- Review
- Open access
- Published:
Modulating autism spectrum disorder pathophysiology using a trace amine-focused approach: targeting the gut
Molecular Medicine volume 31, Article number: 198 (2025)
Abstract
Autism spectrum disorder (ASD) affects approximately 1% of the population directly, but also a much higher proportion (family and caregivers) indirectly. Although ASD is characterized by high prevalence of anxiety and poor gastrointestinal health, current treatment strategies are mainly focused on neurological symptomatic treatment, with little to no attention to gut health. Furthermore, many psychiatric drugs used for management of secondary neurological symptoms, are known to exacerbate gut health issues and neurological dysregulation across the gut-brain axis.
Trace amines are neurotransmitter-like substances synthesized endogenously in the human brain – in trace amounts – but also in high abundance by the microbiome. Emerging evidence suggests dysregulation of the trace amine system in ASD. Since trace aminergic signalling is central to regulatory system homeostasis, we hypothesize targeting this system in the ASD context. Given the various sources of trace amines, we suggest that normalization of functional dysbiosis in terms of trace aminergic signalling – rather than microbial compositional dysbiosis – should be a focus in medicines development. In addition, a holistic consideration including also other factors at play in determining trace aminergic signalling outcome – such as receptor binding, enzymatic role players, etc. – is required to fully elucidate and therapeutically modify the pathophysiology of regulatory systems implicated in ASD.
This review firstly provides a brief overview of trace amine dysregulation in ASD for context. Secondly, we formulate our hypothesis on how this may therapeutically address symptomology, with consideration of cellular and molecular mechanism interplay across the gut-brain axis. Finally, we provide a critical assessment of advances in therapeutics development and drug re-purposing, gaps in knowledge and priorities for medicines development going forward.
Introduction
Despite the fact that non-neurological pathophysiological concerns such as poor gastrointestinal (GI) health are reported in the majority of individuals with autism spectrum disorder (ASD) (Wang et al. 2022a), these are not currently prioritised in the management of ASD. Current treatment strategies are limited to a combination of behavioural, educational, and pharmaceutical therapies for the treatment of neurological symptoms and neurological co-morbidities (anxiety, depression, ADHD). However, commonly prescribed pharmaceuticals, such as risperidone and aripiprazole, may further compromise gut health (Aishworiya et al. 2022), possibly due to their effect to alter gut microbiota composition (Bretler et al. 2019) thereby inducing or exacerbating gut dysbiosis (Angoa-Pérez and Kuhn 2021; Rukavishnikov et al. 2023; Seeman 2021) – although heterogenous study designs have limited conclusions on exact mechanisms at play (Dias et al. 2024). Nevertheless, altered microbial composition and microbially derived metabolite actions in the gut may in turn exacerbate neurological symptoms via gut-brain signalling, suggesting that ASD-associated gut-brain axis alternations can contribute to an ASD phenotype. Indeed, a functional gut-brain axis architecture has been correlated to ASD phenotypic heterogeneity and is characterized by altered macronutrient metabolism that correlates to restrictive dietary patterns, pro-inflammatory cytokine profile and changes in brain-derived gene expression profiles (Morton et al. 2023). This fact emphasises that consideration of gut health should be prioritised in ASD. In addition, these studies (Angoa-Pérez and Kuhn 2021; Rukavishnikov et al. 2023; Seeman 2021) acknowledge that a relative gut dysbiosis may affect the pharmacokinetic profile and efficacy of these drugs.
Acknowledging the heterogeneity of ASD aetiology and symptomology, it would be naïve to propose that a single biochemical pathway may be responsible for the full spectrum of ASD disease pathology. However, we have identified the trace aminergic system, which functions at the level of both the brain and gut (albeit at different levels of abundance), as potential target for modulation in the context of ASD. For example, an altered trace amine load (due to altered microbial composition or consumption of trace amine rich foods) and downstream trace aminergic signalling is anticipated in the ASD context but remains to be confirmed. Importantly, trace aminergic signalling comprises many role players (e.g. trace amines, their receptors and metabolic enzymes) and seems to contribute an additional level of regulation to multiple major regulatory process in the body, such as redox status, inflammatory pathways and neuronal function (Gwilt et al. 2020). While modulation of trace amine signalling is unlikely to modify the genetic foundation of ASD pathology, it may significantly impact psychological and peripheral symptomology, ultimately enhancing overall quality of life and functionality of individuals with ASD.
The purpose of this hypothesis paper is to explore the avenues by which modulation of trace amine signalling may have therapeutic benefit in the ASD context. We first provide a brief summary of known ASD pathologies (neurological, gastrointestinal and mitochondrial). We then provide an integrated discussion of the available literature informing on the potential involvement of aberrant trace amine signalling in the context of ASD symptomology. Finally, we formulate our hypothesis on potential targets for therapeutic intervention in this context and make recommendations for research priorities going forward.
ASD pathology
Neurological pathology in ASD
As summarised in Table 1, abnormalities in neuronal connectivity and synaptogenesis may to a large extent account for the primary pathophysiology of ASD. However, the relative contribution of neurotransmitter system abnormalities to primary disease outcome remains to be confirmed. Nevertheless, the most consistent changes related to ASD-associated behaviours seem to be associated with alterations in serotonergic and dopaminergic signalling, but links to other neurotransmitters also exist. For example, decreased gamma-aminobutyric acid (GABA) levels align with the high incidence of anxiety in ASD (Lever and Geurts 2016; Simonoff et al. 2022; Wang et al. 2018a), as well as incidence of seizure (Frye et al. 2016; Tallarico et al. 2023) and mitochondrial functional abnormalities (Frye et al. 2019) in ASD. Importantly, limited available data seems to suggest that central (brain) and peripheral (e.g. circulation, gut) neurotransmitter profiles may differ e.g. evidence for low central vs. high peripheral 5-HT levels (Muller et al. 2016). This suggests another level of (or differential) regulation of neuronal signaling in the peripheral vs. central compartments, but insufficient information limits firm conclusions at this point.
Mitochondrial dysfunction as central mechanism
There is increasing evidence suggesting that neurodevelopmental disorders are affected by mitochondrial dysfunction and the resulting increased oxidative stress load (Anitha et al. 2023; Wang et al. 2022b). Abnormal mitochondrial biomarkers have been reported in 30–50% of individuals with ASD, while the reported prevalence of electron transport chain (ETC) abnormalities in immune cells are as high as 80% (Frye et al. 2019; Giulivi et al. 2010; Napoli et al. 2014; Rossignol and Frye 2012). Importantly, these characteristic mitochondrial functional abnormalities have been demonstrated in human ASD studies (summarised in Table 2) and reflect a generally reduced mitochondrial respiration and functional capacity. Unfortunately, a relative paucity of data exists on mitochondrial function in ASD specifically. This is a significant oversight, since not all data generated in the brain vs. the periphery align (Table 2). As such, a more holistic profile of mitochondrial dysfunction across compartments may significantly contribute to our understanding of ASD as mitochondrial disease, as well as identification of therapeutic targets.
Furthermore, the contribution of mitochondrial dysfunction to poor GI health in ASD is well-recognised (Rose et al. 2018a). Mitochondria were suggested to be the biological link between environmental stressors (toxicants, medications, microbial secretory products) and enterocyte dysfunction (Frye et al. 2015). In this case – in addition to genetically determined mitochondrial abnormalities in ASD (Frye et al. 2024) – the high environmental challenge endured by the gut enterocytes specifically, could further exacerbate mitochondrial dysfunction, rendering the enterocytes even less functional (Basivireddya et al. 2002; Guerbette et al. 2022; Madsen et al. 1995; Moschandrea et al. 2024). While evidence is not yet conclusive, increased intestinal permeability in patients with ASD has been suggested by several groups (de Magistris et al. 2010; Esnafoglu et al. 2017; Teskey et al. 2021). This would result in a higher antigenic load and increased immune activation, contributing to low-grade GI and systemic inflammation. Clearly, mitochondrial function (and its normalisation) should be an important consideration in ASD management.
ASD-associated gastrointestinal dysfunction
We present a summary of mechanisms likely to be involved in ASD-associated GI dysfunction in Fig. 1. As illustrated, ASD-associated poor gut health is the net outcome of interplay of multiple contributors. For example, central nervous system (CNS)-relevant gene mutations have been proposed to affect the development of the enteric nervous system (ENS) in ASD (Bernier et al. 2014; Campbell et al. 2009; Fröhlich et al. 2019; Hosie et al. 2019; James et al. 2019; Leembruggen et al. 2020; Margolis et al. 2016), impacting its structure and function throughout life (Niesler and Rappold 2021). Of particular interest, ASD-associated serotonin transporter (SERT) polymorphisms are likely to disrupt serotonin (5-HT) metabolism and alter the regulation of mucosal immune responses (Shajib et al. 2017), as well as the activation of enteric reflexes that underlie gut motility, secretion, and sensation (Mawe and Hoffman 2013). However, not all individuals with these mutations exhibit GI dysfunction, aligning with the notion of genetic and environmental interplay in aetiology of poor gut health in ASD.
Potential pathogenesis of gastrointestinal dysfunction in autism spectrum disorder (ASD) as extrapolated from non-ASD literature. Abbreviations: ENS, enteric nervous system; GIT, gastrointestinal tract; MET, mesenchymal-epithelial transition factor. Red ‘X’ indicates genetic abnormalities that result in altered structure and/or function
Moving on to diet, individuals with ASD present with significant food aversions and selectivity that typically lead them to consume a diet high in carbohydrates and processed foods (Cermak et al. 2010; Esposito et al. 2023). An unbalanced diet exacerbate GI symptomology (Valenzuela-Zamora et al. 2022) primarily via disruption of gut microbiome homeostasis (Singh et al. 2017; Zhang 2022), resulting in altered gut microbial composition and secretome profiles (Coretti et al. 2018; De Angelis et al. 2013; Finegold et al. 2002; Hughes et al. 2018; Iglesias-Vazquez et al. 2020; Wang et al. 2012). As previously mentioned, the balance of microbial secretory products and their metabolites, may significantly impact mitochondrial function, and thus GI function. Furthermore, many of these microbial secretory products – specifically trace amines – are also produced endogenously in the brain, where they play important roles in neurological function. We believe that going forward, a gut-brain approach – and with that, trace amine signalling in particular – should be a major focus in the design of therapeutic strategies in ASD.
Interconnectedness of brain and gut in ASD: a role for trace amines
While the benefit of several therapeutic strategies (prebiotic/probiotic therapy, faecal microbial transplant and dietary interventions) aimed at regulating gut microbial composition remains to be conclusively proven (Fattorusso et al. 2019; Geier et al. 2011; Grimaldi et al. 2017; Grossi et al. 2016; Kang et al. 2013; Ooi et al. 2015), we propose a somewhat different approach to gut-brain modulation in ASD, shifting the focus from compositional to functional dysbiosis. In line with this, metabolomic investigations of serum, faeces and urine have revealed altered abundance of a number of molecules in ASD that are of microbial origin (Needham et al. 2021; Sharon et al. 2019). Furthermore, many individuals with ASD present with metabolic imbalances, immune dysregulation, GI dysfunction and altered gut-brain signalling, all of which are significantly impacted by gut microbial metabolites – these include both non-trace amine (e.g. short chain fatty acids (SCFA) originating from bacterial fermentation of dietary carbohydrates, (Rogers et al. 2016)) and trace aminergic role players (e.g. ρ-tyramine, β-phenethylamine and tryptamine) (Gwilt et al. 2020). This emphasises the potential impact of gut microbial secretory products (and thus a functional dysbiosis) on ASD pathophysiological outcome.
Since trace amines are involved in the regulation of several pertinent contributors to comorbid ASD pathophysiology, altered trace aminergic signalling – and the therapeutic normalisation thereof – will be discussed in gut-vs-brain and redox/mitochondrial contexts below.
Trace aminergic signalling pathways of relevance to ASD
Currently, due to a relative paucity of information, firm conclusions in terms of potential trace amine-based causality in primary ASD pathology is not possible, and thus this is not the topic of this review. Rather, the focus will be on the potential to therapeutically target trace amine signalling for improvement of symptomology of ASD across the gut-brain axis.
