ArticlePDF Available

Metabolomics in Schizophrenia and Major Depressive Disorder

Authors:
  • Harvard TH Chan School of Public Health

Abstract

Defining pathophenotype, a systems level consequence of a disease genotype, together with environmental and stochastic influences, is an arduous task in psychiatry. It is also an appealing goal, given growing need for appreciation of brain disorders biological complexity, aspiration for diagnostic tests development and ambition to identify novel drug targets. Here, we focus on the Schizophrenia and Major Depressive Disorder and highlight recent advances in metabolomics research. As a systems biology tool, metabolomics holds a promise to take part in elucidating interactions between genes and environment, in complex behavioral traits and psychopathology risk translational research.
REVIEW
Metabolomics in Schizophrenia and Major Depressive Disorder
Iva Petrovchich
2, *
, Alexandra Sosinsky
3, *
, Anish Konde
4, *
, Abigail Archibald
5
, David Henderson
6
, Mirjana Maletic-Savatic
7
,
Snezana Milanovic ()
1
1
Massachusetts General Hospital, Department of Psychiatry, MGH Clinical Trials Network Institute, MGH Division of Global Psychiatry,
MGH Depression Clinical and Research Program, Boston, MA 02114, USA
2
School of Nursing, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
3
Massachusetts General Hospital, Department of Psychiatry, MGH Center for Womens Mental Health, Boston, MA 02114, USA
4
Louisiana State University Health Science Center, Department of Internal Medicine, Lafayette, LA 70112, USA
5
Massachusetts General Hospital, Department of Psychiatry, MGH Depression Clinical and Research Program, Boston, MA 02114, USA
6
Boston Medical Center, Department of Psychiatry, Boston, MA 02118, USA
7
Baylor College of Medicine, Neurology Research Institute, Houston, TX77030, USA
© Higher Education Press and Springer-Verlag Berlin Heidelberg 2016
Abstract Dening pathophenotype, a systems level consequence of a disease genotype, together with environmental
and stochastic inuences, is an arduous task in psychiatry. It is also an appealing goal, given growing need for
appreciation of brain disorders biological complexity, aspiration for diagnostic tests development and ambition to
identify novel drug targets. Here, we focus on the Schizophrenia and Major Depressive Disorder and highlight recent
advances in metabolomics research. As a systems biology tool, metabolomics holds a promise to take part in elucidating
interactions between genes and environment, in complex behavioral traits and psychopathology risk translational
research.
Keywords Schizophrenia, Major Depressive Disorder, omics, metabolomics, systems biology
Introduction
A major component of psychiatric illness risk is polygenic:
heritability that arises from many genetic loci having a small
effect.Polygenicity and rare large-effect genetic loci are at
the core of complex quest to elucidate under print of
psychiatric disorders. Given the limited biological insight
obtained through Genome Wide Association Studies
(GWAS), the focus of genetic research has shifted to
exploring major effect-size contributions (rare copy variants
and whole exome sequencing), improving phenotype deni-
tion, and attempts to connect various genetic risks to brain
mechanisms. In parallel, advances in the bioinformatics,
multivariate statistics, and the high-throughput analytical
approaches utilized for processing of microarray data have
paved the way for further development of the systems biology
panomicview of the organism, which will be necessary for
the successful unlockingof psychiatric disorders. However,
integration of so called omicssciences (genomics, tran-
scriptomics, proteomics, metabolomics) is a formidable task,
as it aims to capture the cross-talk between different levels of
molecular organization, represented by individual omics
sciences. Nevertheless, the implications of such development
are very signicant, not only for the further advances of our
knowledge of molecular mechanisms, but also for psychiatric
practice, which lacks specic diagnostic, prognostic, and
therapeutic biomarkers in a wide range of diseases.
Metabolites are the nal product of interactions between
gene expression, protein expression, and the cellular
environment. Metabolome presents a state regulated by
interactions between genes and environment, and possibly
genotype and phenotype. As such, it has the potential to be an
informative target for genetic studies of intermediate
phenotypes in brain disorders and help illustrate their
heterogeneity.
Schizophrenia
Schizophrenia is a severe complex mental disorder character-
ized by psychotic symptoms such as hallucinations, delu-
Received March 16, 2016; accepted April 15, 2016
Correspondence: Snezana Milanovic
E-mail: SMILANOVIC@mgh.harvard.edu
*These authors contributed equally to this work.
Front. Biol. 2016, 11(3): 222231
DOI 10.1007/s11515-016-1400-8
sions, and decits in executive function (Whiteld-Gabrieli et
al., 2009; Milanovic et al., 2011). It affects 0.5%1%of the
world population. Similar to other psychiatric disorders,
diagnosis of schizophrenia is based on patients and familys
reports and thus subjective clinical evaluation. The under-
lying molecular mechanisms of schizophrenia are poorly
understood (Yao et al., 2010; He et al., 2012). Symptomatic
onset is in early adulthood- factors such as environment, drug
abuse, and childhood trauma play important roles, as well as
common genetic variations and rare variants. Estimated
heritability is 0.80. Although initial genetic studies demon-
strated increased risk of de novo mutations, larger sample
sizes failed to replicate these data. So far, identication of the
individual risk genes using Whole Exome Sequencing has
had no signicant yield. A variation in the Major Histocom-
patibility Complex locus was shown to have a strong
association with schizophrenia at the population level. A
follow up study (Sekar et al., 2016) identied complement
complex alleles playing the role in synapse elimination during
postnatal development. This nding is consistent with
reported excessive loss of gray matter and synapses in
schizophrenia. In spite of high heritability, clinical hetero-
geneity remains a major limitation in schizophrenia genetic
research. Schizophrenia is not one disease entity, but instead a
cluster of clinical symptoms. Identical genomic causation is
probably shared only by subsets of schizophrenia patients
(Yao et al., 2010; Hosak, 2013). The actual schizophrenia risk
will likely be governed by a combination of alleles of small
effect size, rare alleles, copy number variations, and
epigenetic effects. Complex relationship between genotype
and phenotype leading to different clinical presentations in
patient sub-cohorts has to be tested, from the genetic variation
level to the underlying biologic etiology.
Using downstream products of gene expression, omics
methods (transcriptomics, proteomics and metabolomics)
facilitate the search for plausible disease risk pathways
(Allen et al., 2011; Arnold et al., 2015; Botas et al., 2015). A
recent metabolomics study (He et al., 2012) demonstrated
differences in the amino acid and lipid metabolism in
medicated and non-medicated schizophrenia patients when
compared to the control group. Subsequent network analysis
of these potentially relevant metabolites and known schizo-
phrenia risk genes identied glutamine and arginine signaling
pathways as possible risk factors. Another study (Orešičet al.,
2011) raised a possibility that there are at least two different
schizophrenia related risk pathways, glucoregulatory and
proline metabolism.
Amino acids
In addition to glutamine and arginine metabolism, amino
acids altered in plasma or cerebrospinal liquid of schizo-
phrenia patients are also involved in nitrogen compound
biosynthetic processes. Removal of the α-amino group is the
rst step in amino acid catabolism, a process essential for the
energy production. Removed nitrogen can be excreted as urea
or incorporated into other compounds during the anabolic
processes. Amonia and asparatate are two sources of urea
nitrogen provided by transamination and oxidative deamina-
tion.
Arginine is the essential element in the urea cycle. It is
produced by various tissues, but hydrolyzed into urea and
ornithine exclusively by the liver. Decreased arginine found
in schizophrenia could indicate an increase in ammonia and/
or nitric oxide (NO) metabolism. Monoamines are one of the
sources of ammonia. In the brain, monoamines serve as
hormones or neurotransmitters. Altered cerebrospinal uid
monoamine turnover rates and ratios were implicated in
genetics of psychosis and schizophrenia (Andreou et al.,
2014; Luykx et al., 2014). NO is CNS neurotransmitter. It is
also involved in the storage, uptake, and release of other
neurotransmitters and oxidative stress. Arginine is one of
three substrates for NO synthesis. It is plausible that an
increase in arginine consumption may be related to aberrant
nitric oxide metabolism in schizophrenia (Yanik et al., 2003;
Bernstein et al., 2005; He et al., 2012; Weber et al., 2014)
Data on tryptophan (Trp) metabolites in schizophrenia are
inconsistent, possibly due to patient cohort characteristics
(rst episode vs chronic illness) or the source of biologic
sample (blood or cerebrospinal uid). Trp is metabolized to 5-
hydroxytryptamine (5-HT or serotonine) by tryptophan
hydrolase (TH) and aromatic amino acid decarboxylase
(AAD) in the serotonin pathway. 5-HT is further metabolized
to melatonin, which possesses anti-oxidant activity (Reiter et
al., 2008). Changes in Trp could reect a compensatory
mechanism due to increased oxidative stress in schizophrenia.
Work in the rst episode patients (Yao et al., 2010) revealed
increased conversion of serotonin to N-acetylserotonin,
possibly driving a drop in serum tryptophan levels. Reduction
in tryptophan serum availability was also detected in
treatment naïve patients (Manowitz et al., 1973; Xuan et al.,
2011). In contrast, medicated schizophrenia cohorts demon-
strate either increase (Fukushima et al., 2014) or decrease
(Manowitz et al., 1973) in Trp and decrease in 5-
hydroxyindoleacetic acid (5HIAA) level in plasma (Fukush-
ima et al., 2014) or cerebrospinal uid (Ashcroft et al., 1966).
Earlier investigations suggest that hyperserotonemia may
play an etiological role in some forms of schizophrenia
(Garelis et al., 1975; Jackman et al., 1983; Stahl et al., 1983).
Trp is metabolized to Kyn by tryptophan 2,3-dioxygenase or
indoleamine 2,3-dioxygenase in the Kyn pathway (Olney and
Farber, 1995; Schwarcz et al., 2001; Fukushima et al., 2014).
The kynurenine pathway produces neurotoxic (3-hydroxy-
kynurenine and quinolinic acid) and neuro-inhibitory
(kynurenic acid) compounds. At high concentrations,
kynurenic acid is a competitive antagonist of the glycine
site of N-methyl-D-asparate (NMDA) receptors (Stone, 1993)
and a noncompetitive antagonist of the α-7-nicotinic
acetylcholine receptor at a low concentration (Hilmas et al.,
Iva Petrovchich et al. 223
2001; Alkondon et al., 2004). Thus, increased levels of
kynurenic acid may be associated with a spatial working
memory dysfunction (Alkondon et al., 2004) though NMDA
receptor action. Schwarcz et al. (2001) have shown increased
cortical levels of kynurenic acid in schizophrenia, which may
be related to cognitive impairment. (Yao et al., 2010).
Increases in kynurenic acid levels were also reported in the
post-mortem brain tissue (Schwarcz et al., 2001) or
cerebrospinal uids (Erhardt et al., 2001) from schizophrenia
patients. However, patients serum kynurenic acid levels
remained unaltered, indicating that increased kynurenic acid
levels might occur only locally in the brain (Fukushima et al.,
2014).
Glutamine is a precursor of g-GluCys. Lower concentra-
tion of glutamine was detected in plasma and elevated
glutamate in serum and cortex of patients (He et al., 2012;
Fukushima et al., 2014). Interestingly, amonia obtained by
removal of alpha amino acid amino group is combined with
glutamate to form glutamine, which is transported to the liver
to be catabolized to glutamate and ammonia. Additionally,
glutamine is an important building block for purine and
pirimidine synthesis. The glutamate/glutamine hypothesis of
schizophrenia and associated brain metabolic abnormalities in
patients and their rst degree relatives is well documented
(Tandon et al., 2013). A single-nucleotide polymorphism
(SNP) in glutamine-dependent carbamoyl-phosphate
synthase enzyme involved in glutamine hydrolysis is possibly
linked to schizophrenia
,
(Stone et al., 2007; He et al., 2012;
Fukushima et al., 2014). As evidenced by genetic studies, a
broader metabolic abnormality might exist in schizophrenia.
