ArticlePDF AvailableLiterature Review

Metabolomics of ischemic stroke: insights into risk prediction and mechanisms

Authors:

Abstract and Figures

Ischemic stroke (IS) is the most prevalent type of stroke. The early diagnosis and prognosis of IS are crucial for successful therapy and early intervention. Metabolomics, a tool in systems biology based on several innovative technologies, can be used to identify disease biomarkers and unveil underlying pathophysiological processes. Accordingly, in recent years, an increasing number of studies have identified metabolites from cerebral ischemia patients and animal models that could improve the diagnosis of IS and prediction of its outcome. In this paper, metabolomic research is comprehensively reviewed with a focus on describing the metabolic changes and related pathways associated with IS. Most clinical studies use biofluids (e.g., blood or plasma) because their collection is minimally invasive and they are ideal for analyzing changes in metabolites in patients of IS. We review the application of animal models in metabolomic analyses aimed at investigating potential mechanisms of IS and developing novel therapeutic approaches. In addition, this review presents the strengths and limitations of current metabolomic studies on IS, providing a reference for future related studies.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Metabolic Brain Disease
https://doi.org/10.1007/s11011-022-01011-7
REVIEW ARTICLE
Metabolomics ofischemic stroke: insights intorisk prediction
andmechanisms
RuijieZhang1,2· JiajiaMeng1,2,3· XiaojieWang4· LiyuanPu1,2· TianZhao1,2· YiHuang5,6,7· LiyuanHan1,2
Received: 7 February 2022 / Accepted: 16 May 2022
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2022
Abstract
Ischemic stroke (IS) is the most prevalent type of stroke. The early diagnosis and prognosis of IS are crucial for suc-
cessful therapy and early intervention. Metabolomics, a tool in systems biology based on several innovative technolo-
gies, can be used to identify disease biomarkers and unveil underlying pathophysiological processes. Accordingly, in
recent years, an increasing number of studies have identified metabolites from cerebral ischemia patients and animal
models that could improve the diagnosis of IS and prediction of its outcome. In this paper, metabolomic research is
comprehensively reviewed with a focus on describing the metabolic changes and related pathways associated with IS.
Most clinical studies use biofluids (e.g., blood or plasma) because their collection is minimally invasive and they are
ideal for analyzing changes in metabolites in patients of IS. We review the application of animal models in metabo-
lomic analyses aimed at investigating potential mechanisms of IS and developing novel therapeutic approaches. In
addition, this review presents the strengths and limitations of current metabolomic studies on IS, providing a refer-
ence for future related studies.
Keywords Metabolomics· Stroke· Ischemic stroke· Molecular diagnosis· Metabolism· Biomarkers
Introduction
Stroke is a major cause of adult disability and the sec-
ond leading cause of mortality worldwide (Collaborators
etal. 2018). Epidemiological studies have reported that
ischemic stroke (IS) accounts for 85% of all stroke cases
(Fatahzadeh and Glick 2006). Research has revealed that
early diagnosis and treatment improves the prognosis of
IS patients, but current imaging-based diagnostic tools
for IS, such as magnetic resonance imaging and non-
contrast computed tomography, are time-consuming,
costly, and limited in their sensitivity and availability
(Laborde etal. 2012; Goyal etal. 2020; Montaner etal.
2020). Additionally, the complex etiology of IS means
that its pathogenesis remains unclear (Latchaw etal.
2009; Makris etal. 2018; Shin etal. 2020). In this regard,
the emerging omics science of metabolomics has great
potential to reveal the underlying pathobiology of IS,
identify novel biomarkers, and guide the development
of therapies.
Ruijie Zhang and Jiajia Meng contributed equally to this paper.
* Yi Huang
huangy102@gmail.com
* Liyuan Han
hanliyuan@ucas.ac.cn
1 Hwa Mei Hospital, University ofChinese Academy
ofSciences, Ningbo315010, Zhejiang, China
2 Ningbo Institute ofLife andHealth Industry, University
ofChinese Academy ofSciences, Ningbo315010, Zhejiang,
China
3 Xihu District Center forDisease Control andPrevention,
Hangzhou310013, Zhejiang, China
4 Department ofNeurology, Shenzhen Qianhai Shekou Free
Trade Zone Hospital, Shenzhen518067, Guangdong, China
5 Department ofNeurosurgery, Ningbo First Hospital,
Ningbo315010, Zhejiang, China
6 Key Laboratory ofPrecision Medicine forAtherosclerotic
Diseases ofZhejiang Province, Ningbo315010, Zhejiang,
China
7 Medical Research Center, Ningbo First Hospital,
Ningbo315010, Zhejiang, China
Metabolic Brain Disease
1 3
Metabolomics andits analytical approach
Metabolomics is a powerful tool in systems biology that
is used to qualify and quantify endogenous small mol-
ecules (≤ 1.5ka) in various types of biological samples,
such as feces, biological fluids, and tissues (Nicholson
etal. 2002; Sidorov etal. 2019; Yang etal. 2020; Li
etal. 2021). Metabolomics is positioned below genomics
(DNA), transcriptomics (RNA), and proteomics (protein)
in the hierarchy of systems biology tools and provides the
most accurate representation of the chemical intermedi-
ates or endpoints in cellular processes (Peng etal. 2015;
Ussher etal. 2016; Qureshi etal. 2017; Dang etal. 2018;
Rinschen etal. 2019). Thus, metabolomics best reflects
the actual phenotype of a cell or organism and can depict
its biological status. It can be used to identify biomarkers
for disease diagnosis and monitoring and to delineate the
molecular mechanisms of pathological processes (Laborde
etal. 2012; Sidorov etal. 2019; Donatti etal. 2020).
Recently, metabolomics has been increasingly applied
in biomedical research to investigate various diseases, such
as IS and other cerebrovascular diseases. It has been used
to analyze various biological specimens, such as serum,
plasma, and urine (Nicholson and Lindon 2008; Sidorov
etal. 2019; Donatti etal. 2020). Most analytical platforms
that have been used in such studies are based on nuclear
magnetic resonance spectroscopy and mass spectroscopy,
which offer comprehensive profiles of the metabolic state
of an organism (Liu etal. 2016b; Au 2018).
An appropriate experimental design is essential for the
comprehensive measurement of all metabolites in metabo-
lomic studies and may be used to address related biological
and medical questions (Fig.1). The two strategies applied in
IS metabolomics are untargeted metabolomics and targeted
metabolomics, and each has different objectives (Dang etal.
2018; Saorin etal. 2020; Zahoor etal. 2021).
Untargeted metabolomics involves the detection of all
metabolites in a given sample and generates an unbiased,
Fig.1 General workflow for metabolomics analyses, which can be
regarded as 3 distinct phases. A An experiment is designed, per-
formed, and specimen collection/preparation for metabolomics
assays. B Metabolite features of samples are profiled and detected by
nuclear magnetic resonance (NMR) or mass spectrometry (MS). C
Statistical analyses are implemented to identify significant metabolite
features, metabolic pathway/cluster analyses, and mechanism/model
formulation. GC indicates gas chromatography; LC, liquid chroma-
tography; PCA, principal components analysis and PLS-DA, Partial
least squares discrimination analysis
Metabolic Brain Disease
1 3
comprehensive dataset of metabolites. It examines a wide
range of metabolite classes by comprehensively analyzing all
metabolite sets and thus often discovers previously unknown
metabolites that must be characterized by comparing their
mass spectral data with those of standards and/or with data
in mass spectral databases (Au 2018; Zahoor etal. 2021).
Untargeted metabolomics is used to screen for differences
between the metabolic profiles of diseased and healthy con-
trol groups, which enables the discovery of novel metabolic
biomarkers and reveals underlying metabolic networks (Au
2018; Zhang etal. 2021). However, this approach is only
semi-quantitative at best and is thus not sufficiently precise
or accurate for diagnostic purposes (Tokarz etal. 2017).
Targeted metabolomics is designed for hypothesis-driven
studies and involves the measurement of a predefined set of
metabolites in samples (Tokarz etal. 2020). This approach is
typically used to validate potential biomarkers that are iden-
tified during the discovery phase or to evaluate the response
of a specific metabolic pathway to a treatment (Wishart
2019; Zahoor etal. 2021). Targeted metabolomics is supe-
rior to untargeted metabolomics in terms of precision, accu-
racy, and calibration ranges and enables the absolute con-
centrations of each metabolite of interest to be determined
(Au 2018; Tokarz etal. 2020; Zahoor etal. 2021). However,
the finite number of known metabolites that are analyzed in
targeted metabolomics means that key compounds or path-
ways may remain undetected (Au 2018).
Metabolomic data analysis
Metabolomic datasets are extremely complex and must
be pretreated to reduce systematic biases during analysis
(Fig.1) (Saorin etal. 2020). Then, appropriate statistical
methods—either univariate or multivariate statistical meth-
ods—must be used to reveal the metabolic profiles of dif-
ferent types of samples. Univariate methods, such as a t-test
or a one-way analysis of variance (ANOVA), are used to
measure one response variable in metabolomic data (Au
2018). Multivariate methods, such as unsupervised princi-
pal component analysis and supervised partial least-squares
discriminant analysis (PLS-DA) are widely used to identify
disease biomarkers in metabolomic data (Zhou etal. 2012).
Other multivariate methods, such as multivariate analy-
sis of variance, ANOVA-simultaneous component analy-
sis, orthogonal partial least-squares (OPLS) analysis, and
OPLS-DA, are also used to analyze metabolomic data (Au
2018; Saorin etal. 2020). Metabolomic data have also been
analyzed using machine learning techniques, such as soft
independent modeling of class analogy, hierarchical cluster
analysis, self-organizing maps, support-vector machines, and
random forests (Au 2018; Azad and Shulaev 2019; Tiedt
etal. 2020).
Metabolites can influence various biological systems and
thus affect genetic, epigenetic, and physiological responses
to disease processes and environmental stimuli (Nichol-
son and Lindon 2008; Qureshi etal. 2017; Montaner etal.
2020). Consequently, the metabolic profile of an organism
may directly reflect its physiological and pathological states.
Metabolomics therefore has great potential to facilitate the
integrated mapping of specific processes underlying physi-
ological and pathological states (Johnson etal. 2016). In
the context of IS, the primary goal of metabolomics is
to identify circulating metabolites that provide systems-
level information on the molecular pathways that underlie
IS development and progression, thereby revealing novel
diagnostic and prognostic biomarkers (Qureshi etal. 2017;
Montaner etal. 2020). Metabolite set enrichment analysis
and metabolic network analysis can also be used to provide
key insights into how metabolic changes mediate the biol-
ogy of disease processes or pathological phenomena of IS
(Liu etal. 2016a). Commonly used pathway databases are
the Kyoto Encyclopedia of Genes and Genomes, MetaCyc,
the Small Molecule Pathway Database, and MetaboLights.
These databases are used to meet the increasing demand
for biologically meaningful and informative correlations
between molecular metabolites and a physiological process
or pathological phenotype (Cambiaghi etal. 2017; Hernan-
dez-de-Diego etal. 2018; Li etal. 2021). Additionally, vari-
ous bioinformatics tools (such as MetaboAnalyst, 3Omics,
PaintOmics, integrOmics, and MetScape) are being devel-
oped for determining the relationships between metabolites
and their biological functions (Cambiaghi etal. 2017; Her-
nandez-de-Diego etal. 2018; Li etal. 2021).
Overview ofmetabolomic studies ofIS
Several types of study designs can be used in metabolomics
studies to diagnose and prognose IS. Diagnostic studies have
event-based designs and are used to determine the differ-
ences between control and diseased subjects for differential
disease diagnoses or to investigate the efficacy of treatments
(Fig.2A). For example, in one diagnostic study, a patient’s
post-IS metabolite metrics were measured and compared
with those of a healthy individual, and the differences were
interpreted metabolically (Everett etal. 2019).
Recently, a case–control study of 969 participants was
conducted to identify metabolites that are significantly
associated with IS. It revealed significant differences in the
serum concentrations of 41 endogenous metabolites between
the IS group and the control group (Table1); the AUC for 30
of these metabolites was 0.961, thus confirming significant
differences between the IS patients and the control group.
The study also discovered that the serum concentrations
of pregnenolone sulfate, adenosine, and asymmetric and
Metabolic Brain Disease
1 3
symmetric dimethylarginine effectively discriminated IS
patients from patients with stroke mimics (Tiedt etal. 2020).
