Nutrition & Diabetes www.nature.com/nutd /\RY|£LE OPEN Check for updates Multi-omics signatures in new-onset diabetes predict metabolic response to dietary inulin: findings from an observational study followed by an interventional trial N. Ďáskova1 1 2 , I. M o d o s 2 , M. Krbcová 3 , M . K u z m a 4 , H. Pelantová4 , J . Hradecký5 , M . Heczková2 , M . Bratova2 , P. Vídeňská6 , P. Šplíchalová7 , M . Králová8 1 2 , M. Heniková 3 , J . Potočková3 , A. Ouřadová3 , R. Landberg 9 , T. K ü h n 1 0 1 1 1 3 , M . Cahová 2 1 3 H and J. Gojda 3 1 3 © The Author(s) 2023 AIM: The metabolic performance of the gut microbiota contributes to the onset of type 2 diabetes. However, targeted dietary interventions are limited by the highly variable inter-individual response. We hypothesized (1) that the composition of the complex gut microbiome and metabolome (MIME) differ across metabolic spectra (lean-obese-diabetes); (2) that specific MIME patterns could explain the differential responses to dietary inulin; and (3) that the response can be predicted based on baseline MIME signature and clinical characteristics. METHOD: Forty-nine patients with newly diagnosed pre/diabetes (DM), 66 metabolically healthy overweight/obese (OB), and 32 healthy lean (LH) volunteers were compared in a cross-sectional case-control study integrating clinical variables, dietary intake, gut microbiome, and fecal/serum metabolomes (16S rRNA sequencing, metabolomics profiling). Subsequently, 27 D M were recruited for a predictive study: 3 months of dietary inulin (10g/day) intervention. RESULTS: MIME composition was different between groups. While the D M and LH groups represented opposite poles of the abundance spectrum, OB was closer to DM. Inulin supplementation was associated with an overall improvement in glycemic indices, though the response was very variable, with a shift in microbiome composition toward a more favorable profile and increased serum butyric and propionic acid concentrations. The improved glycemic outcomes of inulin treatment were dependent on better baseline glycemic status and variables related to the gut microbiota, including the abundance of certain bacterial taxa (i.e., Blautia, Eubacterium halii group, Lachnoclostridium, Ruminiclostridium, Dialister, or Phascolarctobacterium), serum concentrations of branched-chain amino acid derivatives and asparagine, and fecal concentrations of indole and several other volatile organic compounds. CONCLUSION: W e demonstrated that obesity is a stronger determinant of different MIME patterns than impaired glucose metabolism. The large inter-individual variability in the metabolic effects of dietary inulin was explained by differences in baseline glycemic status and MIME signatures. These could be further validated to personalize nutritional interventions in patients with newly diagnosed diabetes. Nutrition and Diabetes (2023)13:7; https://doi.org/10.1038/s41387-023-00235-5 Obesity and its associated metabolic diseases, including type 2 diabetes, currently represent o n e of the greatest challenges to global health care [1]. Recently, it has been suggested that the composition and performance of the gut microbiota contribute to individual risks. The critical role of the gut microbiota in the development of obesity was suggested by a seminal study by Turnbaugh [2], followed by others confirming differences in microbiota composition between lean and obese individuals [3,4]. Further research showed an association between the gut microbiota and the development of type 2 diabetes [5-8], with evidence of a specific gut microbiota signature characteristic of prediabetes [9, 10]. However, while many studies suggest that type 2 diabetes is associated with gut dysbiosis [11], results on the composition and function of the microbiota are inconsistent and sometimes contradictory. For example, a-diversity has been reported to be significantly lower [6, 12, 13], not significantly 'First Faculty o f M e d i c i n e , Charles University, Prague, C z e c h Republic, i n s t i t u t e for Clinical a n d E x p e r i m e n t a l M e d i c i n e , Prague, C z e c h Republic, d e p a r t m e n t o f Internal M e d i c i n e , Kralovske V i n o h r a d y University Hospital a n d Third Faculty o f M e d i c i n e , Charles University, Prague, C z e c h Republic, i n s t i t u t e o f M i c r o b i o l o g y o f t h e CAS, Prague, C z e c h Republic. 5 F a c u l t y o f Forestry a n d W o o d Sciences, C z e c h University o f Life Sciences, Prague, C z e c h Republic. 6 M e n d e l University, D e p a r t m e n t o f C h e m i s t r y a n d Biochemistry, Brno, C z e c h Republic. 7 R E C E T O X , Faculty o f Science Masaryk University, Brno, C z e c h Republic. 8 A m b i s University, D e p a r t m e n t o f E c o n o m i c s a n d M a n a g e m e n t , Prague, C z e c h Republic. ' D i v i s i o n o f F o o d a n d Nutrition Science, D e p a r t m e n t o f Biology a n d Biological E n g i n e e r i n g , C h a l m e r s University o f T e c h n o l o g y , G o t e b o r g , S w e d e n , ' " i n s t i t u t e o f G l o b a l F o o d Security, Q u e e n ' s University Belfast, Belfast, UK. " H e i d e l b e r g Institute o f G l o b a l Health (HIGH), M e d i c a l Faculty a n d University Hospital, H e i d e l b e r g University, H e i d e l b e r g , G e r m a n y . 1 2 P r e s e n t address: Institute for Clinical a n d Experimental M e d i c i n e , Prague, C z e c h Republic. 1 3 T h e s e authors c o n t r i b u t e d equally: T. Kúhn, M . Cahová, J . Gojda. H e m a i l : m o n i k a . c a h o v a @ i k e m . c z Received: 9 August 2022 Revised: 22 March 2023 Accepted: 6 A p r i l 2023 Published online: 21 April 2023 SPRINGER NATURE N. Ďáskova et al. reduced [14], or comparable to nondiabetic subjects in patients with T2D [15,16]. Most studies report significant differences in the composition of the gut microbiota between diseased a n d healthy subjects [17], but they differ greatly with respect to specific taxa. Some studies show that T2D is associated with an increased Firmicutes/Bacteroidetes ratio [6, 13, 14, 18, 19], whereas others report a significant increase [14, 18] or decrease [6, 13] in Proteobacteria. A t the genus level, there are f e w dysregulated taxa that have been consistently reported, i.e., an increase in Streptococcus [9, 15, 20], Escherichia [15, 21, 22], Veillonella [6, 21], Lactobacillus [13, 18, 23], a n d Collinsella [12, 15]; decrease in Akkermansia [15, 18], Dialister [15, 19], Haemophilus [12, 15], Roseburia [12,15], a n d Faecalibacterium [10,12,13], whereas many others show changes in both directions [17]. Diet composition is a known risk factor for the d e v e l o p m e n t of type 2 diabetes. In addition to direct effects o n host physiology, diet plays an important role in shaping the microbiome, thereby influencing its metabolic program [24]. Therefore, dietary interventions focused on modulating the composition and/or performance of the gut microbiota appear to be a promising therapeutic target. Supplementation with prebiotic supplements, a n d dietary fiber in particular, is often r e c o m m e n d e d as a beneficial treatment for non-communicable diseases, but controlled clinical trials indicate pronounced differences in response to treatment, with considerable personal variability [25]. The underlying causes are not yet clear, but strong inter-individual differences in microbial response to dietary fiber likely play a key role [26, 27]. Therefore, the identification of the microbial taxa that mediate the beneficial effects of dietary fiber may o p e n n e w avenues for individualized treatment approaches [28]. In the present study, w e aimed to determine (i) whether the composition of the complex gut microbiome a n d metabolome (MIME) differ in lean healthy, obese healthy, a n d obese diabetic drug-naive type 2 diabetic patients; (ii) whether the effects of inulin o n glucose tolerance a n d insulin sensitivity can be explained, at least in part, by the response of the gut microbiota to inulin intervention; a n d (iii) whether this response can be predicted from the initial MIME signature. MATERIAL AND METHODS The current study was performed within t h e framework o f t h e TRIEMA project: Treatment o f Insulin Resistance b y Modification o f Gut Microbiota (ClinicalTrials.gov Identifier: NCT03710850). The first study from the project has been already published [24]. Study design and population Observational study. Forty-nine newly diagnosed patients with pre/ diabetes ( D M : BMI >25, fasting glycemia > 5 . 