Comorbid type 2 diabetes mellitus, cardiac autonomic
dysregulation, and numerous autoimmune disorders have
the strongest familiar predisposition in schizophrenia (Fer-
entinos and Dikeos, 2012).
Serum glutamate (Glu) is elevated in all psychoses
compared to controls. Glutamate is a precursor of ammonia
and aspartate nitrogen in the urea cycle. Consequently,
glutamate-related metabolic abnormalities may reect a
common, amino-acid pathway abnormality, across different
psychoses types (Cherlyn et al., 2010). Serum Glu level is
reported to be elevated in treatment resistant schizophrenia, in
concert with other amino acid abnormalities, which further
supports amino-acid pathway aberrance in schizophrenia
(Tortorella et al., 2001).
Upregulation of serum proline was reported in schizo-
phrenia. There is evidence that polymorphisms in the
PRODH gene encoding proline oxidase are associated with
schizophrenia risk (Liu et al., 2002; Kempf et al., 2008) and
that the related hyperprolinemia is negatively associated with
cognitive performance (Orešičet al., 2011).
Low concentrations of histidine in whole blood and high
levels in cerebrospinal uid were identied in schizophrenic
patients (He et al., 2012). Histidine is the histamine precursor.
Histamine is an ubiquitous neurotransmitter but also a
chemical messenger modulating allergic and inammatory
reactions (He et al., 2012), postulate that downregulation of
TCF4, a transcription factor crucially involved in fetal brain
development, could probably release its inhibition on
histidine decarboxylase, leading to accelerated histidine
decarboxylation in schizophrenia (Prell et al., 1995; Fernán-
dez-Novoa and Cacabelos, 2001; He et al., 2012).
Ornithine is another amino acid implicated in schizo-
phrenia, due to low plasma levels found in patients. Urea is a
major disposal product derived of amino acids amino groups.
Ornithine is synthetized in the cytoplasm of liver cells and
transported to mitochondria, where it participates in the nal
stages of urea formation. Increased plasma ornithine implies
suppression in the ornithine decarboxylation process (Mid-
dleton et al., 2002; He et al., 2012) as a possible aberrant
mechanism in schizophrenia patients. Arginine, glutamine,
histidine, and ornithine metabolic pathways were associated
with schizophrenia genetic susceptibility, based on metabo-
lomic data and schizophrenia risk genes molecular network
analyses (He et al., 2012).
Lipids, fatty acids and amino acids
associated with phospholipid synthesis
Decreased phosphatidylcholines (PC ae C38:6) in the plasma
of schizophrenia patients (He et al., 2012) could imply
aberrations in choline metabolism, immune-related signaling,
or neurotrophin signaling. Choline is a precursor and
metabolite of the neurotransmitter acetylcholine and an
essential component of neuronal membrane phospholipids.
Phosphatidylcholine serves as substrate for phosphatidylser-
ine synthesis, a major phospholipid located in the inner
neuronal membrane. An increase in phospholipase A2
activity in schizophrenia (Gattaz et al., 1987; Gattaz et al.,
1990) could implicate acceleration in the breakdown of
membrane phospholipids in schizophrenia. Alternatively, an
increase in phosphatidylcholine demand would lead to its
depletion. The increase in phosphatidylcholine synthesis is a
common feature of neuronal differentiation, with nerve
growth factor directly modulating this process through
immediate early genes and the ERK 1/2 pathway (Paoletti
et al., 2011). Outlined ndings are consistent with demon-
strated cell membrane abnormalities associated with dis-
ordered phospholipids composition and metabolism (He et
al., 2012).
Saturated triglycerides, small-molecular clusters contain-
ing branched chain amino acids, phenylalanine and tyrosine,
and proline, glutamic (Fukushima et al., 2014), lactic and
pyruvic acids were increased in schizophrenia serum samples,
suggesting possible relevance between glucoregulatory path-
ways, proline metabolism, and schizophrenia (He et al.,
2012). A global metabolomic analysis approach coupled with
the network analysis identied the lipid cluster LC9,
containing saturated and longer chain triglycerides, as the
strongest link to schizophrenia (Orešičet al., 2011). In this
study, schizophrenia patients were also insulin resistant with
224 Metabolomics in Schizophrenia and Major Depressive Disorder
elevated fasting serum insulin levels, which calls for a
replication study prodrome or rst episode schizophrenia
patients. A separate lipidomic study (Kotronen et al., 2009)
revealed that the lipids found in LC9 are associated with
insulin resistance. Together, these ndings imply that
schizophrenia, independent of antipsychotic medication and
metabolic co-morbidity, is characterized by insulin resistance,
enhanced hepatic very low density lipoprotein production,
(Kotronen and Yki-Järvinen, 2008) and thus elevated serum
concentrations of specic triglycerides (Orešičet al., 2011).
Consequently, it is not surprising that fatty acids and ketone
bodies were found to be signicantly elevated in both the
serum and urine sample of the patients, as demonstrated by
metabolic proling (Yang et al., 2013). These metabolic
ndings could be interpreted as upregulated fatty acid
catabolism as a result of insufcient brain of glucose supply
(Orešičet al., 2011; He et al., 2012).
Liver mitochondria have a capacity to convert acetyl CoA
derived from fatty acid oxygenation into ketone bodies,
including 3-hydroxybutyrate. Ketone bodies are used by
peripheral tissue as an energy source. Data imply there might
be a subset of schizophrenia patients characterized by
downregulated fatty acid metabolism, including a decrease
in 3-hydroxybutyrate (Yang et al., 2013; Fukushima et al.,
2014). 3-hydroxybutyrate also plays a role as the inhibitor of
class I histone deacetylases (HDACs). Consequently, a
decrease in 3-hydroxybutyrate would downregulate histone
acetylation and change DNA transcription, potentially
leaving the cell more vulnerable to the oxidative stress
(Bitanihirwe and Woo, 2011; Shimazu et al., 2013).
Decreases in linoleic and arachidonic acids (Ramos-Loyo
et al., 2013; Fukushima et al., 2014) were identied in some
schizophrenia patients. Linoleic acid (LA) is a precursor of w
arachidonic acid (AA) and αlinoleic acid, which is
metabolized to w-3 fatty acids, essential in growth and
development. Arachidonic acid belongs to long-chain poly-
unsaturated fatty acids (LCPUFA), with roles in neuronal
development and neurodegeneration. LCPUFA deciency is
associated with schizophrenia and attention decit hyper-
activity disorder (ADHD). In biological uids, LA and AA
are bound to fatty acid binding protein (FABP), which
governs the sequesteration of circulating PUFAs. Increase in
tissue expression of FABP is closely associated with the
etiology of schizophrenia (Maekawa et al., 2011), high-
lighting one of the possible biological mechanisms poten-
tially responsible for decrease in LA and AA found in
schizophrenia.
Glutathione (GSH) can chemically detoxify hydrogen
peroxide (H
2
O
2
). H
2
O
2
is a by-product of anaerobic
metabolism, with the potential to cause serious damage to
DNA, proteins, or unsaturated lipids which can lead to cell
death. Decreases in the GSH level of some schizophrenia
patients (Raffa et al., 2009; Fukushima et al., 2014) indicate
schizophreniaconsistent with the notion of free radical
mediated neurotoxicity in schizophrenia (Yao et al., 2010). g-
glutamylcysteine (g-GluCys) is a precursor of an endogenous
antioxidant, glutathione (GSH), produced from Glu and Cys
by glutamatecysteine ligase (GCL). Interstingly, GCL activity
is a rate-limiting enzyme for GSH synthesis, and genetic
polymorphisms in GCL signicantly modulate schizophrenia
risk (Gysin et al., 2007; Nichenametla et al., 2008; Fukushima
et al., 2014).
Serine is a substrate for phosphatidylserine synthesis,
required for the membrane production. Interestingly, while
low levels of serine are found in the peripheral blood of
schizophrenia subjects (Hashimoto et al., 2003; Fukushima et
al., 2014), the prefrontal cortex is reported to have high level
of phosphatidylserine (Wood and Holderman, 2015). Conse-
quently, a dysfunction of oligodendrocyte glycosynapses in
the schizophrenia brain could be implicated in peripheral D-
serine depletion. D-serine is also a co-agonist of the glycine
site of the NMDA receptor. The glutamate hypothesis of
schizophrenia etiology suggests that endogenous D-serine is a
crucial factor related to the hypofunction of the N-methyl-D-
aspartate (NMDA) receptor (Schell et al., 1997; Hashimoto et
al., 2003). Interestingly, genetic studies identify D-amino acid
oxidase (DAO), a molecule highly expressed in the brain
where it oxidizes d-serine, as a possible schizophrenia risk
(Chumakov et al., 2002; Madeira et al., 2008).
Glucose and lactate
Schizophrenic patients have higher baseline serum levels of
glucose and lactate (Xuan et al., 2011). Linkage studies in
schizophrenia highlight the possible importance of genes
related to glycolysis (Stone et al., 2004). Lactate is a nal
product of the anaerobic glycolysis in the cells. It signals
disordered energy homeostasis: lactate correlates with obesity
and type 2 diabetes and modulates diastolic blood pressure.
Importantly, stem cell proliferation is ensured through the
anabolic state of glycolysis. In contrast, differentiation to
adult cells is governed by mitochondrial oxidative metabo-
lism. High levels of lactate decrease neurogenesis in animal
models, likely through excessive inammatory response
(Inoue et al., 2015). Similarly, schizophrenia metabolic shift
favoring an increase in lactate level could lead to neuronal
loss.
Major Depressive Disorder
Major Depressive Disorder is a common psychiatric illness. It
is estimated that 10%15%of people will experience Major
Depressive Disorder during their lifetime. Depression is
predicted to become the second most common world health
problem by 2020. It has relatively low heritability (~0.4) and
is clinically heterogeneous. Treatment success is highly
variable; less than 50%of patients respond to any given
Iva Petrovchich et al. 225
antidepressant. Such poor response reects our lack of
knowledge of depression biomarkers and their poor clinical
characterization. Genetic loci identied by biologic candidate
gene studies were not supported by genome-wide studies
(GWAS). Failure to identify loci associated with depression
from large-scale unbiased GWAS is in part due to a need for a
bigger sample size (75 000100 000) in order to reach
comparable power to identify risk loci as has been done in
schizophrenia genome-wide studies (Smoller, 2016). Lack of
homogeneity in clinical cohorts is an additional signicant
challenge.
Research on gene-environment interactions (GE) is
growing: there is a strong correlation between stressful life
events and depression risk calling for quantication of the
stress diathesis model. The allostatic load (AL) is an
interdisciplinary approach aimed at quantifying chronic stress
in relation to various life pathologies. Allostatic load
encompasses the denition of stress as a multidimensional
model. Integral parts of this model are genetic and epigenetic
factors, early adversities that shape brain development, and
hypothalamo-pituitary-adrenal axis responses. These factors
include moderation or mediation by environmental toxins,
distinctions among socioeconomic status, sex, gonadal
hormones and their effects on the brain, and endocrine,
metabolic and immune systems. Consistent with AL hypoth-
esis, data indicate that while some Major Depressive Disorder
patients have immune related dysfunctions leading to
depression, others have energy metabolism impairment,
which could be a gate to a disorder (Martins-de-Souza,
2014). Metabolomic analysis ndings might improve strati-
cation of heterogeneous Major Depressive Disorder cohorts
and facilitate G E studies. Currently, stratication is based
on so called depressive subtypes (by sex, recurrent depression
or recurrent vs. early-onset).
