossRef]
  170. Lee, Y.H.; Bae, S.C.; Kim, J.H.; Seo, Y.H.; Choi, S.J.; Ji, J.D.; Song, G.G. Meta-analysis of SLC22A4 and RUNX1 polymorphisms: Associations with rheumatoid arthritis susceptibility. Z. Rheumatol. 2015, 74, 351–358. [Google Scholar] [CrossRef]
  171. Ren, T.L.; Han, Z.J.; Yang, C.J.; Hang, Y.X.; Fang, D.Y.; Wang, K.; Zhu, X.; Ji, X.J.; Zhou, F.F. Association of SLC22A4 gene polymorphism with Rheumatoid arthritis in the Chinese population. J. Biochem. Mol. Toxicol. 2014, 28, 206–210. [Google Scholar] [CrossRef] [PubMed]
  172. Ding, Y.; Cong, L.; Ionita-Laza, I.; Lo, S.H.; Zheng, T. Constructing gene association networks for rheumatoid arthritis using the backward genotype-trait association (BGTA) algorithm. BMC Proc. 2007, 1 (Suppl. S1), S13. [Google Scholar] [CrossRef] [PubMed]
  173. Jung, J.; Song, J.J.; Kwon, D. Allelic based gene-gene interactions in rheumatoid arthritis. BMC Proc. 2009, 3 (Suppl. S7), S76. [Google Scholar] [CrossRef] [PubMed]
  174. Pawlik, A.; Paradowska-Gorycka, A.; Safranow, K.; Dziedziejko, V.; Dutkiewicz, G.; Slucznowska-Glabowska, S.; Juzyszyn, Z.; Drozdzik, M. SLC22A5 polymorphism associated with risk of extra-articular manifestations in rheumatoid arthritis patients. Reumatologia 2019, 57, 3–7. [Google Scholar] [CrossRef] [PubMed]
  175. Nakahara, S.; Arimura, Y.; Saito, K.; Goto, A.; Motoya, S.; Shinomura, Y.; Miyamoto, A.; Imai, K. Association of SLC22A4/5 polymorphisms with steroid responsiveness of inflammatory bowel disease in Japan. Dis. Colon. Rectum. 2008, 51, 598–603. [Google Scholar] [CrossRef] [PubMed]
  176. Long, G.; Zhang, G.; Zhang, F.; Ye, D.; Yang, D.; Yang, Y. Relationship Between SLC22A1 and SLC22A4 Gene Polymorphisms and Risk of Type 2 Diabetes in Chinese Han Population. Clin. Lab. 2018, 64, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
  177. Weersma, R.K.; Zhou, L.; Nolte, I.M.; van der Steege, G.; van Dullemen, H.M.; Oosterom, E.; Bok, L.; Peppelenbosch, M.P.; Faber, K.N.; Kleibeuker, J.H.; et al. Runt-related transcription factor 3 is associated with ulcerative colitis and shows epistasis with solute carrier family 22, members 4 and 5. Inflamm. Bowel. Dis. 2008, 14, 1615–1622. [Google Scholar] [CrossRef]
  178. Yamase, Y.; Horibe, H.; Ueyama, C.; Fujimaki, T.; Oguri, M.; Kato, K.; Arai, M.; Watanabe, S.; Yamada, Y. Association of TOMM40 and SLC22A4 polymorphisms with ischemic stroke. Biomed. Rep. 2015, 3, 491–498. [Google Scholar] [CrossRef]
  179. Zou, D.; Lou, J.; Ke, J.; Mei, S.; Li, J.; Gong, Y.; Yang, Y.; Zhu, Y.; Tian, J.; Chang, J.; et al. Integrative expression quantitative trait locus-based analysis of colorectal cancer identified a functional polymorphism regulating SLC22A5 expression. Eur. J. Cancer 2018, 93, 1–9. [Google Scholar] [CrossRef]
  180. Sebastian-delaCruz, M.; Olazagoitia-Garmendia, A.; Gonzalez-Moro, I.; Santin, I.; Garcia-Etxebarria, K.; Castellanos-Rubio, A. Implication of m6A mRNA Methylation in Susceptibility to Inflammatory Bowel Disease. Epigenomes 2020, 4, 16. [Google Scholar] [CrossRef]