Modulation of neurotransmitter signalling pathways
Given the interconnectedness of neurotransmitter and trace amine metabolic pathways, altered trace amine metabolism may significantly affect neurotransmitter flux. This is illustrated in Fig. 2, where substantial overlap in the regulation of neurotransmitter signalling by the trace aminergic system exists in both brain and gut compartments, suggesting a potential for therapeutic intervention at both sites via trace aminergic modulation. For example, metabolomics of ASD rodent liver samples (as proxy for general metabolic status) reported increased intensity of β-phenethylamine (β-PEA) (Cao et al. 2023), which – given the common precursor (L-phenylalanine) for β-PEA and dopamine (DA) – may suggest altered substrate availability for DA synthesis and subsequent activity, although correlations between liver and central levels and the implications for dopaminergic activity in ASD remains to be confirmed. In addition, DA synthesis is also dependent on ρ-tyramine metabolism, from their shared precursor, L-tyrosine. Although dysregulated ρ-tyramine levels have not been reported in ASD, a slight increase in faecal m-tyramine (a catabolite downstream from DA) was reported for ASD vs. typically developed controls (Needham et al. 2021), suggesting altered dopaminergic metabolism. Also, ρ-tyramine metabolites octopamine and synephrine have been linked to the ASD hallmarks aggression (Jia et al. 2021; Sherer et al. 2020) and adverse redox outcome (Ribeiro et al. 2019), respectively. This suggests potential alterations in ρ-tyramine flux towards production of octopamine and synephrine rather than DA (see Fig. 2) may indicate another avenue for dysregulation at the cost of DA synthesis in ASD. Furthermore, trace amine associated receptor (TAAR)-1 agonism (functions of both β-PEA and ρ-tyramine) is known to reduce presynaptic DA signalling via TAAR1/dopamine-D2 receptor (DD2R) dimerization-induced inhibition of presynaptic DA release and reuptake via dopamine transporter (DAT) (Halff et al. 2023). Given that ASD is characterised by reduced DA signalling, increased TAAR1 activation – and subsequent TAAR1/DD2R dimerization – may therefore play a role in altered signalling cascades in ASD. Moreover, TAAR1 is expressed in nearly all immune and neuroimmune cells (Fleischer et al. 2018) and its activation and/or dimerization with TAAR2 has been linked to peripheral pro-inflammatory outcome (Babusyte et al. 2013; Panas et al. 2012), accumulation of intracellular calcium and adverse redox outcome (Moiseenko et al. 2024). Thus, skewing of phenylalanine and tyrosine metabolism to favour β-PEA and ρ-tyramine signalling, may contribute significantly to the neurological, oxidative and pro-inflammatory characteristics of ASD.
The intricate nature of dysregulation at this level, was further illustrated in a rodent valproic acid (VPA)-model for ASD, which reported the “dopaminergic hypofunction” effect to be TAAR1- and sex-specific, with females reportedly less affected, which is aligned with the higher male prevalence of ASD reported clinically (Hara et al. 2015). In line with this, oestradiol was recently reported to attenuate ρ-tyramine-associated GI dysregulation in the context of intestinal inflammation (Pretorius and Smith 2023). While it remained to be confirmed how these direct actions of exogenous oestradiol (to reduce ρ-tyramine-associated inflammation and oxidative stress and improve tight junction protein expression) in a gut context may connect to reduced female vs. male prevalence of ASD, a lower probability of females to develop intestinal permeability has been postulated as an etiological mechanism that could be attributed to the protective effect of oestrogen in the ASD context (El-Ansary et al. 2020). In addition to this, the effect of oestradiol to affect the sex dimorphism of ASD through sex-dependent microbially-derived metabolite production has also been suggested (Hao et al. 2020). These data may suggest that females may be protected from ASD outcomes associated with ρ-tyramine or its metabolites, although this remains to be confirmed. Importantly, the interplay of hormones and trace amine signalling warrants further investigation - especially given the relative lack of inclusion of females in mechanistic studies until relatively recently – and as recently implied (Hokanson et al. 2024), this may indicate that ASD in males and females may require different treatment approaches to address different therapeutic targets, and as such considering the impact of sex in ASD treatment is imperative.
Proposed trace aminergic regulation in the brain (A) and gut (B), as it relates to ASD-related pathology and symptomology. Information in frame A reflects data from human studies only, but due to the paucity of human data on the gut, information in frame B was also summarised from preclinical studies. The trace aminergic system is represented in purple, with known ASD pathologies indicated with the use of red arrows. All enzymes are in grey. Abbreviations: ENS, enteric nervous system; 5-HT, serotonin; TAAR, trace amine-associated receptor; AADC, aromatic L-amino acid decarboxylase; MAO-A/B, monoamine oxidase-A/B; 5-HTP, 5-hydroxytryptophan; TPH1/2, tryptophan hydroxylase 1/2; L-Trp, L-tryptophan; L-Tyr, L-tyrosine; L-Phe, L-phenylalanine; TRP, tryptamine; r-TYR, r-tyramine; β-PEA, β-phenethylamine; GI, gastrointestinal; TA, trace amine; AA, amino acid; ASD, autism spectrum disorder; DMT, N,N-Dimethyltryptamine; NMTRP, N-methyltryptamine; IAA, Indoleacetic acid; INMT, indolethylamine-N-methyltransferase; SERT, serotonin transporter; 5-HT1A/2AR, serotonin 1 A/2A receptor; 5-HIAA, 5-hydroxyindoleacetic acid; PAH, phenylalanine hydroxylase; TH, tyrosine hydroxylase; PAA, phenylacetic acid; NMPEA, N-methylphenethylamine; NMTYR, N-methyltyramine; PNMT, phenylethanolamine N-methyltransferase; OCT, octopamine; SYN, synephrine; HPA, 4-hydroxyphenylacetaldehyde; DBH, dopamine beta-hydroxylase; m-TYR/3-MT, 3-Methoxytyramine; COMT, catechol-O-methyltransferase; DAT, dopamine transporter; HVA, homovanillic acid; L-DOPA, levodopa; DDC, DOPA decarboxylase; DA, dopamine; VMAT2, vesicular monoamine transporter 2; DD2R, dopamine receptor D2; βARR2, beta-arrestin-2; ECC, enterochromaffin
Furthermore, although decreased 5-HT signalling has long been established in ASD, very little progress has been made in terms of therapeutics development (Pourhamzeh et al. 2022), highlighting the complexity of dysregulation in this system. Trace amines may also contribute to ASD symptomology here. For example, pineal dysfunction has been implicated in ASD; apart from accounting for the reduced melatonin levels reported in ASD, abnormal pineal metabolism of N, N-dimethyltryptamine (DMT) - a breakdown product of the trace amine tryptamine and thought to act via both TAARs and serotonin 2 A receptor (5-HT2AR) - may be responsible for DMT accumulation, which in turn results in abnormal neurogenesis, including the aberrant synaptic organisation characteristic of ASD (Shomrat and Nesher 2019). This suggests potential alterations in L-tryptophan flux towards production of tryptamine and DMT rather the 5-HT in ASD (see Fig. 2) that may at least in part, underpin some of the central serotonergic abnormalities associated with ASD, although this remains to be confirmed. However, this outcome contrasts the anxiolytic benefit ascribed to low level DMT in human studies (Jacob and Presti 2005) and again highlights the requirement for intricate regulation of absolute levels of any of the trace amines, to avoid adverse outcome.
Moreover, altered trace amine levels have been reported in the context of common neurological ASD comorbidities. Of interest, seemingly contradictory roles have been reported for β-PEA. In addition to its undesired link to ASD symptomology already mentioned, both ADHD and depression are associated with decreased urinary β-PEA and its major metabolite phenylacetic acid (Baker et al. 1991; Sabelli and Javaid 1995; Zametkin et al. 1984), while β-PEA supplementation ameliorated corticosterone-induced depression in a murine model (Lee et al. 2020). These data suggest that modulatory effects of β-PEA are dose- or receptor-dependent. Given the complexity of ASD in terms of severity spectrum and heterogeneity of comorbid symptomology, causality if unlikely to rest with one dysregulated trace amine. Unfortunately, limited parameter panel and sampling diversity used for trace amine quantification, limits interpretation on dysregulation of the balance between different role players at pathway or tissue compartment levels, or how these limited observations in one sample type (e.g. urine or faeces) may relate to pathology elsewhere (e.g. brain). Thus, it remains unclear whether or not trace amines are accurate biomarkers of ASD pathology or comorbid symptomology. Nevertheless, these studies support the notion that neurotransmitter signalling may be normalised via modulation of trace amine signalling.
Modulation of gut function
Moving on to the gut context, the notion that trace amines are physiologically active in the gut is strongly supported by faecal metabolomic studies reporting trace amines at relatively high (micromolar) levels (Jacobs et al. 2016; Kisuse et al. 2018; Ponnusamy et al. 2011; Santoru et al. 2017; Zhai et al. 2023), as well as by data from several in vivo and ex vivo studies demonstrating altered gut function (e.g. increased gut transit via promotion of colonic ion and 5-HT secretion) after supplementation with either trace amines at physiologically relevant concentrations, or specific trace amine-producing bacteria (Bhattarai et al. 2018; Williams et al. 2014; Yano et al. 2015; Zhai et al. 2023). Since modulation of intestinal secretion and motility can have profound effects on factors that affect intestinal homeostasis (luminal pH, mucosal immune response, and delivery of nutrients to gut microbes and host enterocytes), it is not surprising that many GI disorders (with high prevalences of comorbid neuropsychiatric symptomologies) report altered faecal trace amine levels. For example, in various irritable bowel syndrome (IBS) cohorts, levels of β-PEA have been reported as increased (Zhai et al. 2023) or decreased (Han et al. 2022), while faecal tryptamine levels seem consistently increased (Ponnusamy et al. 2011; Zhai et al. 2023). Moreover, in patients with inflammatory bowel disease (IBD), increased levels of β-PEA, putrescine and cadaverine have been reported in a Crohn’s disease sub-group (Jacobs et al. 2016; Santoru et al. 2017), while increased levels of cadaverine, trimethylamine-N-oxide and ρ-tyramine were reported in the ulcerative colitis sub-group (Santoru et al. 2017). It is important to note that changes in trace amine levels have not only been associated with changes in specific bacterial groups, but also disease severity and even co-morbid symptomology. Indeed, in patients with diarrhoea-predominant IBS, elevated levels of faecal β-PEA and tryptamine were associated with severity of diarrhoeal symptoms, while elevated peripheral 5-HT levels (induced via β-PEA/tryptamine-TAAR1 signalling) directly implicates altered trace aminergic signalling in IBS pathology (Zhai et al. 2023). In addition, tryptamine levels were reportedly elevated in an IBS group with co-morbid depression (approximately 30% of the IBS cohort) when compared to healthy controls (Han et al. 2022), with tryptamine/tryptophan ratio relatively increased with increased depression severity in IBS patients, suggesting that co-morbid neurological disorders may be exacerbated by specific alterations in trace amine load in the gut. Of significance in the medicines development context, a rescue effect (via experimental TAAR-1 inhibition) was demonstrated in pseudo germ-free mice colonized with diarrhoea-predominant IBS (IBS-D)-derived microbiota (Zhai et al. 2023), suggesting that normalisation of trace aminergic signalling (and thus also neurotransmitter signalling) can occur despite microbial compositional dysbiosis. While these specific pathways have not yet been investigated in ASD, elevated blood 5-HT was the first biomarker identified in autism research (reviewed by (Gabriele et al. 2014), suggesting that a related pathology may exist to increase peripheral 5-HT and cause GI symptomology. Moreover, given the important role of 5-HT in neuronal formation and presynaptic balance (keeping in mind that ASD is associated with neuronal malformations and dysregulated neurotransmission), the contribution of altered ENS signalling or metabolite load from gut-associated changes cannot be underplayed in the ASD context.
In the context of immune system activation (and thus inflammatory and redox outcome), altered trace amine homeostasis has already been hypothesised to result in hyperactivity of the immune system, specifically mucosal immunity (Christian and Berry 2018) – which is known to be dysregulated in ASD when co-morbid GI symptomologies are also present (Ashwood et al. 2004; Ashwood and Wakefield 2006; Butera et al. 2024; Rose et al. 2018a). Again, these studies suggest that trace amine homeostasis is crucial for GI homeostasis. However, even in ASD individuals without clinical GI symptoms, low level dysregulation of cellular processes such as mitochondrial function would still be present in the gut, which may benefit from trace amine modulation.
Targeting mitochondria
In the context of another common comorbid ASD symptom – anxiety – the role of trace amines in determining redox outcome is clearly illustrated. For example, the role of agmatine (plasma levels of which are decreased in ASD (Esnafoglu and Irende 2018) and its metabolites in the regulation of mitochondrial function, is illustrated in Fig. 3. In the context of ASD specifically, murine gestational agmatine treatment was shown to normalise autistic behaviour – including anxiety – in offspring (Chen et al. 2024), which was associated with increased dephosphorylation of ERK and CREB in the ERK/CREB/BDNF pathway. In a non-autism model, agmatine was shown to reduce anxiety via its inhibition of nitric oxide synthase (Gawali et al. 2017), suggesting a redox-associated mechanism. In line with this, in a rodent VPA model of autism, both prenatal and postnatal acute agmatine administration were recently reported to rescue impaired social behaviour in male offspring (Khoram-Abadi et al. 2024; Kim et al. 2017). While no benefit was observed in terms of plasma total antioxidant capacity, reduced malonaldehyde activity were reported in this model, pointing toward some redox benefit. In line with this, agmatine administration in ASD has been shown to influence cellular energy metabolism to decrease glycolysis in favour of oxidative mechanisms, increasing mitochondrial activity (Hipkiss 2018).