Fatty acid and lipid metabolism
β-oxidation is the major fatty acids catabolic pathway, taking
place in mitochondia. Carnitine is a molecule essential for the
carnitine shufe,a process during which a long chain fatty
acyl group is transported across the inner mitochondrial
membrane. Recent metabolomics analysis (Liu et al., 2015)
identied decrease in various plasma acyl carnitine molecules
(carnitine C10:1, carnitine C10:2, carnitine C14:2, carnitine
C14:3, carnitine C6:0, carnitine C8:0, carnitine C8:1,
carnitine C10:0, carnitine C12:1, carnitine C3:0) as possible
Major Depressive Disorder biomarker set. Consistent with
this nding, there is evidence that Acetyl-l-carnitine may be
effective in depression, as adjunct treatment in Major
Depressive Disorder or monotherapy in Disthymia (Wang et
al., 2014).
Decreased stearic amide and palmitic amide (Liu et al.,
2015) levels were reported in plasma of some Major
Depressive Disorder patients. Palmitic acid has 16 carbons
(16:0) and stearic 18 (18:0). In animal models, palmitic acid
induces anxiety like behaviors (Moon et al., 2014). Interest-
ingly, running, which has anxiolytic effect in animal models,
decreases palmitic acid concentration in the cortex (Santos-
Soto et al., 2013). Palmitic acid was shown to reduces
neuronal progenitor cells proliferation (Park et al., 2011)
while, stearic acid seems to have neuroprotective effects
(Wang et al., 2006).
A variety of lysophospholipids, monoglycerophospholi-
pids, and phosphatidylethanolamines (Liu et al., 2015) were
reported as increased in plasma of Major Depressive Disorder
patients (16:1 sn-1, LPC 16:1 sn-2, LPE 16:0 sn-1, LPE 16:0
sn-2, LPE 16:1 sn-1, LPE 18:1 sn-2, LPE 22:5 sn-1, LPC 16:0
sn-2d, LPC 18:1 sn-1, LPC 18:1 sn-2, LPC 20:1 sn-2, LPC
22:4 sn-1, LPC 22:4 sn-2, LPC P-16:0, LPE 16:1 sn-2, LPE
20:3 sn-2, LPE 22:5 sn-2, LPE 18:0 sn-1, LPE 18:0 sn-2, LPE
18:2 sn-1, LPE 18:2 sn-2, LPE 20:3 sn-1, PC 32:0, PC 32:1,
PC O 36:2, PE 34:2, PE 36:4, PE O 36:6, PE O 34:3, PE O
36:5, PE O 38:7), accompanied with a concomitant decrease
in the free fatty acids (FFA 16:2d). Lysophospholipids,
monoglycerophospholipids, and phosphatidylethanolamines
participate in fuel and energy storage, cell signaling, and
ensuring membrane integrity and stability. On the cellular
level, the lysophospholipatidic acid receptor modulates
survival and apoptosis and serves as a novel cell survival
and apoptotic factor (Santin et al., 2009). Antidepressant
treatment was shown to increase the release of lysopho-
spholipids in the cortex of mice (Lee et al., 2009).
Decreased plasma levels of lithocholic, deoxycholic,
glycodesoxycholic, glycoursodeoxycholic and taurocheno-
deoxycholic acid (Liu et al., 2015) suggest possible
dysregulation of the bile acid metabolism in Major Depres-
sive Disorder. Glycoursodeoxycholic and taurochenodeoxy-
cholic acid were shown to have neuroprotective roles (Vaz et
al., 2015; Nunes et al., 2012).
Amino acid and glucose metabolites
Serum glutamate (Glu) and asparate levels are elevated in
Major Depressive Disorder compared to controls (Liu et al.,
2015). As outlined in Schizophrenia section, glutamate is a
precursor of ammonia and aspartate nitrogen in the urea
cycle, which occurs exclusively in the liver, and changes in its
concentration imply amino-acid pathway abnormality. Serum
increases in glutamate and aspartate levels could also reect a
global dysfunction in NMDA receptor function. Ketamine is
an NMDA antagonist, blocking glutamate neuronal actions.
Rapid antidepressant actions of ketamine underscore clinical
applicability of the glutamate Major Depressive Disorder
hypothesis for at least a subpopulation of patients.
Higher levels of alanine, taurine, citrate, formate, glycine,
isobutyrate, and nicotinate were identied in the urine of
226 Metabolomics in Schizophrenia and Major Depressive Disorder
medication naïve, rst episode Major Depressive Disorder
patients (Zheng et al., 2013). Alanine is a key gluconeogen-
esis amino acid. High levels of alanine in combination with
low brain glucose might imply inefcient gluconeogenesis
process. Taurine is a major constituent of bile. In addition to
bile acid conjugation, taurine fosters proliferation of human
neural stem/progenitor neural cells during fetal brain devel-
opment (Hernández-Benítez et al., 2013). Citrate could be
viewed as a high energy signal. It enables the transfer of
acetate units from mitochondria to cytosol, initiating de novo
fatty acid synthesis. Increases in citrate, in conjunction with
high levels of alanine and low glucose, highlights possible
gluconeogenesis inefciency and a shift in the metabolic
balance toward fatty acid synthesis. Deceased levels of
plasma glucose, lactate, and pyruvate (Zheng et al., 2012)
further support putative imbalance between glycolysis and
gluconeogenesis energy cycles in Major Depressive Disorder.
In addition to being building blocks for proteins, amino
acids are precursors of nitrogen-containing molecules with
important biochemical functions: hormones, neurotransmit-
ters, pyrines, and pyrimidines. The serotonergic system is one
of the key neurotransmitter systems implicated in Major
Depressive Disorder pathophysiology. Seratonin production
is highly dependent on plasma tryptophan utilization. A
decrease in tryptophan levels directly affects the brain
serotonergic system, and metabolomics data reafrm the
tryptophan depletion hypothesis of depression (Moreno et al.,
2010; Martins-de-Souza, 2014; Liu et al., 2015). Tyrosine is a
building block for cateholamines, dopamine, norepinephrine,
and epinephrine. A decrease in tyrosine (Martins-de-Souza,
2014; Liu et al., 2015) could imply increased conversion to
cateholamines or decit in protein catabolism. Cerebrospinal
uid shows reductions in metabolites associated with
tryptophan and tyrosine pathways in remitted depression
(Kaddurah-Daouk et al., 2012). Tryptophan, tyrosine and
methionine pathways might reveal differences between
remitted and non-remitted Major Depressive Disorder sub-
jects (Kaddurah-Daouk et al., 2012).
The tricarboxylic acid (TCA) cycle (also known as the
Krebs cycle) presents the nal pathway for the oxidative
catabolism of carbohydrates, amino acids, and fatty acids.
Depressed patientsurine (Zheng et al., 2013) was shown to
have a decrease in the molecules feeding into the TCA cycle
(glucose and piruvate) and an increase in TCA cycle
associated metabolites (α-ketoglutarate, succinate, malonate,
methylmalonate, and succinyl-CoA). This nding conrms
the postulated compromise of glucose metabolism and a shift
toward fatty acid and/or amino acid catatabolism with the aim
of meeting cell energetic and anabolic needs.
Decreases (Liu et al., 2015) or increases (Woo et al., 2015)
in methionine were reported in the plasma of Major
Depressive Disorder patients. Methionine derivative is S-
adenosyl methionine (SAM), a molecule fundamental to
DNA methylation (Cantoni et al., 1989; Kaddurah-Daouk et
al., 2012).
Other metabolites and pathways
Serotonin, dopamine, and norepinephrine metabolites (5-
hydroxyindoleacetic acid [5-HIAA], and homovanillic acid
[HVA]) were decreased in CSF of patients with melancholic
depression (Asberg et al., 1984) implying altered serotonine
and dopamine brain function, consistent with monoaminergis
hypothesis of depression.
Decreased N-methylnicotinamide (NMNA) (Zheng et al.,
2013). N-methylnicotinamide is a metabolite of niacin (or
nicotinamide) and a decrease in urine concentration could
imply niacin deciency. Biologicaly active coenzyme forms
are nicotinamid adenine dinucleotide (NAD
+
) and its
phosphorylated form, NADP
+
. Both molecules serve as
coenzymes in oxidation-reduction reactions, which could be
compromised in depression.
Conclusion
The Omicssystems biology approach opens a new venue
for the quest to identify diagnostic and treatment outcome
biomarkers in psychiatry. Unbiased genome-wide studies and
pathway analysis hold a promise to identify reproducible
disease-risk gene associations. Complex challenges persist,
such as disease heterogeneity, polygenicity, and low pene-
trance of most genetic variants, to name a few. Omicstools
have a potential to facilitate dening intermediate disease
phenotypes (Maletic-Savatic et al., 2008; Vingara et al., 2013;
Peterson et al., 2013). Additionally, in vivoor in vitro
metabolomics may provide valuable insight about GE
interactions.
Recently, genetics studies discovered a possible role of
mobile DNA transposomes in the brain cellular heterogeneity
(Singer et al., 2010; Evrony et al., 2015). DNA transposomes
are transposable autonomous elements, scattered throughout
the genome and inherited from one generation to the next.
These DNA elements are active and capable of jumping
during neuronal differentiation, altering individual cell
transcriptome and metabolome proles and thereby modulat-
ing functional output through G E interactions. The human
hippocampus has an astonishing 13.7 somatic insertions per
neuron (Upton et al., 2015). Bundo et al. (2014) found that
transposomes may play a role in psychiatric diseases-
increases in the copy numbers were evident in cortical
neurons of schizophrenia patients and had a positive trend in
mood disorders. Moreover, induced pluripitent cell-derived
neurons from patients with schizophrenia with 22q11 deletion
also had an increase in transposome copy numbers. Human
brain somatic transposition was shown to inuence the
biosynthesis of more than 250 metabolites (Arbusán, 2012),
including those linked to schizophrenia.
Hippocampal and/or cortical neuronal mosaicism may be
evolutionary imprinted to ensure genomic diversity and
greater adaptability to the environment (Glinsky, 2015). It
Iva Petrovchich et al. 227
could also pose a psychiatric risk and contribute to disease
heterogeneity. It is in this context that in vivo(brain) and in
vitro(blood) metabolomics proling in patient populations
could help elucidate G x E interactions and offer an additional
tool set for much needed biomarker discovery.
Acknowledgements
This work was supported by Brain and Behavior Research Foundation
2012 Young Investigator Award (19722) to Dr. S. Milanovic.
Compliance with ethics guidelines
Iva Petrovchich, Alexandra Sosinsky, Anish Konde4, Abigail Archibald,
David Henderson, Mirjana Maletic-Savatic and Snezana Milanovic
declare that they have no conict of interest. This manuscript is a review
article and does not involve a research protocol requiring approval by the
relevant institutional review board or ethics committee.
