ArticlePDF Available

Abstract and Figures

Aims/hypothesis Type 2 diabetes is a chronic condition that is caused by hyperglycaemia. Our aim was to characterise the metabolomics to find their association with the glycaemic spectrum and find a causal relationship between metabolites and type 2 diabetes. Methods As part of the Innovative Medicines Initiative - Diabetes Research on Patient Stratification (IMI-DIRECT) consortium, 3000 plasma samples were measured with the Biocrates AbsoluteIDQ p150 Kit and Metabolon analytics. A total of 911 metabolites (132 targeted metabolomics, 779 untargeted metabolomics) passed the quality control. Multivariable linear and logistic regression analysis estimates were calculated from the concentration/peak areas of each metabolite as an explanatory variable and the glycaemic status as a dependent variable. This analysis was adjusted for age, sex, BMI, study centre in the basic model, and additionally for alcohol, smoking, BP, fasting HDL-cholesterol and fasting triacylglycerol in the full model. Statistical significance was Bonferroni corrected throughout. Beyond associations, we investigated the mediation effect and causal effects for which causal mediation test and two-sample Mendelian randomisation (2SMR) methods were used, respectively. Results In the targeted metabolomics, we observed four (15), 34 (99) and 50 (108) metabolites (number of metabolites observed in untargeted metabolomics appear in parentheses) that were significantly different when comparing normal glucose regulation vs impaired glucose regulation/prediabetes, normal glucose regulation vs type 2 diabetes, and impaired glucose regulation vs type 2 diabetes, respectively. Significant metabolites were mainly branched-chain amino acids (BCAAs), with some derivatised BCAAs, lipids, xenobiotics and a few unknowns. Metabolites such as lysophosphatidylcholine a C17:0, sum of hexoses, amino acids from BCAA metabolism (including leucine, isoleucine, valine, N-lactoylvaline, N-lactoylleucine and formiminoglutamate) and lactate, as well as an unknown metabolite (X-24295), were associated with HbA1c progression rate and were significant mediators of type 2 diabetes from baseline to 18 and 48 months of follow-up. 2SMR was used to estimate the causal effect of an exposure on an outcome using summary statistics from UK Biobank genome-wide association studies. We found that type 2 diabetes had a causal effect on the levels of three metabolites (hexose, glutamate and caproate [fatty acid (FA) 6:0]), whereas lipids such as specific phosphatidylcholines (PCs) (namely PC aa C36:2, PC aa C36:5, PC ae C36:3 and PC ae C34:3) as well as the two n-3 fatty acids stearidonate (18:4n3) and docosapentaenoate (22:5n3) potentially had a causal role in the development of type 2 diabetes. Conclusions/interpretation Our findings identify known BCAAs and lipids, along with novel N-lactoyl-amino acid metabolites, significantly associated with prediabetes and diabetes, that mediate the effect of diabetes from baseline to follow-up (18 and 48 months). Causal inference using genetic variants shows the role of lipid metabolism and n-3 fatty acids as being causal for metabolite-to-type 2 diabetes whereas the sum of hexoses is causal for type 2 diabetes-to-metabolite. Identified metabolite markers are useful for stratifying individuals based on their risk progression and should enable targeted interventions. Graphical Abstract
This content is subject to copyright. Terms and conditions apply.
Vol:.(1234567890)
Diabetologia (2024) 67:2804–2818
https://doi.org/10.1007/s00125-024-06282-6
ARTICLE
Role ofhuman plasma metabolites inprediabetes andtype 2 diabetes
fromtheIMI‑DIRECT study
SapnaSharma1· QiulingDong1,2· MarkHaid3· JonathanAdam1,4· RobertoBizzotto5· JuanJ.Fernandez‑Tajes6·
AngusG.Jones7· AndreaTura5· AnnaArtati3· CorneliaPrehn3· GabiKastenmüller8· RobertW.Koivula9·
PaulW.Franks10· MarkWalker11· IanM.Forgie12· GiuseppeGiordano13· ImrePavo14· HartmutRuetten15·
ManolisDermitzakis16,17,18· MarkI.McCarthy6· OlufPedersen19,20· JochenM.Schwenk21·
KonstantinosD.Tsirigos22· FedericoDeMasi22· SorenBrunak20,23· AnaViñuela24· AndreaMari5·
TimothyJ.McDonald25· TarjaKokkola26· JerzyAdamski27,28,29· EwanR.Pearson12· HaraldGrallert1,4
Received: 28 February 2024 / Accepted: 29 July 2024 / Published online: 30 September 2024
© The Author(s) 2024
Abstract
Aims/hypothesis Type 2 diabetes is a chronic condition that is caused by hyperglycaemia. Our aim was to characterise the
metabolomics to find their association with the glycaemic spectrum and find a causal relationship between metabolites and
type 2 diabetes.
Methods As part of the Innovative Medicines Initiative - Diabetes Research on Patient Stratification (IMI-DIRECT) consor-
tium, 3000 plasma samples were measured with the Biocrates AbsoluteIDQ p150 Kit and Metabolon analytics. A total of 911
metabolites (132 targeted metabolomics, 779 untargeted metabolomics) passed the quality control. Multivariable linear and
logistic regression analysis estimates were calculated from the concentration/peak areas of each metabolite as an explanatory
variable and the glycaemic status as a dependent variable. This analysis was adjusted for age, sex, BMI, study centre in the basic
model, and additionally for alcohol, smoking, BP, fasting HDL-cholesterol and fasting triacylglycerol in the full model. Statis-
tical significance was Bonferroni corrected throughout. Beyond associations, we investigated the mediation effect and causal
effects for which causal mediation test and two-sample Mendelian randomisation (2SMR) methods were used, respectively.
Results In the targeted metabolomics, we observed four (15), 34 (99) and 50 (108) metabolites (number of metabolites
observed in untargeted metabolomics appear in parentheses) that were significantly different when comparing normal glucose
regulation vs impaired glucose regulation/prediabetes, normal glucose regulation vs type 2 diabetes, and impaired glucose
regulation vs type 2 diabetes, respectively. Significant metabolites were mainly branched-chain amino acids (BCAAs), with
some derivatised BCAAs, lipids, xenobiotics and a few unknowns. Metabolites such as lysophosphatidylcholine a C17:0,
sum of hexoses, amino acids from BCAA metabolism (including leucine, isoleucine, valine, N-lactoylvaline, N-lactoylleucine
and formiminoglutamate) and lactate, as well as an unknown metabolite (X-24295), were associated with HbA1c progression
rate and were significant mediators of type 2 diabetes from baseline to 18 and 48 months of follow-up. 2SMR was used to
estimate the causal effect of an exposure on an outcome using summary statistics from UK Biobank genome-wide association
studies. We found that type 2 diabetes had a causal effect on the levels of three metabolites (hexose, glutamate and caproate
[fatty acid (FA) 6:0]), whereas lipids such as specific phosphatidylcholines (PCs) (namely PC aa C36:2, PC aa C36:5, PC ae
C36:3 and PC ae C34:3) as well as the two n-3 fatty acids stearidonate (18:4n3) and docosapentaenoate (22:5n3) potentially
had a causal role in the development of type 2 diabetes.
Conclusions/interpretation Our findings identify known BCAAs and lipids, along with novel N-lactoyl-amino acid metabo-
lites, significantly associated with prediabetes and diabetes, that mediate the effect of diabetes from baseline to follow-up (18
and 48 months). Causal inference using genetic variants shows the role of lipid metabolism and n-3 fatty acids as being causal
for metabolite-to-type 2 diabetes whereas the sum of hexoses is causal for type 2 diabetes-to-metabolite. Identified metabo-
lite markers are useful for stratifying individuals based on their risk progression and should enable targeted interventions.
Sapna Sharma, Qiuling Dong and Mark Haid contributed equally to
this study.
Extended author information available on the last page of the article
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2805Diabetologia (2024) 67:2804–2818
Keywords Causality· Glycaemic traits· HbA1c· IMI-DIRECT· Mediation· Metabolomics· N-lactoylaminoacids· Patient
stratification· Targeted metabolomics· Type 2 diabetes· Untargeted metabolomics
Abbreviations
2SMR Two-sample MR
GWAS Genome-wide association study
H1 Hexoses
IGR Impaired glucose regulation
IGT Impaired glucose tolerance
IMI-DIRECT Innovative Medicines Initiative - Diabetes
Research on Patient Stratification
Lac-Phe N-lactoyl-phenylalanine
LysoPC Lysophosphatidylcholine
MOVE Multi-omics variational autoencoders
MR Mendelian randomisation
NA Unidentified metabolite
NGR Normal glucose regulation
PC Phosphatidylcholine
Introduction
Type 2 diabetes is a complex and common metabolic disor-
der, resulting from the body’s ineffective use of insulin. It can
be characterised by hyperglycaemia (high blood sugar) due to
impaired insulin secretion and insulin resistance, with most
affected people being overweight or obese [1]. Impaired glu-
cose tolerance (IGT) and impaired fasting glucose, together
known as impaired glucose regulation (IGR) or prediabetes,
characterise an intermediate condition before converging
towards diabetes. Recent studies show that a complex inter-
play of genetic susceptibility, environmental factors, lifestyle
(including diet, physical activity, smoking and alcohol con-
sumption), clinical heterogeneity, drugs and gut microbiome
orchestrates the development of type 2 diabetes [2]. Over
time, individuals with type 2 diabetes are more likely to have
a higher risk for heart attacks, strokes [3], neuropathy (nerve
damage), retinopathy (causing blindness) and kidney failure
as well as several infectious diseases including COVID-19,
reducing life quality and causing social burden [4, 5].
Metabolomics profiles involve a set of low-molecular-
weight biochemicals (metabolites) that includes sugars,
amino acids, organic acids, nucleotides, lipids, xenobiot-
ics and other compound classes. Identifying biochemi-
cal changes occurring between prediabetes and diabetes
improves risk prediction for better-targeted prevention [6,
7]. In addition, genetic composition can be used to make
predictions regarding disease susceptibility. Genome-wide
association studies (GWAS) show that more than 400 loci
influence the risk of type 2 diabetes [8] and that 900 genetic
variants have been associated with BMI [9]. Therefore,
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2806 Diabetologia (2024) 67:2804–2818
linking metabolites with genetics gives access to genetics
influence on the metabolic compositions [1013], provid-
ing comprehensive molecular understanding of the disease.
In the Innovative Medicines Initiative - Diabetes Research
on Patient Stratification (IMI-DIRECT), we characterised
132 metabolites from targeted measurements and 779
metabolites from untargeted measurements profiled in 3000
individuals at baseline. The study population was stratified
by following ADA 2011 glycaemic categories as follows:
23.89% (n=692) had normal glucose regulation (NGR) with
fasting glucose 5.23 (SD=0.39) mmol/l; 48.91% (n=1418)
had IGR with fasting glucose 5.90 (SD=0.51) mmol/l; and
27.2% (n=890) had type 2 diabetes with fasting glucose 7.15
(SD=1.39) mmol/l [14]. For the integration of non-omics
data such as health status, lifestyle and medication with
metabolomics, advanced statistical techniques were applied
to analyse the data (see Methods). Beyond multivariate and
association analyses we performed causal mediation analysis
to evaluate potential causal roles of mediators on outcome
[15, 16]. A study on drug–omics associations in type 2 dia-
betes [17] used an unsupervised deep learning framework
of multi-omics variational autoencoders (MOVE) to extract
significant drug response patterns from 789 individuals
newly diagnosed with type 2 diabetes in the IMI-DIRECT
cohort. We integrated the polypharmacy effect on metab-
olomics knowledge from MOVE and compared with our
molecular findings in this study.
Our aims in this study were as follows: (1) to characterise
911 small molecular (132 targeted, 779 untargeted metabo-
lomics analysis approach) features associated with prediabetes/
IGR and type 2 diabetes; (2) to identify baseline metabolites
associated with progression rate estimated from cross-sectional
data; (3) to investigate potential mediation effects of metabo-
lites from baseline glycaemic status to follow-up using media-
tion analysis; and (4) to identify causal relationships between
metabolites and type 2 diabetes using genetics drivers using
two-sample Mendelian randomisation (2SMR) tests.