  181. Prieto-Perez, R.; Solano-Lopez, G.; Cabaleiro, T.; Roman, M.; Ochoa, D.; Talegon, M.; Baniandres, O.; Lopez-Estebaranz, J.L.; de la Cueva, P.; Dauden, E.; et al. Polymorphisms Associated with Age at Onset in Patients with Moderate-to-Severe Plaque Psoriasis. J. Immunol. Res. 2015, 2015, 101879. [Google Scholar] [CrossRef] [PubMed]
  182. de Ridder, L.; Weersma, R.K.; Dijkstra, G.; van der Steege, G.; Benninga, M.A.; Nolte, I.M.; Taminiau, J.A.; Hommes, D.W.; Stokkers, P.C. Genetic susceptibility has a more important role in pediatric-onset Crohn’s disease than in adult-onset Crohn’s disease. Inflamm. Bowel Dis. 2007, 13, 1083–1092. [Google Scholar] [CrossRef] [PubMed]
  183. Rose, E.C.; di San Filippo, C.A.; Ndukwe Erlingsson, U.C.; Ardon, O.; Pasquali, M.; Longo, N. Genotype-phenotype correlation in primary carnitine deficiency. Hum. Mutat. 2012, 33, 118–123. [Google Scholar] [CrossRef] [PubMed]
  184. Koizumi, A.; Nozaki, J.; Ohura, T.; Kayo, T.; Wada, Y.; Nezu, J.; Ohashi, R.; Tamai, I.; Shoji, Y.; Takada, G.; et al. Genetic epidemiology of the carnitine transporter OCTN2 gene in a Japanese population and phenotypic characterization in Japanese pedigrees with primary systemic carnitine deficiency. Hum. Mol. Genet. 1999, 8, 2247–2254. [Google Scholar] [CrossRef] [PubMed]
  185. Jaruskova, M.; Curik, N.; Hercog, R.; Polivkova, V.; Motlova, E.; Benes, V.; Klamova, H.; Pecherkova, P.; Belohlavkova, P.; Vrbacky, F.; et al. Genotypes of SLC22A4 and SLC22A5 regulatory loci are predictive of the response of chronic myeloid leukemia patients to imatinib treatment. J. Exp. Clin. Cancer Res. 2017, 36, 55. [Google Scholar] [CrossRef] [PubMed]
  186. Makhseed, N.; Vallance, H.D.; Potter, M.; Waters, P.J.; Wong, L.T.; Lillquist, Y.; Pasquali, M.; Amat di San Filippo, C.; Longo, N. Carnitine transporter defect due to a novel mutation in the SLC22A5 gene presenting with peripheral neuropathy. J. Inherit. Metab. Dis. 2004, 27, 778–780. [Google Scholar] [CrossRef] [PubMed]
  187. Mayatepek, E.; Nezu, J.; Tamai, I.; Oku, A.; Katsura, M.; Shimane, M.; Tsuji, A. Two novel missense mutations of the OCTN2 gene (W283R and V446F) in a patient with primary systemic carnitine deficiency. Hum. Mutat. 2000, 15, 118. [Google Scholar] [CrossRef]
  188. Ben Said, M.; Grati, M.; Ishimoto, T.; Zou, B.; Chakchouk, I.; Ma, Q.; Yao, Q.; Hammami, B.; Yan, D.; Mittal, R.; et al. A mutation in SLC22A4 encoding an organic cation transporter expressed in the cochlea strial endothelium causes human recessive non-syndromic hearing loss DFNB60. Hum. Genet. 2016, 135, 513–524. [Google Scholar] [CrossRef]
Figure 1. Major tissue localization of OCTNs and role in transport of inflammation-related metabolites. Intestine epithelial cell on left and kidney epithelial cell on right are depicted with brush border membranes; airway cell on top, nervous tissue and immune cells at bottom; blood–brain barrier (BBB) is represented as pink barrier. Exo, exosome. Sketch of some human or gut bacteria pathways are depicted: TMA, trimethylamine; TMAO, trimethylamine oxide; ChAT, choline acetyltransferase. OCTNs in dotted lines represent controversial localization [20,21]; Ergo, ergothioneine; Ach, acetylcholine; Cho, choline; Acar, acetylcarnitine; Bet, betaine; γ-BB, γ-butyrobetaine; OC, organic cation. Arrows indicate transport direction. Thin arrows refer to low-affinity transport.