References
Abrusán G (2012). Somatic transposition in the brain has the potential to
inuence the biosynthesis of metabolites involved in Parkinsons
disease and schizophrenia. Biol Direct, 7(1): 41
Alkondon M, Pereira E F, Yu P, Arruda E Z, Almeida L E, Guidetti P,
Fawcett W P, Sapko M T, Randall W R, Schwarcz R, Tagle D A,
Albuquerque E X (2004). Targeted deletion of the kynurenine
aminotransferase ii gene reveals a critical role of endogenous
kynurenic acid in the regulation of synaptic transmission via α7
nicotinic receptors in the hippocampus. J Neurosci, 24(19): 4635
4648
Allen G I, Maletić-SavatićM (2011). Sparse non-negative generalized
PCA with applications to metabolomics. Bioinformatics, 27 (21):
30293035
Andreou D, Söderman E, Axelsson T, Sedvall G C, Terenius L, Agartz I,
Jönsson E G (2014). Polymorphisms in genes implicated in
dopamine, serotonin and noradrenalin metabolism suggest associa-
tion with cerebrospinal uid monoamine metabolite concentrations in
psychosis. Behav Brain Funct, 10(1): 26
Appleton K M, Rogers P J, Ness A R (2008). Is there a role for n-3 long-
chain polyunsaturated fatty acids in the regulation of mood and
behaviour? A review of the evidence to date from epidemiological
studies, clinical studies and intervention trials. Nutr Res Rev, 21(1):
1341
Arai M, Yuzawa H, Nohara I, Ohnishi T, Obata N, Iwayama Y, Haga S,
Toyota T, Ujike H, Arai M, Ichikawa T, Nishida A, Tanaka Y,
Furukawa A, Aikawa Y, Kuroda O, Niizato K, Izawa R, Nakamura
K, Mori N, Matsuzawa D, Hashimoto K, Iyo M, Sora I, Matsushita
M, Okazaki Y, Yoshikawa T, Miyata T, Itokawa M (2010). Enhanced
carbonyl stress in a subpopulation of schizophrenia. Arch Gen
Psychiatry, 67(6): 589597
Arnold J M, Choi W T, Sreekumar A, Maletić-SavatićM(2015).
Analytical strategies for studying stem cell metabolism, Front Biol,
10 (2): 141153
Asberg M, Bertilsson L, Mårtensson B, Scalia-Tomba G P, Thorén P,
Träskman-Bendz L (1984). CSF monoamine metabolites in mel-
ancholia. ActaPsychiatrScand, 69(3): 201219
Ashcroft G W, Crawford T B, Eccleston D, Sharman D F, MacDougall E
J, Stanton J B, Binns J K (1966). 5-hydroxyindole compounds in the
cerebrospinal uid of patients with psychiatric or neurological
diseases. Lancet, 2(7472): 10491052
Bernstein H G, Bogerts B, Keilhoff G (2005). The many faces of nitric
oxide in schizophrenia. A review. Schizophr Res, 78(1): 6986
Bitanihirwe B K, Woo T U (2011). Oxidative stress in schizophrenia: an
integrated approach. NeurosciBiobehav Rev, 35(3): 878893
Botas A, Campbell H M,Han X , Maletic-Savatic M(2015). Metabo-
lomics of neurodegenerative diseases, Int Rev Neurobiol, 122: 5380
Bowers M BJr (1973). 5-Hydroxyindoleacetic acid (5HIAA) and
homovanillic acid (HVA) following probenecid in acute psychotic
patients treated with phenothiazines. Psychopharmacologia, 28(4):
309318
Bundo M, Toyoshima M, Okada Y, Akamatsu W, Ueda J, Nemoto-
Miyauchi T, Sunaga F, Toritsuka M, Ikawa D, Kakita A, Kato M,
Kasai K, Kishimoto T, Nawa H, Okano H, Yoshikawa T, Kato T,
Iwamoto K (2014). Increased l1 retrotransposition in the neuronal
genome in schizophrenia. Neuron, 81(2): 306313
Cantoni G L, Mudd S H, Andreoli V (1989). Affective disorders and S-
adenosylmethionine: a new hypothesis. Trends Neurosci, 12(9):
319324
Capuron L, Neurauter G, Musselman D L, Lawson D H, Nemeroff C B,
Fuchs D, Miller A H (2003). Interferon-alpha-induced changes in
tryptophan metabolism. relationship to depression and paroxetine
treatment. Biol Psychiatry, 54(9): 906914
Cherlyn S Y, Woon P S, Liu J J, Ong W Y, Tsai G C, Sim K (2010).
Genetic association studies of glutamate, GABA and related genes in
schizophrenia and bipolar disorder: a decade of advance. Neurosci
Biobehav Rev, 34(6): 958977
Chumakov I, Blumenfeld M, Guerassimenko O, Cavarec L, Palicio M,
Abderrahim H, Bougueleret L, Barry C, Tanaka H, La Rosa P, Puech
A, Tahri N, Cohen-Akenine A, Delabrosse S, Lissarrague S, Picard F
P, Maurice K, Essioux L, Millasseau P, Grel P, Debailleul V, Simon
A M, Caterina D, Dufaure I, Malekzadeh K, Belova M, Luan J J,
Bouillot M, Sambucy J L, Primas G, Saumier M, Boubkiri N, Martin-
Saumier S, Nasroune M, Peixoto H, Delaye A, Pinchot V, Bastucci
M, Guillou S, Chevillon M, Sainz-Fuertes R, Meguenni S, Aurich-
Costa J, Cherif D, Gimalac A, Van Duijn C, Gauvreau D, Ouellette G,
Fortier I, Raelson J, Sherbatich T, Riazanskaia N, Rogaev E,
Raeymaekers P, Aerssens J, Konings F, Luyten W, Macciardi F,
Sham P C, Straub R E, Weinberger D R, Cohen N, Cohen D (2002).
Genetic and physiological data implicating the new human gene G72
and the gene for D-amino acid oxidase in schizophrenia. Proc Natl
Acad Sci U S A, 99(21): 1367513680
Craft S, Watson G S (2004). Insulin and neurodegenerative disease:
shared and specicmechanisms. Lancet Neurol, 3(3): 169178
Domino E F, Krause R R (1974). Free and bound serum tryptophan in
drug-free normal controls and chronic schizophrenic patients. Biol
Psychiatry, 8(3): 265279
Erhardt S, Blennow K, Nordin C, Skogh E, Lindström L H, Engberg G
(2001). Kynurenic acid levels are elevated in the cerebrospinal uid
of patients with schizophrenia. Neurosci Lett, 313(1-2): 9698
Evrony G D, Lee E, Mehta B K, Benjamini Y, Johnson R M, Cai X,
Yang L, Haseley P, Lehmann H S, Park P J, Walsh C A (2015). Cell
lineage analysis in human brain using endogenous retroelements.
228 Metabolomics in Schizophrenia and Major Depressive Disorder
Neuron, 85(1): 4959
Ferentinos P, Dikeos D (2012). Genetic correlates of medical
comorbidity associated with schizophrenia and treatment with
antipsychotics. Curr Opin Psychiatry, 25(5): 381390
Fernández-Novoa L, Cacabelos R (2001). Histamine function in brain
disorders. Behav Brain Res, 124(2): 213233
Fukushima T, Iizuka H, Yokota A, Suzuki T, Ohno C, Kono Y,
Nishikiori M, Seki A, Ichiba H, Watanabe Y, Hongo S, Utsunomiya
M, Nakatani M, Sadamoto K, Yoshio T (2014). Quantitative analyses
of schizophrenia-associated metabolites in serum: serum D-lactate
levels are negatively correlated with gamma-glutamylcysteine in
medicated schizophrenia patients. PLoS One, 9(7): e101652
Garelis E, Gillin J C, Wyatt R J, Neff N (1975). Elevated blood serotonin
concentration in unmedicated chronic schizophrenic patients. Am J
Psychiatry, 132(2): 184186
Gattaz W F, Brunner J, Schmitt A, Maras A (1994). Accelerated
breakdown of membrane phospholipids in schizophreniaimplica-
tions for the hypofrontality hypothesis. Fortschr Neurol Psychiatr, 62
(12): 489496
Gattaz W F, Hübner C V, Nevalainen T J, Thuren T, Kinnunen P K
(1990). Increased serum phospholipase A2 activity in schizophrenia:
a replication study. Biol Psychiatry, 28(6): 495501
Gattaz W F, Köllisch M, Thuren T, Virtanen J A, Kinnunen P K J (1987).
Increased plasma phospholipase-A2 activity in schizophrenic
patients: reduction after neuroleptic therapy. Biol Psychiatry, 22
(4): 421426
Gillin J C, Kaplan J A, Wyatt R J (1976). Clinical effects of tryptophan in
chronic schizophrenic patients. Biol Psychiatry, 11(5): 635639
Glinsky G V (2015). Transposable elements and DNA methylation
create in embryonic stem cells human-specic regulatory sequences
associated with distal enhancers and noncoding RNAs. Genome Biol
Evol, 7(6): 14321454
Gysin R, Kraftsik R, Sandell J, Bovet P, Chappuis C, Conus P, Deppen
P, Preisig M, Ruiz V, Steullet P, Tosic M, Werge T, Cuénod M, Do K
Q (2007). Impaired glutathione synthesis in schizophrenia: con-
vergent genetic and functional evidence. Proc Natl Acad Sci U S A,
104(42): 1662116626
Hashimoto K, Fukushima T, Shimizu E, Komatsu N, Watanabe H,
Shinoda N, Nakazato M, Kumakiri C, Okada S, Hasegawa H, Imai K,
Iyo M (2003). Decreased serum levels of D-serine in patients with
schizophrenia: evidence in support of the N-methyl-D-aspartate
receptor hypofunction hypothesis of schizophrenia. Arch Gen
Psychiatry, 60(6): 572576
Hashimoto K, Shimizu E, Iyo M (2005). Dysfunction of glia-neuron
communication in pathophysiology of schizophrenia. Curr Psychia-
try Rev, 1(2): 151163
He Y, Yu Z, Giegling I, Xie L, Hartmann A M, Prehn C, Adamski J,
Kahn R, Li Y, Illig T, Wang-Sattler R, Rujescu D (2012).
Schizophrenia shows a unique metabolomics signature in plasma.
Transl Psychiatry, 2(8): e149
Hernández-Benítez R, Vangipuram S D, Ramos-Mandujano G, Lyman
W D, Pasantes-Morales H (2013). Taurine enhances the growth of
neural precursors derived from fetal human brain and promotes
neuronal specication. Dev Neurosci, 35(1): 4049
Hilmas C, Pereira E F, Alkondon M, Rassoulpour A, Schwarcz R,
Albuquerque E X (2001). The brain metabolite kynurenic acid
inhibits α7 nicotinic receptor activity and increases non-α7 nicotinic
receptor expression: physiopathological implications. J Neurosci, 21
(19): 74637473
Hosak L (2013). New ndings in the genetics of schizophrenia. World J
Psychiatry, 3(3): 5761
Inoue K, Okamoto M, Shibato J, Lee M C, Matsui T, Rakwal R, Soya H
(2015). Long-term mild, rather than intense, exercise enhances adult
hippocampal neurogenesis and greatly changes the transcriptomic
prole of the hippocampus. PLoS One, 10(6): e0128720
Iwayama Y, Hattori E, Maekawa M, Yamada K, Toyota T, Ohnishi T,
Iwata Y, Tsuchiya K J, Sugihara G, Kikuchi M, Hashimoto K, Iyo M,
Inada T, Kunugi H, Ozaki N, Iwata N, Nanko S, Iwamoto K, Okazaki
Y, Kato T, Yoshikawa T (2010). Association analyses between brain-
expressed fatty-acid binding protein (FABP) genes and schizophrenia
and bipolar disorder. Am J Med Genet B Neuropsychiatr Genet,
153B(2): 484493
Jackman H, Luchins D, Meltzer H Y (1983). Platelet serotonin levels in
schizophrenia: relationship to race and psychopathology. Biol
Psychiatry, 18(8): 887902
Joseph M H, Owen F, Baker H F, Bourne R C (1977). Platelet serotonin
concentration and monoamine oxidase activity in unmedicated
chronic schizophrenic and in schizoaffective patients. Psychol
Med, 7(1): 159162
Kaddurah-Daouk R, Yuan P, Boyle S H, Matson W, Wang Z, Zeng Z B,
Zhu H, Dougherty G G, Yao J K, Chen G, Guitart X, Carlson P J,
Neumeister A, Zarate C, Krishnan R R, Manji H K, Drevets W
(2012). Cerebrospinal uid metabolome in mood disorders-remission
state has a unique metabolic prole. Sci Rep, 2(667): 667
Kempf L, Nicodemus K K, Kolachana B, Vakkalanka R, Verchinski B A,
Egan M F, Straub R E, Mattay V A, Callicott J H, Weinberger D R,
Meyer-Lindenberg A (2008). Functional polymorphisms in PRODH
are associated with risk and protection for schizophrenia and fronto-
striatal structure and function. PLoS Genet, 4(11): e1000252
Kolakowska T, Molyneux S G (1987). Platelet serotonin concentration
in schizophrenic patients. Am J Psychiatry, 144(2): 232234
Kotronen A, Velagapudi V R, Yetukuri L, Westerbacka J, Bergholm R,
Ekroos K, Makkonen J, Taskinen M R, Oresic M, Yki-Järvinen H
(2009). Serum saturated fatty acids containing triacylglycerols are
better markers of insulin resistance than total serum triacylglycerol
concentrations. Diabetologia, 52(4): 684690
Kotronen A, Yki-Järvinen H (2008). Fatty liver: a novel component of
the metabolic syndrome. Arterioscler Thromb Vasc Biol, 28(1): 27
38
Lee L H, Shui G, Farooqui A A, Wenk M R, Tan C H, Ong W Y (2009).