Methods
DIRECT cohort
The Diabetes Research on Patient Stratification (DIRECT)
cohort encompasses 24,682 European participants at varying
risk of glycaemic deterioration, identified and enrolled into
a prospective cohort (study 1) of prediabetes (n=2235) and
type 2 diabetes (n=830). Using ADA 2011 glycaemic catego-
ries in study 1, 33% (n=692) of cohort 1 (prediabetes risk)
had NGR, 67% (n=1418) had IGR and 108 were excluded.
In study 2, 789 samples were included and 41 samples were
excluded. From study 1, 101 excluded samples entered study
2 (n=890). The ratio of self-reported sex varied in each study.
Detailed characteristics on inclusion and exclusion criteria,
along with the protocol timeline for visits and tests for both
studies, have been described elsewhere [14, 18]. In summary,
venous blood fasting samples were obtained, followed by per-
formance of DNA extractions and additional biochemical
analyses. Metabolomics measurements for distinct samples
at the baseline is considered in this study.
Targeted metabolomics (AbsoluteIDQ p150 Kit)
Blood samples in the study were analysed with the Abso-
luteIDQ p150 Kit (BIOCRATES Life Sciences, Innsbruck,
Austria) (see electronic supplementary material [ESM]
Methods for details) [19]. After data export, lower and upper
outliers were defined as samples with >33% of metabolite
concentrations below 25% quantile (±1.5 × IQR). Metabolite
traits with too many zero-concentration samples and uniden-
tified metabolites (NAs, >50%) were excluded (none). The
CV was calculated in reference samples for each metabolite
over all plates. Metabolite traits with CV>0.25 were excluded.
After quality control, 132 metabolites were included in
this study (ESM Table1). Metabolite concentrations were
loge-transformed and scaled (mean=0, SD=1) to ensure com-
parability between the metabolites.
Untargeted metabolomics (Metabolon platform)
Untargeted LC/MS-based techniques covers a broad spec-
trum of metabolites, in contrast to the targeted techniques
wherein metabolites are limited to a predefined set of
molecules. For details on sample preparation, measure-
ment and identification of metabolites, see ESM Methods.
Incomplete databases and the presence of unknown or
novel metabolites have been reported with an asterisk (*)
against the metabolite name. The measured volume of the
datasets contained 12% missing values. We screened for
outlier remover (see ESM Fig.1 for an example), which
added 4% more missing values onto existing missing val-
ues (ESM Table2). Peaks were quantified using AUC.
For studies spanning multiple days, a data normalisation
step was performed to correct variation resulting from
instrument inter-day tuning differences. Essentially, each
compound was corrected in run-day blocks by register-
ing the medians to equal one and normalising each data
point proportionately (termed the ‘block correction’; ESM
Fig.2). Principal component analysis was performed on the
metabolite dataset and checked for technical effects such as
centre and sex (see ESM Fig.3). The data missing pattern
was tested using logistic regression considering missing as
0 and non-missing as 1; there was no significant association
between missing and regressors indicating the missing-at-
random pattern. The K-nearest neighbour (KNN)-based
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2807Diabetologia (2024) 67:2804–2818
imputation method was applied using K=10 as suggested
and optimised from German Cohort KORA F4 [20].
Statistics
Multivariable logistic regression and linear regression Identi-
fying metabolites specifically associated with the presence of
IGR and type 2 diabetes, we ran the logistic regression with
adjustment for age, sex, BMI and centre as the basic model,
and adjusted additionally for alcohol consumption, smok-
ing, BP, fasting HDL-cholesterol and fasting triacylglycerol
as the full model. The concentration of each metabolite was
loge-transformed and scaled to have a mean of zero and an
SD of 1. Each metabolite was taken as exposure and a binary
NGR-IGR, NGR-type 2 diabetes (NGR-T2D) or IGR-type
2 diabetes (IGR-T2D) variable as an outcome. The OR of
outcomes was calculated using the β coefficient from logistic
regression, where OR>1 indicates higher odds of outcome
and OR<0 shows lower odds of outcome. To account for
multiple testing, the p values from regression analyses were
adjusted for multiple testing using the Bonferroni correction
(pfdr values). To stratify sex-dependent metabolites, men and
women were separated to test the associations by performing
the logistic regression full models.
For incidents of IGR and type 2 diabetes analysis, a
binary NGR-IGR, NGR-T2D or IGR-T2D variable at fol-
low-up times of 18 months and 48 months was taken as the
outcome; transformed metabolites and the same risk factors
in the full model were taken as exposure and covariates,
respectively. The same p correction method was adopted.
The linear regression model was used to explore the asso-
ciation between HbA1c progression rate and metabolites at
the baseline. HbA1c progression rate was computed with
a conditional linear mixed effect model and adjusted for
changes in BMI and diabetes medications [21]. Each trans-
formed metabolite was taken as the independent variable and
HbA1c concentration as the dependent variable, with adjust-
ment for age and sex. Bonferroni correction was performed
for p correction.
Mediation analysis Mediation analysis followed the basic
steps suggested by Baron and Kenny [22], and the signifi-
cance of the mediation effect was tested with a non-para-
metric causal mediation analysis [22, 23]. Each identified
metabolite was taken as a mediator, glycaemic category sta-
tus at the baseline as the independent variable and glycaemic
category at the follow-up (18 months and 48 months) as
the dependent variable. R package ‘mediation (4.5.0)’ was
used to calculate the p value and proportion of the mediation
effect by bootstrapping with 1000 resamples.
Mendelian randomisation We used 2SMR approaches from
the MRInstruments (0.3.2) and TwoSampleMR library
(v0.5.6) to check causal inference [24]. The 2SMR technique
enables the establishment of a causal relationship between
two observational studies (ESM Fig.4), solely relying on
summary statistics obtained from GWAS [24, 25]. To evalu-
ate the influence of type 2 diabetes on metabolite levels, we
conducted a 2SMR examination. Type 2 diabetes instruments
were obtained from the genome-wide genotyping study [26]
and the corresponding SNP estimates on metabolites were
extracted from the metabolite-GWAS [10, 27]. Prior to per-
forming Mendelian randomisation (MR) analysis, exposure
and outcome data were harmonised by aligning the SNPs
on the same effect allele. We employed the inversevariance
weighting [10, 26, 27] to estimate the causal effect.
Results
Study populations
After stringent quality control (see ESM Methods), we iden-
tified 132 (ESM Table1) and 779 (ESM Table2) metabolites
from targeted and untargeted metabolomics measurements,
respectively, that were profiled for 3000 samples (ESM
Table3) [28]. Baseline characteristics (Table1) revealed
that there were significant differences in BMI, fasting vari-
ables and health status observed between NGR, IGR and
type 2 diabetes groups. No significant differences in age and
smoking status were observed between these three groups.
In addition, the study was conducted across seven countries;
type 2 diabetes participants were recruited in all centres
while participants with NGR or IGR were only recruited
in the Amsterdam, Copenhagen, Kuopio and Lund centres.
Metabolites associated withprediabetes
anddiabetes fromtargeted metabolomics
measurements
A multivariable logistic regression model was used with
known diabetes-related variables as covariates to identify
significant metabolites. Study centre, sex, age and BMI were
covariates in the basic model while the additional variables
systolic BP, fasting HDL-cholesterol, fasting triacylglycerol,
smoking status, alcohol status and health status were added in
the full model. Based on the full model, four metabolites dif-
fered significantly between the NGR and IGR groups (Fig.1a).
Of these, hexoses (H1) showed the strongest association (OR
1.81 [95% CI 1.59, 2.06], pfdr=3.97×10−17) and served as a
positive control throughout our analysis. Thirty-four and 50
metabolites differed significantly between NGR and IGR vs
type 2 diabetes, respectively (Fig.1b,c). As a general pattern,
phosphatidylcholines (PCs) and lysophosphatidylcholine
(lysoPC) were negatively associated with progression to type
2 diabetes, while branched-chain and aromatic amino acids as
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2808 Diabetologia (2024) 67:2804–2818
well as valeryl/glutaryl-related acylcarnitines were positively
associated with type 2 diabetes.
H1 (OR 9.67 [95% CI 6.54, 14.32], pfdr=1.13×10−27) also
had the strongest associations in NGR-T2D while C5-M-DC
(OR=5.31 [95% CI 4.16, 6.77], pfdr=1.07×10−38) had the
strongest association in IGR-T2D. Three metabolites (H1,
lysoPC a C17:0, lysoPC a C18:0) were significantly dif-
ferent in all comparisons (NGR-IGR, NGR-T2D and IGR-
T2D), suggesting their important roles in diabetes indica-
tion and severity. Detailed statistics for the basic model
and full model are shown in ESM Tables3–8. As there
were many more male participants than female participants
Table 1 Baseline characteristics
of the DIRECT participants
based on their glycaemic
