Next Article in Journal
The Impact of Varying Pasture Levels on the Metabolomic Profile of Bovine Ruminal Fluid
Previous Article in Journal
An Overview of Pre-Analytical Factors Impacting Metabolomics Analyses of Blood Samples
Previous Article in Special Issue
Metabolic Connectome and Its Role in the Prediction, Diagnosis, and Treatment of Complex Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Peripheral Lipid Signatures, Metabolic Dysfunction, and Pathophysiology in Schizophrenia Spectrum Disorders

1
Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON M6J 1H3, Canada
2
Institute of Medical Sciences, University of Toronto, Toronto, ON M5T 1R8, Canada
3
Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
4
Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4,Canada
5
Clinical Pharmacy Department, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
6
Department of Pharmacy, Michigan Medicine Health System, Ann Arbor, MI 48109, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors share senior co-authorship.
Metabolites 2024, 14(9), 475; https://doi.org/10.3390/metabo14090475
Submission received: 21 July 2024 / Revised: 19 August 2024 / Accepted: 21 August 2024 / Published: 28 August 2024
(This article belongs to the Special Issue Metabolomics in Disease Mechanisms and Drug Targets)

Abstract

:
Metabolic dysfunction is commonly observed in schizophrenia spectrum disorders (SSDs). The causes of metabolic comorbidity in SSDs are complex and include intrinsic or biological factors linked to the disorder, which are compounded by antipsychotic (AP) medications. The exact mechanisms underlying SSD pathophysiology and AP-induced metabolic dysfunction are unknown, but dysregulated lipid metabolism may play a role. Lipidomics, which detects lipid metabolites in a biological sample, represents an analytical tool to examine lipid metabolism. This systematic review aims to determine peripheral lipid signatures that are dysregulated among individuals with SSDs (1) with minimal exposure to APs and (2) during AP treatment. To accomplish this goal, we searched MEDLINE, Embase, and PsychINFO databases in February 2024 to identify all full-text articles written in English where the authors conducted lipidomics in SSDs. Lipid signatures reported to significantly differ in SSDs compared to controls or in relation to AP treatment and the direction of dysregulation were extracted as outcomes. We identified 46 studies that met our inclusion criteria. Most of the lipid metabolites that significantly differed in minimally AP-treated patients vs. controls comprised glycerophospholipids, which were mostly downregulated. In the AP-treated group vs. controls, the significantly different metabolites were primarily fatty acyls, which were dysregulated in conflicting directions between studies. In the pre-to-post AP-treated patients, the most impacted metabolites were glycerophospholipids and fatty acyls, which were found to be primarily upregulated and conflicting, respectively. These lipid metabolites may contribute to SSD pathophysiology and metabolic dysfunction through various mechanisms, including the modulation of inflammation, cellular membrane permeability, and metabolic signaling pathways.

1. Introduction

Schizophrenia spectrum disorders (SSDs) represent a series of chronic mental illnesses affecting approximately 1% of the world’s population [1]. Beyond functional impairment caused by the illness itself, patients with SSDs are at increased risk of suffering from obesity, dyslipidemia, and type 2 diabetes [2]. These metabolic comorbidities contribute to a 2–3-fold increased risk of developing cardiovascular disease and a decreased lifespan of 15–20 years in SSDs compared to the general population [3,4,5].
Metabolic comorbidities in SSDs are understood to be intrinsic to the disease itself and further worsened over time by antipsychotic (AP) medications and lifestyle factors. For example, in AP-naive patients with SSDs, in whom the confounding effects of APs and illness duration are minimal, disturbances in glucose homeostasis and lipid profiles are already observed [5,6,7]. However, over time, lifestyle and social factors including inactivity, dietary habits, and reduced access to medical care contribute to the progressive worsening of metabolic comorbidity throughout the illness [8,9]. Finally, APs are associated with well-established significant metabolic side effects including lipid abnormalities, glucose dysregulation, and weight gain [5,8,10,11,12].
While the exact mechanisms underlying the pathophysiology of SSDs, intrinsic metabolic dysfunction, and AP-induced metabolic dysfunction are unknown, the role of lipid metabolism has been implicated in multiple studies across different stages of illness. In particular, states of dyslipidemia, adiposity, and insulin resistance, which are observed in both AP-naive and AP-treated patients with SSDs, have been associated with altered lipid metabolism [13]. In terms of potential mechanisms, phospholipids are an important component of neuronal membranes. As such, abnormalities in brain phospholipid metabolism can alter neuronal and hence brain morphology, and this has been suggested to underly the pathophysiology of SSDs [14].
Lipidomics is a high-throughput approach that allows for the comprehensive characterization of lipid metabolism. This is achieved through analyzing lipid metabolites in biological samples. Currently, mass spectrometry (MS)-based techniques are the most powerful tools available for detecting, identifying, and quantifying lipid metabolites [15]. Furthermore, MS-based techniques can also be divided into targeted and untargeted approaches, with targeted lipidomics aiming to quantify specific lipid species and untargeted lipidomics examining all detectable lipid metabolites [16]. When applied to studying disease states, lipidomics can lead to the identification of dysregulated lipid metabolites, also known as lipidomic signatures. These lipidomic signatures can facilitate the discovery of biomarkers and underlying pathophysiological mechanisms [17,18].
Over the past decade, a growing number of studies have applied lipidomic tools in SSDs. Lipidomic signatures identified in studies may provide clues to candidate biomarkers and underlying associations of pathophysiology and metabolic dysfunction within SSDs. In particular, the application of lipidomics to minimally AP-treated patients can help to elucidate metabolic and pathophysiological dysregulations that are present without the confounding effects of AP treatment. Therefore, through a systematic search, this review aims to (1) identify peripheral lipidomic signatures observed in minimally AP-treated patients and (2) investigate the impact of APs on lipidomic signatures by comparing AP-treated patients to healthy controls (HCs) or by comparing pre-to-post AP treatment in patients.

2. Materials and Methods

This study was conducted in accordance with the preferred reporting items for systematic reviews and meta-analyses (PRISMA) methodology and reporting standard. A protocol for this review was submitted to the PROSPERO international database of prospectively registered systematic reviews (PROSPERO #CRD42022298057).

2.1. Search Strategy

Studies were identified through a systematic search. MEDLINE, Embase, and PsycINFO databases were searched for relevant peer-reviewed studies published prior to February 2024. The following search string was used: (lipidom* OR metabolom* OR metabonom*) AND (schizophrenia OR schizophreniform OR schizoaffective OR psychosis OR antipsychotic*). The search string details are provided in Supplementary Table S1. The search was limited to studies written in English and conducted on humans.

2.2. Study Selection

Study selection and de-duplication took place in Covidence. The resulting articles were first screened based on title and abstract, followed by full-text screening by six reviewers (S.W., J.L., K.J.P., D.L., E.C.C.S., and K.M.). Agreement from a minimum of two authors was required for a study to be included or excluded. Disagreements were resolved between all authors by reviewing source papers. Studies were selected based on the following criteria: (1) Study Design: Any clinical trials and observational studies including cohort, case–control, and cross-sectional studies were included. We excluded systematic reviews, meta-analyses, case reports, case series, conference proceedings and abstracts, ideas, editorials, opinions, and animal research studies. While systematic reviews and meta-analyses were excluded from the final review, we mined their reference lists to identify additional references that met the eligibility criteria. (2) Study Population: Populations of interest included individuals diagnosed with SSDs according to a recognized diagnostic criterion (e.g., International Classification of Diseases, ICD; Diagnostic and Statistical Manual of Mental Disorders, DSM). We included both AP-treated and minimally AP-treated patients, where minimally AP-treated was defined as having no prior exposure to APs (AP-naive) or not using APs for more than 3 weeks in the past 3 months (AP-free). We excluded a study if most participants presented with comorbidities or concomitant medications deemed to have a clinically significant impact on metabolic homeostasis such as metformin or antidiabetic drugs [19]. We also excluded post-mortem studies. (3) Comparator: For review question 1, we included studies that identified lipidomic signatures differentiating minimally AP-treated patients from HCs. For review question 2, we included studies that investigated the impact of APs on lipidomic signatures by comparing AP-treated patients with SSDs to HCs or by comparing pre-to-post AP treatment in patients. For both questions, HCs were defined as not having a history of psychiatric illness, no previous AP treatment, and no other medical conditions or use of medications deemed to impact lipid signatures. (4) Outcomes: Included studies were required to assess lipid signatures peripherally. Lipid signatures were defined as lipids measured using lipidomic approaches that were reported to be significantly dysregulated. Lipid signatures that differed from the comparator group across at least two studies for each objective were included in this review. In addition, only studies that reported on the direction of change (up- or downregulated) for each metabolite were included.

2.3. Data Extraction

All data were independently extracted and reviewed by five authors (J.L., S.W., K.J.P., D.L., and E.C.C.S.). Lipid metabolites that significantly differed in minimally AP-treated patients and AP-treated patients compared to controls were extracted separately. For each significant metabolite, the direction of change was noted. If an equal number of studies reported a metabolite as being upregulated vs. downregulated, then the overall direction was defined as “conflicting”. For studies that examined a combination of minimally AP-treated, AP-treated, and pre-to-post-treated patients, the outcomes were extracted separately for each group if the data were reported in this manner. Otherwise, if the authors identified lipidomic signatures for both minimally AP-treated and AP-treated patients combined, then the study was classified based on the AP condition group that represented 50% or more of the included participants. One minimally AP-treated study examined male and female participants separately, with no lipidomic investigation across both sexes [20]. As such, for this study, the lipidomic signatures were extracted separately for males and females. In addition to lipidomic signatures, other pertinent information, including author(s), publication year, study population, study design, number of cases, number of controls, sex ratio, age, AP treatment status of patients, types of APs used, analytical tool used, Human Metabolome Database (HMDB) IDs [21], and peripheral tissue analyzed, were also collected.

2.4. Risk of Bias Assessment

The Johanna Briggs Institute (JBI) Critical Appraisal Checklist for Case Control Studies and the JBI Critical Appraisal Checklist for Cohort Studies were used to assess bias in the context of our outcomes of interest (i.e., peripheral lipid signatures) [22]. Risk of bias assessments were conducted by four independent reviewers (S.W., D.L., K.J.P., E.C.C.S.), with two individuals assigned per study, and conflicts were resolved through group discussion and consensus.

