Next Article in Journal
Optimized Mass Spectrometry Detection of Thyroid Hormones and Polar Metabolites in Rodent Cerebrospinal Fluid
Previous Article in Journal
The Kidney–Gut Axis as a Novel Target for Nutritional Intervention to Counteract Chronic Kidney Disease Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lipids in Psychiatric Disorders: Functional and Potential Diagnostic Role as Blood Biomarkers

1
Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia
2
Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
3
Moscow Center for Healthcare Innovations, 123473 Moscow, Russia
4
Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academi of Science, 142290 Moscow, Russia
5
Russia Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 2, Kurchatov Square, 123182 Moscow, Russia
*
Author to whom correspondence should be addressed.
Metabolites 2024, 14(2), 80; https://doi.org/10.3390/metabo14020080
Submission received: 9 November 2023 / Revised: 7 December 2023 / Accepted: 19 December 2023 / Published: 23 January 2024

Abstract

:
Lipids are a crucial component of the human brain, serving important structural and functional roles. They are involved in cell function, myelination of neuronal projections, neurotransmission, neural plasticity, energy metabolism, and neuroinflammation. Despite their significance, the role of lipids in the development of mental disorders has not been well understood. This review focused on the potential use of lipids as blood biomarkers for common mental illnesses, such as major depressive disorder, anxiety disorders, bipolar disorder, and schizophrenia. This review also discussed the impact of commonly used psychiatric medications, such as neuroleptics and antidepressants, on lipid metabolism. The obtained data suggested that lipid biomarkers could be useful for diagnosing psychiatric diseases, but further research is needed to better understand the associations between blood lipids and mental disorders and to identify specific biomarker combinations for each disease.

1. Introduction

The most common mental illnesses are affective disorders and schizophrenia. Thus, among them are major depressive disorder (MDD), anxiety disorders (ADs), bipolar disorder (BPD), and schizophrenia (SCZ) [1]. These illnesses result in long-term disability and cause invalidity. Their disease courses have been characterized by emotional and cognitive disturbances, mood disorders, impaired functioning, and social isolation [1]. In recent years, advances in technology have allowed for the identification of many biological markers of mental illnesses, such as genomic, epigenomic, metabolic, and proteomic markers. However, much less attention has been paid to the lipid markers.
In 2005, the International Committee on the Classification and Nomenclature of Lipids identified eight classes of lipids; they have been displayed in the LIPID MAPS Structure Database [2]. Two fundamental ‘building blocks’ (ketoacyl groups and isoprene groups) form the basis of the LIPID MAPS classification system. Therefore, lipids are defined as hydrophobic or amphipathic small molecules that can arise, in whole or in part, from two types of condensation: based on the carbanions of ketoacyl thioethers and/or based on the carbocations of isoprene units. This classification system segregates eight categories of lipids: fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, saccharolipids, polyketides (derived from the condensation of ketoacyl subunits), sterol lipids, and prenol lipids (derived from the condensation of isoprene subunits). The classification of these lipids is shown in Figure 1.
The brain is the organ that is enriched in lipids. Only adipose tissue contains a larger amount of lipids compared to brain tissue [3]. The entire variety of brain lipids is involved in a range of essential processes, the disruption of which can cause significant damage to the central nervous system (CNS).
Lipids are the structural components of cell membranes, which are involved in a set of processes in the cell, such as myelination, neurotransmission, synaptic plasticity, energy, metabolic processes, and inflammatory processes. Interventions in these processes may influence the development of psychiatric disorders and contribute to their pathogenesis [4].
The state of the cell membrane is extremely important for the functions of neurons and glial cells. In nerve cells, lipids comprise 50–60% of cell membrane components [5]. Lipids form a phospholipid bilayer, the basic structural unit of the membrane, which participates in the regulation of permeability. The three major classes of membrane lipids are glycerophospholipids (e.g., phosphatidylcholine (PC), phosphatidylserine (PS), phosphatidylethanolamine (PE), and phosphatidylinositol (PI)), cholesterol, and sphingolipids. The outer layer of the plasma membrane mainly consists of PC and sphingomyelin (SM); PE and PS represent the predominant phospholipids of the inner layer. PI is also localized in the inner part of the membrane and plays an important role in cell signaling [5]. Sphingolipids (SPs) contain long-chain fatty acids, which provide inter-lipid associations in the lipid bilayer [6]. Cholesterol acts as a “strengthening link” in the structures of membranes, providing them with the necessary strength and stability. Cholesterol affects membrane fluidity and increases the level of friction between membrane flaps [7]. The relative size and degree of fatty acid saturation in lipids affect membrane curvature, fluidity, and thickness [8]. Moreover, lipids can modulate the activity of membrane proteins with lipid-binding domains by recruiting them to specific membrane compartments or subdomains [9]. Lipids also participate in intracellular signaling, where they act as secondary messengers. The most common are diacylglycerol (DG) and inositol triphosphate (IP3) [10].
Myelination plays a crucial role in signal transduction and the proper functioning of the CNS. The main function of myelin is to provide electrical insulation of axons for sufficiently efficient transmission of action potentials. Myelin is characterized by an extremely high lipid content (~80% of dry weight) and a peculiar lipid composition in which the ratio of cholesterol to phospholipids (mainly ethanolamine phosphatide and phosphatidylcholine) to glycolipids (e.g., galactosylceramide and sulfatide) is approximately 2:2:1. Specific SPs and glycerides are substances that cover nerve fibers and accelerate the transmission of nerve impulses [11]. Myelin is particularly high in saturated and monounsaturated lower and higher fatty acids. Phospholipids containing such fatty acids may contribute to the electrical insulation of axons by reducing their membrane fluidity [12].
The functioning and activity of cellular receptors depend on the interactions between the proteins and lipids that comprise the bilipid layer of the cell membrane [13]. Firstly, lipids are involved in the regulation of synapse development and plasticity. For example, levels of tropomyosin receptor kinase B, a crucial protein in synapse development, are regulated via cholesterol levels [14]. Secondly, lipids participate in the release of presynaptic vesicles [15]. Third, lipids regulate neurotransmitter receptors independently, mostly through direct interactions. For example, cholesterol has been shown to function as a direct allosteric regulator of G protein-coupled receptors [16]. Abnormal phospholipid changes have been reported to disturb the functions of ion channels, neurotransmitters, and cell signaling [17].
Lipids also control neuroplasticity. Glycerophospholipids (GPs) and phosphoinositides are important regulators of dendritic spine plasticity. Lipids also influence dendritic spine plasticity by covalently binding to key synaptic proteins via palmitoylation, which can reversibly modulate protein function [18]. Neutral sphingomyelinases also regulate synaptic potentiation. Previous studies have demonstrated at least two different functions of lipids in plasticity processes: altering the functions of synaptic proteins through the palmitoylation mechanism and linking cytoskeletal regulators to membrane remodeling [19]. SPs modulate structural plasticity and neuronal dynamics through lipid–cytoskeletal interactions [20]. Neuronal activity can induce rapid changes in lipid metabolism. It rapidly modulates GP and SP levels. Several studies have shown the effects of ceramide (Cer) metabolism on neuronal susceptibility to death and plasticity process [21]. Cholesterol-deficient neuronal cells exhibit reduced synaptic transmission and impaired synaptic plasticity [4].
Brain tissue needs a large amount of energy. Neurons in the adult brain mainly depend on glucose as an energy source. However, about 20% of the total energy requirements of the adult brain are provided through the oxidation of fatty acids. It has been considered that fatty acid oxidation occurs almost exclusively in astrocytes, and carnitine and fatty acids can be transported from the blood to the astrocytes [22]. Brain mitochondria are characterized by a number of special features. The lipid-to-protein ratio of phospholipids, or cholesterol, is lower in brain mitochondria compared to other organelles [23]. In particular, mitochondria do not contain SM and glycosphingolipids. The major phospholipids of mitochondrial membranes are PC and PE, mainly located in the inner membrane. PI and PS are almost equally distributed on both membranes [23].
Inflammatory processes have been observed during many mental illnesses [24]. Microglial cells are activated during inflammation and perform phagocytosis to counteract inflammation [25]. Once abnormalities are detected, complex remodeling of the lipid composition of microglial cells occurs, providing inflammatory signaling and effector functions [26]. Microglial cells contain receptors for low-density lipoproteins (LDLs) that regulate inflammatory signaling [27]. Cers also promote inflammation and microglia activation [21]. Several lipids represent the sources of pro-inflammatory cytokines that contribute to pathologic neuroinflammatory processes. For example, under certain conditions, arachidonic acid (AA) can produce pro-inflammatory mediators, such as prostaglandins (PGs) and leukotrienes [28]. Long-chain PUFAs (polyunsaturated fatty acids), representing a source of eicosanoids and docosanoids, play an important role in neuroprotective and anti-inflammatory effects in the CNS [4]. SMs also participate in neuroinflammation through cytokine release, microglia activation, and other immune processes [29].
The role of lipids in physiological processes important for the functioning of the nervous system is summarized in Figure 2.
Therefore, lipids play a major role in the functioning of the nervous system, and alterations in lipid metabolism may influence mental disorder development.
Changes in lipid metabolism in brain tissue may contribute to the pathogenesis of neuropsychiatric diseases. The presence of the blood–brain barrier prevents the free penetration of compounds into the brain. Small lipophilic molecules can pass into brain tissue via passive diffusion; at the same time, all other lipids enter the brain via transcytosis or special transport proteins [23]. For example, unbound long-chain fatty acids can diffuse through the membranes. Meanwhile, cholesterol is almost entirely synthesized in the brain, so its concentration in the blood cannot reflect the processes occurring in the central nervous system. It has been hypothesized that fatty acid metabolism in specific regions of the hypothalamus functions as sensors of nutrient availability that are involved in integrating energy balance through the control of multiple nutritional and hormonal signals. In other brain regions, no differences in glucose and fatty acid metabolism were found, depending on nutritional status [23]. The study of brain lipid composition is an important and urgent task from the point of view of fundamental science. However, for practical purposes, it is necessary to study its associations with blood lipid content. In this review, we have chosen to specifically focus on blood biomarkers in order to determine the potential of using lipids in the potential diagnostics of mental illness. Our review summarized recent data on the associations of lipid blood composition with mental disorders such as SCZ, BPD, MDD, and AD. The first part of this review addressed the effects of changes in blood lipid composition on mental illness, while the second part of this review discussed the effects of drugs used in the treatment of mental disorders on lipid metabolism.

