You are on page 1of 10

Journal of Neuroimmunology 354 (2021) 577526

Contents lists available at ScienceDirect

Journal of Neuroimmunology
journal homepage: www.elsevier.com/locate/jneuroim

Gut microbial metabolite TMAO portends prognosis in acute


ischemic stroke
Jianli Zhang a, 1, *, Liankun Wang b, 1, Jinle Cai a, Aidi Lei a, Caiwen Liu a, Ruidian Lin a, Li Jia c, **,
Yingzi Fu d, ***
a
Department of Neurology, Xiamen Fifth Hospital, Xiamen, China
b
Department of Neuroelectrophysiology, Heilongjiang Province Hospital, Harbin, China
c
Center of Health Laboratory Technology, Public Health Department of Harbin Medical University, Harbin, China
d
Department of Neurology, Heilongjiang Province Hospital, Harbin, China

A R T I C L E I N F O A B S T R A C T

Keywords: Background: Over the recent years, the role of trimethylamine N-oxide (TMAO) as a gut-derived metabolite
Trimethylamine N-oxide mediating cardiovascular disease pathogenesis has been under particularly intense scrutiny. The aim was to
Gut microbial metabolism explore whether TMAO levels were associated with clinical severity or functional outcome in Chinese patients
Ischemic stroke
with ischemic stroke.
Prognosis
Mortality
Methods: This is a single-center, prospective cohort study from Xiamen, China. We examined the relationship
between fasting TMAO and 2 of its nutrient precursors, choline and betaine, vs. 3-month functional outcome and
mortality in 351 first-ever patients with acute ischemic stroke.
Results: The median value of the plasma level of TMAO was 6.1 μM (IQR, 3.7–9.9 μM), which was higher than in
those control cases (4.0; 2.4–5.9 μM). Patients with a poor outcome and nonsurvivors had significantly increased
TMAO levels on admission (P < 0.001). Following adjustments for traditional risk factors, increased TMAO
concentrations remained predictive of both poor outcome and mortality risks in stroke patients [e.g., quartiles 4
vs 1, odd ratio 5.65 (95% CI, 2.87–13.45), P < 0.001; and 5.84 (95% CI, 3.05–16.12), P < 0.001, respectively]. In
multivariate analysis, TMAO was an independent predictor of functional outcome and mortality and improved
the prognostic accuracy of the NIHSS to predict functional outcome (combined areas under the curve, 0.82; 95%
CI 0.77–0.89, P = 0.003) and mortality (combined areas under the curve, 0.85; 95% CI: 0.80–0.90, P = 0.002).
Conclusions: Fasting plasma concentrations of gut microbial TMAO are higher in patients with ischemic stroke
and portend higher poor functional outcome events and mortality.

1. Introduction microbiota to cardiometabolic diseases with mechanistic links to gut


microbial choline metabolism (Tang et al., 2017). Trimethylamine-N-
China has 2.5 million new stroke cases and 1.6 million stroke mor­ oxide (TMAO) is a circulating organic compound produced by dietary
tality each year, and 7.5 million stroke survivors, which has exceeded L-carnitine and choline metabolism, which was recently found to induce
heart disease to become the leading cause of death and adult disability atherosclerosis in rodents (Stubbs et al., 2016) directly. Intestinal bac­
(Tu et al., 2017). A previous study suggested that in 2016, the global teria metabolize both L-carnitine and choline to trimethylamine, a
lifetime risk of stroke from the age of 25 years onward was approxi­ metabolite that is absorbed from the intestine and subsequently oxidized
mately 25%, and China had the highest estimated risk (39.3%; 95% via hepatic flavin monooxygenase enzymes to form TMAO (LangDH
uncertainty interval, 37.5 to 41.1) (The GBD, 2016). et al., 1998). Wang et al. (Wang et al., 2011) reported that plasma
Recent studies indicate a pathophysiological contribution of gut concentrations of metabolites of dietary phosphatidylcholine, including

* Correspondence to: J. Zhang, No.101, Min’an Road, Xiang’an District, Xiamen City, Fujian Province, China.
** Correspondence to: L. Jia, No.157, Baojian Road, Nangang District, Harbin City, Heilongjiang Province, China.
*** Correspondence to: Y. Fu, Guo Ge Li street 405, Nangang District, Harbin, Heilongjiang Province, China.
E-mail addresses: zhangjianli1223@163.com (J. Zhang), jiali1013@163.com (L. Jia), yingzibest@126.com (Y. Fu).
1
Jianli Zhang and Liankun Wang contributed equally to this work as co-first authors.

https://doi.org/10.1016/j.jneuroim.2021.577526
Received 15 October 2020; Received in revised form 17 February 2021; Accepted 17 February 2021
Available online 20 February 2021
0165-5728/© 2021 Elsevier B.V. All rights reserved.
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