3. Results

The search identified 1405 records across three databases (Figure 1). Of these, 560 duplicates were removed, resulting in 845 studies that went through title and abstract screening. After title and abstract screening, 560 studies were excluded based on the inclusion and exclusion criteria. Subsequently, 285 studies were assessed for eligibility based on the full text, leading to 240 additional studies being excluded. Thus, a total of 46 studies were included in this review (one study was found during review mining).
Of the 46 included studies, 17 studies investigated lipidomic signatures between minimally AP-treated patients with SSDs and HCs [20,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38], 28 studies investigated lipidomic signatures between AP-treated patients and HCs [33,36,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64], and 13 studies looked at patients pre- vs. post-treatment [20,26,27,29,31,33,34,59,60,62,65,66,67]. Two studies were included in both the minimally AP-treated and AP-treated comparisons as they had sub-groups investigating each aim [33,36]. Additionally, seven studies in the minimally AP-treated group [20,26,27,29,31,33,34] and four studies in the AP-treated group [33,59,60,62] were included in the pre-to-post-treated comparison as there were separate sub-groups for this aim. The most common techniques for lipidomic analysis were liquid chromatography–MS and gas chromatography–MS. A full list of the types of techniques and the participants’ demographic data can be found in Table 1, Table 2 and Table 3. None of the studies included in this review were deemed to be of low quality or have a high risk of bias in domains such as the selection of participants, measurement of outcomes, selection of reported results, and bias due to missing data. Supplemental Tables S2–S4 provide additional characteristics about the included studies such as nicotine use and concomitant medications.
The lipid classes that comprised the altered metabolite signatures in minimally AP-treated patients vs. HCs, AP-treated patients vs. HCs, and pre-to-post AP treatment were primarily glycerophospholipids and fatty acyls. For minimally AP-treated patients, it was observed that the most common class was glycerophospholipids, and they were primarily downregulated, while the most common direction across all the lipid metabolites in this comparison was conflicting (i.e., equal number of upregulated and downregulated). For the lipid signatures in AP-treated patients, the most common metabolite class was fatty acyls, and they had approximately equal numbers in the conflicting and upregulated directions. Interestingly, the downregulated metabolites for this comparison mostly comprised glycerophospholipids. For the pre-to-post AP treatment comparison, there were similar numbers of dysregulated fatty acyls and glycerophospholipids, with fatty acyls predominately being in a conflicting direction and glycerophospholipids being mostly upregulated. Table 4, Table 5 and Table 6 detail the metabolites that appeared at least twice across different studies per comparison and additional study information such as whether a correction for multiple comparisons was applied, significant metabolic indices (i.e., weight, body mass index (BMI), and clinical measures of glucose and cholesterol parameters), and/or symptom scale differences between the comparison groups, if reported.

4. Discussion

This review explored alterations in lipid metabolites associated with SSDs and AP use to describe potential associations that may underlie SSD pathophysiology and metabolic dysfunction intrinsic and extrinsic to SSDs. We focused on three comparator groups: minimally AP-treated patients with SSDs vs. HCs, AP-treated patients vs. HCs, and pre-to-post AP treatment.
Glycerophospholipids and fatty acyls were the metabolite classes that were most frequently impacted across the three comparison groups, suggesting a potential role in the pathophysiology of and metabolic dysfunction within SSDs. Glycerophospholipids represent a class of metabolites that are primarily found in the cellular membrane and are involved in membrane stability and fluidity [68]. Ensuring the stability and fluidity of the cellular membrane is integral for normal cell functioning as the cellular membrane is involved in cellular signaling and provides protection for intracellular components [69]. Glycerophospholipids may play a role in glucose metabolism as low membrane fluidity has been linked to impaired insulin signaling [70]. Additionally, glycerophospholipids are linked to brain structure as oral supplementation can increase hippocampal cell morphology [71]. Glycerophospholipids can also contribute to dendritic development, and abnormalities within dendrites have been associated with neuropsychiatric disorders [72,73].
Fatty acyls have a multitude of functions such as providing energy to cells, mediating signal transduction pathways, and acting as a structural unit in cell membranes [74]. Fatty acids, the largest sub-type of fatty acyls, are important components for metabolism as they provide energy to tissues through β-oxidation. Previous research has shown that morphological brain changes, such as improving white matter microstructure, increasing grey matter volume, and improvements in learning and memory, can occur with polyunsaturated fatty acid (PUFA) supplementation [75,76]. Additionally, fatty acyls can potentially be associated with insulin resistance through inflammation. For example, saturated fatty acids are viewed as pro-inflammatory molecules and inflammation is linked with insulin resistance [77]. Unsaturated fatty acids may play an opposing role in insulin action as they are viewed as anti-inflammatory molecules and may potentially have protective effects [77]. Figure 2 summarizes the potential associations between glycerophospholipids, fatty acyls, and SSDs.

4.1. Aim 1: Lipidomic Signatures in Minimally AP-Treated Patients Compared to Healthy Controls

In minimally AP-treated patients with SSDs, it was observed that glycerophospholipids were the most common class impacted, and they were primarily downregulated. This downregulation is consistent with potential mechanisms involved in SSD pathophysiology such as increased phospholipase A2 (PLA2) activity and dendritic spine alterations [78,79]. PLA2s are a family of enzymes that hydrolyze glycerophospholipids, and increased activity of PLA2s is associated with an increased breakdown of the cell membrane [78]. Therefore, lower levels of glycerophospholipids found in minimally AP-treated patients may help explain illness pathophysiology as the breakdown of the cell membrane can impact cell stability and signaling. Dendritic spine alterations have been found in SSDs including reductions in density and arborization [79]. Given that glycerophospholipids are involved in dendritic growth, these alterations may be linked to decreased levels of glycerophospholipids [80]. Additionally, the reduction in dendritic spine density and arborization may help explain structural brain changes reported and/or cognitive deficits observed in SSDs [81,82,83]. As all the studies included in this comparison involved patients that were minimally exposed to APs, the observed downregulation of glycerophospholipids may represent a biomarker intrinsic to SSDs.
Past research has shown that glycerophospholipids may also be associated with metabolic dysfunction as reduced levels have been observed in individuals with impaired fasting glucose and type 2 diabetes [84]. However, this association was not observed in our review as the majority of included studies did not find significant differences in metabolic parameters between patients and HCs. A potential reason for this observation is that most studies only used BMI as a metabolic parameter and the authors may have matched patients and HCs based on this value to reduce potential group differences. As such, critical parameters like fasting glucose and insulin were not collected in most of the studies, making it difficult to identify any differences based on these parameters. The results also showed that the most common direction of change for the lipid metabolites was conflicting, which could potentially reflect the inherent heterogeneity of SSDs.

4.2. Aim 2: Investigate the Impact of APs on Lipidomic Signatures by Comparing AP-Treated Patients with SSDs to HCs or by Comparing Pre-to-Post AP Treatment in Patients

In AP-treated patients vs. HCs, a trend within the results was that half of the downregulated metabolites were from the glycerophospholipid class. Interestingly, glycerophospholipids are a major component of mitochondria, and a decrease in levels can lead to an increase in mitochondrial permeability, which has been linked to metabolic disorders [85]. Taken together, the possibility may exist that the observed downregulation in glycerophospholipids could be related to the metabolic dysfunction associated with APs [86,87]. It was also observed that fatty acyls were primarily in the conflicting direction in AP-treated patients. One fatty acyl, nervonic acid, was increased across two studies, and previous research suggests that this change may be related to symptom control in relation to AP treatment. For example, one study found that decreased levels of nervonic acid were associated with the development of psychosis in ultra-high-risk individuals [88] while another found that increases in polyunsaturated fatty acids were associated with symptom improvement. As nervonic acid is a monounsaturated (rather than polyunsaturated) fatty acid, it is possible that the observed association may not hold; however, given the convergence of our results with these findings, future research is warranted. Furthermore, it is difficult to parse out the cumulative effects of lifestyle habits (diet and physical activity), intrinsic and extrinsic metabolic risk, and the effects of the illness itself, which can all have potential effects on lipid metabolites. Differing dietary habits between patients and HCs have been noted. For example, patients have been shown to consume higher total dietary fat and increased caloric intake [89,90]. Research has also shown that the lipidome can be impacted by dietary fat, which may further explain some of the conflicting metabolites. The AP exposure of the patients also varied widely in dose, duration, and type, which may also help to explain the variation in the results [11]. Furthermore, in the studies that found downregulated glycerophospholipids, only a couple of studies [50,61] observed significantly different metabolic parameters; however, similar to the minimally AP-treated aim, most studies only measured BMI.
For the pre-to-post AP-treated patients, fatty acyls and glycerophospholipids were the classes most impacted, with fatty acyls primarily being in a conflicting direction and glycerophospholipids being upregulated. Potential reasons for fatty acyls having a conflicting direction may be due to differing follow-up lengths (2–78 weeks), the specific AP agent, and possibly the AP dose. Additionally, not all studies included AP-naive patients at baseline, and authors often failed to report on the washout periods from any pre-existing AP treatments. Three studies found significant differences in linoleic acid [34,60,62], a PUFA, two of which found that it was upregulated after AP treatment and that it correlated with significant associated improvements in overall and positive symptom scores [60,62]. The latter finding is consistent with previous research demonstrating an association between PUFA deficiency and symptom severity [76,91]. Lysophosphatidylcholines (LPCs), a sub-type of glycerophospholipids, were found to be upregulated across studies post-AP treatment. This is also supported by previous research that identified negative associations between LPCs and symptom severity in SSDs [66].
A few of the studies included in the pre-to-post comparison examined lipid signatures in relation to metabolic outcomes. For example, across two studies examining AP-naive patients at baseline, it was reported that cholesterol ester (22:6) and phosphocholine (38:6) were reduced after AP treatment [31,65], and this occurred in association with weight gain [65]. Additionally, LPCs (14:0), (18:0), (18:1), and (20:3) were found to be upregulated post-AP treatment [27,29,34,59,65,66]. In keeping with these observations, past research has demonstrated that increased levels of these metabolites are associated with weight gain, with LPCs (14:0) representing an independent contributor after a regression analysis [92]. As such, these longitudinal studies provide additional support for the idea that lipid signatures could represent biomarkers linked to AP-induced weight gain [93]. However, once again, differences in AP type, duration, and dose as well as the heterogeneity of SSDs may have had potentially confounding effects on the observed results.