Figure 1. Major tissue localization of OCTNs and role in transport of inflammation-related metabolites. Intestine epithelial cell on left and kidney epithelial cell on right are depicted with brush border membranes; airway cell on top, nervous tissue and immune cells at bottom; blood–brain barrier (BBB) is represented as pink barrier. Exo, exosome. Sketch of some human or gut bacteria pathways are depicted: TMA, trimethylamine; TMAO, trimethylamine oxide; ChAT, choline acetyltransferase. OCTNs in dotted lines represent controversial localization [20,21]; Ergo, ergothioneine; Ach, acetylcholine; Cho, choline; Acar, acetylcarnitine; Bet, betaine; γ-BB, γ-butyrobetaine; OC, organic cation. Arrows indicate transport direction. Thin arrows refer to low-affinity transport.
Biomolecules 14 00392 g001
Figure 2. A comparison of OCTNs’ structural features. (A) The lateral view of the AlphaFold structures of human OCTN1 (SLC22A4) and OCTN2 (SLC22A5) are depicted in light blue using a ribbon representation. Cysteine residues are highlighted in yellow using a space-filled representation; the dotted red circles highlight the four cysteines in the extracellular loop. The putative ATP binding site is shown in red. (B) Amino acid sequences of human OCTNs. Conserved amino acid residues among members are indicated by stars.
Figure 2. A comparison of OCTNs’ structural features. (A) The lateral view of the AlphaFold structures of human OCTN1 (SLC22A4) and OCTN2 (SLC22A5) are depicted in light blue using a ribbon representation. Cysteine residues are highlighted in yellow using a space-filled representation; the dotted red circles highlight the four cysteines in the extracellular loop. The putative ATP binding site is shown in red. (B) Amino acid sequences of human OCTNs. Conserved amino acid residues among members are indicated by stars.
Biomolecules 14 00392 g002
Figure 3. Modulation of OCTN expression. Cells of indicated tissues are surrounded by grey boxes. Arrows indicate gene/protein upregulation (green) or downregulation (red).
Figure 3. Modulation of OCTN expression. Cells of indicated tissues are surrounded by grey boxes. Arrows indicate gene/protein upregulation (green) or downregulation (red).
Biomolecules 14 00392 g003
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

Pochini, L.; Galluccio, M.; Console, L.; Scalise, M.; Eberini, I.; Indiveri, C. Inflammation and Organic Cation Transporters Novel (OCTNs). Biomolecules 2024, 14, 392. https://doi.org/10.3390/biom14040392

AMA Style

Pochini L, Galluccio M, Console L, Scalise M, Eberini I, Indiveri C. Inflammation and Organic Cation Transporters Novel (OCTNs). Biomolecules. 2024; 14(4):392. https://doi.org/10.3390/biom14040392

Chicago/Turabian Style

Pochini, Lorena, Michele Galluccio, Lara Console, Mariafrancesca Scalise, Ivano Eberini, and Cesare Indiveri. 2024. "Inflammation and Organic Cation Transporters Novel (OCTNs)" Biomolecules 14, no. 4: 392. https://doi.org/10.3390/biom14040392

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

Article Metrics

Back to TopTop