2. Lipids and Their Role in Neuropsychiatric Disorders

Lipids influence several pathophysiologic pathways that are involved in the development of psychiatric illnesses [30]. The most pronounced effect in the literature has been shown for SCZ and MDD.
Disruption of lipid function is one of the components of SCZ pathogenesis [31]. Yao et al. demonstrated a direct link between abnormal phospholipid levels and disrupted neurochemical parameters, such as SCZ-associated abnormal dopamine and glutamate levels [32]. Phospholipid metabolism abnormalities occur during the progression of SCZ. Most notably, phospholipase A2 (PLA2) activity increases and the level of PUFA integration into phospholipids decreases [31]. The association between PLA2 activity and the dopamine system has also been demonstrated [33]. In particular, it was demonstrated that PUFA dissociation and saturated fatty acid (SFA) incorporation in membrane phospholipids are enhanced in SCZ patients. Decreased levels of membrane phospholipid precursors in the brains of SCZ patients indicate reduced synthesis of PC and PE. Abnormal expression of enzymes and impaired homeostasis of membrane lipids in patients have been associated with the imbalance of phospholipid breakdown and remodeling under the influence of increased oxidative stress. Phospholipid metabolism plays a critical role in the process of synaptic growth, and its dysfunction has been associated with abnormal neuronal development in SCZ [17]. SM and Cers also exert an effect on the presynaptic release of dopamine [34].
In the field of depression research, Andreas Walther et al. proposed their model of lipid involvement in the pathogenesis of depression [35]. This model is based on the chronic stress effects. Chronic stress has been thought to trigger two main pathways: the hypothalamic–pituitary–adrenal axis (HPA) and neuroinflammation [35].
Chronic stress leads to HPA hyperactivity. Elevated glucocorticoid levels increase phospholipase D activity [35]. Increased phospholipase D activity enhances the conversion of PC and PE into phosphatidic acid, as well as lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE). Due to its chemical properties, phosphatidic acid is rapidly converted into DG. DG, LPC, and LPE cause membrane buckling and destabilization, allowing for a greater glucocorticoid influx into the cell. Together with the above mechanism, elevated glucocorticoid levels decrease triacylglycerol hydrolase expression and enhance triacylglycerol (TAG) biosynthesis by increasing the level of diacylglycerolacyltransferase 2. Decreased triacylglycerol hydrolase expression and increased TAG biosynthesis raise the level of TAG [35]. TAG, in turn, is associated with increased glucocorticoid levels.
Chronic stress also leads to the dysregulation of inflammation. Excess pro-inflammatory cytokines and phasic reagents increase the level of PLA2 [35]. Increased PLA2 activity induces the conversion of PC-containing linoleic acid into AA. AA is subsequently converted into PGs, including pro-inflammatory cytokines (e.g., PGA2, PGD2, PGE2, PGF2, PGH2, and PGI2) [35]. PGs further enhance inflammatory responses.
Increased saturated fatty acid-rich phospholipids, namely lysoPS (16:0), lysoPS (18:0), and SM (24:0), have been associated with inflammation and oxidative stress responses in depressed patients [35]. The elevation of δ-6 desaturase activity in patients with depressive symptoms has been demonstrated. δ-6 desaturase converts linoleic acid into AA, which is a precursor of pro-inflammatory products [36].
Several studies have suggested that omega-3 fatty acid deficiency can decrease dopamine levels, D2 receptor expression and mRNA, presynaptic dopamine vesicle amount, and increase dopamine cleavage [37]. Moreover, its deficiency also downregulates tyrosine hydroxylase activity, which results in reduced dopamine levels and depressive symptomatology development [38].
Increased Cer concentrations may also contribute to the progression of depression, as Cers may affect dopamine transporter function by decreasing dopamine transport and increasing 5HT transport [39]. Moreover, it has been reported that increased Cers may affect monoamine neurotransmitter reuptake and initiate a biological cascade that leads to the downregulation of serotoninergic neurotransmission, which represents another pathophysiologic hallmark of depression [38].
Most of the changes accompanying psychiatric disorders primarily affect brain structures. Lipid metabolism impairments during mental diseases also primarily occur in the CNS tissues. Nevertheless, it is necessary to use the available research methods for diagnostic purposes in clinical practice. In this regard, the second part of this review is devoted to the study of blood lipid composition in mental disorders, as these data may have diagnostic value. Although the role of lipids in the pathophysiological pathways of mental illnesses have not been sufficiently investigated and mainly concern the mechanisms of schizophrenia and depression, a significant number of studies have analyzed lipids as potential biomarkers of psychiatric diseases.
Changes in lipid profiles have been consistently observed in the blood serum and plasma across patients with psychiatric diagnoses.
In particular, a lot of studies have focused on identifying reliable blood lipid indicators in SCZ. In total, over 29 studies have assessed lipid changes in the blood of patients with SCZ compared to healthy controls (Table S1). Two studies were conducted without a control group (Table S1). Most of the observed studies included patients that received treatment, but three of them also included patients with first-episode psychosis (FEP) (Table S1). However, only nine of the described studies involved more than 100 individuals (Table S1).
Nine studies evaluated the broad panels of different lipid markers in patients with SCZ [31,34,40,41,42,43,44,45,46] and detected statistical differences in the following lipid classes: fatty acyls, sterols, glycerolipids, sphingolipids, glycerophospholipids, and products of lipid metabolism. Several studies included information about lipids associated with the membranes of erythrocytes [31,47,48]. The most consistent data from the reviewed studies were obtained for PC, PE, SM, and triacylglycerols (TGs). In particular, reduction in these lipid species was mostly demonstrated. Malondialdehyde—the marker of oxidative stress—was also increased in all the concerned studies. Moreover, a meta-analysis considering malondialdehyde in SCZ was conducted [49]. It was shown that medically treated SCZ patients were more affected by the increased oxidative stress, but malondialdehyde levels were elevated in both the treated and untreated groups, in contrast to other markers of oxidative stress. Bile acids were investigated in two studies, and their levels decreased in both of them [40,50]. Calcifediol was reduced in one observed study [50]. High-density lipoprotein (HDL) was decreased in two studies [51,52]. Studies considering fatty acyl and Cer levels received inconsistent results: some lipid types were decreased and some were increased. The levels of PC and PE did not differ significantly in FEP patients and medicated patients [53,54]. The data addressed to PUFA concentrations were found to be inconsistent.
It is worth highlighting those studies that do not identify differences between healthy individuals and patients with SCZ but rather make associations between blood lipids and symptoms of the illness. In some studies, changes in lipid levels in SCZ patients have been associated with Positive and Negative Syndrome Scale (PANSS) scores. For example, such correlations have been detected for shorter-chain TGs [41] and oxysterols [55]. Nandeesha (2023) [52] showed that total cholesterol (TC) and TG levels were negatively correlated with cognitive scores. Plasma calcifediol levels and the ratio of cholestanol to tchol were found to be negatively correlated with Montreal Cognitive Assessment (MOCA) scores [55]. Baseline membrane linoleic acid levels in SCZ with ultra-high risk (UHR) were associated with conversion to psychosis. Sterol, fatty acid, and phospholipid membrane compositions improved the prediction of the psychosis onset [47]. TC levels were positively associated with the Repeated Battery for the Assessment of Neuropsychological Status (RBANS) subscale scores of immediate memory and language [56]. These results further suggest the potential use of blood lipid profiles for the assessment of SCZ symptomatology.
For MDD, we described nineteen studies (Table S1), which included one study considering postpartum depression [57] and four studies considering depression symptoms in the healthy population (Table S1). Four studies above them were conducted on drug-naïve patients (Table S1). Only four studies on MDD patients included more than one hundred individuals (Table S1). Six studies evaluated a broad panel of different lipid markers [34,58,59,60,61,62]. Consensual data were received for LPC and LPE measurements, which were increased in the observed studies. PC and malondialdehyde levels were also mostly elevated in the described studies. On the contrary, acylcarnitine (CAR), calcifediol, SM, and bile acids were mostly decreased in the reviewed studies. Inconsistent data were received for PE, PI, Cers, TGs, PUFAs, and SFAs. Two studies on MDD patients and one on postpartum depression individuals reported a reduction in HDL levels. One study indicated an increased level of LDL. Researchers have investigated the levels of cholesteryl ester (CE), TC, sterols, and calcifediol in healthy people with depressive symptoms. Associations with mental symptoms were shown for TC and sterol lipids in women. Several studies have indicated an association between the lipid concentrations of octadecyl-phosphatidylethanolamine (PE-O) [58], SM, and PC-O [63] and symptom severity according to the specific scales.
Fourteen studies were dedicated to the investigation of lipid changes in BPD, including two conducted on drug-naïve patients (Table S1). Compared to the studies on SCZ and MDD, fewer studies were conducted on patients with BPD. Only one study evaluated the associations of CAR, CE, calcifediol, PE, PC, LPC, LPE, PS, and SM with disease symptoms (Table S1). Cer and PI levels were increased in two and three studies, respectively. PUFA, TC, and TG changes showed inconsistent associations.
We described 24 studies devoted to the evaluation of blood lipid biomarkers in AD (Table S1). Eleven studies considered blood lipid constitution during general anxiety disorder (GAD), or AD, including one performed on pregnant women (Table S1). One study described post-stroke anxiety [64]. Some studies have focused on comorbid psychiatric pathology, such as comorbid AD and MDD or comorbid AD and Parkinson’s disease. The other six works included population studies, which investigated blood lipid biomarkers in individuals with anxiety symptoms (Table S1). The majority of this research included healthy control or other comparison groups with mental disorders, and only eight did not. Six studies have been conducted on a broad sample of individuals (more than 300) (Table S1). Nevertheless, only a few large metabolomic studies assessing lipid blood constitution in AD were performed [63,65,66]. These broad metabolomic studies have revealed changes in a number of lipids of various classes: fatty acyls, sterol lipids, GP, SP, and glycerolipids [63,65,66].
Most of these studies have focused on investigating the changes in lipoproteins, TGs, and cholesterol. Regarding lipoprotein levels, the results were questionable and multidirectional. Nevertheless, in almost all of the papers that were reviewed, anxiety symptomatology was accompanied by an increase in TGs [67,68,69,70,71] and, in only one, by a reduction [65]. For cholesterol, the reviewed results were also found to be inconsistent. A number of studies have investigated the change in PUFAs in blood during anxiety states and mainly demonstrated their decrease [65,72,73]. Regarding the SFA elevation, multidirectional results have been shown. One study identified a decreased carnitine (propionylcarnitine) level [66]. Bile acid changes were also shown in one study reflecting anxiety symptomatology in MDD [74]. Alterations in Cers have also been found in comorbid pathologies. In particular, Xing et al. demonstrated a positive association of Cer C 20:0 levels with anxiety symptoms in Parkinson’s disease [75]. Unidirectional changes were detected when studying the levels of 20-oxo-22,23,24,25,26,27-hexanorvitamin D3 and malondialdehyde. Thus, decreased levels of 20-oxo-22,23,24,25,26,26,27-hexanorvitamin D3 accompanied anxiety symptomatology [76,77]. Malondialdehyde, on the contrary, was increased in AD patients [62,77]. No changes were identified for calcifediol [78].
Among the addressed studies, one was dedicated to the transdiagnostic lipid markers between four illnesses: MDD, BPD, AD, and SCZ [79]. The authors tried to indicate these transdiagnostic lipid subtypes. Researchers have suggested that 10 lipids can be used for diagnostics across psychiatric disorders. Along with these lipid types, the marker of oxidative stress, malondialdehyde, was increased in all the mentioned psychiatric disorders according to the observed studies. The levels of CAR and SM were also decreased in all reviewed psychiatric disorders. Thus, a number of lipid biomarkers were altered in these mental illnesses. The unidirectionality of some of these changes may indicate the diagnostic potential of blood lipid estimation.
Taken together, these studies highlight the need for systematic analysis of the robustness of observed lipidome alterations and their specificity to a single disorder. Future studies would also need to consider the comorbidities commonly linked to psychiatric disorders. The summarized information on lipids as diagnostic biomarkers is presented in Table 1. The description of all the reviewed studies is presented in Table S1.

3. Effect of Medications on Lipid Metabolism

Among the possible confounding factors affecting lipidome measurements, psychopharmacologic treatment effects have been particularly well documented. In this section, two classes of psychotropic drugs will be considered: neuroleptics and antidepressants.

3.1. Antipsychotics

Treatment with antipsychotic drugs has been associated with metabolic disturbances, and side effects of these drugs, such as obesity, hypertriglyceridemia, and glucose dysregulation, have been linked to these processes [129]. Among others, neuroleptic therapy affects lipid homeostasis [130,131]. The large amount of data concerns cholesterol and lipoprotein changes. In particular, it was shown that typical and atypical antipsychotics increase the levels of cholesterol, TG, and LDL. Antipsychotic drugs produce metabolic side effects with a different extent depending on their type. For example, olanzapine and clozapine demonstrated the worst side effect profiles, while the most favorable profiles have been shown for aripiprazole, brexiprazole, cariprazine, lurasidone, and ziprasidone [131]. The negative effect of olanzapine on lipid homeostasis has been detected even in patients with a first episode of psychosis [132]. Comparative research revealed that haloperidol and quetiapine can increase lipid levels; ziprasidone probably improves lipid levels, while risperidone can produce both effects [133]. Both atypical and typical antipsychotics may worsen lipid peroxidation [49]. However, the potential mechanisms of dyslipidemia induced by antipsychotics are of the greatest interest.
Neuroleptics inhibit cholesterol biosynthesis in vitro by reducing the activities of the enzymes involved in this pathway, leading to the accumulation of various sterol intermediates [134,135]. In vitro, clozapine was found to be the most prominent stimulator of fatty acid, TG, and phospholipid biosynthesis. Antipsychotic drugs induce the inhibition of cholesterol biosynthesis, affecting the same enzymes with a different relative activity: ziprasidone > haloperidol > risperidone [129]. Inhibition of its biosynthesis leads to impaired hormone signaling for insulin and somatostatin in vitro [135]. In addition, antipsychotics disrupt intracellular cholesterol trafficking by inhibiting cholesterol efflux from endolysosomes, thereby reducing the transport of endocytosed LDL cholesterol into the endoplasmic reticulum and Golgi apparatus [136]. As cationic amphiphilic drugs, antipsychotics alkalinize lysosomes, affecting lysosomal function, as has been shown with haloperidol [134]. In addition, antipsychotics increase LDL receptor transcription, thereby stimulating LDL endocytosis and exacerbating the intracellular accumulation of LDL-derived lipids [136].
Chlorpromazine and the antidepressant imipramine can increase cholesterol content in lysosomes and disrupt sterol regulatory element-binding protein (SREBP) mediated by the cholesterol-sensing system in the endoplasmic reticulum. Antipsychotic drugs induce the transcriptional activation of cholesterol and FA biosynthesis genes under the control of the transcription factors SREBP1 and SREBP2 [129]. It was shown that treatment with clozapine or risperidone enhanced lipogenesis and cholesterogenesis via the inhibition of PGRMC1/INSIG-2 and activation of SCAP/SREBP expression in rats. However, similar metabolic disturbances were not observed in rats treated with aripiprazole or haloperidol. Moreover, additional treatment with mifepristone effectively reversed the lipid abnormalities induced by atypical antipsychotics [137]. Thus, neuroleptics increase lipogenesis, decrease lipolysis, and enhance the antilipolytic effect of insulin in adipocytes. As a result, this leads to lipid accumulation in adipocytes [133].
Antipsychotics can alter membrane compartmentalization, which may differentially modulate the signaling cascade of the dopamine D2 receptor [138]. Antipsychotics demonstrate a higher affinity for SM compared to phosphatidylcholine. Cholesterol increased the affinity of these drugs to the lipid bilayer and resulted in the following ranking of neuroleptics by this factor and corresponding structural changes: risperidone >9-OH- risperidone> haloperidol. Studies performed on single lipids and mixtures consisting of lipids of biological origin demonstrated that antipsychotics can also modify D2 receptor activity by altering the lipid environment of the receptor [138].
The disruption of other lipid species during antipsychotic treatment has also been shown in human studies. SCZ patients treated with risperidone showed decreased levels of dihydroceramide, very long-chain Cers, and lysoPC in mononuclear cells [136]. Phosphorous magnetic resonance spectroscopy (2D chemical shift imaging (CSI)) allows for the study of membrane phospholipids and high-energy phosphates in vivo. Using this technique, the authors showed that risperidone stimulates the remodeling of neuronal and synaptic phospholipids. This drug increased the level of adenosine triphosphate in the left dorsolateral prefrontal cortex, left anterior temporal cortex, left insular cortex, basal ganglia, and anterior cerebellum, and increased the levels of phosphomonoesters, phosphodiesters, and phosphocreatine in these brain regions [139].
Neuroleptics, at doses recommended for the treatment of acute episodes of SCZ, can also cause distinct changes in the plasma levels of lipid peroxidation products. For example, quetiapine, which is also used in the treatment of depressive disorders, demonstrates the strongest antioxidant properties, in contrast to the pro-oxidant effects of risperidone, ziprasidone, haloperidol, and clozapine at low doses [140].
Almeida [141] investigated the lipidome changes in their blood plasma samples before and after 6 weeks of treatment with either risperidone, olanzapine, or quetiapine. Risperidone affected DG, ceramide 1-phosphates, TG, SM, and ceramide phosphoinositols. Olanzapine mainly affected the PS, PC, glycerophosphatidic acid PA, and glycerophosphoglycerol PG lipid classes. Quetiapine affected the lipid profiles of patients to a smaller extent. After medication with risperidone or olanzapine, the levels of LysoPC, PC, PE, C16 sphinganine, and adrenic acid were significantly increased, while the levels of linoleic acid, oleic acid, palmitoleic acid, γ-linolenic acid, and oxoglutaric acid were significantly decreased [50].