4.3. Limitations and Recommendations for Future Studies

There was a paucity of longitudinal studies examining lipid signatures pre-to-post AP treatment, which may have contributed to the conflicting lipidomic signatures observed across studies. Similarly, for many of the differing lipid signatures identified, there were disagreements among studies across the three population groups regarding the direction of dysregulation. The inconsistencies in the direction of dysregulation may also be indicative of the complex mechanisms by which lipid metabolism may influence psychopathology and metabolic dysfunction. As discussed above, factors such as sex, diet, duration of illness, smoking or other substance use, activity levels, and AP type, duration, and dose may impact lipid signatures, and many studies did not account for these [9,94]. For example, very few studies matched their cases and controls based on these factors, which may have confounded the results. Another limitation is that cultural differences may impact the results. Approximately half of the studies were conducted in Western societies while the other half were conducted in Eastern societies. The respective individualistic vs. collectivistic culture may impact the results. For example, greater social support in some societies can be a protective factor and diet can also vary between cultures [95]. In addition, previous research has shown that APs can vary in their metabolic risk, with potentially differential effects on the lipidome [11]. Since the included studies used a variety of different APs, it was challenging to discern how lipid metabolites may be impacted by APs with high vs. low metabolic risk profiles. As such, future studies should control for the differing risk profiles of these medications. Furthermore, the studies included in our review employed a range of lipidomic tools to assess peripheral lipid signatures in our population of interest. As each technique has different advantages and disadvantages, this may influence the selectivity, sensitivity, and accuracy of the observed results. Moreover, most of the included studies used targeted metabolomic techniques, which may limit the number of identifiable lipids. Future studies should also limit the heterogeneity of reportable outcomes by standardizing methods such as fasting blood sample collection and corrections for multiple comparisons.

5. Conclusions

In the present review, we demonstrate that certain lipidomic signatures may represent biomarkers related to SSDs, as assessed using minimally AP-treated patients. Specifically, we found that the majority of lipid metabolites were from glycerophospholipids, and they were mostly downregulated. Additionally, we attempted to elucidate the effects of APs on lipid metabolites by comparing AP-treated patients to HCs and pre-to-post AP-treated patients. For AP-treated patients, it was found that the most affected class was fatty acyls, and they were mainly in the conflicting direction. For the pre-to-post AP treatment comparison, there were similar numbers of dysregulated fatty acyls and glycerophospholipids, with fatty acyls mostly being in a conflicting direction and glycerophospholipids being predominantly upregulated. These signatures in turn may be associated with SSD pathophysiology as well as intrinsic and AP-induced metabolic dysfunction through various mechanisms, including the modulation of inflammation, cellular membrane permeability, and metabolic signaling pathways. As such, identifying these lipidomic signatures may aid in the development of better diagnostic tools, possibly leading to novel treatment regimens; however, there is a need for further, well-designed prospective longitudinal studies that assess lipidomic changes in relation to metabolic alterations, AP use, psychopathology, and treatment outcomes.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/metabo14090475/s1, Table S1: Search String. Table S2: Additional characteristics of cross-sectional components of studies examining minimally antipsychotic-treated patients; Table S3: Additional characteristics of cross-sectional components of studies examining antipsychotic-treated patients; Table S4: Additional characteristics of included studies examining pre-to-post antipsychotic-treated patients.

Author Contributions

All authors contributed to drafting and revising the manuscript and approved its final version. J.L. and M.H. were involved in the concept and design of the study. S.W., K.J.P., J.L., D.L., E.C.C.S. and K.M. were involved in screening the articles. S.W., K.J.P., D.L., E.C.C.S. and B.H. were involved in data extraction. S.W., K.J.P., D.L. and E.C.C.S. were involved in risk of bias assessments. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. S.W. is supported by the Banting and Best Diabetes Centre Novo-Nordisk Graduate Studentship and the Ontario Graduate Scholarship. K.J.P. is supported by the Margaret and Howard Gamble Research Grant and the Dalton Whitebread Scholarship Fund. E.E.C.S. is supported by the Peterborough K.M. Hunter Charitable Foundation Graduate Award and the IMS Open Fellowship. K.M. is supported by the Peterborough K.M. Hunter Charitable Foundation Graduate Award, the Hilda and William Courtney Clayton Paediatric Research Fund and the Ontario Graduate Scholarship. T.A. is supported by the Canadian Graduate Masters Scholarship and the Banting and Best Diabetes Centre Novo-Nordisk Graduate Studentship. S.M.A. is supported in part by an Academic Scholars Award from the Department of Psychiatry, University of Toronto (U of T) and the Centre for Addiction and Mental Health (CAMH) Discovery Fund. M.H. is supported by the Banting and Best Diabetes Centre (BBDC) and Drucker Funds, U of T, the Canadian Institutes of Health Research (CIHR) (PJT-153262), a research grant from the Investigator-Initiated Studies Program of Merck Canada Inc. (MISP), and holds the CAMH and U of T Kelly and Michael Meighen Chair in Psychosis Prevention.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

S.M.A. has received honoraria from HLS Therapeutics and has served on the advisory board for Boehringer Ingelheim, Canada. Kristen Ward has served on the advisory board for BioXcel Therapeutics. M.H. has received consultant and speaker fees from Alkermes and consultant fees from Merck.