Trace aminergic regulation in the mitochondria, as it relates to ASD-related pathology and symptomology. Mitochondrial location of enzymes is indicted with a blue asterisk (*). Red font indicates abnormalities associated with ASD. Abbreviations: ARG, L-arginine; ARG I / II, arginase I / II; ORN, ornithine; PUT, putrescine; AGM, agmatine; ODC, ornithine decarboxylase; PAO, polyamine oxidase; SSAT, spermidine/spermine-N(1)-acetyltransferase; SPDS, spermidine synthase; SPMS, spermine synthase; SMOX, spermine oxidase; AGMAT, agmatinase; SPM, spermine; SPD, spermidine; TA, trace amines; MOA, monoamine oxidase; ROS, reactive oxygen species; SOD 1 / 2, superoxide dismutase 1 / 2; PrxR, peroxiredoxin reductase; Prx(SH)2, reduced peroxiredoxin; Prx-S2, oxidized peroxiredoxin; CAT, catalase; GSSG, glutathione disulfide; GSH, glutathione; GR, glutathione reductase; NAD+, oxidized Nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate; Trx-S2, oxidized thioredoxin; Trx(SH)2, reduced thioredoxin; TrxR, thioredoxin reductase; mPTP, mitochondrial permeability transition pore; I – V, mitochondrial electron transport chain complex I – V; Cyt, c – cytochrome c; ATP, adenosine triphosphate; ADP, ddenosine diphosphate; NADH, reduced nicotinamide-adenine dinucleotide; FADH2, reduced flavin adenine dinucleotide; FAD, flavin adenine dinucleotide; β-PEA, β-phenylethylamine; QH2, dihydroxyquinone; CoQ, coenzyme Q; GPx, glutathione peroxidase; GR, glutathione reductase; ∆Ψ, mitochondrial membrane potential
As discussed for β-PEA, the requirement for fine control of absolute levels of trace amines – and their metabolites – is also evident from the agmatine literature. For example, in a model of putrescine depletion, lower spermidine/spermine and glutamate/GABA ratios were also strongly associated with anxiety-like behaviour (Gupta et al. 2009). In the same study, higher spermidine levels were associated with good water maze performances and decreased anxiety, whereas spermine had the opposite effect, suggesting differential roles for spermidine and spermine in behavioural outcome. In contrast, an increased spermidine/spermine ratio (resulting from a spermine synthase gene mutation in mice) was linked to increased anxiety, downregulated mitochondrial oxidative phosphorylation and ribosomal protein synthesis in the cerebral cortex, as well as impaired mitochondrial bioenergetics in isolated primary fibroblasts (Akinyele et al. 2024), emphasizing the differential dose-dependent effect of and importance of regulation of these polyamines on behavioural and functional outcome. While these studies suggest that conversion of agmatine into its downstream metabolites may be the key to beneficial outcome, more research is required to understand homeostatic thresholds that result in differential outcomes.
Similarly, in the context of intestinal inflammation, while agmatine administration in human intestinal cell cultures – where normal clearance of agmatine via conversion into its metabolites were likely suboptimal – had detrimental effects on endothelial cell health and barrier integrity (Pretorius and Smith 2022), the same treatment showed protective benefit in an in vivo model – where pathways to metabolise agmatine were intact (Pretorius and Smith 2023). Together, these reports highlight the importance of considering not only the levels of trace amines themselves in development of therapeutics, but also the levels and activity of the enzymes involved in their synthesis or metabolism, as well as the abundance of their downstream metabolites. To our knowledge, potential dysregulation of trace amine-relevant enzymes has not been studied in the context of ASD.
Regarding cellular redox status, it is well known that polyamines contribute to cellular oxidative balance (Lenis et al. 2017). On the one hand, spermine and spermidine reportedly mediate protection against oxidative damage (Belle et al. 2004; Rider et al. 2007), while on the other hand elevated levels of polyamines seem to have toxic effects and are associated with several diseases (Agostinelli 2020; Fiori and Turecki 2008). The basis of this toxicity is likely oxidative stress/damage caused by the production of reactive species (such as hydrogen peroxide and acrolein) during polyamine catabolism. Similarly, the effects of agmatine on cellular redox status and mitochondrial functionality seem to be dependent on a homeostatic balance and regulation of its pleiotropic effects. For example, agmatine has been related to neuroprotective outcomes (Kotagale et al. 2019) via reduction in oxidative stress and preservation of mitochondrial integrity and function (Condello et al. 2011; El-Sayed et al. 2019). However, a hormetic effect of agmatine on the mitochondrial permeability transition mechanism has been reported in rat liver samples (Martinis et al. 2020), with mitochondrial permeability transition induced by low concentrations but inhibited by high concentrations. This suggests a very complex role of agmatine on mitochondrial physiology but emphasises the need for homeostatic regulation to achieve cellular redox homeostasis.
A further consideration is that some trace amines are metabolised by monoamine oxidase (MAO) enzymes, a catabolism process that also produces hydrogen peroxide as byproduct (Bortolato et al. 2008). In healthy cells, this oxidizing agent is neutralised by catalase or glutathione peroxidase into non-toxic H2O. However, any abnormality in antioxidant capacity or activity (such as in diseased states), could lead to oxidative stress-induced cellular damage. Since MAO is mitochondrial bound, mitochondria may be subject to excessive reactive oxygen species if increased trace amine metabolic flux were to occur (i.e. in states of increase trace amine load). This, in conjunction with the fact that MAO inhibition seems to prevent mitochondrial dysfunction (Sorato et al. 2014; Venegas et al. 2024), suggests that altered trace amine metabolism may contribute to, or exacerbate, mitochondrial dysfunction. In addition, dose-dependent effects of β-PEA to generate hydroxyl radicals in vitro and in vivo via significant inhibition of NADH-ubiquinone oxidoreductase activity have been reported (Sengupta and Mohanakumar 2010. This suggests that β-PEA-induced inhibition of oxidative phosphorylation may result in excess hydroxyl radical generation (Sengupta and Mohanakumar 2010) and strengthens the point that altered trace amine load and subsequent metabolism may contribute to mitochondrial dysfunction.
Trace amine-focused therapeutics development: do we know enough?
From the reviewed literature, it is clear that dysregulated trace amine signalling underpins ASD pathology along multiple pathways. A major limitation to trace amine-focused medicines development in ASD, is the relative absence of GI data on trace amines. For conditions with a clear gut-brain link – such as ASD – execution of more studies including parallel assessment at gut and brain level should be a priority. Methodological advancements allowing the assessment of concentrations of comprehensive panels of trace amines across different matrices, are only now emerging (de Bruyn et al. 2024; Henning et al. 2024) and will contribute substantially to our understanding of dysregulation in this regard, in ASD. Furthermore, ASD research data including females are minimal and scattered – inclusion of sex as consideration in research going forward will not only advance management of female individuals on the spectrum but may also provide clues for therapeutics development.
Currently, two viable therapeutic approaches seem to have been identified, namely TAAR1 agonism and agmatine administration. Firstly, in terms of agmatine, we have already discussed its potential. However, also in this context, many gaps in our knowledge still remain. For example, while direct NMDA receptor antagonism of agmatine has been established as therapeutic mechanism centrally (Khoram-Abadi et al. 2024; Kim et al. 2017), the role of agmatine in the gut itself or from the gut (i.e. affecting change indirectly at locations outside of the gut) may be more complex. For example, oral administration of agmatine in a model of experimental depression, was shown to confer benefit via normalisation of gut barrier integrity (cellular junction proteins), but also neuroinflammation and serotonergic signalling in the hippocampus and prefrontal cortex (Rahangdale et al. 2024). A potential limitation to this study was that depression was induced via total eradication of the gut microbiome with antibiotics, so that these benefits of agmatine is not yet conclusive in a more physiologically relevant scenario. Thorough simultaneous investigation of mechanisms and outcomes in an ASD-specific model – both centrally and peripherally – is still lacking.
Secondly, TAAR1 agonism (using the experimental agonist RO5263397) has been linked to beneficial effects on neurological symptomology – such as frustration, irritability and aggression – in a rodent VPA model (L. Wang et al. 2024a, b). In a non-ASD context, TAAR1 antagonism by methamphetamine was shown to decrease cellular calcium flux (Proulx et al. 2024), which supports TAAR1 agonism in ASD to potentially improve mitochondrial respiration. In addition, other experimental compounds have been reported to show benefits in terms of metabolic regulation (specifically glycaemic control and satiety) (Dedic et al. 2024; Raab et al. 2016) that could reduce risk for cardiovascular disease, which is increased in ASD. However, while TAAR1 agonism has also been shown to delay stomach emptying – which at first glance may be the mechanism for satiety (Dedic et al. 2024; Milanovic et al. 2024) – this effect may in fact pose risk in ASD, where repetitive behaviour in terms of dietary patterns may result in relative ineffectiveness of satiety to reduce food intake. Indeed, the same study (Milanovic et al. 2024) reported a significant increase in gut disorders, and we have previously shown that delayed stomach emptying without a change in dietary habits (in rodents) resulted in significant morbid cardiovascular complications (Smith and Krygsman 2014). On a technical note, the majority of preclinical protocols producing beneficial results for TAAR1 agonism, employed intraperitoneal delivery as mode of administration. This bypasses the gut and potential gut-based effects of TAAR1 agonism, e.g. its previously demonstrated effects on serotonin signalling (Zhai et al. 2023). Thus, as is the case for agmatine, given the narrow focus of most preclinical investigations, a holistic picture of benefit vs. risk across tissues and compartments is still lacking. Furthermore, reports of contradictory inflammatory outcome after experimental modulation (by both agonists and antagonists) of TAAR1 signalling (Barnes et al. 2021; Gwilt et al. 2019; Polini et al. 2023), suggests that several contributing factors need to be considered in development of drugs therapeutically targeting TAAR1. These include specificity of ligand, tissue, cellular localisation and dose, as well as the nature of TAAR1 dimerisation. Studies of this scope is not available yet, but the hope is that drug trial designs will incorporate investigations at multiple levels to address these significant gaps in our knowledge.
In terms of progress on re-purposing existing drugs, the versatility of the TAAR1 binding pocket highlights its potential to recognise various ligands (Jiang et al. 2024), suggesting that drug re-purposing may be a feasible avenue for the therapeutics discovery in this context. Indeed, drug re-purposing strategies have led to the identification of several TAAR1 agonists, such as asenapine (atypical antipsychotic), fenoldopam (D1R partial agonist), guanfacine and guanabenz (both α2-adenoreceptor agonists) (Cichero et al. 2023; Shajan et al. 2024). The action of asenapine seems relatively broadly targeting (several neurotransmitter receptors), but other TAAR1 agonists with more specific actions, warrant consideration within the complex ASD scenario, as outcome may be relatively more predictable for these drugs. Nevertheless, a complexity that remains, is the fact that many TAAR1 agonists have lower selectivity for TAAR1 than other monoaminergic G-protein coupled receptors, increasing the risk of off-target effects. As such, besides conventional side effects (most commonly somnolence, headaches and hypotension), off-target receptor activation should be considered. In this case multiple receptors signalling and the related crosstalk should be investigated in the ASD context to confirm drug re-purposing applicability.
In terms of the role(s) of other TAARs in ASD, a study using gene express ion characteristic analyses and weighted gene co-expression network analyses, identified taar7h and taar7b as hub genes that significantly downregulated neuroactive ligand-receptor interaction pathways in hippocampal samples in VPA-treated rats (Huang et al. 2016), implicating TAARs and related gene regulatory networks as risk factors in ASD. Since taar7 is a pseudogene in humans, the relevance of these findings is still unclear. However, peptidome analysis revealed increased serum TAAR6 levels in children with ASD vs. controls (Yang et al. 2018), which may represent the translation from the rodent data, but more research is required before firm conclusions can be made.
Finally, an area of substantial neglect is our understanding of the extent to which enzymes involved in trace amine synthesis and metabolism, may be affected in ASD. Isolated enzymes have been investigated in pre-clinical models, e.g. elucidating the decreased levels of hippocampal glutamic acid decarboxylase enzyme (GAD67) as potential role player in the decreased GABA-signalling in VPA-associated ASD in rodents (Win-Shwe et al. 2018), but a holistic profile of potential enzyme dysregulation eludes. An added complexity here is that different isoforms of enzymes exist in different body compartments. For example, in contrast to the mentioned rodent data, increased plasma GAD65 levels were previously associated with severe autism in children (Vazifekhah et al. 2022), which complicates integrated interpretation of small data sets across different models. Also here, methodology has been a limiting factor, with population-wide gene-association studies yielding limited success (Buttenschon et al. 2009; Yu et al. 2016). However, in the current era of -omics-based research, significant strides should be possible in terms of elucidating dysregulated enzymatic processes, as well as the roles of environmental factors such as pharmaceuticals.
Together, these reports highlight the importance of a comprehensive assessment including all potential role players, i.e. levels of neurotransmitters and trace amines themselves, but also enzymes important for their synthesis or degradation, as well as the receptors through which they signal, across multiple tissue compartments, for a holistic understanding of the pathology that requires normalisation. Without this, biomarker-driven therapeutics development is not possible. Furthermore, returning to the heterogeneous nature of ASD symptomology, once more comprehensive preclinical data on therapeutic mechanisms has been generated, the next step would be assessment of the general applicability of any particular drug across the spectrum of ASD symptomatic diversity and severity, to determine the need for individualisation of therapeutics.
