ArticlePDF Available

Identification of Human Plasma Metabolites Exhibiting Time-of-Day Variation Using an Untargeted Liquid Chromatography–Mass Spectrometry Metabolomic Approach

Taylor & Francis
Chronobiology International
Authors:

Abstract and Figures

Although daily rhythms regulate multiple aspects of human physiology, rhythmic control of the metabolome remains poorly understood. The primary objective of this proof-of-concept study was identification of metabolites in human plasma that exhibit significant 24-h variation. This was assessed via an untargeted metabolomic approach using liquid chromatography-mass spectrometry (LC-MS). Eight lean, healthy, and unmedicated men, mean age 53.6 (SD ± 6.0) yrs, maintained a fixed sleep/wake schedule and dietary regime for 1 wk at home prior to an adaptation night and followed by a 25-h experimental session in the laboratory where the light/dark cycle, sleep/wake, posture, and calorific intake were strictly controlled. Plasma samples from each individual at selected time points were prepared using liquid-phase extraction followed by reverse-phase LC coupled to quadrupole time-of-flight MS analysis in positive ionization mode. Time-of-day variation in the metabolites was screened for using orthogonal partial least square discrimination between selected time points of 10:00 vs. 22:00 h, 16:00 vs. 04:00 h, and 07:00 (d 1) vs. 16:00 h, as well as repeated-measures analysis of variance with time as an independent variable. Subsequently, cosinor analysis was performed on all the sampled time points across the 24-h day to assess for significant daily variation. In this study, analytical variability, assessed using known internal standards, was low with coefficients of variation <10%. A total of 1069 metabolite features were detected and 203 (19%) showed significant time-of-day variation. Of these, 34 metabolites were identified using a combination of accurate mass, tandem MS, and online database searches. These metabolites include corticosteroids, bilirubin, amino acids, acylcarnitines, and phospholipids; of note, the magnitude of the 24-h variation of these identified metabolites was large, with the mean ratio of oscillation range over MESOR (24-h time series mean) of 65% (95% confidence interval [CI]: 49-81%). Importantly, several of these human plasma metabolites, including specific acylcarnitines and phospholipids, were hitherto not known to be 24-h variant. These findings represent an important baseline and will be useful in guiding the design and interpretation of future metabolite-based studies.
Content may be subject to copyright.
Identification of Human Plasma Metabolites Exhibiting Time-of-Day
Variation Using an Untargeted Liquid ChromatographyMass
Spectrometry Metabolomic Approach
Joo Ern Ang,
1
Victoria Revell,
2
Anuska Mann,
2
Simone Mäntele,
2
Daniella T. Otway,
2
Jonathan D. Johnston,
2
Alfred E. Thumser,
2
Debra J. Skene,
2
and Florence Raynaud
1
1
Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton,
Surrey, UK,
2
Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
Although daily rhythms regulate multiple aspects of human physiology, rhythmic control of the metabolome remains
poorly understood. The primary objective of this proof-of-concept study was identification of metabolites in human
plasma that exhibit significant 24-h variation. This was assessed via an untargeted metabolomic approach using liquid
chromatographymass spectrometry (LC-MS). Eight lean, healthy, and unmedicated men, mean age 53.6 (SD ± 6.0) yrs,
maintained a fixed sleep/wake schedule and dietary regime for 1 wk at home prior to an adaptation night and
followed by a 25-h experimental session in the laboratory where the light/dark cycle, sleep/wake, posture, and calorific
intake were strictly controlled. Plasma samples from each individual at selected time points were prepared using
liquid-phase extraction followed by reverse-phase LC coupled to quadrupole time-of-flight MS analysis in positive
ionization mode. Time-of-day variation in the metabolites was screened for using orthogonal partial least square
discrimination between selected time points of 10:00 vs. 22:00 h, 16:00 vs. 04:00 h, and 07:00 (d 1) vs. 16:00 h, as well
as repeated-measures analysis of variance with time as an independent variable. Subsequently, cosinor analysis was
performed on all the sampled time points across the 24-h day to assess for significant daily variation. In this study,
analytical variability, assessed using known internal standards, was low with coefficients of variation <10%. A total of
1069 metabolite features were detected and 203 (19%) showed significant time-of-day variation. Of these, 34
metabolites were identified using a combination of accurate mass, tandem MS, and online database searches. These
metabolites include corticosteroids, bilirubin, amino acids, acylcarnitines, and phospholipids; of note, the magnitude of
the 24-h variation of these identified metabolites was large, with the mean ratio of oscillation range over MESOR (24-h
time series mean) of 65% (95% confidence interval [CI]: 4981%). Importantly, several of these human plasma
metabolites, including specific acylcarnitines and phospholipids, were hitherto not known to be 24-h variant. These
findings represent an important baseline and will be useful in guiding the design and interpretation of future
metabolite-based studies. (Author correspondence: Jooern.Ang@icr.ac.uk or Florence.Raynaud@icr.ac.uk)
Keywords: Acylcarnitines, Daily variation, Human, Liquid chromatographymass spectrometry, Metabolomics, Plasma
metabolites
INTRODUCTION
Metabolomics is the study of small molecule (<1 kDa)
metabolic profiles in biological systems, and comp-
lements genomic and proteomic approaches in providing
global views of biological processes. Metabolic profiles
capture endogenous and exogenous influences on a
living organism and may provide better representation
of its functional phenotype than changes in DNA, RNA,
and proteins (Allen et al., 2003; Nicholson et al., 2002).
Hence, metabolic perturbations caused by such disparate
factors as genetic changes, microbes, diseases, food, and
therapeutic interventions may be investigated using a
metabolomic approach (Allen et al., 2003; Nicholson
et al., 2002).
Recently published studies have identified clear and
pervasive circadian influence on the murine hepatic
metabolome (Eckel-Mahan et al., 2012; Fustin et al.,
2012). Plasma metabolites in a mouse model that vary
significantly with time-of-day have crucially been
Address correspondence to Dr. Joo Ern Ang or Dr. Florence Raynaud, Drug Metabolism, Pharmacokinetics & Metabolomics Team, Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United
Kingdom. Tel.: +44 (0)2087224383; Fax: +44 (0)2087224309; E-mail: Jooern.Ang@icr.ac.uk(Joo Ern Ang) or Florence.Raynaud@icr.ac.uk
(Florence Raynaud)
Submitted January 12, 2012, Returned for revision February 16, 2012, Accepted May 22, 2012
Chronobiology International, 29(7): 868881, (2012)
Copyright © Informa Healthcare USA, Inc.
ISSN 0742-0528 print/1525-6073 online
DOI: 10.3109/07420528.2012.699122

Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
identified in a landmark study; 14 oscillating metabolites
were identified by liquid chromatographymass spec-
trometry (LC-MS) and 28 by capillary electrophoresis
mass spectrometry (Minami et al., 2009). Direct trans-
lation of these animal data to humans, however, cannot
be made. In addition to the obvious problem of compar-
ing diurnal and nocturnal species, the timing and
amount of feeding, activity, rest, sleep, and posture
were not controlled in these animal experiments.
Although daily rhythms regulate multiple aspects of
human physiology, rhythmic control of the plasma meta-
bolome remains poorly understood. Previous studies
have investigated time-of-day variation in the human me-
tabolome by comparing metabolomic profiles of morning
with early evening urine samples (Slupsky et al., 2007;
Walsh et al., 2006). Although both studies identified sig-
nificant changes in metabolite levels, such as increased
creatinine and dimethylamine in the morning samples,
comparison of samples collected at only two time points
does not provide comprehensive overview of variation
across the 24-h day. Furthermore, these studies did not
control for sleep, activity, medication, alcohol/caffeine
intake, or environmental lighting, and thus, the results
obtained are extremely unlikely to show nonconfounded
time-of-day variation. To overcome this, a recent study
employed a targeted platform to study the human
plasma metabolome under controlled laboratory con-
ditions (Dallmann et al., 2012). The analytical platform,
however, was limited to 281 metabolites, and plasma
samples from all subjects were pooled at each time
point, precluding analyses of intersubject variability and
intrasubject daily variation and potentially reducing
the sensitivity of this methodology. The current study
circumvented these limitations by the use of a global,
untargeted metabolomic approach in the analysis of indi-
vidual plasma samples collected from healthy human vol-
unteers at different time points. The aim of this proof-of-
concept study was characterization of 24-h variation of
the plasma metabolome in human subjects maintained
under highly controlled conditions of food, posture,
light/dark cycle, sleep/wake schedule, and prior exposure
to pharmacologic agents. The technological platform with
LC-MS used in this study has been previously validated by
our group with over a thousand metabolites in plasma
being reproducibly detected (Pandher et al., 2009, 2012).
MATERIALS AND METHODS
All aspects of the study were conducted in accordance
with the Declaration of Helsinki and conformed to
international ethical standards (Portaluppi et al., 2010).
A favorable ethical opinion was obtained from the
Surrey Research Ethics Committee and the University of
Surrey Ethics Committee. Written informed consent
was obtained from all participants.
Eligibility for the study was determined via self-
completed questionnaires, including General Health
Questionnaire, General Sleep Questionnaire, Horne-Östberg
Questionnaire, Pittsburgh Sleep Quality Index, Beck
Depression Inventory, and Epworth Sleepiness Scale,
to assess general health, sleep patterns, and diurnal pre-
ference. Full details of the screening process have been
previously presented (Otway et al., 2011). To be in-
cluded, subjects needed to report a regular sleep sche-
duleofbetween6and8hindurationandnotbe
extreme morning or evening chronotype according to
the Horne-Östberg Questionnaire (Horne & Östberg,
1976). Subjects were excluded if they were taking
regular medication or food supplements known to influ-
ence metabolism, inflammatory markers, endothelial
markers, sleep, or the circadian system, or if they con-
sumed more than four caffeinated beverages per day.
Subjects with a history of any of the following were
also excluded: (a) circadian or sleep disorder; (b)
metabolic, cardiovascular, or chronic infectious/inflam-
matory disease; (c) psychiatric or neurological disease;
and (d) drug and alcohol abuse.