choline, betaine, and TMAO, were associated with atherosclerotic car­ Stroke Treatment (TOAST) criteria: (1) large-artery arteriosclerosis; (2)
diovascular risks in patients. In animal models, they also confirmed that cardioembolism; (3) small-artery occlusion; (4) the other causative
those biomarkers mechanistically linked to atherosclerosis development factor, and (5) undetermined causative factor (Adams Jr et al., 1993).
(Wang et al., 2011). Functional impairment was assessed at 3-month using the modified
Elevated TMAO levels have been shown to predict future risk of Rankin scale (mRS), and a good functional outcome was defined as mRS
major adverse cardiac events (MACE), an increased prevalence of car­ of 0–2 points, while a poor outcome was defined as 3–6 points (Bonita
diovascular diseases (CVD), and have shown a relationship with the and Beaglehole, 1988). The secondary end-point was all-cause mortality
number of diseased coronary vessels (Wang et al., 2011; Tang et al., within 3-month. The diagnosis of stroke was based on the results of a
2013; Koeth et al., 2014). A meta-analysis found that higher circulating strict neurological examination by magnetic resonance imaging (MRI).
TMAO may independently predict the risk of subsequent cardiovascular The infarct volume was calculated using the formula 0.5 × a × b × c
events and mortality (Qi et al., 2018). Interestingly, a higher plasma (where a is longest dimension in axis x, b is longest perpendicular
TMAO level was suggested to be associated with worse long-term sur­ dimension to axis x (y), and c is total length in z dimension) (Sims et al.,
vival in patients with chronic kidney disease (Tang et al., 2015) and with 2009).
heart failure (Tang et al., 2014; Troseid et al., 2015).
Furthermore, one study demonstrated a graded relation between 2.3. Blood collection and quantification
TMAO levels and the risk of subsequent cardiovascular events in patients
with recent prior ischemic stroke (Haghikia et al., 2018). Another study The plasma of all patients was collected under fasting at 8:00 on the
proposed that higher TMAO levels were associated with increased risk of first morning of admission for TMAO determination (within 0–6 [n =
the first stroke in hypertensive patients (Nie et al., 2018). Also, TMAO 78], 6–12 [n = 95], 12–24 [n = 88], and 24–48 [n = 90] hours from
was an independent predictor of ischemic stroke, potentially refining symptom onset). Plasma blood samples were collected using EDTA tubes
stroke stratification in patients with atrial fibrillation (Liang et al., and immediately processed and frozen at − 80 ◦ C until analysis. TMAO,
2019). Shafi et al. (Shafi et al., 2017) reported that TMAO had a linear choline, and betaine levels in plasma were determined using stable
increase in risk with cardiovascular mortality in white hemodialysis isotope dilution high-performance liquid chromatography with online
patients, but not in black patients, suggesting the role of TMAO might electrospray ionization tandem mass spectrometry (LC/MS/MS) on an
differ by race and ethnic groups. However, most prior studies came from AB SCIEX 5500 triple quadrupole mass spectrometer using d9-(tri­
Western countries. Research on China populations, where dietary habits methyl)-labelled internal standards as described previously (Stubbs
are not exactly the same as those of Western countries, is limited. Thus, et al., 2016). TMAO and d9-TMAO were monitored in the positive
in this study, the aim was to explore whether TMAO levels were asso­ multiple reaction monitoring mass spectrometry mode using charac­
ciated with clinical severity or functional outcome in Chinese patients teristic precursor–production transitions, including m/z 76/58 and m/z
with ischemic stroke. 85/66, respectively. To calculate the TMAO concentration, various
TMAO standards were added to control plasma to prepare the calibra­
2. Patients and methods tion curves. The assay described shows good inter- and intraday repro­
ducibility (all CVs < 6.0%), accuracy (>98.5% across low, mid, and high
2.1. Patients values). Other biomarkers were also tested by standard assays, such as
serum levels of C-reactive protein (CRP), glucose, and interleukin-6 (IL-
This was a prospective cohort study at one hospital in Fujian (Xiamen 6). An estimated glomerular filtration rate (eGFR; in mL/min per 1.73
Fifth Hospital), China. During March 2017–June 2018, consecutive m2) was calculated using the Modification of Diet in Renal Disease study
patients with first-ever ischemic stroke (symptoms onset was less than equation (Levey et al., 1999).
24 h) were included. Ischemic stroke was diagnosed and identified ac­
cording to World Health Organization recommendations. Exclusion 2.4. Statistical analyses
criteria were: (1) malignant tumor; (2) metabolic syndrome; (3) renal
and liver insufficiency; (4) any surgical procedure within the previous The results of the categorical variable (percentage) and continuous
month; (5) inability to consent (e.g., dementia); (6) Other neurological variable (median value [interquartile ranges, IQRs]) were described.
diseases (such as intracerebral hemorrhage, cerebral hemorrhage, Par­ Chi-square and Mann-Whitney U tests were applied for comparing
kinson’s disease, and Alzheimer’s disease) and (7) presence of cardio­ groups. Bivariate correlation was analyzed using Spearman’s rank
genic shock, sepsis, pneumonia; acute coronary syndromes (ACS). correlation.
One hundred and fifty age and gender-matched healthy volunteers The relationship between TMAO level and clinical severity (dichot­
from our hospital medical center were assigned to as the healthy control omized as NIHSS<6 and NIHSS≥6) (Daubail et al., 2013) or functional
group. All control subjects were also clinically examined by a neurolo­ outcomes (mortality) was assessed by logistic regression. The results
gist (not an author) to exclude any sub-clinical stroke features. The were described as odds ratios (OR) with 95% confidence intervals (CI).
median age of those control cases was 66 (IQR, 55–74) years, and 50% Adjustments were made for individual traditional risk factors including
were men. age, sex, BMI, traditional risk factors, NIHSS and Infarct volume at
admission, stroke subtype, pre-stroke treatment, acute stroke treatment,
2.2. Clinical variables and laboratory testing eGFR, plasma levels of glucose, CRP, IL-6, Choline, Betaine, and TMAO.
We also divided plasma TMAO levels into quartiles.
At baseline, demographic data (age and sex), body mass index (BMI), The accuracy of the TMAO level for predicting the outcome of stroke
and history of conventional vascular risk factors (smoking, hyperten­ was evaluated with Receiver operating characteristic (ROC) curves, and
sion, type 2 diabetes mellitus, hypercholesterolemia, atrial fibrillation, results were calculated with the Area under the curve (AUC). The clin­
coronary heart disease, previous myocardial infarction, peripheral ical value of adding TMAO to the existing risk factors for predicting the
vascular disease, and transient ischemic attack) were collected. Pre- outcome of stroke was calculated with the integrated discrimination
stroke therapy, including oral anticoagulants, antiplatelet agents, anti­ improvement (IDI) index and net reclassification improvement (NRI)
hypertensive treatment, and statins, as well as acute treatment (IV index (Pencina et al., 2011). Furthermore, to study the ability of TMAO
thrombolysis and/or mechanical thrombectomy), was also recorded. for mortality prediction, we calculated Kaplan–Meier survival curves
National Institutes of Health Stroke Scale (NIHSS) was used to and stratified patients by TMAO cut-off, which had been defined by the
evaluate clinical severity at admission by a neurologist (Tu et al., 2014). ROC curve. All statistics were analyzed with SPSS software. The ROC R
Ischemic stroke was classified according to Trial of Org 10,172 in Acute package (version 1.0–2) and GraphPad Prism 5.0 were also applied for

2
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

plotting. P < 0.05 was defined as statistical significance. 4 (IQR, 3–6). Forty-seven patients died, and the mortality rate was
13.4% (95%CI:9.8%–17.0%). The general information of the stroke
2.5. Ethics patients was presented (Table 1).