Conclusion
Large gaps still exist in the ASD literature, but some common maladaptation in the trace amine niche is clear, highlighting the potential to target this system for therapeutic modulation. Further knowledge gain in terms of trace amine metabolism, as well as receptor and enzymatic role players, may refine drug discovery approaches by elucidating exact mechanisms at play. Nevertheless, preclinical intervention studies can already contribute to therapeutic development by assessing the feasibility of achieving neurological modulation in ASD via intervention at the level of the gut. We propose that this may be achieved via modulation of trace amine signalling within the gut, using “clean” models such as larval zebrafish or germ-free rodents, where the relative contribution of the gut microbiome can be experimentally manipulated and accounted for. Given the known different scales in terms of trace amine abundance in the brain vs. the gut, the use of such models would be particularly useful in assessing “off-target” effects in compartments or organs other than the primary target.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- 5-HIAA:
-
5-hydroxyindoleacetic acid
- 5-HT:
-
Serotonin
- 5-HT2A/1AR:
-
Serotonin 1/2A receptor
- 5-HTP:
-
5-Hydroxytryptophan
- AA:
-
Amino acid
- AADC:
-
Aromatic L-amino acid decarboxylase
- ACC:
-
Anterior cingulate gyrus
- ADC:
-
Agmatine decarboxylase
- ADHD:
-
Attention deficit hyperactivity disorder
- ADP:
-
Adenosine diphosphate
- AGM:
-
Agmatine
- AGMAT:
-
Agmatinase
- Amg:
-
Amygdala
- βARR2:
-
Beta-arrestin-2
- ARG:
-
L-Arginine
- ARG I/II:
-
Arginase I/II
- ASD:
-
Autism spectrum disorder
- ATP:
-
Adenosine triphosphate
- BDNF:
-
Brain-derived neurotrophic factor
- CAT:
-
Catalase
- Cereb:
-
Cerebellum
- CN:
-
Caudate nucleus
- CNS:
-
Central nervous system
- COMT:
-
Catechol-O-methyltransferase
- CoQ:
-
Coenzyme Q
- CREB:
-
cAMP-response element binding protein
- Cyt c:
-
Cytochrome c
- DD1/D2R:
-
Dopamine D1/2 receptor
- DA:
-
Dopamine
- DAT:
-
Dopamine transporter
- DBH:
-
Dopamine beta-hydroxylase
- DDC:
-
DOPA decarboxylase
- DMT:
-
N, N-Dimethyltryptamine
- DRn:
-
Dorsal raphe nucleus
- Drp1:
-
Dynamin-related protein 1
- EC:
-
Entorhinal cortex
- ECC:
-
Enterochromaffin cell
- ENS:
-
Enteric nervous system
- ERK:
-
Extracellular signal-regulated kinases
- ETC:
-
Electron transport chain
- FAD/FADH2 :
-
Flavin adenine dinucleotide
- FC:
-
Frontal cortex
- FDOPA:
-
Fluorine-18-labelled fluorodopa
- FG:
-
Fusiform gyrus
- Fis1:
-
Mitochondrial fission 1 protein
- GABA:
-
Gamma-aminobutyric acid
- GABAA1/A2/A3/B3R:
-
GABA A1/A2/A3/B3 receptor(s)
- GAD:
-
Glutamic acid decarboxylase enzyme
- GI:
-
Gastrointestinal
- GIT:
-
Gastrointestinal tract
- Glu:
-
Glutamate
- GPx:
-
Glutathione peroxidase
- GR:
-
Glutathione reductase
- GSH:
-
Glutathione
- GSSG:
-
Glutathione disulfide
- Hipp:
-
Hippocampus
- HPA:
-
4-Hydroxyphenylacetaldehyde
- HVA:
-
Homovanillic acid
- I – V:
-
Mitochondrial electron transport chain complex I - V
- IAA:
-
Indoleacetic acid
- IBD:
-
Inflammatory bowel disease
- IBS-D:
-
Diarrhoea predominant IBS
- IBS:
-
Irritable bowel syndrome
- INMT:
-
Indolethylamine-N-methyltransferase
- L-DOPA:
-
Levodopa
- L-Phe:
-
L-Phenylalanine
- L-Trp:
-
L-Tryptophan
- L-Tyr:
-
L-Tyrosine
- m-TYR/3-MT:
-
3-Methoxytyramine
- MAO-A/B:
-
Monoamine oxidase-A/B
- MDA:
-
Malondialdehyde
- MET:
-
Mesenchymal-epithelial transition factor
- MFN1/2:
-
Mitofusin 1/2
- MnSOD:
-
Manganese superoxide dismutase
- mPTP:
-
Mitochondrial permeability transition pore
- mRNA:
-
Messenger RNA
- MSNs:
-
Medium spiny neurons
- mtDNA:
-
Mitochondrial DNA
- MTX2:
-
Metaxin 2
- NAc:
-
Nucleus accumbens
- NAD+/NADH:
-
Nicotinamide adenine dinucleotide
- NADP+/NADPH:
-
Nicotinamide adenine dinucleotide phosphate
- NEFL:
-
Neurofilament protein
- NMPEA:
-
N-Methylphenethylamine
- NMTRP:
-
N-Methyltryptamine
- NMTYR:
-
N-Methyltyramine
- NST:
-
Nigrostriatal pathway
- OCT:
-
Octopamine
- ODC:
-
Ornithine decarboxylase
- OPA1:
-
Optic atrophy 1
- ORN:
-
Ornithine
- PAA:
-
Phenylacetic acid
- PAH:
-
Phenylalanine hydroxylase
- PAO:
-
Polyamine oxidase
- PCC:
-
Posterior cingulate cortex
- β-PEA:
-
β-Phenethylamine
- PFC:
-
Prefrontal cortex
- PHC:
-
Parahippocampal cortex
- Pir:
-
Piriform
- PNMT:
-
Phenylethanolamine N-methyltransferase
- PPP:
-
Platelet poor plasma
- Prx(SH)2/Prx-S2 :
-
Peroxiredoxin reduced/oxidised
- PrxR:
-
Peroxiredoxin reductase
- Pu:
-
Putamen
- PUT:
-
Putrescine
- QH2 :
-
Dihydroxyquinone
- ROS:
-
Reactive oxygen species
- SCFA:
-
Short chain fatty acid
- SERT:
-
Serotonin transporter
- SMOX:
-
Spermine oxidase
- SNPs:
-
Single nucleotide polymorphisms
- SOD1/2:
-
Superoxide dismutase 1/2
- SPD:
-
Spermidine
- SPDS:
-
Spermidine synthase
- SPM:
-
Spermine
- SPMS:
-
Spermine synthase
- SSAT:
-
Spermidine/spermine N1-acetyltransferase
- STC:
-
Superior temporal cortex
- SYN:
-
Synephrine
- TA:
-
Trace amine
- TAAR1/2/6/7:
-
Trace amine associated receptor 1/2/6/7
- TH:
-
Tyrosine hydroxylase
- TH1-ir:
-
Tyrosine hydroxylase 1 immunoreactive
- TPH1/2:
-
Tryptophan hydroxylase 1/2
- TPH-ir:
-
Tryptophan hydroxylase immunoreactive
- TRP:
-
Tryptamine
- Trx(SH)2/Trx-S2 :
-
Thioredoxin reduced/oxidised
- TrxR:
-
Thioredoxin reductase
- r-TYR:
-
R-Tyramine
- VMAT2:
-
Vesicular monoamine transporter 2
- VNTR:
-
Variable number tandem repeat
- VPA:
-
Valproic acid
- ZF:
-
Zebrafish
References
Aaron E, Montgomery A, Ren X, Guter S, Anderson G, Carneiro AMD, Jacob S, Mosconi M, Pandey GN, Cook E, Veenstra-VanderWeele J. Whole blood serotonin levels and platelet 5-HT2A binding in autism spectrum disorder. J Autism Dev Disord. 2019;49(6):2417–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-019-03989-z.
Abdulamir HA, Abdul-Rasheed OF, Abdulghani EA. Serotonin and serotonin transporter levels in autistic children. Saudi Med J. 2018;39(5):487–94. https://doiorg.publicaciones.saludcastillayleon.es/10.15537/smj.2018.5.21751.
Agostinelli E. Biochemical and pathophysiological properties of polyamines. Amino Acids. 2020;52(2):111–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00726-020-02821-8.
Aishworiya R, Valica T, Hagerman R, Restrepo B. An update on psychopharmacological treatment of autism spectrum disorder. Neurotherapeutics. 2022;19(1):248–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13311-022-01183-1.
Akinyele O, Munir A, Johnson MA, Perez MS, Gao Y, Foley JR, Nwafor A, Wu Y, Murray-Stewart T, Casero RA Jr, Bayir H, Kemaladewi DU. Impaired polyamine metabolism causes behavioral and neuroanatomical defects in a mouse model of Snyder-Robinson syndrome. Dis Model Mech. 2024;17(6). https://doiorg.publicaciones.saludcastillayleon.es/10.1242/dmm.050639.
Al Sagheer T, Haida O, Balbous A, Francheteau M, Matas E, Fernagut P-O, Jaber M. Motor impairments correlate with social deficits and restricted neuronal loss in an environmental model of autism. Int J Neuropsychopharmacol. 2018;21(9):871–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ijnp/pyy043.
Alabdali A, Al-Ayadhi L, El-Ansary A. Association of social and cognitive impairment and biomarkers in autism spectrum disorders. J Neuroinflamm. 2014;11(1):4. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1742-2094-11-4.
Ali H, Elgoly A. Combined prenatal and postnatal Butyl Paraben exposure produces autism-like symptoms in offspring: comparison with valproic acid autistic model. Pharmacol Biochem Behav. 2013;111:102–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pbb.2013.08.016.
Altun H, Şahin N, Kurutaş EB, Karaaslan U, Sevgen FH, Fındıklı E. Assessment of malondialdehyde levels, superoxide dismutase, and catalase activity in children with autism spectrum disorders. Psychiatry Clin Psychopharmacol. 2018;28(4):408–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/24750573.2018.1470360.
Angoa-Pérez M, Kuhn DM. Evidence for modulation of substance use disorders by the gut microbiome: hidden in plain sight. Pharmacol Rev. 2021;73(2):571–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/pharmrev.120.000144.
Anitha A, Nakamura K, Thanseem I, Matsuzaki H, Miyachi T, Tsujii M, Iwata Y, Suzuki K, Sugiyama T, Mori N. Downregulation of the expression of mitochondrial electron transport complex genes in autism brains. Brain Pathol. 2013;23(3):294–302. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bpa.12002.
Anitha A, Thanseem I, Iype M, Thomas SV. Mitochondrial dysfunction in cognitive neurodevelopmental disorders: cause or effect? Mitochondrion. 2023;69:18–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mito.2023.01.002
Ashwood P, Wakefield AJ. Immune activation of peripheral blood and mucosal CD3 + lymphocyte cytokine profiles in children with autism and Gastrointestinal symptoms. J Neuroimmunol. 2006;173(1–2):126–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jneuroim.2005.12.007.
Ashwood P, Anthony A, Torrente F, Wakefield AJ. Spontaneous mucosal lymphocyte cytokine profiles in children with autism and Gastrointestinal symptoms: mucosal immune activation and reduced counter regulatory Interleukin-10. J Clin Immunol. 2004;24(6):664–73.
Azmitia EC, Singh JS, Whitaker-Azmitia PM. Increased serotonin axons (immunoreactive to 5-HT transporter) in postmortem brains from young autism donors. Neuropharmacology. 2011;60(7):1347–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuropharm.2011.02.002.
Babusyte A, Kotthoff M, Fiedler J, Krautwurst D. Biogenic amines activate blood leukocytes via trace amine-associated receptors TAAR1 and TAAR2. J Leukoc Biol. 2013;93(3):387–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1189/jlb.0912433.
Baker GB, Bornstein RA, Rouget AC, Ashton SE, van Muyden JC, Coutts RT. Phenylethylaminergic mechanisms in Attention-Deficit disorder. Biol Psychiatry. 1991;29:15–22.
Barnes DA, Galloway DA, Hoener MC, Berry MD, Moore CS. TAAR1 expression in human macrophages and brain tissue: A potential novel facet of MS neuroinflammation. Int J Mol Sci. 2021;22(21). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms222111576.
Baronio D, Puttonen HAJ, Sundvik M, Semenova S, Lehtonen E, Panula P. Embryonic exposure to valproic acid affects the histaminergic system and the social behaviour of adult zebrafish (Danio rerio). Br J Pharmacol. 2018;175(5):797–809. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bph.14124.
Basivireddya J, Vasudevana A, Jacobb M, Balasubramaniana KA. Indomethacin-induced mitochondrial dysfunction and oxidative stress in villus enterocytes. Biochem Pharmacol. 2002;64:339–49.
Bauman ML. Microscopic neuroanatomic abnormalities in autism. Pediatrics. 1991;87(5 Pt 2):791–6.
Bauman M, Kemper TL. Histoanatomic observations of the brain in early infantile autism. Neurology. 1985;35(6):866–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1212/wnl.35.6.866.
Belle NA, Dalmolin GD, Fonini G, Rubin MA, Rocha JB. Polyamines reduces lipid peroxidation induced by different pro-oxidant agents. Brain Res. 2004;1008(2):245–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainres.2004.02.036.
Bernier R, Golzio C, Xiong B, Stessman HA, Coe BP, Penn O, Witherspoon K, Gerdts J, Baker C, Vulto-van Silfhout AT, Schuurs-Hoeijmakers JH, Fichera M, Bosco P, Buono S, Alberti A, Failla P, Peeters H, Steyaert J, Vissers L, Eichler EE. Disruptive CHD8 mutations define a subtype of autism early in development. Cell. 2014;158(2):263–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2014.06.017.