Another study analyzed serum samples from 38 IS patients
and 46 control subjects and found that the IS patients had
higher serum concentrations of arginine, vaccenyl carnitine
(C18:1), palmitoylcarnitine (C16), and 3-hydroxylbutyryl-
carnitine (C4OH) and a lower citrulline/arginine ratio than
the controls (Sun etal. 2019a).
In another metabolomic study, the serum samples of 40
IS patients were found to have abnormal concentrations of
amino acids and their metabolites compared with 29 control
subjects. Specifically, IS patients had lower serum concen-
trations of alanine, glycine, isoleucine, leucine, serine, tyros-
ine, methionine, tryptophan, urea, purine, hypoxanthine, and
proline, reflecting disrupted amino acid metabolism (Wang
etal. 2017). In contrast, Goulart etal. (2019) found that
serum concentrations of proline, leucine, and glycine were
significantly higher in IS patients than in the controls (Gou-
lart etal. 2019). These studies have also detailed how serum
concentrations of amino acids differed between acute and
chronic IS phases. Finally, dyslipidemia increases the risk
of atherosclerosis, which in turn increases the risk of IS,
and studies have found that serum samples of IS patients
contained lower concentrations of lysophosphatidylcholines
(lysoPCs) and phosphatidylcholines (PCs) and higher con-
centrations of acylcarnitines than those of healthy subjects,
indicating that IS disrupts phospholipid metabolism and
fatty acid oxidation (Liu etal. 2017; Sun etal. 2017).
Prognostic studies have outcome-based designs and
aim to predict IS outcomes from baseline or longitudinal
metabolic analyses, e.g., the occurrence of future IS or the
prognosis of existing IS in groups of subjects (Fig.2B). A
related type of baseline study involves the identification of
prognostically relevant metabolites based on the metabo-
lomic profiling of biofluids obtained from epidemiological
or biobank studies of IS patients and controls. Longitudinal
studies involve the measurement of metabolic responses in
the biofluid samples of a cohort of IS patients to determine
Fig. 2 Schematic Representations of the Three Key Experimental
Designs in IS Metabonomics research. Subjects are on the left-hand
side followed by the procedures and targets of metabonomics analysis
on the right-hand side. A Case–control design for diagnostic meta-
bonomics approach, differentiating healthy individuals (blue) from
subjects with a disease (red) at a given time point. B Nested case–
control design for predictive metabonomics approach. In this case,
there is a difference (although not complete discrimination) in meta-
bolic profiles before IS between two subgroups(Cambridge blue and
dark blue). C Case-time-control design for diagnostic and prognos-
tic metabonomics approach, differentiating patients with IS in accute
stage (red) from subjects in chronic stage (blue) at a given time point
Metabolic Brain Disease
1 3
Table 1 Metabolomics studies in patients with Ischemic stroke
Study Published
date
Study-
design
Group Validation Average
age
Targeted/
Untargeted
Study
objective
Specimens Analysis
platform
Suggested biomarker
Steffen Tiedt etal.
(Tiedt etal.
2020)
2020 Case–con-
trol
Stage1(IS = 74,NC
= 72),Stage3(SM
= 33, IS = 40);
Stage2(IS = 40,
NC = 40),Stage4(SM = 105,
IS = 105),Stage5(SM = 211,
IS = 289)
70.0 Untargeted Diagnostic
bio-
marker
Serum UPLC-
MS/MS
Asymmetrical and sym-
metrical dimethylar-
ginine, pregnenolone
sulfate, adenosine
Evgeny Sidorov
etal.(Sidorov
etal. 2020a)
2020 Case-time-
control
AS = 20, CS = 20 None 57.0 Targeted Biomarkers
for acute
ischemic
stroke
Serum and
urine
LC–MS Asparagine, tyrosine,
xylose,glycine, acetyl-
carnitine
Laurent Suissa
etal.(Suissa
etal. 2020)
2020 Nested
case–
control
FO = 18,
UFO = 23
None 74.8 Untargeted Predicting
biomark-
ers of
outcome
Cerebral
thrombi
LC–MS Sorbitol
Evgeny Sidorov
etal. (Sidorov
etal. 2020b)
2020 Case-time-
control
AS = 60, CS = 60 None 60.0 Untargeted Discovery
of serum
bio-
markers
related
to infarct
volume
Serum UPLC-
MS/MS
4 unknown metabolites
Nai-Fang Chi etal.
(Chi etal. 2021)
2020 Nested
case–
control
FO = 77, UFO = 73 None 67.4 Untargeted Biomarkers
of func-
tional
recovery
of stroke
Serum UPLC-
MS/MS
147 metabolites
Daokun Sun
etal. (Sun etal.
2019a)
2019 Nested
case–
control
IS = 346,NC = 3558 IS = 114,NC = 112 53.5 Untargeted Predicting
inci-
dent of
ischemic
stroke
Serum GC–MS tetradecanedioate, hexa-
decanedioate
Vânia A. M. Gou-
lart etal.(Goulart
etal. 2019)
2019 Case–con-
trol
Ath = 13, Car = 7,
Lac = 10, Und = 8,
NC = 16
None 61.6 Targeted Stroke
etiology
Plasma GC–MS Proline, lysine, phenyla-
lanine, leucine, glycine,
methionine, alanine
Ruitan Sun etal.
(Sun etal.
2019b)
2019 Case–con-
trol
IS = 38,NC = 46 None 66.6 Targeted Diagnostic
bio-
marker
Serum direct-
infusion
mass
spec-
trometry
Vaccenylcarnitine
(C18:1), palmitoylcar-
nitine (C16),
3-hydroxylbutyrylcarni-
tine (C4OH), arginine,
arginine /ornithine,
citrulline /arginine
Metabolic Brain Disease
1 3
Table 1 (continued)
Study Published
date
Study-
design
Group Validation Average
age
Targeted/
Untargeted
Study
objective
Specimens Analysis
platform
Suggested biomarker
Xiaofan Guo etal.
(Guo etal. 2019)
2019 Nest case–
control
IS = 66,NC = 66 None 60.8 Untargeted Predicting
inci-
dent of
ischemic
stroke
Plasma UPLC-
MS/MS
9-cis-Retinal, DL-
Indole-3-Lactic Acid,
(2S)-OMPT, 2-methyl-
1-pyrroline, d-pipeco-
linic Acid, 7,7-dime-
thyl-5,8-eicosadienoic
acid, PC(15:0/0:0)
[U], DL-dihydrosphin-
gosine, bufexamic
acid, 10- L-thyroxine,
L-thyroxine
Michael V.
Holmes(Holmes
etal. 2018)
2018 Nest case–
control
MI = 912,IS = 1146
,ICH = 1138,NC
= 1466
None 47.0 Targeted Investigat-
ing the
associa-
tions of
metabolic
makers
with
different
CVD
subtypes
Plasma H.1-NMR Glycoprotein-acetyls,
β-hydroxybutyrate,
glucose, acetoacetate,
docosahexaenoic acid
Yeseung Lee etal.
(Lee etal. 2017)
2017 Nest case–
control
IS = 62,NC = 348 IS = 99,NC = 301 62.1 Targeted Predicting
incident
ischemic
stroke
Serum LC–MS N6-acetyl-L-lysine,
cadaverine,
nicotinamide,2-
oxoglutarate, L-valine,
S-(2-methylpropionyl)-
dihydrolipoamide-
E,ubiquinone,5-
aminopentanoate,
homocysteine, sulfinic
acid, lysine
Peifang Liu etal.
(Liu etal. 2017)
2017 Case–con-
trol
IS = 40,NC = 40 IS = 26,NC = 23 58.1 Untargeted Diagnostic
bio-
marker
Serum UPLC-
MS/
MS,GC–
MS
serine, isoleucine,
betaine, PC(5:0/5:0),
LysoPE(18:2)
Dian Wang etal.
(Wang etal.
2017)
2017 Case–con-
trol
IS = 40,NC = 29 None 63.7 Targeted Diagnostic
bio-
marker
Serum GC–MS Tyrosine, lactate, tryp-
tophan
Hongxue Sun etal.
(Sun etal. 2017)
2017 Case–con-
trol
IS = 30,NC = 30 None 57.6 Targeted Diagnostic
bio-
marker
Serum LC–MS Uric acid, sphinganine,
adrenoyl ethanolamide
Metabolic Brain Disease
1 3
their metabolic trajectories over time (Fig.2C) (Everett
etal. 2019). These prognostic (or predictive) studies aim to
discover prognostic indicators in cohorts of IS patients and
better interpret details of IS progression.
Most of the baseline or longitudinal studies of IS have
examined the metabolic profiles of blood samples to deter-
mine IS incidence or predict its recurrence. Several prospec-
tive studies have reported significantly altered blood concen-
trations of some organic acids and lipids, such as bufexamac
acid, D-pipecolinic acid, dodecanoic acid, and lysoPCs. For
example, one of the first untargeted metabolomic studies
metabolically profiled plasma samples from 293 patients
with recurrent IS. Plasma concentrations of 1-monopal-
mitin, dodecanoic acid, meso-erythritol, threonate, and
lysoPCs were decreased in these patients. Sun etal. (2019a)
quantified 245 metabolites in the serum samples of 3,904
participants (IS = 346, Control = 3558) and compared these
samples to validated samples from 114 IS patients and 112
healthy controls to investigate the association between the
245 metabolites and IS incidence. They found that tetrade-
canedioate and hexadecanedioate were associated with IS
incidence. Holmes etal. (2018) quantified 225 metabolites
in baseline plasma samples from 912 myocardial infarction
(MI) patients, 1,146 IS patients, and 1,466 healthy control
subjects to investigate the association of lipids and related
metabolites with the risks of IS and MI. Their findings
demonstrated that low-density lipoprotein cholesterol and
triglycerides were positively associated with IS (Holmes
etal. 2018). Glutamate functions as an indicator of neu-
ral excitation and the presence of neurotoxins in the brain
and contributes to neuronal injury (Castellanos etal. 2008;
Meng etal. 2015). Serum concentrations of 2-oxoglutarate,
which is a derivative of glutamate, were found to be lower
in patients at risk of thrombotic stroke (a type of IS), which
suggests that glutamate can exhibit increased excitotoxic
activity (Papes etal. 2001; Kimberly etal. 2013; Lee etal.
2017). This finding was supported by another study by Wang
etal. (Wang etal. 2017). A further two longitudinal studies
performed metabolomic profiling of serum samples obtained
from patients over time, which revealed that higher serum
concentrations of asparagine, tyrosine, and xylose were sig-
nificantly associated with acute IS (Sidorov etal. 2020a) and
that four unknown metabolites were associated with infarc-
tion volume in acute IS (Sidorov etal. 2020b).
The studies described above only utilized surrogate bio-
fluids (i.e., serum or plasma) to depict a global overview of
metabolic changes associated with IS. The collection of bio-
fluids is minimally invasive and is feasible for biomonitoring
large samples. However, biofluids are reflective of many bio-
chemical responses across various tissues within the body,
not just the brain, which is the target organ of stroke. Studies
have shown that only a few altered metabolites, caused by
only certain diseases, can be found in both the brain and
Table 1 (continued)
Study Published
date
Study-
design
Group Validation Average
age
Targeted/
Untargeted
Study
objective
Specimens Analysis
platform
Suggested biomarker
P.A. Vorkas etal.
(Vorkas etal.
2016)
2016 Case–con-
trol
SYM = 5,
ASYM = 5
None None Untargeted Diagnostic
bio-
marker
Carotid
Plaques
UPLC-MS butyrylcarnitine, hex-
anoylcarnitine, palmi-
toylcarnitine, TG(58:6),
PC(16:0/20:4),
PC(16:0/18:1),
PE(18:1/18:0), arachi-
donic acid (AA)
Mariona Jové etal.
(Jove etal. 2015)
2015 Nest case–
control
SR = 20,NC = 111 IS = 15,NC = 147 71.7 Untargeted Predicts
stroke
recur-
rence
Plasma LC–MS 1-monopalmitin, dode-
canoic acid, meso-
erythritol, threonate,
LysoPC[16:0], myris-
toyl-ethanolamine,
LysoPC(20:4)
IS, ischemic stroke; NC, normal control; SM, stroke mimics; Ath,atherothrombotic stroke; Car, cardioembolic stroke; Lac,lacunar stroke; Und, undetermined stroke; MI, myocardial infarction;
ICH, intracerebral hemorrhage; AS, acute stage of ischemic stroke; CS, chronic stage of ischemic stroke; SR, stroke recurrence; FO, favorable outcome; UFO, unfavorable outcome; SYM, cer-
ebrovascular symptoms of carotid origin; ASYM, cerebrovascular asymptoms of carotid origin
Metabolic Brain Disease
1 3
the blood. This is disadvantageous for pathological inter-
pretations of disease, as pathway analyses depend on these
altered metabolites. The few studies of stroke that are based
on metabolomic techniques have used brain tissue as a biosa-
mple. The molecular mechanism through which metabolic
dysregulation in brain tissue leads to stroke remains unclear.