Lipidomic analyses of the mouse brain after antidepressant treatment:
evidence for endogenous release of long-chain fatty acids? Int J
Neuropsychopharmacol, 12(7): 953964
Liu H, Heath S C, Sobin C, Roos J L, Galke B L, Blundell M L, Lenane
M, Robertson B, Wijsman E M, Rapoport J L, Gogos J A,
Karayiorgou M (2002). Genetic variation at the 22q11 PRODH2/
DGCR6 locus presents an unusual pattern and increases suscept-
ibility to schizophrenia. Proc Natl Acad Sci U S A, 99(6): 3717
3722
Liu X, Zheng P, Zhao X, Zhang Y, Hu C, Li J, Zhao J, Zhou J, Xie P, Xu
G (2015). Discovery and validation of plasma biomarkers for major
depressive disorder classication based on liquid chromatography-
mass spectrometry. J Proteome Res, 14(5): 23222330
Luykx J J, Bakker S C, Lentjes E, Neeleman M, Strengman E, Mentink
Iva Petrovchich et al. 229
L, DeYoung J, de Jong S, Sul J H, Eskin E, van Eijk K, van Setten J,
Buizer-Voskamp J E, Cantor R M, Lu A, van Amerongen M, van
Dongen E P, Keijzers P, Kappen T, Borgdorff P, Bruins P, Derks E
M, Kahn R S, Ophoff R A (2014). Genome-wide association study of
monoamine metabolite levels in human cerebrospinal uid. Mol
Psychiatry, 19(2): 228234
Madeira C, Freitas M E, Vargas-Lopes C, Wolosker H, Panizzutti R
(2008). Increased brain D-amino acid oxidase (DAAO) activity in
schizophrenia. Schizophr Res, 101(1-3): 7683
Maekawa M, Owada Y, Yoshikawa T (2011). Role of polyunsaturated
fatty acids and fatty acid binding protein in the pathogenesis of
schizophrenia. Curr Pharm Des, 17(2): 168175
Maletić-SavatićM, Vingara L K, Manganas L N, Li Y, Zhang S, Sierra
A, Hazel R, Smith D, Wagshul M E, Henn F, Krupp L, Enikolopov
G, Benveniste H, DjurićP M, Pelczer I (2008). Metabolomics of
neural progenitor cells: a novel approach to biomarker discovery.
Cold Spring Harb Symp Quant Biol, 73:389401
Manowitz P, Gilmour D G, Racevskis J (1973). Low plasma tryptophan
levels in recently hospitalized schizophrenics. Biol Psychiatry, 6(2):
109118
Martins-de-Souza D (2014). Proteomics, metabolomics, and protein
interactomics in the characterization of the molecular features of
major depressive disorder. Dialogues Clin Neurosci, 16(1): 6373
Middleton F A, Mirnics K, Pierri J N, Lewis D A, Levitt P (2002). Gene
expression proling reveals alterations of specic metabolic path-
ways in schizophrenia. J Neurosci, 22(7): 27182729
Milanovic S M, Thermenos H W, Goldstein J M, Brown A, Gabrieli S
W, Makris N, Tsuang M T, Buka S L, Seidman L J(2011). Medial
prefrontal cortical activation during working memory differentiates
schizophrenia and bipolar psychotic patients: a pilot fMRI study.
Schizophr Res, 129(2-3): 208210
Moon M L, Joesting J J, Lawson M A, Chiu G S, Blevins N A, Kwakwa
K A, Freund G G (2014). The saturated fatty acid, palmitic acid,
induces anxiety-like behavior in mice. Metabolism, 63(9): 1131
1140
Moreno F A, Parkinson D, Palmer C, Castro W L, Misiaszek J, El
Khoury A, Mathé A A, Wright R, Delgado P L (2010). CSF
neurochemicals during tryptophan depletion in individuals with
remitted depression and healthy controls. Eur Neuropsychopharma-
col, 20(1): 1824
Mück-Seler D, JakovljevićM, DeanovićZ (1988). Time course of
schizophrenia and platelet 5-HT level. Biol Psychiatry, 23(3): 243
251
Nichenametla S N, Ellison I, Calcagnotto A, Lazarus P, Muscat J E,
Richie J P Jr (2008). Functional signicance of the GAG
trinucleotide-repeat polymorphism in the gene for the catalytic
subunit of gamma-glutamylcysteine ligase. Free Radic Biol Med, 45
(5): 645650
Nunes A F, Amaral J D, Lo A C, Fonseca M B, Viana R J, Callaerts-
Vegh Z, DHooge R, Rodrigues C M (2012). TUDCA, a bile acid,
attenuates amyloid precursor protein processing and amyloid-β
deposition in APP/PS1 mice. Mol Neurobiol, 45(3): 440454
Olney J W, Farber N B (1995). Glutamate receptor dysfunction and
schizophrenia. Arch Gen Psychiatry, 52(12): 9981007
OrešičM, Tang J, Seppänen-Laakso T, Mattila I, Saarni S E, Saarni S I,
Lönnqvist J, Sysi-Aho M, Hyötyläinen T, Perälä J, Suvisaari J
(2011). Metabolome in schizophrenia and other psychotic disorders:
a general population-based study. Genome Med, 3(3): 19
Paoletti L, Elena C, Domizi P, Banchio C (2011). Role of
phosphatidylcholine during neuronal differentiation. IUBMB Life,
63(9): 714720
Park H R, Kim J Y, Park K Y, Lee J (2011). Lipotoxicity of palmitic
Acid on neural progenitor cells and hippocampal neurogenesis.
Toxicol Res, 27(2): 103110
Payne I R, Walsh E M, Whittenburg E J (1974). Relationship of dietary
tryptophan and niacin to tryptophan metabolism in schizophrenics
and nonschizophrenics. Am J Clin Nutr, 27(6): 565571
Peterson C, Vannucci M, KarakasC, Choi W, Ma L, Maletic-Savatic M
(2013). Inferring metabolic networks using the Bayesian adaptive
graphical lasso with informative priors. Stat Interface, 6(4): 547558
Prell G D, Green J P, Kaufmann C A, Khandelwal J K, Morrishow A M,
Kirch D G, Linnoila M, Wyatt R J (1995). Histamine metabolites in
cerebrospinal uid of patients with chronic schizophrenia: their
relationships to levels of other aminergic transmitters and ratings of
symptoms. Schizophr Res, 14(2): 93104
Raffa M, Mechri A, Othman L B, Fendri C, Gaha L, Kerkeni A (2009).
Decreased glutathione levels and antioxidant enzyme activities in
untreated and treated schizophrenic patients. Prog Neuropsycho-
pharmacol Biol Psychiatry, 33(7): 11781183
Ramos-Loyo J, Medina-Hernández V, Estarrón-Espinosa M, Canales-
Aguirre A, Gómez-Pinedo U, Cerdán-Sánchez L F (2013). Sex
differences in lipid peroxidation and fatty acid levels in recent onset
schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry, 44:
154161
Reiter R J, Tan D X, Jou M J, Korkmaz A, Manchester L C, Paredes S D
(2008). Biogenic amines in the reduction of oxidative stress:
melatonin and its metabolites. Neuro Endocrinol Lett, 29(4): 391
398
Santin L J, Bilbao A, Pedraza C, Matas-Rico E, López-Barroso D,
Castilla-Ortega E, Sánchez-López J, Riquelme R, Varela-Nieto I, de
la Villa P, Suardíaz M, Chun J, De Fonseca F R, Estivill-Torrús G
(2009). Behavioral phenotype of maLPA1-null mice: increased
anxiety-like behavior and spatial memory decits. Genes Brain
Behav, 8(8): 772784
Santos-Soto I J, Chorna N, Carballeira N M, Vélez-Bartolomei J G,
Méndez-Merced A T, Chornyy A P, Peña de Ortiz S (2013).
Voluntary running in young adult mice reduces anxiety-like behavior
and increases the accumulation of bioactive lipids in the cerebral
cortex. PLoS One, 8(12): e81459
Schell M J, Brady R O Jr, Molliver M E, Snyder S H (1997). D-serine as
a neuromodulator: regional and developmental localizations in rat
brain glia resemble NMDA receptors. J Neurosci, 17(5): 16041615
Schwarcz R, Rassoulpour A, Wu H Q, Medoff D, Tamminga C A,
Roberts R C (2001). Increased cortical kynurenate content in
schizophrenia. Biol Psychiatry, 50(7): 521530
Sekar A, Bialas A R, de Rivera H, Davis A, Hammond T R, Kamitaki N,
Tooley K, Presumey J, Baum M, Van Doren V, Genovese G, Rose S
A, Handsaker R E, Daly M J, Carroll M C, Stevens B, McCarroll S A,
and the Schizophrenia Working Group of the Psychiatric Genomics
Consortium (2016). Schizophrenia risk from complex variation of
complement component 4. Nature, 530(7589): 177183
Shimazu T, Hirschey M D, Newman J, He W, Shirakawa K, Le Moan N,
Grueter C A, Lim H, Saunders L R, Stevens R D, Newgard C B,
Farese R VJr, de Cabo R, Ulrich S, Akassoglou K, Verdin E (2013).
230 Metabolomics in Schizophrenia and Major Depressive Disorder
Suppression of oxidative stress by β-hydroxybutyrate, an endogen-
ous histone deacetylase inhibitor. Science, 339(6116): 211214
Singer T, McConnell M J, Marchetto M C, Coufal N G, Gage F H
(2010). LINE-1 retrotransposons: mediators of somatic variation in
neuronal genomes? Trends Neurosci, 33(8): 345354
Smith Q R (2000). Transport of glutamate and other amino acids at the
blood-brain barrier. J Nutr, 130(4SSuppl): 1016S1022S
Smoller J W (2016). The genetics of stress-related disorders: PTSD,
depression, and anxiety disorders. Neuropsychopharmacology, 41
(1): 297319
Stahl S M, Woo D J, Mefford I N, Berger P A, Ciaranello R D (1983).