3.2. Antidepressants

Along with antipsychotic drugs, antidepressants also cause changes in blood lipid composition. Many studies have shown the negative effect of antidepressants on lipid metabolism [142,143]. In particular, tricyclic antidepressants and mirtazapine treatment induce weight gain increases in TG and LDL levels [144]. Cholesterol elevation and a temporary increase in TGs were also observed after mirtazapine treatment in healthy volunteers compared to the placebo group [145]. Kopf et al. [146] have also demonstrated the increase in TG levels following their amitriptyline therapy. However, these authors considered these changes as a positive therapeutic effect due to the fact that this increase was only observed in patients responding to this therapy.
Despite the previously assumed absence of a pronounced negative effect of selective serotonin reuptake inhibitors on fat metabolism, many studies have shown the opposite. Thus, a number of studies revealed an increase in the level of TC, concentration of TGs, and LDL in the blood of depressed patients following treatment with selective serotonin reuptake inhibitors [143,147,148,149]. Various effects have been shown for different types of antidepressants. For example, Olguner Eker et al. [149] detected an increase in HDL after their escitalopram application, but not following the applications of fluoxetine, sertraline, and venlafaxine. In Beyazyuz’s work, metabolic changes were only observed after the use of paroxetine, citalopram, and escitalopram, but not fluoxetine [148]. Conversely, several studies did not reveal pronounced changes in lipid metabolism following treatment with antidepressants of different classes [150]. The presumed mechanism of the negative effect of selective serotonin reuptake inhibitors on lipid metabolism is the excessive accumulation of TGs in the liver tissues. Thus, it has been shown that fluoxetine injection causes an increase in the levels of lipogenic enzymes and a decrease in the levels of lipolytic enzymes in the livers of mice with modeled depression [143]. Previously, an in vitro experiment also demonstrated that fluoxetine induces lipid accumulation in primary hepatocyte cultures by inhibiting the AMP-activated protein kinase signaling pathway [151].
Along with the negative effects on lipid metabolism, some studies, on the contrary, have revealed a positive role of antidepressants. For example, Hsiao showed that venlafaxine treatment induces a decrease in depressive symptoms accompanied by a decrease in dehydroepiandrosterone, a steroid hormone whose precursor is cholesterol [152]. Venlafaxine treatment in the drug-sensitive patients also caused a decrease in AA levels compared to the drug-resistant group [153]. In addition, Hummel et al. demonstrated that the improvement of patients’ symptoms after the antidepressant treatment was accompanied by an improvement in the LDL/HDL ratio [154].
Several studies have suggested that lipid metabolism itself may play a significant role in response to antidepressant therapy. For example, Sonawalla et al. showed that patients with elevated cholesterol levels may demonstrate lower response to fluoxetine therapy [155]. The authors of another study suggested that baseline-elevated LDL level may increase the binding ability of the serotonin type 1A receptor and thus provide a more pronounced therapeutic effect [156].
Extensive metabolomic studies of depression and antidepressant therapies have identified a number of lipid metabolites that undergo changes with treatment. Analyses of the plasma lipid profile following citalopram and escitalopram administration revealed changes in several types of PC, namely alkyl-PC, lysopPC, and SM (e.g., increases in PC (36:2), PC (30:0), PC (34:3), PC-O (34:2), PC-O (36:3), LPC (24:0), and SM (24:0) and decreases in PC (36:4), PC (38:6), LPC (20:4), and SM (18:1)) [157,158,159]. In view of the fact that phosphatidylcholines are involved in membrane construction and remodeling, the increase in PC may be associated with an increase in the activity of membrane proteins, such as carnitine palmitoyltransferase 1, which is involved in fatty acid beta-oxidation [159]. The increase in the important antioxidant phospholipid PC-O can probably be related to counteracting the oxidative stress that accompanies depressive disorders. Thus, such changes in lipid profiles may indicate a favorable outcome of antidepressant treatments.
However, most studies have not compared patients with diagnosed depression following their treatment and a healthy control group. This makes it difficult to assess the ability of antidepressants to affect lipid metabolism and restore normal lipid profiles [158].

4. Conclusions

Lipids represent an extremely significant structural component of the brain, performing various functions related to both the maintenance of cellular function and nerve cell physiology. However, the influence of lipids on the pathologic pathway mechanisms of psychiatric diseases has been undeservedly poorly studied. Disruption of lipid metabolism leads to impaired brain function and the development of neuropsychiatric diseases. Despite the large amount of data concerning the properties of lipids, many of their functions remain poorly understood. In this review, we have demonstrated the role of lipids as potential biomarkers of the most common psychiatric diseases.
We identified a number of studies that observed changes in lipids of various classes during the progression of mental illnesses. Moreover, the altered lipid profile induced by medications, such as antipsychotics and antidepressants, was also described. Lipid markers have the potential to serve as biomarkers for the diagnosis and prognosis of mental illnesses. They can also provide valuable insights into the underlying biological mechanisms of these disorders. In conclusion, while much progress has been made in identifying biological markers of mental illnesses, there is still a need for further research on lipid markers. Understanding the role of lipids in the pathophysiology of mental illnesses could lead to the development of novel diagnostic tools and therapeutic interventions.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/metabo14020080/s1, Table S1: Studies assessing blood lipid alterations in psychiatric disorders at the level of individual lipid species.

Author Contributions

Writing—original draft preparation, Y.Z., V.U. and A.O.; literature search, A.T., O.A., V.S., I.M. and D.R.; writing—review and editing, A.M.; supervision, G.K. and A.M.; project administration, A.G. and I.A.; funding acquisition, A.G. and I.A. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by grant NO 2707-2/22 from the Moscow Center for Innovative Technologies in Healthcare. The publication of this manuscript was sponsored by the Moscow Center for Innovative Technologies in Healthcare.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Acknowledgments

We would like to thank Philipp Khaitovich for his helpful edits.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAArachidonic acid
ADAnxiety disorder
ApoApolipoprotein
BPDBipolar disorder
CARAcylcarnitine
CDCAChenodeoxycholic acid
CECholesteryl ester
CerCeramide
CNSCentral nervous system
DGDiacylglycerol
DHADocosahexaenoic acid
FEPFirst-episode psychosis
GADGeneral anxiety disorder
GLCAGlycolithocholic acid
GMMonosialotetrahexosylganglioside
GPGlycerophospholipid
HDLHigh-density lipoprotein
HPAHypothalamic–pituitary–adrenal axis
IP3Inositol triphosphate
LCALithocholic acid
LDLLow-density lipoprotein
LPCLysophosphatidylcholine
LPC-OLysoplasmanyl-phosphatidylcholine
LPC-PLysoplasmenyl-phosphatidylcholine
LPELysophosphatidylethanolamine
LPILysophosphatidylinositol
MDDMajor depressive disorder
MOCAMontreal Cognitive Assessment
PANSSPositive and Negative Syndrome Scale
PCPhosphatidylcholine
PC-OOctadecyl-phosphatidylcholine
PC-PPlasmenyl-phosphatidylcholine
PEPhosphatidylethanolamine
PE-OOctadecyl-phosphatidylethanolamine
PE-PPlasmenyl-phosphatidylethanolamine
PGProstaglandin
PIPhosphatidylinositol
PIP Phosphatidylinositol phosphate
PLA2Phospholipase A2
PSPhosphatidylserine
PUFAsPolyunsaturated fatty acids
RBANSRepeated Battery for the Assessment of Neuropsychological Status
SCZSchizophrenia
SFASaturated fatty acid
SMSphingomyelin
SPSphingolipid
SREBPSterol regulatory element-binding protein
TAGTriacylglycerol
TCTotal cholesterol
TGTriacylglycerol
TGHTriacylglycerol hydrolase
TLCATaurolithocholic acid
UHRUltra-high risk
VLDLVery low-density lipoprotein