Overview ofmetabolomic studies onmiddle
cerebral artery occlusion (MCAO) models
ofIS
Given the inherent ethical and safety concerns regarding the
use of tissue samples from human IS patients for studying
thromboembolic occlusions, various animal models of stroke
have been developed. These models are indispensable for the
following reasons: (1) Unlike human IS, which has diverse
manifestations, experimental IS is a highly reproducible,
controllable, and standardized system, which allows patho-
physiological processes and the effects of potential therapeu-
tics to be analyzed accurately. (2) The affected brain tissue
of animals can be subjected to molecular, biochemical, and
physiological analyses. (3) The pathophysiological changes
associated with IS are detectable by imaging techniques
within a minute of IS occurrence in animal models; such
an assessment is not practical in humans. (4) Perfusion and
vasculature can be modeled in animals, but not invitro (Fluri
etal. 2015; Shin etal. 2020).
The MCAO model is most widely used to investigate the
mechanisms underlying IS and to develop novel therapies.
However, neither the MCAO model nor any other animal
models of IS perfectly replicate the pathological features of
the human disease (Sommer 2017; Kaiser and West 2020).
For example, the MCAO model has several limitations, such
as hyperthermia or hypothermia problems, uncontrollable
infarct location and size, a high risk of vessel rupture with
certain suture types, and unsuitability for neuroprotective
studies (Fluri etal. 2015). In addition, there are macro-
scopic and microscopic differences between humans and the
MCAO model, such that the model fails to fully simulate
human IS; these include differences in brain anatomy, func-
tional organization, genetics and epigenetics, and treatment
reactions (Sommer 2017). Furthermore, subtypes of stroke
in clinical settings can be atherothrombotic or cardioembolic
in origin, while stroke in an animal model is mainly pro-
voked by arterial occlusion. Thus, an animal model cannot
simulate all of the metabolic changes seen in human stroke
subtypes. The results of an animal study therefore cannot be
directly compared to the results of a clinical study. Neverthe-
less, the MCAO model is extremely useful for providing new
perspectives on the diagnosis of IS and for the evaluation of
clinical disease activity.
IS-related metabolites, such as amino acids, lipids, poly-
amines, and nucleotides, have been explored in animal mod-
els to understand disease mechanisms in cerebral ischemia
(Table2). Metabolites have been identified in various types
of biological samples, such as rat plasma, brain tissue,
liver tissue, and cerebrospinal fluid. A liquid chromatog-
raphy–mass spectrometry (LC–MS) analysis by Guo etal.
(2020) found that the serum concentrations of 37 metabo-
lites in an MCAO group were significantly different from
those in a control group (Guo etal. 2020). These metabo-
lites included sphingomyelin, lysoPC, stearidonic acid, PCs,
organic acids, amino acids, and carnitine derivatives, and
pathway enrichment analyses suggested that key metabo-
lites were involved in the biosynthesis of unsaturated fatty
acids, ɑ-linolenic acid metabolism, fatty acid metabolism,
the synthesis and degradation of ketone bodies, and pentose
and glucuronate interconversions (Guo etal. 2020). Simi-
larly, Wu and Liu used LC–MS to reveal that there were
decreased concentrations of PCs, phosphatidylethanola-
mines (PEs), lysoPEs, and sphingosine-1-phosphate in the
blood samples of a rat MCAO model, reflecting perturba-
tions in lipid metabolism pathways (Liu etal. 2016a; Wu
etal. 2020). Paik investigated lipid metabolism in the blood
of rats subjected to MCAO surgery and found that the sur-
gery caused changes in the concentrations of 19 free fatty
acid metabolites (Paik etal. 2009). Additionally, the blood
concentrations of lactate, glucose, glucose 6-phosphate, suc-
cinic acid, malic acid, and citric acid were increased in these
animals compared with control rats; as these metabolites are
associated with energy metabolism, this indicated that the
glucose and anaerobic glycolysis metabolism pathways were
perturbed (Wang etal. 2019). In contrast, Wang etal. (2014)
found that an MCAO rat model had lower blood concentra-
tions of lactate (L-lactic acid) than the control rats, as well as
significantly lower blood concentrations of leucine, isoleu-
cine, and valine, which is consistent with human cohort stud-
ies and indicates perturbed amino acid metabolism (Wang
etal. 2014). We observed changes in the concentrations of
the same metabolites (e.g., lactate, creatine, glycine, alanine,
leucine, and lysine) in the brain tissue, cerebrospinal fluid,
and plasma of MCAO rats, but the changes varied across
these matrices (Kimberly etal. 2013; Wang etal. 2013; Luo
etal. 2019; Wesley etal. 2019).
We collected the identified metabolites published in clini-
cal metabolomics studies and animal studies, with a com-
parison of their consistency in blood between the two types
of studies. A total of 190 metabolites identified in Murine
model studies share 15 ones in clinical studies. We exam-
ined whether their tendency of 15 metabolites is the same
between clinical studies and animal studies. If it showed
the same change in or more than two of three studies, we
determined it in the same trend. Of 15 metabolites, three
have the same tendency in both types of studies, including
Metabolic Brain Disease
1 3
Table 2 Metabolic alterations associated with MCAo in rats model
Study Group Specimens or
subjects
Analysis platform Elevated metabo-
lites
Reduced metabolites Metabolites type pathway
Bingfeng Lin etal.
(Lin etal. 2021)
NC = 15,
MCAo = 15,AEGE = 15,Nim = 15
Plasma LC–MS Tyramine, thy-
mine, lysine,
L-carnitine,
L-Histidine,
L-acetylcarnitine,
deoxycytidine,
lauroyl diethanol-
amide, glutamyl-
phenylalanine,
5-methylthioad-
enosine, phyto-
sphingosine,
corticosterone,
11b,17a,21-tri-
hydroxypreg-
nenolone, netilm-
icin, LysoPC
[P-18:1 (9z)],
LysoPC(P-18:0)
3-Indolehydracrylic
acid, LysoPC [20:3
(5Z,8Z,11Z)/0:0],
LysoPC [20:1 (11Z)],
LysoPC(20:0/0:0),
LysoPE (0:0/24:0),
LysoPC [22:1 (13Z)/0:0],
arachidonic acid,
5a-tetrahydrocortisol,
lansiumarin C, Scuti-
geral, Mammeigin,
LysoPE (18:0/0:0)
amino acids,
lipids,other
metabolites
phenylalanine, tyros-
ine, and trypto-
phan biosynthesis;
phenylalanine
metabolism; histi-
dine metabolism;
sphingolipid
metabolism; pyrimi-
dine metabolism;
cysteine and
methionine
metabolism; tyrosine
metabolism;
steroid-hormone
biosynthesis
Wenjun Guo etal.
(Guo etal. 2020)
NC = 10, MCAo = 10 Serum LC–MS Acetoacetic acid,
l-acetylcarnitine,
deoxyuridine,
2-hydroxybutyric
acid, leucylpro-
line, hippuric
acid, decenedioic
acid, 3-oxodo-
decanoic acid,
l-palmitoylcar-
nitine, 3-oxotet-
radecanoic acid,
eicosapentaenoic
acid, palmitic
amide, hexa-
decadienoic acid,
alpha-linolenic
acid, palmitoleic
acid, docosap-
entaenoic acid
(22n-3), dihomo-
alpha-linolenic
acid, oleic acid,
eicosadienoic
acid, PC (36:4),
SM (d18:0/18:1)
Proline, molybdate,
trichloroethanol glucu-
ronide, 2-phenylethanol
glucuronide, glycocholic
acid, chenodeoxyglyco-
cholic acid, 2-hydrox-
yhexadecanoylcarnitine,
hydroxyhexadecanoic
acid, goshuyic acid,
LysoPC (20:3), LysoPC
(20:2), myristoleic acid,
LysoPC (20:1), steari-
donic acid, palmitic acid,
SM (d18:1/20:0)
Lipids,organic
acids, amino
acids,other
metabolites
Unsaturated fatty
acids, alpha-
linolenic acid
metabolism, fatty
acid metabolism,
synthesis and deg-
radation of ketone
bodies, pentouse
and glucuronate
interconversions
Metabolic Brain Disease
1 3
Table 2 (continued)
Study Group Specimens or
subjects
Analysis platform Elevated metabo-
lites
Reduced metabolites Metabolites type pathway
Lanlan Wu etal.
(Wu etal. 2020)
NC = 11, MCAo = 11 Serum LC–MS L-lactic acid,
isobutyrylgly-
cine, sebacic
acid, linoleic
acid, gluco-
sylgalactosyl
hydroxylysine,
docosahexaenoic
acid, oxoglutaric
acid
Hydroxyphenylacetyl-
glycine, N-methyl-
nicotinium, N2-suc-
cinyl-L-ornithine,
tauroursodeoxycholic
acid, sphingosine
1-phosphate,sphinganine
1-phosphate,2-hydrox-
yestradiol-3-methyl ether,
glutamylphenylalanine,
vitamin D2 3-glucuron-
ide, neoxanthin, stearoyl-
carnitine, petroselinic
acid
Lipids, organic
acids, amino
acids, other
metabolites
Di-unsaturated fatty
acid β-oxidation;
glycine, serine,
alanine, threo-
nine metabolism;
glycolysis and
gluconeogenesis;
glycosphingolipid
metabolism;
linoleate metabo-
lism; TCA cycle;
urea cycle, metab-
olism of arginine,
proline, glutamate,
aspartate, and
asparagine
Umadevi V.
Wesleya etal.
(Wesley etal.
2019) ξ
NC = 3,MCAo2d = 3, MCAo3d = 3,
MCAo7d = 3, MCAo14d = 3
Plasma 1H-NMR Glycine, isoleu-
cine, o-acetylcar-
nitine,, lysine
Methionine,Choline, Cit-
rate, Proline, Threonine,
Tyrosine
Amino acids,
lipids, other
metabolites
None
Umadevi V.
Wesleya etal.
(Wesley etal.
2019).ψ
NC = 3,MCAo2d = 3, MCAo3d = 3,
MCAo7d = 3, MCAo14d = 3
Liver 1H-NMR Choline,
3-hydroxybu-
tyrate, Creatine,
Taurine
Tyrosine, glutathione, tryp-
tophan, serine, alanine,
histamine, PC
Amino acids,
lipids,other
metabolites
None
Umadevi V.
Wesleya etal.
(Wesley etal.
2019)ξ
NC = 3, MCAo2d = 3, MCAo3d = 3,
MCAo7d = 3, MCAo14d = 3
Brain tissue 1H-NMR 3-hydroxybutyrate,
creatine, taurine,
glycine, methio-
nine, isoleucine,
ethanolamine,
fumarate, glyc-
erol, pantothen-
ate, tryptophan
Glutathione, tryptophan,
serine, alanine, hista-
mine, N-acetylaspartate
Amino acids,
lipids,other
metabolites
None
Lan Luo etal. (Luo
etal. 2019)
NC = 16, MCAo = 16 Brain tissue 1H-NMR Lactate, acetate
(Ace), myo-
inositol(m-Ins),
γ-aminobutyric
acid (GABA),
aspartate (Asp),
alanine (Ala),
leucine (Leu),
isoleucine (Ile),
choline (Cho)
Creatine/phosphocreatine
(Cr/PCr), N-acetyl-aspar-
tate (NAA), N-acetylas-
partylglutamate (NAAG)
Organic acids,
amino acids,
other metabolites
None
Metabolic Brain Disease
1 3
Table 2 (continued)
Study Group Specimens or
subjects
Analysis platform Elevated metabo-
lites
Reduced metabolites Metabolites type pathway
Lan Luo etal. (Luo
etal. 2019)
NC = 16, MCAo = 16 Urine 1H-NMR creatinine (Crn),
acetoacetate,
acetone (Aco),
N-acetyl-glyco-
protein (NAG),
dimethyl-
amine (DMA),
3-hydroxybu-
tyrate
_ Organic acids,
polyamines,
other metabolites
None
Lan Luo etal.(Luo
etal. 2019)
NC = 16, MCAo = 16 Plasma 1H-NMR Lactate, trimethyl-
amine-N-oxide/
betaine
VLDL/LDL, 3-hydroxybu-
tyrate (3-HB), ace-
toacetate (AcAc), poly
unsaturated fatty acid
(PUFA)
Organic acids,
lipids, other
metabolites
None
Metabolic Brain Disease
1 3
Table 2 (continued)
Study Group Specimens or
subjects
Analysis platform Elevated metabo-
lites
Reduced metabolites Metabolites type pathway
Yang Wang etal.