Hyperserotonemia and platelet serotonin uptake and release in
schizophrenia and affective disorders. Am J Psychiatry, 140(1): 26
30
Steffens D C, Jiang W, Krishnan K R, Karoly E D, Mitchell M W,
OConnor C M, Kaddurah-Daouk R (2010). Metabolomic differ-
ences in heart failure patients with and without major depression. J
Geriatr Psychiatry Neurol, 23(2): 138146
Stone J M, Morrison P D, Pilowsky L S (2007). Glutamate and dopamine
dysregulation in schizophreniaa synthesis and selective review. J
Psychopharmacol, 21(4): 440452
Stone T W (1993). Neuropharmacology of quinolinic and kynurenic
acids. Pharmacol Rev, 45(3): 309379
Stone W S, Faraone S V, Su J, Tarbox S I, Van Eerdewegh P, Tsuang M
T (2004). Evidence for linkage between regulatory enzymes in
glycolysis and schizophrenia in a multiplex sample. Am J Med Genet
B Neuropsychiatr Genet, 127B(1): 510
Tandon N, Bolo N R, Sanghavi K, Mathew I T, Francis A N, Stanley J A,
Keshavan M S (2013). Brain metabolite alterations in young adults at
familial high risk for schizophrenia using proton magnetic resonance
spectroscopy. Schizophr Res, 148(1-3): 5966
Tortorella A, Monteleone P, Fabrazzo M, Viggiano A, De Luca L, Maj
M (2001). Plasma concentrations of amino acids in chronic
schizophrenics treated with clozapine. Neuropsychobiology, 44(4):
167171
UptonK r,GerhardtD J, Jesuadian J S, Richardson S R, Sánchez-Luque F
J, Bodea G O, Ewing A D, Salvador-PalomequeC,van der Knaap M
S, Brennan P M, Vanderver A, Faulkner G J(2015). Ubiquitous L1
mosaicism in hippocampal neurons. Cell, 161(2): 228239
Vaz A R, Cunha C, Gomes C, Schmucki N, Barbosa M, Brites D (2015).
Glycoursodeoxycholic acid reduces matrix metalloproteinase-9 and
caspase-9 activation in a cellular model of superoxide dismutase-1
neurodegeneration. Mol Neurobiol, 51(3): 864877
Vingara L K, Yu H J,Wagshul M E , Seran D,Christodoulou C , Pelczer
I, Krupp L B, Maletić-SavatićM(2013). Metabolomic approach to
human brain spectroscopy identies associations between clinical
features and the frontal lobe metabolome in multiple sclerosis.
Neuroimage, 82: 586594
Wang S M, Han C, Lee S J, Patkar A A, Masand P S, Pae C U (2014). A
review of current evidence for acetyl-l-carnitine in the treatment of
depression. J Psychiatr Res, 53: 3037
Wang Z J, Li G M, Tang W L, Yin M (2006). Neuroprotective effects of
stearic acid against toxicity of oxygen/glucose deprivation or
glutamate on rat cortical or hippocampal slices. Acta Pharmacol
Sin, 27(2): 145150
Weber H, Klamer D, Freudenberg F, Kittel-Schneider S, Rivero O,
Scholz C J, Volkert J, Kopf J, Heupel J, Herterich S, Adolfsson R,
Alttoa A, Post A, Grußendorf H, Kramer A, Gessner A, Schmidt B,
Hempel S, Jacob C P, Sanjuán J, Moltó M D, Lesch K P, Freitag C M,
Kent L, Reif A (2014). The genetic contribution of the NO system at
the glutamatergic post-synapse to schizophrenia: further evidence
and meta-analysis. Eur Neuropsychopharmacol, 24(1): 6585
Whiteld-Gabrieli S, Thermenos H W, Milanovic S, Tsuang M T,
Faraone S V, McCarley R W, Shenton M E, Green A I, Nieto-
Castanon A, LaViolette P, Wojcik J, Gabrieli J D, Seidman L J
(2009). Hyperactivity and hyperconnectivity of the default network
in schizophrenia and in rst-degree relatives of persons with
schizophrenia. Proc Natl Acad Sci U S A. 106(4): 12791284
Wichers M C, Koek G H, Robaeys G, Verkerk R, Scharpé S, Maes M
(2005). IDO and interferon-alpha-induced depressive symptoms: a
shift in hypothesis from tryptophan depletion to neurotoxicity. Mol
Psychiatry, 10(6): 538544
Woo H I, Chun M R, Yang J S, Lim S W, Kim M J, Kim S W, Myung W
J, Kim D K, Lee S Y (2015). Plasma amino acid proling in major
depressive disorder treated with selective serotonin reuptake
inhibitors. CNS Neurosci Ther, 21(5): 417424
Wood P L (2014). Accumulation of N-acylphosphatidylserines and N-
acylserines in the frontal cortex in schizophrenia. Neurotransmitter
(Houst), 1(1): e263
Wood P L, Holderman N R (2015). Dysfunctional glycosynapses in
schizophrenia: disease and regional specicity. Schizophr Res, 166
(1-3): 235237
Wyatt R J, Vaughan T, Galanter M, Kaplan J, Green R (1972).
Behavioral changes of chronic schizophrenic patients given L-5-
hydroxytryptophan. Science, 177(4054): 11241126
Xuan J, Pan G, Qiu Y, Yang L, Su M, Liu Y, Chen J, Feng G, Fang Y, Jia
W, Xing Q, He L (2011). Metabolomic proling to identify potential
serum biomarkers for schizophrenia and risperidone action. J
Proteome Res, 10(12): 54335443
Yang J, Chen T, Sun L, Zhao Z, Qi X, Zhou K, Cao Y, Wang X, Qiu Y,
Su M, Zhao A, Wang P, Yang P, Wu J, Feng G, He L, Jia W, Wan C
(2013). Potential metabolite markers of schizophrenia. Mol
Psychiatry, 18(1): 6778
Yanik M, Vural H, Kocyigit A, Tutkun H, Zoroglu S S, Herken H, Savaş
H A, Köylü A, Akyol O (2003). Is the arginine-nitric oxide pathway
involved in the pathogenesis of schizophrenia? Neuropsychobiology,
47(2): 6165
Yao J K, Dougherty G GJr, Reddy R D, Keshavan M S, Montrose D M,
Matson W R, Rozen S, Krishnan R R, McEvoy J, Kaddurah-Daouk R
(2010). Altered interactions of tryptophan metabolites in rst-episode
neuroleptic-naive patients with schizophrenia. Mol Psychiatry, 15
(9): 938953
Yao J K, Reddy R (2011). Oxidative stress in schizophrenia:
pathogenetic and therapeutic implications. Antioxid Redox Signal,
15(7): 19992002
Zheng P, Gao H C, Li Q, Shao W H, Zhang M L, Cheng K, Yang Y, Fan
S H, Chen L, Fang L, Xie P (2012). Plasma metabonomics as a novel
diagnostic approach for major depressive disorder. J Proteome Res,
11(3): 17411748
Zheng P, Wang Y, Chen L, Yang D, Meng H, Zhou D, Zhong J, Lei Y,
Melgiri N D, Xie P (2013). Identication and validation of urinary
metabolite biomarkers for major depressive disorder. Mol Cell
Proteomics, 12(1): 207214
Iva Petrovchich et al. 231
... In addition to glutamine and arginine metabolism, amino acids altered in plasma or CSF of schizophrenia patients have been linked to nitrogen compound biosynthetic processes. The finding of changes in certain lipids, fatty acids, and amino acids has implicated phospholipid synthesis [85]. The use of metabolic profiles in CSF from drug-naïve patients compared with matched controls found elevated glucose concentrations in patients, whereas the serum glucose concentration showed no differences [7]. ...
... Tele-methylhistamine [85,86] CSF GC-MS Methyl phosphate [40] PBMC GC-MS Myo-inositol [72,76] Serum GC-MS GC-TOFMS Norepinephrine (NE) [77] Blood LC-ESI/MS/MS Octadecanoic acid (stearic acid) [72,76] Serum GC-MS GC-TOFMS (9Z)-Octadec-9-enoic acid (oleic acid) [72,80] Serum GC-MS GC-TOFMS Octanoic acid [40] PBMC GC-MS Ornithine [8] Plasma MS 2-Oxoglutarate [72] Serum GC-TOFMS 1-Oxoproline [80] Serum GC-MS PC ae C38:6 [8] Plasma MS Pentadecanoic acid [80] Serum GC-MS Pentane [79,82] Breath GC-MS 4-Pentenoic acid [72] Urine GC-TOFMS Phenylalanine [72] Serum GC-TOFMS Pipecolinic acid [72] Urine GC-TOFMS (continued) has been reported to occur in other diseases such as heart attack, rheumatoid arthritis, and nutritional deficiency [79,82]. ...
Chapter
Psychiatric disorders are some of the most impairing human diseases. Among them, bipolar disorder and schizophrenia are the most common. Both have complicated diagnostics due to their phenotypic, biological, and genetic heterogeneity, unknown etiology, and the underlying biological pathways, and molecular mechanisms are still not completely understood. Given the multifactorial complexity of these disorders, identification and implementation of metabolic biomarkers would assist in their early detection and diagnosis and facilitate disease monitoring and treatment responses. To date, numerous studies have utilized metabolomics to better understand psychiatric disorders, and findings from these studies have begun to converge. In this chapter, we briefly describe some of the metabolomic biomarkers found in these two disorders.
... The individual biochemical fingerprint represents the complex interplay of the genetic make-up, and environmentally shaped gene and protein expression of a person 10 , and provides the highest resolution to assess metabolic changes associated with pathophysiological conditions at a certain point in time. For this reason, metabolomics might be able to contribute to the elucidation of gene-environment interactions 11 leading to a better understanding of the biological and psychosomatic consequences of early life stress. ...
... 13,14 ), cancer 15 , or aging 16 . Only a limited number of studies has been conducted that applied untargeted metabolomics in the field of psychiatry, in particular on schizophrenia and MDD (see refs 11,17 for an overview). A few recent studies investigated the metabolite profiles of autism 18 , bipolar disorder 19 , PTSD 20 , and smoking behavior 21 . ...
Article
Full-text available
Childhood maltreatment (CM) can increase the risk of adverse health consequences in adulthood. A deeper insight in underlying biological pathways would be of high clinical relevance for early detection and intervention. The untargeted investigation of all detectable metabolites and lipids in biological samples represents a promising new avenue to identify so far unknown biological pathways associated with CM. Using an untargeted approach, liquid chromatography-mass spectrometry (LC-MS) was performed on peripheral blood serum samples collected three months postpartum from 105 women with varying degrees of CM exposure. Comprehensive univariate and multivariate statistical analyses consistently identified eight biomarker candidates putatively belonging to antioxidant-, lipid-, and endocannabinoid-associated pathways, which differentiated between women with and without CM. Classification algorithms allowed for clear prediction of the CM status with high accuracy scores (~80-90%). Similar results were obtained when excluding all women with a lifetime psychiatric diagnosis. In order to confirm the identities of these promising biomarker candidates, LC-MS/MS analysis was applied, confirming one of the metabolites as bilirubin IXa, a potent antioxidant with immunomodulatory properties. In sum, our results suggest novel pathways that could explain long-term effects of CM on health and disease by influencing biological patterns associated with energy metabolism, inflammation, and oxidative stress.
... Metabolites are biomolecules that reflect changes in biochemical pathways in the body. [50][51][52] Factors such as sex, age, disease, environment, and nutrition can lead to differences in metabolite profiling and metabolite-metabolite networks in the blood between individuals. In a previous study, serum pro- The male and female groups are indicated by the blue and red bars, respectively. ...