References

  1. WHO. Transforming Mental Health for All; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  2. LIPID MAPS. Available online: https://www.lipidmaps.org/ (accessed on 8 November 2023).
  3. Mesa-Herrera, F.; Taoro-González, L.; Valdés-Baizabal, C.; Diaz, M.; Marín, R. Lipid and Lipid Raft Alteration in Aging and Neurodegenerative Diseases: A Window for the Development of New Biomarkers. Int. J. Mol. Sci. 2019, 20, 3810. [Google Scholar] [CrossRef] [PubMed]
  4. Yoon, J.H.; Seo, Y.; Jo, Y.S.; Lee, S.; Cho, E.; Cazenave-Gassiot, A.; Shin, Y.S.; Moon, M.H.; An, H.J.; Wenk, M.R.; et al. Brain Lipidomics: From Functional Landscape to Clinical Significance. Sci. Adv. 2022, 8, eadc9317. [Google Scholar] [CrossRef] [PubMed]
  5. Cooper, G.M. The Cell: A Molecular Approach, 2nd ed.; Sinauer Associates: Sunderland, MA, USA, 2000; ISBN 0-87893-106. [Google Scholar]
  6. Kraft, M.L. Sphingolipid Organization in the Plasma Membrane and the Mechanisms that Influence It. Front. cell Dev. Biol. 2016, 4, 154. [Google Scholar] [CrossRef] [PubMed]
  7. Arriaga, L.R.; Rodríguez-García, R.; Moleiro, L.H.; Prévost, S.; López-Montero, I.; Hellweg, T.; Monroy, F. Dissipative Dynamics of Fluid Lipid Membranes Enriched in Cholesterol. Adv. Colloid Interface Sci. 2017, 247, 514–520. [Google Scholar] [CrossRef] [PubMed]
  8. Stieger, B.; Steiger, J.; Locher, K.P. Membrane Lipids and Transporter Function. Biochim. Biophys. Acta-Mol. Basis Dis. 2021, 1867, 166079. [Google Scholar] [CrossRef]
  9. Itri, R.; Junqueira, H.C.; Mertins, O.; Baptista, M.S. Membrane Changes under Oxidative Stress: The Impact of Oxidized Lipids. Biophys. Rev. 2014, 6, 47–61. [Google Scholar] [CrossRef]
  10. Hilgemann, D.W.; Dai, G.; Collins, A.; Lariccia, V.; Magi, S.; Deisl, C.; Fine, M. Lipid Signaling to Membrane Proteins: From Second Messengers to Membrane Domains and Adapter-Free Endocytosis. J. Gen. Physiol. 2018, 150, 211–224. [Google Scholar] [CrossRef]
  11. Podbielska, M.; Ariga, T.; Pokryszko-Dragan, A. Sphingolipid Players in Multiple Sclerosis: Their Influence on the Initiation and Course of the Disease. Int. J. Mol. Sci. 2022, 23, 5330. [Google Scholar] [CrossRef]
  12. Lauwers, E.; Goodchild, R.; Verstreken, P. Membrane Lipids in Presynaptic Function and Disease. Neuron 2016, 90, 11–25. [Google Scholar] [CrossRef]
  13. Martín, M.G.; Dotti, C.G. Plasma Membrane and Brain Dysfunction of the Old: Do We Age from Our Membranes? Front. cell Dev. Biol. 2022, 10, 1031007. [Google Scholar] [CrossRef]
  14. Postila, P.A.; Róg, T. A Perspective: Active Role of Lipids in Neurotransmitter Dynamics. Mol. Neurobiol. 2020, 57, 910–925. [Google Scholar] [CrossRef] [PubMed]
  15. Puchkov, D.; Haucke, V. Greasing the Synaptic Vesicle Cycle by Membrane Lipids. Trends Cell Biol. 2013, 23, 493–503. [Google Scholar] [CrossRef] [PubMed]
  16. Manna, M.; Niemelä, M.; Tynkkynen, J.; Javanainen, M.; Kulig, W.; Müller, D.J.; Rog, T.; Vattulainen, I. Mechanism of Allosteric Regulation of Β2-Adrenergic Receptor by Cholesterol. eLife 2016, 5, e18432. [Google Scholar] [CrossRef] [PubMed]
  17. Zhuo, C.; Hou, W.; Tian, H.; Wang, L.; Li, R. Lipidomics of the Brain, Retina, and Biofluids: From the Biological Landscape to Potential Clinical Application in Schizophrenia. Transl. Psychiatry 2020, 10, 391. [Google Scholar] [CrossRef]
  18. Ji, B.; Skup, M. Roles of Palmitoylation in Structural Long-Term Synaptic Plasticity. Mol. Brain 2021, 14, 8. [Google Scholar] [CrossRef] [PubMed]
  19. Buszka, A.; Pytyś, A.; Colvin, D.; Włodarczyk, J.; Wójtowicz, T. S-Palmitoylation of Synaptic Proteins in Neuronal Plasticity in Normal and Pathological Brains. Cells 2023, 12, 387. [Google Scholar] [CrossRef]
  20. Vaughen, J.P.; Theisen, E.; Clandinin, T.R. From Seconds to Days: Neural Plasticity Viewed through a Lipid Lens. Curr. Opin. Neurobiol. 2023, 80, 102702. [Google Scholar] [CrossRef]
  21. Bernal-Vega, S.; García-Juárez, M.; Camacho-Morales, A. Contribution of Ceramides Metabolism in Psychiatric Disorders. J. Neurochem. 2023, 164, 708–724. [Google Scholar] [CrossRef]
  22. Panov, A.; Orynbayeva, Z.; Vavilin, V.; Lyakhovich, V. Fatty Acids in Energy Metabolism of the Central Nervous System. Biomed Res. Int. 2014, 2014, 472459. [Google Scholar] [CrossRef]
  23. Lajtha, A.; Gibson, G.E.; Dienel, G.A. Handbook of Neurochemistry and Molecular Neurobiology: Brain Energetics. Integration of Molecular and Cellular Processes; Springer: New York, NY, USA, 2007; ISBN 9780387303468. [Google Scholar]
  24. Gorji, A. Neuroinflammation: The Pathogenic Mechanism of Neurological Disorders. Int. J. Mol. Sci. 2022, 23, 5744. [Google Scholar] [CrossRef]
  25. Yang, D.; Wang, X.; Zhang, L.; Fang, Y.; Zheng, Q.; Liu, X.; Yu, W.; Chen, S.; Ying, J.; Hua, F. Lipid Metabolism and Storage in Neuroglia: Role in Brain Development and Neurodegenerative Diseases. Cell Biosci. 2022, 12, 106. [Google Scholar] [CrossRef] [PubMed]
  26. Muzio, L.; Viotti, A.; Martino, G. Microglia in Neuroinflammation and Neurodegeneration: From Understanding to Therapy. Front. Neurosci. 2021, 15, 742065. [Google Scholar] [CrossRef] [PubMed]
  27. Yang, L.; Liu, C.C.; Zheng, H.; Kanekiyo, T.; Atagi, Y.; Jia, L.; Wang, D.; N’songo, A.; Can, D.; Xu, H.; et al. LRP1 Modulates the Microglial Immune Response via Regulation of JNK and NF-ΚB Signaling Pathways. J. Neuroinflammation 2016, 13, 304. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, B.; Wu, L.; Chen, J.; Dong, L.; Chen, C.; Wen, Z.; Hu, J.; Fleming, I.; Wang, D.W. Metabolism Pathways of Arachidonic Acids: Mechanisms and Potential Therapeutic Targets. Signal Transduct. Target. Ther. 2021, 6, 94. [Google Scholar] [CrossRef] [PubMed]
  29. Lee, J.Y.; Jin, H.K.; Bae, J.S. Sphingolipids in Neuroinflammation: A Potential Target for Diagnosis and Therapy. BMB Rep. 2020, 53, 28–34. [Google Scholar] [CrossRef] [PubMed]
  30. Schneider, M.; Levant, B.; Reichel, M.; Gulbins, E.; Kornhuber, J.; Müller, C.P. Lipids in Psychiatric Disorders and Preventive Medicine. Neurosci. Biobehav. Rev. 2017, 76, 336–362. [Google Scholar] [CrossRef] [PubMed]
  31. Li, M.; Gao, Y.; Wang, D.; Hu, X.; Jiang, J.; Qing, Y.; Yang, X.; Cui, G.; Wang, P.; Zhang, J.; et al. Impaired Membrane Lipid Homeostasis in Schizophrenia. Schizophr. Bull. 2022, 48, 1125–1135. [Google Scholar] [CrossRef]
  32. Yao, J.K.; Dougherty, G.G.; Gautier, C.H.; Haas, G.L.; Condray, R.; Kasckow, J.W.; Kisslinger, B.L.; Gurklis, J.A.; Messamore, E. Prevalence and Specificity of the Abnormal Niacin Response: A Potential Endophenotype Marker in Schizophrenia. Schizophr. Bull. 2016, 42, 369–376. [Google Scholar] [CrossRef]
  33. Ong, W.-Y.; Farooqui, T.; Farooqui, A.A. Involvement of Cytosolic Phospholipase A(2), Calcium Independent Phospholipase A(2) and Plasmalogen Selective Phospholipase A(2) in Neurodegenerative and Neuropsychiatric Conditions. Curr. Med. Chem. 2010, 17, 2746–2763. [Google Scholar] [CrossRef]
  34. Wang, F.; Guo, L.; Zhang, T.; Cui, Z.; Wang, J.; Zhang, C.; Xue, F.; Zhou, C.; Li, B.; Tan, Q.; et al. Alterations in Plasma Lipidomic Profiles in Adult Patients with Schizophrenia and Major Depressive Disorder. Medicina 2022, 58, 1509. [Google Scholar] [CrossRef]
  35. Walther, A.; Cannistraci, C.V.; Simons, K.; Durán, C.; Gerl, M.J.; Wehrli, S.; Kirschbaum, C. Lipidomics in Major Depressive Disorder. Front. Psychiatry 2018, 9, 459. [Google Scholar] [CrossRef] [PubMed]
  36. Chan, P.; Suridjan, I.; Mohammad, D.; Herrmann, N.; Mazereeuw, G.; Hillyer, L.M.; Ma, D.W.L.; Oh, P.I.; Lanctôt, K.L. Novel Phospholipid Signature of Depressive Symptoms in Patients with Coronary Artery Disease. J. Am. Heart Assoc. 2018, 7, e008278. [Google Scholar] [CrossRef] [PubMed]
  37. Healy-Stoffel, M.; Levant, B. N-3 (Omega-3) Fatty Acids: Effects on Brain Dopamine Systems and Potential Role in the Etiology and Treatment of Neuropsychiatric Disorders. CNS Neurol. Disord.-Drug Targets 2018, 17, 216–232. [Google Scholar] [CrossRef]
  38. Hussain, G.; Anwar, H.; Rasul, A.; Imran, A.; Qasim, M.; Zafar, S.; Imran, M.; Kamran, S.K.S.; Aziz, N.; Razzaq, A.; et al. Lipids as Biomarkers of Brain Disorders. Crit. Rev. Food Sci. Nutr. 2020, 60, 351–374. [Google Scholar] [CrossRef] [PubMed]
  39. Riddle, E.L.; Rau, K.S.; Topham, M.K.; Hanson, G.R.; Fleckenstein, A.E. Ceramide-Induced Alterations in Dopamine Transporter Function. Eur. J. Pharmacol. 2003, 458, 31–36. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, Y.; Song, X.; Liu, X.; Pu, J.; Gui, S.; Xu, S.; Tian, L.; Zhong, X.; Zhao, L.; Wang, H.; et al. Alteration of Lipids and Amino Acids in Plasma Distinguish Schizophrenia Patients from Controls: A Targeted Metabolomics Study. Psychiatry Clin. Neurosci. 2021, 75, 138–144. [Google Scholar] [CrossRef] [PubMed]
  41. Tkachev, A.; Stekolshchikova, E.; Anikanov, N.; Zozulya, S.; Barkhatova, A.; Klyushnik, T.; Petrova, D. Shorter Chain Triglycerides Are Negatively Associated with Symptom Improvement in Schizophrenia. Biomolecules 2021, 11, 720. [Google Scholar] [CrossRef]
  42. Yan, L.; Zhou, J.; Wang, D.; Si, D.; Liu, Y.; Zhong, L.; Yin, Y. Unbiased Lipidomic Profiling Reveals Metabolomic Changes during the Onset and Antipsychotics Treatment of Schizophrenia Disease. Metabolomics 2018, 14, 80. [Google Scholar] [CrossRef]
  43. Orešič, M.; Seppänen-Laakso, T.; Sun, D.; Tang, J.; Therman, S.; Viehman, R.; Mustonen, U.; van Erp, T.G.; Hyötyläinen, T.; Thompson, P.; et al. Phospholipids and Insulin Resistance in Psychosis: A Lipidomics Study of Twin Pairs Discordant for Schizophrenia. Genome Med. 2012, 4, 1. [Google Scholar] [CrossRef]
  44. Costa, A.C.; Riça, L.B.; van de Bilt, M.; Zandonadi, F.S.; Gattaz, W.F.; Talib, L.L.; Sussulini, A. Application of Lipidomics in Psychiatry: Plasma-Based Potential Biomarkers in Schizophrenia and Bipolar Disorder. Metabolites 2023, 13, 600. [Google Scholar] [CrossRef]
  45. Wang, D.; Sun, X.; Maziade, M.; Mao, W.; Zhang, C.; Wang, J.; Cao, B. Characterising Phospholipids and Free Fatty Acids in Patients with Schizophrenia: A Case-Control Study. World J. Biol. Psychiatry 2021, 22, 161–174. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, D.; Cheng, S.L.; Fei, Q.; Gu, H.; Raftery, D.; Cao, B.; Sun, X.; Yan, J.; Zhang, C.; Wang, J. Metabolic Profiling Identifies Phospholipids as Potential Serum Biomarkers for Schizophrenia. Psychiatry Res. 2019, 272, 18–29. [Google Scholar] [CrossRef] [PubMed]