(Wang etal.
2019)
NC = 10, MCAo = 10 Serum GC–MS 4-Hydroxy-proline,
allantoic acid,
beta-alanine, cre-
atine, creatinine,
homocysteine,
isoleucine, ser-
ine, methionine
sulfoxide, orni-
thine, Arabitol,
glucose, ribose,
xylose, glucose
6-phosphate,
ribonolactone,
caproic acid,
docosahex-
aenoic acid,
dodecanoic acid,
heptadecanoic
acid, linoleic
acid, myristic
acid, myristoleic
acid, oleic acid,
Palmitic acid,
palmitoleic acid,
pentadecanoic
acid, stearic
acid, cholesterol,
o-phosphoe-
thanolamine,
pregnenolone,
guanosine,
inosine, pipecolic
acid, malic acid,
Petroselinic acid,
Picolinic acid,
succinic acid,
Dopamine, pan-
tothenic acid
Arachidonic acid Lipids, organic
acids, amino
acids, carbo-
hydrates, other
metabolites
Linoleic acid metab-
olism, homocyst-
eine degradation,
fatty acid biosyn-
thesis, amino acid
metabolism
Metabolic Brain Disease
1 3
Table 2 (continued)
Study Group Specimens or
subjects
Analysis platform Elevated metabo-
lites
Reduced metabolites Metabolites type pathway
Mengting Liu etal.
(Liu etal. 2016b)
NC = 10, MCAo = 10 Plasma UPLC/TOF–MS Cholic acid,
pseudouridine/
uridine, pyruvate,
taurine, lactate,
tryptophan,
glutamine, his-
tidine, cytidine,
creatine, alanine,
phenylalanine,
5-hydroxyin-
doleaceticacid,
norepinephrine,
palmitoyl-
L-carnitine,
N-stearoylserine
Acetone, PC(14:0/0:0),
PE(17:0/0:0),
PS(20:0/0:0), PC(14:1),
LysoPE(0:0/16:0),
LysoPE(20:1),
LysoPE(0:0/24:1(15Z)),
LysoPE(20:0/0:0),
LysoPE(24:0)
Nucleotide, lipids,
organic acids,
other metabolites
Monoamine
neurotransmitter
metabolism,amino
acid
metabolism,energy
metabolism, lipid
metabolism
Yun Wang etal.
(Wang etal.
2014)*
NC = 10, MCAO0 = 10,
MCAO0.5 = 10, MCAO1 = 10,
MCAO3 = 10, MCAO6 = 10,
MCAO12 = 10, MCAO24 = 10
Serum 1H-NMR Malonic acid,
citric acid, gly-
cine, ornithine,
pyruvic acid,
glutamic acid,
succinic acid
L-lactic acid, betaine,
L-serine, acetic acid,
L-valine, L-alanine
Organic acid,
amino acids
TCA
Yun Wang etal.
(Wang etal.
2013)*
NC = 10, MCAO0 = 10,
MCAO0.5 = 10, MCAO1 = 10,
MCAO3 = 10, MCAO6 = 10
Cerebrospinal fluid 1H-NMR L-lactic acid,
L-Alanine,
glutamic acid
None Lipids, organic
acids, amino
acids
None
W. Taylor
Kimberly etal.
(Kimberly etal.
2013)
NC = 7,SMCAo = 6,LMCAo = 7 Cerebrospinal fluid LC–MS Xanthosine, lysine Leucine, isoleucine, valine Amino acids, other
metabolites
None
W. Taylor
Kimberly etal.
(Kimberly etal.
2013)
NC = 7,SMCAo = 6,LMCAo = 7 Plasma LC–MS Xanthosine, carno-
sine, glutamate
Leucine, isoleucine, valine,
niacinamide, phenyla-
lanine
Amino acids, other
metabolites
None
Metabolic Brain Disease
1 3
Table 2 (continued)
Study Group Specimens or
subjects
Analysis platform Elevated metabo-
lites
Reduced metabolites Metabolites type pathway
Man Jeong Paik
etal.(Paik etal.
2009)
NC = 8, MCAo = 6 Plasma GC–MS Caproic
acid(C6:0),
caprylic
acid(C8:0), dece-
noic acid(C10:1),
capric
acid(C10:0), lau-
ric acid(C12:0),
myristoleic
acid(C14:1),
myristic
acid(C14:0),
γ-linolenic
acid(C18:3n6),
eicosapentaenoic
acid(C20:5n3),
docosahexaenoic
acid (C22:6n3),
docsapentaenoic
acid (C22:5n3),
erucic acid
(C22:1), nervonic
acid (C24:1),
lignoceric acid
(C24:0), hexa-
cosanoic acid
(C26:0)
Palmitic acid(C16:0), lin-
oleic acid(C18:2n6), ara-
chidonic acid(C20:4n6),
eicosenoic acids(C20:1)
Lipids None
ξ To investigate metabolite changes in cerebral ischemia/reperfusion (I/R) injury, 3-month rats were euthanized at day 2, 3, 7 and 14 of I/R. ψ For study of ischemia/reperfusion injury related to
time, 12-month rats were grouped to reperfusion time of day 2, 3, 7 and 14. *To investigate metabolite changes in cerebral ischemia/reperfusion injury, 0, 0.5, 1, 3, 6, 12, 24h of I/R
Metabolic Brain Disease
1 3
lower levels of proline, tyrosine, and valine. It suggests that
cerebral artery occlusion will cause turbulent amino acid
metabolism, both in the rat of MCAO model and patients of
IS. On the contrary, the four inconsistent ones were isoleu-
cine, glycine, lysine, and alanine, with higher concentrations
in the rat of MCAO model rat while lower levels in patients
of IS.
Benets andlimits ofthemetabolomic
analyses ofISinthe current studies
Metabolomic studies of stroke focus on changes to metabo-
lites in blood samples because the collection of these bio-
markers is easy and minimally invasive, making for a speedy
and convenient choice for clinical diagnoses. Inflammatory
mediators released in the early stage of IS occurrence dam-
age the blood–brain barrier, resulting in the leakage of pro-
teins and metabolites from the brain to peripheral blood,
where they induce changes in metabolites. This makes
metabolomic studies ideal for early diagnosis and predic-
tion of functional outcomes. However, the majority of serum
or plasma biomarkers may have low specificity due to their
lack of organ-specificity. Furthermore, expression of specific
biomarkers could be the result of altered conditions in spe-
cific cellular context of an organ. This issue can be remedied
by the application of a combination of multiple biomark-
ers to improve diagnosis and prognosis efficiency. When
blood samples are used for metabolic pathway analysis, it
is difficult to achieve a comprehensive and direct metabolic
pathway, compared with the samples of the central nervous
system. In addition, for many metabolites, the plasma lev-
els are highly regulated, and a local change could lead to a
non-detectable change in the blood concentration. However,
in some cases, blood concentrations are poorly controlled.
Other types of samples (e.g., urine) should be analyzed to
obtain a more comprehensive understanding of metabolic
pathways for IS patients.
In clinical studies, most subjects are patients within the
first 24h of stroke onset. Levels of metabolites change in
the serum or plasma of patients because of brain injury. The
altered metabolites are not only identified as biomarkers but
also investigated through metabolomic analyses of the serum
or plasma to reflect the real-time pathological condition result-
ing from the trauma of stroke. The identified pathological con-
ditions are related to clinical signatures and help to understand
the molecular mechanisms underlying clinical features. Several
days after stroke onset, biomarkers are collected and analyzed
to predict outcomes. These biomarkers reflect the biological
processes that occur when the patient is recovering from an
IS; different biochemical processes are related to favorable
or unfavorable patient outcomes. Such analyses further our
understanding of the molecular mechanisms underlying IS.
Cerebral ischemia/reperfusion leads to a series of biological
reactions such as oxidative stress, inflammation, mitochon-
drial dysfunction, reduction of antioxidative agents, and ATP
depletion(Wu etal. 2018; Liao etal. 2020). In clinical settings,
it is not feasible to confirm cell death in the ischemic region or
detect metabolic alteration in brain tissue. The animal model
is thus a good substitute for exploring the biological processes
of ischemia/reperfusion. Studies using animal models have
shown that some metabolite levels change differently, depend-
ing on whether they are in blood or brain tissue after ischemia/
reperfusion. One example is methionine, which is downregu-
lated in plasma but upregulated in brain tissue (Wesley etal.
2019). This demonstrates that changes in metabolites in the
ischemic/reperfused region of the brain do not necessarily lead
to corresponding changes in the blood. Important information
would be lost if we performed analyses of dysregulated path-
ways according to disordered metabolites in the blood alone.
This suggests that the use of blood analysis for the interpreta-
tion of biological mechanisms occurring in brain tissue during
IS and after reperfusion is limited.
Given the ethical and safety concerns involved with har-
vesting tissue samples from human IS patients, the use of
tissue samples in metabolomic studies of IS is rare. To cir-
cumvent this issue, scientists use MCAO models to imitate
human stroke. In animal studies, metabolomic techniques
are used to explore the potential biomarkers for early diag-
nosis of ischemic stroke and novel therapeutic targets. For
this purpose, blood samples are collected before and during
cerebral artery occlusion and after reperfusion to record the
integral metabolic trajectory. In contrast, blood samples of
stroke patients are generally collected several hours or days
after stroke onset; thus, obtaining metabolomic data upon
stroke onset in humans is difficult. However, published study
have demonstrated that acylcarnitines and adipokines are
associated with the destabilization and rupture of plaque
(Vorkas etal. 2016). Low levels of neurosteroids were
related to poor outcome in many brain pathologies (Adib-
hatla and Hatcher 2008). Furthermore, sorbitol and glucose
levels in the thrombi are correlated with functional outcomes
(Suissa etal. 2020). Through metabolomic analyses, we can
speculate how the metabolites change and obtain informa-
tion on the active pathways at stroke onset, which can pro-
mote improved etiological diagnosis of stroke and patient
outcomes (Jia etal. 2021; Chumachenko etal. 2022).
Conclusion
IS is a complex and multifactorial disease that accounts
for many deaths and cases of serious disability worldwide.
Thus, there is an urgent need for new approaches to disease-
specific diagnosis and prognosis, which will facilitate the
development of more precise and personalized treatment.
Metabolic Brain Disease
1 3
Metabolomics is a powerful tool in systems biology that
affords new insights into the pathological status of disease,
and studies in the past decade have demonstrated its poten-
tial utility for diagnosing and prognosing IS. In particular,
findings from the vast majority of studies suggest that many
metabolomic signatures could serve as biomarkers for the
early-stage diagnosis of IS, provide better prognostic evalu-
ations, and enable better evaluation of drug responses. How-
ever, despite the many IS-associated metabolites that have
been identified thus far, a clinical biomarker has yet to be
identified. Moreover, studies have been affected by con-
founders, such as drug-taking, diet, and comorbidity, which
profoundly influence metabolomic profiles and biomarker
discovery. Furthermore, the results of studies are inconsist-
ent in some areas because of study heterogeneity and the
range of experimental conditions and analytical techniques
used for metabolic profiling. Thus, the results are far from
being applicable to real-world practice.
Metabolomic profiling has been used to investigate IS for
more than 10years, with recent research having focused on eti-
ological, predictive, diagnostic, and prognostic aspects. Despite
the aforementioned limitations of these studies, the large vol-
ume of exploratory data has enhanced understanding of disease
mechanisms and generated a list of candidate biomarkers.
Future metabolomic studies should use standardized
experimental processes, upgraded analytical platforms, and
standard cohorts and perform refined data analyses. Insights
into disease mechanisms and disease biomarkers should be
achieved in part by integrated approaches based on a combi-
nation of metabolomics, other omics fields, and analyses of
the gut microbiota. The resulting candidate metabolites should
be subjected to validation studies before clinical application.
Authors’ contributions Meng Jiajia, Zhang Ruijie and Han Liyuan
wrote and revised the manuscript. All authors participated in critical
revision of the manuscript and approved the final version.
Funding This study is supported by the National Natural Science
Foundation of China (82173648), Innovative Talent Support Plan
of the Medical and Health Technology Project in Zhejiang Prov-
ince(2021422878), Internal Fund of Ningbo Institute of Life and Health
Industry, University of Chinese Academy of Sciences(2020YJY0212),
Sanming Project of Medicine in Shenzhen (SZSM201803080), Zhe-
jiang Provincial Public Service and Application Research Foundation
(LGF20H250001 and GC22H264267), Public Welfare Foundation of
Ningbo (2021S108), Ningbo Science and Technology Innovation 2025
Specific Project (2020Z096),and Shenzhen Nanshan District Science
and Technology Bureau (2020075).
Data Availability The datasets supporting the conclusions of this article
are available from the corresponding author.
Declarations
Ethics approval Not applicable.
Consent to participate Not applicable for animal studies.
Consent for publication Not applicable.
Competing Interests The authors have no relevant financial or non-
financial interests to disclose.
References
Adibhatla RM, Hatcher JF (2008) Altered lipid metabolism in brain
injury and disorders. Subcell Biochem 49:241–268. https:// doi.
org/ 10. 1007/ 978-1- 4020- 8831-5_9
Au A (2018) Metabolomics and Lipidomics of Ischemic Stroke. Adv
Clin Chem 85:31–69. https:// doi. org/ 10. 1016/ bs. acc. 2018. 02. 002
Azad RK, Shulaev V (2019) Metabolomics technology and bioin-
formatics for precision medicine. Br Bioinform 20:1957–1971.
https:// doi. org/ 10. 1093/ bib/ bbx170
Cambiaghi A, Ferrario M, Masseroli M (2017) Analysis of metab-
olomic data: tools, current strategies and future challenges for
omics data integration. Br Bioinform 18:498–510. https:// doi. org/
10. 1093/ bib/ bbw031
Castellanos M, Sobrino T, Pedraza S etal (2008) High plasma gluta-
mate concentrations are associated with infarct growth in acute
ischemic stroke. Neurology 71:1862–1868. https:// doi. org/ 10.
1212/ 01. wnl. 00003 26064. 42186. 7e
Chi NF, Chang TH, Lee CY etal (2021) Untargeted metabolomics
predicts the functional outcome of ischemic stroke. J Formos Med
Assoc 120:234–241. https:// doi. org/ 10. 1016/j. jfma. 2020. 04. 026
Chumachenko MS, Waseem TV, Fedorovich SV (2022) Metabolomics
and metabolites in ischemic stroke. Rev Neurosci 33:181–205.
https:// doi. org/ 10. 1515/ revne uro- 2021- 0048
Collaborators GBDLR of S, Feigin VL, Nguyen G etal (2018) Global,
Regional, and Country-Specific Lifetime Risks of Stroke, 1990
and 2016. N Engl J Med 379:2429–2437. https:// doi. org/ 10. 1056/
NEJMo a1804 492
Dang VT, Huang A, Werstuck GH (2018) Untargeted Metabolomics
in the Discovery of Novel Biomarkers and Therapeutic Targets
for Atherosclerotic Cardiovascular Diseases. Cardiovasc Hematol
Disord Drug Targets 18:166–175. https:// doi. org/ 10. 2174/ 18715
29X18 66618 04201 70108
Donatti A, Canto AM, Godoi AB, etal (2020) Circulating Metabolites
as Potential Biomarkers for Neurological Disorders-Metabolites in
Neurological Disorders. Metabolites 10. https:// doi. org/ 10. 3390/
metab o1010 0389
Everett JR, Holmes E, Veselkov KA etal (2019) A Unified Conceptual
Framework for Metabolic Phenotyping in Diagnosis and Progno-
sis. Trends Pharmacol Sci 40:763–773. https:// doi. org/ 10. 1016/j.
tips. 2019. 08. 004
Fatahzadeh M, Glick M (2006) Stroke: epidemiology, classification,
risk factors, complications, diagnosis, prevention, and medical
and dental management. Oral Surg Oral Med Oral Pathol Oral
Radiol Endod 102:180–191. https:// doi. org/ 10. 1016/j. tripl eo.
2005. 07. 031
Fluri F, Schuhmann MK, Kleinschnitz C (2015) Animal models of
ischemic stroke and their application in clinical research. Drug
Des Devel Ther 9:3445–3454. https:// doi. org/ 10. 2147/ DDDT.
S56071
Goulart VAM, Sena MM, Mendes TO etal (2019) Amino Acid Biosig-
nature in Plasma among Ischemic Stroke Subtypes. Biomed Res
Int 2019:8480468. https:// doi. org/ 10. 1155/ 2019/ 84804 68
Goyal M, Ospel JM, Menon B etal (2020) Challenging the Ischemic
Core Concept in Acute Ischemic Stroke Imaging. Stroke 51:3147–
3155. https:// doi. org/ 10. 1161/ STROK EAHA. 120. 030620
Metabolic Brain Disease
1 3
Guo X, Li Z, Zhou Y etal (2019) Metabolic profile for prediction of
ischemic stroke in Chinese hypertensive population. J Stroke Cer-
ebrovasc Dis 28:1062–1069. https:// doi. org/ 10. 1016/j. jstro kecer
ebrov asdis. 2018. 12. 035
Guo W, Wang Y, Fan M etal (2020) Integrating metabolomics and
network pharmacology to explore the protective effect of gross
saponins of Tribulus terrestris L. fruit against ischemic stroke in
rat. J Ethnopharmacol 263:113202. https:// doi. org/ 10. 1016/j. jep.
2020. 113202
Hernandez-de-Diego R, Tarazona S, Martinez-Mira C etal (2018)
PaintOmics 3: a web resource for the pathway analysis and visu-
alization of multi-omics data. Nucleic Acids Res 46:W503–W509.
https:// doi. org/ 10. 1093/ nar/ gky466
Holmes MV, Millwood IY, Kartsonaki C etal (2018) Lipids, Lipo-
proteins, and Metabolites and Risk of Myocardial Infarction and
Stroke. J Am Coll Cardiol 71:620–632. https:// doi. org/ 10. 1016/j.
jacc. 2017. 12. 006
Jia J, Zhang H, Liang X etal (2021) Application of Metabolomics to
the Discovery of Biomarkers for Ischemic Stroke in the Murine
Model: a Comparison with the Clinical Results. Mol Neurobiol
58:6415–6426. https:// doi. org/ 10. 1007/ s12035- 021- 02535-2
Johnson CH, Ivanisevic J, Siuzdak G (2016) Metabolomics: beyond
biomarkers and towards mechanisms. Nat Rev Mol Cell Biol
17:451–459. https:// doi. org/ 10. 1038/ nrm. 2016. 25
Jove M, Mauri-Capdevila G, Suarez I etal (2015) Metabolomics pre-
dicts stroke recurrence after transient ischemic attack. Neurology
84:36–45. https:// doi. org/ 10. 1212/ WNL. 00000 00000 001093
Kaiser EE, West FD (2020) Large animal ischemic stroke models:
replicating human stroke pathophysiology. Neural Regen Res
15:1377–1387. https:// doi. org/ 10. 4103/ 1673- 5374. 274324
Kimberly WT, Wang Y, Pham L etal (2013) Metabolite profiling
identifies a branched chain amino acid signature in acute cardi-
oembolic stroke. Stroke 44:1389–1395. https:// doi. org/ 10. 1161/
STROK EAHA. 111. 000397
Laborde CM, Mourino-Alvarez L, Akerstrom F etal (2012) Potential
blood biomarkers for stroke. Expert Rev Proteomics 9:437–449.
https:// doi. org/ 10. 1586/ epr. 12. 33
Latchaw RE, Alberts MJ, Lev MH etal (2009) Recommendations for
imaging of acute ischemic stroke: a scientific statement from the
American Heart Association. Stroke 40:3646–3678. https:// doi.
org/ 10. 1161/ STROK EAHA. 108. 192616
Lee Y, Khan A, Hong S etal (2017) A metabolomic study on high-risk
stroke patients determines low levels of serum lysine metabolites:
a retrospective cohort study. Mol Biosyst 13:1109–1120. https://
doi. org/ 10. 1039/ c6mb0 0732e
Li JT, Zeng N, Yan ZP etal (2021) A review of applications of metabo-
lomics in osteoarthritis. Clin Rheumatol 40:2569–2579. https://
doi. org/ 10. 1007/ s10067- 020- 05511-8
Liao S, Apaijai N, Chattipakorn N, Chattipakorn SC (2020) The pos-
sible roles of necroptosis during cerebral ischemia and ischemia
/ reperfusion injury. Arch Biochem Biophys 695:108629. https://
doi. org/ 10. 1016/j. abb. 2020. 108629
Lin B, Chen R, Wang Q etal (2021) Transcriptomic and metabolomic
profiling reveals the protective effect of Acanthopanax senticosus
(Rupr. & Maxim.) harms combined with Gastrodia Elata Blume
on cerebral Ischemia- Reperfusion injury. Front Pharmacol
12:619076. https:// doi. org/ 10. 3389/ fphar. 2021. 619076
Liu M, Liu X, Wang H etal (2016a) Metabolomics study on the effects
of Buchang Naoxintong capsules for treating cerebral ischemia
in rats using UPLC-Q/TOF-MS. J Ethnopharmacol 180:1–11.
https:// doi. org/ 10. 1016/j. jep. 2016. 01. 016
Liu M, Tang L, Liu X etal (2016b) An Evidence-Based Review of
Related Metabolites and Metabolic Network Research on Cerebral
Ischemia. Oxid Med Cell Longev 2016:9162074. https:// doi. org/
10. 1155/ 2016/ 91620 74
Liu P, Li R, Antonov AA etal (2017) Discovery of Metabolite Bio-
markers for Acute Ischemic Stroke Progression. J Proteome Res
16:773–779. https:// doi. org/ 10. 1021/ acs. jprot eome. 6b007 79
Luo L, Kang J, He Q, etal (2019) A NMR-Based Metabonomics
Approach to Determine Protective Effect of a Combination of
Multiple Components Derived from Naodesheng on Ischemic
Stroke Rats. Molecules 24. https:// doi. org/ 10. 3390/ molec ules2
40918 31
Makris K, Haliassos A, Chondrogianni M, Tsivgoulis G (2018) Blood
biomarkers in ischemic stroke: potential role and challenges in
clinical practice and research. Crit Rev Clin Lab Sci 55:294–328.
https:// doi. org/ 10. 1080/ 10408 363. 2018. 14611 90
Meng XE, Li N, Guo DZ etal (2015) High plasma glutamate levels
are associated with poor functional outcome in acute ischemic
stroke. Cell Mol Neurobiol 35:159–165. https:// doi. org/ 10. 1007/
s10571- 014- 0107-0
Montaner J, Ramiro L, Simats A etal (2020) Multilevel omics
for the discovery of biomarkers and therapeutic targets for
stroke. Nat Rev Neurol 16:247–264. https:// doi. org/ 10. 1038/
s41582- 020- 0350-6
Nicholson JK, Connelly J, Lindon JC, Holmes E (2002) Metabonomics:
a platform for studying drug toxicity and gene function. Nat Rev
Drug Discov 1:153–161. https:// doi. org/ 10. 1038/ nrd728
Nicholson JK, Lindon JC (2008) Systems biology: Metabonomics.
Nature 455:1054–1056. https:// doi. org/ 10. 1038/ 45510 54a
Paik MJ, Li WY, Ahn YH etal (2009) The free fatty acid metabolome
in cerebral ischemia following human mesenchymal stem cell
transplantation in rats. Clin Chim Acta 402:25–30. https:// doi.
org/ 10. 1016/j. cca. 2008. 12. 022
Papes F, Surpili MJ, Langone F etal (2001) The essential amino acid
lysine acts as precursor of glutamate in the mammalian central
nervous system. FEBS Lett 488:34–38. https:// doi. org/ 10. 1016/
s0014- 5793(00) 02401-7
Peng B, Li H, Peng XX (2015) Functional metabolomics: from bio-
marker discovery to metabolome reprogramming. Protein Cell
6:628–637. https:// doi. org/ 10. 1007/ s13238- 015- 0185-x
Qureshi MI, Vorkas PA, Coupland AP etal (2017) Lessons from Meta-
bonomics on the Neurobiology of Stroke. Neuroscientist 23:374–
382. https:// doi. org/ 10. 1177/ 10738 58416 673327
Rinschen MM, Ivanisevic J, Giera M, Siuzdak G (2019) Identifi-
cation of bioactive metabolites using activity metabolomics.