Eight lean, male volunteers were recruited with a
mean age of 53.6 (SD ±6.0) yrs, mean body mass index
(BMI) of 23.2 kg/m
2
(SD ±1.4), and fasting glucose of
4.2 mmol/L (SD ±.7). One participant was a smoker,
but refrained from smoking for 1 wk prior to study. No
participant had undertaken shiftwork within 5 yrs or
crossed any time zones within 1 mo of the study. For 1
wk prior to the laboratory study, volunteers were required
to maintain scheduled daily meal times (monitored by
food diaries) and a fixed sleep/wake schedule (23:00
07:00 h), confirmed via wrist actigraphy (Actiwatch-L;
Cambridge Neurotechnology, Cambridge, UK), sleep
diaries, and calling a time-stamped voicemail (Otway
et al., 2011). Participants also abstained from eating
fatty or sugary foods and drinking alcohol or caffeine
throughout this baseline week. For the final 3 d of this
week, participants were provided with meals of specific
nutritional content: the daily calorific content was 1.5-
fold the basal metabolic rate (estimated in calories =
11.5 × body weight [kg] + 873), with 35% of energy
derived from fat (Schofield, 1985).
In-laboratory Session
The in-laboratory session was conducted at the Surrey
Clinical Research Centre. Following an adaptation night
in the laboratory, subjects were woken at 06:30 h and
commenced a 25-h experimental session throughout
which they maintained a semi-recumbent posture to
minimize the impact of exogenous factors on the
measured parameters. Subjects remained awake in
normal room lighting (range 440825 lux in the direction
of gaze) between 06:30 and 22:30 h and were allowed to
sleep between 22:30 and 06:30 h in 0 lux. During the
waking period, participants were provided with hourly
nutritional drinks (Fortisip; Nutricia, Schiphol, The
Netherlands) and were allowed to drink water ad
libitum. The hourly consumption of this drink met the
protein, carbohydrate, fat, and fiber requirements of
each participant. Daily energy intake was 1.1-fold basal
Time-of-Day Variation in Human Plasma Metabolites 
© Informa Healthcare USA, Inc.
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
metabolic rate spread equally over the waking hours.
Throughout the protocol, including overnight, blood
samples were collected via an indwelling cannula by a
qualified person, and attempts were made to minimize
any disruption of the participantssleep. Blood samples
were collected at selected time points (.5 mL/time point
for metabolomic analysis) into lithium heparin tubes,
and the plasma fraction was separated by centrifugation
(3000 × g, 10 min, 4°C) and stored at 80°C.
Reagents and Solutions
Water (LC-MS grade), acetonitrile (LC-MS grade), and
formic acid (Aristar grade) were purchased from Fisher
Scientic (Loughborough, UK), and leucine enkephalin
was purchased from Sigma (Poole, UK). External stan-
dards, creatine (CAS number: 57-00-1), and colchicine
(CAS number: 64-86-8), were purchased from Sigma.
Plasma Preparation
Samples from selected time points (07:00, 10:00, 16:00,
22:00, 04:00, and 07:00 h [d 2]) were extracted by
mixing 1 volume of heparinized plasma with 4 volumes
of methanol/ethanol 1:1, followed by centrifugation at
18 000 × gfor 15 min at 4°C.
Assessment of Analytical Variability
Pooled plasma from the eight human subjects from all
the sampled time points (i.e., 48 samples) served as
quality controls and was analyzed throughout the exper-
imental batch to continuously monitor the analytical
variability of the system. These quality-control samples
(1/10 injections) were spiked with 1 mM colchicine and
creatine. Variability of these spiked compounds and
endogenous metabolites, including carnitine, phenyl-
alanine, and lysophosphatidylcholine (lysoPC(16:0)),
were evaluated.
LC-MS
Experiments were carried out on two LC-MS systems,
namely Acquity UPLC coupled to QTOF Premier mass
spectrometer (Waters Corporation, Manchester, UK) and
Agilent 1290 Series UPLC connected to a hybrid quadru-
pole-time-of-flight Agilent 6510 mass spectrometer
(Agilent, Waldbronn, Germany); the first system was used
for full-scan analysis and the second system for MS/MS.
The robustness, reproducibility, and cross-platform vali-
dation of the two systems in studying the exo-metabolome,
including plasma, have been previously published
(Pandher et al., 2009, 2012). To minimize systematic
analytical drift from use of large sample numbers and as
the analytical reproducibility of the system is high, one
analytical replicate from each individual per time point
was used. An electrospray ionization source in positive
mode was used for both LC-MS setups in this proof-of-
concept study, as the ions detected in the positive mode
are known to represent a large proportion of the exo-meta-
bolome in our analytical system (Pandher et al., 2009).
Briefly, chromatographic separation was performed
on a Waters Acquity HSS T3 C18 (100 × 2.1 mm, internal
diameter [I.D.] 1.8 µm) column. Mobile phase A was LC-
MS-grade water containing .1% formic acid and mobile
phase B was LC-MS-grade acetonitrile containing .1%
formic acid. The column and the autosampler were
maintained at a temperature of 50°C and 4°C, respect-
ively. A 13-min linear gradient elution was performed
as follows: 100% mobile phase A for the first .5 min, chan-
ging to 100% B over 7.5 min, holding at 100% B up to 9.5
min, and finally back to 100% A at 10 min and holding for
3 min. The flow rate was .6 mL/min, with an injection
volume of 10 µL. The MS instrument and data acquisition
parameters were as previously described (Pandher et al.,
2009, 2012).
Data Handling and Statistical Considerations
Raw data were detected, aligned, and processed using
MarkerLynx application manager software (version 4.1;
Waters, Milford, MA, USA), with parameters documented
previously (Pandher et al., 2009). Each metabolite
feature was characterized by a unique combination of
mass/charge ratio and retention time. The data matrix
obtained was subsequently subjected to multivariate stat-
istical analysis using (i) SIMCA-P v11.0 software (Ume-
trics AB, Umeå, Sweden): metabolite features that were
differentially expressed in one or more of three chosen
sets of selected time points10:00 vs. 22:00 h, 16:00 h
vs. 04:00 h, and 07:00 h (d 1) vs. 16:00 hwere identified
using orthogonal partial least squares-discriminant
analysis (OPLS-DA) with a low threshold of |p(corr)| > .5
on the OPLS-DA S-plot (Wiklund et al., 2008); and (ii)
repeated-measures analysis of variance (ANOVA) with
time as an independent variable; statistical significance
was deemed to be achieved at p< .05. Extracted ion chro-
matograms (EICs) of the selected metabolite features
were then generated using QuanLynx application
manager software (version 4.1; Waters). Finally, cosinor
analysis using the mean peak height of EICs of all metab-
olite features of interest at each time point by the method
of least squares (period of 24 h) was carried out to derive
estimates by the cosine curve approximation of
MESOR (24-h time series mean), amplitude (one-half
peak-to-trough variation), acrophase (peak) time, and
pvalue for test of the null hypothesis that the amplitude
of the fitted curve was 0; (Nelson et al., 1979); rhythm
detection was considered statistically significant when
p< .05 for the zero-amplitude test.
Metabolite Identification
The accurate mass and tandem MS fragmentation
pattern of each metabolite feature of interest was
ascertained and identification performed by database
searching (including Human Metabolome Database,
Lipid maps, and Metlin) and/or comparison with pure
commercial standards. MS/MS was performed on the
Agilent system with a default iso-width (width half-
maximum of the quadrupole mass bandpass used
 J. E. Ang et al.
Chronobiology International
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
during MS/MS precursor isolation) of 4 m/zusing a fixed
collision energy of 15V and data acquired in the range of
50 to 800 Da.
RESULTS
Analytical Reproducibility
Within the pooled plasma quality-control samples, coef-
ficients of variation for endogenous metabolites (carni-
tine, phenylalanine, and lysoPC(16:0)) were 1.7%, 3.0%,
and 8%, respectively, whereas that of spiked exogenous
compounds (creatine and colchicine) were 3.6% and
7.7%, respectively.
Details of Workflow
Figure 1 summarizes our data analysis workflow. In the
present study, a total of 1069 metabolite features were
detected across all analyzed plasma samples. Of these,
318 features passed the OPLS-DA filter using three pair-
wise comparisons of 10:00 vs. 22:00 h, 16:00 vs. 04:00 h,
and 07:00 vs. 16:00 h. Subsequently, 167 features were
confirmed to be significantly 24-h variant using cosinor
analysis of EIC data ( p< .05). In parallel, using repeated-
measures ANOVA, 254 putative features were detected,
and 203 were confirmed to exhibit 24-h variation. The
203 confirmed features detected by repeated-measures
ANOVA represented the sum total of all the temporally
variant features detected in this study (19% of all detected
features in this study) and included all 167 features ident-
ified by OPLS-DA and 36 features additionally identified
by repeated-measures ANOVA.
Of a total of 203 24-h variant features, the levels of 110
(54%) were significantly different between 16:00 and
04:00 h, whereas those of 68 (33%) and 65 (32%) were sig-
nificantly different between 10:00 and 22:00 h, and 07:00
and 16:00 h, respectively. Repeated-measures ANOVA
detected 36 features unique to those obtained from the
stated paired comparisons. These results are summar-
ized in Supplementary Figure 1 and Supplementary
Table 1. Using a combination of accurate mass, tandem
MS, and online database searches, the identities of 34
metabolites were determined from the 203 rhythmic
features. Fragmentation patterns and properties of
these compounds on our LC-MS system are summarized
in Table 1.
Variation of Human Plasma Metabolite Levels Across
Time-of-Day
Metabolites showing significant 24-h variation were from
a variety of chemical classes and included acylcarnitines,
lysophospholipids, bilirubin, corticosteroids, and amino
acids. For these identified compounds, the mean ratio
of oscillation range relative to the MESOR was 65%
(95% confidence interval [CI]: 4981%). Biological varia-
bility was consistently greater than analytical variability;
the mean analytical coefficient of variation of these ions
was 8% (95% CI: 610%).
The fitted peak times of the identified 24-h varying
plasma metabolites were spread across the day, but
appeared to be clustered around early morning, after-
noon, and evening (Table 1). Levels of long-chain un-
saturated acylcarnitines (C14:1, C14:2, and C18:1)
peaked before the long-chain saturated acylcarnitines
(C10, C12, C14, C16, and C18) in the morning,
whereas the acrophases of short-chain acylcarnitines
were observed at radically different times across the
day (C2 at 4.9 h, C6 at 5.4 h, C3 at 14.3 h, and C4 at
17.7 h). Bilirubin and cortisol peaked after the start
of the light phase, whereas the levels of detected
amino acids, such as methionine, tyrosine, proline,
lysine, phenylalanine, and leucine, were highest from
mid- to late afternoon. By contrast, levels of carnitine,
alanine, arginine, tryptophan, and valine (Supplemen-
tary Table 1) did not vary significantly over the 24 h.