The study’s design was reviewed and approves by the investigational 3.2. Main results
review board of the Xiamen Fifth Hospital, according to the principles of
the Declaration of Helsinki. Informed consent was obtained from pa­ 3.2.1. TMAO and severity of stroke
tients or their relatives (patients unable to communicate) prior to their As a continuous variable, a modest correlation was obtained between
inclusion in this study. NIHSS score and plasma TMAO level (r = 0.413; P < 0.001). At
admission, there were 184 minor strokes (52.4%, NIHSS<6). The me­
3. Results dian TMAO levels in those patients were lower compared to those of
patients with moderate-to-high stroke (4.9 μM [IQR, 2.9–6.6] vs. 8.6 μM
3.1. Patients [5.2–11.6], Z = 7.521; P < 0.001). In univariate and multivariable
models, when TMAO was increased by each 1 μM, the unadjusted and
From 503 screened patients with suspected first-ever ischemic adjusted (variables including age, sex, BMI, traditional risk factors,
stroke, acute ischemic stroke was diagnosed in 388 patients, and 351 infarct volume at admission, stroke subtype, pre-stroke treatment, eGFR,
completed 3-month follow-up and were finally included (Fig. 1). plasma levels of glucose, CRP, IL-6, Choline, and Betaine) risk of
The median value of the plasma level of TMAO was 6.1 μM (IQR, moderate-to-high stroke were increased by 21% (OR = 1.21 [95%CI:
3.7–9.9 μM), which was higher than in those control cases (4.0; 2.4–5.9 1.13–1.30], P < 0.001) and 10% (1.10 [1.03–1.21], P = 0.005),
μM), Fig. 2A. The included patients’ median age was 66 (IQR, 55–74) respectively. Also, TMAO levels paralleled lesion size. There was a
years, and 50.4% were men. On admission, the median NIHSS score and positive correlation between infarct volume and plasma TMAO level (r
infarct volumes were 5 (IQR, 3–12) points and 15.5(6.2–24.1) ml, = 0.244; P < 0.001). Furthermore, correlation analysis showed that
respectively. A total 49 patients (14.0%) received IV thrombolysis and/ plasma TMAO level positively corrected with age, IL-6, Choline, and
or mechanical thrombectomy. In these patients, the median TMAO level Betaine, while negative with eGFR (P < 0.05, all). Patients of the male
was lower than that observed in other patients (4.4 μM [IQ, 2.6–7.3] vs. sex, older age, suffered from diabetes, and received glucose-lowing
6.3 μM [4.0–10.1], P = 0.003). In addition, 39 out of those 49 patients drugs were more likely to had higher levels of TMAO(p < 0.05).
who received IV thrombolysis and/or mechanical thrombectomy were
successfully reperfused after thrombectomy. In these patients, the me­ 3.2.2. Copeptin and functional outcome after 3 months
dian TMAO level was no different from that of non-reperfused patients TMAO plasma levels in patients with a poor outcome were signifi­
(4.4 μM [IQ, 2.5–7.5] vs. 4.4 μM [2.8–6.9], P = 0.92). A poor functional cantly higher than those in patients with a good outcome (5.0 [IQR,
outcome was found in 117 patients (33.3%) with a median mRS score of 3.0–7.0] vs. 9.4 [IQR, 6.2–12.2] μM; P < 0.001; Fig. 2B). In univariate

Fig. 1. Study profile/flow sheet of the study.

3
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

Fig. 2. Plasma levels of TMAO in different


groups. (A) Plasma levels of TMAO in pa­
tients with ischemic stroke and controls. (B)
Plasma levels of TMAO in patients with a
good outcome and poor outcome. A good
functional outcome was defined as an mRS
score of 0 to 2 points, while poor outcome
was defined as 3–6 points. (C) TMAO levels
in survivors and nonsurvivors of stroke. All
data are medians and interquartile ranges
(IQR). P values refer to Mann-Whitney U
tests for differences between groups. TMAO,
Trimethylamine N-oxide.

logistic regression analysis, with an unadjusted OR of 1.35 (95% CI, 3.2.3. TMAO and death within 3 months
1.25–1.46), TMAO is strongly associated with poor functional outcomes. Copeptin plasma levels in 47 patients who died were nearly two
After adjusting for all other significant outcome predictors, the OR times as compared with patients who survived (10.6 [IQR, 8.5–13.2] vs.
adjusted for all other risk factors was 1.21 (95% CI, 1.07–1.35), sug­ 5.8 [IQR, 3.4–8.5] μM; p < 0.001; see Fig. 2C). In univariate and
gesting that TMAO was still an independent factor for predicting func­ multivariate logistic regression analysis models, TMAO was correlated
tional outcome, Table 2. As shown in Table 2, age, stroke severity, large- with all-cause mortality, and the mortality risk was increased by 36%
vessel occlusive, IL-6, and acute treatment remained significant outcome (OR 1.36, 95% CI: 1.23–1.49) 24% (1.24, 1.06–1.38), respectively. As
predictors, unlike all others assessed. Interestingly, Choline and Betaine shown in Table 2, age, stroke severity, IL-6, and acute treatment
were outcome predictors in univariate analysis. However, this trend remained significant mortality predictors, unlike all others assessed.
disappeared in the multivariable models. Again, Choline and Betaine were mortality predictors in univariate
The poor outcome was distributed across TMAO quartiles, from analysis. However, this trend disappeared in the multivariable models.
11.2% (first quartile, Q1) to 62.1% (fourth quartile, Q4). In the analysis All-cause mortality was distributed across TMAO quartiles, between
of multivariable models, the second, third, and fourth quartiles (Q2, Q3, 3.4% (Q1) to 32.2% (Q4). In multivariable models, Q2, Q3, and Q4 of
and Q4) of TMAO levels were compared against Q1 (Fig. 3). The results TMAO plasma levels were compared against Q1 (Fig. 5). The results
showed that TMAO levels in Q3 and Q4 were correlated with functional suggested that TMAO levels in Q4 were correlated with all-cause mor­
outcome, and the poor outcome risk was increased by 202% (OR 3.02, tality, and the risk was increased by 484% (OR 5.84, 95% CI
95% CI 1.34–6.12) and 465% (5.65, 2.87–13.45), respectively (Fig. 3). 3.05–16.12). The results showed that TMAO levels in Q2 and Q3 were
The independence of the correlation between TMAO and functional not correlated with mortality, and the OR were 2.29(0.83–6.03) and
outcome was tested by the likelihood ratio test (P = 0.001). In addition, 0.89(0.43–3.87), respectively (Fig. 5). The independence of the TMAO
plasma choline and betaine levels in Q4 (vs. Q1) were correlated with correlation with mortality was also confirmed by the likelihood ratio test
functional outcome (Sup table I). (P = 0.005). In addition, plasma choline and betaine levels in Q4 (vs.
The ROC curve was applied for choosing the cut-off value of TMAO in Q1) were correlated with mortality (Sup table I).
predicting functional outcome. An optimal amount of 5.8 μM produced a The ROC curve was also applied for exploring the cut-off value of
sensitivity of 57.5% and a specificity of 74.9%, with AUC of 0.78 (95% TMAO in predicting mortality. An optimal amount of 7.8 μM produced a
CI 0.72–0.83, Fig. 4A), which showed a significantly greater discrimi­ sensitivity of 80.9% and a specificity of 72.0%, with AUC of 0.80 (95%
natory ability as compared with age, CRP, and IL-6, and was within the CI 0.74–0.87; Fig. 4B), which showed a significantly greater discrimi­
range of the NIHSS score (Table 3). In a combined model, the AUC of the natory ability as compared with age, CRP, and IL-6, and was within the
NIHSS score could be increased with TMAO (0.82; 95% CI 0.77–0.89, P range of the NIHSS score (Table 3). In a combined model, the AUC of the
= 0.003). Furthermore, TMAO improved the established risk factors NIHSS score could be increased with TMAO (0.85; 95% CI: 0.80–0.90, P
(AUC of the combined model, 0.83; 95% CI, 0.78–0.90; p < 0.001). This = 0.002). Furthermore, TMAO improved the established risk factors
improvement was stable in an internal 5-fold cross-validation that (AUC of the combined model, 0.86; 95% CI, 0.80–0.93; P < 0.001). This
resulted in an average AUC (standard error) of 0.805 (0.032) for the improvement was stable in an internal 5-fold cross-validation that
established risk factors and 0.834(0.026) for the combined model, cor­ resulted in an average AUC (standard error) of 0.837 (0.025) for the
responding to a difference of 0.029 (0.008). The NRI statistic results established risk factors and 0.861(0.020) for the combined model, cor­
indicated that the correct reclassification of functional outcomes could responding to a difference of 0.024 (0.005). The NRI statistic results
be improved after adding TMAO to the existing risk factors (P = 0.02). indicated that functional outcomes’ correct reclassification could be
Furthermore, this result also is confirmed with the IDI statistic (P = improved after adding TMAO to the existing risk factors (P = 0.003).
0.01) (Table 4). However, this result could not be confirmed with the IDI statistic (P =
0.11) (Table 4).