References

  1. Tandon, R.; Nasrallah, H.A.; Keshavan, M.S. Schizophrenia, “Just the Facts” 4. Clinical Features and Conceptualization. Schizophr. Res. 2009, 110, 1–23. [Google Scholar] [CrossRef]
  2. De Hert, M.; Schreurs, V.; Vancampfort, D.; Van Winkel, R. Metabolic Syndrome in People with Schizophrenia: A Review. World Psychiatry 2009, 8, 15. [Google Scholar] [CrossRef] [PubMed]
  3. Hennekens, C.H.; Hennekens, A.R.; Hollar, D.; Casey, D.E. Schizophrenia and Increased Risks of Cardiovascular Disease. Am. Heart J. 2005, 150, 1115–1121. [Google Scholar] [CrossRef] [PubMed]
  4. Rajkumar, A.P.; Horsdal, H.T.; Wimberley, T.; Cohen, D.; Mors, O.; Børglum, A.D.; Gasse, C. Endogenous and Antipsychotic-Related Risks for Diabetes Mellitus in Young People With Schizophrenia: A Danish Population-Based Cohort Study. Am. J. Psychiatry 2017, 174, 686–694. [Google Scholar] [CrossRef]
  5. Correll, C.U.; Robinson, D.G.; Schooler, N.R.; Brunette, M.F.; Mueser, K.T.; Rosenheck, R.A.; Marcy, P.; Addington, J.; Estroff, S.E.; Robinson, J.; et al. Cardiometabolic Risk in Patients with First-Episode Schizophrenia Spectrum Disorders: Baseline Results from the RAISE-ETP Study. JAMA Psychiatry 2014, 71, 1350–1363. [Google Scholar] [CrossRef]
  6. Misiak, B.; Stańczykiewicz, B.; Łaczmański, Ł.; Frydecka, D. Lipid Profile Disturbances in Antipsychotic-Naive Patients with First-Episode Non-Affective Psychosis: A Systematic Review and Meta-Analysis. Schizophr. Res. 2017, 190, 18–27. [Google Scholar] [CrossRef] [PubMed]
  7. Kucukgoncu, S.; Kosir, U.; Zhou, E.; Sullivan, E.; Srihari, V.H.; Tek, C. Glucose Metabolism Dysregulation at the Onset of Mental Illness Is Not Limited to First Episode Psychosis: A Systematic Review and Meta-Analysis. Early Interv. Psychiatry 2019, 13, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
  8. Mitchell, A.J.; Vancampfort, D.; Sweers, K.; Van Winkel, R.; Yu, W.; De Hert, M. Prevalence of Metabolic Syndrome and Metabolic Abnormalities in Schizophrenia and Related Disorders—A Systematic Review and Meta-Analysis. Schizophr. Bull. 2013, 39, 306. [Google Scholar] [CrossRef]
  9. Heald, A.; Pendlebury, J.; Anderson, S.; Narayan, V.; Guy, M.; Gibson, M.; Haddad, P.; Livingston, M. Lifestyle Factors and the Metabolic Syndrome in Schizophrenia: A Cross-Sectional Study. Ann. Gen. Psychiatry 2017, 16, 12. [Google Scholar] [CrossRef]
  10. Rummel-Kluge, C.; Komossa, K.; Schwarz, S.; Hunger, H.; Schmid, F.; Lobos, C.A.; Kissling, W.; Davis, J.M.; Leucht, S. Head-to-Head Comparisons of Metabolic Side Effects of Second Generation Antipsychotics in the Treatment of Schizophrenia: A Systematic Review and Meta-Analysis. Schizophr. Res. 2010, 123, 225–233. [Google Scholar] [CrossRef]
  11. 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]
  12. Newcomer, J.W. Second-Generation (Atypical) Antipsychotics and Metabolic Effects: A Comprehensive Literature Review. CNS Drugs 2005, 19 (Suppl. 1), 1–93. [Google Scholar] [CrossRef] [PubMed]
  13. Monnerie, S.; Comte, B.; Ziegler, D.; Morais, J.A.; Pujos-Guillot, E.; Gaudreau, P. Metabolomic and Lipidomic Signatures of Metabolic Syndrome and Its Physiological Components in Adults: A Systematic Review. Sci. Rep. 2020, 10, 669. [Google Scholar] [CrossRef] [PubMed]
  14. Horrobin, D.F. The Membrane Phospholipid Hypothesis as a Biochemical Basis for the Neurodevelopmental Concept of Schizophrenia. Schizophr. Res. 1998, 30, 193–208. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, Z.; Bagarolo, G.I.; Thoröe-Boveleth, S.; Jankowski, J. “Lipidomics”: Mass Spectrometric and Chemometric Analyses of Lipids. Adv. Drug Deliv. Rev. 2020, 159, 294–307. [Google Scholar] [CrossRef] [PubMed]
  16. Cajka, T.; Fiehn, O. Toward Merging Untargeted and Targeted Methods in Mass Spectrometry-Based Metabolomics and Lipidomics. Anal. Chem. 2016, 88, 524–545. [Google Scholar] [CrossRef]
  17. Sethi, S.; Hayashi, M.A.F.; Barbosa, B.S.; Pontes, J.G.M.; Tasic, L.; Brietzke, E. Lipidomics, Biomarkers, and Schizophrenia: A Current Perspective. Adv. Exp. Med. Biol. 2017, 965, 265–290. [Google Scholar] [CrossRef]
  18. 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]
  19. Udhane, S.S.; Legeza, B.; Marti, N.; Hertig, D.; DIserens, G.; Nuoffer, J.M.; Vermathen, P.; Flück, C.E. Combined Transcriptome and Metabolome Analyses of Metformin Effects Reveal Novel Links between Metabolic Networks in Steroidogenic Systems. Sci. Rep. 2017, 7, 8652. [Google Scholar] [CrossRef]
  20. Bicikova, M.; Hill, M.; Ripova, D.; Mohr, P.; Hampl, R. Determination of Steroid Metabolome as a Possible Tool for Laboratory Diagnosis of Schizophrenia. J. Steroid Biochem. Mol. Biol. 2013, 133, 77–83. [Google Scholar] [CrossRef]
  21. Wishart, D.S.; Guo, A.C.; Oler, E.; Wang, F.; Anjum, A.; Peters, H.; Dizon, R.; Sayeeda, Z.; Tian, S.; Lee, B.L.; et al. HMDB 5.0: The Human Metabolome Database for 2022. Nucleic Acids Res. 2022, 50, D622–D631. [Google Scholar] [CrossRef]
  22. Moola, S.; Munn, Z.; Tufanaru, C.; Aromataris, E.; Sears, K.; Sfetcu, R.; Currie, M.; Qureshi, R.; Mattis, P.; Lisy, K.; et al. Chapter 7: Systematic Reviews of Etiology and Risk. Available online: https://synthesismanual.jbi.global (accessed on 14 April 2023).
  23. Cai, H.L.; Li, H.D.; Yan, X.Z.; Sun, B.; Zhang, Q.; Yan, M.; Zhang, W.Y.; Jiang, P.; Zhu, R.H.; Liu, Y.P.; et al. Metabolomic Analysis of Biochemical Changes in the Plasma and Urine of First-Episode Neuroleptic-Naïve Schizophrenia Patients after Treatment with Risperidone. J. Proteome Res. 2012, 11, 4338–4350. [Google Scholar] [CrossRef] [PubMed]
  24. Cui, G.; Qing, Y.; Li, M.; Sun, L.; Zhang, J.; Feng, L.; Li, J.; Chen, T.; Wang, J.; Wan, C. Salivary Metabolomics Reveals That Metabolic Alterations Precede the Onset of Schizophrenia. J. Proteome Res. 2021, 20, 5010–5023. [Google Scholar] [CrossRef]
  25. Kaddurah-Daouk, R.; Mcevoy, J.; Baillie, R.; Zhu, H.; Yao, J.K.; 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]
  26. Kriisa, K.; Leppik, L.; Balõtšev, R.; Ottas, A.; Soomets, U.; Koido, K.; Volke, V.; Rgen 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]
  27. 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] [PubMed]
  28. 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]
  29. Qiao, Y.; Zhang, L.; He, S.; Wen, H.; Yu, Y.-M.; Cao, C.-H.; Li, H.-F. Plasma Metabonomics Study of First-Episode Schizophrenia Treated with Olanzapine in Female Patients. Neurosci. Lett. 2016, 617, 270–276. [Google Scholar] [CrossRef]
  30. Schwarz, E.; Whitfield, P.; Nahnsen, S.; Wang, L.; Major, H.; Leweke, F.M.; Koethe, D.; Lio, P.; Bahn, S. Alterations of Primary Fatty Acid Amides in Serum of Patients with Severe Mental Illness. Front. Biosci. (Elite Ed) 2011, 3, 308–314. [Google Scholar] [CrossRef]
  31. Yan, L.; Zhou, J.; Wang, D.; Si, D.; Liu, Y.; Zhong, L.; Yin, Y.; Yan, L. Unbiased Lipidomic Profiling Reveals Metabolomic Changes during the Onset and Antipsychotics Treatment of Schizophrenia Disease. Metabolomics 2018, 14, 80. [Google Scholar] [CrossRef]
  32. Lee, J.; Costa-Dookhan, K.; Panganiban, K.; MacKenzie, N.; Treen, Q.C.; Chintoh, A.; Remington, G.; Müller, D.J.; Sockalingam, S.; Gerretsen, P.; et al. Metabolomic Signatures Associated with Weight Gain and Psychosis Spectrum Diagnoses: A Pilot Study. Front. Psychiatry 2023, 14, 1169787. [Google Scholar] [CrossRef] [PubMed]
  33. Shang, P.; Man-Choi Ho, A.; Tufvesson-Alm, M.; Lindberg, D.R.; Grant, C.W.; Orhan, F.; Eren, F.; Bhat, M.; Engberg, G.; Schwieler, L.; et al. Identification of Cerebrospinal Fluid and Serum Metabolomic Biomarkers in First Episode Psychosis Patients. Transl. Psychiatry 2022, 12, 229. [Google Scholar] [CrossRef]
  34. 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]
  35. Wang, Z.; Yuan, X.; Zhu, Z.; Pang, L.; Ding, S.; Li, X.; Kang, Y.; Hei, G.; Zhang, L.; Zhang, X.; et al. Multiomics Analyses Reveal Microbiome-Gut-Brain Crosstalk Centered on Aberrant Gamma-Aminobutyric Acid and Tryptophan Metabolism in Drug-Naïve Patients with First-Episode Schizophrenia. Schizophr. Bull. 2024, 50, 187–198. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, X.; Xie, J.; Ma, H.; Li, G.; Li, M.; Li, S.; Sun, X.; Zhao, Y.; Sun, W.; Yang, S.; et al. The Relationship between Alterations in Plasma Metabolites and Treatment Responses in Antipsychotic-Naïve Female Patients with Schizophrenia. World J. Biol. Psychiatry 2024, 25, 106–115. [Google Scholar] [CrossRef] [PubMed]
  37. Su, Q.; Bi, F.; Yang, S.; Yan, H.; Sun, X.; Wang, J.; Qiu, Y.; Li, M.; Li, S.; Li, J. Identification of Plasma Biomarkers in Drug-Naïve Schizophrenia Using Targeted Metabolomics. Psychiatry Investig. 2023, 20, 818–825. [Google Scholar] [CrossRef]
  38. Liu, M.L.; Zheng, P.; Liu, Z.; Xu, Y.; Mu, J.; Guo, J.; Huang, T.; Meng, H.Q.; Xie, P. GC-MS Based Metabolomics Identification of Possible Novel Biomarkers for Schizophrenia in Peripheral Blood Mononuclear Cells. Mol. Biosyst. 2014, 10, 2398–2406. [Google Scholar] [CrossRef]
  39. Al Awam, K.; Haußleiter, I.S.; Dudley, E.; Donev, R.; Brüne, M.; Juckel, G.; Thome, J. Multiplatform Metabolome and Proteome Profiling Identifies Serum Metabolite and Protein Signatures as Prospective Biomarkers for Schizophrenia. J. Neural Transm. 2015, 122, 111–122. [Google Scholar] [CrossRef]
  40. Avigdor, B.E.; Yang, K.; Shinder, I.; Orsburn, B.C.; Rais, R.; Kano, S.I.; Sawa, A.; Pevsner, J. Characterization of Antipsychotic Medications, Amino Acid Signatures, and Platelet-Activating Factor in First-Episode Psychosis. Biomark. Neuropsychiatry 2021, 5, 100045. [Google Scholar] [CrossRef]
  41. Campeau, A.; Mills, R.H.; Stevens, T.; Rossitto, L.A.; Meehan, M.; Dorrestein, P.; Daly, R.; Nguyen, T.T.; Gonzalez, D.J.; Jeste, D.V.; et al. Multi-Omics of Human Plasma Reveals Molecular Features of Dysregulated Inflammation and Accelerated Aging in Schizophrenia. Mol. Psychiatry 2022, 27, 1217–1225. [Google Scholar] [CrossRef]
  42. Cui, G.; Qing, Y.; Hu, X.; Wang, P.; Sun, L.; Yang, X.; Jiang, J.; Zhang, J.; Wang, H.; Feng, L.; et al. Serum Metabolomic Profiling Based on Fourier Transform-Ion Cyclotron Resonance-Mass Spectrometry: Do the Dysfunctions of Metabolic Pathways Reveal a Universal Risk of Oxidative Stress in Schizophrenia? Antioxid. Redox Signal. 2020, 33, 679–688. [Google Scholar] [CrossRef] [PubMed]