6 m M , and/or 2hOGTT glycemia >7.8mM), 66 metabolically healthy overweight/obese (OB: BMI >25) a n d 32 lean healthy (LH: BMI <25) subjects were screened a n d enrolled in t h e cross-sectional case-control study. A clinical visit was scheduled after enrollment. Volunteers were examined after a 12-h overnight fast; blood a n d urine samples were collected; a clinical examination, bioimpedance analysis, a n d oral glucose tolerance test (OGTT, 75 g glucose) were performed. A prospective 3-day dietary record and stool samples were collected from each participant. Dietary records and stool samples were obtained n o longer than a week after t h e clinical visit. Prospective study. Twenty-seven patients (DM) were then enrolled in a onearm, non-controlled intervention study in which they were fed 10 g of inulin daily for 3 months. The sample size determination for the intervention study was calculated for the primary outcome, glucose disposal (GD). According t o GD, standard deviations ranged from 1.8 t o 2.5 mg/kg/min in both insulinsensitive and insulin-resistant individuals, with high insulin levels (i.e., 80 m i l / m2 ) showing less variability with SD u p t o 0.51 [29]. W e anticipate that participants will respond individually t o the intervention, and w e will divide them into tertiles (responders, neutral, and non-responders). If we consider a difference between changes of 2 0 % (i.e., ~1.5 mg/kg/min) t o be significant t o have 9 0 % power to detect a difference at the 0.05 alpha level, we must have 6 subjects in each group. To account for dropouts or incomplete data, we aimed t o have at least 9 subjects in each group (i.e., responders vs. nonresponders). Baseline and post-intervention examinations were identical t o those described above. In addition, indirect calorimetry and a two-step glucose clamp (10 and 8 0 m l U / m 2 BSA insulin dose) were performed [30], Insulin sensitivity (IS) of adipose tissue was expressed as the change in nonesterified fatty acids (NEFA) and plasma glycerol levels from baseline t o the steady state of the first step of the clamp, whereas IS o f skeletal muscle was expressed as space-corrected glucose infusion rate per kg fat-free mass (Mcor mg/kg FFM/min) and metabolic clearance of glucose divided by steady-state insulinemia (MCR/I, ml/kg FFM/min) at the steady state of the second step. Detailed calculations are described in Supplementary Material. All participants signed an informed consent before enrollment in each respective study. The research protocol was approved by the Ethics Committee of University Hospital Kralovske Vinohrady (EK-VP /26/0/2017) in accordance with the Declaration of Helsinki. The study was registered under NCT03710850. Gut microbiome analysis DNA from stool samples was isolated using the QIAmp PowerFecal D N A Kit (Qiagen, Hilden, Germany), and the V 4 region o f the bacterial 16 S rRNA gene was amplified b y PCR. Sequencing was performed using the Miseq reagent kit V2 with a MiSeq instrument (lllumina, Hayward, CA, USA). The raw sequences were processed using a D A D A 2 A m p l i c o n Denoiser [31]. Short-chain fatty acids (SCFA) in plasma SCFA were analyzed in plasma by LC-MS according t o a method described before [32]. Volatile compounds (VOCs) analysis in feces Volatile fingerprinting o f fecal samples was performed using an Agilent 7890B gas chromatograph (Santa Clara, California, USA) coupled t o a Pegasus 4 D time-of-flight mass spectrometer (LECO, Geleen, The Netherlands). Data acquisition and initial data processing were performed using instrumental SW ChromaTOF by LECO. NMR analyses Serum samples (after protein precipitation) were measured o n a 600 M H z Bruker Avance III spectrometer (Bruker BioSpin, Rheinstetten, Germany) equipped with a 5 m m TCI cryogenic probe head. The concentrations of individual metabolites, identified by comparison o f proton a n d carbon chemical shift with t h e H M D B database, were expressed as P Q N [33] normalized intensities of corresponding signals in C P M G spectra. The list of quantified metabolites with corresponding ' H and 1 3 C chemical shifts is given in Table S1. The representative 1 H N M R spectrum is shown in Fig. S1. Statistics The statistical analyses were performed using R software packages and inhouse scripts [34]. T h e microbiome a n d VOCs data were treated as compositional (proportions o f total read count in each sample or proportion of the total area o f selected masses), and before all statistical analyses, the data were transformed by centered log-ratio (clr) transformation with a multiplicative simple replacement for handling zero values. According t o their abundance and prevalence, the bacteria were classified as "core microbial taxa" w h e n fulfilling t h e following conditions, i.e. abundance o f > 0 . 1 % and prevalence o f > 7 5 % at least in o n e experimental group. Other microbial taxa were classified as rare. All methods are described in detail in Supplementary Material. RESULTS Observational study: clinical characteristics The clinical characteristics of the study participants are shown in Table 1. As expected, the groups differed in terms of glycemic indices, insulin sensitivity, a n d beta cell function. Biomarkers of lipid metabolism were significantly elevated in both the OB a n d D M groups compared with LH. Observational study: fecal microbiome composition In all samples, w e f o u n d 44,332 amplicon sequence variants (ASVs) and identified 13 phyla, 30 classes, 56 orders, 104 families, a n d 367 genera. Considering only the ASVs, all a-diversity indices were SPRINGER NATURE Nutrition and Diabetes (2023)13:7 N. Ďáskova et al. Table 1. G r o u p characteristics for lean (LH), o b e s e (OB) a n d persons w i t h pre/diabetes (DM). LH DM OB K-W test DMCT LH vs OB LH vs DM OB vs DM General characteristics Sex (F/M) 16/16 26/23 47/19 W e i g h t (kg) 74.8 [23.1] 99.5 [17.4] 87.2 [25.8] "0.05 "0.001 "0.001 "0.01 A g e (years) 30.9 [11.0] 58.3 [13.1] 51.3 [14.2] "0.05 "0.001 "0.001 n.s. BMI (kg/m2 ) 23.0 [4.0] 34.9 [9.1] 30.8 [6.6] "0.05 "0.001 "0.001 "0.05 W H R 0.8 [0.1] 1.0 [0.1] 0.9 [0.1] "0.05 "0.001 "0.001 "0.05 Body composition Fat (kg) 14.2 [4.8] 39.5 [22.3] 32.9[14.7] "0.05 "0.001 "0.001 n.s. F F M (kg) 56.5 [22.5] 61.3[14.8] 51.9[17.3] "0.05 n.s. n.s. "0.05 T B W (kg) 41.4 [16.5] 44.9[10.8] 38.0[12.6] '0.05 n.s. n.s. "0.05 Macronutrient intake Total e n e r g y (kcal/day) 2101[1583] 2017 [879] 1777[555] n.s. N/A N/A N/A Proteins (g/day) 81 [29] 82 [33] 72 [28.5] n.s. N/A N/A N/A Lipids (g/day) 83 [49] 79 [40] 65 [35.5] "0.05 "0.05 n.s. n.s. Carbohydrates (g/day) 232 [98] 207 [96] 197 [73.5] n.s. N/A N/A N/A Dietary fiber (g/day) 18 [19] 16 [9] 15 [7.5] n.s. N/A N/A N/A Glucose metabolism Fasting g l u c o s e (mmol/l) 4.8 [0.3] 5.9 [0.8] 5.3 [0.6] "0.05 "0.001 "0.001 "0.001 2 h O G T T g l u c o s e (mmol/l) 5.7 [1.1] 8.9 [3.1] 6.4 [1.6] "0.05 n.s. "0.001 "0.001 A U C for O G T T g l u c o s e ( m m o l / 1 x 1 2 0 m i n " 1 ) 254 [114] 499 [282] 239 [150] "0.05 n.s. "0.001 "0.001 A U C for O G T T insulin (mlU/l x 120 m i n - 1 ) 3890[2707] 8948[6596] 6453[4122] "0.05 "0.01 "0.001 "0.05 Insulin (mlU/l) 4.0 [2.7] 15.9 [8.6] 9.5 [5.7] "0.05 "0.001 "0.001 "0.001 C-peptide (pmol/l) 233 [97] 769 [357] 5.3 [0.6] "0.05 "0.001 "0.001 "0.01 H b A l c (mmol/mol) 32 [2] 38 [7] 6.4 [1.6] "0.05 "0.001 "0.001 "0.001 M a t s u d a index 10.2 [6.4] 2.0 [1.7] 4.0 [3.4] "0.05 "0.01 "0.001 "0.001 Insulinogenic index 0.8 [0.7] 0.8 [1.0] 1.1 [1.0] "0.05 n.s. n.s. n.s. Oral disposition index 6.7 [4.9] 1.9 [1.2] 4.9 [5.7] "0.001 n.s. "0.001 "0.001 Beta cell index 163 [134] 45 [25] 108 [145] "0.001 n.s. "0.001 "0.001 TyG i n d e x 0.51 [0.67] 1.54[0.59] 1.01 [0.60] "0.05 "0.001 "0.001 "0.01 Lipid metabolism Total cholesterol (mmol/l) 4.30 [1.09] 5.01 [1.23] 5.15 [1.24] "0.05 "0.01 "0.05 n.s. HDL-C (mmol/l) 1.67 [0.47] 1.26 [0.30] 1.39 [0.56] "0.05 "0.05 "0.001 n.s. LDL-C (mmol/l) 2.37 [1.15] 3.05 [1.40] 3.06 [1.16] "0.05 "0.001 "0.05 n.s. Triacylglycerols (mmol/l) 0.69 [0.52] 1.53 [0.93] 1.10 [0.71] "0.05 "0.001 "0.001 "0.05 Inflammatory markers CRP (mg/l) 0.7 [0.9] 3.3 [4.5] 2.3 [4.0] "0.05 "0.001 "0.001 n.s. Stool characteristics p H in feces 7.26 [0.67] 7.04 [0.52] 7.27 [0.50] n.s. N/A N/A N/A dry mass (%) 25.1 [8.9] 24.5 [9.9] 23.0 [6.9] n.s. N/A N/A N/A Data were given as median [71], AUC area under the curve during oral glucose tolerance test, BMI body mass index, CRP C-reactive protein, DMCT Dunn's multiple comparison test, FFM fat-free mass, HDL-C high-density lipoprotein-cholesterol, HbAl glycated hemoglobin, K-W Kruskal-Wallis test, LDL-C low-density lipoprotein-cholesterol, N/A not applicable, ns not significant, TyG index In (fasting triglyceride x fasting glucose)/2; TBW total body water, WHR waist-hip ratio. Insulinogenic index (AINS 0-30/ A G L U 0-30), ISI-M Matsuda-deFronzo index; oral disposition index (IGI*ISI); beta cell index (iAUCi n s u M n /iAUCg |u c o s e )*ISI. *p < 0.05, **p < 0.01, ***p < 0.001. significantly lower in O B a n d D M compared with LH, whereas no differences were f o u n d between the D M a n d O B groups (Fig. S2). W h e n ASVs were aggregated a n d classified at t h e genus level, only the Shannon index remained significantly lower in O B and D M c o m p a r e d with LH (Fig. S3). At t h e p h y l u m level, t h e m i c r o b i o t a c o m p o s i t i o n was d o m i n a t e d by Firmicutes a n d Bacteroidetes, f o l l o w e d by m u c h less a b u n d a n t Actinobacteria, Proteobacteria, a n d Verrucomicrobia. T h e m e d i a n a b u n d a n c e o f all other phyla was less than 0 . 