  47. Frajerman, A.; Chaumette, B.; Farabos, D.; Despres, G.; Simonard, C.; Lamazière, A.; Krebs, M.O.; Kebir, O. Membrane Lipids in Ultra-High-Risk Patients: Potential Predictive Biomarkers of Conversion to Psychosis. Nutrients 2023, 15, 2215. [Google Scholar] [CrossRef] [PubMed]
  48. Li, N.; Yang, P.; Tang, M.; Liu, Y.; Guo, W.; Lang, B.; Wang, J.; Wu, H.; Tang, H.; Yu, Y.; et al. Reduced Erythrocyte Membrane Polyunsaturated Fatty Acid Levels Indicate Diminished Treatment Response in Patients with Multi- versus First-Episode Schizophrenia. Schizophrenia 2022, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  49. Goh, X.X.; Tang, P.Y.; Tee, S.F. Blood-Based Oxidation Markers in Medicated and Unmedicated Schizophrenia Patients: A Meta-Analysis. Asian J. Psychiatr. 2022, 67, 102932. [Google Scholar] [CrossRef] [PubMed]
  50. Song, M.; Liu, Y.; Zhou, J.; Shi, H.; Su, X.; Shao, M.; Yang, Y.; Wang, X.; Zhao, J.; Guo, D.; et al. Potential Plasma Biomarker Panels Identification for the Diagnosis of First-Episode Schizophrenia and Monitoring Antipsychotic Monotherapy with the Use of Metabolomics Analyses. Psychiatry Res. 2023, 321, 115070. [Google Scholar] [CrossRef] [PubMed]
  51. Wei, Y.; Wang, T.; Li, G.; Feng, J.; Deng, L.; Xu, H.; Yin, L.; Ma, J.; Chen, D.; Chen, J. Investigation of Systemic Immune-Inflammation Index, Neutrophil/High-Density Lipoprotein Ratio, Lymphocyte/High-Density Lipoprotein Ratio, and Monocyte/High-Density Lipoprotein Ratio as Indicators of Inflammation in Patients with Schizophrenia and Bipolar Disorder. Front. Psychiatry 2022, 13, 941728. [Google Scholar] [CrossRef]
  52. Nandeesha, H.; Keshri, N.; Rajappa, M.; Menon, V. Association of Hyperglycaemia and Hyperlipidaemia with Cognitive Dysfunction in Schizophrenia Spectrum Disorder. Arch. Physiol. Biochem. 2023, 129, 497–504. [Google Scholar] [CrossRef]
  53. McEvoy, J.; Baillie, R.A.; Zhu, H.; Buckley, P.; Keshavan, M.S.; Nasrallah, H.A.; Dougherty, G.G.; Yao, J.K.; Kaddurah-Daouk, R. Lipidomics Reveals Early Metabolic Changes in Subjects with Schizophrenia: Effects of Atypical Antipsychotics. PLoS ONE 2013, 8, e68717. [Google Scholar] [CrossRef]
  54. Kaddurah-Daouk, R.; McEvoy, J.; Baillie, R.; Zhu, H.K.; Yao, J.; Nimgaonkar, V.L.; Buckley, P.F.; Keshavan, M.S.; Georgiades, A.; Nasrallah, H.A. Impaired Plasmalogens in Patients with Schizophrenia. Psychiatry Res. 2012, 198, 347–352. [Google Scholar] [CrossRef]
  55. Guidara, W.; Messedi, M.; Naifar, M.; Maalej, M.; Khrouf, W.; Grayaa, S.; Maalej, M.; Bonnefont-Rousselot, D.; Lamari, F.; Ayadi, F. Plasma Oxysterols in Drug-Free Patients with Schizophrenia. J. Steroid Biochem. Mol. Biol. 2022, 221, 106123. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, G.; Ye, X.; Wang, X.; Lin, Y.; Zhu, C.; Pan, J.; Yin, X.; Ye, M.; Lv, W.; Tang, W.; et al. Serum Total Cholesterol Levels Associated with Immediate Memory Performance in Patients with Chronic Schizophrenia. Schizophr. Res. 2023, 255, 256–260. [Google Scholar] [CrossRef] [PubMed]
  57. Pillai, R.R.; Wilson, A.B.; Premkumar, N.R.; Kattimani, S.; Sagili, H.; Rajendiran, S. Low Serum Levels of High-Density Lipoprotein Cholesterol (HDL-c) as an Indicator for the Development of Severe Postpartum Depressive Symptoms. PLoS ONE 2018, 13, e0192811. [Google Scholar] [CrossRef]
  58. Liu, X.; Li, J.; Zheng, P.; Zhao, X.; Zhou, C.; Hu, C.; Hou, X.; Wang, H.; Xie, P.; Xu, G. Plasma Lipidomics Reveals Potential Lipid Markers of Major Depressive Disorder. Anal. Bioanal. Chem. 2016, 408, 6497–6507. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, X.; Zheng, P.; Zhao, X.; Zhang, Y.; Hu, C.; Li, J.; Zhao, J.; Zhou, J.; Xie, P.; Xu, G. Discovery and Validation of Plasma Biomarkers for Major Depressive Disorder Classification Based on Liquid Chromatography-Mass Spectrometry. J. Proteome Res. 2015, 14, 2322–2330. [Google Scholar] [CrossRef] [PubMed]
  60. Kim, E.Y.; Lee, J.W.; Lee, M.Y.; Kim, S.H.; Mok, H.J.; Ha, K.; Ahn, Y.M.; Kim, K.P. Serum Lipidomic Analysis for the Discovery of Biomarkers for Major Depressive Disorder in Drug-Free Patients. Psychiatry Res. 2018, 265, 174–182. [Google Scholar] [CrossRef]
  61. Zhang, T.; Guo, L.; Li, R.; Wang, F.; Yang, W.M.; Yang, J.B.; Cui, Z.Q.; Zhou, C.H.; Chen, Y.H.; Yu, H.; et al. Alterations of Plasma Lipids in Adult Women with Major Depressive Disorder and Bipolar Depression. Front. Psychiatry 2022, 13, 927817. [Google Scholar] [CrossRef]
  62. Kuwano, N.; Kato, T.A.; Setoyama, D.; Sato-Kasai, M.; Shimokawa, N.; Hayakawa, K.; Ohgidani, M.; Sagata, N.; Kubo, H.; Kishimoto, J.; et al. Tryptophan-Kynurenine and Lipid Related Metabolites as Blood Biomarkers for First-Episode Drug-Naïve Patients with Major Depressive Disorder: An Exploratory Pilot Case-Control Study. J. Affect. Disord. 2018, 231, 74–82. [Google Scholar] [CrossRef]
  63. Demirkan, A.; Isaacs, A.; Ugocsai, P.; Liebisch, G.; Struchalin, M.; Rudan, I.; Wilson, J.F.; Pramstaller, P.P.; Gyllensten, U.; Campbell, H.; et al. Plasma Phosphatidylcholine and Sphingomyelin Concentrations Are Associated with Depression and Anxiety Symptoms in a Dutch Family-Based Lipidomics Study. J. Psychiatr. Res. 2013, 47, 357–362. [Google Scholar] [CrossRef]
  64. Liu, Z.; Cai, Y.; Zhang, X.; Zhu, Z.; He, J. High Serum Levels of Malondialdehyde and Antioxidant Enzymes Are Associated with Post-Stroke Anxiety. Neurol. Sci. 2018, 39, 999–1007. [Google Scholar] [CrossRef]
  65. Dong, M.X.; Hu, L.; Wei, Y.D.; Chen, G.H. Metabolomics Profiling Reveals Altered Lipid Metabolism and Identifies a Panel of Lipid Metabolites as Biomarkers for Parkinson’s Disease Related Anxiety Disorder. Neurosci. Lett. 2021, 745, 135626. [Google Scholar] [CrossRef] [PubMed]
  66. Kui, H.; Su, H.; Wang, Q.; Liu, C.; Li, Y.; Tian, Y.; Kong, J.; Sun, G.; Huang, J. Serum Metabolomics Study of Anxiety Disorder Patients Based on LC-MS. Clin. Chim. Acta. 2022, 533, 131–143. [Google Scholar] [CrossRef] [PubMed]
  67. De Berardis, D.; Serroni, N.; Campanella, D.; Marini, S.; Rapini, G.; Valchera, A.; Iasevoli, F.; Mazza, M.; Fornaro, M.; Perna, G.; et al. Alexithymia, Suicide Ideation, C-Reactive Protein, and Serum Lipid Levels Among Outpatients with Generalized Anxiety Disorder. Arch. Suicide Res. 2017, 21, 100–112. [Google Scholar] [CrossRef] [PubMed]
  68. Sevincok, L.; Buyukozturk, A.; Dereboy, F. Serum Lipid Concentrations in Patients with Comorbid Generalized Anxiety Disorder and Major Depressive Disorder. Can. J. Psychiatry 2001, 46, 68–71. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, J.; Jiang, C.; Chen, L.; Wu, S.; Lin, J.; Gao, L.; Xie, B.; Zheng, D.; Yang, R.; Li, S.; et al. A Cross-Sectional Study to Investigate the Correlation between Depression Comorbid with Anxiety and Serum Lipid Levels. Compr. Psychiatry 2016, 69, 163–168. [Google Scholar] [CrossRef] [PubMed]
  70. Hu, M.X.; Penninx, B.W.J.H.; de Geus, E.J.C.; Lamers, F.; Kuan, D.C.H.; Wright, A.G.C.; Marsland, A.L.; Muldoon, M.F.; Manuck, S.B.; Gianaros, P.J. Associations of Immunometabolic Risk Factors with Symptoms of Depression and Anxiety: The Role of Cardiac Vagal Activity. Brain. Behav. Immun. 2018, 73, 493–503. [Google Scholar] [CrossRef] [PubMed]
  71. Pistorio, E.; Luca, M.; Luca, A.; Messina, V.; Calandra, C. Autonomic Nervous System and Lipid Metabolism: Findings in Anxious-Depressive Spectrum and Eating Disorders. Lipids Health Dis. 2011, 10, 192. [Google Scholar] [CrossRef]
  72. Thesing, C.S.; Bot, M.; Milaneschi, Y.; Giltay, E.J.; Penninx, B.W.J.H. Omega-3 and Omega-6 Fatty Acid Levels in Depressive and Anxiety Disorders. Psychoneuroendocrinology 2018, 87, 53–62. [Google Scholar] [CrossRef]
  73. Verly-Miguel, M.V.B.; Farias, D.R.; Pinto, T.d.J.P.; Lepsch, J.; Nardi, A.E.; Kac, G. Serum Docosahexaenoic Acid (DHA) Is Inversely Associated with Anxiety Disorders in Early Pregnancy. J. Anxiety Disord. 2015, 30, 34–40. [Google Scholar] [CrossRef]
  74. MahmoudianDehkordi, S.; Bhattacharyya, S.; Brydges, C.R.; Jia, W.; Fiehn, O.; Rush, A.J.; Dunlop, B.W.; Kaddurah-Daouk, R. Gut Microbiome-Linked Metabolites in the Pathobiology of Major Depression with or without Anxiety—A Role for Bile Acids. Front. Neurosci. 2022, 16, 937906. [Google Scholar] [CrossRef]
  75. Xing, Y.; Tang, Y.; Zhao, L.; Wang, Q.; Qin, W.; Ji, X.; Zhang, J.; Jia, J. Associations between Plasma Ceramides and Cognitive and Neuropsychiatric Manifestations in Parkinson’s Disease Dementia. J. Neurol. Sci. 2016, 370, 82–87. [Google Scholar] [CrossRef] [PubMed]
  76. Huang, J.Y.; Arnold, D.; Qiu, C.F.; Miller, R.S.; Williams, M.A.; Enquobahrie, D.A. Association of Serum Vitamin D with Symptoms of Depression and Anxiety in Early Pregnancy. J. Women’s Heal. 2014, 23, 588. [Google Scholar] [CrossRef] [PubMed]
  77. Chen, L.; Zhu, H.; Harshfield, G.A.; Treiber, F.A.; Pollock, J.S.; Pollock, D.; Okereke, O.I.; Su, S.; Dong, Y. Serum 25-Hydroxyvitamin D Concentrations Are Associated with Mental Health and Psychosocial Stress in Young Adults. Nutrients 2020, 12, 1938. [Google Scholar] [CrossRef] [PubMed]
  78. Casseb, G.A.S.; Ambrósio, G.; Rodrigues, A.L.S.; Kaster, M.P. Levels of 25-Hydroxyvitamin D3, Biochemical Parameters and Symptoms of Depression and Anxiety in Healthy Individuals. Metab. Brain Dis. 2019, 34, 527–535. [Google Scholar] [CrossRef] [PubMed]
  79. Tao, S.; Zhang, Y.; Wang, Q.; Qiao, C.; Deng, W.; Liang, S.; Wei, J.; Wei, W.; Yu, H.; Li, X.; et al. Identifying Transdiagnostic Biological Subtypes across Schizophrenia, Bipolar Disorder, and Major Depressive Disorder Based on Lipidomics Profiles. Front. cell Dev. Biol. 2022, 10, 969575. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, X.; Sun, L.; Zhao, A.; Hu, X.; Qing, Y.; Jiang, J.; Yang, C.; Xu, T.; Wang, P.; Liu, J.; et al. Serum Fatty Acid Patterns in Patients with Schizophrenia: A Targeted Metabonomics Study. Transl. Psychiatry 2017, 7, e1176. [Google Scholar] [CrossRef]
  81. Zhou, X.; Long, T.; Haas, G.L.; Cai, H.L.; Yao, J.K. Reduced Levels and Disrupted Biosynthesis Pathways of Plasma Free Fatty Acids in First-Episode Antipsychotic-Naïve Schizophrenia Patients. Front. Neurosci. 2020, 14, 784. [Google Scholar] [CrossRef]
  82. Zhou, X.; Liu, L.; Lan, X.; Cohen, D.; Zhang, Y.; Ravindran, A.V.; Yuan, S.; Zheng, P.; Coghill, D.; Yang, L.; et al. Polyunsaturated Fatty Acids Metabolism, Purine Metabolism and Inosine as Potential Independent Diagnostic Biomarkers for Major Depressive Disorder in Children and Adolescents. Mol. Psychiatry 2019, 24, 1478–1488. [Google Scholar] [CrossRef]