  43. Dickens, A.M.; Borgan, F.; Laurikainen, H.; Lamichhane, S.; Marques, T.; Rönkkö, T.; Veronese, M.; Lindeman, T.; Hyötyläinen, T.; Howes, O.; et al. Links between Central CB1-Receptor Availability and Peripheral Endocannabinoids in Patients with First Episode Psychosis. NPJ Schizophr. 2020, 6, 21. [Google Scholar] [CrossRef]
  44. Du, Y.; Chen, L.; Li, X.S.; Li, X.L.; Xu, X.D.; Tai, S.B.; Yang, G.L.; Tang, Q.; Liu, H.; Liu, S.H.; et al. Metabolomic Identification of Exosome-Derived Biomarkers for Schizophrenia: A Large Multicenter Study. Schizophr. Bull. 2021, 47, 615–623. [Google Scholar] [CrossRef] [PubMed]
  45. Fukushima, T.; Iizuka, H.; Yokota, A.; Suzuki, T.; Ohno, C.; Kono, Y.; Nishikiori, M.; Seki, A.; Ichiba, H.; Watanabe, Y.; et al. Quantitative Analyses of Schizophrenia-Associated Metabolites in Serum: Serum D-Lactate Levels Are Negatively Correlated with Gamma-Glutamylcysteine in Medicated Schizophrenia Patients. PLoS ONE 2014, 9, e101652. [Google Scholar] [CrossRef] [PubMed]
  46. 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]
  47. Koike, S.; Bundo, M.; Iwamoto, K.; Suga, M.; Kuwabara, H.; Ohashi, Y.; Shinoda, K.; Takano, Y.; Iwashiro, N.; Satomura, Y.; et al. A Snapshot of Plasma Metabolites in First-Episode Schizophrenia: A Capillary Electrophoresis Time-of-Flight Mass Spectrometry Study. Transl. Psychiatry 2014, 4, e379. [Google Scholar] [CrossRef]
  48. Liu, L.; Zhao, J.; Chen, Y.; Feng, R. Metabolomics Strategy Assisted by Transcriptomics Analysis to Identify Biomarkers Associated with Schizophrenia. Anal. Chim. Acta 2020, 1140, 18–29. [Google Scholar] [CrossRef]
  49. Mednova, I.A.; Chernonosov, A.A.; Kasakin, M.F.; Kornetova, E.G.; Semke, A.V.; Bokhan, N.A.; Koval, V.V.; Ivanova, S.A. Amino Acid and Acylcarnitine Levels in Chronic Patients with Schizophrenia: A Preliminary Study. Metabolites 2021, 11, 34. [Google Scholar] [CrossRef]
  50. Orešič, M.; Tang, J.; Seppänen-Laakso, T.; Mattila, I.; Saarni, S.E.; Saarni, S.I.; Lönnqvist, J.; Sysi-Aho, M.; Hyötyläinen, T.; Perälä, J.; et al. Metabolome in Schizophrenia and Other Psychotic Disorders: A General Population-Based Study. Genome Med. 2011, 3, 1–14. [Google Scholar] [CrossRef]
  51. 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]
  52. Paredes, R.M.; Quinones, M.; Marballi, K.; Gao, X.; Valdez, C.; Ahuja, S.S.; Velligan, D.; Walss-Bass, C. Metabolomic Profiling of Schizophrenia Patients at Risk for Metabolic Syndrome. Int. J. Neuropsychopharmacol. 2014, 17, 1139–1148. [Google Scholar] [CrossRef]
  53. Tasic, L.; Pontes, J.G.M.; Carvalho, M.S.; Cruz, G.; Dal Mas, C.; Sethi, S.; Pedrini, M.; Rizzo, L.B.; Zeni-Graiff, M.; Asevedo, E.; et al. Metabolomics and Lipidomics Analyses by 1H Nuclear Magnetic Resonance of Schizophrenia Patient Serum Reveal Potential Peripheral Biomarkers for Diagnosis. Schizophr. Res. 2017, 185, 182–189. [Google Scholar] [CrossRef]
  54. Tasic, L.; Larcerda, A.L.T.; Pontes, J.G.M.; da Costa, T.B.B.C.; Nani, J.V.; Martins, L.G.; Santos, L.A.; Nunes, M.F.Q.; Adelino, M.P.M.; Pedrini, M.; et al. Peripheral Biomarkers Allow Differential Diagnosis between Schizophrenia and Bipolar Disorder. J. Psychiatr. Res. 2019, 119, 67–75. [Google Scholar] [CrossRef] [PubMed]
  55. Tessier, C.; Sweers, K.; Frajerman, A.; Bergaoui, H.; Ferreri, F.; Delva, C.; Lapidus, N.; Lamaziere, A.; Roiser, J.P.; De Hert, M.; et al. Membrane Lipidomics in Schizophrenia Patients: A Correlational Study with Clinical and Cognitive Manifestations. Transl. Psychiatry 2016, 6, e906. [Google Scholar] [CrossRef]
  56. Wang, T.; Li, P.; Meng, X.; Zhang, J.; Liu, Q.; Jia, C.; Meng, N.; Zhu, K.; Lv, D.; Sun, L.; et al. An Integrated Pathological Research for Precise Diagnosis of Schizophrenia Combining LC-MS/1H NMR Metabolomics and Transcriptomics. Clin. Chim. Acta 2022, 524, 84–95. [Google Scholar] [CrossRef] [PubMed]
  57. 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]
  58. 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] [PubMed]
  59. 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]
  60. 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]
  61. 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]
  62. Xuan, J.; Pan, G.; Qiu, Y.; Yang, L.; Su, M.; Liu, Y.; Chen, J.; Feng, G.; Fang, Y.; Jia, W.; et al. Metabolomic Profiling to Identify Potential Serum Biomarkers for Schizophrenia and Risperidone Action. J. Proteome Res. 2011, 10, 5433–5443. [Google Scholar] [CrossRef] [PubMed]
  63. Qing, Y.; Wang, P.; Cui, G.; Zhang, J.; Liang, K.; Xia, Z.; Wang, P.; He, L.; Jia, W. Targeted Metabolomics Reveals Aberrant Profiles of Serum Bile Acids in Patients with Schizophrenia. Schizophrenia 2022, 8, 65. [Google Scholar] [CrossRef] [PubMed]
  64. Parksepp, M.; Haring, L.; Kilk, K.; Taalberg, E.; Kangro, R.; Zilmer, M.; Vasar, E. A Marked Low-Grade Inflammation and a Significant Deterioration in Metabolic Status in First-Episode Schizophrenia: A Five-Year Follow-Up Study. Metabolites 2022, 12, 983. [Google Scholar] [CrossRef]
  65. Qiu, Y.; Dong, Y.; Sun, W.; Li, G.; Li, M.J.; Zhao, Y.; Jiang, C.; Li, J. Metabolic Biomarkers of Risperidone-Induced Weight Gain in Drug-Naïve Patients with Schizophrenia. Front. Psychiatry 2023, 14, 1144873. [Google Scholar] [CrossRef] [PubMed]
  66. Liu, J.; Xiu, M.; Liu, H.; Wang, J.; Li, X. Plasma Lysophosphatidylcholine and Lysophosphatidylethanolamine Levels Were Associated With the Therapeutic Response to Olanzapine in Female Antipsychotics-Naïve First-Episode Patients With Schizophrenia. Front. Pharmacol. 2021, 12, 735196. [Google Scholar] [CrossRef] [PubMed]
  67. Cao, B.; Jin, M.; Brietzke, E.; McIntyre, R.S.; Wang, D.; Rosenblat, J.D.; Ragguett, R.M.; Zhang, C.; Sun, X.; Rong, C.; et al. Serum Metabolic Profiling Using Small Molecular Water-Soluble Metabolites in Individuals with Schizophrenia: A Longitudinal Study Using a Pre–Post-Treatment Design. Psychiatry Clin. Neurosci. 2019, 73, 100–108. [Google Scholar] [CrossRef]
  68. Farooqui, A.A.; Horrocks, L.A.; Farooqui, T. Glycerophospholipids in Brain: Their Metabolism, Incorporation into Membranes, Functions, and Involvement in Neurological Disorders. Chem. Phys. Lipids 2000, 106, 1–29. [Google Scholar] [CrossRef]
  69. Van Meer, G.; Voelker, D.R.; Feigenson, G.W. Membrane Lipids: Where They Are and How They Behave. Nat. Rev. Mol. Cell Biol. 2008, 9, 112. [Google Scholar] [CrossRef]
  70. Pilon, M. Revisiting the Membrane-Centric View of Diabetes. Lipids Health Dis. 2016, 15, 167. [Google Scholar] [CrossRef]
  71. Reddan, J.M.; White, D.J.; Macpherson, H.; Scholey, A.; Pipingas, A. Glycerophospholipid Supplementation as a Potential Intervention for Supporting Cerebral Structure in Older Adults. Front. Aging Neurosci. 2018, 10, 49. [Google Scholar] [CrossRef]
  72. Quach, T.T.; Stratton, H.J.; Khanna, R.; Kolattukudy, P.E.; Honnorat, J.; Meyer, K.; Duchemin, A.M. Intellectual Disability: Dendritic Anomalies and Emerging Genetic Perspectives. Acta Neuropathol. 2021, 141, 139–158. [Google Scholar] [CrossRef] [PubMed]
  73. Penzes, P.; Cahill, M.E.; Jones, K.A.; Vanleeuwen, J.E.; Woolfrey, K.M. Dendritic Spine Pathology in Neuropsychiatric Disorders. Nat. Neurosci. 2011, 14, 285–293. [Google Scholar] [CrossRef]
  74. De Carvalho, C.C.C.R.; Caramujo, M.J. The Various Roles of Fatty Acids. Molecules 2018, 23, 2583. [Google Scholar] [CrossRef] [PubMed]
  75. Witte, A.V.; Kerti, L.; Hermannstädter, H.M.; Fiebach, J.B.; Schreiber, S.J.; Schuchardt, J.P.; Hahn, A.; Flöel, A. Long-Chain Omega-3 Fatty Acids Improve Brain Function and Structure in Older Adults. Cereb. Cortex 2014, 24, 3059–3068. [Google Scholar] [CrossRef] [PubMed]
  76. Dighriri, I.M.; Alsubaie, A.M.; Hakami, F.M.; Hamithi, D.M.; Alshekh, M.M.; Khobrani, F.A.; Dalak, F.E.; Hakami, A.A.; Alsueaadi, E.H.; Alsaawi, L.S.; et al. Effects of Omega-3 Polyunsaturated Fatty Acids on Brain Functions: A Systematic Review. Cureus 2022, 14, e30091. [Google Scholar] [CrossRef] [PubMed]
  77. Sears, B.; Perry, M. The Role of Fatty Acids in Insulin Resistance. Lipids Health Dis. 2015, 14, 1–9. [Google Scholar] [CrossRef]
  78. Law, M.H.; Cotton, R.G.H.; Berger, G.E. The Role of Phospholipases A2 in Schizophrenia. Mol. Psychiatry 2006, 11, 547–556. [Google Scholar] [CrossRef]
  79. Moyer, C.E.; Shelton, M.A.; Sweet, R.A. Dendritic Spine Alterations in Schizophrenia. Neurosci. Lett. 2015, 601, 46–53. [Google Scholar] [CrossRef]
  80. Ziegler, A.B.; Tavosanis, G. Glycerophospholipids—Emerging Players in Neuronal Dendrite Branching and Outgrowth. Dev. Biol. 2019, 451, 25–34. [Google Scholar] [CrossRef]
  81. Haijma, S.V.; Van Haren, N.; Cahn, W.; Koolschijn, P.C.M.P.; Hulshoff Pol, H.E.; Kahn, R.S. Brain Volumes in Schizophrenia: A Meta-Analysis in over 18 000 Subjects. Schizophr. Bull. 2013, 39, 1129–1138. [Google Scholar] [CrossRef]
  82. Bowie, C.R.; Harvey, P.D. Cognitive Deficits and Functional Outcome in Schizophrenia. Neuropsychiatr. Dis. Treat. 2006, 2, 531. [Google Scholar] [CrossRef] [PubMed]
  83. Kasai, H.; Fukuda, M.; Watanabe, S.; Hayashi-Takagi, A.; Noguchi, J. Structural Dynamics of Dendritic Spines in Memory and Cognition. Trends Neurosci. 2010, 33, 121–129. [Google Scholar] [CrossRef] [PubMed]
  84. Du, J.; Xi, L.; Zhang, Z.; Ge, X.; Li, W.; Peng, W.; Jiang, X.; Liu, W.; Zhao, N.; Wang, X.; et al. Metabolic Remodeling of Glycerophospholipids Acts as a Signature of Dulaglutide and Liraglutide Treatment in Recent-Onset Type 2 Diabetes Mellitus. Front. Endocrinol. 2023, 13, 1097612. [Google Scholar] [CrossRef]
  85. Chen, W.; Zhao, H.; Li, Y. Mitochondrial Dynamics in Health and Disease: Mechanisms and Potential Targets. Signal Transduct. Target. Ther. 2023, 8, 1–25. [Google Scholar] [CrossRef] [PubMed]
  86. Belosludtsev, K.N.; Belosludtseva, N.V.; Dubinin, M.V. Diabetes Mellitus, Mitochondrial Dysfunction and Ca2+-Dependent Permeability Transition Pore. Int. J. Mol. Sci. 2020, 21, 6559. [Google Scholar] [CrossRef]
  87. Hishikawa, D.; Hashidate, T.; Shimizu, T.; Shindou, H. Diversity and Function of Membrane Glycerophospholipids Generated by the Remodeling Pathway in Mammalian Cells. J. Lipid Res. 2014, 55, 799–807. [Google Scholar] [CrossRef] [PubMed]
  88. Amminger, G.P.; Schäfer, M.R.; Klier, C.M.; Slavik, J.M.; Holzer, I.; Holub, M.; Goldstone, S.; Whitford, T.J.; McGorry, P.D.; Berk, M. Decreased Nervonic Acid Levels in Erythrocyte Membranes Predict Psychosis in Help-Seeking Ultra-High-Risk Individuals. Mol. Psychiatry 2012, 17, 1150–1152. [Google Scholar] [CrossRef]