0 1 % . There were n o significant differences in t h e Nutrition and Diabetes (2023)13:7 SPRINGER NATURE N. Ďáskova et al. representation of individual phyla (Table S2). The separation of individual samples at the genus level is visualized in Fig. 1A. M u l t i v a r i a t e statistics revealed significant differences in p-diversity (p< 0.001), a n d pairwise analysis confirmed significant differences between OB vs. LH ( p < 0.001) a n d D M vs. LH (p< 0.001), but not between D M a n d OB. Using univariable analysis, w e identified 37 taxa that had significantly different abundance a m o n g groups; 15 of t h e m met the criteria of "core" microbiota, i.e., an abundance of >0.05% a n d a prevalence of > 7 5 % in at least o n e group (Fig. 1B a n d Table S3), accounting for 4 5 % of all core genera. Thirteen core genera were more abundant in LH c o m p a r e d to the other t w o groups, while E • G r o u p s LH O B • M B Dim1 ( 1 0 . 8 % ) Erysipelotricha U C G - 0 0 3 L a c h n o s p i r a c e a e incertae sedis Lachnospiraceae N D 3 0 0 7 g r o u p * Bifidobacterium Anaerostipes * Fusicatenibacter L a c h n o s p i r a c e a e _ u n a s s i g n e d (*) [Eubacterium] hallii g r o u p * Blautia (*) D o r e a Lachnospiraceae N K 4 A 1 3 6 g r o u p " Faecalibacterium * Christensenellaceae R - 7 g r o u p * Pseudobutyrivibrio (*) Lachnoclostridium (*) 0.5 Lachnospiraceae F C S 0 2 group [Ruminococcus] gauvreauii group Marvinbryantia * Lachnospiraceae U C G - 0 0 8 Family XIII A D 3 0 1 1 g r o u p Prevotella 7 Prevotellaceae Tyzzerella 3 Catenibacterium (*) Tyzzerella 4 Alloprevotella Mitsuokella (*) Megasphaera * Fusobacterium (*) B a c t e r o i d a l e s _ u n a s s i g n e d Ruminococcaceae U C G - 0 0 4 Flavonifractor * Desulfovibrio Succinivibrio Slackia Megamonas L H O B D M Fig. 1 Fecal microbiome composition. A 2D PCA scores plot on genera level after clr transformation. The explained variance of each component is included in the axis labels. The large points represent the centroids of each group. B Abundances of all significant genera (FDR <0.1). Proportional data were used. Each cell then represents the mean in each group for the corresponding genera. Rows were z-scaled. Core genera are defined by the condition abundance >0.05% and prevalence >75% at least in one group. Genera marked by * are confirmed butyrate producers, and genera marked by (*) are potential butyrate producers. Pseudobutyrivibrio a n d Lachnoclostridium were enriched only in DM. Confirmed butyrate producers, i.e., Anaerostipes, Eubacterium halii, Faecalibacterium, Christensenellaceae R-7 group, were more abundant in the core microbiota LH than in the core microbiota O B or D M . Most of the taxa enriched in D M and/or OB belong to the "non-core" taxa. A m o n g them, potentially harmful genera were identified {Fusobacterium, Megasphera, and Desulfovibrio). Significant positive correlations were found between Fusobacterium abundance a n d C-peptide concentration in all groups. The c o m m o n or unique taxa specific to the groups are shown in Fig. S4. The discrimination of the groups as a function of microbiome composition was investigated using a machine learning approach (LASSO regression model). This model, which has an accuracy of 5 1 % and a sensitivity of 6 6 % (LH), 5 0 % (OB), and 4 3 % (DM), does not reliably classify LH, OB, and D M (Fig. S5). When w e grouped OB and DM, the accuracy of the model increased to 7 5 % and the sensitivity to 6 5 % (LH) (Fig. S6). Observational study: fecal metabolome In the fecal metabolome, w e identified 185 different VOCs. Within this subset, 113 VOCs were of very low abundance ("0.1%), 54 VOCs each accounted for 0.1-1% of the total, 12 VOCs accounted for 1-5% of the total, a n d six were very abundant (>5%). The separation of individual samples is visualized in Fig. 2. Multivariable statistics revealed significant differences in B-diversity (p = 0.0017). The pairwise analysis confirmed significant differences between the D M vs. LH groups (p<0.01) a n d OB vs. LH (p < 0.05), but not between D M and OB. Univariable analysis followed by effect size analysis revealed ten VOCs with significantly different abundance between groups (FDR ; • • • • • i • •• • • T V " " " * * •— r * : » V • • — — — • • • • G r o u p s LH O B D M Dim1 ( 2 6 % ) B L H O B D M N o n a n o i c acid • i B e nze ne a ceta Ide hyd e 2-Octanol 0.5 Propyl acetate D e c a n e 0 Tetradecanal t r a n s - O c i m e n e H u m u l e n e -0.5 H u m u l e n e -0.5 Methyl pentanoate • -1 Fig. 2 Fecal metabolome composition. A 2D PCA scores plot on VOCs abundances after clr transformation. Only VOCs meeting condition A U C x p > 0 . 1 % AUCt o tai,p are shown. The explained variance of each component is included in the axis labels. The large points represent the centroids of each group. B Abundances of significant metabolites. Proportional data were used. Each cell then represents the median in each group for the corresponding metabolite. Rows were z-scaled. SPRINGER NATURE Nutrition a n d Diabetes (2023)13:7 N. Ďáskova et al. p < 0.1) (Fig. 2B and Table S4). Nonanoic acid was more abundant, while all other compounds, including SCFA esters, were less abundant in the OB and D M groups compared to LH. Only methyl pentanoate showed an opposite pattern in the D M and OB groups (DM>LH = OB) (Fig. S7). Nonanoic acid correlated positively with the TyG index in all groups. A LASSO model created for the classification of tested subjects into three categories (LH vs OB vs DM) achieved only 5 2 % accuracy and only 4 8 % (LH), 5 4 % (OB), and 5 3 % (DM) sensitivity (Fig. S8). When we combined subjects from OB and D M into one category, classification accuracy increased to 80.5%, but sensitivity remained low at 5 2 % (Fig. S9). Observational study: serum/plasma metabolome To determine the composition of the serum metabolome, we used an untargeted NMR approach and LC-MS analyzes that allows accurate determination of SCFA concentration in plasma. In total, we identified 35 quantified analytes by NMR and nine SCFAs by LC-MS, only acetate/acetic acid was identified by both methods. PERMANOVA analysis suggested the separation of the groups, and subsequent pairwise tests revealed significant differences (p< 0.001) in serum metabolome composition between all compared pairs. The univariable analysis identified 21 metabolites that were significantly different in abundance between groups (Fig. 3B and Table S5). Based o n the univariable analysis, we identified LH, OB, and DM-specific groups of serum metabolites. For most metabolites, the D M and LH groups represented the opposite poles of the abundance spectra, with OB closer to the D M group. All three groups differed in serum concentrations of intermediates of saccharide metabolism (glucose, lactate, and mannose) and two amino acids (AA) (glutamine, alanine). The concentration of seven compounds, including three SCFA (propionic acid, succinic acid, valeric acid), two A A (tyrosine, histidine), and glycerol was comparable at OB and D M , but differed from LH. Six compounds, including two branched-chain amino acid (BCAA) derivatives (2oxoisovalerate, 3-methyl-2-oxovalerate), 2-hydroxybutyrate, acetone, 2-propanol, and formic acid, presented a specific DMassociated signature (Fig. S10). A LASSO -model based on serum metabolome data was able to classify unknown subjects into the categories LH, OB, or D M with an accuracy of 7 4 % and a sensitivity of 9 0 % (LH), 7 2 % (OB), or 6 5 % (DM) (Fig. S11). When we grouped subjects from OB and D M groups together, model accuracy increased to 8 9 % and sensitivity (LH) increased to 8 8 % (Fig. S12). None of the models selected glucose as a key discriminant. Observational study: integrative analysis We further investigated whether a combination of all variables would allow better classification between groups. With this integrated LASSO model, an unknown subject could be assigned to one of the three groups (LH, OB, and DM) with an accuracy of 7 7 % and a sensitivity of 8 8 % (LH), 7 9 % (OB), and 6 6 % (DM), respectively. LASSO coefficients included five variables from the microbiome dataset, one variable from the fecal metabolome dataset, and nine variables from the serum metabolome dataset (Fig. S13). When we constructed the LASSO model only for two groups (LH vs. OB + DM), we were able to classify an unknown subject with 9 1 % accuracy and 8 9 % sensitivity. Ten microbes, five fecal VOCs, and 11 serum metabolites contributed to the discrimination between groups (Fig. S14). Finally, w e looked for a possible complex interaction between different MIME components in individual groups. Figure 4 depicts the positive and negative Spearman correlations among datasets filtered by |p| > 0.5; these correlations unravel differences in interaction networks within each group. In the LH group, we observed a rich network among variables both within and outside the datasets, whereas the complexity in OB and D M was much lower. Groups Dim1 (13.6%) B Saccharide metabolism Amino acids i 0.5 0 - 0 . 5 - 1 LH O B D M BCAA metabolism intermediates NAD+ regeneration Lipid metabolism G l u c o s e M a n n o s e Lactate Pyruvate Glutamine Asparagine Histidine Glycine Alanine Tyrosine 3-Methyl-2-oxovalerate 2-Oxoisovalerate Valeric acid Formate Propionic acid Succinic acid Acetone | 2-Propanol | 2-Hydroxybutyrate Glycerol I Ethanol LH O B D M Fig. 3 Serum metabolome composition. A 2D PCA scores plot. The explained variance of each component is included in the axis labels. The large points represent the centroids of each group. B Abundances of significant metabolites. Each cell then represents the median in each group for the corresponding metabolite. Rows were z-scaled. Prospective study: effect of inulin on omics signature Twenty-seven newly diagnosed D M subjects participated in a threemonth, single-arm, non-controlled intervention study in which they were administered inulin (10g/day) without other antidiabetic medications and/or lifestyle interventions. No clinically significant adverse events occurred, and all subjects completed the study. The inulin intervention was associated with a significant change in microbiota composition (PERMANOVA p< 0.001) and a significant decrease in a-diversity (Fig. 5A, B). At the phylum level, the abundance of Bacteroidetes and Proteobacteria significantly decreased, whereas the proportion of Actinobacteria and Verrucomicrobia significantly increased (Table S6). Univariable analysis revealed 28 taxa with significantly different abundance before and after inulin treatment (Fig. 5C and Table S7). The abundance of 16 bacterial taxa (genera or higher taxonomic units), including confirmed butyrate producers such as Faecalibacterium, Anaerostipes, and Eubacterium halii or bacteria considered beneficial such as Lactobacillus, Bifidobacterium, and Akkermansia, increased after treatment. The abundance of 12 taxa, including Alistipes, Odoribacter, or Bacteroides, decreased. In serum and feces, inulin intake was not associated with a shift in total metabolome composition, but using univariable analysis, we identified several metabolites that were significantly different Nutrition and Diabetes (2023)13:7 SPRINGER NATURE N. Ďáskova et al. A Positive correlations B Negative correlations Fig. 4 Correlation chord diagrams between variables of different datasets. S p e a r m a n c o r r e l a t i o n s w e r e c a l c u l a t e d f o r e a c h g r o u p (LH, O B , D M ) separately. O n l y c o r r e l a t i o n s a m o n g variables f r o m d i f f e r e n t d a t a s e t s (clinical variables, m i c r o b i o m e , s e r u m , a n d fecal m e t a b o l o m e ) a n d c h a r a c t e r i z e d b y |p| > 0.5 are p r e s e n t e d . Positive (A, C, E) a n d n e g a t i v e (B, D, F) c o r r e l a t i o n s are s h o w n separately. T h e c o l o r s o n t h e circuit c o d e i n d i v i d u a l datasets, t h e c o l o r o f t h e e d g e s c o r r e s p o n d s t o o n e o f t h e d a t a s e t s t h a t are l i n k e d b y t h e e d g e . Blue: m i c r o b i o m e ; g r e e n : fecal m e t a b o l o m e ; y e l l o w : clinical variables; violet: s e r u m m e t a b o l o m e . SPRINGER NATURE Nutrition and Diabetes (2023)13:7 N. Ďáskova et al. B Q CD O b s e r v e d s p e c i e s p < 0.001 13 co 1000 CD 1 A B Timepoint C h a o l p < 0.001 A B Timepoint S h a n n o n p = 0.016 1, A B Timepoint InvSimpson p = 0.057 Ii rv A B Timepoint Dim1 (14.6%) *Anaerostipes *[Eubacterium] hallii Lactobacillus Lachnospiraceae FCS020 *Megasphaera *Lachnospiraceae ND3007 Actinomyces Fusicatenibacter (*) Blautia Bifidobacterium i Lachnospiraceae_unassigned *Faecalibacterium (*) Akkermansia Dorea 'Coprococcus 1 *Collinsella *Barnesiella "Butyhcimonas Sutterella Lachnospiraceae NC2004 Bacteroides Lachnospiraceae UCG-008 *Lachnospiraceae UCG-004 (*) Parabacteroides (*) Clostridials (*) Odoribacter Alistipes -1.0 Timepoint -•- A -•- B -0.5 0.0 Cliffs delta 0.5 1.0 Fig. 5 Effect of inulin on fecal microbiome composition. A Alpha diversity calculated on rarefied ASV data, p value represents the result of Wilcoxon test; B 2D PCA scores plot on genera level. The explained variance of each component is included in the axis labels. The large points represent the centroids of each group. C Biomarker bacterial genera. Prior to all calculations, data were clr transformed. Biomarkers were generated from univariable discriminant analysis (FDR >0.1), with effect size estimated by Cliff's delta with a 9 5 % confidence interval. A, time point prior to intervention; B, time point post-intervention. Core genera (bold) are defined by the condition abundance >0.05% and prevalence >75% at least in one group. Genera marked by * are confirmed butyrate producers, and genera marked by (*) are potential butyrate producers. ASV, amplicon sequence variant. Nutrition and Diabetes (2023)13:7 SPRINGER NATURE N. Ďáskova et al. W CD W 03 O CD E C W CD W 03 O D E C CD cn 03 O D E C CD cn 03 o E C 10.0 7.5 5.0 2.5 0.0 4 2 0 10.0 7.5 5.0 2.5 0.0 AUC OGTT glucose I -50 -30 -10 10 30 AUC OGTT insulin -30 -10 10 30 Mcorr (FFM) I 50 -50 -30 -10 10 30 50 70 90 C peptide I I I I I I 50 -50 -30 -10 10 30 50 70 100*(B-A)/A[%] 90 9- 6- 3- 0- 2hr OGTT glucose I -70 -50 -30 -10 10 6- 4- 2- 0Fasting insulinemia -70 -50 -30 -10 10 30 50 70 90 -30 -10 10 6- 4- 2- 0MCR/I (FFM) I 30 • HbA1c I I I 1 30 -70 -50 -30 -10 10 30 50 70 90 100 * (B-A)/A[%] Fig. 6 Effect of inulin supplementation on selected markers of glucose metabolism. Data are expressed as the percentual change baseline to post-intervention. A, time point prior to intervention; B, time point post-intervention. Dashed line, 0 % ; dotted lines, ±10% range. before and after the intervention. In serum, the concentration of butyric acid, propionic acid, and asparagine increased significantly, whereas the concentration of glycerol and 2-propanol decreased after inulin treatment (Fig. S15 and Table S8). In feces, three VOCs were significantly different in abundance (p< 0.05) before and after inulin treatment, including two propionic acid esters (increased) and 1-hexanol (decreased) (Table S9). However, the significance disappeared after multiple comparisons. Prospective study: effect of inulin on glucose metabolism Inulin intake affected markers of glucose tolerance and insulin sensitivity, but the individual response was highly variable; we observed positive, no, or negative changes for each of the variables (Fig. 6 and Table S10). In the entire intervention group, we observed a significant improvement in glucose tolerance (120 min OGTT glucose) and a trend toward a reduction in AUC for OGTT glucose and fasting glycemia. Skeletal muscle insulin sensitivity, measured by glucose clamp and expressed as MCR/I value, increased by more than 1 0 % after the intervention compared with baseline in 14 subjects (from +11.4 to +62.4%), whereas it did not change or decrease in 13 subjects (from +4.8 to -48.7%). A similar distribution was observed for other indices of insulin sensitivity (Mcorr corrected for FFM, A U C OGTT insulin, and fasting insulinemia). Prospective study: predictors of the metabolic effect of inulin Because we replicated previous findings of large inter-individual differences in metabolic responses to inulin, we sought to identify predictors of these differences. To this end, we built linear regression models for all glucose metabolism parameters studied SPRINGER NATURE Nutrition and Diabetes (2023)13:7 N. Ďáskova et al. Table 2. Predictors o f t h e inulin treatment effect o n glucose homeostasis parameters. outcome predictor p val ßx ßy p val ßy R2 A U C O G T T glucose Ruminidostridium - 4 1 . 6 0 0.015 -0.11 0.236 0.393 Lachnospiraceaejncertae sedis 40.37 0.015 - 0 . 1 8 0.044 0.249 Lachnodostridium 35.83 0.033 - 0 . 1 6 0.083 0.317 3-methyl-2-oxovalerate 37.78 0.018 - 0 . 1 8 0.044 0.326 alanine 36.28 0.024 - 0 . 1 9 0.040 0.287 ethanol -51.31 0.001 -0.21 0.008 0.501 2 h O G T T glucose A U C O G T T insulin 1.00 0.027 - 0 . 0 3 0.876 0.286 fasting insulinemia 0.91 0.037 - 0 . 1 0 0.536 0.181 H O M A INS 0.88 0.046 - 0 . 1 2 0.487 0.215 Eubacterium halii group 0.99 0.032 - 0 . 1 7 0.321 0.280 3-methyl-2-oxovalerate 1.08 0.012 - 0 . 0 9 0.585 0.287 3-hydroxyisobutyrate 0.98 0.024 - 0 . 1 4 0.392 0.278 2-oxoisocaproate 0.93 0.033 - 0 . 1 2 0.462 0.229 pyruvate 0.93 0.034 - 0 . 0 9 0.580 0.245 indole 1.33 0.002 -0.11 0.465 0.350 tridecanol 1.17 0.008 - 0 . 1 3 0.426 0.300 c-Dodecalactone 1.13 0.012 - 0 . 1 8 0.276 0.269 methyl h e p t e n o n e 1.05 0.020 - 0 . 1 6 0.360 0.217 2-undecanone 1.05 0.021 - 0 . 1 6 0.341 0.240 methyl butanal 1.02 0.024 -0.11 0.527 0.214 MCR/I (FFM) ISI (Matsuda) 0.01 0.005 - 0 . 6 7 0.008 0.313 A U C O G T T insulin -0.01 0.005 - 0 . 5 3 0.017 0.291 2 hr O G T T insulinemia -0.01 0.024 - 0 . 4 5 0.051 0.251 H O M A INS -0.01 0.027 - 0 . 5 7 0.029 0.208 fasting insulinemia -0.01 0.030 - 0 . 5 6 0.032 0.269 H O M A IR -0.01 0.036 - 0 . 5 5 0.035 0.205 IGI -0.01 0.045 - 0 . 4 0 0.080 0.189 Blautia -0.01 0.027 - 0 . 2 2 0.329 0.222 [Eubarterium] hallii group -0.01 0.030 - 0 . 1 6 0.451 0.180 asparagine 0.01 0.011 -0.31 0.121 0.230 A M c o r r (FFM) ISI (Matsuda) 1.02 0.001 - 0 . 5 3 0.025 0.474 A U C O G T T insulin - 0 . 9 0 0.002 -0.31 0.132 0.335 H O M A INS - 0 . 9 0 0.003 - 0 . 3 8 0.080 0.318 Fasting insulinemia - 0 . 8 8 0.003 - 0 . 3 7 0.090 0.326 H O M A IR - 0 . 8 6 0.006 - 0 . 3 8 0.091 0.243 IGI - 0 . 7 5 0.009 - 0 . 2 0 0.367 0.243 2 h O G T T insulinemia - 0 . 6 6 0.029 - 0 . 2 4 0.293 0.201 Dialister - 0 . 5 8 0.038 -0.01 0.979 0.172 Phascolardobacterium 0.55 0.048 0.08 0.705 0.203 asparagine 0.75 0.011 - 0 . 2 6 0.239 0.210 The data shown in the table are derived from the linear regression model described by the equation Y ( B ) - Y , A ) = p0 + p Y Y , A ) + PxX, A ) + e, where Y ( A ) stands for outcome variable at time A; Y ( B ) stands for outcome variable at time B, B > A; X ( A ) stands for a standardized variable at time A representing in each model any single clinical, metabolome or microbiome variable; px , p y are model coefficients; e stands for random error. Fecal metabolites were filtered by the condition Z A U C X >0.1 % X A U C t o t a i across all samples; bacteria were filtered by the condition median abundance >0.1 % of the total X of bacteria across all samples. HbAlC glycosylated hemoglobin, Mcorr glucose disposal space corrected and adjusted t o fat-free mass, MCR/I metabolic clearance rate of glucose space corrected and adjusted to fat-free mass divided by steady-state insulinemia, OGTT oral glucose tolerance test, R2 proportion of variation in y explained by the predictors obtained using bootstrapping (50 iterations). as o u t c o m e variables, with all clinical or omics variables as predictors; w e omitted variables with significant coefficients that had high leverage (Figs. S16-S19). Despite t h e limitations of our model, it showed several potentially interesting findings (summarized in Table 2). For example, t h e effect o f inulin o n skeletal muscle insulin sensitivity (Mcorr a n d MCR/I) could be predicted from pre-intervention glycemic measures. In contrast, the MIME predictors of the inulin effect were mostly not associated with preintervention o u t c o m e variables. Change in A U C OGTT glucose was negatively associated with an initial abundance of Nutrition and Diabetes (2023)13:7 SPRINGER NATURE N. Ďáskova et al. Ruminiclostridium, whereas increases in Mcorr a n d MCR/I were associated with higher initial serum asparagine (both parameters) and lower Dialister (Mcorr) or Blautia and Eubacterium halii (MCR/I). Initial serum concentrations of BCAA derivatives were positively associated with increases in A U C and 2-hour OGTT glucose. All results are summarized in Table 2. DISCUSSION Our main findings are: (i) obesity is the dominant factor determining the MIME signature, whereas glycemic status has a lesser additional influence; (ii) the metabolic response to inulin supplementation in individuals with newly diagnosed prediabetes/diabetes is highly variable but can be predicted, at least in part, from baseline clinical characteristics and MIME signatures. Indeed, more insulin-resistant individuals with poorer glycemic indices and elevated circulating BCAA derivatives and fecal indole and p-cresol are less likely to respond to inulin supplementation. Observational study: gut microbiome and metabolome Obesity is a prominent risk factor for the development of type 2 diabetes. Numerous studies have identified groups of bacterial taxa that are enriched or depleted in obesity and type 2 diabetes, and despite considerable heterogeneity in the results, some common observations have been noted. First, type 2 diabetes is associated with the depletion of potentially beneficial bacteria rather than the presence of some dominant potentially harmful bacteria. Second, the abundance of butyrate producers and the functional potential for butyrate production is reduced in type 2 diabetes [10, 20, 35]. Third, the diversity of the microbiota is lower in diseased individuals compared with healthy controls [6, 36]. Some of our results are consistent with the above, whereas others are contradictory. In contrast to the results of Wu [10], the change in the composition of the gut microbiota in our study was not related to glycemic status but mainly to obesity. The dominant butyrate producers, such as Faecalibacterium, Anaerostipes, Eubacterium halii, or Blautia were significantly less abundant in the microbiota of D M and OB, but w e did not detect lower SCFA concentrations in either feces or serum. In contrast, MCFA, nonanoic and decanoic acids were elevated in OB and DM. MCFA can originate from dietary sources [35], but also from microbial or yeast fermentation [37]. SCFA and MCFA have different immunomodulatory properties; whereas SCFA attenuate inflammation, MCFA have the opposite effect [35, 38, 39]. In addition, MCFA may enhance intestinal permeation because of their physicochemical properties as anionic surfactants [40]. Based on these findings, we might suggest that it is not the lower level of SCFA but the increased level of MCFA in the lumen that contributes to the complications associated with obesity, such as impaired intestinal barrier function or chronic low-grade inflammation. Observational study: serum metabolome The serum metabolome signature of obesity and diabetes overlapped greatly in the study. Compared to lean subjects, both the OB a n d D M signatures follow the same direction a n d differ only in magnitude. The "adiposity signature," which is similar in both OB and D M , includes SCFA (succinic and propionic acid increased, while valeric acid decreased), aromatic A A tyrosine (increased), and two other A A (histidine a n d asparagine, decreased). The concentration shift of five other metabolites, i.e., intermediates of saccharide metabolism (glucose, lactate, and mannose, increased) a n d A A glutamine a n d alanine, follows the concordant direction to LH, but there is a significant difference among all three groups. Six metabolites are specific for DM. This signature consists of three BCAA derivatives, formic acid, 2hydroxybutyrate, acetone, and 2-propanol. Our findings are consistent with previously published observations [41, 42]. Some signature metabolites could be attributed to altered saccharide metabolism in obesity and diabetes, such as glucose, mannose, and lactate. 2-propanol, acetone, and 2-hydroxybutyrate might be related to NADH/NAD+ redox imbalance, which has been proposed as o n e of the features of T2D [43]. Some other signature metabolites, i.e., SCFA and BCAA, are located at the interface between the host and microbiota. SCFA in serum have not previously been described as components of an obesity-related serum signature, probably because of the analytical difficulties associated with their determination in serum. They are exclusively microbial products, some of which (circulating butyric acid a n d propionic acid) have been associated with beneficial effects [44]. Elevated circulating BCAAs have been associated with insulin-resistance conditions such as obesity, diabetes [45], a n d even cancer [46]. For mammals, BCAAs are essential and must be supplied from external sources. Recent research has deciphered the importance of the gut microbiota in modulating the availability of many necessary compounds, including BCAA, to the host [47]. Inulin intervention and the effects on microbiota composition and performance Three months of regular consumption of 10 g inulin/day was associated with a significant shift in the composition of the microbiota, characterized by a marked increase in potentially beneficial bacteria, many of which are capable of butyrate production [48]. At the same time, several bacterial taxa were depleted, such as those associated with the fermentation of proteins [49, 50]. This observation is largely consistent with previously published reports [51, 52]. We did not detect a significant shift in the composition of the fecal metabolome, although there was a non-significant trend toward an increase in SCFA esters content. Participants were asked not to change their dietary habits, and the only difference before and after the intervention was the amount of inulin consumed. This change could primarily increase the production of SCFA, but these compounds are readily utilized by other microbes or colonocytes at the site of their production, and only about 5 % of SCFA are excreted in the feces [53]. A small fraction of SCFA from the intestine may enter the bloodstream, and indeed w e observed a significant increase in serum butyric and propionic acid concentrations at the e n d of the intervention. Muller et al. [54] have previously reported that it is not fecal but circulating SCFA, particularly butyrate, that can provide a link between the gut microbiota and whole-body insulin sensitivity. SCFA are ligands of the G protein-coupled receptors GPR41 a n d GPR43, which are expressed in many tissues, including adipose tissue a n d skeletal muscle [55, 56]. Animal studies have shown that oral administration of SCFA or intravenous infusion improves insulin sensitivity [54]. Predicting the individual effect of an inulin intervention The increasing understanding of the role of the microbiome in host physiology opened new avenues for research focused o n the possibility of predicting the outcome of a given intervention based on the individual MIME setting. Clinically relevant results have been obtained in cancer research, e.g., the success of therapy with Anti-programmed Cell Death Protein-1 (PD—1) has been shown to depend significantly on the baseline composition of the patient's gut microbiota [57-60]. MIME has also been successfully used to predict the response of IBD patients to a low FODMAP diet [61] or anti-TNF treatment [62], the efficacy of synbiotic treatment of gastrointestinal disease in children [63], or the prediction of the clinical outcome of bariatric surgery [64]. The gut microbiota may serve as a biomarker for selecting the most effective drugs for the treatment of rheumatoid arthritis [65], and gut bacterial signatures have even been described to characterize the diagnosis and predict treatment outcomes in bipolar depression [66]. SPRINGER NATURE Nutrition a n d Diabetes (2023)13:7 N. Ďáskova et al. Inulin-type dietary fiber is thought to alleviate several features of metabolic syndrome; however, results from human studies are inconsistent. A recent systematic review [67], which included 33 RCTs, showed that inulin intake (average 11 g/day) significantly reduced blood glucose, total cholesterol, and TAG in individuals with prediabetes and diabetes. However, a c o m m o n feature of all included studies was the wide heterogeneity of individual responses to treatment, making clear dietary recommendations difficult. Therefore, w e sought to identify factors that would allow a personalized assessment of the efficacy of inulin treatment. We found that patients with a profile suggestive of less impaired glucose homeostasis were likely to improve metabolically. In addition, w e identified several other potential predictors that were not dependent on pre-intervention glycemic indices, including lower serum BCAA derivatives (3-methy-2-oxovalerate, 2-oxoisocaproate), serum 3-hydroxyisobutyrate (product of NADH oxidation), fecal indole, and/or various bacteria {Ruminiclostridium, Lachnoclostridium, Eubacterium halii, etc.), which could allow a more accurate prediction of inulin intervention outcomes. In the prediabetes phase, patients are often advised to change their lifestyle a n d diet. Despite initial adherence to advice, outcomes may be highly variable, and patients w h o have failed despite their best efforts may be demotivated to adhere to further recommendations. The tool of predicting the individual appropriateness of a particular intervention, in this case, the administration of inulin, would help personalize treatment so that it has a higher chance of success in potential responders and does not expose potential non-responders to repeated failures. Strengths and limitations of the study There are several strengths of the study. First, the D M group included only participants with newly diagnosed type 2 diabetes prior and/or concomitant treatment, thus excluding confounding effects of antidiabetic drugs on the effects of inulin. Second, we did not rely solely on the measurement of fecal SCFA as the only indicator of SCFA production in the colon, but used a highly sensitive LC-MS method that allows its quantification in serum. Third, w e evaluated the complex effects of the inulin intervention using a multi-omics approach. Nevertheless, the study is limited by several factors. First, w e were able to include only a limited number of subjects, a n d the results were not validated in an independent cohort. For this, the results were internally validated by permutation tests. Second, the lean healthy subjects differed from the OB or D M groups by age, because obesity and associated comorbidities are more c o m m o n in older populations. A g e is one of the external factors affecting microbiota composition, but this is especially true for very young children or the elderly (over 70 years of age). In adolescence a n d adulthood, the composition of the microbiome is remarkably stable in terms of diversity indices, PCA metrics, or representation of selected taxa [68-70]. Therefore, we believe that the age difference in our population did not result in a significant bias. Third, w e did not control dietary intake during the prospective intervention study with inulin because w e did not want to further burden participants a n d increase the risk of dropping out of the study, but all participants were explicitly asked to maintain their usual dietary habits. A n indirect measure of adherence to the habitual diet may be the BMI of participants, which did not change significantly during the intervention period. Finally, the prospective study design was a single-arm, noncontrolled intervention study, so the causality of the effect of inulin on metabolic outcomes cannot be inferred. The small number of participants in the prospective study did not allow us to build more complex models to account for possible synergies among predictors. Because the study aimed to explore predictors, and w e found several novel biomarkers that predict response to inulin treatment, these will need to be validated on a larger scale in future studies. In summary, w e showed that the gut microbiota and metabolome profiles in OB and D M differed from those of lean healthy individuals, whereas the differences between OB and D M were less pronounced. We identified several omics-derived biomarkers that may play a central role in the development of obesity-associated metabolic changes. In patients with newly diagnosed pre/diabetes, w e observed substantial inter-individual variability in the effects of inulin on glucose homeostasis and identified several predictors of treatment response. If replicated in further studies with other populations, the identified predictors could facilitate the estimation of inulin intervention outcomes, paving the way for the concept of personalized dietary management of early diabetes. DATA AVAILABILITY T h e s e q u e n c i n g data w e r e available at t h e S R A d a t a b a s e u n d e r t h e accession n u m b e r PRJNA823864. T h e o t h e r datasets g e n e r a t e d a n d a n a l y z e d d u r i n g t h e current study are available f r o m t h e c o r r e s p o n d i n g a u t h o r o n request. REFERENCES 1. C h e n g HL, M e d l o w S, S t e i n b e c k K. T h e health c o n s e q u e n c e s o f obesity in y o u n g a d u l t h o o d . Curr O b e s Rep. 2016;5:30-7. 2. T u r n b a u g h PJ, Ley RE, M a h o w a l d M A , M a g r i n i V, Mardis ER, G o r d o n Jl. A n obesityassociated g u t m i c r o b i o m e w i t h increased capacity f o r energy harvest. Nature. 2006;444:1027-31. 3. Cornejo-Pareja I, M u n o z - G a r a c h A , C l e m e n t e - P o s t i g o M , T i n a h o n e s FJ. Importance o f g u t m i c r o b i o t a in obesity. Eur J Clin Nutr. 2019;72:26-37. 4. L i m YY, L e e YS, O o i D S Q . E n g i n e e r i n g t h e g u t m i c r o b i o m e f o r t r e a t m e n t o f obesity: a review o f current u n d e r s t a n d i n g a n d progress. B i o t e c h n o l J . 2020;15:e2000013. 5. Q i n J , Li Y, Cai Z, Li S, Z h u J , Z h a n g F, et al. A m e t a g e n o m e - w i d e association study of g u t m i c r o b i o t a in t y p e 2 diabetes. Nature. 2012;490:55-60. 6. Nuli R, C a i J , Kadeer A , Z h a n g Y, M o h e m a i t i P. Integrative analysis t o w a r d different g l u c o s e tolerance-related g u t m i c r o b i o t a a n d diet. Front E n d o c r i n o l . 2019;10:295. 7. Z h o u W, Sailani MR, C o n t r e p o i s K, Z h o u Y, A h a d i S, L e o p o l d SR, et al. L o n g i t u d i n a l multi-omics o f host-microbe d y n a m i c s in prediabetes. Nature. 2019;569:663-71. 8. Forslund K, H i l d e b r a n d F, Nielsen T, Falony G, Le Chatelier E, S u n a g a w a S, et al. D i s e n t a n g l i n g t y p e 2 diabetes a n d m e t f o r m i n t r e a t m e n t signatures in t h e h u m a n gut m i c r o b i o t a . Nature. 2015;528:262-6. 9. Allin K H , Tremaroli V, Caesar R, J e n s e n B A H , D a m g a a r d M T F , Bahl M l , et a l . A b e r r a n t intestinal m i c r o b i o t a in individuals with prediabetes. D i a b e t o l o g i a . 2018;61:810-20. 