Of the detected phospholipids, lysophosphatidyletha-
nolamines (lysoPEs) peaked in the late afternoon
and early evening, followed by the phosphatidyl-
cholines (PCs), which peaked later in the evening.
In contrast, two other phosphocholines (lysoPC(16:0)
and lysoPC(18:1)) (Supplementary Table 2) detected
in our analytical system had levels that did not
change significantly over the day.
The peak height versus time profiles of four selected
metabolites are presented in Figure 2, whereas Figure 3
additionally shows the time profile of one of the metabolites
of interest, acetylcarnitine, in each of the eight subjects.
DISCUSSION
In this study, an untargeted, global metabolomic
approach was employed to discover novel 24-h rhythmic
FIGURE 1. Flowchart summarizing data analysis workflow.
Time-of-Day Variation in Human Plasma Metabolites 
© Informa Healthcare USA, Inc.
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
metabolites in human plasma obtained under highly
controlled conditions. In this context, a UPLC-QTOF
MS platform was chosen for its high reproducibility, sen-
sitivity, and mass accuracy. It is stressed that rigorous
conditions of sample collection are necessary, as the
plasma metabolome is known to be exquisitely sensitive
to a broad range of intrinsic and extrinsic factors (Lawton
et al., 2008; Lewis et al., 2010; Psychogios et al., 2011).
A panel of 24-h rhythmic metabolites from a range
of chemical classes were identified, including bilirubin,
corticosteroids, amino acids, acylcarnitines, and phos-
pholipids. Nineteen percent of the metabolite features
detected using our analytical platform exhibited signifi-
cant daily variation, with the mean oscillation range
over MESOR ratio of 65% (95% CI: 4981%) in the ident-
ified metabolites, pointing to the presence of clinically
significant time-of-day biological variation.
In order to identify daily rhythms in metabolites, two
sets of time points 12-h apart were selected for compari-
son, the rationale being they have a periodicity of 24 h
and typically oscillate in a manner that approximates a
cosine curve with the peak and nadir occurring 12 h
out of phase. To identify such rhythms in metabolites,
sets of time points 912 h apart were selected for
comparison to provide a good probability of detecting
something that approximates to the maximum temporal
difference. Repeated-measures ANOVA was additionally
used to capitalize on the paired nature of the data,
TABLE 1. LC-MS characteristics of identified, 24-h variant plasma metabolites and key parameters characterizing temporal variation
Identification
Mass
(Da)
Retention
time
(min)
Analytical
CV (%)
Elemental
composition Fragments
Oscillation
range/
MESOR (%)
Peak
time
(dec·h)
p
value
Octenoylcarnitine (C8:1) 285.20 3.2 4.9 C
15
H
27
NO
4
286.202, 227.1197, 85.0229, 60.077 7.5 1.4 .020
Acetylcarnitine (C2:0) 203.12 .4 4.2 C
9
H
17
NO
4
204.124, 85.010, 60.064 94.4 4.9 <.001
Oleoylcarnitine (C18:1) 425.36 6.3 9.7 C
25
H
47
NO
4
426.358, 85.032, 60.084 43.6 5.2 .039
Hexanoylcarnitine (C6:0) 259.18 2.8 15.1 C
13
H
25
NO
4
260.181, 201.115,
183.010, 85.029, 60.080
98.8 5.4 <.001
Tetradenenoylcarnitine
(C14:1)
369.30 5.2 6.9 C
21
H
39
NO
4
370.296, 85.029, 60.082 70.8 5.5 <.001
Tetradecadienoylcarnitine
(C14:2)
367.28 4.9 11.2 C
21
H
37
NO
4
368.280, 85.023, 60.076 63.8 6.1 .002
Palmitoylcarnitine (C16:0) 399.34 6.1 17.8 C
23
H
45
NO
4
400.343, 341.268, 85.029, 60.081 109.8 6.4 <.001
Stearoylcarnitine (C18:0) 427.37 6.7 8.5 C
25
H
49
NO
4
428.375, 369.304, 85.030, 60.0811 45.6 6.5 .019
Tetradecanoylcarnitine
(C14:0)
371.31 5.6 12.8 C
21
H
41
NO
4
372.309, 313.235, 85.028, 60.081 146.0 6.6 <.001
Dodecanoylcarnitine
(C12:0)
343.28 5.0 10.3 C
19
H
37
NO
4
344.276, 285.201, 133.085, 85.027,
60.079
122.6 7.1 <.001
Bilirubin 584.27 8.2 5.0 C
33
H
36
N
4
O
6
585.274, 568.243, 299.139, 285.123 51.2 7.1 <.001
Octanoylcarnitine (C8:0) 287.21 3.6 7.4 C
15
H
29
NO
4
288.213, 85.027, 60.079 68.6 7.4 .003
Decanoylcarnitine (C10:0) 315.25 4.3 9.1 C
17
H
33
NO
4
316.249, 85.029, 60.080 77.2 7.8 <.001
Cortisol 362.22 3.7 8.1 C
21
H
30
O
5
363.209, 327.194, 121.063 194.6 8.4 <.001
Cortisone 360.19 3.8 19.5 C
21
H
28
O
5
361.201, 301.118, 163.071, 105.068 143.0 8.6 <.001
LysoPC(18:4) 515.31 5.5 3.0 C
25
H
46
NO
7
P 516.306, 184.976, 104.107, 86.095 9.8 14.1 .020
Propionylcarnitine (C3:0) 217.14 .5 3.3 C
10
H
19
NO
4
218.132, 100.986, 85.014, 60.076 28.3 14.3 .009
Methionine 149.06 .5 6.5 C
5
H
11
NO
2
S 150.057, 133.031, 104.054, 61.010,
56.050
20.2 15.3 <.001
Tyrosine 181.08 .5 10.3 C
9
H
11
NO
3
182.086, 165.051, 136.075,
123.042, 119.050, 91.054
28.4 15.5 <.001
LysoPE(16:0) 453.29 5.9 2.3 C
21
H
44
NO
7
P 454.290, 313.295, 62.058 27.8 16.5 .003
Proline 115.07 .4 3.1 C
5
H
9
NO
2
116.067, 70.056 47.8 16.6 <.001
LysoPE(18:3) 475.28 5.9 11.2 C
23
H
42
NO
7
P 476.276, 335.269 35.0 16.9 .023
Butyrylcarnitine (C4:0) 231.16 1.9 9.9 C
11
H
21
NO
4
232.151, 85.026, 60.082 53.4 17.7 <.001
LysoPE(20:4) 501.30 6.1 17.7 C
25
H
44
NO
7
P 502.294, 484.283, 361.274, 62.059 113.2 18.3 <.001
LysoPE(18:2) 477.29 5.7 13.2 C
23
H
44
NO
7
P 478.292, 337.270 110.4 18.8 <.001
LysoPE(18:1) 479.31 6.1 2.2 C
23
H
46
NO
7
P 480.309, 339.289, 62.060 132.0 19.1 <.001
Lysine 146.11 6.8 .1 C
6
H
14
N
2
O
2
147.113, 130.088, 84.082 7.8 19.1 .014
Phenylalanine 165.09 1.8 2.6 C
9
H
11
NO
2
166.085, 120.081 18.6 19.3 <.001
LysoPC(18:2) 519.34 5.7 2.5 C
26
H
50
NO
7
P 520.340, 184.073, 104.107, 86.096 44.6 19.6 .001
LysoPC(20:5) 541.32 5.7 2.3 C
28
H
48
NO
7
P 542.324, 524.313, 184.073,
104.107, 86.097
23.7 19.8 .04
LysoPC(20:3) 545.69 6.0 18.6 C
28
H
52
NO
7
P 546.354, 184.072, 104.107, 86.096 57.1 20.0 .02
LysoPC(18:3) 517.32 5.4 5.9 C
26
H
48
NO
7
P 518.324, 184.071, 86.096 67.2 20.5 .002
Leucine 131.10 .5 3.9 C
6
H
13
NO
2
132.102, 86.098, 44.051 15.6 21.5 .048
LysoPC(20:1) or PC
(18:1/2:0)
549.39 6.7 8.9 C
28
H
56
NO
7
P 550.385, 184.070, 104.105, 86.098 26.0 23.1 .020
 J. E. Ang et al.
Chronobiology International
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
FIGURE 2. Time profiles of four plasma metabolites with different acrophases: (a) acetylcarnitine, (b) LysoPE(18:1), (c) proline, and (d)
cortisol. On the horizontal axis, black bar indicates lights-off (0 lux) and white bar lights-on (440825 lux). Internal standard (IS) shows the
analytical variation of each ion in the pooled, replicate samples analyzed throughout the LC-MS run.
FIGURE 3. Plasma acetylcarnitine profiles for eight individuals maintained under controlled light/dark, sleep/wake, posture, and calorific
intake conditions. Black bar indicates lights-off (0 lux) and white bar lights-on (440825 lux).
Time-of-Day Variation in Human Plasma Metabolites 
© Informa Healthcare USA, Inc.
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
i.e., plasma was collected from eight individuals across
time. This latter method was the most sensitive and
yielded 36 features in addition to OPLS-DA comparisons
(Supplementary Figure 1). Not only did this validate
results already obtained by OPLS-DA, but it also demon-
strated the effect of intersubject variability as well as the
relatively higher sensitivity of this approach (Supplemen-
tary Figure 1). To illustrate this point, the 24-h temporal
profile of a metabolite feature is presented in Sup-
plementary Figure 2, which highlights the significant
intersubject variation in this ion compared to intrasub-
ject time-of-day changes. In principle, other multivariate
tools, such as the multilevel partial least square projec-
tion to latent structures (PLS), could be used instead of
OPLS-DA for each pairwise comparison in the screening
step, as it offers greater statistical power (Westerhuis
et al., 2010). In this study, multilevel PLS was not
carried out; instead, repeated-measures ANOVA was per-
formed, making use of all data across all time points.