4
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

Table 1 Table 2
Characteristics of stroke patients and control cases. Predictors of poor functional outcome and all-cause mortality.
Ischemic Control cases Predictor variable OR (95%CI) P
stroke a
Multivariate Analysis for Functional Outcome
N 351 150 TMAO (increase per unit) 1.21(1.07–1.35) <0.001
Age(years), median (IQR) 66(55–74) 66(55–74) IL-6 (increase per unit) 1.30(1.18–1.52) 0.005
Male, n (%) 177(50.4) 75(50.0) Age (increase per unit) 1.05(1.02–1.08) 0.003
BMI, kg/m2, median (IQR) 25.6 24.8 Stroke severity, NIHSS (>5) 4.45(2.85–8.16) <0.001
(23.7–27.4) (22.5–26.2) Large-vessel occlusive VS. other 8.15 0.004
Prior vascular risk factors, n (%) (2.15–20.44)
Hypertension 224(63.8) 62(41.3) IV thrombolysis and/or mechanical 0.45(0.23–0.70) <0.001
Diabetes 77(21.9) 25(16.7) thrombectomy
Hypercholesterolemia 85(24.2) 33(22.0) Multivariate analysis for mortalitya
Atrial fibrillation 71(21.1) 2(1.3) TMAO (increase per unit) 1.24(1.06–1.38) <0.001
Prior TIA 42(12.0) 3(2.0) IL-6 (increase per unit) 1.28(1.10–1.64) 0.015
Smoking 61(17.4) 25(16.7) Age (increase per unit) 1.06(1.03–1.08) 0.001
PVD 15(4.3) 1 (0.7) Stroke severity, NIHSS (>5) 5.05(2.75–8.77) <0.001
Pre-stroke treatment, n (%) IV thrombolysis and/or mechanical 0.40(0.19–0.64) <0.001
Antihypertensive 218(62.1) 38(25.3) thrombectomy
Anticoagulant 38(10.8) 5(3.3)
Antiplatelet agents 65(18.5) 6(4.0) OR, odds ratio; NIHSS, National Institutes of Health Stroke Scale; IL-6, inter­
Statins 72(20.5) 6(4.0) leukin-6; CRP, C–reactive protein; TMAO, Trimethylamine N-oxide; eGFR,
Acute treatment, n (%) estimated glomerular filtration rate.
a
IV thrombolysis 43(12.2) – Multivariable model included all of the following variables: age, sex, BMI,
Mechanical thrombectomy 10(2.8) – traditional risk factors, NIHSS and Infarct volume at admission, stroke subtype,
IV thrombolysis and/or mechanical 49(14.0) – pre-stroke treatment, acute stroke treatment, eGFR, plasma levels of glucose,
thrombectomy CRP, IL-6, Choline, Betaine and TMAO. OR expressed as OR (95% confidence
Stroke causative factors, n (%)
interval).
Small-vessel occlusive 75(21.4) –
Large-vessel occlusive 73(20.8) –
Cardioembolic 135(38.5) – 5. Discussions
Other 45(12.8) –
Unknown 23(6.6)
TMAO is an oxidized metabolite mediated by gut microbial meta­

Laboratory findings, median (IQR)
Glucose level, mmol/l 5.95 5.68 bolism of choline-containing lipids and carnitine-based molecules
(5.05–7.29) (4.95–6.38) (Suzuki et al., 2017). The gut microbial TMAO pathway has been shown
C–reactive protein, mg/l 6.5(3.8–11.6) 4.4(2.6–7.3) to possess numerous clinical and mechanistic links with the promotion
IL-6, pg/ml 7.8(5.5–9.4) 4.9(3.5–7.4)
of atherosclerosis (Wang et al., 2011; Miao et al., 2015) and with mor­
eGFR, ml⋅min− 1⋅(1.73⋅m2)− 1
83(60–97) 72(55–88)
TMAO, μM 6.1(3.7–9.9) 4.0(2.4–5.9) tality and adverse cardiovascular outcomes for cardiorenal disorders (e.
Choline, μM 10.1(7.2–12.6) 8.2(5.9–10.3) g., coronary artery disease (Wang et al., 2011; Senthong et al., 2016),
Betaine, μM 43.5 35.2 heart failure (Troseid et al., 2015) and chronic kidney disease (Tang
(20.7–54.1) (15.4–47.3) et al., 2015; Rhee et al., 2013)) and cerebrovascular diseases (Yin et al.,
NIHSS at admission, median (IQR) 5(3− 12)
2015).

Infarct volume, ml. median (IQR) 15.5(6.2–24.1) –
This study was the first population-based study on the correlation
NIHSS, National Institutes of Health Stroke Scale; TIA, transient ischemic attack; between circulating TAMO levels and functional outcome(mortality)
IL-6, interleukin-6; IQR, interquartile ranges; TMAO, Trimethylamine N-oxide; risk in Chinese patients with ischemic stroke. This study’s findings were
PVD, peripheral vascular disease; eGFR, estimated glomerular filtration rate.
as follows: First, plasma levels of TMAO were dose-dependently asso­
ciated with 3-month poor functional outcome and all-cause mortality
The time to death was analyzed by Kaplan–Meier survival curves risk among patients with ischemic stroke. Furthermore, TMAO
based on TMAO optimal cut-off value, which had been defined by the improved the prognostic accuracy of the NIHSS to predict functional
ROC curve(7.8 μM). The Kaplan–Meier estimates of all-cause mortality outcome and mortality. Second, we confirmed previous reports that
stratified by baseline TMAO (elevated vs. normal) was shown in Fig. 6. suggested patients with ischemic stroke had higher TMAO than healthy
Patients with elevated TMAO had shorter survival than patients with controls. Third, increased fasting TMAO concentrations were observed
normal TMAO (P < 0.001, log-rank test). The OR between those two to be associated with stroke severity according to the NIHSS score and
groups (elevated vs. normal) was 3.48 (95% CI 1.58–6.46). lesion size. Fourth, we observe that the prognostic value of increased
choline and betaine are only observed among those with increased
4. Sub-group analysis TMAO.
A previous study finished by Haghikia et al. (Haghikia et al., 2018)
we also conducted analyses separately among cases who experienced reported a graded relation between TMAO levels and the risk of subse­
with eGFR <90 mL/min/1.73 m2 and ≥ 90 mL/min/1.73 m2. In the quent cardiovascular events in patients with recent prior ischemic
multivariate regression analysis, the data suggested that for each 1 μM stroke, suggesting that TMAO-related increase of proinflammatory
increase of plasma concentration of TMAO, the association with poor monocytes may add to an elevated cardiovascular risk of patients with
outcome was stronger among cases who experienced with eGFR increased TMAO levels. Consistent with our findings, Suzuki et al.
<90 mL/min/1.73 m2 (OR = 1.25, 95%CI: 1.10–1.38; P < 0.001) versus (Suzuki et al., 2017) reported that TMAO levels showed an association
≥90 mL/min/1.73 m2 (OR = 1.16, 95%CI: 1.03–1.33; P = 0.005). with poor prognosis at two years and superiority over current bio­
Interestingly, the predictive value of TMAO to predict all-cause mor­ markers for patients hospitalized due to acute myocardial infarction.
tality in cases who experienced with eGFR <90 mL/min/1.73 m2 (OR = Elevated levels were associated with poor prognosis at one year, and a
1.30, 95%CI: 1.13–1.43; p < 0.001) was stronger than in ≥90 mL/min/ combination of TMAO and NT-pro-BNP provided additional prognostic
1.73 m2 (1.18 [1.03–1.40]; P = 0.008). information in acute heart failure (Suzuki et al., 2016). Similarly, Tang
et al. (Tang et al., 2014) reported that high TMAO levels were observed
in patients with HF, and elevated TMAO levels portended higher long-