  89. Strassnig, M.; Brar, J.S.; Ganguli, R. Dietary Fatty Acid and Antioxidant Intake in Community-Dwelling Patients Suffering from Schizophrenia. Schizophr. Res. 2005, 76, 343–351. [Google Scholar] [CrossRef]
  90. Stogios, N.; Smith, E.; Asgariroozbehani, R.; Hamel, L.; Gdanski, A.; Selby, P.; Sockalingam, S.; Graff-Guerrero, A.; Taylor, V.H.; Agarwal, S.M.; et al. Exploring Patterns of Disturbed Eating in Psychosis: A Scoping Review. Nutrients 2020, 12, 3883. [Google Scholar] [CrossRef]
  91. Berger, M.; Nelson, B.; Markulev, C.; Yuen, H.P.; Schäfer, M.R.; Mossaheb, N.; Schlögelhofer, M.; Smesny, S.; Hickie, I.B.; Berger, G.E.; et al. Relationship between Polyunsaturated Fatty Acids and Psychopathology in the NEURAPRO Clinical Trial. Front. Psychiatry 2019, 10, 393. [Google Scholar] [CrossRef]
  92. Barber, M.N.; Risis, S.; Yang, C.; Meikle, P.J.; Staples, M.; Febbraio, M.A.; Bruce, C.R. Plasma Lysophosphatidylcholine Levels Are Reduced in Obesity and Type 2 Diabetes. PLoS ONE 2012, 7, e41456. [Google Scholar] [CrossRef]
  93. Liu, J.H.; Chen, N.; Guo, Y.H.; Guan, X.N.; Wang, J.; Wang, D.; Xiu, M.H. Metabolomics-Based Understanding of the Olanzapine-Induced Weight Gain in Female First-Episode Drug-Naïve Patients with Schizophrenia. J. Psychiatr. Res. 2021, 140, 409–415. [Google Scholar] [CrossRef] [PubMed]
  94. Castellani, L.N.; Costa-Dookhan, K.A.; McIntyre, W.B.; Wright, D.C.; Flowers, S.A.; Hahn, M.K.; Ward, K.M. Preclinical and Clinical Sex Differences in Antipsychotic-Induced Metabolic Disturbances: A Narrative Review of Adiposity and Glucose Metabolism. J. Psychiatr. Brain Sci. 2019, 4, e190013. [Google Scholar] [CrossRef] [PubMed]
  95. Sariah, A.E.; Outwater, A.H.; Malima, K.I.Y. Risk and Protective Factors for Relapse among Individuals with Schizophrenia: A Qualitative Study in Dar Es Salaam, Tanzania. BMC Psychiatry 2014, 14, 240. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Preferred reporting items for systematic reviews and meta-analyses (PRISMA) flow chart of included studies.
Figure 1. Preferred reporting items for systematic reviews and meta-analyses (PRISMA) flow chart of included studies.
Metabolites 14 00475 g001
Figure 2. Potential associations between glycerophospholipids, fatty acyls, and schizophrenia spectrum disorders; ↑ = increased, ↓ = decreased.
Figure 2. Potential associations between glycerophospholipids, fatty acyls, and schizophrenia spectrum disorders; ↑ = increased, ↓ = decreased.
Metabolites 14 00475 g002
Table 1. Characteristics of cross-sectional components of studies (N = 17) examining minimally antipsychotic-treated schizophrenia spectrum disorder patients.
Table 1. Characteristics of cross-sectional components of studies (N = 17) examining minimally antipsychotic-treated schizophrenia spectrum disorder patients.
Author,
Year
Analytical ToolDiagnosisAP-Naive
Window
AP-NaiveControlsSignificantly Different Metabolic Parameters
(SSDs Compared to HCs)
NSex, Female (%)Mean Age (SD)NSex, Female (%)Mean Age (SD)
Bicikova 2013 #
[20]
GC–MSSSDsAP-naive2138Males: Median = 31 (range: 22–52)
Females: Median = 35 (range: 24–52)
3241Males: Median = 35 (range: 23–53)
Females: Median = 35 (range: 23–51)
TG *: ↑
Cholesterol *: ↑
HDL *: ↓
* Only in AP-naive FEP males compared to HCs
Cai 2012
[23]
UPLC–MS/MS and H-NMRSSDsAP-naive1145.4527.6 (9.5)1145.4527.6 (9.5)FBG: ↓
Insulin: ↑
Cui 2021
[24]
UPLC–QTOF-MS/MSFESAP-naive8342.225.60 (7.42)785023.03 (6.45)NR
Kaddurah-Daouk 2012
[25]
GC + flame ionization detectorSSDsAP-naive203527.0 (9.8)176637.9 (9.1)None
Kriisa 2017 #
[26]
FIA–MS/MS and LC–MSFEP, SSDsAP-naive38NRRange: 18–4537NRNRNone
Lee 2023
[32]
LC–MSPSDsAP-naive or AP-free (3 weeks or less in past 3 months)253221.8 (4.1)65025.2 (3.5)None
Leppik 2020 #
[27]
FIA–MS/MS and LC–MSFEP; SSDsAP-naive5339.6026.2 (6.0)3756.8024.8 (5.3)NR
Liu 2014
[38]
GC–MSFES and SSDsAP-naive (42.2% AP-treated)456033.22 (12.9)5056.0037.26 (8.67)NR
Liu 2021
[28]
LC–MS-based multiple reaction monitoringSSDsAP-naive3857.934.82 (13.33)254033.7 (12.5)None
Qiao 2016 #
[29]
LC–MSFESAP-naive15100.0028.25 (5.34)1510027.6 (6.3)None
Schwarz 2011
[30]
LC–MSFEP, SSDsAP-naive7034.2029.3 (10.0)5944.127.5 (5.9)NR
Shang 2022 *,#
[33]
UPLC–TOF-MSFEPNot treated for more than 30 days; mean (SD): 10 (2.9) days254831.40 (1.96) a216225.81 (1.34) aNone
Song 2023 #
[34]
UPLC–QTOF-MSSSDsAP-naive434227.3 (6.0)295527.1 (4.2)None
Su 2023
[37]
FIA/LS–MSSSDsAP-naive607238.08 (10.48)365638.03 (9.66)BMI: ↓
Wang X 2024 *
[36]
FIA–MS/MS and LC–MS/MSSSDsAP-naive3810039.74 (9.83)1910040 (9.1)None
Wang Z 2024
[35]
LC–MSSSDsAP-naive12752.821.65 (7.55)926222.95 (2.59)None
Yan 2018 #
[31]
LC–MSSSDsAP-naive2045.0032.1 (9.1)2937.932.4 (7.8)NR
a = standard error of the mean; AP = antipsychotic; BMI = body mass index; FBG = fasting blood glucose; FIA = flow injection analysis; FEP = first-episode psychosis; FES = first-episode schizophrenia; GC = gas chromatography; HDL = high-density lipoprotein; H-NMR = proton nuclear magnetic resonance; LC = liquid chromatography; MS = mass spectrometry; MS/MS = tandem mass spectrometry; NR = not reported; PSDs = psychotic spectrum disorders; QTOF = quadrupole time-of-flight; SD = standard deviation; SSDs = schizophrenia spectrum disorders; TG = triglycerides; UPLC = ultra performance liquid chromatography; ↑ = increased; ↓ = decreased; * = shared studies between minimally AP-treated and AP-treated groups; # = shared studies between minimally AP-treated and pre-to-post AP treatment groups.
Table 2. Characteristics of cross-sectional components of studies (N = 28) examining antipsychotic-treated patients.
Table 2. Characteristics of cross-sectional components of studies (N = 28) examining antipsychotic-treated patients.
Author, YearAnalytical ToolAntipsychoticsDiagnosisAntipsychotic-Treated PatientsControlsSignificantly Different Metabolic Parameters
(SSDs Compared to HCs)
NSex, Female (%)Mean Age (SD)NSex, Female (%)Mean Age (SD)
AlAwam 2015
[39]
GC–MSARI, CLO, CLZ, FLU, HAL, LEV, MEL, OLA, PAL, PRO, QUE, RIS, ZUCSSDs2623.137.27 (12.4)2623.137.0 (10.7)NR
Avigdor 2021
[40]
UHPLC–MSARI, CLZ, LUR, QUE, RIS, TRI, ZIPSSDs3138.723.77 (4.4)3545.723.74 (3.0)NR
Campeau 2022
[41]
LC–MSRISSSDs5453.753.8 (10.0)514952.1 (11.5)NR
Cui 2020
[42]
LC–MSNRSSDs5466.638.4 (11.6)5475.933.6 (9.87)None
Dickens 2020
[43]
LC–QqQMSARI, OLA, PER, RISSSDs8026.4 (3.6)10027.18 (5.9)None
Du 2021
[44]
UPLC–MSNRSSDs7814.1031.7 (7.7)6642.4024.5 (5.1)NR
Fukushima 2014
[45]
HPLC–MSARI, BLO, FLU, LEV, OLA, PAL, QUE, RIS, SUL,SSDs255628.2 (4.4)2755.626.5 (5.6)BMI: ↑
He 2012
[46]
LC–MSAMI, CLZ, HAL, OLA, QUE, RISSSDs21338.0036.9 (11.7)21648.10%38.9 (10.6)None
Kaddurah-Daouk 2007 #
[59]
GC + flame ionization detectorARI, OLA, RISSSDs2785.132.3 (5.0)16MatchedMatchedNone
Koike 2014
[47]
CE–TOF-MSNRSSDs1827.7823.2 (5.4)1421.4325.7 (6.1)NR
Li 2022 #
[60]
GC–MSARI, AMI, CLZ, OLA, RIS,
Others
SSDs3275532.70 (10.40)15952.2032.86 (10.09)None
Liu 2020
[48]
LC–TOF-MS and H-NMRNRSSDs5563.633.4 (12.8)5763.244.4 (13.5)TC: ↓
TG: ↓
HDL: ↓
Mednova 2021
[49]
LC–MS/MSNRSSDs3748.65Median: 35 (IQR: 31.00. 39.00)3638.89Median: 32.5
(IQR: 28.75; 40.25)
None
Oresic 2011
[50]
UPLC–MSAtypical and typical APsSSDs4557.853.7 (12.9)4557.853.7 (12.9)T2D (n): ↑
FBG: ↑
TG: ↑
Insulin: ↑
HOMA-IR: ↑
WC: ↑
HDL: ↓
Oresic 2012
[51]
LC–TOF-MSAtypical APsSSDs1968.4Median: 51 (IQR: 46.4, 55.6)3470.6Median: 53.4 (IQR: 50.2, 56.6)BMI: ↑
Paredes 2014
[52]
LC–MSARI, CLZ, OLA. QUE, RIS, ZIPSSDs6023.342.5 (2.6)203041.1. (2.6)Insulin: ↑
Parksepp 2022
[64]
(FIA)−MS/MS + (LC)−MS/MSVarious APs, types NRFEP385731.8 (5.9)585624.7 (4.5)NR
Qing 2022
[63]
UPLC–MS/MSNRSSDs596137.48 (11.80)605836.80 (9.74)None
Shang 2022 *,#
[33]
UPLC–TOF-MSARI, CLZ, HAL, OLA, QUE, RISFEP254831.40 (1.96) a216225.81 (1.34) aNone
Tasic 2017
[53]
H-NMROLA, QUESSDs273736 (10.3)2665.436 (13.1)NR
Tasic 2019
[54]
H-NMRCLZ, HAL, OLA, QUE, RISSSDs504835.4 (9.5)607036 (10.5)NR
Tessier 2016
[55]
LC–MSAMI, ARI, CLZ, CPZ, FLU, HAL,
OLA, RIS, SER
SSDs7435.243.8 (9.3)404042.6 (13.2)NR
Wang 2021
[61]
LC–MSNRSSDs11956.3029.0
(IQR: 25, 33.3)
10966.130
(IQR: 26, 33)
TC: ↑
Wang 2022
[56]
LC–MS and H-NMRNRSSDs64NR44.56 (9.53)40NR43.76 (13.87)NR
Wang X 2024 *
[36]
FIA–MS/MS and LC–MS/MSOLA, PAL, RISSSDs3810039.74 (9.83)1910040 (9.1)None
Wood 2015
[57]
MS/MSAtypical APsSSDs2321.7Median: 47 (range: 25–66)2766.7Median: 47
(range: 25–65)
None
Xuan 2011 #
[62]
GC–MSNRSSDs1844.441.3 (16.1)1844.441 (15.0)NR
Yang 2017
[58]
UPLC–QTOF-MSNRSSDs606037.2 (12.0)615936.9 (9.7)BMI: ↓
a = standard error of the mean; AMI = amisulpride; ARI = aripiprazole; BLO = blonanserin; BMI = body mass index; CE = capillary electrophoresis; CLO = chlorprothixene; CLZ = clozapine; CPZ = chlorpromazine; FBG = fasting blood glucose; FEP = first-episode psychosis; FLU = flupentixol; GC = gas chromatography; HAL = haloperidol; HDL = high-density lipoprotein; H-NMR = proton nuclear magnetic resonance; HPLC = high performance LC; HOMA-IR = Homeostatic Model Assessment for Insulin Resistance; IQR = interquartile range; LC = liquid chromatography; LEV = levomepromazine; LUR = lurasidone; MEL = melperone; MS = mass spectrometry; MS/MS = tandem mass spectrometry; NR = not reported; OLA = olanzapine; PAL= paliperidone; PER= perphenazine; PRO = promethazine; QqQMS = quadrupole tandem MS; QTOF = quadrupole time-of-flight; QUE = quetiapine; RIS = risperidone; SD = standard deviation; SER = sertindole; SSDs = schizophrenia spectrum disorders; SUL = sulpiride; TC = total cholesterol; TG = triglycerides; TOF-MS = time-of-flight mass spectrometry; TRI = trifluoperazine; T2D = type 2 diabetes; UHPLC = ultra-high performance liquid chromatography; UPLC = ultra-performance liquid chromatography; WC = waist circumference; ZIP = ziprasidone; ZUC = zuclopenthixol; ↑ = increased; ↓ = decreased; * = shared studies between minimally AP-treated and AP-treated groups; # = shared studies between AP-treated and pre-to-post AP treatment groups.
Table 3. Characteristics of included studies (N = 13) examining pre-to-post antipsychotic-treated patients.
Table 3. Characteristics of included studies (N = 13) examining pre-to-post antipsychotic-treated patients.
Author, Year;
Duration Treated
Analytical ToolAP-Naive at BaselineDiagnosis; AP TypeSSD GroupSignificantly Different Metabolic
Parameters
(Post-Treatment Compared to
Pre-Treatment)
Significantly Different Symptom
Scale Scores
(Post-Treatment Compared to
Pre-Treatment)
NSex, Female (%)Mean Age (SD)
Bicikova 2013 *;
26 weeks
[20]
GC–MSAP-naiveSSDs;