category
Quantitative variables are expressed as mean ± SD; categorical variables are expressed as n (%)
The significant difference of population characteristics between the individuals with IGR/type 2 diabetes
and the normal participants (NGR) was calculated. Test statistics for categorical variables were calculated
via the χ2 test and Student’s t test for continuous variables
T2D, type 2 diabetes; TG, triacylglycerol
Characteristic NGR IGR T2D p value
Sample size 692 1418 890
Male sex 519 (75.0) 1074 (75.7) 525 (59.0) <0.001
Centre <0.001
Amsterdam 167 (24.1) 300 (21.2) 183 (20.6)
Copenhagen 54 (7.8) 223 (15.7) 97 (10.9)
Dundee 0 0 164 (18.4)
Exeter 0 0 142 (16.0)
Kuopio 407 (58.8) 820 (57.8) 34 (3.8)
Lund 64 (9.2) 75 (5.3) 104 (11.7)
Newcastle 0 0 166 (18.7)
Age, years 62.15±6.43 62.08±6.19 61.99±7.96 0.894
BMI, kg/m227.15±3.65 28.33±4.06 30.59±4.92 <0.001
Systolic BP, mmHg 128.48±15.21 131.62±15.20 132.02±15.78 <0.001
Diastolic BP, mmHg 79.18±8.73 81.20±8.97 76.48±9.88 <0.001
Fasting glucose, mmol/l 5.23±0.39 5.90±0.51 7.13±1.39 <0.001
Fasting HDL-cholesterol, mmol/l 1.37±0.35 1.30±0.36 1.18±0.38 <0.001
Fasting LDL-cholesterol, mmol/l 3.21±0.90 3.19±0.95 2.43±1.00 <0.001
Fasting TG, mmol/l 1.22±0.53 1.44±0.66 1.56±0.88 <0.001
Fasting cholesterol, mmol/l 5.14±0.97 5.15±1.01 4.33±1.17 <0.001
Fasting HbA1c, mmol/mol 35.34±2.22 37.86±2.88 45.86±5.94 <0.001
Fasting HbA1c, % 5.38±0.20 5.61±0.26 6.35±0.54 <0.001
Fasting insulin, pmol/l 50.84±30.90 72.42±50.22 96.56±72.69 <0.001
Smoking status 0.717
Current smoker 93 (13.4) 215 (15.2) 117 (13.2)
Ex-smoker 326 (47.1) 681 (48.0) 445 (50.1)
Never 272 (39.3) 520 (36.7) 326 (36.7)
Not Known 1 (0.1) 2 (0.1) 1 (0.1)
Alcohol consumption status 0.004
Never 96 (13.9) 166 (11.7) 140 (15.7)
Occasionally 134 (19.4) 282 (19.9) 214 (24.1)
Regularly 462 (66.8) 968 (68.3) 534 (60.1)
Not known 0 2 (0.1) 1 (0.1)
Health status <0.001
Poor 1 (0.1) 10 (0.7) 28 (3.1)
Fair 49 (7.1) 74 (5.2) 34 (3.8)
Good 331 (47.8) 744 (52.5) 428 (48.1)
Very good 213 (30.8) 396 (27.9) 239 (26.9)
Excellent 49 (7.1) 74 (5.2) 34 (3.8)
Not known 4 (0.6) 11 (0.8) 19 (2.1)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2809Diabetologia (2024) 67:2804–2818
Lipid_lysoPC a C17:0
Lipid_lysoPC a C18:0
Lipid_PC aa C34:4
Sugar_H1
1.0 1.5 2.0
OR
Metabolites
a
Acylcarnitines_C14:2−OH
Acylcarnitines_C5
Acylcarnitines_C5−M−DC
Acylcarnitines_C5−OH (C3−DC−M)
Acylcarnitines_C5:1
Amino acid_Phe
Amino acid_Tyr
Amino acid_Val
Amino acid_xLeu
Lipid_lysoPC a C14:0
Lipid_lysoPC a C16:0
Lipid_lysoPC a C16:1
Lipid_lysoPC a C17:0
Lipid_lysoPC a C18:0
Lipid_lysoPC a C18:1
Lipid_lysoPC a C18:2
Lipid_PC aa C32:3
Lipid_PC aa C34:2
Lipid_PC aa C34:3
Lipid_PC aa C40:1
Lipid_PC ae C30:2
Lipid_PC ae C32:1
Lipid_PC ae C34:2
Lipid_PC ae C34:3
Lipid_PC ae C36:0
Lipid_PC ae C40:0
Lipid_PC ae C40:1
Lipid_PC ae C42:0
Lipid_PC ae C42:3
Lipid_SM (OH) C14:1
Lipid_SM C16:0
Lipid_SM C16:1
Lipid_SM C24:1
Sugar_H1
0510 15
OR
Metabolites
b
Acylcarnitines_C14
Acylcarnitines_C14:2−OH
Acylcarnitines_C3−DC (C4−OH)
Acylcarnitines_C5−M−DC
Acylcarnitines_C5−OH (C3−DC−M)
Amino acid_Gly
Amino acid_Tyr
Amino acid_Val
Amino acid_xLeu
Lipid_lysoPC a C14:0
Lipid_lysoPC a C16:0
Lipid_lysoPC a C16:1
Lipid_lysoPC a C17:0
Lipid_lysoPC a C18:0
Lipid_lysoPC a C18:1
Lipid_lysoPC a C18:2
Lipid_lysoPC a C20:3
Lipid_PC aa C32:0
Lipid_PC aa C32:3
Lipid_PC aa C34:2
Lipid_PC aa C34:3
Lipid_PC aa C36:0
Lipid_PC aa C36:2
Lipid_PC aa C38:0
Lipid_PC aa C40:1
Lipid_PC aa C40:2
Lipid_PC ae C30:2
Lipid_PC ae C32:1
Lipid_PC ae C34:1
Lipid_PC ae C34:2
Lipid_PC ae C34:3
Lipid_PC ae C36:0
Lipid_PC ae C36:2
Lipid_PC ae C36:3
Lipid_PC ae C38:0
Lipid_PC ae C40:0
Lipid_PC ae C40:1
Lipid_PC ae C42:0
Lipid_PC ae C42:2
Lipid_PC ae C42:3
Lipid_PC ae C42:4
Lipid_SM (OH) C14:1
Lipid_SM (OH) C16:1
Lipid_SM (OH) C22:2
Lipid_SM C16:0
Lipid_SM C16:1
Lipid_SM C18:1
Lipid_SM C24:0
Lipid_SM C24:1
Sugar_H1
246
OR
Metabolites
Category
Acylcarnitines
Amino acid
Lipid
Sugar
c
Fig. 1 Flag plots representing the results of the multivariable logistic regression models for NGR vs IGR (a), NGR vs type 2 diabetes (b) and IGR vs type 2 diabetes (c) as dependent variables
and the metabolites as independent variables, adjusted for study centre, sex, age, BMI, BP, fasting HDL-cholesterol, fasting triacylglycerol, smoking status, alcohol status and health status. The
x-axis shows OR (95% CI) and the y-axis shows each significant metabolite; metabolite classes are represented by different colours. SM, sphingomyelin
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2810 Diabetologia (2024) 67:2804–2818
enrolled in the study, a sensitivity analysis stratified by
sex was conducted, and is reported in ESM Results, ESM
Tables9–14 and ESM Fig.5.
Metabolites associated withprediabetes
anddiabetes fromuntargeted metabolomics
measurements
Fifteen metabolites were significantly changed between
NGR and IGR based on the logistic regression analyses
in the full model (Fig.2a). Fructosyl lysine had the high-
est statistically significant association with progression
to IGR (OR 1.53 [95% CI 1.37, 1.71], pfdr=8.64×10−12).
Similarly, 99 and 108 metabolites differed significantly
between NGR or IGR and type 2 diabetes, respectively
(Fig.2b,c). As a general pattern, lipids were negatively
associated and amino acids were positively associated
with progression to type 2 diabetes. 1-(1-Enyl-palmitoyl)-
2-oleoyl-GPC (P-16:0_18:1)* (OR 0.23 [95% CI 0.17,
0.31], pfdr=3.48×10−18) had the strongest association for
the NGR-T2D comparison, while cysteine-S-sulphate
(OR 3.25 [95% CI 2.55, 4.15], pfdr=3.11×10−18) was sig-
nificantly associated in the IGR-T2D comparison. Seven
metabolites (fructosyl lysine, glutamate, 1-stearoyl-
GPC (18:0), N-lactoylphenylalanine, N-lactoylvaline,
picolinoyl glycine, mannonate) appeared significant in
all comparison groups, suggesting their important roles as
diabetes risk indicators. Detailed statistics are presented
in ESM Tables15–20. A sex-based sensitivity analysis of
metabolomics data from the untargeted measurements is
reported in ESM Results, ESM Table21–26, ESM Fig.6.
Metabolites associated with HbA1c progression rate
HbA1c progression rate was computed with a condi-
tional linear mixed effect model and adjusted for changes
in BMI and diabetes medications [21]. In multivariable
linear regression analysis, lysoPC a C17:0 (β −0.0535
[95% CI −0.08, −0.0269], pfdr=0.0109), glycine (Gly) (β
−0.0509 [95% CI −0.0782, −0.0236], pfdr=0.0347) and
H1 (β 0.0481 [95% CI 0.0218, 0.0745], pfdr=0.0452) were
significantly correlated with HbA1c progression rate and
all were related to glycaemic-deterioration traits as well.
In untargeted metabolomic profiling, 20 metabolites were
significantly related to HbA1c progression rate, with pyru-
vate (β 0.0877 [95% CI 0.0609, 0.114], pfdr=1.28×10−7)
showing the strongest association. Besides pyruvate,
N-lactoylleucine, lactate, N-lactoylphenylalanine, X-15245,
N-lactoylisoleucine, N-lactoylvaline, 1-(1-enyl-palmitoyl)-
2-oleoyl-GPC (P-16:0/18:1)*, cortolone glucuronide,
X-24295, formiminoglutamate and N-lactoyltyrosine were
also significantly associated with glycaemic categories.
Tables2 and 3 show the metabolites with significant asso-
ciations, while the complete results are reported in ESM
Tables27–28.
Metabolite association withincident diabetes (IGR/
type 2 diabetes)
Several metabolites were identified to be significantly
associated with HbA1c progression rate as well as glycae-
mic category: three targeted metabolites (lysoPC a C17:0;
glycine, H1); and 12 untargeted metabolites (pyruvate,
N-lactoylleucine, lactate, N-lactoylphenylalanine, X-15245,
N-lactoylisoleucine, N-lactoylvaline, 1-[1-enyl-palmitoyl[-
2-oleoyl-GPC* [PC(P-16:0/18:1)], cortolone glucuronide,
X-24295, formiminoglutamate, N-lactoyltyrosine). Next,
we investigated their predictive value for IGR and type 2
diabetes by including baseline metabolite concentrations
and incident IGT or type 2 diabetes in follow-up timelines
in multivariable logistic regression. As shown in Table4,
lysoPC a C17:0 concentration at baseline was observed to
significantly differ in 244 incident IGR individuals com-
pared with 398 NGR control individuals after 18 months.
The sum of H1 at baseline concentrations showed significant
differences between incident IGR (at 48 month follow-up)
and NGR or incident type 2 diabetes and IGR at both the 18
month and the 48 month follow-up.
In untargeted metabolomic profiling, lactate and X-24295
baseline concentrations were significantly correlated with
IGR or type 2 diabetes incidence at the 18 month and 48
month follow-up (Table5). Formiminoglutamate, N-lac-
toylleucine and N-lactoylvaline significantly differed in 244
incident IGT individuals compared with 398 NGT control
individuals after 18 months. We did not find any significant
metabolites from untargeted measurements to predict the
incidence of IGR from NGR at 48 months.
Mediation analysis
Causal mediation analysis was employed to explore the
potential mediation effects of the identified metabolites
from baseline glycaemic status to follow-up. Consistent
with incidence results, lysoPC a C17:0 showed strong sig-
nificance (proportion of mediation by 13%, mediation effect
p=0.034, Fig.3a), indicating that this metabolite partially
mediated the glycaemic deterioration from NGR to IGR at
18 months. The positive control H1 exhibited significant
mediation effects in all groups (between 6% and 9%) as it is
mainly represented by blood glucose.