5
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

Fig. 3. Multivariate analyses for poor functional outcome according to TMAO quartiles in stroke patients (first quartile as the reference). The numbers in the first
column represent stroke patients, and the second column represents patients with poor outcome. §Multivariable model included all of the following variables: age,
sex, BMI, traditional risk factors, NIHSS and Infarct volume at admission, stroke subtype, pre-stroke treatment, acute stroke treatment, eGFR, plasma levels of
glucose, CRP, IL-6, Choline, and Betaine. OR was expressed as OR (95% confidence interval). NIHSS, National Institutes of Health Stroke Scale; CRP, C-reactive
protein; BMI, body mass index; IL-6, interleukin-6; TMAO, Trimethylamine N-oxide.

Fig. 4. ROC curves were utilized to evaluate the accuracy of plasma TMAO levels to predict functional outcome and mortality. (A) ROC curves were used to assess
the accuracy of plasma TMAO levels to predict poor functional outcomes. (B) ROC curves were utilized to assess the accuracy of plasma TMAO levels to predict
mortality. A poor outcome was defined as an mRS score of 3 to 6 points. TMAO, Trimethylamine N-oxide; ROC, receiver operating characteristic.

Table 3 Table 4
Prediction of poor functional outcomes and morality according to ROC. Plasma levels of TMAO at admission prediction of functional outcome and all-
Parameter AUC 95% CI p
cause mortality with AUROC.
End points AUROC
Prediction of functional outcome
TMAO 0.78 0.72 0.83 – TMAO Risk Risk Incremental NRI(P) IDI(P)
NIHSS 0.75 0.70 0.79 0.072 factorsa factors area (P)b
Age 0.70 0.64 0.75 0.003 with
IL-6 0.65 0.59 0.70 <0.001 TMAOa
CRP 0.63 0.56 0.69 <0.001
Combined score (TMAO/NIHSS) 0.82 0.77 0.89 0.011 Functional 0.78 0.81 0.83 0.03(0.01) 0.18 0.03
Prediction of mortality (0.02) (0.01)
TMAO 0.80 0.74 0.87 – Morality 0.80 0.84 0.86 0.02(0.02) 0.28 0.02
NIHSS 0.82 0.77 0.89 0.15 (0.003) (0.11)
Age 0.73 0.67 0.78 0.005 NIHSS, National Institutes of Health Stroke Scale; CRP, C-reactive protein; BMI,
IL-6 0.70 0.65 0.76 <0.001
body mass index; IL-6, interleukin-6; TMAO, Trimethylamine N-oxide.
CRP 0.69 0.63 0.74 <0.001 a
Established risk factors including: age, sex, BMI, traditional risk factors,
Combined score (TMAO/NIHSS) 0.85 0.80 0.90 <0.001
NIHSS and Infarct volume at admission, stroke subtype, pre-stroke treatment,
AUC, area under the curve; CI, confidence interval; CRP, C-reactive protein; acute stroke treatment, eGFR, plasma levels of glucose, CRP, IL-6, Choline, and
NIHSS, National Institutes of Health Stroke Scale; IL-6, interleukin-6; TMAO, Betaine.
Trimethylamine N-oxide. b
Comparison of AUROCs: established risk factors without TMAO levels vs.
established risk factors with TMAO levels.

6
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

Fig. 5. Multivariate analyses for mortality according to TMAO quartiles in stroke patients (first quartile as the reference). The numbers in the first column represent
stroke patients, and the second column represents non-survivors. §Multivariable model included all of the following variables: age, sex, BMI, traditional risk factors,
NIHSS and Infarct volume at admission, stroke subtype, pre-stroke treatment, acute stroke treatment, eGFR, plasma levels of glucose, CRP, IL-6, Choline, and Betaine.
OR was expressed as OR (95% confidence interval). NIHSS, National Institutes of Health Stroke Scale; CRP, C-reactive protein; BMI, body mass index; IL-6, inter­
leukin-6; TMAO, Trimethylamine N-oxide.

Fig. 6. Kaplan-Meier analysis of all-cause mortality according to the TMAO cut-off value (defined by the ROC curve). The patients with elevated TMAO had a higher
risk of mortality (log-rank test P < 0.001). TMAO, Trimethylamine N-oxide; ROC, receiver operating characteristic.

term mortality risk independent of traditional risk factors and car­ studies, they were not associated with all-cause mortality, cardiovas­
diorenal indexes. Increased TMAO concentrations correlate with coro­ cular death, or hospitalizations (Kaysen et al., 2015). Another study
nary atherosclerosis burden and may associate with long-term mortality showed that TMAO was associated with all-cause mortality in subjects
in patients with chronic kidney disease (CKD) undergoing coronary with any degree of renal function impairment, but not in subjects with
angiography (Stubbs et al., 2016). Another study also suggested that normal renal function (Gruppen et al., 2017).
plasma TMAO levels were elevated in patients with CKD and portend Furthermore, one study showed that high choline and betaine levels
lower long-term survival (Tang et al., 2015). are only associated with a higher risk of future MACE with a concomi­
Similarly, fasting plasma concentrations of TMAO were higher in tant increase in TMAO (Wang et al., 2014), confirmed by our results.
diabetic patients and portend higher major adverse cardiac events Also, Gruppen et al. (Gruppen et al., 2017) found that TMAO was
(MACE) and mortality risks independent of other factors (Tang et al., associated with all-cause mortality, particularly in subjects with eGFR
2017). Although TMAO levels were found to be increased in hypergly­ <90 mL/min/1.73 m2. We also found that the prognostic value of TMAO
cemic subjects and patients with impaired kidney function in other was more pronounced in subjects with eGFR <90 mL/min/1.73 m2. one