Nat Rev Mol Cell Biol 20:353–367. https:// doi. org/ 10. 1038/
s41580- 019- 0108-4
Saorin A, Di Gregorio E, Miolo G, etal (2020) Emerging Role of
Metabolomics in Ovarian Cancer Diagnosis. Metabolites 10.
https:// doi. org/ 10. 3390/ metab o1010 0419
Shin TH, Lee DY, Basith S, etal (2020) Metabolome Changes in Cer-
ebral Ischemia. Cells 9. https:// doi. org/ 10. 3390/ cells 90716 30
Sidorov E, Bejar C, Xu C etal (2020a) Potential Metabolite Biomarkers
for Acute Versus Chronic Stage of Ischemic Stroke: A Pilot Study.
J Stroke Cerebrovasc Dis 29:104618. https:// doi. org/ 10. 1016/j.
jstro kecer ebrov asdis. 2019. 104618
Sidorov E, Sanghera DK, Vanamala JKP (2019) Biomarker for
Ischemic Stroke Using Metabolome: A Clinician Perspective. J
Stroke 21:31–41. https:// doi. org/ 10. 5853/ jos. 2018. 03454
Sidorov EV, Bejar C, Xu C etal (2020b) Novel Metabolites as Potential
Indicators of Ischemic Infarction Volume: a Pilot Study. Transl
Stroke Res. https:// doi. org/ 10. 1007/ s12975- 020- 00876-z
Sommer CJ (2017) Ischemic stroke: experimental models and real-
ity. Acta Neuropathol 133:245–261. https:// doi. org/ 10. 1007/
s00401- 017- 1667-0
Suissa L, Guigonis JM, Graslin F etal (2020) Metabolome of cerebral
thrombi reveals an association between high glycemia at stroke
onset and good clinical outcome. Metabolites 10:1–15. https:// doi.
org/ 10. 3390/ metab o1012 0483
Metabolic Brain Disease
1 3
Sun D, Tiedt S, Yu B etal (2019a) A prospective study of serum metab-
olites and risk of ischemic stroke. Neurology 92:e1890–e1898.
https:// doi. org/ 10. 1212/ WNL. 00000 00000 007279
Sun R, Li Y, Cai M etal (2019b) Discovery of a new biomarker pattern
for differential diagnosis of acute ischemic stroke using targeted
metabolomics. Front Neurol 10:1011. https:// doi. org/ 10. 3389/
fneur. 2019. 01011
Sun H, Zhao J, Zhong D, Li G (2017) Potential serum biomarkers and
metabonomic profiling of serum in ischemic stroke patients using
UPLC/Q-TOF MS/MS. PLoS ONE 12:e0189009. https:// doi. org/
10. 1371/ journ al. pone. 01890 09
Tiedt S, Brandmaier S, Kollmeier H etal (2020) Circulating
Metabolites Differentiate Acute Ischemic Stroke from Stroke
Mimics. Ann Neurol 88:736–746. https:// doi. org/ 10. 1002/ ana.
25859
Tokarz J, Adamski J, Rizner TL (2020) Metabolomics for Diagnosis
and Prognosis of Uterine Diseases? A Systematic Review. J Pers
Med 10. https:// doi. org/ 10. 3390/ jpm10 040294
Tokarz J, Haid M, Cecil A etal (2017) Endocrinology Meets Metabo-
lomics: Achievements, Pitfalls, and Challenges. Trends Endo-
crinol Metab 28:705–721. https:// doi. org/ 10. 1016/j. tem. 2017.
07. 001
Ussher JR, Elmariah S, Gerszten RE, Dyck JR (2016) The Emerging
Role of Metabolomics in the Diagnosis and Prognosis of Cardio-
vascular Disease. J Am Coll Cardiol 68:2850–2870. https:// doi.
org/ 10. 1016/j. jacc. 2016. 09. 972
Vorkas PA, Shalhoub J, Lewis MR etal (2016) Metabolic Phenotypes
of Carotid Atherosclerotic Plaques Relate to Stroke Risk: An
Exploratory Study. Eur J Vasc Endovasc Surg 52:5–10. https://
doi. org/ 10. 1016/j. ejvs. 2016. 01. 022
Wang D, Kong J, Wu J etal (2017) GC-MS-based metabolomics
identifies an amino acid signature of acute ischemic stroke.
Neurosci Lett 642:7–13. https:// doi. org/ 10. 1016/j. neulet. 2017.
01. 039
Wang Y, Wang Y, Li M etal (2013) (1)H NMR-based metabolomics
exploring biomarkers in rat cerebrospinal fluid after cerebral
ischemia/reperfusion. Mol Biosyst 9:431–439. https:// doi. org/
10. 1039/ c2mb2 5224d
Wang Y, Wang YG, Ma TF etal (2014) Dynamic metabolites profile
of cerebral ischemia/reperfusion revealed by (1)H NMR-based
metabolomics contributes to potential biomarkers. Int J Clin Exp
Pathol 7:4067–4075
Wang Y, Zhao H, Liu Y, etal (2019) GC-MS-Based Metabolomics to
Reveal the Protective Effect of Gross Saponins of Tribulus ter-
restris Fruit against Ischemic Stroke in Rat. Molecules 24. https://
doi. org/ 10. 3390/ molec ules2 40407 93
Wesley UV, Bhute VJ, Hatcher JF etal (2019) Local and systemic
metabolic alterations in brain, plasma, and liver of rats in response
to aging and ischemic stroke, as detected by nuclear magnetic
resonance (NMR) spectroscopy. Neurochem Int 127:113–124.
https:// doi. org/ 10. 1016/j. neuint. 2019. 01. 025
Wishart DS (2019) Metabolomics for Investigating Physiological and
Pathophysiological Processes. Physiol Rev 99:1819–1875. https://
doi. org/ 10. 1152/ physr ev. 00035. 2018
Wu L, Chen C, Li Y etal (2020) UPLC-Q-TOF/MS-Based Serum
Metabolomics Reveals the Anti-Ischemic Stroke Mechanism of
Nuciferine in MCAO Rats. ACS Omega 5:33433–33444. https://
doi. org/ 10. 1021/ acsom ega. 0c053 88
Wu MY, Yiang GT, Liao WT etal (2018) Current Mechanistic Con-
cepts in Ischemia and Reperfusion Injury. Cell Physiol Biochem
46:1650–1667. https:// doi. org/ 10. 1159/ 00048 9241
Yang L, Wang Y, Cai H etal (2020) Application of metabolomics
in the diagnosis of breast cancer: a systematic review. J Cancer
11:2540–2551. https:// doi. org/ 10. 7150/ jca. 37604
Zahoor I, Rui B, Khan J etal (2021) An emerging potential of
metabolomics in multiple sclerosis: a comprehensive over-
view. Cell Mol Life Sci 78:3181–3203. https:// doi. org/ 10. 1007/
s00018- 020- 03733-2
Zhang Y, Li J, Zhang X etal (2021) Advances of Mechanisms-Related
Metabolomics in Parkinson’s Disease. Front Neurosci 15:614251.
https:// doi. org/ 10. 3389/ fnins. 2021. 614251
Zhou B, Xiao JF, Tuli L, Ressom HW (2012) LC-MS-based metabolomics.
Mol Biosyst 8:470–481. https:// doi. org/ 10. 1039/ c1mb0 5350g
Publisher's note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
... Studies in recent past have demonstrated that serum metabolomics analysis can be particularly valuable for probing mitochondrial damage/dysfunction (Demine et al. 2014;Arya et al. 2021). Biofluid samples extracted from various sources such as tissue, blood plasma/serum, and whole organism can be analyzed using metabolomics approaches for discovery of metabolic biomarkers and exploration of pathological mechanism (Gibney et al. 2005, Gupta and Sharma 2021, Wen et al. 2023, Zhang et al. 2022. Recently, the metabolomics studies have also been explored to evaluate the protective as well as therapeutic efficacy of promising treatment strategies including those based on combination of bioactive components/compounds derived from natural sources (Luo et al. 2019, Jia et al. 2021, Wang et al. 2020). ...
Article
Full-text available
Brain stroke (BS, also known as a cerebrovascular accident), represents a serious global health crisis. It has been a leading cause of permanent disability and unfortunately, frequent fatalities due to lack of timely medical intervention. While progress has been made in prevention and management, the complexities and consequences of stroke continue to pose significant challenges, especially, its impact on patient’s quality of life and independence. During stroke, there is a substantial decrease in oxygen supply to the brain leading to alteration of cellular metabolic pathways, including those involved in mitochondrial-damage, leading to mitochondrial-dysfunction. The present proof-of-the-concept metabolomics study has been performed to gain insights into the metabolic pathways altered following a brain stroke and discover new potential targets for timely interventions to mitigate the effects of cellular and mitochondrial damage in BS. The serum metabolic profiles of 108 BS-patients were measured using 800 MHz NMR spectroscopy and compared with 60 age and sex matched normal control (NC) subjects. Compared to NC, the serum levels of glutamate, TCA-cycle intermediates (such as citrate, succinate, etc.), and membrane metabolites (betaine, choline, etc.) were found to be decreased BS patients, whereas those of methionine, mannose, mannitol, phenylalanine, urea, creatine and organic acids (such as 3-hydroxybutyrate and acetone) were found to be elevated in BS patients. These metabolic changes hinted towards hypoxia mediated mitochondrial dysfunction in BS-patients. Further, the area under receiver operating characteristic curve (ROC) values for five metabolic features (methionine, mannitol, phenylalanine, mannose and urea) found to be more than 0.9 suggesting their high sensitivity and specificity for differentiating BS from NC subjects. Graphical abstract
... At the same time, no significant side effects of SHD on other organs were observed, suggesting that SHD is safe at the doses at which it exerts its therapeutic effects. In recent years, a growing number of studies have identified the same signature metabolites from patients with cerebral ischemia and animal models that improve the diagnosis and predict the outcome of IS (32). Gender is an important variable in the prevalence of stroke. ...
Article
Full-text available
San Hua Decoction (SHD) is a traditional four-herbal formula that has long been used to treat stroke. Our study used a traditional pharmacodynamic approach combined with systematic and untargeted metabolomics analyses to further investigate the therapeutic effects and potential mechanisms of SHD on ischemic stroke (IS). Male Sprague-Dawley rats were randomly divided into control, sham-operated, middle cerebral artery occlusion reperfusion (MCAO/R) model and SHD groups. The SHD group was provided with SHD (7.2 g/kg, i.g.) and the other three groups were provided with equal amounts of purified water once a day in the morning for 10 consecutive days. Our results showed that cerebral infarct volumes were reduced in the SHD group compared with the model group. Besides, SHD enhanced the activity of SOD and decreased MDA level in MCAO/R rats. Meanwhile, SHD could ameliorate pathological abnormalities by reducing neuronal damage, improving the structure of damaged neurons and reducing inflammatory cell infiltration. Metabolomic analysis of brain and serum samples with GC - MS techniques revealed 55 differential metabolites between the sham and model groups. Among them, the levels of 12 metabolites were restored after treatment with SHD. Metabolic pathway analysis showed that SHD improved the levels of 12 metabolites related to amino acid metabolism and carbohydrate metabolism, 9 of which were significantly associated with disease. SHD attenuated brain inflammation after ischemia-reperfusion. The mechanisms underlying the therapeutic effects of SHD in MCAO/R rats are related to amino acid and carbohydrate metabolism.
... Blood is the most used sample source for metabolomics identification because it contains numerous detectable metabolites and can be easily obtained in large sample sizes, facilitating the screening of circulating biomarkers for stroke risk [47]. Current metabolomics studies have revealed alterations in the metabolic profiles of stroke patients, with the most reported metabolites being amino acids, lipids, polyamines, and nucleotides [48]. Our study has confirmed the existence of stroke-specific metabolic profiles and identified crucial metabolites and metabolic pathways correlated with the onset of stroke and its related phenotypes. ...