  83. Assies, J.; Pouwer, F.; Lok, A.; Mocking, R.J.T.; Bockting, C.L.H.; Visser, I.; Abeling, N.G.G.M.; Duran, M.; Schene, A.H. Plasma and Erythrocyte Fatty Acid Patterns in Patients with Recurrent Depression: A Matched Case-Control Study. PLoS ONE 2010, 5, e10635. [Google Scholar] [CrossRef]
  84. Scola, G.; Versace, A.; Metherel, A.H.; Monsalve-Castro, L.A.; Phillips, M.L.; Bazinet, R.P.; Andreazza, A.C. Alterations in Peripheral Fatty Acid Composition in Bipolar and Unipolar Depression. J. Affect. Disord. 2018, 233, 86–91. [Google Scholar] [CrossRef]
  85. Pomponi, M.; Janiri, L.; La Torre, G.; Di Stasio, E.; Di Nicola, M.; Mazza, M.; Martinotti, G.; Bria, P.; Lippa, S.; Natili, R.; et al. Plasma Levels of N-3 Fatty Acids in Bipolar Patients: Deficit Restricted to DHA. J. Psychiatr. Res. 2013, 47, 337–342. [Google Scholar] [CrossRef]
  86. Sobczak, S.; Honig, A.; Christophe, M.; Maes, M.; Helsdingen, R.W.C.; De Vriese, S.; Reidel, W.J. Lower High-Density Lipoprotein Cholesterol and Increased Omega-6 Polyunsaturated Fatty Acids in First-Degree Relatives of Bipolar Patients. Psychol. Med. 2004, 34, 103–112. [Google Scholar] [CrossRef]
  87. McNamara, R.K.; Jandacek, R.; Tso, P.; Blom, T.J.; Welge, J.A.; Strawn, J.R.; Adler, C.M.; DelBello, M.P.; Strakowski, S.M. First-Episode Bipolar Disorder Is Associated with Erythrocyte Membrane Docosahexaenoic Acid Deficits: Dissociation from Clinical Response to Lithium or Quetiapine. Psychiatry Res. 2015, 230, 447–453. [Google Scholar] [CrossRef]
  88. Igarashi, M.; Ma, K.; Gao, F.; Kim, H.W.; Greenstein, D.; Rapoport, S.I.; Rao, J.S. Brain Lipid Concentrations in Bipolar Disorder. J. Psychiatr. Res. 2010, 44, 177–182. [Google Scholar] [CrossRef]
  89. Saunders, E.F.H.; Reider, A.; Singh, G.; Gelenberg, A.J.; Rapoport, S.I. Low Unesterified: Esterified Eicosapentaenoic Acid (EPA) Plasma Concentration Ratio Is Associated with Bipolar Disorder Episodes, and Omega-3 Plasma Concentrations Are Altered by Treatment. Bipolar Disord. 2015, 17, 729–742. [Google Scholar] [CrossRef]
  90. Mongan, D.; Healy, C.; Jones, H.J.; Zammit, S.; Cannon, M.; Cotter, D.R. Plasma Polyunsaturated Fatty Acids and Mental Disorders in Adolescence and Early Adulthood: Cross-Sectional and Longitudinal Associations in a General Population Cohort. Transl. Psychiatry 2021, 11, 321. [Google Scholar] [CrossRef]
  91. Li, X.; Yuan, X.; Pang, L.; Zhang, S.; Li, Y.; Huang, X.; Fan, X.; Song, X. The Effect of Serum Lipids and Short-Chain Fatty Acids on Cognitive Functioning in Drug-Naïve, First Episode Schizophrenia Patients. Psychiatry Res. 2022, 313, 114582. [Google Scholar] [CrossRef]
  92. Cao, B.; Wang, D.; Pan, Z.; McIntyre, R.S.; Brietzke, E.; Subramanieapillai, M.; Nozari, Y.; Wang, J. Metabolic Profiling for Water-Soluble Metabolites in Patients with Schizophrenia and Healthy Controls in a Chinese Population: A Case-Control Study. World J. Biol. Psychiatry 2020, 21, 357–367. [Google Scholar] [CrossRef]
  93. Jadoon, A.; Chiu, C.C.; McDermott, L.; Cunningham, P.; Frangou, S.; Chang, C.J.; Sun, I.W.; Liu, S.I.; Lu, M.L.; Su, K.P.; et al. Associations of Polyunsaturated Fatty Acids with Residual Depression or Anxiety in Older People with Major Depression. J. Affect. Disord. 2012, 136, 918–925. [Google Scholar] [CrossRef] [PubMed]
  94. Kriisa, K.; Leppik, L.; Balõtšev, R.; Ottas, A.; Soomets, U.; Koido, K.; Volke, V.; Innos, J.; Haring, L.; Vasar, E.; et al. Profiling of Acylcarnitines in First Episode Psychosis before and after Antipsychotic Treatment. J. Proteome Res. 2017, 16, 3558–3566. [Google Scholar] [CrossRef] [PubMed]
  95. Cao, B.; Wang, D.; Pan, Z.; Brietzke, E.; McIntyre, R.S.; Musial, N.; Mansur, R.B.; Subramanieapillai, M.; Zeng, J.; Huang, N.; et al. Characterizing Acyl-Carnitine Biosignatures for Schizophrenia: A Longitudinal Pre- and Post-Treatment Study. Transl. Psychiatry 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  96. Mamalakis, G.; Jansen, E.; Cremers, H.; Kiriakakis, M.; Tsibinos, G.; Kafatos, A. Depression and Adipose and Serum Cholesteryl Ester Polyunsaturated Fatty Acids in the Survivors of the Seven Countries Study Population of Crete. Eur. J. Clin. Nutr. 2006, 60, 1016–1023. [Google Scholar] [CrossRef] [PubMed]
  97. Beydoun, M.A.; Beydoun, H.A.; Dore, G.A.; Fanelli-Kuczmarski, M.T.; Evans, M.K.; Zonderman, A.B. Total Serum Cholesterol, Atherogenic Indices and Their Longitudinal Association with Depressive Symptoms among US Adults. Transl. Psychiatry 2015, 5, e518. [Google Scholar] [CrossRef] [PubMed]
  98. Ribeiro, H.C.; Klassen, A.; Pedrini, M.; Carvalho, M.S.; Rizzo, L.B.; Noto, M.N.; Zeni-Graiff, M.; Sethi, S.; Fonseca, F.A.H.; Tasic, L.; et al. A Preliminary Study of Bipolar Disorder Type I by Mass Spectrometry-Based Serum Lipidomics. Psychiatry Res. 2017, 258, 268–273. [Google Scholar] [CrossRef] [PubMed]
  99. Huang, Y.J.; Tsai, S.Y.; Chung, K.H.; Chen, P.H.; Huang, S.H.; Kuo, C.J. State-Dependent Alterations of Lipid Profiles in Patients with Bipolar Disorder. Int. J. Psychiatry Med. 2018, 53, 273–281. [Google Scholar] [CrossRef] [PubMed]
  100. Peter, H.; Hand, I.; Hohagen, F.; Koenig, A.; Mindermann, O.; Oeder, F.; Wittich, M. Serum Cholesterol Level Comparison: Control Subjects, Anxiety Disorder Patients, and Obsessive-Compulsive Disorder Patients. Can. J. Psychiatry 2002, 47, 557–561. [Google Scholar] [CrossRef] [PubMed]
  101. De Lacerda, A.L.T.; Caetano, D.; Caetano, S.C. Cholesterol Levels in Panic Disorder, Generalized Anxiety Disorder and Major Depression. Arq. Neuropsiquiatr. 2000, 58, 408–411. [Google Scholar] [CrossRef]
  102. Cenik, B.; Cenik, C.; Snyder, M.P.; Brown, E.S. Plasma Sterols and Depressive Symptom Severity in a Population-Based Cohort. PLoS ONE 2017, 12, e0184382. [Google Scholar] [CrossRef]
  103. Sun, N.; Zhang, J.; Wang, J.; Liu, Z.; Wang, X.; Kang, P.; Yang, C.; Liu, P.; Zhang, K. Abnormal Gut Microbiota and Bile Acids in Patients with First-Episode Major Depressive Disorder and Correlation Analysis. Psychiatry Clin. Neurosci. 2022, 76, 321–328. [Google Scholar] [CrossRef]
  104. Köhnke, C.; Herrmann, M.; Berger, K. Associations of Major Depressive Disorder and Related Clinical Characteristics with 25-Hydroxyvitamin D Levels in Middle-Aged Adults. Nutr. Neurosci. 2022, 25, 1209–1218. [Google Scholar] [CrossRef]
  105. Boerman, R.; Cohen, D.; Schulte, P.F.J.; Nugter, A. Prevalence of Vitamin D Deficiency in Adult Outpatients With Bipolar Disorder or Schizophrenia. J. Clin. Psychopharmacol. 2016, 36, 588–592. [Google Scholar] [CrossRef] [PubMed]
  106. Wood, P.L.; Unfried, G.; Whitehead, W.; Phillipps, A.; Wood, J.A. Dysfunctional Plasmalogen Dynamics in the Plasma and Platelets of Patients with Schizophrenia. Schizophr. Res. 2015, 161, 506–510. [Google Scholar] [CrossRef] [PubMed]
  107. Leppik, L.; Parksepp, M.; Janno, S.; Koido, K.; Haring, L.; Vasar, E.; Zilmer, M. Profiling of Lipidomics before and after Antipsychotic Treatment in First-Episode Psychosis. Eur. Arch. Psychiatry Clin. Neurosci. 2020, 270, 59–70. [Google Scholar] [CrossRef]
  108. He, Y.; Yu, Z.; Giegling, I.; Xie, L.; Hartmann, A.M.; Prehn, C.; Adamski, J.; Kahn, R.; Li, Y.; Illig, T.; et al. Schizophrenia Shows a Unique Metabolomics Signature in Plasma. Transl. Psychiatry 2012, 2, e149. [Google Scholar] [CrossRef] [PubMed]
  109. Stirton, H.; Meek, B.P.; Edel, A.L.; Solati, Z.; Surendran, A.; Aukema, H.; Modirrousta, M.; Ravandi, A. Oxolipidomics Profile in Major Depressive Disorder: Comparing Remitters and Non-Remitters to Repetitive Transcranial Magnetic Stimulation Treatment. PLoS ONE 2021, 16, e0246592. [Google Scholar] [CrossRef] [PubMed]
  110. Costa, A.C.; Joaquim, H.P.G.; Talib, L.L.; Serpa, M.H.; Zanetti, M.V.; Gattaz, W.F. Plasma Metabolites in First Episode Psychoses. Schizophr. Res. 2019, 206, 468–470. [Google Scholar] [CrossRef]
  111. Cai, H.L.; Cao, T.; Li, N.N.; Fang, P.F.; Xu, P.; Wu, X.X.; Zhang, B.K.; Xiang, D.X. Quantitative Monitoring of a Panel of Stress-Induced Biomarkers in Human Plasma by Liquid Chromatography–Tandem Mass Spectrometry: An Application in a Comparative Study between Depressive Patients and Healthy Subjects. Anal. Bioanal. Chem. 2019, 411, 5765–5777. [Google Scholar] [CrossRef]
  112. Tkachev, A.I.; Stekolshchikova, E.A.; Morozova, A.Y.; Anikanov, N.A.; Zorkina, Y.A.; Alekseyeva, P.N.; Khobta, E.B.; Andreyuk, D.S.; Zozulya, S.A.; Barkhatova, A.N.; et al. Ceramides: Shared Lipid Biomarkers of Cardiovascular Disease and Schizophrenia. Consort. Psychiatr. 2021, 2, 35–43. [Google Scholar] [CrossRef]
  113. Gracia-Garcia, P.; Rao, V.; Haughey, N.J.; Banduru, V.V.R.; Smith, G.; Rosenberg, P.B.; Lobo, A.; Lyketsos, C.G.; Mielke, M.M. Elevated Plasma Ceramides in Depression. J. Neuropsychiatry Clin. Neurosci. 2011, 23, 215–218. [Google Scholar] [CrossRef]
  114. Brunkhorst-Kanaan, N.; Klatt-Schreiner, K.; Hackel, J.; Schröter, K.; Trautmann, S.; Hahnefeld, L.; Wicker, S.; Reif, A.; Thomas, D.; Geisslinger, G.; et al. Targeted Lipidomics Reveal Derangement of Ceramides in Major Depression and Bipolar Disorder. Metabolism 2019, 95, 65–76. [Google Scholar] [CrossRef]
  115. Brunkhorst-Kanaan, N.; Trautmann, S.; Schreiber, Y.; Thomas, D.; Kittel-Schneider, S.; Gurke, R.; Geisslinger, G.; Reif, A.; Tegeder, I. Sphingolipid and Endocannabinoid Profiles in Adult Attention Deficit Hyperactivity Disorder. Biomedicines 2021, 9, 1173. [Google Scholar] [CrossRef] [PubMed]
  116. Solberg, D.K.; Bentsen, H.; Refsum, H.; Andreassen, O.A. Lipid Profiles in Schizophrenia Associated with Clinical Traits: A Five Year Follow-up Study. BMC Psychiatry 2016, 16, 299. [Google Scholar] [CrossRef] [PubMed]
  117. Maes, M.; Mihaylova, I.; Kubera, M.; Uytterhoeven, M.; Vrydags, N.; Bosmans, E. Lower Plasma Coenzyme Q10 in Depression: A Marker for Treatment Resistance and Chronic Fatigue in Depression and a Risk Factor to Cardiovascular Disorder in That Illness. Neuro-Endocrinol. Lett. 2009, 30, 462–469. [Google Scholar] [PubMed]
  118. Hurşitoğlu, O.; Orhan, F.Ö.; Kurutaş, E.B.; Doğaner, A.; Durmuş, H.T.; Kopar, H. Diagnostic Performance of Increased Malondialdehyde Level and Oxidative Stress in Patients with Schizophrenia. Noro Psikiyatr. Ars. 2021, 58, 184–188. [Google Scholar] [CrossRef] [PubMed]
  119. Günes, M.; Camkurt, M.A.; Bulut, M.; Demir, S.; Ibiloglu, A.O.; Kaya, M.C.; Atli, A.; Kaplan, I.; Sir, A. Evaluation of Paraoxonase, Arylesterase and Malondialdehyde Levels in Schizophrenia Patients Taking Typical, Atypical and Combined Antipsychotic Treatment. Clin. Psychopharmacol. Neurosci. 2016, 14, 345–350. [Google Scholar] [CrossRef]
  120. Uddin, S.M.N.; Sultana, F.; Uddin, M.G.; Dewan, S.M.R.; Hossain, M.K.; Islam, M.S. Effect of Antioxidant, Malondialdehyde, Macro-Mineral, and Trace Element Serum Concentrations in Bangladeshi Patients with Schizophrenia: A Case-Control Study. Health Sci. Rep. 2021, 4, e291. [Google Scholar] [CrossRef]
  121. Islam, M.R.; Islam, M.R.; Ahmed, I.; Moktadir, A.A.; Nahar, Z.; Islam, M.S.; Shahid, S.F.B.; Islam, S.N.; Islam, M.S.; Hasnat, A. Elevated Serum Levels of Malondialdehyde and Cortisol Are Associated with Major Depressive Disorder: A Case-Control Study. SAGE Open Med. 2018, 6, 2050312118773953. [Google Scholar] [CrossRef]
  122. Alvarez-Mon, M.A.; Ortega, M.A.; García-Montero, C.; Fraile-Martinez, O.; Lahera, G.; Monserrat, J.; Gomez-Lahoz, A.M.; Molero, P.; Gutierrez-Rojas, L.; Rodriguez-Jimenez, R.; et al. Differential Malondialdehyde (MDA) Detection in Plasma Samples of Patients with Major Depressive Disorder (MDD): A Potential Biomarker. J. Int. Med. Res. 2022, 50, 3000605221094995. [Google Scholar] [CrossRef]
  123. Aydemir, Ö.; Çubukçuoğlu, Z.; Erdin, S.; Taş, C.; Onur, E.; Berk, M. Oxidative Stress Markers, Cognitive Functions, and Psychosocial Functioning in Bipolar Disorder: An Empirical Cross-Sectional Study. Rev. Bras. Psiquiatr. 2014, 36, 293–297. [Google Scholar] [CrossRef]
  124. Islam, M.R.; Ahmed, M.U.; Islam, M.S.; Sayeed, M.S.B.; Sadia, F.; Chowdhury, Z.S.; Nahar, Z.; Hasnat, A. Comparative Analysis of Serum Malondialdehyde, Antioxidant Vitamins and Immunoglobulin Levels in Patients Suffering from Generalized Anxiety Disorder. Drug Res. 2014, 64, 406–411. [Google Scholar] [CrossRef]
  125. Uyar, B.; Budak, E.A. Effects of Major Depressive Disorder on Monocytes, High-Density Lipoprotein (HDL) and Monocyte to HDL Ratio: A Case-Control Study. Psychiatr. Danub. 2022, 34, 687–694. [Google Scholar] [CrossRef] [PubMed]
  126. Lieberman, H.R.; Kellogg, M.D.; Kramer, F.M.; Bathalon, G.P.; Lesher, L.L. Lipid and Other Plasma Markers Are Associated with Anxiety, Depression, and Fatigue. Health Psychol. 2012, 31, 210–216. [Google Scholar] [CrossRef] [PubMed]
  127. Veen, G.; Giltay, E.J.; DeRijk, R.H.; van Vliet, I.M.; van Pelt, J.; Zitman, F.G. Salivary Cortisol, Serum Lipids, and Adiposity in Patients with Depressive and Anxiety Disorders. Metabolism 2009, 58, 821–827. [Google Scholar] [CrossRef] [PubMed]
  128. Huang, T.L.; Wu, S.C.; Chiang, Y.S.; Chen, J.F. Correlation between Serum Lipid, Lipoprotein Concentrations and Anxious State, Depressive State or Major Depressive Disorder. Psychiatry Res. 2003, 118, 147–153. [Google Scholar] [CrossRef] [PubMed]
  129. Skrede, S.; Steen, V.M.; Fernø, J. Antipsychotic-Induced Increase in Lipid Biosynthesis: Activation through Inhibition? J. Lipid Res. 2013, 54, 307–309. [Google Scholar] [CrossRef] [PubMed]
  130. Kaddurah-Daouk, R.; McEvoy, J.; Baillie, R.A.; Lee, D.; Yao, J.K.; Doraiswamy, P.M.; Krishnan, K.R.R. Metabolomic Mapping of Atypical Antipsychotic Effects in Schizophrenia. Mol. Psychiatry 2007, 12, 934–945. [Google Scholar] [CrossRef]
  131. Pillinger, T.; McCutcheon, R.A.; Vano, L.; Mizuno, Y.; Arumuham, A.; Hindley, G.; Beck, K.; Natesan, S.; Efthimiou, O.; Cipriani, A.; et al. Comparative Effects of 18 Antipsychotics on Metabolic Function in Patients with Schizophrenia, Predictors of Metabolic Dysregulation, and Association with Psychopathology: A Systematic Review and Network Meta-Analysis. Lancet Psychiatry 2020, 7, 64–77. [Google Scholar] [CrossRef]
  132. Huang, J.; Xiao, J.; Peng, Z.; Shao, P.; Sun, M.; Long, Y.; Wang, X.; Shen, M.; Kang, D.; Yang, Y.; et al. PCSK9 Mediates Dyslipidemia Induced by Olanzapine Treatment in Schizophrenia Patients. Psychopharmacology 2022, 239, 83–91. [Google Scholar] [CrossRef]
  133. Chaggar, P.S.; Shaw, S.M.; Williams, S.G. Effect of Antipsychotic Medications on Glucose and Lipid Levels. J. Clin. Pharmacol. 2011, 51, 631–638. [Google Scholar] [CrossRef]
  134. Canfrań-Duque, A.; Casado, M.E.; Pastor, Ó.; Sánchez-Wandelmer, J.; De La Penã, G.; Lerma, M.; Mariscal, P.; Bracher, F.; Lasunción, M.A.; Busto, R. Atypical Antipsychotics Alter Cholesterol and Fatty Acid Metabolism in Vitro. J. Lipid Res. 2013, 54, 310–324. [Google Scholar] [CrossRef]
  135. Sánchez-Wandelmer, J.; Dávalos, A.; de la Peña, G.; Cano, S.; Giera, M.; Canfrán-Duque, A.; Bracher, F.; Martín-Hidalgo, A.; Fernández-Hernando, C.; Lasunción, M.A.; et al. Haloperidol Disrupts Lipid Rafts and Impairs Insulin Signaling in SH-SY5Y Cells. Neuroscience 2010, 167, 143–153. [Google Scholar] [CrossRef] [PubMed]
  136. Canfrán-Duque, A.; Pastor, Ó.; García-Seisdedos, D.; Molina, Y.L.; Babiy, B.; Lerma, M.; Sánchez-Castellano, C.; Martínez-Botas, J.; Gómez-Coronado, D.; Lasunción, M.A.; et al. The Antipsychotic Risperidone Alters Dihydroceramide and Ceramide Composition and Plasma Membrane Function in Leukocytes In Vitro and In Vivo. Int. J. Mol. Sci. 2021, 22, 3919. [Google Scholar] [CrossRef] [PubMed]
  137. Cai, H.L.; Tan, Q.Y.; Jiang, P.; Dang, R.L.; Xue, Y.; Tang, M.M.; Xu, P.; Deng, Y.; Li, H.D.; Yao, J.K. A Potential Mechanism Underlying Atypical Antipsychotics-Induced Lipid Disturbances. Transl. Psychiatry 2015, 5, e661. [Google Scholar] [CrossRef] [PubMed]
  138. Alves, I.; Staneva, G.; Tessier, C.; Salgado, G.F.; Nuss, P. The Interaction of Antipsychotic Drugs with Lipids and Subsequent Lipid Reorganization Investigated Using Biophysical Methods. Biochim. Biophys. Acta 2011, 1808, 2009–2018. [Google Scholar] [CrossRef]
  139. Smesny, S.; Langbein, K.; Rzanny, R.; Gussew, A.; Burmeister, H.P.; Reichenbach, J.R.; Sauer, H. Antipsychotic Drug Effects on Left Prefrontal Phospholipid Metabolism: A Follow-up 31P-2D-CSI Study of Haloperidol and Risperidone in Acutely Ill Chronic Schizophrenia Patients. Schizophr. Res. 2012, 138, 164–170. [Google Scholar] [CrossRef] [PubMed]
  140. Dietrich-Muszalska, A.; Kolińska-Łukaszuk, J. Comparative Effects of Aripiprazole and Selected Antipsychotic Drugs on Lipid Peroxidation in Plasma. Psychiatry Clin. Neurosci. 2018, 72, 329–336. [Google Scholar] [CrossRef] [PubMed]
  141. de Almeida, V.; Alexandrino, G.L.; Aquino, A.; Gomes, A.F.; Murgu, M.; Dobrowolny, H.; Guest, P.C.; Steiner, J.; Martins-de-Souza, D. Changes in the Blood Plasma Lipidome Associated with Effective or Poor Response to Atypical Antipsychotic Treatments in Schizophrenia Patients. Prog. Neuropsychopharmacol. Biol. Psychiatry 2020, 101, 109945. [Google Scholar] [CrossRef]
  142. Gabriel, A. Changes in Plasma Cholesterol in Mood Disorder Patients: Does Treatment Make a Difference? J. Affect. Disord. 2007, 99, 273–278. [Google Scholar] [CrossRef]
  143. Pan, S.J.; Tan, Y.L.; Yao, S.W.; Xin, Y.; Yang, X.; Liu, J.; Xiong, J. Fluoxetine Induces Lipid Metabolism Abnormalities by Acting on the Liver in Patients and Mice with Depression. Acta Pharmacol. Sin. 2018, 39, 1463–1472. [Google Scholar] [CrossRef]
  144. McIntyre, R.S.; Soczynka, J.K.; Konarski, J.Z.; Kennedy, S.H. The Effect of Antidepressants on Lipid Homeostasis: A Cardiac Safety Concern? Expert Opin. Drug Saf. 2006, 5, 523–537. [Google Scholar] [CrossRef]
  145. Nicholas, L.M.; Ford, A.L.; Esposito, S.M.; Ekstrom, R.D.; Golden, R.N. The Effects of Mirtazapine on Plasma Lipid Profiles in Healthy Subjects. J. Clin. Psychiatry 2003, 64, 883–889. [Google Scholar] [CrossRef] [PubMed]
  146. Kopf, D.; Westphal, S.; Luley, C.W.; Ritter, S.; Gilles, M.; Weber-Hamann, B.; Lederbogen, F.; Lehnert, H.; Henn, F.A.; Heuser, I.; et al. Lipid Metabolism and Insulin Resistance in Depressed Patients: Significance of Weight, Hypercortisolism, and Antidepressant Treatment. J. Clin. Psychopharmacol. 2004, 24, 527–531. [Google Scholar] [CrossRef]
  147. Yosmaoǧlu, A.; Fistikci, N.; Keyvan, A.; Hacioǧlu, M.; Erten, E.; Saatçioǧlu, Ö.; Kora, K. Correlation of Selective Serotonin Re-Uptake Inhibitor Use with Weight Gain and Metabolic Parameters. Anatol. J. Psychiatry 2013, 14, 245–251. [Google Scholar] [CrossRef]
  148. Beyazyüz, M.; Albayrak, Y.; Eğilmez, O.B.; Albayrak, N.; Beyazyüz, E. Relationship between SSRIs and Metabolic Syndrome Abnormalities in Patients with Generalized Anxiety Disorder: A Prospective Study. Psychiatry Investig. 2013, 10, 148–154. [Google Scholar] [CrossRef] [PubMed]
  149. Olguner Eker, Ö.; Özsoy, S.; Eker, B.; Doğan, H. Metabolic Effects of Antidepressant Treatment. Noro Psikiyatr. Ars. 2017, 54, 49–56. [Google Scholar] [CrossRef] [PubMed]
  150. Deisenhammer, E.A.; Kramer-Reinstadler, K.; Liensberger, D.; Kemmler, G.; Hinterhuber, H.; Fleischhacker, W.W. No Evidence for an Association between Serum Cholesterol and the Course of Depression and Suicidality. Psychiatry Res. 2004, 121, 253–261. [Google Scholar] [CrossRef]
  151. Xiong, J.; Yang, H.; Wu, L.; Shang, W.; Shan, E.; Liu, W.; Hu, G.; Xi, T.; Yang, J. Fluoxetine Suppresses AMP-Activated Protein Kinase Signaling Pathway to Promote Hepatic Lipid Accumulation in Primary Mouse Hepatocytes. Int. J. Biochem. Cell Biol. 2014, 54, 236–244. [Google Scholar] [CrossRef]
  152. Hsiao, C.C. Difference in Pre- and Post-Treatment Plasma DHEA Levels Were Significantly and Positively Correlated with Difference in Pre- and Post-Treatment Hamilton Depression Scores Following Successful Therapy for Major Depression. Psychoneuroendocrinology 2006, 31, 839–846. [Google Scholar] [CrossRef]
  153. Mocking, R.J.T.; Nap, T.S.; Westerink, A.M.; Assies, J.; Vaz, F.M.; Koeter, M.W.J.; Ruhé, H.G.; Schene, A.H. Biological Profiling of Prospective Antidepressant Response in Major Depressive Disorder: Associations with (Neuro)Inflammation, Fatty Acid Metabolism, and Amygdala-Reactivity. Psychoneuroendocrinology 2017, 79, 84–92. [Google Scholar] [CrossRef]
  154. Hummel, J.; Westphal, S.; Weber-Hamann, B.; Gilles, M.; Lederbogen, F.; Angermeier, T.; Luley, C.; Deuschle, M.; Kopf, D. Serum Lipoproteins Improve after Successful Pharmacologic Antidepressant Treatment: A Randomized Open-Label Prospective Trial. J. Clin. Psychiatry 2011, 72, 885–891. [Google Scholar] [CrossRef]
  155. Sonawalla, S.B.; Papakostas, G.I.; Petersen, T.J.; Yeung, A.S.; Smith, M.M.; Sickinger, A.H.; Gordon, J.; Israel, J.A.; Tedlow, J.R.; Lamon-Fava, S.; et al. Elevated Cholesterol Levels Associated with Nonresponse to Fluoxetine Treatment in Major Depressive Disorder. Psychosomatics 2002, 43, 310–316. [Google Scholar] [CrossRef]