N-Lactoylvaline (proportion of mediation 24%, mediation
effect p<2×10−16), lactate (proportion of mediation 22%,
mediation effect p=0.002), N-lactoylleucine (proportion
of mediation 20%, mediation effect p=0.006), formimino-
glutamate (proportion of mediation 11%, mediation effect
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2811Diabetologia (2024) 67:2804–2818
Amino acid_1−carboxyethylphenylalanine
Amino acid_1−carboxyethylvaline
Amino acid_creatine
Amino acid_fructosyllysine
Amino acid_glutamate
Carbohydrate_lactate
Carbohydrate_pyruvate
Lipid_1−myristoyl−2−arachidonoyl−GPC (14:0/20:4)*
Lipid_1−stearoyl−GPC (18:0)
Lipid_2−hydroxyarachidate*
Lipid_cortolone glucuronide (1)
Lipid_picolinoylglycine
NA_X − 15492
NA_X − 17357
Xenobiotics_mannonate*
0.9 1.2 1.5
OR
Metabolites
a
Amino acid_1−carboxyethylisoleucine
Amino acid_1−carboxyethylleucine
Amino acid_1−carboxyethylphenylalanine
Amino acid_1−carboxyethyltyrosine
Amino acid_1−carboxyethylvaline
Amino acid_2−hydroxybutyrate/2−hydroxyisobutyrate
Amino acid_4−hydroxyglutamate
Amino acid_4−hydroxyphenylpyruvate
Amino acid_cystathionine
Amino acid_cysteine s−sulfate
Amino acid_cysteine−glutathione disulfide
Amino acid_formiminoglutamate
Amino acid_fructosyllysine
Amino acid_glutamate
Amino acid_glycine
Amino acid_hypotaurine
Amino acid_N−acetylglycine
Amino acid_phenylpyruvate
Amino acid_taurine
Carbohydrate_pyruvate
Carbohydrate_ribitol
Cofactors and vitamins_beta−cryptoxanthin
Cofactors and vitamins_carotene diol (1)
Cofactors and vitamins_carotene diol (2)
Cofactors and vitamins_carotene diol (3)
Cofactors and vitamins_nicotinamide
Energy_alpha−ketoglutarate
Lipid_1−(1−enyl−palmitoyl)−2−linoleoyl−GPC (P−16:0/18:2)*
Lipid_1−(1−enyl−palmitoyl)−2−oleoyl−GPC (P−16:0/18:1)*
Lipid_1−(1−enyl−palmitoyl)−2−palmitoleoyl−GPC (P−16:0/16:1)*
Lipid_1−(1−enyl−palmitoyl)−GPE (P−16:0)*
Lipid_1−(1−enyl−stearoyl)−GPE (P−18:0)*
Lipid_1−linoleoyl−2−linolenoyl−GPC (18:2/18:3)*
Lipid_1−linoleoyl−GPC (18:2)
Lipid_1−linoleoyl−GPE (18:2)*
Lipid_1−oleoyl−GPC (18:1)
Lipid_1−palmitoleoyl−2−linolenoyl−GPC (16:1/18:3)*
Lipid_1−palmitoyl−GPE (16:0)
Lipid_1−stearoyl−2−docosahexaenoyl−GPE (18:0/22:6)*
Lipid_1−stearoyl−2−linoleoyl−GPI (18:0/18:2)
Lipid_1−stearoyl−2−oleoyl−GPI (18:0/18:1)*
Lipid_1−stearoyl−GPC (18:0)
Lipid_1−stearoyl−GPE (18:0)
Lipid_1,2−dilinoleoyl−GPC (18:2/18:2)
Lipid_choline phosphate
Lipid_cortolone glucuronide (1)
Lipid_deoxycholic acid 12−sulfate*
Lipid_dihomo−linoleate (20:2n6)
Lipid_glycerophosphoethanolamine
Lipid_glycerophosphorylcholine (GPC)
Lipid_glycochenodeoxycholate glucuronide (1)
Lipid_glycosyl ceramide (d18:1/20:0, d16:1/22:0)*
Lipid_glycosyl ceramide (d18:2/24:1, d18:1/24:2)*
Lipid_glycosyl−N−palmitoyl−sphingosine (d18:1/16:0)
Lipid_glycosyl−N−stearoyl−sphingosine (d18:1/18:0)
Lipid_lactosyl−N−palmitoyl−sphingosine (d18:1/16:0)
Lipid_lignoceroyl sphingomyelin (d18:1/24:0)
Lipid_linoleoylcarnitine (C18:2)*
Lipid_margarate (17:0)
Lipid_myristoyl dihydrosphingomyelin (d18:0/14:0)*
Lipid_N−palmitoyl−sphingadienine (d18:2/16:0)*
Lipid_palmitate (16:0)
Lipid_phosphoethanolamine
Lipid_picolinoylglycine
Lipid_sphingomyelin (d17:1/14:0, d16:1/15:0)*
Lipid_sphingomyelin (d18:1/20:2, d18:2/20:1, d16:1/22:2)*
Lipid_sphingomyelin (d18:2/14:0, d18:1/14:1)*
Lipid_sphingomyelin (d18:2/18:1)*
Lipid_sphingomyelin (d18:2/23:0, d18:1/23:1, d17:1/24:1)*
Lipid_sphingomyelin (d18:2/23:1)*
Lipid_sphingomyelin (d18:2/24:2)*
Lipid_sphingosine 1−phosphate
Lipid_stearate (18:0)
Lipid_tricosanoyl sphingomyelin (d18:1/23:0)*
NA_X − 13844
NA_X − 15245
NA_X − 15492
NA_X − 16087
NA_X − 17340
NA_X − 17357
NA_X − 17653
NA_X − 17654
NA_X − 21467
NA_X − 21471
NA_X − 23636
NA_X − 23639
NA_X − 24295
NA_X − 24306
NA_X − 24334
Nucleotide_2'−deoxyuridine
Nucleotide_adenosine 5'−monophosphate (AMP)
Nucleotide_orotate
Peptide_gamma−glutamylglycine
Peptide_gamma−glutamylisoleucine*
Peptide_isoleucylglycine
Xenobiotics_cinnamoylglycine
Xenobiotics_erythritol
Xenobiotics_gluconate
Xenobiotics_mannonate*
012345
OR
Metabolites
b
Amino acid_1−carboxyethylisoleucine
Amino acid_1−carboxyethylleucine
Amino acid_1−carboxyethylphenylalanine
Amino acid_1−carboxyethyltyrosine
Amino acid_1−carboxyethylvaline
Amino acid_2−hydroxybutyrate/2−hydroxyisobutyrate
Amino acid_3−hydroxyisobutyrate
Amino acid_4−hydroxyglutamate
Amino acid_beta−hydroxyisovalerate
Amino acid_cysteine s−sulfate
Amino acid_cysteine−glutathione disulfide
Amino acid_formiminoglutamate
Amino acid_fructosyllysine
Amino acid_glutamate
Amino acid_glycine
Amino acid_hypotaurine
Amino acid_imidazole lactate
Amino acid_N−acetylglycine
Amino acid_N−acetyltaurine
Amino acid_pro−hydroxy−pro
Amino acid_taurine
Carbohydrate_1,5−anhydroglucitol (1,5−AG)
Carbohydrate_mannitol/sorbitol
Carbohydrate_ribitol
Cofactors and vitamins_beta−cryptoxanthin
Cofactors and vitamins_carotene diol (1)
Cofactors and vitamins_carotene diol (2)
Energy_alpha−ketoglutarate
Lipid_1−(1−enyl−palmitoyl)−2−linoleoyl−GPC (P−16:0/18:2)*
Lipid_1−(1−enyl−palmitoyl)−2−oleoyl−GPC (P−16:0/18:1)*
Lipid_1−(1−enyl−palmitoyl)−2−oleoyl−GPE (P−16:0/18:1)*
Lipid_1−(1−enyl−palmitoyl)−2−palmitoleoyl−GPC (P−16:0/16:1)*
Lipid_1−(1−enyl−palmitoyl)−GPE (P−16:0)*
Lipid_1−(1−enyl−stearoyl)−2−arachidonoyl−GPE (P−18:0/20:4)*
Lipid_1−(1−enyl−stearoyl)−2−linoleoyl−GPE (P−18:0/18:2)*
Lipid_1−(1−enyl−stearoyl)−GPE (P−18:0)*
Lipid_1−linolenoyl−GPC (18:3)*
Lipid_1−linoleoyl−2−linolenoyl−GPC (18:2/18:3)*
Lipid_1−linoleoyl−GPC (18:2)
Lipid_1−linoleoyl−GPE (18:2)*
Lipid_1−oleoyl−GPC (18:1)
Lipid_1−oleoylglycerol (18:1)
Lipid_1−palmitoleoyl−2−linolenoyl−GPC (16:1/18:3)*
Lipid_1−palmitoleoylglycerol (16:1)*
Lipid_1−palmitoyl−GPE (16:0)
Lipid_1−stearoyl−2−docosahexaenoyl−GPE (18:0/22:6)*
Lipid_1−stearoyl−2−oleoyl−GPI (18:0/18:1)*
Lipid_1−stearoyl−GPC (18:0)
Lipid_1−stearoyl−GPE (18:0)
Lipid_1,2−dilinoleoyl−GPC (18:2/18:2)
Lipid_10−heptadecenoate (17:1n7)
Lipid_10−nonadecenoate (19:1n9)
Lipid_ceramide (d18:1/14:0, d16:1/16:0)*
Lipid_deoxycholate
Lipid_deoxycholic acid 12−sulfate*
Lipid_dihomo−linoleate (20:2n6)
Lipid_dihomo−linolenate (20:3n3 or n6)
Lipid_glycerophosphoethanolamine
Lipid_glycerophosphorylcholine (GPC)
Lipid_glycosyl ceramide (d18:1/20:0, d16:1/22:0)*
Lipid_glycosyl ceramide (d18:2/24:1, d18:1/24:2)*
Lipid_glycosyl−N−palmitoyl−sphingosine (d18:1/16:0)
Lipid_glycosyl−N−stearoyl−sphingosine (d18:1/18:0)
Lipid_lactosyl−N−palmitoyl−sphingosine (d18:1/16:0)
Lipid_lignoceroyl sphingomyelin (d18:1/24:0)
Lipid_linoleoyl−arachidonoyl−glycerol (18:2/20:4) [2]*
Lipid_margarate (17:0)
Lipid_myristoyl dihydrosphingomyelin (d18:0/14:0)*
Lipid_N−palmitoyl−sphingadienine (d18:2/16:0)*
Lipid_palmitate (16:0)
Lipid_phosphoethanolamine
Lipid_picolinoylglycine
Lipid_sphingomyelin (d17:1/14:0, d16:1/15:0)*
Lipid_sphingomyelin (d18:1/20:2, d18:2/20:1, d16:1/22:2)*
Lipid_sphingomyelin (d18:1/21:0, d17:1/22:0, d16:1/23:0)*
Lipid_sphingomyelin (d18:1/25:0, d19:0/24:1, d20:1/23:0, d19:1/24:0)*
Lipid_sphingomyelin (d18:2/14:0, d18:1/14:1)*
Lipid_sphingomyelin (d18:2/18:1)*
Lipid_sphingomyelin (d18:2/21:0, d16:2/23:0)*
Lipid_sphingomyelin (d18:2/23:0, d18:1/23:1, d17:1/24:1)*
Lipid_sphingomyelin (d18:2/23:1)*
Lipid_sphingomyelin (d18:2/24:2)*
Lipid_sphingosine 1−phosphate
Lipid_stearate (18:0)
Lipid_tricosanoyl sphingomyelin (d18:1/23:0)*
NA_docosapentaenoate (n6 DPA; 22:5n6)
NA_X − 13728
NA_X − 15220
NA_X − 15461
NA_X − 16087
NA_X − 17653
NA_X − 17654
NA_X − 18887
NA_X − 21339
NA_X − 21467
NA_X − 21471
NA_X − 23639
NA_X − 24306
Nucleotide_2'−deoxyuridine
Nucleotide_adenosine 5'−monophosphate (AMP)
Nucleotide_orotate
Peptide_gamma−glutamylglycine
Peptide_gamma−glutamylisoleucine*
Peptide_isoleucylglycine
Xenobiotics_1−methylurate
Xenobiotics_7−methylxanthine
Xenobiotics_erythritol
Xenobiotics_mannonate*
1234
OR
Metabolites
Category
Amino acid
Carbohydrate
Cofactors and vitamins
Energy
Lipid
Nucleotide
Peptide
Xenobiotics
NA
c
Fig. 2 Flag plots representing the results of the multivariable logistic regression models for NGR vs IGR (a), NGR vs type 2 diabetes (b) and IGR vs type 2 diabetes (c) as dependent variables
and the metabolites as independent variables, adjusted for study centre, sex, age, BMI, BP, fasting HDL-cholesterol, fasting triacylglycerol, smoking status, alcohol status and health status. The
x-axis shows OR (95% CI) and the y-axis shows each significant metabolite; metabolite classes are represented by different colours. Asterisks (*) indicate the presence of unknown or novel
metabolites
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2812 Diabetologia (2024) 67:2804–2818
p=0.034) and X-24295 (proportion of mediation 11%, medi-
ation effect p=0.042) were all observed to show significant
mediation effects from baseline NGR to IGR at 18 months’
follow-up (Fig. 3b). Furthermore, formiminoglutamate
(proportion of mediation 23%, mediation effect p=0.006)
showed a significant mediation effect from NGR to IGR at
48 months. These results suggest that these metabolites own
a significant mediation effect on glycaemic deterioration.
MR
The availability of genetic data on type 2 diabetes makes
the use of MR particularly compelling. To assess bidi-
rectional causal relationships between type 2 diabe-
tes and metabolites (Fig.4), we employed 2SMR tests.
After multiple testing correction only the concentration
of the sum of H1 was determined by type 2 diabetes
(p<0.05/117=0.00042). For untargeted metabolites we
found instruments for only 19% of the metabolites (i.e.
151 out of 779). For example, instruments are from genes
TCF7L2, IGF2BP2, NOTCH2, CDKAL1, PABPC4, FTO
and JAZF1, known to be associated with diabetes and that
have been further significantly associated with the metab-
olites. Following multiple testing correction, it suggests
that the change in an amino acid (glutamate) and a lipid
(caproate, FA C6:0) was caused by change in type 2 diabe-
tes status (p<0.05/151=0.000331). However, metabolites
that are causal for type 2 diabetes (meaning that the change
in metabolite caused change in the disease status)included
several phosphatidylcholines, namely PC aa C36:2, PC aa
C36:5, PC ae C36:3 and PC ae C34:3, from the targeted
metabolomics dataset. From the untargeted metabolomics
Table 2 Metabolites from targeted measurements significantly associ-
ated with HbA1c progression rate from a linear regression model
The dependent variable is the HbA1c progression rate while the inde-
pendent variable is the loge-transformed and standardised baseline
concentration of a given metabolite, adjusted by age and sex
The pfdr values represent the adjusted p value for multiple testing by
Bonferroni correction
Metabolite β (95% CI) p value pfdr value
LysoPC a C17:0 −0.053 (−0.080,
−0.027)
8.25×10−5 0.011
Gly −0.051 (−0.078,
−0.024)
2.63×10−4 0.0345
H1 0.048 (0.022, 0.075) 3.42×10−4 0.045
Table 3 Metabolites from
untargeted metabolomics
measurements significantly
associated with HbA1c
progression rate from a linear
regression model
The dependent variable is the HbA1c progression rate while the independent variable is the
loge-transformed and standardised baseline concentration of a given metabolite, adjusted by age and sex.