Bhattarai Y, Williams BB, Battaglioli EJ, Whitaker WR, Till L, Grover M, Linden DR, Akiba Y, Kandimalla KK, Zachos NC, Kaunitz JD, Sonnenburg JL, Fischbach MA, Farrugia G, Kashyap PC. Gut Microbiota-Produced tryptamine activates an epithelial G-Protein-Coupled receptor to increase colonic secretion. Cell Host Microbe. 2018;23(6):775–e785775. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.chom.2018.05.004.
Bortolato M, Chen K, Shih JC. Monoamine oxidase inactivation: from pathophysiology to therapeutics. Adv Drug Deliv Rev. 2008;60(13–14):1527–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2008.06.002.
Bortolato M, Godar SC, Alzghoul L, Zhang J, Darling RD, Simpson KL, Bini V, Chen K, Wellman CL, Lin RC. Monoamine oxidase A and A/B knockout mice display autistic-like features. Int J Neuropsychopharmacol. 2013;16(4):869–88.
Brandenburg C, Blatt GJ. Differential serotonin transporter (5-HTT) and 5-HT2 receptor density in limbic and neocortical areas of adults and children with autism spectrum disorders: implications for selective serotonin reuptake inhibitor efficacy. J Neurochem. 2019;151(5):642–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jnc.14832.
Brandenburg C, Soghomonian JJ, Zhang K, Sulkaj I, Randolph B, Kachadoorian M, Blatt GJ. Increased dopamine type 2 gene expression in the dorsal striatum in individuals with autism spectrum disorder suggests alterations in indirect pathway signaling and circuitry. Front Cell Neurosci. 2020;14:577858. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fncel.2020.577858.
Bretler T, Weisberg H, Koren O, Neuman H. The effects of antipsychotic medications on Microbiome and weight gain in children and adolescents. BMC Med. 2019;17(1):112. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12916-019-1346-1.
Bu X, Wu D, Lu X, Yang L, Xu X, Wang J, Tang J. Role of SIRT1/PGC-1alpha in mitochondrial oxidative stress in autistic spectrum disorder. Neuropsychiatr Dis Treat. 2017;13:1633–45. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/NDT.S129081.
Buck TR, Viskochil J, Farley M, Coon H, McMahon WM, Morgan J, Bilder DA. Psychiatric comorbidity and medication use in adults with autism spectrum disorder. J Autism Dev Disord. 2014;44(12):3063–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-014-2170-2.
Butera A, De Simone R, Potenza RL, Sanchez M, Armida M, Campanile D, Di Carlo N, Trenta F, Boirivant M, Ricceri L. Effects of a gut-selective integrin-targeted therapy in male mice exposed to early immune activation, a model for the study of autism spectrum disorder. Brain Behav Immun. 2024;115:89–100. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbi.2023.09.024.
Buttenschon HN, Lauritsen MB, El Daoud A, Hollegaard M, Jorgensen M, Tvedegaard K, Hougaard D, Borglum A, Thorsen P, Mors O. A population-based association study of glutamate decarboxylase 1 as a candidate gene for autism. J Neural Transm (Vienna). 2009;116(3):381–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00702-008-0142-4.
Campbell DB, Buie TM, Winter H, Bauman M, Sutcliffe JS, Perrin JM, Levitt P. Distinct genetic risk based on association of MET in families with co-occurring autism and Gastrointestinal conditions. Pediatrics. 2009;123(3):1018–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1542/peds.2008-0819.
Cao C, Wang D, Zou M, Sun C, Wu L. Untargeted metabolomics reveals hepatic metabolic disorder in the BTBR mouse model of autism and the significant role of liver in autism. Cell Biochem Funct. 2023;41(5):553–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cbf.3811.
Cermak SA, Curtin C, Bandini LG. Food selectivity and sensory sensitivity in children with autism spectrum disorders. J Am Diet Assoc. 2010;110(2):238–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jada.2009.10.032.
Cezar LC, Kirsten TB, da Fonseca CCN, de Lima APN, Bernardi MM, Felicio LF. Zinc as a therapy in a rat model of autism prenatally induced by valproic acid. Prog Neuropsychopharmacol Biol Psychiatry. 2018;84:173–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pnpbp.2018.02.008.
Chauhan A, Chauhan V, Brown WT, Cohen I. Oxidative stress in autism: increased lipid peroxidation and reduced serum levels of ceruloplasmin and transferrin–the antioxidant proteins. Life Sci. 2004;75(21):2539–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2004.04.038.
Chauhan A, Gu F, Essa MM, Wegiel J, Kaur K, Brown WT, Chauhan V. Brain region-specific deficit in mitochondrial electron transport chain complexes in children with autism. J Neurochem. 2011;117(2):209–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1471-4159.2011.07189.x.
Chen S, Xu Q, Zhao L, Zhang M, Xu H. The prenatal use of agmatine prevents social behavior deficits in VPA-exposed mice by activating the ERK/CREB/BDNF signaling pathway. Birth Defects Res. 2024;116(4):e2336. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/bdr2.2336.
Christian SL, Berry MD. Trace Amine-Associated receptors as novel therapeutic targets for Immunomodulatory disorders. Front Pharmacol. 2018;9:680. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2018.00680.
Chugani DC, Muzik O, Behen M, Rothermel R, Janisse JJ, Lee J, Chugani HT. Developmental changes in brain serotonin synthesis capacity in autistic and nonautistic children. Ann Neurol. 1999;45(3):287–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/1531-8249(199903)45:3%3C287::aid-ana3%3E3.0.co;2-9.
Cichero E, Francesconi V, Casini B, Casale M, Kanov E, Gerasimov AS, Sukhanov I, Savchenko A, Espinoza S, Gainetdinov RR. Discovery of Guanfacine as a novel TAAR1 agonist: A combination strategy through molecular modeling studies and biological assays. Pharmaceuticals. 2023;16:1632. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph16111632.
Condello S, Curro M, Ferlazzo N, Caccamo D, Satriano J, Ientile R. Agmatine effects on mitochondrial membrane potential and NF-kappaB activation protect against rotenone-induced cell damage in human neuronal-like SH-SY5Y cells. J Neurochem. 2011;116(1):67–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1471-4159.2010.07085.x.
Coretti L, Paparo L, Riccio MP, Amato F, Cuomo M, Natale A, Borrelli L, Corrado G, Comegna M, Buommino E, Castaldo G, Bravaccio C, Chiariotti L, Canani B, R., Lembo F. Gut microbiota features in young children with autism spectrum disorders. Front Microbiol. 2018;9:3146. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmicb.2018.03146.
Courchesne E, Press GA, Yeung-Courchesne R. Parietal lobe abnormalities detected with MR in patients with infantile autism. AJR Am J Roentgenol. 1993;160(2):387–93. https://doiorg.publicaciones.saludcastillayleon.es/10.2214/ajr.160.2.8424359.
Courchesne E, Mouton PR, Calhoun ME, Semendeferi K, Ahrens-Barbeau C, Hallet MJ, Barnes CC, Pierce K. Neuron number and size in prefrontal cortex of children with autism. JAMA. 2011a;306(18):2001–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jama.2011.1638.
Courchesne E, Campbell K, Solso S. Brain growth across the life span in autism: age-specific changes in anatomical pathology. Brain Res. 2011b;1380:138–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainres.2010.09.101.
De Angelis M, Piccolo M, Vannini L, Siragusa S, De Giacomo A, Serrazzanetti DI, Cristofori F, Guerzoni ME, Gobbetti M, Francavilla R. Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified. PLoS ONE. 2013;8(10):e76993. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0076993.
de Bruyn K, Diekman EF, van der Ley CP, van Faassen M, Kema IP. Simultaneous mass spectrometric quantification of trace amines, their precursors and metabolites. J Chromatogr B Analyt Technol Biomed Life Sci. 2024;1238:124098. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jchromb.2024.124098.
de Magistris L, Familiari V, Pascotto A, Sapone A, Frolli A, Iardino P, Carteni M, De Rosa M, Francavilla R, Riegler G, Militerni R, Bravaccio C. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51(4):418–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/MPG.0b013e3181dcc4a5.
Dedic N, Wang L, Hajos-Korcsok E, Hecksher-Sorensen J, Roostalu U, Vickers SP, Wu S, Anacker C, Synan C, Jones PG, Milanovic S, Hopkins SC, Bristow LJ, Koblan KS. TAAR1 agonists improve glycemic control, reduce body weight and modulate neurocircuits governing energy balance and feeding. Mol Metab. 2024;80:101883. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molmet.2024.101883.
Dias MF, Nogueira YJA, Romano-Silva MA, de Marques D. Effects of antipsychotics on the Gastrointestinal microbiota: A systematic review. Psychiatry Res. 2024;336:115914. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.psychres.2024.115914.
El-Ansary A, Bhat RS, Zayed N. Gut Microbiome and sex Bias in autism spectrum disorders. Curr Behav Neurosci Rep. 2020;7(1):22–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40473-020-00197-3.
El-Sayed EK, Ahmed A, Morsy EE, Nofal S. Neuroprotective effect of Agmatine (decarboxylated l-arginine) against oxidative stress and neuroinflammation in rotenone model of Parkinson’s disease. Hum Exp Toxicol. 2019;38(2):173–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0960327118788139.
Elesawy RO, El-Deeb OS, Eltokhy AK, Arakeep HM, Ali DA, Elkholy SS, Kabel AM. Postnatal Baicalin ameliorates behavioral and neurochemical alterations in valproic acid-induced rodent model of autism: the possible implication of sirtuin-1/mitofusin-2/ Bcl-2 pathway. Biomed Pharmacother. 2022;150:112960. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2022.112960.
Ernst M, Zametkin AJ, Matochik JA, Pascualvaca D, Cohen RM. Low medial prefrontal dopaminergic activity in autistic children. Lancet. 1997;350(9078):638. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(05)63326-0.
Esnafoglu E, Irende I. Decreased plasma agmatine levels in autistic subjects. J Neural Transm (Vienna). 2018;125(4):735–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00702-017-1836-2.
Esnafoglu E, Cirrik S, Ayyildiz SN, Erdil A, Erturk EY, Dagli A, Noyan T. Increased serum Zonulin levels as an intestinal permeability marker in autistic subjects. J Pediatr. 2017;188:240–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jpeds.2017.04.004.
Esposito M, Mirizzi P, Fadda R, Pirollo C, Ricciardi O, Mazza M, Valenti M. Food selectivity in children with autism: guidelines for assessment and clinical interventions. Int J Environ Res Public Health. 2023;20(6). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijerph20065092.
Fatemi SH, Reutiman TJ, Folsom TD, Thuras PD. GABA(A) receptor downregulation in brains of subjects with autism. J Autism Dev Disord. 2009;39(2):223–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-008-0646-7.
Fattorusso A, Di Genova L, Dell’Isola GB, Mencaroni E, Esposito S. Autism spectrum disorders and the gut microbiota. Nutrients. 2019;11(3). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu11030521.
Finegold SM, Molitoris D, Song Y, Liu C, Vaisanen M-L, Bolte E, McTeague M, Sandler R, Wexler H, Marlowe EM, Collins MD, Lawson PA, Summanen P, Baysallar M, Tomzynski TJ, Read E, Johnson E, Rolfe R, Nasir P, Kaul A. Gastrointestinal microflora studies in Late-Onset autism. Clin Infect Dis. 2002;35:S6–16.
Fiori LM, Turecki G. Implication of the polyamine system in mental disorders. J Psychiatry Neurosci. 2008;33(2):102–10.
Fleischer LM, Somaiya RD, Miller GM. Review and meta-analyses of TAAR1 expression in the immune system and cancers. Front Pharmacol. 2018;9:683. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2018.00683.
Fröhlich H, Kollmeyer ML, Linz VC, Stuhlinger M, Groneberg D, Reigl A, Zizer E, Friebe A, Niesler B, Rappold G. Gastrointestinal dysfunction in autism displayed by altered motility and achalasia in Foxp1+/− mice. Proc Natl Acad Sci U S A. 2019;116(44):22237–22245. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1911429116.
Frye RE, Rose S, Slattery J, MacFabe DF. Gastrointestinal dysfunction in autism spectrum disorder: the role of the mitochondria and the enteric microbiome. Microb Ecol Health Dis. 2015;26:27458. https://doiorg.publicaciones.saludcastillayleon.es/10.3402/mehd.v26.27458.
Frye RE, Casanova MF, Fatemi SH, Folsom TD, Reutiman TJ, Brown GL, Edelson SM, Slattery JC, Adams JB. Neuropathological mechanisms of seizures in autism spectrum disorder [Mini Review]. Front NeuroSci. 2016;10. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnins.2016.00192.
Frye RE, Vassall S, Kaur G, Lewis C, Karim M, Rossignol D. Emerging biomarkers in autism spectrum disorder: a systematic review. Ann Transl Med. 2019;7(23):792. https://doiorg.publicaciones.saludcastillayleon.es/10.21037/atm.2019.11.53.