  156. Noordam, R.; Aarts, N.; De Keyser, C.E.; Hofman, A.; Stricker, B.H.; Visser, L.E. Antidepressants with a High Serotonin Reuptake Transporter Affinity and Serum Lipid Levels in a Population-Based Study in Older Adults. J. Psychopharmacol. 2015, 29, 1112–1118. [Google Scholar] [CrossRef]
  157. Homorogan, C.; Nitusca, D.; Enatescu, V.; Schubart, P.; Moraru, C.; Socaciu, C.; Marian, C. Untargeted Plasma Metabolomic Profiling in Patients with Major Depressive Disorder Using Ultra-High Performance Liquid Chromatography Coupled with Mass Spectrometry. Metabolites 2021, 11, 466. [Google Scholar] [CrossRef]
  158. Pinto, B.; Conde, T.; Domingues, I.; Domingues, M.R. Adaptation of Lipid Profiling in Depression Disease and Treatment: A Critical Review. Int. J. Mol. Sci. 2022, 23, 2032. [Google Scholar] [CrossRef]
  159. MahmoudianDehkordi, S.; Ahmed, A.T.; Bhattacharyya, S.; Han, X.; Baillie, R.A.; Arnold, M.; Skime, M.K.; John-Williams, L.S.; Moseley, M.A.; Thompson, J.W.; et al. Alterations in Acylcarnitines, Amines, and Lipids Inform about the Mechanism of Action of Citalopram/Escitalopram in Major Depression. Transl. Psychiatry 2021, 11, 153. [Google Scholar] [CrossRef]
Figure 1. The main lipid groups and the examples of their representatives. The illustrations were prepared using the following program: https://molview.org/ accessed on 18 December 2023).
Figure 1. The main lipid groups and the examples of their representatives. The illustrations were prepared using the following program: https://molview.org/ accessed on 18 December 2023).
Metabolites 14 00080 g001aMetabolites 14 00080 g001b
Figure 2. Lipids and their functions in the nervous system.
Figure 2. Lipids and their functions in the nervous system.
Metabolites 14 00080 g002
Table 1. Studies assessing blood lipid alterations in psychiatric disorders at the level of individual lipid species. ↑ increased and ↓ decreased.
Table 1. Studies assessing blood lipid alterations in psychiatric disorders at the level of individual lipid species. ↑ increased and ↓ decreased.
FATTY ACYLS
Type of LipidDisorder
SCZMDDBPDAD
PUFAs↑ 8 types [45],
↑ monounsaturated fatty acids and ω-6 PUFAs [80], and
↓ 5 types [40] ↓ 6 types [81]
↓ including eicosapentaenoic acid and arachidonic acid [82],
↑ in plasma,
mostly ↓ membrane fatty acids, exp octadectetraenoic, gamma-linolenic acid, and docosadienoic acid [83].
↑ AA: eicosapentaenoic acid (EPA) ratio [84].
↑ omega-6 PUFA, AA: EPA and AA: EPA+ docosahexaenoic acid (DHA) [84].
↑ linoleic acid, AA, α-linolenic acid, EPA [85],
↑ ratio of omega-6/omega-3 [86]
↓ DHA 22:6n-3 decreased in membrane [87], and
↓ DHA (22:6n-3) and AA (20:4n-6) [88]
↓ EPA acid [89].
↓ DHA [85].
↑ n-6:n-3 ratio [90]
↓ N-3 PUFA and ↓ N3:FA ratio in patients with comorbid depressive and anxiety disorder [72].
↓ DHA in pregnant women [73].
↓ 3 types of PUFAs in patients with comorbid Parkinson’s disease and AD [65].
SFA↑ [91],
↑ 4 types [45],
↑ linoleic acid [47],
↓ stearic acid, behenic acid, α-dimorphecolic [50],
↓ lithocholic acid [40], and
↓ 16:0 [92].
Higher levels of total SCFAs, acetic acid, acetic acid/ propionic acid ratio SCZ compared to CTL. The lipid levels were positively associated with acetic acid/propionic acid ratio
↑ azelaic acid,
↓ palmitic acid, dodecanoic acid, and capric acid [82].
↑ plasma,
mostly ↓ membrane exp. palmitic acid [83].
No information.↓ hexacosanoic acid and 10-oxo-nonadecanoic acid in patients with comorbid Parkinson’s disease and AD [65].
↑ 3-Hydroxysebacic acid and
↓ 2-Hydroxy-3-methylpentanoic acid [66].
↑ erythrocyte membrane linoleic acid in patients with anxiety symptoms comorbid to MDD [93].
Carnitine↑ long chain in FEP [94],
↑ CAR 10:2 [44],
↑ 3 types of carnitine [95],
↓ [34],
↓ 3 types of carnitine [40],
↓ medium and high chain [95],
↓ short-chain CAR in FEP [94], and
↓ 18:2 in FEP [42].
↓ 5 types [59].
↓ AcCAR [61].
↓ [61].↓ propionylcarnitine [66].
Transdiagnostic between SCZ, MDD, BPD, and AD:
↓ 10-nitro-9Z,12Z-octadecadienoic acid decreased;
↑ 9,12-octadecadienal, cyclopentaneoctanoic acid, hexadecandioic acid, 12-tridecynoic acid, and caprylic acid [79].
SACCHAROLIPIDS
Type of lipidDisorder
SCZMDDBPDAD
MGDGNo information.↓ [61].↓ [61].No information.
STEROL LIPIDS
Type of lipidDisorder
SCZMDDBPDAD
Cholesteryl esters↑ in FEP [42].
↓ CE 16:1 [44].
Plasma tryptophan-kynurenine metabolites and CEs were significantly correlated in the MDD group, but not in the HC group [62].
No difference [96].
↓ CE [60].
↓ [86]No information.
Total cholesterol↓ [52].↑ in women with depressive symptoms [97].
↓ in postpartum depression [57].
↑ [98].
↓ [51,99].
↑ [100]; ↑ in comorbid MDD and GAD [68].
↓ in alexythimic patents [67],
↓ in patients with comorbid Parkinson’s disease and AD [65],
↓ in the anxious–depressive disorder group [71], and
not changed compared to MDD and BPD [101].
Sterols↑ the ratios of cholestane-3β,5α,6β-triol, 27-hydroxycholesterol, and cholestanol to tchol [55].
↓ several types of sterol lipids [44].
↑ 7-dihydrocholesterol, ↓ desmosterol, and 14-desmethyl lanosterol in people with depressive symptoms [102].No information↓ 11-acetoxy-3β,6α-dihydroxy-9,11-seco-5α-cholest-7-en-9-one in patients with comorbid Parkinson’s disease and AD [67].
Bile acids↓ [50].
↓ lithocholic acid [40].
↑ 23-nordeoxycholic acid,
↓ taurolithocholic acid (TLCA), glycolithocholic acid (GLCA), and lithocholic acid (LCA) 3-sulfate ↓ [103].
↓ chenodeoxycholic acid (CDCA) [74].
No information.↓ CDCA in highly anxious participants compared to participants with less severe symptoms [74].
↑ LCA [74].
Calcifediol↓ [50].↓ [104];
not changed depressive symptoms in healthy people [78].
↓ [105].Not changed [78].
↓ 20-oxo-22,23,24,25,26,27-hexanorvitamin D3 [79] transdiagnostic↓ [79] transdiagnostic.
↓ anxiety symptoms [76,77].
GLYCEROPHOSPHOLIPIDS
Type of lipidDisorder
SCZMDDBPDAD
PC↑ 32 types of PC [40],
↓ PC [50], ↓ 46:7 [44],
↓ PC (O-34:2) [40],
mostly ↓ membrane PC [31],
↓ PC [45,54,106], PC-O in FEP [107],
↓ PC-P in FEP [42],
↓ PC-O [45,46],
↓ PC-O 38:6 [108],
↓ PUFA-containing PC [53], and
↓ 14 types and ↑ 11 types [34].
↑ [58],
↑ PC 32:1 [59],
↑ PC-O [63]
↑ PC(8:0e/6:0) [34] ↓ 3 types ↑ 5 types [82]
↑ [109]
and ↓ PC-O 36:2 [59]
↑ PC [110].↓ PC O 36:4 (anxiety symptoms) [63] and
↓ LysoPC(0:0/16:0) [66].
↑ PC-O 16:0-18:1+2O [79] transdiagnostic
PE↑ PE 34:2 [40],
↓ [50],
↓ 40:7 [44],
↓ PE (O-34:3), (O-36:6) [40],
mostly ↓ membrane PE [31],
↓ PE-P [54,106],
↓ PE-P in FEP [42],
↓ PE-O [46],
↓ PUFA-containing PE [53], and
↓ 2 ↑ 5 [45].
↑ [58],
↑ PE 34:2 36:4 [59],
↓ PE-O [58], ↓ PE-O [59],
↓ PE (16:0/22:6) PE(18:0/22:6) [111],
↑ PE(18:1/0:0), and PE(18:2/0:0) [82].
↓ [61].No information.
LPC↑ LPC [40],
↑ LPC 18:1 [44],
↓ [34,43,46]
↓ in FEP [42],
mostly ↓ membrane LPC [31]
↑ 4 ↓ 19 [45]
↑ [58,59]
↑ LysoPC (16:0) and LysoPC (18:0) [111], and
↑ LPC [106],
↑ LPC [106].No information.
Lysophosphatidylethanolamine (LPE)↑ [34,40]
mostly ↓ membrane LPE [31]
3 ↓ 9 ↑ [45]
↑ [58,59]↑ [61]↑ lysoPE(18:2(9Z,12Z)/0:0) [66].
PS↑ LPS 21:0 [44],
↓ 43:2 [44],
↓ 10, ↑ 13 membrane PS [31],
↓ [61]↓ [61]No information.
↑ DGTS 16:0/18:1 [79] transdiagnostic
SPHINGOLIPIDS
Type of lipidDisorder
SCZMDDBPDAD
Sphingomyelin↑ SM with PUFA (C18:1 and C24:1), ↓ 12 types [44]
mostly ↓ SMs with SFA (C16:0, C20:0, and C24:0) [45], and
mostly ↓ membrane SM [31]
↑ [63],
↓ PhSM [61], and
↓SM 39:1 [58].
↓ SM and phSM [61].↓ SPM 23:1 (anxiety symptoms) [63].
Ceramide↑ Cer (d18: 1/16: 0), Cer (d18: 1/18: 0) и Cer (d18: 1/24: 1) [112],
↓ 44:1 [44]
↓ 22, ↑ 20 membrane Cer [31]
↑ Cer elevated [113],
↑ Cer and HexCer [114]
↓ CerG2GNAc1(d38:4) [34],
↓ CerG2GNAc1 [61]
↑ Cer and HexCer elevated [114] and
↑ Cer22:0 [115]
↑ Cer 20:0 in Parkinson’s disease patients with anxiety symptoms [75].
GangliosideNo information↑ monosialotetrahexosylganglioside 2 (GM2) [61]↑ GM2 [61].No information.
Other↓ C16 sphinganine [50];
↓ glycosphingolipids [44].
↑ total sphingolipids [68].↓ N-(hexadecanoyl)-deoxysphing-4-enine-1-sulfonate in patients with comorbid Parkinson’s disease and AD [65].
GLYCEROLIPIDS
Type of lipidDisorders
SCZMDDBPDAD
TG↑ 20 types [41],
↑ [43,52,116]
↑ in FEP [42],
↑ membrane TG [31], and
↓ 3 types [44].
↑ [58,59] and ↓ TG [60].
↑ [34].
↑ [51,61,114].
↓ TG [99].
↑ in alexythimic patents [67],
↑ comorbid MDD with anxiety [68,69,71],
↑ anxiety symptoms [70], and
↓ in patients with comorbid Parkinson’s disease and AD [65].
DG↑ membrane [31].Not changed [60].↑ [114].↓ 6 types in patients with comorbid Parkinson’s disease and AD [65]
PRODUCTS OF LIPID METABOLISM
Type of lipidDisorder
SCZMDDBPDAD
Coenzyme Q10No information.↓ [117].↓ [61].No information.
Malondialdehyde↑ [49,118,119,120]↑ [121,122].↑ [123].↑ [124];
↑ post-stroke anxiety [64].
Lipoproteins↓ HDL [52];
↓ HDL, LDL, and ApoE [51].
↑ LDL [34],
↓ HDL [125],
↓ HDL-c postpartum [57], and
↓ HDL-C [62].
HDL ↓, LDL, and apolipoprotein E (ApoE) ↓ [51].↑ LDL [67,68],
↓ LDL in patients with comorbid Parkinson’s disease and AD [65], ↓ LDL (tension-anxiety symptoms) [126],
↓ HDL [67,68,69,127,128],
↑ HDL (tension–anxiety symptoms) [126],
↑ VLDL [67], ↑ LDL/HDL [67], ↑ TC/HDL [67,128],
↑ ApoB [71], ↓ ApoB in patients with comorbid Parkinson’s disease and AD [65], and
↑ ApoA [71].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zorkina, Y.; Ushakova, V.; Ochneva, A.; Tsurina, A.; Abramova, O.; Savenkova, V.; Goncharova, A.; Alekseenko, I.; Morozova, I.; Riabinina, D.; et al. Lipids in Psychiatric Disorders: Functional and Potential Diagnostic Role as Blood Biomarkers. Metabolites 2024, 14, 80. https://doi.org/10.3390/metabo14020080

AMA Style

Zorkina Y, Ushakova V, Ochneva A, Tsurina A, Abramova O, Savenkova V, Goncharova A, Alekseenko I, Morozova I, Riabinina D, et al. Lipids in Psychiatric Disorders: Functional and Potential Diagnostic Role as Blood Biomarkers. Metabolites. 2024; 14(2):80. https://doi.org/10.3390/metabo14020080

Chicago/Turabian Style

Zorkina, Yana, Valeria Ushakova, Aleksandra Ochneva, Anna Tsurina, Olga Abramova, Valeria Savenkova, Anna Goncharova, Irina Alekseenko, Irina Morozova, Daria Riabinina, and et al. 2024. "Lipids in Psychiatric Disorders: Functional and Potential Diagnostic Role as Blood Biomarkers" Metabolites 14, no. 2: 80. https://doi.org/10.3390/metabo14020080

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

Article Metrics

Back to TopTop