The pfdr are adjusted p for multiple testing by Bonferroni correction
Metabolite β (95% CI) p value pfdr value
Pyruvate 0.087 (0.060, 0.114) 1.65×10−10 1.28×10−7
N-Lactoylleucine 0.082 (0.056, 0.109) 8.43×10−10 6.57×10−7
Lactate 0.075 (0.049, 0.102) 3.30×10−8 2.57×10−5
N-Lactoylphenylalanine 0.074 (0.048, 0.100) 3.66×10−8 2.85×10−5
X-15245 0.074 (0.047, 0.100) 6.24×10−8 4.86×10−5
N-Lactoylisoleucine 0.068 (0.042, 0.095) 3.11×10−7 2.42×10−4
N-Lactoylvaline 0.067 (0.041, 0.094) 5.69×10−7 4.43×10−4
X-11444 0.068 (0.041, 0.094) 6.22×10−7 4.84×10−4
Orotidine 0.065 (0.038, 0.091) 1.74×10−6 1.35×10−3
Metabolonic lactone sulphate 0.063 (0.036, 0.089) 2.9 ×10−6 2.28×10−3
3,4-Dihydroxybutyrate 0.060 (0.033, 0.087) 1.11×10−5 8.64×10−3
N4-Acetylcytidine 0.059 (0.033, 0.085) 1.16×10−5 9.06×10−3
X-24337 0.058 (0.032, 0.085) 1.47×10−5 0.011
1-(1-Enyl-palmitoyl)-2-oleoyl-GPC(P-16:0/18:1)* −0.058 (−0.084, −0.032) 1.49×10−5 0.016
X-25828 −0.058 (−0.085, −0.032) 1.50×10−5 0.017
Cortolone glucuronide 0.058 (0.032, 0.085) 1.73×10−5 0.013
X-24295 0.057 (0.031, 0.084) 1.77×10−5 0.014
Formiminoglutamate 0.059 (0.032, 0.088) 2.75×10−5 0.021
1-Palmitoyl-2-oleoyl-GPE (16:0/18:1) 0.056 (0.029, 0.082) 3.59×10−5 0.028
N-Lactoyltyrosine 0.055 (0.029, 0.082) 3.98×10−5 0.031
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2813Diabetologia (2024) 67:2804–2818
dataset,two n-3 fatty acids, namely stearidonate (18:4n3)
and docosapentaenoate (n3 DPA; 22:5n3), were identified
to be causal for type 2 diabetes. Detailed statistics of our
MR analysis are presented in ESM Tables29–32.
Discussion
In this study, we used untargeted metabolomics to provide
semi-quantitative global screening of metabolites in the
development of a disease whereas targeted metabolomics
was used to quantify a pre-selected subset of metabo-
lites with absolute concentrations. However, the overlap
between the two metabolomic techniques was limited to a
few amino acids and lipids. In the current study we report
19 metabolites (three from targeted and 14 from global
profiling, plus one common lysoPC a C18:0 / 1-stearoyl-
GPC [18:0]) that were significantly associated with predia-
betes in the DIRECT cohort. The advantages of global pro-
filing become evident as it allows for the identification of
a broader spectrum of metabolites. Few notable examples
are given here. First, picolinoylglycine (HMDB0059766),
which is potentially a phase II product of picolinic acid,
a degradation product of tryptophan [29] and glycine
[30], and shows potential as a novel marker for glycaemic
deterioration. Prediabetes is often associated with dyslipi-
daemia, marked by an imbalanced lipid profile compared
with individuals with NGR [24]. Second, N-lactoyl amino
acids are not infrequently observed in metabolomic data-
sets. In fact it has come to light that N-lactoyl amino acids
were misidentified in some metabolomic studies and were
erroneously reported as 1-carboxyethyl amino acids. In
particular, N-lactoyl-phenylalanine (Lac-Phe) is known to
act as an appetite suppressant when given to obese mice
[31]. However, in humans Lac-Phe concentrations were
observed to rise following vigorous exercise [32]. In fact,
the most recent study shows that Lac-Phe facilitates the
impact of metformin on both food intake and body weight
[33, 34]. It seems that the exact role of Lac-Phe in the
human body and pathways downstream, such as energy
metabolism, insulin signalling, exercise-induced pathways,
are unclear and needs further research.
We are aware of several limitations to our study.
Although metabolomics screening showcases numerous
valuable attributes in health science, challenges inherent
to this approach continue to exist, especially in the accu-
rate identification of metabolites which is crucial for the
biological interpretation and validation of metabolomics
Table 4 Metabolites from targeted measurements that were signifi-
cantly associated with incidence of IGR and type 2 diabetes in differ-
ent pairwise comparisons
Baseline metabolites were taken as the independent variables with
glycaemic category in different timelines (18 months and 48 months)
as the dependent variables, adjusted by study centre, sex, age, BMI,
BP, fasting HDL-cholesterol, fasting triacylglycerol, smoking status,
alcohol status and health status
ORs and p values were calculated from the logistic regression model
T2D, type 2 diabetes
Comparison OR (95% CI) p value
18 months
398 NGR vs 244 IGR
lysoPC a C17:0 −0.246 (−0.452, −0.043) 0.018
897 IGR vs 71 T2D
H1 0.545 (0.164, 0.945) 0.006
48 months
244 NGR vs 295 IGR
H1 0.433 (0.189, 0.690) 7x10−3
821 IGR vs 128 T2D
H1 0.347 (0.064, 0.642) 0.018
Table 5 Metabolites from untargeted measurements that were signifi-
cantly associated with incidence of IGR and type 2 diabetes in differ-
ent pairwise comparisons
Baseline metabolites were taken as the independent variables with
glycaemic category in different timelines (18 months and 48 months)
as the dependent variables, adjusted by study centre, sex, age, BMI,
BP, fasting HDL-cholesterol, fasting triacylglycerol, smoking status,
alcohol status and health status
ORs and p values were calculated from the logistic regression model
T2D, type 2 diabetes
Comparison OR (95% CI) p value
18 months
398 NGR vs 244 IGR
Formiminoglutamate 0.369 (0.157, 0.588) 7.7×10−4
Lactate 0.373 (0.143, 0.557) 0.002
N-Lactoylleucine 0.294 (0.079, 0.514) 0.008
N-Lactoylvaline 0.248 (0.039, 0.460) 0.021
X-24295 0.225 (0.022, 0.432) 0.031
897 IGR vs 71 T2D
X-24295 0.474 (0.162, 0.801) 3.6x10−3
Lactate 0.409 (0.077, 0.747) 1.6x10−2
48 months
821 IGR vs 128 T2D
X-24295 0.474 (0.162, 0.801) 3.6x10−3
Lactate 0.409 (0.077, 0.747) 1.6x10−2
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2814 Diabetologia (2024) 67:2804–2818
data [35]. Variability in sample collection, preparation
and analytical techniques can impact the reproducibil-
ity and comparability of results across different studies.
Standardisation efforts are ongoing but may not fully
address all sources of variation. The identification of
metabolites, especially in untargeted metabolomics, can
be challenging. Incomplete databases and the presence of
unknown or novel metabolites have been reported with
a metabolite name with an asterisk (*) sign. However,
ongoing advancements in technology, methodology and
standardisation efforts aim to enhance the robustness and
applicability of metabolomics studies [35]. The current
study is predominantly based on White male participants
from the Kuopio region of Europe, and for this reason
an additional sex-based sensitivity analysis has been per-
formed and reported separately (ESM Results 1 and 2).
Challenges in MR studies include limited statistical power,
potential reverse causation, confounding and pleiotropy
[36]. Caution is advised in interpreting causality inference,
considering the various limitations mentioned in the meth-
ods, and precautionary measures were taken by using valid
MR instruments and reporting Bonferroni significance.
A drug–metabolomics associations study [17] was exam-
ined to determine whether or not metabolites linked to type
2 diabetes from the DIRECT study were also associated
with a particular drug. Looking at our results and those of
Allesøe etal [17], we found that 44% (15 out of 34) of tar-
geted metabolites and 3% (three out of 99) of non-targeted
metabolites that were significantly associated with type 2
diabetes also showed a significant association with at least
one of the 20 drugs. This suggests that metabolites linked to
type 2 diabetes may be confounded by polypharmacy.
However, metabolite association with incident predia-
betes or diabetes (IGR-T2D) showed that lysoPC a C17:0
could predict the risk of developing IGR at 18 months and
48 months. It has already been shown that lysoPCs differ
significantly between individuals with incident IGT or type
2 diabetes and individuals with NGR in the KORA study
a
b
Fig. 3 Schematic overview of mediation analysis with lysoPC a
C17:0 and hexoses (a) or N-lactoylvaline, lactate, N-lactoylleucine,
formiminoglutamate and X-24295 (b) as mediators. Numbers above
the red arrows indicate the percentage and significance of mediation
effects. T2D, type 2 diabetes
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2815Diabetologia (2024) 67:2804–2818
[37]. LysoPC a C17:0 was negatively associated with diabe-
tes, a finding that was confirmed in several studies [38, 39].
The aforementioned drug–metabolomics association study
[17] showed that lysoPC a 17:0 was not associated with the
drugs. However, the origin of odd-chain fatty acids (mainly
C15:0 and C17:0) remains elusive. Jenkins etal [40] investi-
gated the origin of circulating odd-chain fatty acids (C17:0,
C15:0) through a combination of animal and human stud-
ies to determine possible contributions of fatty acids from
the gut-microbiota, diet and novel endogenous biosynthesis
[41]. The findings suggested that C15:0 was linked to die-
tary intake, while C17:0 was predominantly biosynthesised,
indicating independent origins and non-homologous roles in
disease causation.
Causal mediation analysis indicated that plasma lactate
strongly mediates the effects of identified metabolites in the
transition from baseline glycaemic status to follow-up [42].
In a longitudinal study of Swedish men, elevated serum lac-
tate was independently linked to a higher incidence of type
2 diabetes, irrespective of obesity measures [43]. Formimi-
noglutamate was confirmed to be associated with a higher
risk of incident type 2 diabetes in older Puerto Ricans [44].
N-lactoylleucine and N-lactoylvaline, derivatives of leucine
and valine, respectively, are ubiquitous pseudodipeptides of
lactic acid and amino acids that are formed by reverse prote-
olysis [32] and are correlated with underivatised amino acids
in human plasma. The Microbiome and Insulin Longitudinal
Evaluation Study (MILES) [45] investigated the association
between ABO haplotypes and insulin-related characteristics,
and explored possible pathways that could mediate these
associations. The study showed that the A1 haplotype poten-
tially enhances favourable insulin sensitivity in non-Hispanic
White individuals, with lactate likely influencing this mecha-
nism, while gut bacteria are not believed to be a contributing
factor.
In MR, causality signifies that modifying exposure leads
to a predictable change in the outcome. Our 2SMR analysis
suggests that the metabolites causal for type 2 diabetes are
PC aa C36:2, PC aa C36:5, PC ae C34:3 and PC ae C36:3
and all these metabolites are significantly associated with
drug–metabolomics. However, from untargeted metabo-
lomics two n-3 fatty acids, namely stearidonate (18:4n3) and
docosapentaenoate DPA 22:5n3), are not further associated
with drugs. In 2012, Banz etal [46] explored the therapeutic
implications of stearidonate acid in preventing or managing
type 2 diabetes. The Fatty Acids and Outcomes Research
Consortium (FORCE) [47] found that higher circulating bio-
markers of seafood-derived n-3 fatty acids were associated
with lower type 2 diabetes risk. On the contrary, branched-
chain amino acids [48] and sphingomyelin [15] have been
shown to have a causal role in type 2 diabetes development,
a correlation not observed in the DIRECT study.