10. W u H, Tremaroli V, S c h m i d t C, L u n d q v i s t A, O l s s o n L M , Kramer M , et al. T h e g u t m i c r o b i o t a in prediabetes a n d diabetes: a p o p u l a t i o n - b a s e d cross-sectional study. Cell M e t a b . 2020;32:379-90.e3. 11. Tilg H, M o s c h e n AR. M i c r o b i o t a a n d diabetes: a n e v o l v i n g relationship. G u t . 2014;63:1513-21. 12. Z h a n g X, S h e n D, F a n g Z, Jie Z, Q i u X, Z h a n g C, et a l . H u m a n g u t m i c r o b i o t a c h a n g e s reveal t h e p r o g r e s s i o n o f g l u c o s e i n t o l e r a n c e . P L o S O N E . 2 0 1 3 ; 8 : e 7 1 1 0 8 . 13. B h u t e SS, Suryavanshi M V , Joshi S M , Yajnik CS, S h o u c h e YS, G h a s k a d b i SS. G u t microbial diversity assessment o f Indian type-2-diabetics reveals alterations in Eubacteria, A r c h a e a , a n d Eukaryotes. Front M i c r o b i o l . 2017;8:214. 14. Z h a o L, L o u H, P e n g Y, C h e n S, Z h a n g Y, Li X. C o m p r e h e n s i v e relationships b e t w e e n g u t m i c r o b i o m e a n d faecal m e t a b o l o m e in individuals with t y p e 2 diabetes a n d its c o m p l i c a t i o n s . Endocrine. 2019;66:526-37. 15. Z h o n g H, Ren H, Lu Y, Fang C, H o u G, Y a n g Z, et al. Distinct g u t m e t a g e n o m i c s a n d m e t a p r o t e o m i c s signatures in prediabetics a n d treatment-naive t y p e 2 diabetics. E B i o M e d i c i n e . 2019;47:373-83. 16. W a n g L, Y u X, X u X, M i n g J , W a n g Z, G a o B, et al. T h e fecal m i c r o b i o t a is already altered in n o r m o g l y c e m i c individuals w h o g o o n t o have t y p e 2 diabetes. Front Cell Infect M i c r o b i o l . 2021;11:598672. 17. L e t c h u m a n a n G, A b d u l l a h N, Marlini M , B a h a r o m N, Lawley B, O m a r MR, et al. G u t m i c r o b i o t a c o m p o s i t i o n in prediabetes a n d newly d i a g n o s e d t y p e 2 diabetes: a systematic review o f observational studies. Front Cell Infect M i c r o b i o l . 2022;12:943427. 18. Gaike A H , Paul D, Bhute S, Dhotre DP, P a n d e P, Upadhyaya S, et al. T h e gut microbial diversity o f newly diagnosed diabetics b u t n o t o f prediabetics is significantly different f r o m that o f healthy nondiabetics. mSystems. 2020;5:e00578-19. Nutrition a n d Diabetes (2023)13:7 SPRINGER NATURE N. Ďáskova et al. 19. Li L, Li C, Lv M, H u Q, G u o L, X i o n g D. Correlation b e t w e e n alterations o f gut m i c r o b i o t a a n d miR-122-5p expression in patients with t y p e 2 diabetes mellitus. A n n Transl M e d . 2020;8:1481. 20. Karlsson FH, Tremaroli V, N o o k a e w I, B e r g s t r o m G, Behre CJ, Fagerberg B, et al. Gut m e t a g e n o m e in European w o m e n with n o r m a l , i m p a i r e d a n d diabetic g l u cose control. Nature. 2013;498:99-103. 21. Diener C, Reyes-Escogido ML, Jimenez-Ceja L M , Matus M, G o m e z - N a v a r r o C M , C h u N D , et al. Progressive shifts in t h e g u t m i c r o b i o m e reflect prediabetes a n d diabetes d e v e l o p m e n t in a treatment-naive M e x i c a n cohort. Front E n d o c r i n o l . 2020;11:602326. 22. G h a e m i F, Fateh A, S e p a h y A A , Z a n g e n e h M, G h a n e i M , Siadat SD. Intestinal m i c r o b i o t a c o m p o s i t i o n in Iranian diabetic, pre-diabetic a n d healthy individuals. J Diabetes M e t a b Disord. 2020;19:1199-203. 23. C h e n PC, Chien YW, Y a n g SC. The alteration o f g u t m i c r o b i o t a in newly d i a g n o s e d type 2 diabetic patients. Nutrition. 2019;63-64:51-6. 24. P r o c h a z k o v a M, Budinska E, K u z m a M, Pelantova H, Hradecky J , H e c z k o v a M , et al. V e g a n diet is associated with favorable effects on t h e m e t a b o l i c p e r f o r m a n c e of intestinal m i c r o b i o t a : a cross-sectional multi-omics study. Front Nutr. 2021;8:783302. 25. C o l a n t o n i o A G , W e r n e r SL, B r o w n M. The effects o f prebiotics a n d substances with prebiotic properties on m e t a b o l i c a n d i n f l a m m a t o r y biomarkers in individuals with t y p e 2 diabetes mellitus: a systematic review. J A c a d Nutr Diet. 2020;120:587-607.e2. 26. Davis L M , Martinez I, Walter J , G o i n C, Hutkins RW. B a r c o d e d p y r o s e q u e n c i n g reveals that c o n s u m p t i o n o f galactooligosaccharides results in a highly specific bifidogenic response in h u m a n s . PLoS ONE. 201 l;6:e25200. 27. Z h a o L, Z h a n g F, D i n g X, W u G, L a m YY, W a n g X, et al. G u t bacteria selectively p r o m o t e d by dietary fibers alleviate t y p e 2 diabetes. Science. 2018;359:1151-6. 28. W a r e h a m NJ. Personalised prevention of t y p e 2 diabetes. D i a b e t o l o g i a . 2022;65:1796-1803. 29. Le DS, Brookshire T, Krakoff J , Bunt JC. Repeatability a n d reproducibility of the hyperinsulinemic-euglycemic c l a m p a n d the tracer dilution t e c h n i q u e in a c o n trolled inpatient setting. M e t a b o l i s m . 2009;58:304-10. 30. D e F r o n z o RA, T o b i n JD, A n d r e s R. G l u c o s e c l a m p t e c h n i q u e : a m e t h o d for quantifying insulin secretion a n d resistance. A m J Physiol. 1979;237:E214-23. 31. Callahan BJ, M c M u r d i e PJ, Rosen MJ, Han A W , J o h n s o n AJ, H o l m e s SP. D A D A 2 : high-resolution s a m p l e inference f r o m lllumina a m p l i c o n data. Nat M e t h o d s . 2016;13:581-3. 32. Han J, Lin K, Sequeira C, Borchers C H . A n isotope-labeled c h e m i c a l derivatization m e t h o d for t h e quantitation o f short-chain fatty acids in h u m a n feces by liquid c h r o m a t o g r a p h y - t a n d e m mass spectrometry. A n a l C h i m Acta. 2015;854:86-94. 33. Dieterle F, Ross A, Schlotterbeck G, S e n n H. Probabilistic q u o t i e n t n o r m a l i z a t i o n as robust m e t h o d to a c c o u n t for dilution o f c o m p l e x biological mixtures. A p p l i c a t i o n in 1H N M R m e t a b o n o m i c s . A n a l C h e m . 2006;78:4281-90. 34. R C o r e T e a m . R: a l a n g u a g e a n d e n v i r o n m e n t for statistical c o m p u t i n g . Vienna, Austria: R F o u n d a t i o n for Statistical C o m p u t i n g . 2017. 35. S a m Q H , Ling H, Y e w WS, Tan Z, Ravikumar S, C h a n g M W , et al. The divergent i m m u n o m o d u l a t o r y effects o f short chain fatty acids a n d m e d i u m chain fatty acids. Int J M o l Sei. 2021;22:6453. 36. Le Chatelier E, Nielsen T, Qin J, Prifti E, H i l d e b r a n d F, Falony G, et al. Richness of h u m a n g u t m i c r o b i o m e correlates w i t h m e t a b o l i c markers. Nature. 2013;500:541-6. 37. Nissen L, Samaei SP, Babini E, Gianotti A. G l u t e n free s o u r d o u g h b r e a d e n r i c h e d with cricket flour for protein fortification: A n t i o x i d a n t i m p r o v e m e n t a n d Volatil o m e characterization. F o o d C h e m . 2020;333:127410. 38. L u s c o m b e VB, Lucy D, Bataille CJR, Russell AJ, Greaves DR. 20 years an o r p h a n : is G P R 8 4 a plausible m e d i u m - c h a i n fatty acid-sensing receptor? D N A Cell Biol. 2020;39:1926-37. 39. Saresella M, M a r v e n t a n o I, Barone M, La Rosa F, Piancone F, M e n d o z z i L, et al. Alterations in circulating fatty acid are associated with g u t m i c r o b i o t a dysbiosis a n d i n f l a m m a t i o n in m u l t i p l e sclerosis. Front I m m u n o l . 2020;11:1390. 40. Brayden DJ, Mäher S, Bahar B, Walsh E. S o d i u m caprate-induced increases in intestinal permeability a n d epithelial d a m a g e are p r e v e n t e d by m i s o p r o s t o l . Eur J P h a r m B i o p h a r m . 2015;94:194-206. 41. Halama A, S u l e i m a n N N , Kulinski M , Bettahi I, Hassoun S, A l k a s e m M, et al. The m e t a b o l i c footprint o f c o m p r o m i s e d insulin sensitivity u n d e r fasting a n d hyperinsulinemic-euglycemic c l a m p c o n d i t i o n s in an A r a b p o p u l a t i o n . Sei Rep. 2020;10:17164. 42. Gall WE, B e e b e K, L a w t o n KA, A d a m KP, Mitchell M W , Nakhle PJ, et al. alphahydroxybutyrate is an early b i o m a r k e r o f insulin resistance a n d g l u c o s e intolera n c e in a n o n d i a b e t i c p o p u l a t i o n . PLoS ONE. 2 0 1 0 ; 5 : e l 0 8 8 3 . 43. S o n g J, Y a n g X, Yan LJ. Role o f p s e u d o h y p o x i a in t h e p a t h o g e n e s i s o f t y p e 2 diabetes. H y p o x i a . 2019;7:33-40. 44. K u m a r J , Rani K, Datt C. M o l e c u l a r link b e t w e e n dietary fibre, gut m i c r o b i o t a a n d health. M o l Biol Rep. 2020;47:6229-37. 45. N e w g a r d CB. Interplay b e t w e e n lipids a n d branched-chain a m i n o acids in d e v e l o p m e n t of insulin resistance. Cell M e t a b . 2012;15:606-14. 46. Rossmeislova L, G o j d a J , S m o l k o v a K. Pancreatic cancer: branched-chain a m i n o acids as putative key m e t a b o l i c regulators? Cancer Metastasis Rev. 2021;40:1115-39. 47. Gojda J, C a h o v a M. G u t m i c r o b i o t a as t h e link b e t w e e n elevated B C A A s e r u m levels a n d insulin resistance. Biomolecules. 2021;11:1414. 