AMI, OLA, RIS
229Males: Median 31 (range: 22–52)
Females: Median 35 (range: 24–52)
NRNR
Cao 2019;
8 weeks
[67]
LC–MSAP-naive or no AP use for 30 daysSSDs;
ARI, CLZ, CPZ, HAL, OLA, PRO, QUE, RIS, SUL, ZIP
12257.428.91 (6.21)BMI: ↑
WC: ↑
TG: ↑
VLDL: ↑
FBG: ↓
HDL: ↓
PANSS Total: ↓
PANSS Positive: ↓
PANSS Negative: ↓
PANS General: ↓
Kaddurah-Daouk 2007 #;
2–3 weeks
[59]
GC + flame ionization detectorNo AP treatment for at least 3 weeksSSDs;
ARI, OLA, RIS
27
(ARI = 4,
OLA = 14,
RIS = 9)
85.132.3 (5.0)NRNR
Kriisa 2017 *;
30 weeks
[26]
FIA–MS/MS and LC–MSAP-naiveSSDs;
ARI, CLZ, OLA, QUE, RIS, SER, ZIP
36NRRange: 18–45BMI: ↑PANSS Total: ↓
PANSS Positive: ↓
PANSS Negative: ↓
PANSS General: ↓
Leppik 2020 *;
30 weeks
[27]
FIA–MS/MS and LC–MSAP-naiveFEP, SSDs;
ARI, CLZ, OLA, PER, QUE, RIS, SER, ZIP
4439.6026.20 (6.00)BMI: ↑BPRS: ↓
Li 2022 #;
4 weeks
[60]
GC–MSNo AP use for 30 daysSSDs;
ARI, AMI, CLZ, OLA, RIS
3275532.70 (10.40)NRPANSS Total: ↓
PANSS Positive: ↓
PANSS General: ↓
Liu 2021;
4 weeks
[66]
UPLC–QTOF-MS/MSAP-naiveFEP, SSDs;
OLA
2510027.4 (7.6)NRPANSS Total: ↓
PANSS Positive: ↓
PANSS General: ↓
Qiao 2016 *;
4 weeks
[29]
LC–MSAP-naiveFES;
OLA
1510028.20 (5.34)LDL: ↑PANSS Total: ↓
PANSS Positive: ↓
PANSS General: ↓
Qiu 2023;
8 weeks
[65]
LC–MS/MS and (FIA–MS/MSAP-naiveSSDs;
RIS
305036.40 (12.10)Weight: ↑
BMI: ↑
WC: ↑
HC: ↑
PANSS Total: ↓
PANSS Positive: ↓
PANSS Negative: ↓
PANSS General: ↓
Shang 2022 *,#;
78 weeks
[33]
UPLC–TOF-MSAP-naiveFEP;
ARI, CLZ, HAL,
OLA, QUE
254831.40 (1.96) aNRPANSS Total: ↓
PANSS Positive: ↓
PANSS General: ↓
Song 2023 *;
4–6 weeks
[34]
UPLC–MSAP-naiveSSDs;
OLA
434227.30 (6.00)BMI: ↑
TG: ↑
PANSS Total: ↓
PANSS Positive: ↓
PANSS Negative: ↓
PANSS General: ↓
Xuan 2011 #;
8 weeks
[62]
GC–MSUnmedicated (duration not specified)SSDs;
RIS
1844.441.3 (16.1)NRPANSS Total: ↓
PANSS Positive: ↓
PANSS Negative: ↓
Yan 2018 *;
8 weeks
[31]
LC–MSAP-naiveSSDs;
CLZ, HAL, OLA, QUE, RIS
2045.0032.1 (9.1)NRNR
a = standard error of the mean; AMI = amisulpride; ARI = aripiprazole; BMI = body mass index; BPRS = Brief Psychiatric Rating Scale; CLZ = clozapine; CPZ = chlorpromazine; FBG = fasting blood glucose; FEP = first-episode psychosis; FES = first-episode schizophrenia; FIA = flow injection analysis; GC = gas chromatography; HAL = haloperidol; HDL = high-density lipoprotein; LC = liquid chromatography; LDL = low-density lipoprotein; MS = mass spectrometry; MS/MS = tandem mass spectrometry; NR = not reported; OLA = olanzapine; PAL= paliperidone; PANSS = Positive and Negative Syndrome Scale; PER = perphenazine; PRO = promethazine; QTOF = quadrupole time-of-flight; QUE = quetiapine; RIS = risperidone; SD = standard deviation; SER= sertindole; SSDs = schizophrenia spectrum disorders; SUL = sulpiride; TG = triglycerides; TOF-MS = time-of-flight mass spectrometry; UPLC = ultra-performance liquid chromatography; VLDL = very low-density lipoprotein; WC = waist circumference; ZIP = ziprasidone; ↑ = increased; ↓ = decreased; * = shared studies between minimally AP-treated and pre-to-post AP treatment groups; # = shared studies between AP-treated and pre-to-post AP treatment groups.
Table 4. Minimally antipsychotic-treated patients’ lipid signatures that appeared in at least two studies when compared to healthy controls.
Table 4. Minimally antipsychotic-treated patients’ lipid signatures that appeared in at least two studies when compared to healthy controls.
ClassLipid, HMDB IDBicikova 2013
[20]
Cai 2012
[23]
Cui 2021
[24]
Kaddurah-Daouk 2012
[25]
Kriisa 2017
[26]
Lee 2023
[32]
Leppik 2020
[27]
Liu 2021
[28]
Qiao 2016
[29]
Song 2023
[34]
Su 2023
[37]
Wang X 2024
[36]
Wang Z 2024
[35]
Yan 2018
[31]
Overall Direction
Fatty AcylsStearic acid, HMDB0000827 - - Downregulated
GlycerophospholipidsLPC (14:0), HMDB0010379 - -Downregulated
GlycerophospholipidsLPC (18:0), HMDB0010384 -- -Downregulated
GlycerophospholipidsPC (O-34:2), HMDB0011151 -- Downregulated
GlycerophospholipidsPC (36:2), HMDB0000593 - - Downregulated
GlycerophospholipidsPC (O-36:0), HMDB0013406 - - Downregulated
GlycerophospholipidsPC (O-36:3), HMDB0013425 - - Downregulated
Fatty AcylsArachidonic acid, HMDB0001043 + - Conflicting
Fatty AcylsBehenic acid, HMDB0000944 + - Conflicting
Fatty AcylsFumarylcarnitine, HMDB0013134 - + Conflicting
GlycerophospholipidsPC (32:1), HMDB0007872 -+ Conflicting
Steroids and Steroid DerivativesGlycohyocholic acid, HMDB0240607 - + Conflicting
Steroids and Steroid DerivativesDHEAS, HMDB0001032+ - Conflicting
GlycerophospholipidsLPC (16:0), HMDB0010382 + + - Upregulated
Steroids and Steroid DerivativesCE 16:1, HMDB0000658 + +Upregulated
Steroids and Steroid DerivativesCE 20:3, HMDB0006736 + +Upregulated
Steroids and Steroid DerivativesCholic acid, HMDB0000619 ++ Upregulated
CE = cholesterol ester; DHEAS = dehydroepiandrosterone sulfate; LPC = lysophosphatidylcholine; PC = phosphatidylcholine.
Table 5. Antipsychotic-treated patients’ lipid signatures that appeared in at least two studies when compared to healthy controls.
Table 5. Antipsychotic-treated patients’ lipid signatures that appeared in at least two studies when compared to healthy controls.
ClassLipid,
HMDB ID
AlAwam 2015
[39]
Avigdor 2021
[40]
Campeau 2022
[41]
Cui 2020
[42]
Dickens 2020
[43]
Du 2020
[44]
Fukushima 2014
[45]
He 2012
[46]
Kaddurah-Daouk 2007
[59]
Li 2022
[60]
Liu 2020
[48]
Mednova 2021
[49]
Oresic 2011
[50]
Oresic 2012
[51]
Paredes 2014
[52]
Parksepp 2022
[64]
Qing 2022
[63]
Tessier 2016
[55]
Wang 2021
[61]
Wang 2022
[56]
Wang X 2024
[36]
Wood 2015
[57]
Xuan 2011
[62]
Yang 2017
[58]
Overall
Direction
Fatty AcylsArachidonic acid, HMDB0001043 +- - - - +Downregulated
Fatty AcylsDocosahexaenoic acid, HMDB0002183 - - Downregulated
Fatty AcylsLinoleic acid, HMDB0000673 - -- - Downregulated
Fatty AcylsOleic acid, HMDB0000207- + - - + -+Downregulated
Fatty AcylsPalmitic acid, HMDB0000220 - - + -+Downregulated
Fatty AcylsStearic acid, HMDB0000827 + - - + - Downregulated
GlycerophospholipidsLPC (16:0), HMDB0010382 - - -- Downregulated
GlycerophospholipidsLPC (17:0), HMDB0012108 -- Downregulated
GlycerophospholipidsLPC (18:0), HMDB0010384 - - -- Downregulated
GlycerophospholipidsLPE (18:0), HMDB0011130 -- Downregulated
GlycerophospholipidsLPE (18:1), HMDB0011506 -- Downregulated
GlycerophospholipidsPC (O-38:6), HMDB0013409 - - Downregulated
Fatty Acyls9-Hydroxylinoleic acid, HMDB0062652 + - Conflicting
Fatty AcylsAcetylcarnitine, HMDB0000201 - + Conflicting
Fatty AcylsAcylcarnitine C16-OH, N/A - + Conflicting
Fatty AcylsAcylcarnitine C16:1, N/A - + Conflicting
Fatty AcylsAcylcarnitine C16:1-OH, N/A - + Conflicting
Fatty AcylsAcylcarnitine C18:1-OH, N/A - + Conflicting
Fatty AcylsArachidic acid, HMDB0002212- + Conflicting
Fatty AcylsHeptadecenoic acid, HMDB0002259- + Conflicting
Fatty AcylsDihomo-gamma-linolenic acid, HMDB0002925 - +Conflicting
Fatty AcylsDocosapentaenoic acid, HMDB0006528 - +Conflicting
Fatty AcylsOleamide, HMDB0002117 + - Conflicting
Fatty AcylsOleoylcarnitine, HMDB0005065 - + Conflicting
Fatty AcylsStearoylcarnitine, HMDB0062532 - + Conflicting
GlycerophospholipidsLPC (14:0), HMDB0010379 + - Conflicting
GlycerophospholipidsLPC (18:1), HMDB0002815 + + - - Conflicting
GlycerophospholipidsLPC (22:6), HMDB0010404 +- Conflicting
GlycerophospholipidsPE (18:0/18:1), HMDB0008993 -+ Conflicting
GlycerophospholipidsPC (18:0/18:2), HMDB0008039 + - Conflicting
GlycerophospholipidsPE (18:2/18:0), HMDB0009090 -+ Conflicting
Steroids and Steroid DerivativesCholic acid, HMDB0000619 - + Conflicting
Steroids and Steroid DerivativesProgesterone, HMDB0001830 - + Conflicting
Fatty AcylsAdrenic acid, HMDB0002226 +-+ + +Upregulated
Fatty AcylsEicosenoic acid, HMDB0002231 + +Upregulated
Fatty AcylsEicosadienoic acid, HMDB0005060 + +Upregulated
Fatty AcylsLinoleamide, HMDB0062656 + + + Upregulated
Fatty AcylsMyristic acid, HMDB0000806 - + + Upregulated
Fatty AcylsNervonic acid, HMDB0002368 + +Upregulated
Fatty AcylsPalmitaldehyde, HMDB0001551 + + Upregulated
GlycerophospholipidsLPE (16:0), HMDB0011503 + + Upregulated
GlycerophospholipidsPC (16:0/18:1), HMDB0007971 ++ Upregulated
GlycerophospholipidsPlatelet-activating factor, HMDB0062195 ++ Upregulated
SphingolipidsSM (d18:1/18:0), HMDB0001348 + + Upregulated
Steroids and Steroid DerivativesCholesterol, HMDB0000067- + + Upregulated
Steroids and Steroid DerivativesSulfolithocholic acid, HMDB0000907 + + Upregulated
LPC = lysophosphatidylcholine; LPE = lysophosphatidylethanolamine; PC = phosphatidylcholine; PE = phosphatidylethanolamine; SM = sphingomyelin.
Table 6. Pre-to-post antipsychotic-treated patients’ lipid signatures. Overall direction is in relation to post-treatment.
Table 6. Pre-to-post antipsychotic-treated patients’ lipid signatures. Overall direction is in relation to post-treatment.
ClassLipid, HMDB IDCao 2019
[67]
Kaddurah-Daouk 2007
[59]
Leppik 2020
[27]
Li 2022
[60]
Liu 2021
[66]
Qiao 2016
[29]
Qiu 2023
[65]
Song 2023
[34]
Xuan 2011
[62]
Yan 2018
[31]
Overall Direction
Fatty AcylsPalmitoleic acid, HMDB0003229 - - Downregulated
GlycerophospholipidsLPC (16:0), HMDB0010382-- + Downregulated
GlycerophospholipidsLPC (22:6), HMDB0010404 - -Downregulated
GlycerophospholipidsPC (38:6), HMDB0007991 - -Downregulated
Steroids and Steroid DerivativesCE (22:6), HMDB0245627 - -Downregulated
Fatty AcylsDocosahexaenoic acid, HMDB0002183 - + Conflicting
Fatty AcylsDocosapentaenoic acid, HMDB0006528 - + Conflicting
Fatty AcylsEicosapentaenoic acid, HMDB0001999 - + Conflicting
Fatty AcylsPalmitic acid, HMDB0000220 - + +-Conflicting
Fatty AcylsStearic acid, HMDB0000827 + - Conflicting
GlycerophospholipidsLPC (15:0), HMDB0010381- + Conflicting
GlycerophospholipidsLPC (18:2), HMDB0061700 + -Conflicting
Steroids and Steroid DerivativesCE (20:5), HMDB0006731 + -Conflicting
Fatty AcylsAdrenic acid, HMDB0002226 + + -Upregulated
Fatty AcylsLinoleic acid, HMDB0000673 + -+ Upregulated
Fatty AcylsOleic acid, HMDB0000207 + -+ Upregulated
GlycerophospholipidsLPC (14:0), HMDB0010379-++ +++ Upregulated
GlycerophospholipidsLPC (16:1), HMDB0010383 + + Upregulated
GlycerophospholipidsLPC (18:0), HMDB0010384 + + + Upregulated
GlycerophospholipidsLPC (18:1), HMDB0002815 + + Upregulated
GlycerophospholipidsLPC (20:3), HMDB0010393 ++ +++ Upregulated
Steroids and Steroid DerivativesCE (18:3), HMDB0010369 + + Upregulated
CE = cholesterol ester; FA = fatty acid; LPC = lysophosphatidylcholines; PC = phosphatidylcholines.
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