Conclusions
Our study demonstrates that alteration in blood plasma
metabolites is associated with glycaemic deterioration. The
progression from prediabetes to diabetes is mediated by novel
0.000197
0.000566
0.000566
0.000914
0.000642
0.000766
6.35 ×1012
3.26 ×106
0.000282
PC aa C36:5
PC ae C34:3
PC ae C36:3
PC aa C36:2
Stearidonate (18:4n3)
Docosapentaenoate (n3 DPA; 22:5n3)
H1
Glutamate
Caproate_(6:0)
−1 01
Estimates
Metabolites
Category
T2D to targeted metabolomics
T2D to untargeted metabolomics
Ta rgeted metabolomics to T2D
Untargeted metabolomics to T2D
Fig. 4 Forest plot representing causal estimates of type 2 diabetes on targeted and untargeted metabolites in the two-sample MR test. T2D, type
2 diabetes
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2816 Diabetologia (2024) 67:2804–2818
metabolites such as picolinoylglycine and N-lactoyl-amino
acids, as demonstrated by evidence from the DIRECT study.
N-lactoyl-amino acids are known to be exercise-induced
metabolites that suppress food intake and influence glucose
homeostasis. Additional functional research and quantifica-
tion are needed to advance the identification of early meta-
bolic biomarkers such as N-lactoyl-amino acids, which have
the potential to forecast the onset of type 2 diabetes. Collec-
tively, these findings direct attention towards novel metabolic
signatures associated with glycaemic deterioration.
Supplementary Information The online version contains peer-reviewed
but unedited supplementary material available at https:// doi. org/ 10.
1007/ s00125- 024- 06282-6.
Acknowledgements We extend our gratitude to the IMI-DIRECT study
participants who willingly participated in phenotyping as well as to the
clinical and technical staff across European study centres for their con-
tributions to participant recruitment and clinical assessment. This pub-
lication’s development has been supported by the Innovative Medicines
Initiative Joint Undertaking under grant agreement 115317 (DIRECT),
with resources derived from the European Union’s Seventh Framework
Programme (FP7/2007–2013) and EFPIA companies’ in-kind contribu-
tions. Special thanks go to the study centre team, L. M. 't Hart, F. Rutters,
J. Vangipurapu and T. H. Hansen, for providing internal review on this
manuscript.
Data availability Access to the molecular and clinical raw data, as
well as the processed data, is restricted. This is in accordance with the
informed consent provided by study participants, the various national
ethical approvals obtained for the study, and compliance with the Euro-
pean General Data Protection Regulation (GDPR). Individual-level
clinical and molecular data cannot be transferred from the centralised
IMI-DIRECT repository. Requests for access will receive guidance on
accessing data through the DIRECT secure analysis platform after sub-
mitting an appropriate application. The IMI-DIRECT data access policy
and additional information about the IMI-DIRECT research consor-
tium’s initiatives and activities can be found at https:// direc tdiab etes. org.
Code used for MR in the study is included as ESM.
Funding Open Access funding enabled and organized by Projekt
DEAL. We would like to thank Helmholtz Munich, German Diabetes
Center (DZD) for their support in current research and China Research
Council (CRC) funding for a PhD student hosted by Helmholtz Munich.
Authors’ relationships and activities The authors declare that there are
no relationships or activities that might bias, or be perceived to bias,
their work.
Contribution statement SS, QD, MH, JA and HG conceptualised the
analysis plan. RWK, PWF, MW, IF, GG, IP, HR, MD, MM, OP, JS, KT,
FDM, SB, AV, AM, TM, TK, JA, EP and HG were involved in concep-
tion and design of the DIRECT study. SS, QD, MH, JA, GK, AA, CP,
RB, JFT, AJ and AT were involved in the data acquisition, pre-pro-
cessing and interpretation of data. SS organised inclusion of outlined
sections and, along with QD, wrote the original draft of the manuscript.
All authors contributed to drafting the article or critically revising it for
significant intellectual content and have provided approval to the final
version to be published. SS and HG are the guarantors of this work and,
as such, had full access to all the data in the study and take responsibil-
ity for the integrity of the data and the accuracy of the data analysis.
Open Access This article is licensed under a Creative Commons Attri-
bution 4.0 International License, which permits use, sharing, adapta-
tion, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in
the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
References
1. Zimmet PZ (2017) Diabetes and its drivers: the largest epidemic
in human history? Clin Diabetes Endocrinol 3:1. https:// doi. org/
10. 1186/ s40842- 016- 0039-3
2. Wesolowska-Andersen A, Brorsson CA, Bizzotto R etal (2022)
Four groups of type 2 diabetes contribute to the etiological and
clinical heterogeneity in newly diagnosed individuals: an IMI
DIRECT study. Cell Rep Med 3(1):100477. https:// doi. org/ 10.
1016/j. xcrm. 2021. 100477
3. Emerging Risk Factors Collaboration, Sarwar N, Gao P etal
(2010) Diabetes mellitus, fasting blood glucose concentration,
and risk of vascular disease: a collaborative meta-analysis of
102 prospective studies. Lancet 375(9733):2215–2222. https://
doi. org/ 10. 1016/ S0140- 6736(10) 60484-9
4. Emerging Risk Factors Collaboration, Di Angelantonio E, Kap-
toge S etal (2015) Association of cardiometabolic multimor-
bidity with mortality. JAMA 314(1):52–60. https:// doi. org/ 10.
1001/ jama. 2015. 7008
5. Rao KondapallySeshasai S, Kaptoge S, Thompson A etal
(2011) Diabetes mellitus, fasting glucose, and risk of cause-
specific death. N Engl J Med 364(9):829–841. https:// doi. org/
10. 1056/ NEJMo a1008 862
6. Wurtz P, Kangas AJ, Soininen P, Lawlor DA, Davey Smith G,
Ala-Korpela M (2017) Quantitative serum nuclear magnetic
resonance metabolomics in large-scale epidemiology: a primer
on -Omic technologies. Am J Epidemiol 186(9):1084–1096.
https:// doi. org/ 10. 1093/ aje/ kwx016
7. Guasch-Ferre M, Hruby A, Toledo E etal (2016) Metabolomics in
prediabetes and diabetes: a systematic review and meta-analysis.
Diabetes Care 39(5):833–846. https:// doi. org/ 10. 2337/ dc15- 2251
8. Cai L, Wheeler E, Kerrison ND etal (2020) Genome-wide asso-
ciation analysis of type 2 diabetes in the EPIC-InterAct study. Sci
Data 7(1):393. https:// doi. org/ 10. 1038/ s41597- 020- 00716-7
9. Yengo L, Sidorenko J, Kemper KE etal (2018) Meta-analysis of
genome-wide association studies for height and body mass index in
approximately 700000 individuals of European ancestry. Hum Mol
Genet 27(20):3641–3649. https:// doi. org/ 10. 1093/ hmg/ ddy271
10. Shin SY, Fauman EB, Petersen AK etal (2014) An atlas of genetic
influences on human blood metabolites. Nat Genet 46(6):543–550.
https:// doi. org/ 10. 1038/ ng. 2982
11. Gieger C, Geistlinger L, Altmaier E etal (2008) Genetics meets
metabolomics: a genome-wide association study of metabolite
profiles in human serum. PLoS Genet 4(11):e1000282. https://
doi. org/ 10. 1371/ journ al. pgen. 10002 82
12. Lanznaster D, Veyrat-Durebex C, Vourc’h P, Andres CR, Blasco
H, Corcia P (2020) Metabolomics: a tool to understand the impact
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2817Diabetologia (2024) 67:2804–2818
of genetic mutations in amyotrophic lateral sclerosis. Genes
(Basel) 11(5):537. https:// doi. org/ 10. 3390/ genes 11050 537
13. Suhre K, Raffler J, Kastenmuller G (2016) Biochemical insights from
population studies with genetics and metabolomics. Arch Biochem
Biophys 589:168–176. https:// doi. org/ 10. 1016/j. abb. 2015. 09. 023
14. Koivula RW, Forgie IM, Kurbasic A etal (2019) Discovery of
biomarkers for glycaemic deterioration before and after the onset
of type 2 diabetes: descriptive characteristics of the epidemio-
logical studies within the IMI DIRECT Consortium. Diabetologia
62(9):1601–1615. https:// doi. org/ 10. 1007/ s00125- 019- 4906-1
15. Zhang Z, Zheng C, Kim C, Van Poucke S, Lin S, Lan P (2016)
Causal mediation analysis in the context of clinical research. Ann
Transl Med 4(21):425. https:// doi. org/ 10. 21037/ atm. 2016. 11. 11
16. Dong Q, Sidra S, Gieger C etal (2023) Metabolic signatures eluci-
date the effect of body mass index on type 2 diabetes. Metabolites
13(2):227. https:// doi. org/ 10. 3390/ metab o1302 0227
17. Allesøe RL, Lundgaard AT, Hernandez Medina R etal (2023) Dis-
covery of drug-omics associations in type 2 diabetes with genera-
tive deep-learning models. Nat Biotechnol 41(3):399–408. https://
doi. org/ 10. 1038/ s41587- 022- 01520-x
18. Koivula RW, Heggie A, Barnett A etal (2014) Discovery of bio-
markers for glycaemic deterioration before and after the onset
of type 2 diabetes: rationale and design of the epidemiologi-
cal studies within the IMI DIRECT Consortium. Diabetologia
57(6):1132–1142. https:// doi. org/ 10. 1007/ s00125- 014- 3216-x
19. Römisch-Margl W, Prehn C, Bogumil R, Röhring C, Suhre K, Adam-
ski J (2012) Procedure for tissue sample preparation and metabolite
extraction for high-throughput targeted metabolomics. Metabo-
lomics. 8:133–142. https:// doi. org/ 10. 1007/ s11306- 011- 0293-4
20. Do KT, Wahl S, Raffler J etal (2018) Characterization of missing
values in untargeted MS-based metabolomics data and evaluation
of missing data handling strategies. Metabolomics 14(10):128.
https:// doi. org/ 10. 1007/ s11306- 018- 1420-2
21. Bizzotto R, Jennison C, Jones AG etal (2021) Processes underlying
glycemic deterioration in type 2 diabetes: an IMI DIRECT study.
Diabetes Care 44(2):511–518. https:// doi. org/ 10. 2337/ dc20- 1567
22. Baron RM, Kenny DA (1986) The moderator-mediator variable
distinction in social psychological research: conceptual, strate-
gic, and statistical considerations. J Pers Soc Psychol 51(6):1173–
1182. https:// doi. org/ 10. 1037/ 0022- 3514. 51.6. 1173
23. Tingley D, Yamamoto T, Hirose K, Keele L, Imai K (2014)
mediation: R package for causal mediation analysis. J Stat Soft
59(5):1–38. https:// doi. org/ 10. 18637/ jss. v059. i05
24. Hemani G, Zheng J, Elsworth B etal (2018) The MR-Base plat-
form supports systematic causal inference across the human phe-
nome. Elife 7. https:// doi. org/ 10. 7554/ eLife. 34408
25. Zaghlool SB, Sharma S, Molnar M etal (2021) Revealing the
role of the human blood plasma proteome in obesity using genetic
drivers. Nat Commun 12(1):1279. https:// doi. org/ 10. 1038/
s41467- 021- 21542-4
26. Xue A, Wu Y, Zhu Z etal (2018) Genome-wide association analy-
ses identify 143 risk variants and putative regulatory mechanisms
for type 2 diabetes. Nat Commun 9(1):2941. https:// doi. org/ 10.
1038/ s41467- 018- 04951-w
27. Draisma HHM, Pool R, Kobl M etal (2015) Genome-wide asso-
ciation study identifies novel genetic variants contributing to vari-
ation in blood metabolite levels. Nat Commun 6:7208. https:// doi.
org/ 10. 1038/ ncomm s8208
28. Brown AA, Fernandez-Tajes JJ, Hong MG etal (2023) Genetic
analysis of blood molecular phenotypes reveals common proper-
ties in the regulatory networks affecting complex traits. Nat Com-
mun 14(1):5062. https:// doi. org/ 10. 1038/ s41467- 023- 40569-3
29. Hu Q, Jin L, Zeng J etal (2020) Tryptophan metabolite-regulated
Treg responses contribute to attenuation of airway inflammation
during specific immunotherapy in a mouse asthma model. Hum
Vaccin Immunother 16(8):1891–1899. https:// doi. org/ 10. 1080/
21645 515. 2019. 16989 00
30. Choi JY, Kim SH, Kim JE etal (2019) Four amino acids as
serum biomarkers for anti-asthma effects in the ovalbumin-
induced asthma mouse model treated with extract of Asparagus
cochinchinensis. Lab Anim Res 35:32. https:// doi. org/ 10. 1186/
s42826- 019- 0033-x
31. Li VL, He Y, Contrepois K etal (2022) An exercise-induc-
ible metabolite that suppresses feeding and obesity. Nature
606(7915):785–790. https:// doi. org/ 10. 1038/ s41586- 022- 04828-5
32. Jansen RS, Addie R, Merkx R etal (2015) N-lactoyl-amino acids
are ubiquitous metabolites that originate from CNDP2-mediated
reverse proteolysis of lactate and amino acids. Proc Natl Acad Sci U
S A 112(21):6601–6606. https:// doi. org/ 10. 1073/ pnas. 14246 38112
33. Xiao S, Li VL, Lyu X etal (2024) Lac-Phe mediates the effects of
metformin on food intake and body weight. Nat Metab 6(4):659–
669. https:// doi. org/ 10. 1038/ s42255- 024- 00999-9
34. Scott B, Day EA, O’Brien KL etal (2024) Metformin and feeding
increase levels of the appetite-suppressing metabolite Lac-Phe
in humans. Nat Metab 6(4):651–658. https:// doi. org/ 10. 1038/
s42255- 024- 01018-7
35. Kirwan JA, Gika H, Beger RD etal (2022) Quality assurance and
quality control reporting in untargeted metabolic phenotyping:
mQACC recommendations for analytical quality management.
Metabolomics 18(9):70. https:// doi. org/ 10. 1007/ s11306- 022- 01926-3
36. Davey Smith G, Hemani G (2014) Mendelian randomization: genetic
anchors for causal inference in epidemiological studies. Hum Mol
Genet 23(R1):R89-98. https:// doi. org/ 10. 1093/ hmg/ ddu328
37. Wang-Sattler R, Yu Z, Herder C etal (2012) Novel biomarkers
for pre-diabetes identified by metabolomics. Mol Syst Biol 8:615.
https:// doi. org/ 10. 1038/ msb. 2012. 43
38. Floegel A, Stefan N, Yu Z etal (2013) Identification of serum
metabolites associated with risk of type 2 diabetes using a targeted
metabolomic approach. Diabetes 62(2):639–648. https:// doi. org/
10. 2337/ db12- 0495
39. Lee H-S, Xu T, Lee Y etal (2016) Identification of putative bio-
markers for type 2 diabetes using metabolomics in the Korea
Association REsource (KARE) cohort. Metabolomics 12(12):178.
https:// doi. org/ 10. 1007/ s11306- 016- 1103-9
40. Jenkins BJ, Seyssel K, Chiu S etal (2017) Odd chain fatty acids;
new insights of the relationship between the gut microbiota,
dietary intake, biosynthesis and glucose intolerance. Sci Rep
7:44845. https:// doi. org/ 10. 1038/ srep4 4845
41. Weitkunat K, Schumann S, Nickel D etal (2017) Odd-chain fatty
acids as a biomarker for dietary fiber intake: a novel pathway
for endogenous production from propionate. Am J Clin Nutr
105(6):1544–1551. https:// doi. org/ 10. 3945/ ajcn. 117. 152702
42. Crawford SO, Hoogeveen RC, Brancati FL etal (2010) Associa-
tion of blood lactate with type 2 diabetes: the Atherosclerosis Risk
in Communities Carotid MRI Study. Int J Epidemiol 39(6):1647–
1655. https:// doi. org/ 10. 1093/ ije/ dyq126
43. Ohlson LO, Larsson B, Bjorntorp P etal (1988) Risk factors for
type 2 (non-insulin-dependent) diabetes mellitus. Thirteen and
one-half years of follow-up of the participants in a study of Swed-
ish men born in 1913. Diabetologia 31(11):798–805. https:// doi.
org/ 10. 1007/ BF002 77480
44. Rivas-Tumanyan S, Pacheco LS, Haslam DE etal (2022) Novel
plasma metabolomic markers associated with diabetes progression
in older puerto ricans. Metabolites 12(6):513. https:// doi. org/ 10.
3390/ metab o1206 0513
45. Li-Gao R, Grubbs K, Bertoni AG etal (2022) The roles of gut
microbiome and plasma metabolites in the associations between
ABO blood groups and insulin homeostasis: the Microbiome and
Insulin Longitudinal Evaluation Study (MILES). Metabolites
12(9):787. https:// doi. org/ 10. 3390/ metab o1209 0787
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2818 Diabetologia (2024) 67:2804–2818
46. Banz WJ, Davis JE, Clough RW, Cheatwood JL (2012) Steari-
donic acid: is there a role in the prevention and management of
type 2 diabetes mellitus? J Nutr 142(3):635S-640S. https:// doi.
org/ 10. 3945/ jn. 111. 146829
47. Qian F, Ardisson Korat AV, Imamura F etal (2021) n-3 fatty acid
biomarkers and incident type 2 diabetes: an individual participant-
level pooling project of 20 prospective cohort studies. Diabetes Care
44(5):1133–1142. https:// doi. org/ 10. 2337/ dc20- 2426
48. Lotta LA, Scott RA, Sharp SJ etal (2016) Genetic predisposition to
an impaired metabolism of the branched-chain amino acids and risk
of type 2 diabetes: a mendelian randomisation analysis. PLoS Med
13(11):e1002179. https:// doi. org/ 10. 1371/ journ al. pmed. 10021 79
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Authors and Aliations
SapnaSharma1· QiulingDong1,2· MarkHaid3· JonathanAdam1,4· RobertoBizzotto5· JuanJ.Fernandez‑Tajes6·
AngusG.Jones7· AndreaTura5· AnnaArtati3· CorneliaPrehn3· GabiKastenmüller8· RobertW.Koivula9·
PaulW.Franks10· MarkWalker11· IanM.Forgie12· GiuseppeGiordano13· ImrePavo14· HartmutRuetten15·
ManolisDermitzakis16,17,18· MarkI.McCarthy6· OlufPedersen19,20· JochenM.Schwenk21·
KonstantinosD.Tsirigos22· FedericoDeMasi22· SorenBrunak20,23· AnaViñuela24· AndreaMari5·
TimothyJ.McDonald25· TarjaKokkola26· JerzyAdamski27,28,29· EwanR.Pearson12· HaraldGrallert1,4
* Sapna Sharma
sapna.sharma@helmholtz-munich.de
* Harald Grallert
harald.grallert@helmholtz-munich.de
1 Research Unit ofMolecular Epidemiology, Institute
ofEpidemiology, German Research Center
forEnvironmental Health, Helmholtz Zentrum München,
Neuherberg, Germany
2 Faculty ofMedicine, Ludwig-Maximilians-University
München, Munich, Germany
3 Metabolomics andProteomics Core, German Research
Center forEnvironmental Health, Helmholtz Zentrum
München, Neuherberg, Germany
4 German Center forDiabetes Research (DZD),
MünchenNeuherberg, Germany
5 Institute ofNeuroscience, National Research Council,
Padova, Italy
6 Wellcome Trust Centre forHuman Genetics, University
ofOxford, Oxford, UK
7 Department ofClinical andBiomedical Sciences, University
ofExeter College ofMedicine & Health, Exeter, UK
8 Institute ofComputational Biology, Helmholtz Zentrum
München, Munich, Germany
9 Oxford Centre forDiabetes, Endocrinology andMetabolism,
University ofOxford, Oxford, UK
10 Department ofClinical Science, Genetic andMolecular
Epidemiology, Lund University Diabetes Centre, Malmö,
Sweden
11 Translational andClinical Research Institute, Faculty
ofMedical Sciences, University ofNewcastle,
NewcastleuponTyne, UK
12 Population Health andGenomics, Ninewells Hospital
andMedical School, University ofDundee, Dundee, UK
13 Department ofClinical Science, Genetic andMolecular
Epidemiology, Lund University Diabetes Centre, Malmö,
Sweden
14 Eli Lilly Regional Operations GmbH, Vienna, Austria
15 Sanofi Partnering, Sanofi-Aventis Deutschland GmbH,
FrankfurtamMain, Germany
16 Department ofGenetic Medicine andDevelopment,
University ofGeneva Medical School, Geneva, Switzerland
17 Institute forGenetics andGenomics inGeneva (iGE3),
University ofGeneva, Geneva, Switzerland
18 Swiss Institute ofBioinformatics, Geneva, Switzerland
19 Center forClinical Metabolic Research, Herlev andGentofte
University Hospital, Copenhagen, Denmark
20 Novo Nordisk Foundation Center forBasic Metabolic
Research, Faculty ofHealth andMedical Sciences,
University ofCopenhagen, Copenhagen, Denmark
21 Science forLife Laboratory, School ofBiotechnology, KTH
- Royal Institute ofTechnology, Solna, Sweden
22 Department ofHealth Technology, Technical University
ofDenmark, KongensLyngby, Denmark
23 Department ofHealth Technology, Technical University
ofDenmark, KongensLyngby, Denmark
24 Biosciences Institute, Faculty ofMedical Sciences,
University ofNewcastle, NewcastleuponTyne, UK
25 Blood Sciences, Royal Devon andExeter NHS Foundation
Trust, Exeter, UK
26 Internal Medicine, Institute ofClinical Medicine, University
ofEastern Finland, Kuopio, Finland
27 Department ofBiochemistry, Yong Loo Lin School
ofMedicine, National University ofSingapore, Singapore,
Singapore
28 Institute ofExperimental Genetics, German Research Center
forEnvironmental Health, Helmholtz Zentrum München,
Neuherberg, Germany
29 Institute ofBiochemistry, Faculty ofMedicine, University
ofLjubljana, Ljubljana, Slovenia
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... N-lactoyltyrosine, which belongs to a class of pseudopeptides, formed by lactic acid and an amino acid (52), was associated with brain health in our study, with higher levels being linked to lower brain volume. N-lactoyl amino acids received some attention in diabetes research recently (53,54); while higher levels of N-lactoyl amino acids (including Nlactoylphenylalanine, N-lactoyltyrosine, and N-lactoylleucine) associate with decreasing is the author/funder, who has granted medRxiv a license to display the preprint in (which was not certified by peer review) preprint ...
Preprint
Full-text available
Increasing evidence suggests the involvement of metabolic alterations in neurological disorders, including Alzheimer’s disease (AD), and highlights the significance of the peripheral metabolome, influenced by genetic factors and modifiable environmental exposures, for brain health. In this study, we examined 1,387 metabolites in plasma samples from 1,082 dementia-free middle-aged participants of the population-based Rotterdam Study. We assessed the relation of metabolites with general cognition (G-factor) and magnetic resonance imaging (MRI) markers using linear regression and estimated the variance of these metabolites explained by genes, gut microbiome, lifestyle factors, common clinical comorbidities, and medication using gradient boosting decision tree analysis. Twenty-one metabolites and one metabolite were significantly associated with total brain volume and total white matter lesions, respectively. Fourteen metabolites showed significant associations with G-factor, with ergothioneine exhibiting the largest effect (adjusted mean difference = 0.122, P = 4.65x10 ⁻⁷ ). Associations for nine of the 14 metabolites were replicated in an independent, older cohort. The metabolite signature of incident AD in the replication cohort resembled that of cognition in the discovery cohort, emphasizing the potential relevance of the identified metabolites to disease pathogenesis. Lifestyle, clinical variables, and medication were most important in determining these metabolites’ blood levels, with lifestyle, explaining up to 28.6% of the variance. Smoking was associated with ten metabolites linked to G-factor, while diabetes and antidiabetic medication were associated with 13 metabolites linked to MRI markers, including N-lactoyltyrosine. Antacid medication strongly affected ergothioneine levels. Mediation analysis revealed that lower ergothioneine levels may partially mediate negative effects of antacids on cognition (31.5%). Gut microbial factors were more important for the blood levels of metabolites that were more strongly associated with cognition and incident AD in the older replication cohort (beta-cryptoxanthin, imidazole propionate), suggesting they may be involved later in the disease process. The detailed results on how multiple modifiable factors affect blood levels of cognition- and brain imaging-related metabolites in dementia-free participants may help identify new AD prevention strategies.
Article
Background Sleep is essential to maintaining health and wellbeing of individuals, influencing a variety of outcomes from mental health to cardiometabolic disease. This study aims to assess the relationships between various sleep-related phenotypes and blood metabolites. Methods Utilising data from the Hispanic Community Health Study/Study of Latinos, we performed association analyses between 40 sleep-related phenotypes, grouped in several domains (sleep disordered breathing (SDB), sleep duration, sleep timing, self-reported insomnia symptoms, excessive daytime sleepiness (EDS), and heart rate during sleep), and 768 metabolites measured via untargeted metabolomics profiling. Network analysis was employed to visualise and interpret the associations between sleep phenotypes and metabolites. Findings The patterns of statistically significant associations between sleep phenotypes and metabolites differed by superpathways, and highlighted subpathways of interest for future studies. For example, primary bile acid metabolism showed the highest cumulative percentage of statistically significant associations across all sleep phenotype domains except for SDB and EDS phenotypes. Several metabolites were associated with multiple sleep phenotypes, from a few domains. Glycochenodeoxycholate, vanillyl mandelate (VMA) and 1-stearoyl-2-oleoyl-GPE (18:0/18:1) were associated with the highest number of sleep phenotypes, while pregnenolone sulfate was associated with all sleep phenotype domains except for sleep duration. N-lactoyl amino acids such as N-lactoyl phenylalanine (lac-Phe), were associated with sleep duration, SDB, sleep timing and heart rate during sleep. Interpretation This atlas of sleep–metabolite associations will facilitate hypothesis generation and further study of the metabolic underpinnings of sleep health. Funding R01HL161012, R35HL135818, R01AG80598.
Article
Full-text available
Metformin is a widely prescribed anti-diabetic medicine that also reduces body weight. There is ongoing debate about the mechanisms that mediate metformin’s effects on energy balance. Here, we show that metformin is a powerful pharmacological inducer of the anorexigenic metabolite N-lactoyl-phenylalanine (Lac-Phe) in cells, in mice and two independent human cohorts. Metformin drives Lac-Phe biosynthesis through the inhibition of complex I, increased glycolytic flux and intracellular lactate mass action. Intestinal epithelial CNDP2⁺ cells, not macrophages, are the principal in vivo source of basal and metformin-inducible Lac-Phe. Genetic ablation of Lac-Phe biosynthesis in male mice renders animals resistant to the effects of metformin on food intake and body weight. Lastly, mediation analyses support a role for Lac-Phe as a downstream effector of metformin’s effects on body mass index in participants of a large population-based observational cohort, the Multi-Ethnic Study of Atherosclerosis. Together, these data establish Lac-Phe as a critical mediator of the body weight-lowering effects of metformin.
Article
Full-text available
Metformin, a widely used first-line treatment for type 2 diabetes (T2D), is known to reduce blood glucose levels and suppress appetite. Here we report a significant elevation of the appetite-suppressing metabolite N-lactoyl phenylalanine (Lac-Phe) in the blood of individuals treated with metformin across seven observational and interventional studies. Furthermore, Lac-Phe levels were found to rise in response to acute metformin administration and post-prandially in patients with T2D or in metabolically healthy volunteers.
Article
Full-text available
We evaluate the shared genetic regulation of mRNA molecules, proteins and metabolites derived from whole blood from 3029 human donors. We find abundant allelic heterogeneity, where multiple variants regulate a particular molecular phenotype, and pleiotropy, where a single variant associates with multiple molecular phenotypes over multiple genomic regions. The highest proportion of share genetic regulation is detected between gene expression and proteins (66.6%), with a further median shared genetic associations across 49 different tissues of 78.3% and 62.4% between plasma proteins and gene expression. We represent the genetic and molecular associations in networks including 2828 known GWAS variants, showing that GWAS variants are more often connected to gene expression in trans than other molecular phenotypes in the network. Our work provides a roadmap to understanding molecular networks and deriving the underlying mechanism of action of GWAS variants using different molecular phenotypes in an accessible tissue.
Article
Full-text available
Obesity plays an important role in the development of insulin resistance and diabetes, but the molecular mechanism that links obesity and diabetes is still not completely understood. Here, we used 146 targeted metabolomic profiles from the German KORA FF4 cohort consisting of 1715 participants and associated them with obesity and type 2 diabetes. In the basic model, 83 and 51 metabolites were significantly associated with body mass index (BMI) and T2D, respectively. Those metabolites are branched-chain amino acids, acylcarnitines, lysophospholipids, or phosphatidylcholines. In the full model, 42 and 3 metabolites were significantly associated with BMI and T2D, respectively, and replicate findings in the previous studies. Sobel mediation testing suggests that the effect of BMI on T2D might be mediated via lipids such as sphingomyelin (SM) C16:1, SM C18:1 and diacylphosphatidylcholine (PC aa) C38:3. Moreover, mendelian randomization suggests a causal relationship that BMI causes the change of SM C16:1 and PC aa C38:3, and the change of SM C16:1, SM C18:1, and PC aa C38:3 contribute to T2D incident. Biological pathway analysis in combination with genetics and mice experiments indicate that downregulation of sphingolipid or upregulation of phosphatidylcholine metabolism is a causal factor in early-stage T2D pathophysiology. Our findings indicate that metabolites like SM C16:1, SM C18:1, and PC aa C38:3 mediate the effect of BMI on T2D and elucidate their role in obesity related T2D pathologies.
Article
Full-text available
The application of multiple omics technologies in biomedical cohorts has the potential to reveal patient-level disease characteristics and individualized response to treatment. However, the scale and heterogeneous nature of multi-modal data makes integration and inference a non-trivial task. We developed a deep-learning-based framework, multi-omics variational autoencoders (MOVE), to integrate such data and applied it to a cohort of 789 people with newly diagnosed type 2 diabetes with deep multi-omics phenotyping from the DIRECT consortium. Using in silico perturbations, we identified drug–omics associations across the multi-modal datasets for the 20 most prevalent drugs given to people with type 2 diabetes with substantially higher sensitivity than univariate statistical tests. From these, we among others, identified novel associations between metformin and the gut microbiota as well as opposite molecular responses for the two statins, simvastatin and atorvastatin. We used the associations to quantify drug–drug similarities, assess the degree of polypharmacy and conclude that drug effects are distributed across the multi-omics modalities. Clinical multi-omics data are integrated and analyzed using a generative deep-learning model.
Article
Full-text available
Background Demonstrating that the data produced in metabolic phenotyping investigations (metabolomics/metabonomics) is of good quality is increasingly seen as a key factor in gaining acceptance for the results of such studies. The use of established quality control (QC) protocols, including appropriate QC samples, is an important and evolving aspect of this process. However, inadequate or incorrect reporting of the QA/QC procedures followed in the study may lead to misinterpretation or overemphasis of the findings and prevent future metanalysis of the body of work. Objective The aim of this guidance is to provide researchers with a framework that encourages them to describe quality assessment and quality control procedures and outcomes in mass spectrometry and nuclear magnetic resonance spectroscopy-based methods in untargeted metabolomics, with a focus on reporting on QC samples in sufficient detail for them to be understood, trusted and replicated. There is no intent to be proscriptive with regard to analytical best practices; rather, guidance for reporting QA/QC procedures is suggested. A template that can be completed as studies progress to ensure that relevant data is collected, and further documents, are provided as on-line resources. Key reporting practices Multiple topics should be considered when reporting QA/QC protocols and outcomes for metabolic phenotyping data. Coverage should include the role(s), sources, types, preparation and uses of the QC materials and samples generally employed in the generation of metabolomic data. Details such as sample matrices and sample preparation, the use of test mixtures and system suitability tests, blanks and technique-specific factors are considered and methods for reporting are discussed, including the importance of reporting the acceptance criteria for the QCs. To this end, the reporting of the QC samples and results are considered at two levels of detail: “minimal” and “best reporting practice” levels.
Article
Full-text available
Non-O blood groups are associated with decreased insulin sensitivity and risk of type 2 diabetes. A recent study pinpointed the associations between ABO blood groups and gut microbiome, which may serve as potential mediators for the observed increased disease risks. We aimed to characterize associations between ABO haplotypes and insulin-related traits as well as potential mediating pathways. We assessed insulin homeostasis in African Americans (AAs; n = 109) and non-Hispanic whites (n = 210) from the Microbiome and Insulin Longitudinal Evaluation Study. The ABO haplotype was determined by six SNPs located in the ABO gene. Based on prior knowledge, we included 21 gut bacteria and 13 plasma metabolites for mediation analysis. In the white study cohort (60 ± 9 years, 42% male), compared to the O1 haplotype, A1 was associated with a higher Matsuda insulin sensitivity index, while a lower relative abundance of Bacteroides massiliensis and lactate levels. Lactate was a likely mediator of this association but not Bacteroides massiliensis. In the AAs group (57 ± 8 years, 33% male), we found no association between any haplotype and insulin-related traits. In conclusion, the A1 haplotype may promote healthy insulin sensitivity in non-Hispanic whites and lactate likely play a role in this process but not selected gut bacteria.
Article
Full-text available
Exercise confers protection against obesity, type 2 diabetes and other cardiometabolic diseases1–5. However, the molecular and cellular mechanisms that mediate the metabolic benefits of physical activity remain unclear⁶. Here we show that exercise stimulates the production of N-lactoyl-phenylalanine (Lac-Phe), a blood-borne signalling metabolite that suppresses feeding and obesity. The biosynthesis of Lac-Phe from lactate and phenylalanine occurs in CNDP2⁺ cells, including macrophages, monocytes and other immune and epithelial cells localized to diverse organs. In diet-induced obese mice, pharmacological-mediated increases in Lac-Phe reduces food intake without affecting movement or energy expenditure. Chronic administration of Lac-Phe decreases adiposity and body weight and improves glucose homeostasis. Conversely, genetic ablation of Lac-Phe biosynthesis in mice increases food intake and obesity following exercise training. Last, large activity-inducible increases in circulating Lac-Phe are also observed in humans and racehorses, establishing this metabolite as a molecular effector associated with physical activity across multiple activity modalities and mammalian species. These data define a conserved exercise-inducible metabolite that controls food intake and influences systemic energy balance.
Article
Full-text available
We assessed longitudinal associations between plasma metabolites, their network-derived clusters, and type 2 diabetes (T2D) progression in Puerto Rican adults, a high-risk Hispanic subgroup with established health disparities. We used data from 1221 participants free of T2D and aged 40–75 years at baseline in the Boston Puerto Rican Health and San Juan Overweight Adult Longitudinal Studies. We used multivariable Poisson regression models to examine associations between baseline concentrations of metabolites and incident T2D and prediabetes. Cohort-specific estimates were combined using inverse-variance weighted fixed-effects meta-analyses. A cluster of 13 metabolites of branched chain amino acids (BCAA), and aromatic amino acid metabolism (pooled IRR = 1.87, 95% CI: 1.28; 2.73), and a cell membrane component metabolite cluster (pooled IRR = 1.54, 95% CI: 1.04; 2.27) were associated with a higher risk of incident T2D. When the metabolites were tested individually, in combined analysis, 5 metabolites involved in BCAA metabolism were associated with incident T2D. These findings highlight potential prognostic biomarkers to identify Puerto Rican adults who may be at high risk for diabetes. Future studies should examine whether diet and lifestyle can modify the associations between these metabolites and progression to T2D.
Article
Full-text available
The presentation and underlying pathophysiology of type 2 diabetes (T2D) is complex and heterogeneous. Recent studies attempted to stratify T2D into distinct subgroups using data-driven approaches, but their clinical utility may be limited if categorical representations of complex phenotypes are suboptimal. We apply a soft-clustering (archetype) method to characterize newly diagnosed T2D based on 32 clinical variables. We assign quantitative clustering scores for individuals and investigate the associations with glycemic deterioration, genetic risk scores, circulating omics biomarkers, and phenotypic stability over 36 months. Four archetype profiles represent dysfunction patterns across combinations of T2D etiological processes and correlate with multiple circulating biomarkers. One archetype associated with obesity, insulin resistance, dyslipidemia, and impaired β cell glucose sensitivity corresponds with the fastest disease progression and highest demand for anti-diabetic treatment. We demonstrate that clinical heterogeneity in T2D can be mapped to heterogeneity in individual etiological processes, providing a potential route to personalized treatments.