Our data extend previous findings that an unbiased
metabolomic platform may be used to identify 24-h
variant metabolites (Dallmann et al., 2012; Eckel-
Mahan et al., 2012; Fustin et al., 2012; Minami et al.,
2009; Slupsky et al., 2007; Walsh et al., 2006); some of
the 24-h variant plasma metabolites detected in this
study are already known to show such variation, includ-
ing bilirubin, cortisol, and several amino acids (Eriksson
et al., 1989; Feigin et al., 1967, 1968; Larsson et al., 2009;
Selmaoui & Touitou, 2003; Wurtman et al., 1968), and
this provides further validation of this approach. It is
also noteworthy that numerous metabolites linked to
major metabolic pathways were identified in this study;
the 24-h variant nature of these metabolites has not
been previously demonstrated.
Rhythms in human plasma levels of amino acids have
been previously reported (Eriksson et al., 1989; Feigin
et al., 1967, 1968; Wurtman et al., 1968). Typically,
maximal concentrations were observed in the afternoon/
evening, and minimal concentrations in the early hours
of the morning before waking. Our results are in keeping
with these findings: of the detected amino acids, lysine,
proline, leucine, methionine, phenylalanine, and tyrosine
exhibited significant day/night variation with similar
peak/trough temporal changes. The mechanisms regulat-
ing this 24-h rhythmicity remain obscure, and they may
be related to the periodicity of many other metabolic pro-
cesses (Eckel-Mahan et al., 2012; Feigin et al., 1971). For in-
stance, the tricarboxylic acid cycle, gluconeogenesis, and
lipogenesis utilize amino acid carbon backbones and
may be important in this context.
The present study identified numerous plasma acyl-
carnitines that demonstrated significant time-of-day vari-
ation; only a subset of these has thus far been reported
(Dallmann et al., 2012). Acylcarnitines are key intermedi-
ates in the β-oxidation of fatty acids in mitochondria
(Kompare & Rizzo, 2008), and abnormal levels of these
metabolites have been linked to errors of metabolism
involving fatty acid oxidation and carnitine cycle
(Pasquali et al., 2006). Costa et al. (1999) reported pre-
ferential increase in levels of specific mono-unsaturated
acylcarnitines in fasting individuals, who generally
display increased levels of blood unesterified fatty
acids and hepatic β-oxidation. Fasting alone, however,
is unlikely to account for the changes observed in the
current study, as levels of many of these metabolites
peaked and started to decrease in the morning before
lights were switched on and feeding of hourly caloric
drinks recommenced. Critical components of the bio-
logical system regulating the levels of acylcarnitines
also demonstrate 24-h variation. For instance, using
gene expression profiling data of liver enzymes across
the time-of-day in a mouse model, mRNA levels of key
transporters of long-chain acylcarnitines, including car-
nitine palmitoyltransferase (CPT) 1a and 2, were shown
to exhibit clear day/night oscillation (Hughes et al.,
2009).
It is interesting to note that the time-of-day pattern of
variation differs between short-chain, unsaturated long-
chain, and saturated long-chain acylcarnitines. Levels of
long-chain unsaturated acylcarnitines (C18:1, C14:1, and
C14:2) peaked early in the morning before the long-
chain saturated acylcarnitines (C10, C12, C14, C16, and
C18), whereas the acrophases of short-chain acylcarni-
tines were observed at radically different times, i.e., C2 at
4.9 h, C6 at 5.4 h, C3 at 14.3 h, and C4 at 17.7 h. Abnorm-
alities in particular enzyme and membrane transporter
systems have already been associated with pathognomo-
nic changes in acylcarnitine subtypes in well-defined sub-
types of inborn errors of metabolism (Santra & Hendriksz
2010). Hence, it is plausible that short- and long-chain acyl
dehydrogenases and transporters are regulated differently
across time. Indeed, preliminary evidence suggests the
presence of such a specific regulatory process. Whereas
mRNA levels of CPT1a are rhythmic with an acrophase
just before lights-off in the mouseend of the inactivity
span, levels of long-chain acylcarnitines transported by
this mitochondrial membrane enzyme peak at the end
of the dark phase in human subjectsalso the end of
inactivity span (selected profiles are juxtaposed in time
relative to the activity/rest rhythm of the respective
species in Figure 4). Given that humans and mice are
diurnal and nocturnal species, respectively, agreement
of these patterns of temporal change suggest that (CPT)
1a may be responsible for the 24-h variation observed in
long-chain acylcarnitines. By contrast, expression profiles
of (CPT) 1b and (CPT) 1c did not show significant time-of-
day difference in the mouse, lending additional support
for the hypothesis that (CPT) 1a may be the specific
rhythmic driver of long-chain acylcarnitines (Hughes
et al., 2009). It is important to point out that these cross-
species associations, although interesting, are tentative
and require further biological validation.
In this study, acrophases of specific C16, C18, and C20
lysoPCs and lysoPEs clustered in the afternoon and
evening; no clear exogenous factor related to this could
be identified, including food consumption. Two other
 J. E. Ang et al.
Chronobiology International
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
endogenously expressed plasma phospholipids were
assessed and showed no significant time-of-day
change, suggesting possible specificity of the rhythmic
driver for particular subtypes of lysophospholipids. Of
note, temporal variation in plasma lysophospholipid
levels has also been detected in mouse plasma
(Minami et al., 2009). Enzyme systems currently under-
stood to be mainly responsible for production of these
metabolites are the lecithin:cholesterolacyltransferase
(Lcat), secreted phospholipase A
2
(Pla2), and endothelial
lipase (Lipg) systems (Yamamoto et al., 2011); in a mouse
model, Lcat,Pla2g12a, and Lipg are rhythmically
expressed in the liver (Hughes et al., 2009).
The current results will be of use in a number of areas,
for example, in the evaluation of pharmacodynamic bio-
markers of new drug therapies (Sarker & Workman,
2007). Evaluation of pharmacodynamic biomarkers is
made by drawing intrasubject comparisons across time
with the patient pre-dose used as a control. This
approach, however, may be confounded by the patients
internal circadian and homeostatic systems and external
influences, such as dietary intake. The approach taken
in the current study typifies a way to validate such
pharmacodynamic studies by defining metabolites that
are rhythmic and those that are not.
The findingsof our study have implications on the use of
plasma metabolites in clinical testing. At least 19% of the
metabolite features in our study exhibited significant 24-
h variation, with the mean oscillation range over MESOR
ratio of 65% (95% CI: 4981%) in the identified metabolites.
To improve the predictive utility of clinical biomarkers
used in diagnosis, prognosis, and follow-up,determination
of 24-h variability should become part of the clinical
validation process, especially in the ascertainment of the
degree of 24-h variation relative to the factor(s) in question.
A limitation of the present study is that although a
global, untargeted approach was adopted, only a pro-
portion of the entire plasma metabolome was monitored
using our analytical method. Despite this, the primary ob-
jective of identifying and characterizing novel 24-h variant
plasma metabolites wasmet in this proof-of-concept study.
Indeed, the present study identified all the 24-h variant
metabolites detected by LC-MS in positive ionization
mode in the Dallmann et al. (2012) study that was con-
ducted under constant routineconditions of constant
dim lighting, continuous hourly food supplement, and
no sleep, thereby providing external validation of their
data. Significantly, additional 24-h variant plasma metab-
olites were identified in the present study, suggesting that
these metabolites may be affected by lighting, sleep, and/
or feeding condition. Alternatively, as the samples in the
Dallmann et al. (2012) study were pooled for each
studied time point, the nondetection of these additional
metabolites could be due to lower sensitivity of their ap-
proach. Although only eight subjects were studied with
six time points across a 24-h day, multiple factors that
could confound the rhythms were strictly controlled for.
Specifically, study subjects were lean, healthy, and unme-
dicated and who maintained a fixed sleep/wake schedule
and dietary regime for 1 wk at home prior to an adaptation
night, followed bya 25-h session in the laboratory when the
light/dark and sleep/wake cycle, posture, and calorific
intake were rigorously controlled. In this context, it would
be of interest to disentangle the impact of food, sleep,
and darkness, and prospective clinical studies are
ongoing to addressthis. In addition, the study participants
were all middle-aged men, and the applicability of these
findings to females and other age groups is unclear.
In summary, we have successfully applied the use of
an untargeted metabolomic approach to identify and
characterize 24-h variant metabolites from a range of
chemical classes. These findings represent an important
baseline and will be useful in guiding the proper design
and interpretation of future metabolite-based studies.
Declaration of Interest: The authors report no conflicts
of interest. The authors alone are responsible for the
content and writing of the paper.
This study was funded by Diabetes-UK (grant 08/
0003607), Biotechnology and Biological Sciences Research
FIGURE 4. (Top panel) The temporal profile of palmitoylcarnitine
in human plasma. (Bottom panel) CPT1a gene expression in a
mouse model from an independent study (Hughes et al., 2009).
Black/gray bars indicate lights-off and the white bar lights-on.
Time-of-Day Variation in Human Plasma Metabolites 
© Informa Healthcare USA, Inc.
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
Council (grants BB/D526853/1 and BB/I019405/1) and
Cancer Research UK (grants C309/A8274 and C309/
A2187). J.E.A. receives funding support from the Wellcome
Trust (grant 090952/Z/09/Z) and D.J.S. is a Royal Society
Wolfson Research Merit Award holder. The funders had
no role in study design, data collection and analysis,
decision to publish, or preparation of the manuscript.
REFERENCES
Allen J, Davey HM, Broadhurst D, Heald JK, Rowland JJ, Oliver SG, Kell
DB. (2003). High-throughput classification of yeast mutants for
functional genomics using metabolic footprinting. Nat. Biotechnol.
21:692696.
Costa CC, de Almeida IT, Jakobs C, Poll-The BT, Duran M. (1999).
Dynamic changes of plasma acylcarnitine levels induced by
fasting and sunflower oil challenge test in children. Pediatr. Res.
46:440444.
Dallmann R, Viola AU, Tarokh L, Cajochen C, Brown SA. (2012). The
human circadian metabolome. Proc. Natl. Acad. Sci. U. S. A.
109:26252629.
Eckel-Mahan KL, Patel VR, Mohney RP, Vignola KS, Baldi P, Sassone-
Corsi P. (2012). Coordination of the transcriptome and metabo-
lome by the circadian clock. Proc. Natl. Acad. Sci. U. S. A.
109:55415546.
Eriksson T, Voog L, Walinder J, Eriksson TE. (1989). Diurnal rhythm in
absolute and relative concentrations of large neutral amino acids
in human plasma. J. Psychiatr. Res. 23:241249.
Feigin RD, Klainer AS, Beisel WR. (1967). Circadian periodicity of
blood amino-acids in adult men. Nature 215:512514.
Feigin RD, Klainer AS, Beisel WR. (1968). Factors affecting circadian
periodicity of blood amino acids in man. Metabolism 17:764775.
Feigin RD, Beisel WR, Wannemacher RW (1971). Rhythmicity of
plasma amino acids and relation to dietary intake. Am. J. Clin.
Nutr. 24:329341.
Fustin M, Doi M, Yamada H, Komatsu R, Shimba S, Okamura H.
(2012). Rhythmic nucleotide synthesis in the liver: temporal segre-
gation of metabolites. Cell Reports 1:341349.
Horne JA, Östberg O. (1976). A self-assessment questionnaire to
determine morningness-eveningness in human circadian rhythms.
Int. J. Chronobiol 4:97110.
Hughes ME, DiTacchio L, Hayes KR, Vollmers C, Pulivarthy S, Baggs JE,
Panda S, Hogenesch JB. (2009). Harmonics of circadian gene
transcription in mammals. PLoS Genet. 5:e1000442.
Kompare M, Rizzo WB. (2008). Mitochondrial fatty acid oxidation
disorders. Semin. Pediatr. Neurol. 15:140149.
Larsson A, Hassan M, Ridefelt P, Axelsson J. (2009). Circadian variabil-
ity of bilirubin in healthy men during normal sleep and after an
acute shift of sleep. Chronobiol. Int. 26:16131621.
Lawton KA, Berger A, Mitchell M, Milgram KE, Evans AM, Guo L, Hanson
RW, Kalhan SC, Ryals JA, Milburn MV. (2008). Analysis of the adult
human plasma metabolome. Pharmacogenomics 9:383397.
Lewis GD, Farrell L, Wood MJ, Martinovic M, Arany Z, Rowe GC, Souza
A, Cheng S, McCabe EL, Yang E, Shi X, Deo R, Roth FP, Asnani A,
Rhee EP, Systrom DM, Semigran MJ, Vasan RS, Carr SA, Wang TJ,
Sabatine MS, Clish CB, Gerszten RE. (2010). Metabolic signatures
of exercise in human plasma. Sci. Transl. Med. 2:33ra37.
Minami Y, Kasukawa T, Kakazu Y, Iigo M, Sugimoto M, Ikeda S, Yasui
A, van der Horst GT, Soga T, Ueda HR. (2009). Measurement of
internal body time by blood metabolomics. Proc. Natl. Acad.
Sci. U. S. A. 106:98909895.
Nelson W, Tong YL, Lee JK, Halberg F. (1979). Methods for cosinor-
rhythmometry. Chronobiologia 6:305323.
Nicholson JK, Connelly J, Lindon JC, Holmes E. (2002). Metabonomics:
a platform for studying drug toxicity and gene function. Nat. Rev.
Drug Discov. 1:153161.
Otway DT, Mantele S, Bretschneider S, Wright J, Trayhurn P, Skene DJ,
Robertson MD, Johnston JD. (2011). Rhythmic diurnal gene
expression in human adipose tissue from individuals who are
lean, overweight, and type 2 diabetic. Diabetes 60:15771581.
Pandher R, Ducruix C, Eccles SA, Raynaud FI. (2009). Cross-platform
Q-TOF validation of global exo-metabolomic analysis: application
to human glioblastoma cells treated with the standard PI 3-
Kinase inhibitor LY294002. J. Chromatogr. B Analyt. Technol.
Biomed. Life Sci. 877:13521358.
Pandher R, Naegele E, Fischer SM, Raynaud FI. (2012). Improvement
in the number of analytic features detected by non-targeted meta-
bolomic analysis: influence of the chromatographic system and
ionisation technique. In Metabolomics. Rijeka, Croatia: InTech
Europe, 317328
Pasquali M, Monsen G, Richardson L, Alston M, Longo N. (2006). Bio-
chemical findings in common inborn errors of metabolism.
Am. J. Med. Genet. C Semin. Med. Genet. 142C:6476.
Portaluppi F, Smolensky MH, Touitou Y. (2010). Ethics and methods
for biological rhythm research on animals and human beings.
Chronobiol. Int. 27:19111929.
Psychogios N, Hau DD, Peng J, Guo AC, Mandal R, Bouatra S, Sinelni-
kov I, Krishnamurthy R, Eisner R, Gautam B, Young N, Xia J, Knox
C, Dong E, Huang P, Hollander Z, Pedersen TL, SmithSR, Bamforth
F, Greiner R, McManus B, Newman JW, Goodfriend T, Wishart DS.
(2011). The human serum metabolome. PLoS ONE 6:e16957.
Santra S, Hendriksz C. (2010). How to use acylcarnitine profiles to help
diagnose inborn errors of metabolism. Arch. Dis. Child Educ. Pract.
Ed. 95:151156.
Sarker D, Workman P. (2007). Pharmacodynamic biomarkers for mol-
ecular cancer therapeutics. Adv. Cancer Res. 96:213268.
Schofield WN. (1985). Predicting basal metabolic rate, new standards
and review of previouswork. Hum. Nutr. Clin.Nutr. 39(Suppl1):541.
Selmaoui B, Touitou Y. (2003). Reproducibility of the circadian
rhythms of serum cortisol and melatonin in healthy subjects: a
study of three different 24-h cycles over six weeks. Life Sci.
73:33393349.
Slupsky CM, Rankin KN, Wagner J, Fu H, Chang D, Weljie AM, Saude
EJ, Lix B, Adamko DJ, Shah S, Greiner R, Sykes BD, Marrie TJ.
(2007). Investigations of the effects of gender, diurnal variation,
and age in human urinary metabolomic profiles. Anal. Chem.
79:69957004.
Walsh MC, Brennan L, Malthouse JP, Roche HM, Gibney MJ. (2006).
Effect of acute dietary standardization on the urinary, plasma,
and salivary metabolomic profiles of healthy humans.
Am. J. Clin. Nutr. 84:531539.
Westerhuis JA, van Velzen EJ, Hoefsloot HC, Smilde AK. (2010).
Multivariate paired data analysis: multilevel PLSDA versus
OPLSDA. Metabolomics 6:119128.
Wiklund S, Johansson E, Sjostrom L, Mellerowicz EJ, Edlund U,
Shockcor JP, Gottfries J, Moritz T, Trygg J. (2008). Visualization of
GC/TOF-MS-based metabolomics data for identification of
biochemically interesting compounds using OPLS class models.
Anal. Chem. 80:115122.
Wurtman RJ, Rose CM, Chou C, Larin FF. (1968). Daily rhythms in
the concentrations of various amino acids in human plasma.
N. Engl. J. Med. 279:171175.
Yamamoto K, Isogai Y, Sato H, Taketomi Y, Murakami M. (2011).
Secreted phospholipase A2, lipoprotein hydrolysis, and athero-
sclerosis: integration with lipidomics. Anal. Bioanal. Chem.
400:18291842.
 J. E. Ang et al.
Chronobiology International
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
SUPPLEMENTARY FIGURE 1. Venn diagram showing overlapping sets of metabolite features detected by OPLS-DA of paired time points
and repeated-measures ANOVA across all time points.
SUPPLEMENTARY FIGURE 2. Plasma profiles of metabolite feature with mass/charge 455.19 Da and retention time 5.7 min for eight indi-
viduals detected by repeated-measures ANOVA but not pairwise OPLS-DA comparisons, illustrating higher intersubject variability relative
to intrasubject time-of-day variation. All participants were maintained under controlled light/dark, sleep/wake, posture, and calorific intake
conditions. Black bar indicates lights-off (0 lux) and white bar lights-on (440825 lux).
Time-of-Day Variation in Human Plasma Metabolites 
© Informa Healthcare USA, Inc.
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
SUPPLEMENTARY TABLE 1. List of metabolite features detected in the screening process and considered for tandem MS
Metabolite feature Comparison made
Retention time (min) Mass/charge (Da) 16h vs. 04h 10h vs. 22h 07h vs. 16h Repeated-measures ANOVA
.31 198.94 0 1 0 1
.31 200.97 1 0 0 1
.33 288.92 0 1 0 1
.34 327.05 1 0 0 1
.34 566.89 0 1 0 1
.34 634.88 1 0 0 1
.34 498.90 1 0 0 1
.38 200.86 0 0 0 1
.38 134.02 1 0 0 1
.39 164.03 0 1 0 1
.41 103.01 1 0 0 1
.41 116.07 1 0 0 1
.41 117.08 0 0 0 1
.41 219.03 0 0 0 1
.41 221.01 0 0 0 1
.41 365.11 1 0 1 1
.41 430.70 1 0 0 1
.41 204.12 1 0 1 1
.43 218.14 1 0 0 1
.43 229.16 1 0 1 1
.43 335.92 0 0 1 1
.44 133.00 1 0 0 1
.44 194.02 1 0 0 1
.46 135.00 1 0 0 1
.46 150.06 1 0 0 1
.47 182.08 1 0 0 1
.49 149.02 1 0 1 1
.49 132.10 0 1 0 1
.50 337.02 1 0 0 1
.55 226.05 1 0 0 1
.57 254.16 0 0 1 1
.62 168.99 1 0 0 1
1.66 254.16 0 1 1 1
1.81 103.05 1 0 0 1
1.81 107.05 1 0 0 1
1.81 120.08 0 1 0 1
1.81 131.05 1 1 0 1
1.81 149.06 0 1 0 1
1.81 166.09 1 1 0 1
1.93 232.16 1 0 0 1
2.57 257.17 1 1 1 1
2.58 235.18 1 1 1 1
2.82 260.18 1 0 0 1
2.85 251.18 1 0 1 1
3.12 185.12 1 0 0 1
3.23 286.20 1 0 0 1
3.59 310.20 0 1 0 1
3.62 288.22 0 0 1 1
3.66 363.22 0 1 1 1
3.80 361.20 0 1 1 1
4.17 585.27 0 1 1 1
4.20 243.63 0 0 0 1
4.21 464.28 0 0 0 1
4.24 252.63 0 0 0 1
4.24 412.28 1 0 0 1
4.24 430.30 1 0 0 1
4.24 448.31 0 0 0 1
4.24 488.30 1 0 0 1
4.24 504.27 0 0 0 1
4.24 510.28 0 0 0 1
4.32 316.25 0 0 1 1
4.46 416.32 1 0 0 1
4.46 552.24 0 0 0 1
4.56 470.29 0 0 0 1
 J. E. Ang et al.
Chronobiology International
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
4.75 583.26 0 0 1 1
4.79 244.27 0 0 0 1
4.81 207.62 0 0 0 1
4.81 226.63 1 0 0 1
4.81 235.63 0 0 0 1
4.81 244.63 1 0 0 1
4.81 414.30 1 0 0 1
4.81 432.31 1 0 0 1
4.81 450.32 0 0 0 1
4.81 472.30 0 0 0 1
4.81 488.27 1 0 0 1
4.81 494.29 1 0 0 1
4.81 504.24 1 0 0 1
4.82 368.28 0 0 1 1
4.84 344.28 0 0 1 1
4.88 251.12 0 0 0 1
4.92 450.32 1 0 0 1
4.92 472.30 1 0 0 1
4.98 415.21 1 1 0 1
4.99 286.14 1 0 0 1
4.99 437.19 1 0 0 1
4.99 453.17 1 1 0 1
5.16 387.19 0 0 1 1
5.16 409.18 0 0 1 1
5.16 425.15 0 0 0 1
5.18 370.29 0 0 1 1
5.22 432.24 1 1 0 1
5.24 107.09 0 1 0 1
5.24 119.09 1 1 0 1
5.24 135.08 1 1 0 1
5.24 160.04 1 1 0 1
5.24 227.08 1 1 0 1
5.24 233.06 0 1 0 1
5.24 281.14 1 1 0 1
5.24 295.12 1 1 0 1
5.24 300.11 1 1 0 1
5.24 367.14 1 1 0 1
5.24 380.25 0 0 0 1
5.24 397.20 1 0 0 1
5.24 415.21 1 1 0 1
5.24 434.18 1 1 0 1
5.24 437.19 1 1 0 1
5.24 453.17 1 0 0 1
5.24 499.17 1 1 0 1
5.24 129.05 1 0 0 1
5.29 253.63 1 0 0 1
5.29 468.31 0 0 0 1
5.29 490.29 0 0 0 1
5.30 299.14 1 1 1 1
5.30 585.27 1 0 1 1
5.36 476.28 1 1 1 1
5.39 278.64 0 1 0 1
5.39 500.28 1 0 0 1
5.39 518.32 0 1 0 1
5.39 540.31 0 0 0 1
5.42 290.64 1 0 0 1
5.42 564.31 1 0 0 1
5.48 299.14 0 0 1 1
5.48 585.27 0 0 1 1
5.49 494.32 0 1 0 1
5.49 516.31 1 0 0 1
5.49 513.29 1 0 0 1
5.52 643.28 0 1 0 1
5.52 603.30 0 0 1 1
5.55 372.31 1 0 1 1
5.57 337.27 0 0 0 1
5.59 475.79 1 0 0 1
Continued
Time-of-Day Variation in Human Plasma Metabolites 
© Informa Healthcare USA, Inc.
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
SUPPLEMENTARY TABLE 1. Continued
Metabolite feature Comparison made
Retention time (min) Mass/charge (Da) 16h vs. 04h 10h vs. 22h 07h vs. 16h Repeated-measures ANOVA
5.61 184.07 0 1 0 1
5.61 385.28 0 0 0 1
5.65 398.33 0 0 0 1
5.67 284.23 0 0 0 1
5.67 516.24 1 1 0 1
5.67 497.24 1 1 1 1
5.68 455.19 0 0 0 1
5.68 460.28 1 1 1 1
5.68 473.20 1 1 1 1
5.68 478.30 1 1 1 1
5.68 500.28 1 1 1 1
5.68 526.30 0 0 0 1
5.68 471.16 0 0 1 1
5.69 548.28 0 0 0 1
5.71 520.34 1 1 1 1
5.71 542.32 0 0 1 1
5.71 590.33 1 0 1 1
5.71 798.97 0 1 0 1
5.71 799.47 0 0 0 1
5.71 279.64 1 0 0 1
5.77 580.30 0 0 1 1
5.85 318.24 0 0 1 1
5.88 313.27 0 0 0 1
5.88 436.28 1 0 0 1
5.88 454.29 1 0 0 1
5.88 476.28 1 0 0 1
5.95 546.36 0 0 0 1
5.96 339.29 1 1 1 1
5.98 400.34 1 1 1 1
6.07 457.20 1 0 1 1
6.07 462.30 1 1 1 1
6.07 475.22 1 1 1 1
6.07 480.32 1 1 1 1
6.07 502.30 1 1 1 1
6.08 520.30 0 0 1 1
6.23 405.26 1 0 0 1
6.27 426.35 1 0 1 1
6.47 428.37 0 0 1 1
6.51 459.32 1 0 0 1
6.54 504.31 0 0 0 1
6.56 273.67 0 0 1 1
6.64 419.28 1 0 0 1
6.71 294.67 0 1 0 1
6.72 550.39 0 1 0 1
6.72 572.37 0 1 0 1
6.82 147.11 0 0 1 1
6.83 650.44 1 1 0 1
6.87 672.42 1 1 0 1
6.93 268.26 1 0 0 1
6.98 279.23 1 1 0 1
7.17 329.25 1 0 1 1
7.27 285.93 0 1 0 1
7.29 350.25 0 1 1 1
7.40 563.55 0 0 0 1
7.54 163.15 0 1 0 1
7.59 455.34 0 0 1 1
7.59 354.20 1 0 1 1
7.59 395.22 0 0 1 1
7.59 561.41 0 0 1 1
7.69 339.18 1 0 1 1
7.70 669.50 1 0 0 1
7.74 284.30 1 0 0 1
7.84 381.30 0 0 1 1
7.92 298.31 0 0 0 1
 J. E. Ang et al.
Chronobiology International
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
8.08 780.55 1 0 0 1
8.20 299.14 1 1 1 1
8.20 584.26 0 1 1 1
8.20 585.27 0 1 1 1
8.20 583.26 0 0 1 1
8.21 581.25 0 0 1 1
8.25 389.25 0 1 0 1
8.37 312.33 0 1 1 1
When the feature is significant in a comparison, it is denoted 1and when it is not, it is denoted 0.
SUPPLEMENTARY TABLE 2. LC-MS features of metabolites of interest that did not exhibit significant 24-h variation ( p> .05)
Identification Retention time (min) Mass (Da) Elemental composition Fragments
Arginine .39 174.112 C
6
H
14
N
4
O
2
175.120, 158.070, 116.050, 70.100
Valine .41 117.079 C
5
H
11
NO
2
118.087, 72.070, 55.040
Carnitine .42 161.105 C
7
H
15
NO
3
162.113, 103.040, 85.030, 60.080
Alanine .42 89.048 C
3
H
7
NO
2
90.056, 44.050
Tryptophan 2.03 204.090 C
11
H
12
N
2
O
2
205.098, 188.069, 146.059, 118.064
LysoPC(16:0) 5.90 495.333 C
24
H
50
NO
7
P 496.340, 184.074, 104.105, 86.097
LysoPC(18:1) 6.09 521.348 C
26
H
52
NO
7
P 522.356, 184.073, 104.107, 86.095
Time-of-Day Variation in Human Plasma Metabolites 
© Informa Healthcare USA, Inc.
Chronobiol Int Downloaded from informahealthcare.com by 77.96.98.185 on 07/25/12
For personal use only.
... Timing of collection is another important choice to make for a successful metabolomics study as many metabolites are influenced by circadian rhythm, nutritional status, and other periodic conditions. Within the human metabolome, 15-40% of identifiable metabolites display varying levels impacted by circadian rhythm [32][33][34]. These rhythmic metabolites include fatty acids, lipids, and amino acids [32][33][34]. ...
... Within the human metabolome, 15-40% of identifiable metabolites display varying levels impacted by circadian rhythm [32][33][34]. These rhythmic metabolites include fatty acids, lipids, and amino acids [32][33][34]. The peak times of these metabolites vary throughout the day and are clustered around early morning, afternoon, and evening [33]. ...
... These rhythmic metabolites include fatty acids, lipids, and amino acids [32][33][34]. The peak times of these metabolites vary throughout the day and are clustered around early morning, afternoon, and evening [33]. Furthermore, the metabolome will also change depending on whether the person is fasting or recently ate a meal. ...
Article
Full-text available
As end-products of the intersection between the genome and environmental influences, metabolites represent a promising approach to the discovery of novel biomarkers for diseases. However, many potential biomarker candidates identified by metabolomics studies fail to progress beyond analytical validation for routine implementation in clinics. Awareness of the challenges present can facilitate the development and advancement of innovative strategies that allow improved and more efficient applications of metabolite-based markers in clinical settings. This minireview provides a comprehensive summary of the pre-analytical factors, required analytical validation studies, and kit development challenges that must be resolved before the successful translation of novel metabolite biomarkers originating from research. We discuss the necessity for strict protocols for sample collection, storage, and the regulatory requirements to be fulfilled for a bioanalytical method to be considered as analytically validated. We focus especially on the blood as a biological matrix and liquid chromatography coupled with tandem mass spectrometry as the analytical platform for biomarker validation. Furthermore, we examine the challenges of developing a commercially viable metabolomics kit for distribution. To bridge the gap between the research lab and clinical implementation and utility of relevant metabolites, the understanding of the translational challenges for a biomarker panel is crucial for more efficient development of metabolomics-based precision medicine.
... Under the ad libitum circumstances, the circadian clock, which synchronizes the behaviors and physiology including food taking and wake-sleep circle, is the most critical factor influencing the gut microbial dynamics 15 . As a result of rhythmic feed-taking behavior, the metabolites in the serum undergo robustly oscillating and are relevant to the gut microbiota fluctuation 9,14, [16][17][18] . The direct effect of dietary interventions functions probably through altering the substrates and metabolome profile in the intestine [19][20][21] . ...
Article
Full-text available
Studies on rodents indicate the daily oscillations of the gut microbiota have biological implications for host. However, the responses of fluctuating gut microbes to the dynamic nutrient substrates are not fully clear. In the study, we found that the feed intake, nutrient substrates, microbiota and metabolites in the colon underwent asynchronous oscillation within a day. Short-chain fatty acids (SCFAs) including acetate, propionate, butyrate and valerate peaked during T24 ~ T27 (Timepoint 24, 12:00 pm, T27, 03:00 am) whereas branched SCFAs isobutyrate and isovalerate peaked during T09 ~ T12. Further extended local similarity analysis (eLSA) revealed that the fluctuation of feed intake dynamically correlated with the colonic carbon substrates which further influenced the oscillation of sugar metabolites and acetate, propionate, butyrate and valerate with a certain time shift. The relative abundance of primary degrader Ruminococcaceae taxa was highly related to the dynamics of the carbon substrates whereas the fluctuations of secondary degraders Lactobacillaceae and Streptococcaceae taxa were highly correlated with the sugar metabolites. Meanwhile, colonic nitrogen substrates were correlated with branched amino acids and the branched SCFAs. Furthermore, we validated the evolution of gut microbes under different carbohydrate and protein combinations by using an in vitro fermentation experiment. The study pictured the dynamics of the micro-ecological environment within a day which highlights the implications of the temporal dimension in studies related to the gut microbiota. Feed intake, more precisely substrate intake, is highly correlated with microbial evolution, which makes it possible to develop chronotherapies targeting the gut microbiota through nutrition intervention.
... To date, circadian metabolome have been extensively reported in various biological samples, including human serum, saliva [15], breath [16], urine [17], and tissues from various species. Importantly, these investigations show that a major portion of metabolome is under circadian control [18]. These metabolites exert their influence by modulating rhythmic gene expression through various mechanisms. ...
Article
Full-text available
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders. The management of PD is a challenging aspect for general physicians and neurologists. It is characterized by the progressive loss of dopaminergic neurons. Impaired α-synuclein secretion and dopamine release may cause mitochondrial dysfunction and perturb energy metabolism, subsequently altering the activity and survival of dopaminergic neurons, thus perpetuating the neurodegenerative process in PD. While the etiology of PD remains multifactorial, emerging research indicates a crucial role of circadian dysfunction in its pathogenesis. Researchers have revealed that circadian dysfunction and sleep disorders are common among PD subjects and disruption of circadian rhythms can increase the risk of PD. Hence, understanding the findings of circadian biology from translational research in PD is important for reducing the risk of neurodegeneration and for improving the quality of life. In this review, we discuss the intricate relationship between circadian dysfunction in cellular metabolism and PD by summarizing the evidence from animal models and human studies. Understanding the metabolic basis of circadian dysfunction in PD may shed light on novel therapeutic approaches to restore circadian rhythm, preserve dopaminergic function, and ameliorate disease progression. Further investigation into the complex interplay between circadian rhythm and PD pathogenesis is essential for the development of targeted therapies and interventions to alleviate the burden of this debilitating neurodegenerative disorder.
... Daily rhythms of glucose metabolism and insulin sensitivity are well described (10). We and others have also characterised widespread diurnal and circadian rhythms of circulating metabolites in humans (28,33,(42)(43)(44). However, daily rhythms of metabolic hormones remain poorly understood in humans, especially those who are overweight and have T2D. ...
Article
Full-text available
Obesity is a major cause of type 2 diabetes. Transition from obesity to type 2 diabetes manifests in the dysregulation of hormones controlling glucose homeostasis and inflammation. As metabolism is a dynamic process that changes across 24-hours, we assessed diurnal rhythmicity in a panel of 10 diabetes-related hormones. Plasma hormones were analysed every 2-hours over 24-hours in a controlled laboratory study with hourly isocaloric drinks during wake. To separate effects of body mass from type 2 diabetes, we recruited 3 groups of middle-aged men; an overweight group with type 2 diabetes and 2 control groups (lean and overweight). Average daily concentrations of glucose, triacylglycerol and all the hormones except visfatin were significantly higher in the overweight group compared to the lean group (P < 0.001). In type 2 diabetes, glucose, insulin, C-peptide, glucose-dependent insulinotropic peptide and glucagon-like peptide-1 increased further (P < 0.05), whereas triacylglycerol, ghrelin and plasminogen activator inhibitor-1 concentrations were significantly lower compared to the overweight group (P < 0.001). Insulin, C-peptide, glucose-dependent insulinotropic peptide and leptin exhibited significant diurnal rhythms in all study groups (P < 0.05). Other hormones were only rhythmic in 1 or 2 groups. In every group, hormones associated with glucose regulation (insulin, C-peptide, glucose-dependent insulinotropic peptide, ghrelin and plasminogen activator inhibitor-1), triacylglycerol and glucose peaked in the afternoon whereas glucagon and hormones associated with appetite and inflammation peaked at night. Thus being overweight with or without type 2 diabetes significantly affected hormone concentrations but did not affect the timing of the hormonal rhythms.
... Like other metabolic pathways, lipid metabolism shows daily rhythmicity and is under circadian regulation in mammals (see (7,8)). In particular, there is substantial daily rhythmicity in plasma transport lipids, including diacylglycerols and triacylglycerols (DGs, TGs), free fatty acids, sphingolipids, and phospholipids (e.g., (9)(10)(11)(12)). The fruit fly Drosophila, a long-standing model in circadian biology, is attracting increasing attention as a model to study lipid metabolism in health and disease (13), including circadian aspects (14). ...
Article
Full-text available
Modern lifestyle is often at odds with endogenously driven rhythmicity, which can lead to circadian disruption and metabolic syndrome. One signature for circadian disruption is a reduced or altered metabolite cycling in the circulating tissue reflecting the current metabolic status. Drosophila is a well-established model in chronobiology, but day-time dependent variations of transport metabolites in the fly circulation are poorly characterized. Here, we sampled fly hemolymph throughout the day and analysed diacylglycerols (DGs), phosphoethanolamines (PEs) and phosphocholines (PCs) using LC-MS. In wildtype flies kept on sugar-only medium under a light-dark cycle, all transport lipid species showed a synchronized bimodal oscillation pattern with maxima at the beginning and end of the light phase which were impaired in period01 clock mutants. In wildtype flies under constant dark conditions, the oscillation became monophasic with a maximum in the middle of the subjective day. In strong support of clock-driven oscillations, levels of DGs, PEs and PCs peaked once in the middle of the light phase under time-restricted feeding independent of the time of food intake. When wildtype flies were reared on full standard medium, the rhythmic alterations of hemolymph lipid levels were greatly attenuated. Our data suggest that the circadian clock aligns daily oscillations of DGs, PEs and PCs in the hemolymph to the anabolic siesta phase, with a strong influence of light on phase and modality. This finding raises the question of whether and to what extent the circadian regulation of transport lipid levels in the hemolymph contributes to the health of the fly.
... Over the last decade, many studies have examined factors affecting metabolite stability, mainly in biofluids such as plasma, serum, and urine [3], since these matrices are a convenient specimen type for investigation. For plasma and serum, these factors included (i) sample-related factors (e.g., collection time [4], hemolysis [5], tube additives [6], fasting status [7]), (ii) sample processing (e.g., centrifugation conditions [8], time delay and temperature [9]), (iii) postprocessing (e.g., time delay temperature [10]), and (iv) sample storage (e.g., freeze/thaw cycles [10], storage time [5]). Additional sample processing factors were studied for urine, such as osmolarity, sample volume [11], and filtration [12]. ...
Article
Full-text available
Thermal reactions can significantly alter the metabolomic and lipidomic content of biofluids and tissues during storage. In this study, we investigated the stability of polar metabolites and complex lipids in dry human serum and mouse liver extracts over a three-day period under various temperature conditions. Specifically, we tested temperatures of −80 °C (freezer), −24 °C (freezer), −0.5 °C (polystyrene box with gel-based ice packs), +5 °C (refrigerator), +23 °C (laboratory, room temperature), and +30 °C (thermostat) to simulate the time between sample extraction and analysis, shipping dry extracts to different labs as an alternative to dry ice, and document the impact of higher temperatures on sample integrity. The extracts were analyzed using five fast liquid chromatography-mass spectrometry (LC-MS) methods to screen polar metabolites and complex lipids, and over 600 metabolites were annotated in serum and liver extracts. We found that storing dry extracts at −24 °C and partially at −0.5 °C provided comparable results to −80 °C (reference condition). However, increasing the storage temperatures led to significant changes in oxidized triacylglycerols, phospholipids, and fatty acids within three days. Polar metabolites were mainly affected at storage temperatures of +23 °C and +30 °C.
... Studies on human circadian and time-of-day variation have found that amino acids and lipid metabolism are involved in the pathogenesis of sleep disturbances, which suggests that the characteristics of endogenous metabolites could be used as a way to interpret EDS [4,5]. In addition, patients with ESRD on maintenance PD have significant alterations in their intracellular and plasma amino acid and lipid profiles, which has been attributed to a low-protein diet or inadequate intake, impaired protein or lipid catabolism, and long-term dialysis [6]. ...
Article
Full-text available
Excessive daytime sleepiness (EDS) is associated with quality of life and all-cause mortality in the end-stage renal disease population. This study aims to identify biomarkers and reveal the underlying mechanisms of EDS in peritoneal dialysis (PD) patients. A total of 48 nondiabetic continuous ambulatory peritoneal dialysis patients were assigned to the EDS group and the non-EDS group according to the Epworth Sleepiness Scale (ESS). Ultra-high-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UHPLC-Q-TOF/MS) was used to identify the differential metabolites. Twenty-seven (male/female, 15/12; age, 60.1 ± 16.2 years) PD patients with ESS ≥ 10 were assigned to the EDS group, while twenty-one (male/female, 13/8; age, 57.9 ± 10.1 years) PD patients with ESS < 10 were defined as the non-EDS group. With UHPLC-Q-TOF/MS, 39 metabolites with significant differences between the two groups were found, 9 of which had good correlations with disease severity and were further classified into amino acid, lipid and organic acid metabolism. A total of 103 overlapping target proteins of the differential metabolites and EDS were found. Then, the EDS–metabolite–target network and the protein–protein interaction network were constructed. The metabolomics approach integrated with network pharmacology provides new insights into the early diagnosis and mechanisms of EDS in PD patients.
Preprint
Full-text available
Rhythmicity is a central feature of behavioral and biological processes including metabolism, however, the mechanisms of metabolite cycling are poorly understood. A robust oscillation in a network of key metabolite pathways downstream of glucose is described in humans, then these pathways mechanistically probed through purpose-built 13C6-glucose isotope tracing in Drosophila every 4h. A temporal peak in biosynthesis was noted by broad labelling of pathways downstream of glucose in wild-type flies shortly following lights on. Krebs cycle labelling was generally increased in a hyperactive mutant (fumin) along with glycolysis labelling primarily observed at dawn. Surprisingly, neither underlying feeding rhythms nor the presence of food explains the rhythmicity of glucose processing across genotypes. These results are consistent with clinical data demonstrating detrimental effects of mis-timed energy intake. This approach provides a window into the dynamic range of metabolic processing ability through the day and mechanistic basis for exploring circadian metabolic homeostasis in disease states.
Article
Full-text available
Lipids are biological molecules that play vital roles in all living organisms. They perform many cellular functions, such as 1) forming cellular and subcellular membranes, 2) storing and using energy, and 3) serving as chemical messengers during intra‐ and inter‐cellular signal transduction. The large‐scale study of the pathways and networks of cellular lipids in biological systems is called “lipidomics” and is one of the fastest‐growing omics technologies of the last two decades. With state‐of‐the‐art mass spectrometry instrumentation and sophisticated data handling, clinical studies show how human lipid composition changes in health and disease, thereby making it a valuable medium to collect for clinical applications, such as disease diagnostics, therapeutic decision‐making, and drug development. This review gives a comprehensive overview of current workflows used in clinical research, from sample collection and preparation to data and clinical interpretations. This is followed by an appraisal of applications in 2022 and a perspective on the exciting future of clinical lipidomics.
Article
Full-text available
Endocytosis through Drosophila glia is a significant determinant of sleep amount and occurs preferentially during sleep in glia of the blood brain barrier (BBB). To identify metabolites whose trafficking is mediated by sleep-dependent endocytosis, we conducted metabolomic analysis of flies that have increased sleep due to a block in glial endocytosis. We report that acylcarnitines, fatty acids conjugated to carnitine to promote their transport, accumulate in heads of these animals. In parallel, to identify transporters and receptors whose loss contributes to the sleep phenotype caused by blocked endocytosis, we screened genes enriched in barrier glia for effects on sleep. We find that knockdown of lipid transporters LRP1&2 or of carnitine transporters ORCT1&2 increases sleep. In support of the idea that the block in endocytosis affects trafficking through specific transporters, knockdown of LRP or ORCT transporters also increases acylcarnitines in heads. We propose that lipid species, such as acylcarnitines, are trafficked through the BBB via sleep-dependent endocytosis, and their accumulation reflects an increased need for sleep.
Article
Full-text available
The synthesis of nucleotides in the body is centrally controlled by the liver, via salvage or de novo synthesis. We reveal a pervasive circadian influence on hepatic nucleotide metabolism, from rhythmic gene expression of rate-limiting enzymes to oscillating nucleotide metabolome in wild-type (WT) mice. Genetic disruption of the hepatic clock leads to aberrant expression of these enzymes, together with anomalous nucleotide rhythms, such as constant low levels of ATP with an excess in uric acid, the degradation product of purines. These results clearly demonstrate that the hepatic circadian clock orchestrates nucleotide synthesis and degradation. This circadian metabolome timetable, obtained using state-of-the-art capillary electrophoresis time-of-flight mass spectrometry, will guide further investigations in nucleotide metabolism-related disorders.
Article
Full-text available
The circadian clock orchestrates many aspects of human physiology, and disruption of this clock has been implicated in various pathologies, ranging from cancer to metabolic syndrome and diabetes. Although there is evidence that metabolism and the circadian clockwork are intimately linked on a transcriptional level, whether these effects are directly under clock control or are mediated by the rest-activity cycle and the timing of food intake is unclear. To answer this question, we conducted an unbiased screen in human subjects of the metabolome of blood plasma and saliva at different times of day. To minimize indirect effects, subjects were kept in a 40-h constant routine of enforced posture, constant dim light, hourly isocaloric meals, and sleep deprivation. Under these conditions, we found that ~15% of all identified metabolites in plasma and saliva were under circadian control, most notably fatty acids in plasma and amino acids in saliva. Our data suggest that there is a strong direct effect of the endogenous circadian clock on multiple human metabolic pathways that is independent of sleep or feeding. In addition, they identify multiple potential small-molecule biomarkers of human circadian phase and sleep pressure.
Article
Full-text available
The circadian clock governs a large array of physiological functions through the transcriptional control of a significant fraction of the genome. Disruption of the clock leads to metabolic disorders, including obesity and diabetes. As food is a potent zeitgeber (ZT) for peripheral clocks, metabolites are implicated as cellular transducers of circadian time for tissues such as the liver. From a comprehensive dataset of over 500 metabolites identified by mass spectrometry, we reveal the coordinate clock-controlled oscillation of many metabolites, including those within the amino acid and carbohydrate metabolic pathways as well as the lipid, nucleotide, and xenobiotic metabolic pathways. Using computational modeling, we present evidence of synergistic nodes between the circadian transcriptome and specific metabolic pathways. Validation of these nodes reveals that diverse metabolic pathways, including the uracil salvage pathway, oscillate in a circadian fashion and in a CLOCK-dependent manner. This integrated map illustrates the coherence within the circadian metabolome, transcriptome, and proteome and how these are connected through specific nodes that operate in concert to achieve metabolic homeostasis.
Article
Full-text available
Phospholipase A(2) (PLA(2)) is a group of enzymes that hydrolyze the sn-2 position of glycerophospholipids to yield fatty acids and lysophospholipids. Of many PLA(2)s or related enzymes identified to date, secreted PLA(2)s (sPLA(2)s) comprise the largest family that contains 10 catalytically active isozymes. Besides arachidonic acid released from cellular membranes for eicosanoid synthesis, several if not all sPLA(2)s have recently been implicated in hydrolysis of phospholipids in lipoprotein particles. The sPLA(2)-processed low-density lipoprotein (LDL) particles contain a large amount of lysophospholipids and exhibit the property of "small-dense" or "modified" LDL, which facilitates foam cell formation from macrophages. Transgenic overexpression of these sPLA(2)s leads to development of atherosclerosis in mice. More importantly, genetic deletion or pharmacological inhibition of particular sPLA(2)s significantly attenuates atherosclerosis and aneurysm. In this article, we will give an overview of current understanding of the role of sPLA(2)s in atherosclerosis, with recent lipidomics data showing the action of a subset of sPLA(2)s on lipoprotein phospholipids.
Article
Full-text available
Previous animal studies suggest a functional relationship between metabolism, type 2 diabetes, and the amplitude of daily rhythms in white adipose tissue (WAT). However, data interpretation is confounded by differences in genetic background and diet or limited sampling points. We have taken the novel approach of analyzing serial human WAT biopsies across a 24-h cycle in controlled laboratory conditions. Lean (n = 8), overweight/obese (n = 11), or overweight/obese type 2 diabetic (n = 8) volunteers followed a strict sleep-wake and dietary regimen for 1 week prior to the laboratory study. They were then maintained in controlled light-dark conditions in a semirecumbent posture and fed hourly during wake periods. Subcutaneous WAT biopsies were collected every 6 h over 24 h, and gene expression was measured by quantitative PCR. Lean individuals exhibited significant (P < 0.05) temporal changes of core clock (PER1, PER2, PER3, CRY2, BMAL1, and DBP) and metabolic (REVERBα, RIP140, and PGC1α) genes. The BMAL1 rhythm was in approximate antiphase with the other clock genes. It is noteworthy that there was no significant effect (P > 0.05) of increased body weight or type 2 diabetes on rhythmic gene expression. The robust nature of these rhythms and their relative phasing indicate that WAT now can be considered as a peripheral tissue suitable for the study of in vivo human rhythms. Comparison of data between subject groups clearly indicates that obesity and type 2 diabetes are not related to the amplitude of rhythmic WAT gene expression in humans maintained under controlled conditions.
Article
Full-text available
Continuing improvements in analytical technology along with an increased interest in performing comprehensive, quantitative metabolic profiling, is leading to increased interest pressures within the metabolomics community to develop centralized metabolite reference resources for certain clinically important biofluids, such as cerebrospinal fluid, urine and blood. As part of an ongoing effort to systematically characterize the human metabolome through the Human Metabolome Project, we have undertaken the task of characterizing the human serum metabolome. In doing so, we have combined targeted and non-targeted NMR, GC-MS and LC-MS methods with computer-aided literature mining to identify and quantify a comprehensive, if not absolutely complete, set of metabolites commonly detected and quantified (with today's technology) in the human serum metabolome. Our use of multiple metabolomics platforms and technologies allowed us to substantially enhance the level of metabolome coverage while critically assessing the relative strengths and weaknesses of these platforms or technologies. Tables containing the complete set of 4229 confirmed and highly probable human serum compounds, their concentrations, related literature references and links to their known disease associations are freely available at http://www.serummetabolome.ca.
Article
THE term circadian was introduced by Halberg1 to describe a recurring sequence of events with a cycle of approximately 24 h, and the present knowledge of human circadian rhythms has been reviewed recently by Mills2. The circadian periodicity of a large number of constituents of blood, plasma, and urine has been described. Although a diurnal periodicity of whole blood amino-acids in man, in which concentrations at 2000 h were consistently greater than those at 0800 h on the same day, has been noted in this laboratory3, there were insufficient sampling times to establish a circadian pattern. A circadian periodicity of serum amino-acids in growing chickens4 and whole blood tryptophan in mice5 has been reported. We report here a circadian periodicity of whole blood and serum amino-acids in healthy adults.
Article
An ultra performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC Q-TOF MS) metabonomics approach was employed to study the serum metabolic profiling of adenine-induced chronic renal failure (CRF) rats. Acquired data were subjected to principal component analysis (PCA) for differentiating the CRF and the normal control groups. Potential biomarkers were screened by using S-plot and were identified by the accurate mass, isotopic pattern and MS/MS fragments information obtained from UPLC Q-TOF MS analysis. Significant differences in the serum level of creatinine, amino acids and LysoPCs were observed, indicating the perturbations of amino acid metabolism and phospholipid metabolism in adenine-induced CRF rats. This research proved that metabonomics is a promising tool for disease research.