7
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

study found that higher levels of TMAO were not independently asso­ to TMAO enhanced sub-maximal stimulus-dependent platelet activation
ciated with cardiovascular death after adjustment for age and eGFR from multiple agonists through augmented Ca (2+) release from intra­
(Skagen et al., 2016). However, we found that TMAO was still associated cellular stores (Zhu et al., 2016). TMAO promotes an imbalance in
with mortality after adjustment for age and eGFR. cholesterol uptake and effluxes from macrophages, leading to an
In this study, we found that ischemic stroke had higher TMAO than abundance of pro-atherogenic foam cells migrating to the arterial wall
healthy controls. A previous study found that higher TMAO levels were (Wang et al., 2011; Koeth et al., 2013).
associated with increased risk of the first stroke in hypertensive patients Some study limitations need to be considered. (1) only a single time-
(Nie et al., 2018). However, it should be noted that while the majority of point blood draw after an overnight fast was available, and we did not
studies in different cohorts have observed a positive association between have dietary history to confirm or refute the impact of diet on TMAO. (2)
systemic TMAO levels and incident cardiovascular risks (Shafi et al., the relatively small number of mortalities end-points (N = 47) and lack
2017; Senthong et al., 2016), others have not (Kaysen et al., 2015; of data on cause of death, preventing us from obtaining a more nuanced
Mueller et al., 2015). One study showed that stroke and transient understanding of the relationship between TMAO and cardiovascular
ischemic attack patients showed significant dysbiosis of the gut micro­ mortality. (3) the potential for confounding by unmeasured risk factors,
biota, and their blood TMAO levels were decreased (Yin et al., 2015). such as dietary patterns and proteinuria. Specifically, we did not have
Similarly, a European study of 339 patients undergoing coronary angi­ any information regarding gastrointestinal symptoms and pathology or
ography found no association between plasma TMAO and prevalent prior antibiotic use or knowledge of previous supplements other than
coronary artery disease or incident events over an 8-year follow-up those taken on the day of enrollment. Thus, the effects of antibiotic use
period. (Mueller et al., 2015). In a North American study of 817 in modulating gut microbiome–mediated TMAO synthesis will need to
healthy mixed-race volunteers aged 33 to 45, plasma TMAO levels were be incorporated in future studies in this field. (4) in this study, only
not associated with incident coronary artery calcification over a 10-year ischemic strokes were included. Interestingly, one study demonstrated
follow-up period (Meyer et al., 2016). In this study, the median levels of that TMAO has a stronger association with hemorrhagic stroke than
TMAO in stroke and controls were 6.1 μM and 4.0 μM. A previous study ischemic stroke (Nie et al., 2018). (5) The variants of the FMO3 gene,
in the Chinese population reported that the average and median TMAO which could provide opportunities to assess the impact of reduced
levels of stroke/TIA patients were only 2.68uM and 1.91 uM, respec­ TMAO production on CV disease, were not tested in this study. Inter­
tively, comparable to quartile 1 in a US cohort (Koeth et al., 2013). The estingly, in a separate cohort, no association was identified between 471
range of TMAO levels in different study groups was vast (Supplementary FMO gene polymorphisms and plasma TMAO levels in 3865 subjects
materials). These apparently conflicting data are likely to have resulted referred for cardiac evaluation (Hartiala et al., 2014). (6) TMAO plasma
from various issues, including differences in study methodology, diet, level was measured at only a single time point, and we did not test
ethnicity, environment, and prevalent patterns of gut microbiome TMAO in other time-point and cerebral spinal fluid samples. One study
composition in study subjects. suggested that TMAO can be assessed in human CSF (Del Rio et al.,
Alterations to the normal gut microbiota or gut dysbiosis caused by 2017). (7) It is not known whether decreasing systemic TMAO using
acute brain lesions can affect neuroinflammatory and immune responses antibiotics or diet improve the stroke outcome in humans. Observational
in the brain and worsen neurological function (Singh et al., 2016). studies challenge the proposed relationships between dietary choline or
Commensal gut microbiota exerts protection against ischemic damage, betaine intakes with CVD outcomes (Meyer and Shea, 2017). Unfortu­
and their depletion or dysbiosis increases poststroke mortality in mice nately, as a cross-sectional study, the obtained results could not prove
and influences stroke outcome (Winek et al., 2016). In ischemic stroke, any causal relationship.
gut dysbiosis via stress-mediated intestinal paralysis can alter T-cell
homeostasis, promote migration of T cells from the intestine to the 6. Clinical perspective
ischemic brain, and increase proinflammatory responses resulting in
poor stroke outcome (Benakis et al., 2016). In ischemic stroke, gut In this study, the data suggests that the elevated serum level of gut
dysbiosis, and increased bacterial counts of Lactobacillus ruminis sub­ microbial TMAO is a potential useful biomarker for predicting poor
group in the fecal gut microbiota was associated with elevated systemic prognosis, and therefore it is useful for therapeutic decision making in
inflammation and altered the balance in favor of proinflammatory patients with ischemic stroke. Whether TMAO reduction treatment
mechanism (Arpaia et al., 2013). could improve stroke prognosis need further conform.
Altered TMAO levels have been associated with impaired cardiac
function, heart attack, and heart failure (Zhu et al., 2016). The role of
7. Conclusions
elevated TAMO in stroke prognosis might, through the following
mechanisms: (1) One study suggested that aging increases circulating
Fasting plasma concentrations of gut microbial TMAO are higher in
TMAO levels, which may impair eNOS-derived NO bioavailability by
patients with ischemic stroke and portend higher poor functional
increasing vascular inflammation and oxidative stress, contributing to
outcome events and mortality risks independent of traditional risk fac­
aging-associated endothelial dysfunction in aged rats (Li et al., 2017). In
tors. The present studies suggest a potential clinical utility for clearance
part, endothelial dysfunction resulting from excessive production of
of gut microbiome generated uremic toxins as a possible adjunct therapy
reactive oxygen species and inflammation is an essential mechanism of
for stroke. Future large-scale studies specifically designed to examine
cerebrovascular damage (Moskowitz et al., 2010). Recent studies
the predictive value of TMAO for cardiovascular and mortality outcomes
demonstrated that TMAO promotes vascular inflammation and oxida­
in stroke populations will be necessary to determine the generalizability
tive stress, inhibits eNOS expression and activity, and reduces NO pro­
of our initial observations to these separate groups.
duction, which is associated with endothelial dysfunction and
atherosclerosis (Wang et al., 2011; Seldin et al., 2016; Chen et al., 2016).
(2) TMAO mediated vascular inflammation by activating the nucleotide- Conflict of interest disclosures
binding oligomerization domain-like receptor family pyrin
domain–containing 3 (NLRP3) inflammasome via the sirtuin-3 (SIRT3)– None.
superoxide dismutase (SOD)–mitochondrial reactive oxygen species
(mtROS) signaling pathway (Chen et al., 2017). (3) One study showed Funding/support
that gut microbes, through the generation of trimethylamine N-oxide
(TMAO), directly contribute to platelet hyperreactivity and enhanced This work was supported by the Xiamen Health Care Guidance
thrombosis potential (Zhu et al., 2016), and direct exposure of platelets Program (No. 3502Z20209227).