Frye RE, Rincon N, McCarty PJ, Brister D, Scheck AC, Rossignol DA. Biomarkers of mitochondrial dysfunction in autism spectrum disorder: A systematic review and meta-analysis. Neurobiol Dis. 2024;197:106520. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2024.106520.
Gabriele S, Sacco R, Persico AM. Blood serotonin levels in autism spectrum disorder: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2014;24(6):919–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.euroneuro.2014.02.004.
Gawali NB, Bulani VD, Gursahani MS, Deshpande PS, Kothavade PS, Juvekar AR. Agmatine attenuates chronic unpredictable mild stress-induced anxiety, depression-like behaviours and cognitive impairment by modulating nitrergic signalling pathway. Brain Res. 2017;1663:66–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainres.2017.03.004.
Geier DA, Kern JK, Davis G, King PG, Adams JB, Young JL, Geier MR. A prospective double-blind, randomized clinical trial of levocarnitine to treat autism spectrum disorders. Med Sci Monit. 2011;17(6):15–23.
Giulivi C, Zhang YF, Omanska-Klusek A, Ross-Inta C, Wong S, Hertz-Picciotto I, Tassone F, Pessah IN. Mitochondrial dysfunction in autism. JAMA. 2010;304(21):2389–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jama.2010.1706.
Gotham K, Brunwasser SM, Lord C. Depressive and anxiety symptom trajectories from school age through young adulthood in samples with autism spectrum disorder and developmental delay. J Am Acad Child Adolesc Psychiatry. 2015;54(5):369–e376363. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaac.2015.02.005.
Grimaldi R, Cela D, Swann JR, Vulevic J, Gibson GR, Tzortzis G, Costabile A. In vitro fermentation of B-GOS: impact on faecal bacterial populations and metabolic activity in autistic and non-autistic children. FEMS Microbiol Ecol. 2017;93(2). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/femsec/fiw233.
Grossi E, Melli S, Dunca D, Terruzzi V. Unexpected improvement in core autism spectrum disorder symptoms after long-term treatment with probiotics. SAGE Open Med Case Rep. 2016;4:2050313X16666231. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/2050313X16666231.
Gu F, Chauhan V, Chauhan A. Impaired synthesis and antioxidant defense of glutathione in the cerebellum of autistic subjects: alterations in the activities and protein expression of glutathione-related enzymes. Free Radic Biol Med. 2013a;65:488–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.freeradbiomed.2013.07.021.
Gu F, Chauhan V, Kaur K, Brown WT, LaFauci G, Wegiel J, Chauhan A. Alterations in mitochondrial DNA copy number and the activities of electron transport chain complexes and pyruvate dehydrogenase in the frontal cortex from subjects with autism. Translational Psychiatry. 2013b;3(9):e299–299. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/tp.2013.68.
Guerbette T, Boudry G, Lan A. Mitochondrial function in intestinal epithelium homeostasis and modulation in diet-induced obesity. Mol Metab. 2022;63:101546. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molmet.2022.101546.
Gupta N, Zhang H, Liu P. Behavioral and neurochemical effects of acute Putrescine depletion by Difluoromethylornithine in rats. Neuroscience. 2009;161(3):691–706. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuroscience.2009.03.075.
Gwilt KB, Olliffe N, Hoffing RA, Miller GM. Trace amine associated receptor 1 (TAAR1) expression and modulation of inflammatory cytokine production in mouse bone marrow-derived macrophages: a novel mechanism for inflammation in ulcerative colitis. Immunopharmacol Immunotoxicol. 2019;41(6):577–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/08923973.2019.1672178.
Gwilt K, Gonzalez DP, Olliffe N, Oller H, Hoffing R, Puzan M, El Aidy S, Miller GM. Actions of trace amines in the brain-gut-microbiome axis via trace amine-associated receptor-1 (TAAR1). Cell Mol Neurobiol. 2020;40(2):191–201. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10571-019-00772-7.
Haida O, Al Sagheer T, Balbous A, Francheteau M, Matas E, Soria F, Fernagut PO, Jaber M. Sex-dependent behavioral deficits and neuropathology in a maternal immune activation model of autism. Translational Psychiatry. 2019;9(1):124. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41398-019-0457-y.
Halff EF, Rutigliano G, Garcia-Hidalgo A, Howes OD. Trace amine-associated receptor 1 (TAAR1) agonism as a new treatment strategy for schizophrenia and related disorders. Trends Neurosci. 2023;46(1):60–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tins.2022.10.010.
Han L, Zhao L, Zhou Y, Yang C, Xiong T, Lu L, Deng Y, Luo W, Chen Y, Qiu Q, Shang X, Huang L, Mo Z, Huang S, Huang S, Liu Z, Yang W, Zhai L, Ning Z, Fang X. Altered metabolome and Microbiome features provide clues in Understanding irritable bowel syndrome and depression comorbidity. ISME J. 2022;16(4):983–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41396-021-01123-5.
Hao X, Pan J, Gao X, Zhang S, Li Y. Gut microbiota on gender bias in autism spectrum disorder. Rev Neurosci. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1515/revneuro-2020-0042.
Hara Y, Takuma K, Takano E, Katashiba K, Taruta A, Higashino K, Hashimoto H, Ago Y, Matsuda T. Reduced prefrontal dopaminergic activity in valproic acid-treated mouse autism model. Behav Brain Res. 2015;289:39–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbr.2015.04.022.
Harada M, Taki MM, Nose A, Kubo H, Mori K, Nishitani H, Matsuda T. Non-invasive evaluation of the gabaergic/glutamatergic system in autistic patients observed by MEGA-editing proton MR spectroscopy using a clinical 3 Tesla instrument. J Autism Dev Disord. 2011;41(4):447–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-010-1065-0.
Henning N, Kellermann TA, Smith C. Effect of chronic dolutegravir administration on the trace amine profile in Wistar rats. Drugs R D. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40268-024-00484-4
Hettinger JA, Liu X, Hudson ML, Lee A, Cohen IL, Michaelis RC, Schwartz CE, Lewis SME, Holden JJA. DRD2 and PPP1R1B (DARPP-32) polymorphisms independently confer increased risk for autism spectrum disorders and additively predict affected status in male-only affected sib-pair families. Behav Brain Funct. 2012;8(1):19. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1744-9081-8-19.
Hipkiss AR. Energy metabolism and autism: the ameliorative potential of carnosine and Agmatine. J Neurol Neurocritical Care. 2018;1(1):1–6. https://doiorg.publicaciones.saludcastillayleon.es/10.31038/JNNC.2018101Alan.
Hokanson KC, Hernandez C, Deitzler GE, Gaston JE, David MM. Sex shapes gut-microbiota-brain communication and disease. Trends Microbiol. 2024;32(2):151–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tim.2023.08.013.
Hosie S, Ellis M, Swaminathan M, Ramalhosa F, Seger GO, Balasuriya GK, Gillberg C, Rastam M, Churilov L, McKeown SJ, Yalcinkaya N, Urvil P, Savidge T, Bell CA, Bodin O, Wood J, Franks AE, Bornstein JC, Hill-Yardin EL. Gastrointestinal dysfunction in patients and mice expressing the autism-associated R451C mutation in neuroligin-3. Autism Res. 2019;12(7):1043–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/aur.2127.
Hranilovic D, Bujas-Petkovic Z, Vragovic R, Vuk T, Hock K, Jernej B. Hyperserotonemia in adults with autistic disorder. J Autism Dev Disord. 2007;37(10):1934–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-006-0324-6.
Huang JY, Tian Y, Wang HJ, Shen H, Wang H, Long S, Liao MH, Liu ZR, Wang ZM, Li D, Tao RR, Cui TT, Moriguchi S, Fukunaga K, Han F, Lu YM. Functional genomic analyses identify pathways dysregulated in animal model of autism. CNS Neurosci Ther. 2016;22(10):845–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cns.12582.
Hughes HK, Rose D, Ashwood P. The gut microbiota and dysbiosis in autism spectrum disorders. Curr Neurol Neurosci Rep. 2018;18(11):81. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11910-018-0887-6.
Hutsler JJ, Zhang H. Increased dendritic spine densities on cortical projection neurons in autism spectrum disorders. Brain Res. 2010;1309:83–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainres.2009.09.120.
Iglesias-Vazquez L, Van Ginkel Riba G, Arija V, Canals J. Composition of gut microbiota in children with autism spectrum disorder: A systematic review and Meta-Analysis. Nutrients. 2020;12(3). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu12030792.
Jacob MS, Presti DE. Endogenous psychoactive tryptamines reconsidered: an anxiolytic role for dimethyltryptamine. Med Hypotheses. 2005;64(5):930–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mehy.2004.11.005.
Jacobs JP, Goudarzi M, Singh N, Tong M, McHardy IH, Ruegger P, Asadourian M, Moon BH, Ayson A, Borneman J, McGovern DP, Fornace AJ Jr., Braun J, Dubinsky M. A disease-Associated microbial and metabolomics state in relatives of pediatric inflammatory bowel disease patients. Cell Mol Gastroenterol Hepatol. 2016;2(6):750–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jcmgh.2016.06.004.
James SJ, Cutler P, Melnyk S, Jernigan S, Janak L, Gaylor DW, Neubrander JA. Metabolic biomarkers of increased oxidative stress and impaired methylation capacity in children with autism. Am J Clin Nutr. 2004;80(6):1611–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ajcn/80.6.1611.
James DM, Kozol RA, Kajiwara Y, Wahl AL, Storrs EC, Buxbaum JD, Klein M, Moshiree B, Dallman JE. Intestinal dysmotility in a zebrafish (Danio rerio) shank3a;shank3b mutant model of autism. Mol Autism. 2019;10:3. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13229-018-0250-4.
Jia Y, Jin S, Hu K, Geng L, Han C, Kang R, Pang Y, Ling E, Tan EK, Pan Y, Liu W. Gut microbiome modulates drosophila aggression through octopamine signaling. Nat Commun. 2021;12(1):2698. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-021-23041-y.
Jiang K, Zheng Y, Zeng L, Wang L, Li F, Pu J, Lu Y, Zhao S, Xu F. The versatile binding landscape of the TAAR1 pocket for LSD and other antipsychotic drug molecules. Cell Rep. 2024;43:114505. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2024.114505.
Kang DW, Park JG, Ilhan ZE, Wallstrom G, Labaer J, Adams JB, Krajmalnik-Brown R. Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. PLoS ONE. 2013;8(7):e68322. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0068322.
Khoram-Abadi KM, Haratizadeh S, Basiri M, Parvan M, Pourjafari F, Aghaei I, Amiresmaili S, Nozari M. Autistic-like behaviors are attenuated by agmatine consumption during pregnancy: assessment of oxidative stress profile and histopathological changes in the prefrontal cortex and CA1 region of the hippocampus. Iran J Basic Med Sci. 2024;27(3):335–42. https://doiorg.publicaciones.saludcastillayleon.es/10.22038/IJBMS.2023.74536.16190.
Kim JW, Seung H, Kim KC, Gonzales ELT, Oh HA, Yang SM, Ko MJ, Han SH, Banerjee S, Shin CY. Agmatine rescues autistic behaviors in the valproic acid-induced animal model of autism. Neuropharmacol. 2017;113(Pt A):71–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuropharm.2016.09.014.
Kisuse J, La-Ongkham O, Nakphaichit M, Therdtatha P, Momoda R, Tanaka M, Fukuda S, Popluechai S, Kespechara K, Sonomoto K, Lee YK, Nitisinprasert S, Nakayama J. Urban diets linked to gut microbiome and metabolome alterations in children: a comparative cross-sectional study in Thailand. Front Microbiol. 2018;9:1345. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmicb.2018.01345.
Kong Q, Wang B, Tian P, Li X, Zhao J, Zhang H, Chen W, Wang G. Daily intake of Lactobacillus alleviates autistic-like behaviors by ameliorating the 5-hydroxytryptamine metabolic disorder in VPA-treated rats during weaning and sexual maturation [10.1039/D0FO02375B]. Food Funct. 2021;12(6):2591–604. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/D0FO02375B.
Kotagale NR, Taksande BG, Inamdar NN. Neuroprotective offerings by Agmatine. Neurotoxicology. 2019;73:228–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuro.2019.05.001.
Kumar H, Sharma B. Memantine ameliorates autistic behavior, biochemistry & blood brain barrier impairments in rats. Brain Res Bull. 2016;124:27–39. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainresbull.2016.03.013.
Lee YJ, Kim HR, Lee CY, Hyun SA, Ko MY, Lee BS, Hwang DY, Ka M. 2-Phenylethylamine (PEA) ameliorates corticosterone-induced depression-like phenotype via the BDNF/TrkB/CREB signaling pathway. Int J Mol Sci. 2020;21(23). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21239103.
Leembruggen AJL, Balasuriya GK, Zhang J, Schokman S, Swiderski K, Bornstein JC, Nithianantharajah J, Hill-Yardin EL. Colonic dilation and altered ex vivo gastrointestinal motility in the neuroligin-3 knockout mouse. Autism Res. 2020;13(5):691–701. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/aur.2109.
Lenis YY, Elmetwally MA, Maldonado-Estrada JG, Bazer FW. Physiological importance of polyamines. Zygote. 2017;25(3):244–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1017/S0967199417000120.
Lever AG, Geurts HM. Psychiatric co-occurring symptoms and disorders in young, middle-aged, and older adults with autism spectrum disorder. J Autism Dev Disord. 2016;46(6):1916–1930. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-016-2722-8
Lew CH, Groeniger KM, Hanson KL, Cuevas D, Greiner DMZ, Hrvoj-Mihic B, Bellugi U, Schumann CM, Semendeferi K. Serotonergic innervation of the amygdala is increased in autism spectrum disorder and decreased in Williams syndrome. Mol Autism. 2020;11(1):12. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13229-019-0302-4.
Madsen KL, Yanchar NL, Sigalet DL, Reigel T, Fedorak RN. FK506 increases permeability in rat intestine by inhibiting mitochondrial function. Gasteroenterology. 1995;109:107–14.
Makkonen I, Riikonen R, Kokki H, Airaksinen MM, Kuikka JT. Serotonin and dopamine transporter binding in children with autism determined by SPECT. Dev Med Child Neurol. 2008;50(8):593–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1469-8749.2008.03027.x.
Margolis KG, Li Z, Stevanovic K, Saurman V, Israelyan N, Anderson GM, Snyder I, Veenstra-VanderWeele J, Blakely RD, Gershon MD. Serotonin transporter variant drives preventable gastrointestinal abnormalities in development and function. J Clin Invest. 2016;126(6):2221–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI84877.
Mariggiò MA, Palumbi R, Vinella A, Laterza R, Petruzzelli MG, Peschechera A, Gabellone A, Gentile O, Vincenti A, Margari L. DRD1 and DRD2 receptor polymorphisms: genetic neuromodulation of the dopaminergic system as a risk factor for ASD, ADHD and ASD/ADHD overlap [Original Research]. Front NeuroSci. 2021;15. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnins.2021.705890.
Marler S, Ferguson BJ, Lee EB, Peters B, Williams KC, McDonnell E, Macklin EA, Levitt P, Gillespie CH, Anderson GM, Margolis KG, Beversdorf DQ, Veenstra-VanderWeele J. Brief report: whole blood serotonin levels and gastrointestinal symptoms in autism spectrum disorder. J Autism Dev Disord. 2016;46(3):1124–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-015-2646-8.
Martinis P, Grancara S, Kanamori Y, Garcia-Argaez AN, Pacella E, Dalla Via L, Toninello A, Agostinelli E. Involvement of the biogenic active amine agmatine in mitochondrial membrane permeabilization and release of pro-apoptotic factors. Amino Acids. 2020;52(2):161–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00726-019-02791-6.
Mawe GM, Hoffman JM. Serotonin signalling in the gut–functions, dysfunctions and therapeutic targets. Nat Rev Gastroenterol Hepatol. 2013;10(8):473–86. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrgastro.2013.105.
Melnyk S, Fuchs GJ, Schulz E, Lopez M, Kahler SG, Fussell JJ, Bellando J, Pavliv O, Rose S, Seidel L, Gaylor DW, James SJ. Metabolic imbalance associated with methylation dysregulation and oxidative damage in children with autism. J Autism Dev Disord. 2012;42(3):367–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-011-1260-7.
Milanovic S, Dedic N, Lew R, Burton D, Koblan KS, Camilleri M, Hopkins SC. TAAR1 agonist Ulotaront delays gastric emptying of solids in patients with schizophrenia and concurrent metabolic syndrome with prediabetes. Diabetes Obes Metab. 2024;26(6):2466–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/dom.15569.
Moiseenko VI, Apryatina VA, Gainetdinov RR, Apryatin SA. Trace amine-associated receptors’ role in immune system functions. Biomedicines. 2024;12(4). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines12040893.
Morton JT, Jin DM, Mills RH, Shao Y, Rahman G, McDonald D, Zhu Q, Balaban M, Jiang Y, Cantrell K, Gonzalez A, Carmel J, Frankiensztajn LM, Martin-Brevet S, Berding K, Needham BD, Zurita MF, David M, Averina OV, Taroncher-Oldenburg G. Multi-level analysis of the gut-brain axis shows autism spectrum disorder-associated molecular and microbial profiles. Nat Neurosci. 2023;26(7):1208–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41593-023-01361-0.
Moschandrea C, Kondylis V, Evangelakos I, Herholz M, Schneider F, Schmidt C, Yang M, Ehret S, Heine M, Jaeckstein MY, Szczepanowska K, Schwarzer R, Baumann L, Bock T, Nikitopoulou E, Brodesser S, Kruger M, Frezza C, Heeren J, Pasparakis M. Mitochondrial dysfunction abrogates dietary lipid processing in enterocytes. Nature. 2024;625(7994):385–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-023-06857-0.
Muller CL, Anacker AMJ, Veenstra-VanderWeele J. The serotonin system in autism spectrum disorder: from biomarker to animal models. Neuroscience. 2016;321:24–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuroscience.2015.11.010.
Nakamura K, Sekine Y, Ouchi Y, Tsujii M, Yoshikawa E, Futatsubashi M, Tsuchiya KJ, Sugihara G, Iwata Y, Suzuki K, Matsuzaki H, Suda S, Sugiyama T, Takei N, Mori N. Brain serotonin and dopamine transporter bindings in adults with high-functioning autism. Arch Gen Psychiatry. 2010;67(1):59–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/archgenpsychiatry.2009.137.
Napoli E, Wong S, Hertz-Picciotto I, Giulivi C. Deficits in bioenergetics and impaired immune response in granulocytes from children with autism. Pediatrics. 2014;133(5):e1405–1410. https://doiorg.publicaciones.saludcastillayleon.es/10.1542/peds.2013-1545.
Needham BD, Adame MD, Serena G, Rose DR, Preston GM, Conrad MC, Campbell AS, Donabedian DH, Fasano A, Ashwood P, Mazmanian SK. Plasma and fecal metabolite profiles in autism spectrum disorder. Biol Psychiatry. 2021;89(5):451–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopsych.2020.09.025.
Niesler B, Rappold GA. Emerging evidence for gene mutations driving both brain and gut dysfunction in autism spectrum disorder. Mol Psychiatry. 2021;26(5):1442–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41380-020-0778-5.
Oblak AL, Gibbs TT, Blatt GJ. Reduced GABAA receptors and benzodiazepine binding sites in the posterior cingulate cortex and fusiform gyrus in autism. Brain Res. 2011;1380:218–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.brainres.2010.09.021.
Oblak A, Gibbs TT, Blatt GJ. Reduced serotonin receptor subtypes in a limbic and a neocortical region in autism. Autism Res. 2013;6(6):571–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/aur.1317.
Ooi YP, Weng SJ, Jang LY, Low L, Seah J, Teo S, Ang RP, Lim CG, Liew A, Fung DS, Sung M. Omega-3 fatty acids in the management of autism spectrum disorders: findings from an open-label pilot study in Singapore. Eur J Clin Nutr. 2015;69(8):969–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ejcn.2015.28.
Panas MW, Xie Z, Panas HN, Hoener MC, Vallender EJ, Miller GM. Trace amine associated receptor 1 signaling in activated lymphocytes. J Neuroimmune Pharmacol. 2012;7(4):866–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11481-011-9321-4.
Polini B, Ricardi C, Bertolini A, Carnicelli V, Rutigliano G, Saponaro F, Zucchi R, Chiellini G. T1AM/TAAR1 system reduces inflammatory response and beta-amyloid toxicity in human microglial HMC3 cell line. Int J Mol Sci. 2023;24(14). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms241411569.
Ponnusamy K, Choi JN, Kim J, Lee SY, Lee CH. Microbial community and metabolomic comparison of irritable bowel syndrome faeces. J Med Microbiol. 2011;60(Pt 6):817–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1099/jmm.0.028126-0.
Pourhamzeh M, Moravej FG, Arabi M, Shahriari E, Mehrabi S, Ward R, Ahadi R, Joghataei MT. The roles of serotonin in neuropsychiatric disorders. Cell Mol Neurobiol. 2022;42(6):1671–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10571-021-01064-9.
Pretorius L, Smith C. Aspalathus linearis (Rooibos) and Agmatine May act synergistically to beneficially modulate intestinal tight junction integrity and inflammatory profile. Pharmaceuticals (Basel). 2022;15(9). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph15091097.
Pretorius L, Smith C. Tyramine-induced Gastrointestinal dysregulation is attenuated via estradiol associated mechanisms in a zebrafish larval model. Toxicol Appl Pharmacol. 2023;461:116399. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.taap.2023.116399.
Proulx JM, Park IW, Borgmann K. HIV-1 and methamphetamine co-treatment in primary human astrocytes: taargeting ER/UPR dysfunction. NeuroImmune Pharm Ther. 2024;3(2):139–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1515/nipt-2023-0020.
Raab S, Wang H, Uhles S, Cole N, Alvarez-Sanchez R, Kunnecke B, Ullmer C, Matile H, Bedoucha M, Norcross RD, Ottaway-Parker N, Perez-Tilve D, Conde Knape K, Tschop MH, Hoener MC, Sewing S. Incretin-like effects of small molecule trace amine-associated receptor 1 agonists. Mol Metab. 2016;5(1):47–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molmet.2015.09.015.
Rahangdale S, Deshmukh P, Sammeta S, Aglawe M, Kale M, Umekar M, Kotagale N, Taksande B. Agmatine modulation of gut-brain axis alleviates dysbiosis-induced depression-like behavior in rats. Eur J Pharmacol. 2024;981:176884. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2024.176884.
Raymond GV, Bauman ML, Kemper TL. Hippocampus in autism: a golgi analysis. Acta Neuropathol. 1996;91(1):117–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s004010050401.
Ribeiro DL, Machado ART, da Silva Machado C, Santos P, Aissa AF, Barcelos GRM, Antunes LMG. Analysis of the cytotoxic, genotoxic, mutagenic, and pro-oxidant effect of Synephrine, a component of thermogenic supplements, in human hepatic cells in vitro. Toxicology. 2019;422:25–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tox.2019.04.010.
Ricarte M, Tagkalidou N, Bellot M, Bedrossiantz J, Prats E, Gomez-Canela C, Garcia-Reyero N, Raldúa D. Short-and long-term neurobehavioral effects of developmental exposure to valproic acid in zebrafish. Int J Mol Sci. 2024;25(14):7688.
Rider JE, Hacker A, Mackintosh CA, Pegg AE, Woster PM, Casero RA Jr. Spermine and spermidine mediate protection against oxidative damage caused by hydrogen peroxide. Amino Acids. 2007;33(2):231–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00726-007-0513-4.
Robertson CE, Ratai EM, Kanwisher N. Reduced GABAergic action in the autistic brain. Curr Biol. 2016;26(1):80–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cub.2015.11.019.
Rogers G, Keating DJ, Young RL, Wong M-L, Licinio J, Wesselingh S. From gut dysbiosis to altered brain function and mental illness: mechanisms and pathways. Mol Psychiatry. 2016;21(6):738–48.
Roohi J, DeVincent CJ, Hatchwell E, Gadow KD. Association of a monoamine oxidase-A gene promoter polymorphism with ADHD and anxiety in boys with autism spectrum disorder. J Autism Dev Disord. 2009;39:67–74. https://link.springer.com/article/10.1007/s10803-008-0600-8.
Rose DR, Yang H, Serena G, Sturgeon C, Ma B, Careaga M, Hughes HK, Angkustsiri K, Rose M, Hertz-Picciotto I, Van de Water J, Hansen RL, Ravel J, Fasano A, Ashwood P. Differential immune responses and microbiota profiles in children with autism spectrum disorders and co-morbid Gastrointestinal symptoms. Brain Behav Immun. 2018a;70:354–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbi.2018.03.025.
Rossignol DA, Frye RE. Mitochondrial dysfunction in autism spectrum disorders: a systematic review and meta-analysis. Mol Psychiatry. 2012;17(3):290–314. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/mp.2010.136.
Rukavishnikov G, Leonova L, Kasyanov E, Leonov V, Neznanov N, Mazo G. Antimicrobial activity of antidepressants on normal gut microbiota: results of the in vitro study [Original Research]. Front Behav Neurosci. 2023;17. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnbeh.2023.1132127.
Sabelli HC, Javaid JI. Phenylethylamine modulation of affect: therapeutic and diagnostic implications. J Neuropsychiatry Clin Neurosci. 1995;7(1):6–14.
Santoru ML, Piras C, Murgia A, Palmas V, Camboni T, Liggi S, Ibba I, Lai MA, Orru S, Blois S, Loizedda AL, Griffin JL, Usai P, Caboni P, Atzori L, Manzin A. Cross sectional evaluation of the gut-microbiome metabolome axis in an Italian cohort of IBD patients. Sci Rep. 2017;7(1):9523. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-017-10034-5.
Schalbroeck R, van Velden FHP, de Geus-Oei LF, Yaqub M, van Amelsvoort T, Booij J, Selten JP. Striatal dopamine synthesis capacity in autism spectrum disorder and its relation with social defeat: an [(18)F]-FDOPA PET/CT study. Transl Psychiatry. 2021;11(1):47. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41398-020-01174-w.
Schiavi S, Iezzi D, Manduca A, Leone S, Melancia F, Carbone C, Petrella M, Mannaioni G, Masi A, Trezza V. Reward-related behavioral, neurochemical and electrophysiological changes in a rat model of autism based on prenatal exposure to valproic acid. Front Cell Neurosci. 2019;13:479.