48. Le Bastard Q, C h a p e l e t G, J a v a u d i n F, Lepelletier D, Batard E, Montassier E. The effects of inulin o n g u t microbial c o m p o s i t i o n : a systematic review of e v i d e n c e f r o m h u m a n studies. Eur J Clin M i c r o b i o l Infect Dis. 2020;39:403-13. 49. Diether NE, Willing BP. Microbial f e r m e n t a t i o n o f dietary p r o t e i n : an i m p o r t a n t factor in diet(-)microbe(-)host interaction. M i c r o o r g a n i s m s . 2019;7:19. 50. Yao CK, M u i r JG, G i b s o n PR. Review article: insights into c o l o n i c protein ferm e n t a t i o n , its m o d u l a t i o n a n d potential health implications. A l i m e n t P h a r m Ther. 2016;43:181-96. 51. V a n d e p u t t e D, Tito RY, V a n l e e u w e n R, Falony G, Raes J. Practical considerations for large-scale gut m i c r o b i o m e studies. F E M S M i c r o b i o l Rev. 2017;41:S154-S67. 52. Healey G, M u r p h y R, Butts C, B r o u g h L, W h e l a n K, C o a d J . Habitual dietary fibre intake influences g u t m i c r o b i o t a response t o an inulin-type fructan prebiotic: a r a n d o m i s e d , d o u b l e - b l i n d , placebo-controlled, cross-over, h u m a n intervention study. Br J Nutr. 2018;119:176-89. 53. R e c h k e m m e r G, R o n n a u K, v o n Engelhardt W. F e r m e n t a t i o n o f polysaccharides a n d a b s o r p t i o n o f short chain fatty acids in t h e m a m m a l i a n h i n d g u t . C o m p B i o c h e m Physiol A C o m p Physiol. 1988;90:563-8. 54. Muller M , H e r n a n d e z M A G , G o o s s e n s G H , Reijnders D, Holst JJ, J o c k e n JWE, et al. Circulating but not faecal short-chain fatty acids are related t o insulin sensitivity, lipolysis a n d GLP-1 concentrations in h u m a n s . Sei Rep. 2019;9:12515. 55. T a n g C, A h m e d K, Gille A, Lu S, G r o n e HJ, Tunaru S, et al. Loss of F F A 2 a n d FFA3 increases insulin secretion a n d i m p r o v e s g l u c o s e tolerance in t y p e 2 diabetes. Nat M e d . 2015;21:173-7. 56. Koh A, De V a d d e r F, Kovatcheva-Datchary P, B a c k h e d F. F r o m dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165:1332-45. 57. M a o J, W a n g D, L o n g J, Y a n g X, Lin J, S o n g Y, et al. Gut m i c r o b i o m e is associated with the clinical response t o anti-PD-1 based i m m u n o t h e r a p y in hepatobiliary cancers. J I m m u n o t h e r Cancer. 2021;9:e003334. 58. F a h r m a n n JF, Saini NY, Chia-Chi C, Irajizad E, Strati P, Nair R, et al. A polyaminecentric, b l o o d - b a s e d metabolite panel predictive o f p o o r response t o CAR-T cell therapy in large B cell l y m p h o m a . Cell Rep. M e d . 2022;3:100720. 59. Sannicolo S, Giaj Levra M , Le Gouellec A, A s p o r d C, Boccard J, C h a p e r o t L, et al. Identification o f a predictive m e t a b o l i c signature of response t o i m m u n e c h e c k p o i n t inhibitors in non-small cell lung cancer: METABO-ICI clinical study protocol. Respir M e d Res. 2021 ;80:100845. 60. M c C u l l o c h JA, Davar D, Rodrigues RR, Badger JH, Fang JR, C o l e A M , et al. Intestinal microbiota signatures o f clinical response a n d immune-related adverse events in m e l a n o m a patients treated with anti-PD-1. Nat M e d . 2022;28:545-56. 61. Vervier K, M o s s S, K u m a r N, A d o u m A, Barne M, B r o w n e H, et al. T w o microbiota subtypes identified in irritable b o w e l s y n d r o m e with distinct responses to the low F O D M A P diet. Gut. 2022;71:1821-30. 62. Busquets D, Oliver L, A m o e d o J , Ramio-Pujol S, M a l a g o n M, Serrano M, et al. RAID prediction: pilot study o f fecal microbial signature with capacity to predict response to anti-TNF treatment. Inflamm B o w e l Dis. 2021;27:S63-S6. 63. Tierney BT, Versalovic J, Fasano A, Petrosino JF, C h u m p i t a z i BP, Mayer EA, et al. Functional response to a microbial synbiotic in the gastrointestinal system of children: a r a n d o m i z e d clinical trial. Pediatr Res. 2022. https://doi.org/10.1038/ S41390-022-02289-0. 64. Vaz M, Pereira SS, M o n t e i r o M P . M e t a b o l o m i c signatures after bariatric surgery - a systematic review. Rev E n d o c r M e t a b Disord. 2022;23:503-19. 65. Wei M, C h u C Q . Prediction o f t r e a t m e n t response: p e r s o n a l i z e d m e d i c i n e in the m a n a g e m e n t o f r h e u m a t o i d arthritis. Best Pr Res Clin R h e u m a t o l . 2022;36:101741. 66. Lai J , Li A, Jiang J , Yuan X, Z h a n g P, Xi C, et al. M e t a g e n o m i c analysis reveals gut bacterial signatures for diagnosis a n d t r e a t m e n t o u t c o m e prediction in bipolar depression. Psychiatry Res. 2022;307:114326. 67. Li L, Li P, X u L. Assessing the effects o f inulin-type fructan intake on b o d y weight, b l o o d glucose, a n d lipid profile: a systematic review a n d meta-analysis o f rand o m i z e d c o n t r o l l e d trials. F o o d Sei Nutr. 2021;9:4598-616. 68. O d a m a k i T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, et al. Agerelated changes in g u t m i c r o b i o t a c o m p o s i t i o n f r o m n e w b o r n t o centenarian: a cross-sectional study. B M C M i c r o b i o l . 2016;16:90. 69. R o d r i g u e z J M , M u r p h y K, Stanton C, Ross RP, Kober O l , J u g e N, et al. The c o m position o f the g u t m i c r o b i o t a t h r o u g h o u t life, w i t h an e m p h a s i s on early life. M i c r o Ecol Health Dis. 2015;26:26050. 70. Salazar N, G o n z a l e z S, N o g a c k a A M , Rios-Covian D, A r b o l e y a S, G u e i m o n d e M, et al. M i c r o b i o m e : effects o f a g e i n g a n d diet. Curr Issues M o l Biol. 2020;36:33-62. SPRINGER NATURE Nutrition and Diabetes (2023)13:7 N. Ďáskova et al. 71. Feins EN, Ireland C, G a u v r e a u K, C h a v e z M , Callahan R, Jenkins KJ, et al. P u l m o n a r y vein stenosis: a n a t o m i c considerations, surgical m a n a g e m e n t , a n d o u t c o m e s . J Thorac Cardiovasc Surg. 2022;163:2198-207.e3. ACKNOWLEDGEMENTS This study w a s s u p p o r t e d by t h e Ministry o f Health o f t h e C z e c h Republic, grant NV- 18-01-00040 a n d b y t h e project National Institute for Research o f M e t a b o l i c a n d Cardiovascular Diseases (Program EXCELES, Project N o . L X 2 2 N P O 5 1 0 4 ) — F u n d e d b y t h e E u r o p e a n U n i o n — N e x t G e n e r a t i o n EU. Jan G o j d a is s u p p o r t e d by EFSD m e n t o r s h i p p r o g r a m s u p p o r t e d by A s t r a Z e n e c a . 13 ADDITIONAL INFORMATION Supplementary information T h e o n l i n e version contains s u p p l e m e n t a r y material available at https://doi.org/10.1038/s41387-023-00235-5. Correspondence a n d requests for materials s h o u l d b e a d d r e s s e d t o M . Cahovä. Reprints and permission information is available at http://www.nature.com/ reprints Publisher's note Springer Nature remains neutral with regard t o jurisdictional claims in p u b l i s h e d m a p s a n d institutional affiliations. AUTHOR CONTRIBUTIONS N D , TK, M C , a n d JG w e r e i n v o l v e d in t h e study c o n c e p t a n d d e s i g n . N D , M P , M K , HP, JH, M H e c , M B , PV, PS, M H e n , A O , JP, a n d RL participated in t h e acquisition o f data. N D , IM, a n d M K w e r e i n v o l v e d in statistical analysis. N D , IM, M K , HP, RL, TK, M K , M C , a n d JG participated in interpretation o f data. N D , M C , T K a n d JG drafted t h e manuscript. All authors participated in t h e critical revision o f t h e m a n u s c r i p t a n d a p p r o v e d t h e final version o f t h e manuscript. T h e senior a n d c o r r e s p o n d i n g authors (JG, TK, a n d M C ) take full responsibility f o r t h e w o r k and/or t h e c o n d u c t o f t h e study, they h a d access t o t h e data a n d c o n t r o l l e d t h e decision t o publish. CONFLICT OF INTEREST T h e authors declare n o c o m p e t i n g interests. O p e n Access This article is licensed u n d e r a Creative C o m m o n s Attribution 4.0 International License, w h i c h permits use, sharing, a d a p t a t i o n , distribution a n d r e p r o d u c t i o n in a n y m e d i u m or format, as long as y o u give a p p r o p r i a t e credit t o t h e original author(s) a n d t h e source, p r o v i d e a link t o t h e Creative C o m m o n s license, a n d indicate if c h a n g e s w e r e m a d e . T h e images or other t h i r d party material in this article are i n c l u d e d in t h e article's Creative C o m m o n s license, unless i n d i c a t e d o t h e r w i s e in a credit line t o t h e material. If material is n o t i n c l u d e d in t h e article's Creative C o m m o n s license a n d your i n t e n d e d use is n o t p e r m i t t e d by statutory regulation or exceeds t h e p e r m i t t e d use, y o u will n e e d t o o b t a i n p e r m i s s i o n directly f r o m t h e c o p y r i g h t holder. T o v i e w a c o p y o f this license, visit http:// c r e a t i v e c o m m o n s . o r g / l i c e n ses/by/4.0/. © T h e Author(s) 2 0 2 3 Nutrition and Diabetes (2023)13:7 SPRINGER NATURE