8
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

Acknowledgment containing monooxygenase 3 as a potential player in diabetes-associated


atherosclerosis. Nat. Commun. 6, 6498.
Moskowitz, M.A., Lo, E.H., Iadecola, C., 2010. The science of stroke: mechanisms in
All authors have contributed significantly and agreed with the con­ search of treatments. Neuron. 67, 181–198.
tent of the manuscript. We are grateful to all the people who participated Mueller, D.M., Allenspach, M., Othman, A., et al., 2015. Plasma levels of trimethylamine-
in our study. We also thank Andrea Baird, MD, from Edanz Editing China N-oxide are confounded by impaired kidney function and poor metabolic control[J].
Atherosclerosis 243 (2), 638–644.
(www.liwenbianji.cn/ac), for editing the English text of a draft of this Nie, J., Xie, L., Zhao, B., et al., 2018. Serum trimethylamine N-oxide concentration is
manuscript. positively associated with first stroke in hypertensive patients[J]. Stroke 49 (9),
2021–2028.
Pencina, M.J., Steyerberg, E.W., D’Agostino Sr., R.B., 2011. Extensions of net
Appendix A. Supplementary data reclassification improvement calculations to measure usefulness of new biomarkers.
Stat. Med. 30, 11–21.
Supplementary data to this article can be found online at https://doi. Qi, J., You, T., Li, J., et al., 2018. Circulating trimethylamine N-oxide and the risk of
cardiovascular diseases: a systematic review and meta-analysis of 11 prospective
org/10.1016/j.jneuroim.2021.577526. cohort studies[J]. J. Cell. Mol. Med. 22 (1), 185–194.
Rhee, E.P., Clish, C.B., Ghorbani, A., Larson, M.G., Elmariah, S., McCabe, E., et al., 2013.
References A combined epidemiologic and metabolomic approach improves CKD prediction.
J. Am. Soc. Nephrol. 24, 1330–1338.
Seldin, M.M., Meng, Y., Qi, H., Zhu, W., Wang, Z., Hazen, S.L., Lusis, A.J., Shih, D.M.,
Adams Jr., H.P., Bendixen, B.H., Kappelle, L.J., et al., 1993. Classification of subtype of
2016. Trimethylamine N-oxide promotes vascular inflammation through signaling of
acute ischemic stroke. Definitions for use in a multicenter clinical trial. TOAST. Trial
mitogen-activated protein kinase and nuclear factor-kappaB. J. Am. Heart Assoc. 5,
of Org 10172 in acute stroke treatment. Stroke 24, 35–41.
e002767.
Arpaia, N., Campbell, C., Fan, X., Dikiy, S., van der Veeken, J., de Roos, P., Liu, H.,
Senthong, V., Wang, Z., Li, X.S., et al., 2016. Intestinal microbiota-generated metabolite
Cross, J.R., Pfeffer, K., Coffer, P.J., Rudensky, A.Y., 2013. Metabolites produced by
trimethylamine-N-oxide and 5-year mortality risk in stable coronary artery disease:
commensal bacteria promote peripheral regulatory T-cell generation. Nature 504,
the contributory role of intestinal microbiota in a COURAGE-like patient cohort[J].
451–455.
J. Am. Heart Assoc. 5 (6), e002816.
Benakis, C., Brea, D., Caballero, S., Faraco, G., Moore, J., Murphy, M., Sita, G.,
Shafi, T., Powe, N.R., Meyer, T.W., Hwang, S., Hai, X., Melamed, M.L., et al., 2017.
Racchumi, G., Ling, L., Pamer, E.G., Iadecola, C., Anrather, J., 2016. Commensal
Trimethylamine N-oxide and cardiovascular events in hemodialysis patients. J. Am.
microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat.
Soc. Nephrol. 28, 321–331.
Med. 22, 516–552.
Sims, J.R., Gharai, L.R., Schaefer, P.W., et al., 2009. ABC/2 for rapid clinical estimate of
Bonita, R., Beaglehole, R., 1988. Recovery of motor function after stroke. Stroke. 19 (12),
infarct, perfusion, and mismatch volumes[J]. Neurology 72 (24), 2104–2110.
1497–1500.
Singh, V., Roth, S., Llovera, G., Sadler, R., Garzetti, D., Stecher, B., Dichgans, M.,
Chen, M.L., Yi, L., Zhang, Y., Zhou, X., Ran, L., Yang, J., et al., 2016. Resveratrol
Liesz, A., 2016. Microbiota dysbiosis controls the neuroinflammatory response after
attenuates trimethylamine-N-oxide (TMAO)-induced atherosclerosis by regulating
stroke. J. Neurosci. 36, 7428–7440.
TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota.
Skagen, K., Trøseid, M., Ueland, T., Holm, S., Abbas, A., Gregersen, I., et al., 2016. The
Mbio 7, e02210–e02215.
carnitine-butyrobetaine-trimethylamine-N-oxide pathway and its association with
Chen, M., Zhu, X., Ran, L., et al., 2017. Trimethylamine-N-oxide induces vascular
cardiovascular mortality in patients with carotid atherosclerosis. Atherosclerosis.
inflammation by activating the NLRP3 inflammasome through the SIRT3-SOD2-
247, 64–69.
mtROS signaling pathway[J]. J. Am. Heart Assoc. 6 (9), e006347.
Stubbs, J.R., House J A, Ocque, A.J., et al., 2016. Serum trimethylamine-N-oxide is
Daubail, B., Jacquin, A., Guilland, J.C., et al., 2013. Serum 25-hydroxyvitamin D predicts
elevated in CKD and correlates with coronary atherosclerosis burden[J]. J. Am. Soc.
severity and prognosis in stroke patients[J]. Eur. J. Neurol. 20 (1), 57–61.
Nephrol. 27 (1), 305–313.
Del Rio, D., Zimetti, F., Caffarra, P., et al., 2017. The gut microbial metabolite
Suzuki, T., Heaney, L.M., Bhandari, S.S., Jones, D.J.L., Ng, L.L., 2016. Trimethylamine N-
trimethylamine-N-oxide is present in human cerebrospinal fluid[J]. Nutrients 9 (10),
oxide and prognosis in acute heart failure. Heart 102, 841–848.
1053.
Suzuki, T., Heaney, L.M., Jones, D.J.L., et al., 2017. Trimethylamine N-oxide and risk
Gruppen, E.G., Garcia, E., Connelly, M.A., et al., 2017. TMAO is associated with
stratification after acute myocardial infarction[J]. Clin. Chem. 63 (1), 420–428.
mortality: impact of modestly impaired renal function[J]. Sci. Rep. 7 (1), 13781.
Tang, W.H., Wang, Z., Levison, B.S., Koeth, R.A., Britt, E.B., Fu, X., Wu, Y., Hazen, S.L.,
Haghikia, A., Li, X.S., Liman, T.G., et al., 2018. Gut microbiota–dependent