Seeman MV. The gut Microbiome and antipsychotic treatment response. Behav Brain Res. 2021;396:112886. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbr.2020.112886.
Sengupta T, Mohanakumar KP. 2-Phenylethylamine, a constituent of chocolate and wine, causes mitochondrial complex-I Inhibition, generation of hydroxyl radicals and depletion of striatal biogenic amines leading to psycho-motor dysfunctions in Balb/c mice. Neurochem Int. 2010;57(6):637–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuint.2010.07.013.
Shajan B, Bastiampillai T, Hellyer SD, Nair PC. Unlocking the secrets of trace amine-associated receptor 1 agonists: new horizon in neuropsychiatric treatment. Front Psychiatry. 2024;15:1464550. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fpsyt.2024.1464550.
Shajib MS, Baranov A, Khan WI. Diverse effects of Gut-Derived serotonin in intestinal inflammation. ACS Chem Neurosci. 2017;8(5):920–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acschemneuro.6b00414.
Sharon G, Cruz NJ, Kang DW, Gandal MJ, Wang B, Kim YM, Zink EM, Casey CP, Taylor BC, Lane CJ, Bramer LM, Isern NG, Hoyt DW, Noecker C, Sweredoski MJ, Moradian A, Borenstein E, Jansson JK, Knight R, Mazmanian SK. Human gut microbiota from autism spectrum disorder promote behavioral symptoms in mice. Cell. 2019;177(6):1600–e16181617. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2019.05.004.
Sherer LM, Catudio Garrett E, Morgan HR, Brewer ED, Sirrs LA, Shearin HK, Williams JL, McCabe BD, Stowers RS, Certel SJ. Octopamine neuron dependent aggression requires dVGLUT from dual-transmitting neurons. PLoS Genet. 2020;16(2):e1008609. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pgen.1008609.
Shomrat T, Nesher N. Updated view on the relation of the pineal gland to autism spectrum disorders. Front Endocrinol (Lausanne). 2019;10:37. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fendo.2019.00037.
Simonoff E, Mowlem F, Pearson O, Anagnostou E, Donnelly C, Hollander E, King BH, McCracken JT, Scahill L, Sikich L. Citalopram did not significantly improve anxiety in children with autism spectrum disorder undergoing treatment for core symptoms: secondary analysis of a trial to reduce repetitive behaviors. J Child Adolesc Psychopharmacol. 2022;32(4):233–41.
Singh C, Bortolato M, Bali N, Godar SC, Scott AL, Chen K, Thompson RF, Shih JC. Cognitive abnormalities and hippocampal alterations in monoamine oxidase A and B knockout mice. Proc Natl Acad Sci U S A. 2013;110(31):12816–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1308037110.
Singh RK, Chang HW, Yan D, Lee KM, Ucmak D, Wong K, Abrouk M, Farahnik B, Nakamura M, Zhu TH, Bhutani T, Liao W. Influence of diet on the gut Microbiome and implications for human health. J Transl Med. 2017;15(1):73. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-017-1175-y.
Smith C, Krygsman A. Hoodia gordonii extract targets both adipose and muscle tissue to achieve weight loss in rats. J Ethnopharmacol. 2014;155(2):1284–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jep.2014.07.018.
Sorato E, Menazza S, Zulian A, Sabatelli P, Gualandi F, Merlini L, Bonaldo P, Canton M, Bernardi P, Di Lisa F. Monoamine oxidase inhibition prevents mitochondrial dysfunction and apoptosis in myoblasts from patients with collagen VI myopathies. Free Radic Biol Med. 2014;75:40–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.freeradbiomed.2014.07.006.
Spivak B, Golubchik P, Mozes T, Vered Y, Nechmad A, Weizman A, Strous RD. Low platelet-poor plasma levels of serotonin in adult autistic patients. Neuropsychobiology. 2004;50(2):157–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000079108.
Tallarico M, Leo A, Russo E, Citraro R, Palma E, De Sarro G. Seizure susceptibility to various convulsant stimuli in the BTBR mouse model of autism spectrum disorders [Original Research]. Front Pharmacol. 2023;14. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2023.1155729.
Tang G, Rios G, Kuo P, Akman SH, Rosoklija HO, Tanji G, Dwork K, Schon A, Dimauro EA, Goldman S, J., Sulzer D. Mitochondrial abnormalities in temporal lobe of autistic brain. Neurobiol Dis. 2013;54:349–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2013.01.006.
Teskey G, Anagnostou E, Mankad D, Smile S, Roberts W, Brian J, Bowdish DME, Foster JA. Intestinal permeability correlates with behavioural severity in very young children with ASD: A preliminary study. J Neuroimmunol. 2021;357:577607. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jneuroim.2021.577607.
Trinchese G, Cimmino F, Cavaliere G, Catapano A, Fogliano C, Lama A, Pirozzi C, Cristiano C, Russo R, Petrella L, Meli R, Raso GM, Crispino M, Avallone B, Mollica MP. The hepatic mitochondrial alterations exacerbate meta-inflammation in autism spectrum disorders. Antioxid (Basel). 2022;11(10). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox11101990.
Uljarević M, Hedley D, Rose-Foley K, Magiati I, Cai RY, Dissanayake C, Richdale A, Trollor J. Anxiety and depression from adolescence to old age in autism spectrum disorder. J Autism Dev Disord. 2020;50(9):3155–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10803-019-04084-z.
Vakilzadeh G, Maseko BC, Bartely TD, McLennan YA, Martínez-Cerdeño V. Increased number of excitatory synapsis and decreased number of inhibitory synapsis in the prefrontal cortex in autism. Cereb Cortex. 2024;34(13):121–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cercor/bhad268.
Valenzuela-Zamora AF, Ramirez-Valenzuela DG, Ramos-Jimenez A. Food selectivity and its implications associated with gastrointestinal disorders in children with autism spectrum disorders. Nutrients. 2022;14(13). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu14132660.
Vazifekhah S, Barfi S, Soleimany F, Aliakbar A, Zavvari F, Karimzadeh F. The plasma level of glutamic acid decarboxylase 65 (GAD65) increased in severely autistic Iranian children. Bratisl Lek Listy. 2022;123(5):347–51. https://doiorg.publicaciones.saludcastillayleon.es/10.4149/BLL_2022_054.
Venegas FC, Sanchez-Rodriguez R, Luisetto R, Angioni R, Viola A, Canton M. Oxidative stress by the mitochondrial monoamine oxidase B mediates calcium pyrophosphate crystal-induced arthritis. Arthritis Rheumatol. 2024;76(2):279–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.42697.
Wang L, Christophersen CT, Sorich MJ, Gerber JP, Angley MT, Conlon MA. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig Dis Sci. 2012;57(8):2096–102. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10620-012-2167-7.
Wang CC, Lin HC, Chan YH, Gean PW, Yang YK, Chen PS. 5-HT1A-receptor agonist modified amygdala activity and amygdala-associated social behavior in a valproate-induced rat autism model. Int J Neuropsychopharmacol. 2013;16(9):2027–39. https://doiorg.publicaciones.saludcastillayleon.es/10.1017/s1461145713000473.
Wang X, Kery R, Xiong Q. Synaptopathology in autism spectrum disorders: complex effects of synaptic genes on neural circuits. Prog Neuropsychopharmacol Biol Psychiatry. 2018a;84:398–415.
Wang R, Hausknecht K, Shen R-Y, Haj-Dahmane S. Potentiation of glutamatergic synaptic transmission onto dorsal Raphe serotonergic neurons in the valproic acid model of autism [Original Research]. Front Pharmacol. 2018b;9. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2018.01185.
Wang Y, Li N, Yang J-J, Zhao D-M, Chen B, Zhang G-Q, Chen S, Cao R-F, Yu H, Zhao C-Y, Zhao L, Ge Y-S, Liu Y, Zhang L-H, Hu W, Zhang L, Gai Z-T. Probiotics and fructo-oligosaccharide intervention modulate the microbiota-gut brain axis to improve autism spectrum reducing also the hyper-serotonergic state and the dopamine metabolism disorder. Pharmacol Res. 2020;157:104784. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.phrs.2020.104784.
Wang J, Ma B, Wang J, Zhang Z, Chen O. Global prevalence of autism spectrum disorder and its Gastrointestinal symptoms: A systematic review and meta-analysis. Front Psychiatry. 2022a;13:963102. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fpsyt.2022.963102.
Wang J, Fröhlich H, Torres FB, Silva RL, Poschet G, Agarwal A, Rappold GA. Mitochondrial dysfunction and oxidative stress contribute to cognitive and motor impairment in FOXP1 syndrome. Proc Natl Acad Sci U S A. 2022;119(8):e2112852119. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.2112852119
Wang J, Zou L, Jiang P, Yao M, Xu Q, Hong Q, Zhu J, Chi X. Vitamin A ameliorates valproic acid-induced autism-like symptoms in developing zebrafish larvae by attenuating oxidative stress and apoptosis. Neurotoxicology. 2024a;101:93–101. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuro.2023.12.015.
Wang L, Clark EA, Hanratty L, Koblan KS, Foley A, Dedic N, Bristow LJ. TAAR1 and 5-HT(1B) receptor agonists attenuate autism-like irritability and aggression in rats prenatally exposed to valproic acid. Pharmacol Biochem Behav. 2024b;245:173862. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pbb.2024.173862.
Williams BB, Van Benschoten AH, Cimermancic P, Donia MS, Zimmermann M, Taketani M, Ishihara A, Kashyap PC, Fraser JS, Fischbach MA. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe. 2014;16(4):495–503. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.chom.2014.09.001.
Win-Shwe T-T, Nway NC, Imai M, Lwin T-T, Mar O, Watanabe H. Social behavior, neuroimmune markers and glutamic acid decarboxylase levels in a rat model of valproic acid-induced autism. J Toxicol Sci. 2018;43(11):631–43.
Xiao-Mian S, Jing Y, Chongxuna Z, Min L, Hui-Xing D. Study of 99mTc-TRODAT-1 imaging on human brain with children autism by single photon emission computed tomography. Conf Proc IEEE Eng Med Biol Soc. 2005;5328–5330. https://doiorg.publicaciones.saludcastillayleon.es/10.1109/iembs.2005.1615684
Yang J, Chen Y, Xiong X, Zhou X, Han L, Ni L, Wang W, Wang X, Zhao L, Shao D, Huang C. Peptidome analysis reveals novel serum biomarkers for children with autism spectrum disorder in China. Proteom Clin Appl. 2018;12(5):e1700164. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/prca.201700164.
Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK, Hsiao EY. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161(2):264–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2015.02.047.
Yu H, Liu J, Yang A, Yang G, Yang W, Lei H, Quan J, Zhang Z. Lack of association between polymorphisms in dopa decarboxylase and dopamine Receptor-1 genes with childhood autism in Chinese Han population. J Child Neurol. 2016;31(5):560–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0883073815601496.
Zametkin AJ, Karoum F, Rapoport JL, Brown GL, Wyatt RJ. Phenylethylamine excretion in attention deficit disorder. J Am Acad Child Psychiatry. 1984;23(3):310–4.
Zhai L, Huang C, Ning Z, Zhang Y, Zhuang M, Yang W, Wang X, Wang J, Zhang L, Xiao H, Zhao L, Asthana P, Lam YY, Chow CFW, Huang J-d, Yuan S, Chan KM, Yuan C-S, Lau JY-N, Bian Z-x. Ruminococcus gnavus plays a pathogenic role in diarrhea-predominant irritable bowel syndrome by increasing serotonin biosynthesis. Cell Host Microbe. 2023;31(1):33–e4435. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.chom.2022.11.006.
Zhang P. Influence of foods and nutrition on the gut microbiome and implications for intestinal health. Int J Mol Sci. 2022;23(17). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23179588.
Zieminska E, Toczylowska B, Diamandakis D, Hilgier W, Filipkowski RK, Polowy R, Orzel J, Gorka M, Lazarewicz JW. Glutamate, glutamine and GABA levels in rat brain measured using MRS, HPLC and NMR methods in study of two models of autism [Original Research]. Front Mol Neurosci. 2018;11. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnmol.2018.00418.
Acknowledgements
None.
Funding
The South African National Research Foundation is acknowledged for funding.
Author information
Authors and Affiliations
Contributions
L.P. and C.S. jointly conceptualised the paper, L.P. wrote the first draft sections pertaining to gut health and trace amines, prepared figures for these sections and contributed substantially to the integrated interpretation of the reviewed literature. J.A.C. wrote the first draft of the section related to mitochondrial dysfunction and redox in autism spectrum disorder, including preparation of the relevant table and figures, as well as editing, technical formatting and references. A.P.D.S. wrote the first draft of the sections pertaining to neurological pathology in autism spectrum disorders, including the relevant table. C.S. also wrote the first draft sections for the introduction and therapeutic approaches focused on trace amine system modulation, as well as final conclusions, and substantially contributed to editing of the manuscript and figures. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable. No data is presented.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Pretorius, L., Coetzee, J.A., Santos, A.P.d. et al. Modulating autism spectrum disorder pathophysiology using a trace amine-focused approach: targeting the gut. Mol Med 31, 198 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-025-01232-3
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-025-01232-3