Article
Full-text available
Background Stroke is a common neurological disorder that disproportionately affects middle-aged and elderly individuals, leading to significant disability and mortality. Recently, human blood metabolites have been discovered to be useful in unraveling the underlying biological mechanisms of neurological disorders. Therefore, we aimed to evaluate the causal relationship between human blood metabolites and susceptibility to stroke. Methods Summary data from genome-wide association studies (GWASs) of serum metabolites and stroke and its subtypes were obtained separately. A total of 486 serum metabolites were used as the exposure. Simultaneously, 11 different stroke phenotypes were set as the outcomes, including any stroke (AS), any ischemic stroke (AIS), large artery stroke (LAS), cardioembolic stroke (CES), small vessel stroke (SVS), lacunar stroke (LS), white matter hyperintensities (WMH), intracerebral hemorrhage (ICH), subarachnoid hemorrhage (SAH), transient ischemic attack (TIA), and brain microbleeds (BMB). A two‐sample Mendelian randomization (MR) study was conducted to investigate the causal effects of serum metabolites on stroke and its subtypes. The inverse variance-weighted MR analyses were conducted as causal estimates, accompanied by a series of sensitivity analyses to evaluate the robustness of the results. Furthermore, a reverse MR analysis was conducted to assess the potential for reverse causation. Additionally, metabolic pathway analysis was performed using the web-based MetOrigin. Results After correcting for the false discovery rate (FDR), MR analysis results revealed remarkable causative associations with 25 metabolites. Further sensitivity analyses confirmed that only four causative associations involving three specific metabolites passed all sensitivity tests, namely ADpSGEGDFXAEGGGVR* for AS (OR: 1.599, 95% CI 1.283–1.993, p = 2.92 × 10⁻⁵) and AIS (OR: 1.776, 95% CI 1.380–2.285, p = 8.05 × 10⁻⁶), 1-linoleoylglycerophosph-oethanolamine* for LAS (OR: 0.198, 95% CI 0.091–0.428, p = 3.92 × 10⁻⁵), and gamma-glutamylmethionine* for SAH (OR: 3.251, 95% CI 1.876–5.635, p = 2.66 × 10⁻⁵), thereby demonstrating a high degree of stability. Moreover, eight causative associations involving seven other metabolites passed both sensitivity tests and were considered robust. The association result of one metabolite (glutamate for LAS) was considered non-robust. As for the remaining metabolites, we speculate that they may potentially possess underlying causal relationships. Notably, no common metabolites emerged from the reverse MR analysis. Moreover, after FDR correction, metabolic pathway analysis identified 40 significant pathways across 11 stroke phenotypes. Conclusions The identified metabolites and their associated metabolic pathways are promising circulating metabolic biomarkers, holding potential for their application in stroke screening and preventive strategies within clinical settings.
... 8 Analyses of the pattern and level of metabolites contributed to identifying the disease-related changes, which provided a unique insight into the underlying biological processes. 9,10 In recent years, metabolomics has become an increasingly powerful tool in the identification and validation of biomarkers for various diseases. Methionine (Met) cycle was a complex metabolic pathway with important regulatory functions in cellular processes such as DNA synthesis, methylation, and redox balance. ...
Article
Full-text available
Objective The role of methionine (Met) cycle in the pathogenesis and progression of cardiovascular and cerebrovascular diseases has been established, but its association with moyamoya disease (MMD) has rarely been studied. This study aimed to analyze the levels of Met cycle‐related metabolites and constructed a risk model to explore its association with the risk of MMD. Methods In this prospective study, a total of 302 adult MMD patients and 88 age‐matched healthy individuals were consecutively recruited. The serum levels of Met cycle‐related metabolites were quantified by liquid chromatography‐mass spectrometry (LC–MS). Participants were randomly divided into training set and testing set at a ratio of 1:1. The training set was used to construct the risk score model by LASSO regression. The association between Met cycle‐related risk score and the risk of MMD was analyzed using logistic regression and assessed by ROC curves. The testing set was used for validation. Results The levels of methionine sulfoxide and homocysteine were significantly increased, while the levels of betaine and choline were significantly decreased in MMD and its subtypes compared to healthy controls (p < 0.05 for all). The training set was used to construct the risk model and the risk score of each participant has been calculated. After adjusting for potential confounders, the risk score was independently associated with the risk of MMD and its subtypes (p < 0.05 for all). We then divided the participants into low‐risk and high‐risk groups, the high‐risk score was significantly associated with the risk of MMD and its subtypes (p < 0.05 for all). The risk scores were further assessed as tertiles, the highest tertile was significantly associated with a higher risk of MMD and its subtypes compared to the lowest (p < 0.05 for all). The results were validated in the testing set. Conclusion This study has constructed and validated a risk model based on Met cycle‐related metabolites, which was independently associated with the risk of MMD and its subtypes. The findings provided a new perspective on the risk evaluation and prevention of MMD.
... In recent years, with the further study of multi-omics technologies, researchers have found that the ultimate effect of gene or protein regulatory action was to cause changes in metabolites in body (Holmes et al., 2018;Yang et al., 2018;Zhang et al., 2022). Metabolomics studied endogenous small molecule compounds at the end of metabolic pathways, which could skip the complex regulatory processes of body and directly reflect the final physiological and pathological changes (Ke et al., 2018;Guo et al., 2020;Chumachenko et al., 2022). ...
Article
Full-text available
Objective Methionine sulfoxide (MetO) has been identified as a risk factor for vascular diseases and was considered as an important indicator of oxidative stress. However, the effects of MetO and its association with moyamoya disease (MMD) remained unclear. Therefore, we performed this study to evaluate the association between serum MetO levels and the risk of MMD and its subtypes. Methods We eventually included consecutive 353 MMD patients and 88 healthy controls (HCs) with complete data from September 2020 to December 2021 in our analyzes. Serum levels of MetO were quantified using liquid chromatography-mass spectrometry (LC–MS) analysis. We evaluated the role of MetO in MMD using logistic regression models and confirmed by receiver-operating characteristic (ROC) curves and area under curve (AUC) values. Results We found that the levels of MetO were significantly higher in MMD and its subtypes than in HCs (p < 0.001 for all). After adjusting for traditional risk factors, serum MetO levels were significantly associated with the risk of MMD and its subtypes (p < 0.001 for all). We further divided the MetO levels into low and high groups, and the high MetO level was significantly associated with the risk of MMD and its subtypes (p < 0.05 for all). When MetO levels were assessed as quartiles, we found that the third (Q3) and fourth (Q4) MetO quartiles had a significantly increased risk of MMD compared with the lowest quartile (Q3, OR: 2.323, 95%CI: 1.088–4.959, p = 0.029; Q4, OR: 5.559, 95%CI: 2.088–14.805, p = 0.001). Conclusion In this study, we found that a high level of serum MetO was associated with an increased risk of MMD and its subtypes. Our study raised a novel perspective on the pathogenesis of MMD and suggested potential therapeutic targets.
... Interestingly, decreased valine and isoleucine have been observed in IS as well (28,32). Interestingly, the differential changes in circulating BCAA may relate to the nutritional condition of IS patients in response to anabolic and catabolic alterations or metabolic comorbidities, such as diabetes, hypertension, hypercholesterolemia, or cardiovascular disease (34). Ornithine and citrulline, two metabolites linked to the arginine production pathway, can also exhibit dysregulation in IS patients. ...
Article
Full-text available
Background The pathophysiological processes linked to an acute ischemic stroke (IS) can be reflected in the circulating metabolome. Amino acids (AAs) have been demonstrated to be one of the most significant metabolites that can undergo significant alteration after a stroke. Methods We sought to identify the potential biomarkers for the early detection of IS using an extensive targeted technique for reliable quantification of 27 different AAs based on ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). A cohort with 216 participants was enrolled, including 70 mild to moderate ischemic stroke patients (National Institutes of Health Stroke Scale < 15, MB group), 76 stroke mimics (MM group) and 70 healthy controls (NC group). Results It was found that upon comparing MB and MM to control patients, AAs shifts were detected via partial least squares discrimination analysis (PLS-DA) and pathway analysis. Interestingly, MB and MM exhibited similar AAs pattern. Moreover, ornithine, asparagine, valine, citrulline, and cysteine were identified for inclusion in a biomarker panel for early-stage stroke detection based upon an AUC of 0.968 (95% CI 0.924–0.998). Levels of ornithine were positively associated with infract volume, 3 months mRS score, and National Institutes of Health Stroke Scale (NIHSS) score in MB. In addition, a metabolites biomarker panel, including ornithine, taurine, phenylalanine, citrulline, cysteine, yielded an AUC of 0.99 (95% CI 0.966–1) which can be employed to effectively discriminate MM patients from control. Conclusion Overall, alternations in serum AAs are characteristic metabolic features of MB and MM. AAs could serve as promising biomarkers for the early diagnosis of MB patients since mild to moderate IS patients were enrolled in the study. The metabolism of AAs can be considered as a key indicator for both the prevention and treatment of IS.
... Metabolomics is mainly focused on small molecule metabolites of molecular weight below 1000 (Wishart, 2019). Metabolomics technology has developed rapidly in recent years, and researchers can use it to detect and analyze biomarkers that are closely related to diseases, which can then be used to diagnose diseases, determine the course of diseases and develop treatment options, so it plays a unique advantage in various elds of research (Newgard, 2017;Patel et al., 2022;Zhang et al., 2022). The pathogenesis of depression is complex and involves the alteration of multiple system functions (Peng et al., 2015;Suneson et al., 2021), so the antidepressant mechanism of exercise cannot be comprehensively explained from a single target. ...
Preprint
Full-text available
Objective To study the effects of different types of exercise on the plasma metabolomics of chronic unpredictable mild stress (CUMS)-induced depressed rats based on ¹H-NMR metabolomics techniques, and to explore the potential mechanisms of exercise for the treatment of depression. Methods Rats were randomly divided into blank control group (C), CUMS control group (D), pre-exercise group (P), aerobic exercise group (A), resistance exercise group (R), and aerobic + resistance exercise group (AR). The corresponding protocol intervention was applied to each group of rats. Body weight, sucrose preference and open field tests were performed weekly during the experiment to evaluate the extent of depression in rats. Plasma samples from each group of rats were collected at the end of the experiment, and then the plasma was analyzed by ¹H-NMR metabolomics combined with multivariate statistical analysis methods to identify differential metabolites and perform metabolic pathway analysis. Results (1) Compared with the group D, the body weight, sucrose preference rate, and the number of crossings and standings in the different types of exercise groups were significantly improved (p < 0.05 or p < 0.01). (2) Compared to group C, a total of 15 differential metabolites associated with depression were screened in the plasma of rats in group D, involving 6 metabolic pathways. Group P can regulate the levels of 6 metabolites: valine, lactate, inositol, glucose, phosphocreatine, acetoacetic acid. Group A can regulate the levels of 6 metabolites: N-acetylglycoprotein, leucine, lactate, low density lipoprotein, glucose, acetoacetic acid. Group R can regulate the levels of 6 metabolites: choline, lactate, inositol, glucose, phosphocreatine, acetoacetic acid. Group AR can regulate the levels of 5 metabolites: choline, citric acid, glucose, acetone, acetoacetic acid. Conclusion The different types of exercise groups can improve the depressive symptoms in CUMS rats, and there are common metabolites and metabolic pathways for their mechanism of effects. This study provides a powerful analytical tool to study the mechanism of the antidepressant effect of exercise, and provides an important method and basis for the early diagnosis, prevention and treatment of depression.
Article
Full-text available
Background: Ischemic stroke (IS) is a major cause of death and disability worldwide. Previous studies have reported associations between metabolic disorders and IS. However, evidence regarding the causal relationship between blood metabolites and IS lacking. Methods: A two-sample Mendelian randomization analysis (MR) was used to assess the causal relationship between 1,400 serum metabolites and IS. The inverse variance-weighted (IVW) method was employed to estimate the causal effect between exposure and outcome. Additionally, MR-Egger regression, weighted median, simple mode, and weighted mode approaches were employed as supplementary comprehensive evaluations of the causal effects between blood metabolites and IS. Tests for pleiotropy and heterogeneity were conducted. Results: After rigorous selection, 23 known and 5 unknown metabolites were identified to be associated with IS. Among the 23 known metabolites, 13 showed significant causal effects with IS based on 2 MR methods, including 5-acetylamino-6-formylamino-3-methyluracil, 1-ribosyl-imidazoleacetate, Behenoylcarnitine (C22), N-acetyltyrosine, and N-acetylputrescine to (N (1) + N (8))-acetate,these five metabolites were positively associated with increased IS risk. Xanthurenate, Glycosyl-N-tricosanoyl-sphingadienine, Orotate, Bilirubin (E,E), Bilirubin degradation product, C17H18N2O, Bilirubin (Z,Z) to androsterone glucuronide, Bilirubin (Z,Z) to etiocholanolone glucuronide, Biliverdin, and Uridine to pseudouridine ratio were associated with decreased IS risk. Conclusion: Among 1,400 blood metabolites, this study identified 23 known metabolites that are significantly associated with IS risk, with 13 being more prominent. The integration of genomics and metabolomics provides important insights for the screening and prevention of IS.