Article
Bloodstain age estimation involves measuring time-dependent changes in the levels of biomolecules in bloodstains. Although several studies have identified bloodstain metabolites as markers for estimating bloodstain age, none have considered sex, age-related metabolomic differences, or long-time bloodstain age. Therefore, we aimed to identify metabolite markers for estimating the age of bloodstains at weekly intervals within 28 days and validate them through multiple reaction monitoring. Adenosine 5'-monophosphate, choline, and pyroglutamic acid were selected as markers. Seven metabolites were validated, including five previously reported metabolites, ergothioneine, hypoxanthine, L-isoleucine, L-tryptophan, and pyroglutamic acid. Choline and hypoxanthine can be used to differentiate bloodstains between days 0 and 14 after deposition at weekly intervals, whereas L-isoleucine and L-tryptophan can help distinguish bloodstains between 7 days before and 14 days after deposition. Evaluation of the changes in metabolite levels according to sex and age revealed that the average levels of all seven metabolites were higher in women on day 0. Moreover, the level of ergothioneine was significantly higher in elderly individuals than in young individuals at all time points. In this study, we confirmed the potential effectiveness of metabolites in bloodstains as forensic markers and provided a new perspective on metabolomic approaches linked to forensic science.
... Metabolomics is the study of small molecules (up to 1.5 kDa) called metabolites. Since metabolites are the final downstream products of transcription and translation, they are closest to the phenotype (6) and largely reflect environmental influences, nutritional requirements, effects of xenobiotics and drugs, stress, and various pathological or internal changes in biochemical pathways (7,8). Owing to the importance of these metabolites in biological systems, metabolite analysis is increasingly used in various research fields because it can improve the understanding of many pathological processes through altered metabolic pathways (9,10). ...
Article
Full-text available
Major depressive disorder (MDD) is one of the most common psychiatric disorders that accompany psychophysiological and mood changes. However, the pathophysiology-based disease mechanism of MDD is not yet fully understood, and diagnosis is also conducted through interviews with clinicians and patients. Diagnosis and treatment of MDD are limited due to the absence of biomarkers underlying the pathophysiological mechanisms of MDD. Although various attempts have been made to discover metabolite biomarkers for the diagnosis and treatment response of MDD, problems with sample size and consistency of results have limited clinical application. In addition, it was reported that future biomarker studies must consider exposure to antidepressants, which is the main cause of heterogeneity in depression subgroups. Therefore, the purpose of this study is to discover and validate biomarkers for the diagnosis of depression in consideration of exposure to drug treatment including antidepressants that contribute to the heterogeneity of the MDD subgroup. In the biomarker discovery and validation set, the disease group consisted of a mixture of patients exposed and unexposed to drug treatment including antidepressants for the treatment of MDD. The serum metabolites that differed between the MDD patients and the control group were profiled using mass spectrometry. The validation set including the remission group was used to verify the effectiveness as a biomarker for the diagnosis of depression and determination of remission status. The presence of different metabolites between the two groups was confirmed through serum metabolite profiling between the MDD patient group and the control group. Finally, Acetylcarnitine was selected as a biomarker. In validation, acetylcarnitine was significantly decreased in MDD and was distinguished from remission status. This study confirmed that the discovered acetylcarnitine has potential as a biomarker for diagnosing depression and determining remission status, regardless of exposure to drug treatment including antidepressants.
... A profile of metabolites in plasma, which are accessed relatively easily in clinical situations, can be an intermediate phenotype between the genome/transcriptome and general conditions of the body reflecting phenomena relevant to depression. There is increasing evidence indicating the potential contribution of plasma metabolome profiles to the understanding of depression [1][2][3] . ...
Article
Full-text available
To solve major limitations in algorithms for the metabolite-based prediction of psychiatric phenotypes, a novel prediction model for depressive symptoms based on nonlinear feature selection machine learning, the Hilbert–Schmidt independence criterion least absolute shrinkage and selection operator (HSIC Lasso) algorithm, was developed and applied to a metabolomic dataset with the largest sample size to date. In total, 897 population-based subjects were recruited from the communities affected by the Great East Japan Earthquake; 306 metabolite features (37 metabolites identified by nuclear magnetic resonance measurements and 269 characterized metabolites based on the intensities from mass spectrometry) were utilized to build prediction models for depressive symptoms as evaluated by the Center for Epidemiologic Studies-Depression Scale (CES-D). The nested fivefold cross-validation was used for developing and evaluating the prediction models. The HSIC Lasso-based prediction model showed better predictive power than the other prediction models, including Lasso, support vector machine, partial least squares, random forest, and neural network. l-leucine, 3-hydroxyisobutyrate, and gamma-linolenyl carnitine frequently contributed to the prediction. We have demonstrated that the HSIC Lasso-based prediction model integrating nonlinear feature selection showed improved predictive power for depressive symptoms based on metabolome data as well as on risk metabolites based on nonlinear statistics in the Japanese population. Further studies should use HSIC Lasso-based prediction models with different ethnicities to investigate the generality of each risk metabolite for predicting depressive symptoms.
... O QUINA também é um produto intermediário da metabolização da QUIN, e, ao contrário do AQ, que é um agonista, o QUINA age como antagonista dos receptores NMDA e, portanto, exerce ação neuroprotetora na SCZ (28). O QUINA é sintetizado e liberado por astrócitos e também exerce ação como antagonista em receptores α7nicotínico de acetilcolina (α7nAChR) (47,50). ...
Article
Full-text available
O triptofano (TRP) é um aminoácido essencial, encontrado no plasma principalmente ligado à albumina, e com apenas uma pequena fração encontrada na forma livre. Ao atravessar a barreia hematoencefálica, seus metabólitos exercem diferentes ações no sistema nervoso central. Importantes metabólitos neuroativos resultantes do metabolismo do TRP (como a serotonina, melatonina, 3-hidroxiquinurenina, ácido quinolínico e quinurênico, dentre outros) têm sido associados a doenças neurodegenerativas como o Mal de Alzheimer, a doença de Huntington, o transtorno bipolar (TB) e a esquizofrenia (SCZ). Além disto, há também relatos sobre a sua relação com a depressão, as doenças inflamatórias, diversas alergias e com doenças infecciosas. O objetivo deste trabalho foi reunir informações recentes e atualizadas sobre o que se sabe sobre papel do TRP e as seus metabólitos, enfatizando a relação desse aminoácido e seus metabólitos na fisiopatologia da SCZ, com enfoque na via das quinureninas e sobretudo suas eventuais associações com as doenças mentais.
Article
Full-text available
Schizophrenia (SCZ) is a neuropsychiatric disorder, caused by a combination of genetic and environmental factors. The etiology behind the disorder remains elusive although it is hypothesized to be associated with the aberrant response to neurotransmitters, such as dopamine and glutamate. Therefore, investigating the link between dysregulated metabolites and distorted neurodevelopment holds promise to offer valuable insights into the underlying mechanism of this complex disorder. In this study, we aimed to explore a presumed correlation between the transcriptome and the metabolome in a SCZ model based on patient-derived induced pluripotent stem cells (iPSCs). For this, iPSCs were differentiated towards cortical neurons and samples were collected longitudinally at various developmental stages, reflecting neuroepithelial-like cells, radial glia, young and mature neurons. The samples were analyzed by both RNA-sequencing and targeted metabolomics and the two modalities were used to construct integrative networks in silico. This multi-omics analysis revealed significant perturbations in the polyamine and gamma-aminobutyric acid (GABA) biosynthetic pathways during rosette maturation in SCZ lines. We particularly observed the downregulation of the glutamate decarboxylase encoding genes GAD1 and GAD2, as well as their protein product GAD65/67 and their biochemical product GABA in SCZ samples. Inhibition of ornithine decarboxylase resulted in further decrease of GABA levels suggesting a compensatory activation of the ornithine/putrescine pathway as an alternative route for GABA production. These findings indicate an imbalance of cortical excitatory/inhibitory dynamics occurring during early neurodevelopmental stages in SCZ. Our study supports the hypothesis of disruption of inhibitory circuits to be causative for SCZ and establishes a novel in silico approach that enables for integrative correlation of metabolic and transcriptomic data of psychiatric disease models.
Preprint
Full-text available
Schizophrenia is a neuropsychiatric disorder, caused by a combination of genetic and environmental factors. Recently, metabolomic studies based on patients’ biofluids and post-mortem brain specimens have revealed altered levels of distinct metabolites between healthy individuals and patients with schizophrenia (SCZ). However, a putative link between dysregulated metabolites and distorted neurodevelopment has not been assessed and access to patients’ material is restricted. In this study, we aimed to investigate a presumed correlation between transcriptomics and metabolomics in a SCZ model using patient-derived induced pluripotent stem cells (iPSCs). iPSCs were differentiated towards cortical neurons and samples were collected longitudinally at defined developmental stages, such as neuroepithelium, radial glia, young and mature neurons. Samples were subsequently analyzed by bulk RNA-sequencing and targeted metabolomics. The transcriptomic analysis revealed dysregulations in several extracellular matrix-related genes in the SCZ samples observed in early neurogenesis, including members of the collagen superfamily. At the metabolic level, several lipid and amino acid discrepancies were correlated to the SCZ phenotype. By employing a novel in silico analysis, we correlated the transcriptome with the metabolome through the generation of integrative networks. The network comparison between SCZ and healthy controls revealed a number of consistently affected pathways in SCZ, related to early stages of cortical development, indicating abnormalities in membrane composition, lipid homeostasis and amino acid imbalances. Ultimately, our study suggests a novel approach of correlating in vitro metabolic and transcriptomic data obtained from a patient-derived iPSC model. This type of analysis will offer novel insights in cellular and genetic mechanisms underlying the pathogenesis of complex neuropsychiatric disorders, such as schizophrenia.
Article
Full-text available
Background The mammalian brain is organized into regions with specific biological functions and properties. These regions have distinct transcriptomes, but little is known whether they may also differ in their metabolome. The metabolome, a collection of small molecules or metabolites, is at the intersection of the genetic background of a given cell or tissue and the environmental influences that affect it. Thus, the metabolome directly reflects information about the physiologic state of a biological system under a particular condition. The objective of this study was to investigate whether various brain regions have diverse metabolome profiles, similarly to their genetic diversity. The answer to this question would suggest that not only the genome but also the metabolome may contribute to the functional diversity of brain regions. Methods We investigated the metabolome of four regions of the mouse brain that have very distinct functions: frontal cortex, hippocampus, cerebellum, and olfactory bulb. We utilized gas- and liquid- chromatography mass spectrometry platforms and identified 215 metabolites. Results Principal component analysis, an unsupervised multivariate analysis, clustered each brain region based on its metabolome content, thus providing the unique metabolic profile of each region. A pathway-centric analysis indicated that olfactory bulb and cerebellum had most distinct metabolic profiles, while the cortical parenchyma and hippocampus were more similar in their metabolome content. Among the notable differences were distinct oxidative-anti-oxidative status and region-specific lipid profiles. Finally, a global metabolic connectivity analysis using the weighted correlation network analysis identified five hub metabolites that organized a unique metabolic network architecture within each examined brain region. These data indicate the diversity of global metabolome corresponding to specialized regional brain function and provide a new perspective on the underlying properties of brain regions. Conclusion In summary, we observed many differences in the metabolome among the various brain regions investigated. All four brain regions in our study had a unique metabolic signature, but the metabolites came from all categories and were not pathway-centric. Electronic supplementary material The online version of this article (10.1186/s12918-018-0644-0) contains supplementary material, which is available to authorized users.
Article
The potential role in neuropsychiatric disease risk arising from alterations and derangements of endogenous small molecule metabolites remains understudied. Alterations of endogenous metabolite concentrations can arise in multiple ways. Marked derangements of single endogenous small-molecule metabolites are found in a large group of rare genetic human diseases referred to as “inborn errors of metabolism,” many of which are associated with prominent neuropsychiatric symptomology. Whether such metabolites act neuroactively to directly lead to distinct neural dysfunction has been frequently hypothesized but rarely demonstrated unequivocally. Here we discuss this disease concept in the context of our recent findings demonstrating that neural dysfunction arising from accumulation of the schizophrenia-associated metabolite L-proline is due to L-proline-mediated GABA-mimesis that leads to alterations in GABA-ergic short-term synaptic plasticity. We lay out a broad, systematic approach that can be extended to assessing the potential disruptive neuroactive action of any candidate disease-associated metabolite upon neurotransmitter system function. To address the potentially important and broader role in neuropsychiatric disease, we also consider whether the more subtle yet more ubiquitous variations in endogenous metabolites arising from natural allelic variation may likewise contribute to disease risk but in a more complex and nuanced manner.
Article
Full-text available
Our six-week treadmill running training (forced exercise) model has revealed that mild exercise (ME) with an intensity below the lactate threshold (LT) is sufficient to enhance spatial memory, while intense exercise (IE) above the LT negates such benefits. To help understand the unrevealed neuronal and signaling/molecular mechanisms of the intensity-dependent cognitive change, in this rat model, we here investigated plasma corticosterone concentration as a marker of stress, adult hippocampal neurogenesis (AHN) as a potential contributor to this ME-induced spatial memory, and comprehensively delineated the hippocampal transcriptomic profile using a whole-genome DNA microarray analysis approach through comparison with IE. Results showed that only IE had the higher corticosterone concentration than control, and that the less intense exercise (ME) is better suited to improve AHN, especially in regards to the survival and maturation of newborn neurons. DNA microarray analysis using a 4 x 44 K Agilent chip revealed that ME regulated more genes than did IE (ME: 604 genes, IE: 415 genes), and only 41 genes were modified with both exercise intensities. The identified molecular components did not comprise well-known factors related to exercise-induced AHN, such as brain-derived neurotrophic factor. Rather, network analysis of the data using Ingenuity Pathway Analysis algorithms revealed that the ME-influenced genes were principally related to lipid metabolism, protein synthesis and inflammatory response, which are recognized as associated with AHN. In contrast, IE-influenced genes linked to excessive inflammatory immune response, which is a negative regulator of hippocampal neuroadaptation, were identified. Collectively, these results in a treadmill running model demonstrate that long-term ME, but not of IE, with minimizing running stress, has beneficial effects on increasing AHN, and provides an ME-specific gene inventory
Article
Full-text available
Owing to their capacity for self-renewal and pluripotency, stem cells possess untold potential for revolutionizing the field of regenerative medicine through the development of novel therapeutic strategies for treating cancer, diabetes, cardiovascular and neurodegenerative diseases. Central to developing these strategies is improving our understanding of biological mechanisms responsible for governing stem cell fate and self-renewal. Increasing attention is being given to the significance of metabolism, through the production of energy and generation of small molecules, as a critical regulator of stem cell functioning. Rapid advances in the field of metabolomics now allow for in-depth profiling of stem cells both in vitro and in vivo, providing a systems perspective on key metabolic and molecular pathways which influence stem cell biology. Understanding the analytical platforms and techniques that are currently used to study stem cell metabolomics, as well as how new insights can be derived from this knowledge, will accelerate new research in the field and improve future efforts to expand our understanding of the interplay between metabolism and stem cell biology.
Article
Full-text available
Despite significant progress in the structural and functional characterization of the human genome, understanding of the mechanisms underlying the genetic basis of human phenotypic uniqueness remains limited. Here, I report that transposable element-derived sequences, most notably LTR7/HERV-H, LTR5_Hs, and L1HS, harbor 99.8% of the candidate human-specific regulatory loci (HSRL) with putative transcription factor-binding sites in the genome of human embryonic stem cells (hESC). A total of 4,094 candidate HSRL display selective and site-specific binding of critical regulators (NANOG, POU5F1, CTCF, Lamin B1), and are preferentially located within the matrix of transcriptionally active DNA segments that are hypermethylated in hESC. hESC-specific NANOG-binding sites are enriched near the protein-coding genes regulating brain size, pluripotency long non-coding RNAs, hESC enhancers, and 5-hydroxymethylcytosine-harboring regions immediately adjacent to binding sites. Sequences of only 4.3% of hESC-specific NANOG-binding sites are present in Neanderthals' genome, suggesting that a majority of these regulatory elements emerged in Modern Humans. Comparisons of estimated creation rates of novel TF-binding sites revealed that there was 49.7-fold acceleration of creation rates of NANOG-binding sites in genomes of Chimpanzees compared to the mouse genomes and further 5.7-fold acceleration in genomes of Modern Humans compared to the Chimpanzees genomes. Preliminary estimates suggest that emergence of one novel NANOG-binding site detectable in hESC required 466 years of evolution. Pathway analysis of coding genes that have hESC-specific NANOG-binding sites within gene bodies or near gene boundaries revealed their association with physiological development and functions of nervous and cardiovascular systems, embryonic development, behavior, as well as development of a diverse spectrum of pathological conditions such as cancer, diseases of cardiovascular and reproductive systems, metabolic diseases, multiple neurological and psychological disorders. A proximity placement model is proposed explaining how a 33-47% excess of NANOG, CTCF, and POU5F1 proteins immobilized on a DNA scaffold may play a functional role at distal regulatory elements. © The Author(s) 2015. Published by Oxford University Press on behalf of the Society for Molecular Biology and Evolution.
Article
Full-text available
Somatic LINE-1 (L1) retrotransposition during neurogenesis is a potential source of genotypic variation among neurons. As a neurogenic niche, the hippocampus supports pronounced L1 activity. However, the basal parameters and biological impact of L1-driven mosaicism remain unclear. Here, we performed single-cell retrotransposon capture sequencing (RC-seq) on individual human hippocampal neurons and glia, as well as cortical neurons. An estimated 13.7 somatic L1 insertions occurred per hippocampal neuron and carried the sequence hallmarks of target-primed reverse transcription. Notably, hippocampal neuron L1 insertions were specifically enriched in transcribed neuronal stem cell enhancers and hippocampus genes, increasing their probability of functional relevance. In addition, bias against intronic L1 insertions sense oriented relative to their host gene was observed, perhaps indicating moderate selection against this configuration in vivo. These experiments demonstrate pervasive L1 mosaicism at genomic loci expressed in hippocampal neurons. Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.
Article
Schizophrenia is a heritable brain illness with unknown pathogenic mechanisms. Schizophrenia's strongest genetic association at a population level involves variation in the major histocompatibility complex (MHC) locus, but the genes and molecular mechanisms accounting for this have been challenging to identify. Here we show that this association arises in part from many structurally diverse alleles of the complement component 4 (C4) genes. We found that these alleles generated widely varying levels of C4A and C4B expression in the brain, with each common C4 allele associating with schizophrenia in proportion to its tendency to generate greater expression of C4A. Human C4 protein localized to neuronal synapses, dendrites, axons, and cell bodies. In mice, C4 mediated synapse elimination during postnatal development. These results implicate excessive complement activity in the development of schizophrenia and may help explain the reduced numbers of synapses in the brains of individuals with schizophrenia.
Article
Neurodegenerative diseases are progressive, devastating, and terminal, carrying both personal and societal burden. Currently, their diagnosis depends on their clinical presentation. No quantitative biomarkers exist to enable early verdict and commencement of therapy. The lack of diagnostic biomarkers stems from the unavailability of brain tissue, the complexity and heterogeneity of the brain and neurodegenerative pathology, and the fact that peripheral tissues such as blood, urine, and even cerebrospinal fluid might not reflect early stages of brain pathology. Moreover, accumulated evidence indicates the majority of these diseases are not genetically inherited; rather, the genes bring about the risk to develop them, but the trigger is not known. As metabolites are at the intersection between the genetic background of a cell or a tissue and the environmental effects on the same, metabolomics has emerged as a field with great promise to deliver new, biologically, and clinically relevant biomarkers for neurodegenerative disorders. Here, we review the basic principles of metabolomics and focus on studies performed in most common neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, Multiple sclerosis, and Amyotrophic lateral sclerosis.
Article
Research into the causes of psychopathology has largely focused on two broad etiologic factors: genetic vulnerability and environmental stressors. An important role for familial/heritable factors in the etiology of a broad range of psychiatric disorders was established well before the modern era of genomic research. This review focuses on the genetic basis of three disorder categories--posttraumatic stress disorder (PTSD), major depressive disorder (MDD), and the anxiety disorders-for which environmental stressors and stress responses are understood to be central to pathogenesis. Each of these disorders aggregates in families and is moderately heritable. More recently, molecular genetic approaches, including genomewide studies of genetic variation, have been applied to identify specific risk variants. In this review, I summarize evidence for genetic contributions to PTSD, MDD, and the anxiety disorders including genetic epidemiology, the role of common genetic variation, the role of rare and structural variation, and the role of gene-environment interaction. Available data suggest that stress-related disorders are highly complex and polygenic and, despite substantial progress in other areas of psychiatric genetics, few risk loci have been identified for these disorders. Progress in this area will likely require analysis of much larger sample sizes than have been reported to date. The phenotypic complexity and genetic overlap among these disorders present further challenges. The review concludes with a discussion of prospects for clinical translation of genetic findings and future directions for research.Neuropsychopharmacology accepted article preview online, 31 August 2015. doi:10.1038/npp.2015.266.
Article
While schizophrenia is generally considered a neurodevelopment disorder, our basic understanding of the biochemical processes involved in disease etiology and/or progression is limited. One class of biochemical mediators that has been suggested to play a role in the development of schizophrenia is N-acyl ethanolamine metabolites of N-acylphosphatidylethanolamines. However, no investigations of N-acylphosphatidylserines or their N-acylserine metabolites have been published. We undertook a targeted postmortem lipidomics analysis of N-acylphosphatidylserines (NAPS) and N-acylserines (NAS) in gray matter of the frontal cortex of schizophrenia subjects. Our data are the first to demonstrate that NAPS and NAS are present in human brain. Furthermore, NAPS and their bioactive metabolites, N-acylserines (NAS), were found to be significantly elevated in the frontal cortex of schizophrenia subjects. Elevated levels of NAPS lipid pools in schizophrenia may result in complex alterations in the structural function of neuronal membranes while increases in NAS may alter signal transduction pathways.
Article
Our previous lipidomics studies demonstrated elevated sulfatides, plasmalogens, and N-acylphosphatidylserines in the frontal cortex of schizophrenia subjects. These data suggest that there may be an abnormal function of glycosynapses in schizophrenia. We further examined the disease and anatomical specificity of these observations. We undertook a targeted lipidomics analysis of plasmalogens, sulfatides, and N-acyl-phosphatidylserines in the frontal cortex obtained from schizophrenia, bipolar, and ALS subjects and the cerebellum of schizophrenia subjects. We demonstrate that sulfatides, plasmalogens, and N-acyl-phosphatidylserines are significantly elevated in the frontal cortex of patients suffering from schizophrenia and bipolar depression but not in ALS patients. These lipids were unchanged in the cerebellum of subjects with schizophrenia. Our data suggest that dysfunction of oligodendrocyte glycosynapses may be specific to limbic circuits in schizophrenia and that this dysfunction is also detected in bipolar depression, suggesting that these disorders possess several common pathophysiological features. Copyright © 2015 Elsevier B.V. All rights reserved.