2013. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular
trimethylamine N-oxide predicts risk of cardiovascular events in patients with stroke
risk. N Engl. J. Med. 368, 1575–1584.
and is related to proinflammatory monocytes[J]. Arterioscler. Thromb. Vasc. Biol.
Tang, W.H.W., Wang, Z., Fan, Y., et al., 2014. Prognostic value of elevated levels of
38 (9), 2225–2235.
intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with
Hartiala, J., Bennett, B.J., Tang, W.H., et al., 2014. Comparative genome-wide
heart failure: refining the gut hypothesis[J]. J. Am. Coll. Cardiol. 64 (18),
association studies in mice and humans for trimethylamine N-oxide, a
1908–1914.
proatherogenic metabolite of choline and L-carnitine. Arterioscler. Thromb. Vasc.
Tang, W.H., Wang, Z., Kennedy, D.J., Wu, Y., Buffa, J.A., Agatisa-Boyle, B., et al., 2015.
Biol. 34, 1307–1313.
Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to
Kaysen, G.A., Johansen, K.L., Chertow, G.M., Dalrymple, L.S., Kornak, J., Grimes, B.,
both development of renal insufficiency and mortality risk in chronic kidney disease.
Dwyer, T., Chassy, A.W., Fiehn, O., 2015. Associations of trimethylamine N-oxide
Circ. Res. 116, 448–455.
with nutritional and inflammatory biomarkers and cardiovascular outcomes in
Tang, W.H.W., Wang, Z., Li, X.S., et al., 2017. Increased trimethylamine N-oxide
patients new to dialysis. J. Ren. Nutr. 25, 351–356.
portends high mortality risk independent of glycemic control in patients with type 2
Koeth, R.A., Wang, Z., Levison, B.S., Buffa, J.A., Org, E., Sheehy, B.T., Britt, E.B., Fu, X.,
diabetes mellitus[J]. Clin. Chem. 63 (1), 297–306.
Wu, Y., Li, L., et al., 2013. Intestinal microbiota metabolism of L-carnitine, a nutrient
The GBD, 2018. 2016 lifetime risk of stroke collaborators. Global, regional, and country-
in red meat, promotes atherosclerosis. Nat. Med. 19 (5), 576–585.
specific lifetime risks of stroke, 1990 and 2016. N. Engl. J. Med. 379, 2429–2437.
Koeth, R.A., Levison, B.S., Culley, M.K., et al., 2014. Gamma- Butyrobetaine is a
Troseid, M., Ueland, T., Hov, J.R., Svardal, A., Gregersen, I., Dahl, C.P., Aakhus, S.,
proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO.
Gude, E., Bjorndal, B., Halvorsen, B., Karlsen, T.H., Aukrust, P., Gullestad, L.,
Cell Metab. 20, 799–812.
Berge, R.K., Yndestad, A., 2015. Microbiota-dependent metabolite trimethylamine-
LangDH, YeungCK, Peter, R.M., Ibarra, C., Gasser, R., Itagaki, K., Philpot, R.M., Rettie, A.
N-oxide is associated with disease severity and survival of patients with chronic
E., 1998. Isoform specificity of trimethylamine N-oxygenation by human flavin-
heart failure. J. Intern. Med. 277, 717–726.
containing monooxygenase (FMO) and P450 enzymes: selective catalysis by FMO3.
Tu, W.J., Zhao, S.J., Xu, D.J., et al., 2014. Serum 25-hydroxyvitamin D predicts the short-
Biochem. Pharmacol. 56, 1005–1012.
term outcomes of Chinese patients with acute ischaemic stroke[J]. Clin. Sci. 126 (5),
Levey, A.S., Bosch, J.P., Lewis, J.B., Greene, T., Rogers, N., Roth, D., 1999. A more
339–346.
accurate method to estimate glomerular filtration rate from serum creatinine: a new
Tu, W.J., Zeng, X.W., Deng, A., et al., 2017. Circulating FABP4 (fatty acid–binding
prediction equation. Modification of diet in renal disease study group. Ann. Intern.
protein 4) is a novel prognostic biomarker in patients with acute ischemic stroke[J].
Med. 130, 461–470.
Stroke 48 (6), 1531–1538.
Li, T., Chen, Y., Gua, C., et al., 2017. Elevated circulating trimethylamine N-oxide levels
Wang, Z., Klipfell, E., Bennett, B.J., Koeth, R., Levison, B.S., Dugar, B., et al., 2011. Gut
contribute to endothelial dysfunction in aged rats through vascular inflammation
flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature
and oxidative stress[J]. Front. Physiol. 8, 350.
472, 57–63.
Liang, Z., Dong, Z., Guo, M., et al., 2019. Trimethylamine N-oxide as a risk marker for
Wang, Z., Tang, W.H., Buffa, J.A., Fu, X., Britt, E.B., Koeth, R.A., et al., 2014. Prognostic
ischemic stroke in patients with atrial fibrillation[J]. J. Biochem. Mol. Toxicol. 33
value of choline and betaine depends on intestinal microbiota-generated metabolite
(2), e22246.
trimethylamine-N-oxide. Eur. Heart J. 35, 904–910.
Meyer, K., Shea, J., 2017. Dietary choline and betaine and risk of CVD: a systematic
Winek, K., Engel, O., Koduah, P., Heimesaat, M.M., Fischer, A., Bereswill, S., Dames, C.,
review and meta-analysis of prospective studies[J]. Nutrients 9 (7), 711.
Kershaw, O., Gruber, A.D., Curato, C., Oyama, N., Meisel, C., Meisel, A., Dirnagl, U.,
Meyer, K.A., Benton, T.Z., Bennett, B.J., et al., 2016. Microbiota-dependent metabolite
trimethylamine N-oxide and coronary artery calcium in the coronary artery risk
development in young adults study (CARDIA)[J]. J. Am. Heart Assoc. 5 (10),
e003970.
Miao, J., Ling, A.V., Manthena, P.V., Gearing, M.E., Graham, M.J., Crooke, R.M.,
Croce, K.J., Esquejo, R.M., Clish, C.B., Morbid Obesity Study Group, 2015. Flavin-

9
J. Zhang et al. Journal of Neuroimmunology 354 (2021) 577526

2016. Depletion of cultivatable gut microbiota by broad-spectrum antibiotic trimethylamine-n-oxide level in patients with large-artery atherosclerotic stroke or
pretreatment worsens outcome after murine stroke. Stroke. 47, 1354–1363. transient ischemic attack. J. Am. Heart Assoc. 4, 1–12.
Yin, J., Liao, S.X., He, Y., Wang, S., Xia, G.H., Liu, F.T., Zhu, J.J., You, C., Chen, Q., Zhu, W., Gregory, J.C., Org, E., et al., 2016. Gut microbial metabolite TMAO enhances
Zhou, L., Pan, S.Y., Zhou, H.W., 2015. Dysbiosis of gut microbiota with reduced platelet hyperreactivity and thrombosis risk[J]. Cell 165 (1), 111–124.

10

You might also like