Article
Full-text available
Ischemic stroke (IS) is a multifactorial and heterogeneous disease. Despite years of studies, effective strategies for the diagnosis, management and treatment of stroke are still lacking in clinical practice. Metabolomics is a growing field in systems biology. It is starting to show promise in the identification of biomarkers and in the use of pharmacometabolomics to help patients with certain disorders choose their course of treatment. The development of metabolomics has enabled further and more biological applications. Particularly, metabolomics is increasingly being used to diagnose diseases, discover new drug targets, elucidate mechanisms, and monitor therapeutic outcomes and its potential effect on precision medicine. In this review, we reviewed some recent advances in the study of metabolomics as well as how metabolomics might be used to identify novel biomarkers and understand the mechanisms of IS. Then, the use of metabolomics approaches to investigate the molecular processes and active ingredients of Chinese herbal formulations with anti-IS capabilities is summarized. We finally summarized recent developments in single cell metabolomics for exploring the metabolic profiles of single cells. Although the field is relatively young, the development of single cell metabolomics promises to provide a powerful tool for unraveling the pathogenesis of IS.
Article
Full-text available
Ischemic stroke (IS) is a major cause of mortality and disability worldwide. However, the pathogenesis of IS remains unknown, and methods for early prediction and diagnosis of IS are lacking. Metabolomics can be applied to biomarker discovery and mechanism exploration of IS by exploring metabolic alterations. In this review, 62 IS metabolomics studies in the murine model published from January 2006 to December 2020 in the PubMed and Web of Science databases were systematically reviewed. Twenty metabolites (e.g., lysine, phenylalanine, methionine, tryptophan, leucine, lactate, serine, N-acetyl-aspartic acid, and glutathione) were reported consistently in more than two-third murine studies. The disturbance of metabolic pathways, such as arginine biosynthesis; alanine, aspartate and glutamate metabolism; aminoacyl-tRNA biosynthesis; and citrate cycle, may be implicated in the development of IS by influencing the biological processes such as energy failure, oxidative stress, apoptosis, and glutamate toxicity. The transient middle cerebral artery occlusion model and permanent middle cerebral artery occlusion model exhibit both common and distinct metabolic patterns. Furthermore, five metabolites (proline, serine, LysoPC (16:0), uric acid, glutamate) in the blood sample and 7 metabolic pathways (e.g., alanine, aspartate, and glutamate metabolism) are shared in animal and clinical studies. The potential biomarkers and related pathways of IS in the murine model may facilitate the biomarker discovery for early diagnosis of IS and the development of novel therapeutic targets.
Article
Full-text available
The effects of current treatment strategies used in ischemic stroke are weakened by cerebral ischemia-reperfusion (CIR) injury. Suitable treatment regimens targeting CIR injury are still lacking. Two herbs, namely, Acanthopanax senticosus (Rupr. & Maxim.) Harms (ASE) and Gastrodia elata Blume (GEB), have been used as traditional Chinese medicine and are indicated in the treatment of stroke and cerebrovascular diseases. However, there are no studies that report the effects of ASE combined with GEB in the treatment of CIR injury. In this study, we used the Zea Longa method to induce CIR injury in male Wistar rats. Results of the pharmacodynamic studies revealed that co-administration of ASE and GEB may improve neuronal injury and prevent neuronal apoptosis by reducing oxidative stress and inflammation, and also help prevent CIR injury. On the basis of our hypothesis, we combined the results from transcriptomic and metabonomic analyses and found that ASE and GEB could prevent CIR injury by targeting phenylalanine, pyrimidine, methionine, and sphingolipid metabolism. Therefore, our study provides the basis for the compatibility and efficacy of ASE and GEB.
Article
Full-text available
Parkinson’s disease (PD) is a multifactorial disorder characterized by progressively debilitating dopaminergic neurodegeneration in the substantia nigra and the striatum, along with various metabolic dysfunctions and molecular abnormalities. Metabolomics is an emerging study and has been demonstrated to play important roles in describing complex human diseases by integrating endogenous and exogenous sources of alterations. Recently, an increasing amount of research has shown that metabolomics profiling holds great promise in providing unique insights into molecular pathogenesis and could be helpful in identifying candidate biomarkers for clinical detection and therapies of PD. In this review, we briefly summarize recent findings and analyze the application of molecular metabolomics in familial and sporadic PD from genetic mutations, mitochondrial dysfunction, and dysbacteriosis. We also review metabolic biomarkers to assess the functional stage and improve therapeutic strategies to postpone or hinder the disease progression.
Article
Full-text available
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the nervous system that primarily affects young adults. Although the exact etiology of the disease remains obscure, it is clear that alterations in the metabolome contribute to this process. As such, defining a reliable and disease-specific metabolome has tremendous potential as a diagnostic and therapeutic strategy for MS. Here, we provide an overview of studies aimed at identifying the role of metabolomics in MS. These offer new insights into disease pathophysiology and the contributions of metabolic pathways to this process, identify unique markers indicative of treatment responses, and demonstrate the therapeutic effects of drug-like metabolites in cellular and animal models of MS. By and large, the commonly perturbed pathways in MS and its preclinical model include lipid metabolism involving alpha-linoleic acid pathway, nucleotide metabolism, amino acid metabolism, tricarboxylic acid cycle, d -ornithine and d -arginine pathways with collective role in signaling and energy supply. The metabolomics studies suggest that metabolic profiling of MS patient samples may uncover biomarkers that will advance our understanding of disease pathogenesis and progression, reduce delays and mistakes in diagnosis, monitor the course of disease, and detect better drug targets, all of which will improve early therapeutic interventions and improve evaluation of response to these treatments.
Article
Full-text available
Nuciferine is an aporphine alkaloid monomer that is extracted from the leaves of the lotus species Nymphaea caerulea and Nelumbo nucifera Gaertn. Nuciferine was reported to treat cerebrovascular diseases. However, the potential mechanism of the neuroprotective effects of nuciferine at the metabolomics level is still not unclear. The present research used neurological score, infarct volume, cerebral water content, and ultraperformance liquid chromatography to quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS)-based serum metabolomics to elucidate the anti-ischemic stroke effect and mechanisms of nuciferine. The results showed that nuciferine significantly improved neurological deficit scores and ameliorated cerebral edema and infarction. Multivariate data analysis methods were used to examine the differences in serum endogenous metabolism between groups, and the biomarkers of nuciferine on ischemic stroke were identified. Approximately 19 metabolites and 7 metabolic pathways associated with nuciferine on treatment of stroke were found, which indicated that nuciferine exerted a positive therapeutic action on cerebral ischemic by regulating metabolism. These results provided some data support for the study of anti-stroke effect of nuciferine from the perspective of metabolomics.
Article
Full-text available
This systematic review analyses the contribution of metabolomics to the identification of diagnostic and prognostic biomarkers for uterine diseases. These diseases are diagnosed invasively, which entails delayed treatment and a worse clinical outcome. New options for diagnosis and prognosis are needed. PubMed, OVID, and Scopus were searched for research papers on metabolomics in physiological fluids and tissues from patients with uterine diseases. The search identified 484 records. Based on inclusion and exclusion criteria, 44 studies were included into the review. Relevant data were extracted following the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) checklist and quality was assessed using the QUADOMICS tool. The selected metabolomics studies analysed plasma, serum, urine, peritoneal, endometrial, and cervico-vaginal fluid, ectopic/eutopic endometrium, and cervical tissue. In endometriosis, diagnostic models discriminated patients from healthy and infertile controls. In cervical cancer, diagnostic algorithms discriminated patients from controls, patients with good/bad prognosis, and with/without response to chemotherapy. In endometrial cancer, several models stratified patients from controls and recurrent from non-recurrent patients. Metabolomics is valuable for constructing diagnostic models. However, the majority of studies were in the discovery phase and require additional research to select reliable biomarkers for validation and translation into clinical practice. This review identifies bottlenecks that currently prevent the translation of these findings into clinical practice.
Article
Full-text available
Despite the fact that glucose is the main fuel of the brain, hyperglycemia at hospital admission is generally associated with a poor functional outcome in stroke patients. This paradox may be explained by the lack of information about the blood glucose level at stroke onset. Here, we analyzed the metabolome of blood cells entrapped in cerebral thrombi to gain insight into their metabolism at stroke onset. Fourty-one consecutive stroke patients completely recanalized by mechanical thrombectomy within 6 h were included. The metabolome of retrieved thrombi was analyzed by liquid chromatography tandem with mass spectrometry. Discriminant Analysis (sparse Partial Least Squares Discriminant Analysis (sPLS-DA)) was performed to identify classification models and significant associated features of favorable clinical outcome at 3 months (modified Rankin Scale (mRS) < 2). sPLS-DA of the metabolomes of cerebral thrombi discriminated between stroke patients with a favorable or poor clinical outcome (Area Under the Curve (AUC) = 0.992 (0.931-1)). In addition, our results revealed that high sorbitol and glucose levels in the thrombi positively correlated with favorable clinical outcomes. Sorbitol, a short-term glycemic index reflecting a high blood glucose level at stroke onset, was found to be an independent predictor of good outcome (AUC = 0.908 (0.807-0.995)). This study demonstrates that a high blood glucose level at stroke onset is beneficial to the clinical outcome of the patient.
Article
Stroke is a major reason for disability and the second highest cause of death in the world. When a patient is admitted to a hospital, it is necessary to identify the type of stroke, and the likelihood for development of a recurrent stroke, vascular dementia, and depression. These factors could be determined using different biomarkers. Metabolomics is a very promising strategy for identification of biomarkers. The advantage of metabolomics, in contrast to other analytical techniques, resides in providing low molecular weight metabolite profiles, rather than individual molecule profiles. Technically, this approach is based on mass spectrometry and nuclear magnetic resonance. Furthermore, variations in metabolite concentrations during brain ischemia could alter the principal neuronal functions. Different markers associated with ischemic stroke in the brain have been identified including those contributing to risk, acute onset, and severity of this pathology. In the brain, experimental studies using the ischemia/reperfusion model (IRI) have shown an impaired energy and amino acid metabolism and confirmed their principal roles. Literature data provide a good basis for identifying markers of ischemic stroke and hemorrhagic stroke and understanding metabolic mechanisms of these diseases. This opens an avenue for the successful use of identified markers along with metabolomics technologies to develop fast and reliable diagnostic tools for ischemic and hemorrhagic stroke.
Article
Cell death is a process consequential to cerebral ischemia and cerebral ischemia/reperfusion (I/R) injury. Recent evidence suggest that necroptosis has been involved in the pathogenesis of ischemic brain injury. The mechanism of necroptosis is initiated by an activation of inflammatory receptors including tumor necrosis factor, toll like receptor, and fas ligands. The signals activate the receptor-interacting protein kinase (RIPK) 1, 3, and a mixed-lineage kinase domain-like pseudokinase (MLKL) to instigate necroptosis. RIPK1 inhibitor, necrostatin-1, was developed, and dramatically reduced brain injury following cerebral ischemia in mice. Consequently, necroptosis could be a novel therapeutic target for stroke, which aims to reduce long-term adverse outcomes after cerebral ischemia. Several studies have been conducted to test the roles of necroptosis on cerebral ischemia and cerebral I/R injury, and the efficacy of necrostatin-1 has been tested in those models. Evidence regarding the roles of necroptosis and the effects of necrostatin-1, from in vitro and in vivo studies, has been summarized and discussed. In addition, other therapeutic managements, involving in necroptosis, are also included in this review. We believe that the insights from this review might clarify the clinical perspective and challenges involved in future stroke treatment by targeting the necroptosis pathway.
Article
Osteoarthritis (OA) represents the most prevalent and disabling arthritis worldwide due to its heterogeneous and progressive articular degradation. However, effective and timely diagnosis and fundamental treatment for this disorder are lacking. Metabolomics, a growing field in life science research in recent years, has the potential to detect many metabolites and thus explains the underlying pathophysiological processes. Hence, new specific metabolic markers and related metabolic pathways can be identified for OA. In this review, we aimed to provide an overview of studies related to the metabolomics of OA in animal models and humans to describe the metabolic changes and related pathways for OA. The present metabolomics studies reveal that the pathogenesis of OA may be significantly related to perturbations of amino acid metabolism. These altered amino acids (e.g., branched-chain amino acids, arginine, and alanine), as well as phospholipids, were identified as potential biomarkers to distinguish patients with OA from healthy individuals.