Wu, S.; Panganiban, K.J.; Lee, J.; Li, D.; Smith, E.C.C.; Maksyutynska, K.; Humber, B.; Ahmed, T.; Agarwal, S.M.; Ward, K.; et al. Peripheral Lipid Signatures, Metabolic Dysfunction, and Pathophysiology in Schizophrenia Spectrum Disorders. Metabolites 2024, 14, 475. https://doi.org/10.3390/metabo14090475

AMA Style

Wu S, Panganiban KJ, Lee J, Li D, Smith ECC, Maksyutynska K, Humber B, Ahmed T, Agarwal SM, Ward K, et al. Peripheral Lipid Signatures, Metabolic Dysfunction, and Pathophysiology in Schizophrenia Spectrum Disorders. Metabolites. 2024; 14(9):475. https://doi.org/10.3390/metabo14090475

Chicago/Turabian Style

Wu, Sally, Kristoffer J. Panganiban, Jiwon Lee, Dan Li, Emily C.C. Smith, Kateryna Maksyutynska, Bailey Humber, Tariq Ahmed, Sri Mahavir Agarwal, Kristen Ward, and et al. 2024. "Peripheral Lipid Signatures, Metabolic Dysfunction, and Pathophysiology in Schizophrenia Spectrum Disorders" Metabolites 14, no. 9: 475. https://doi.org/10.3390/metabo14090475

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop