Article Text
Abstract
Background The microbiota is emerging as a key factor in the predisposition to insulin resistance and obesity.
Objective To understand the interplay among gut microbiota and insulin sensitivity in multiple tissues.
Design Integrative multiomics and multitissue approach across six studies, combining euglycaemic clamp measurements (used in four of the six studies) with other measurements of glucose metabolism and insulin resistance (glycated haemoglobin (HbA1c) and fasting glucose).
Results Several genera and species from the Proteobacteria phylum were consistently negatively associated with insulin sensitivity in four studies (ADIPOINST, n=15; IRONMET, n=121, FLORINASH, n=67 and FLOROMIDIA, n=24). Transcriptomic analysis of the jejunum, ileum and colon revealed T cell-related signatures positively linked to insulin sensitivity. Proteobacteria in the ileum and colon were positively associated with HbA1c but negatively with the number of T cells. Jejunal deoxycholic acid was negatively associated with insulin sensitivity. Transcriptomics of subcutaneous adipose tissue (ADIPOMIT, n=740) and visceral adipose tissue (VAT) (ADIPOINST, n=29) revealed T cell-related signatures linked to HbA1c and insulin sensitivity, respectively. VAT Proteobacteria were negatively associated with insulin sensitivity. Multiomics and multitissue integration in the ADIPOINST and FLORINASH studies linked faecal Proteobacteria with jejunal and liver deoxycholic acid, as well as jejunal, VAT and liver transcriptomic signatures involved in the actin cytoskeleton, insulin and T cell signalling. Fasting glucose was consistently linked to interferon-induced genes and antiviral responses in the intestine and VAT. Studies in Drosophila melanogaster validated these human insulin sensitivity-associated changes.
Conclusion These data provide comprehensive insights into the microbiome-gut-adipose-liver axis and its impact on systemic insulin action, suggesting potential therapeutic targets.Cite Now
- OBESITY
- LIVER
- INTESTINAL BACTERIA
Data availability statement
Data are available upon reasonable request.
This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/.
Statistics from Altmetric.com
WHAT IS ALREADY KNOWN ON THIS TOPIC
In recent years, there has been growing recognition of the significant systemic effects of microbiota diversity and composition on the predisposition to obesity, type 2 diabetes and metabolic syndrome. However, most studies investigating the connections between gut microbiota and insulin sensitivity have used single-omics approaches focused on specific tissues and limited to individual human studies. Consequently, our understanding remains fragmented. The limitations of previous studies highlight the necessity for a more integrative approach to comprehend the interplay among gut microbiota, intestine, adipose tissue and liver in influencing systemic insulin action.
WHAT THIS STUDY ADDS
It is the first study to offer comprehensive insights into the microbiome-gut(jejunum, ileum and colon)-adipose-liver axis and its impact on systemic insulin action in humans, underscoring: (1) the pivotal role of Proteobacteria, bile acids, and T-cell related genes in influencing insulin sensitivity, (2) the involvement of Enterobacteria through several of the identified genes, and (3) associations between antiviral response genes across different tissues and fasting glucose. These findings significantly contribute to our understanding of the complex interplay among tissues and the gut microbiota within the context of obesity and insulin resistance.
HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY
The novel insights gained from our findings may pave the way for the development of innovative therapeutic strategies targeting the gut microbiota and the intestine-adiposeliver axis. Such strategies and targets hold the potential to enhance insulin sensitivity and improve metabolic health, representing promising avenues for advancement in clinical practice.
Introduction
Obesity is associated with multiple metabolic alterations, specifically insulin resistance and type 2 diabetes (T2D). The last years witnessed the recognition of strong systemic effects of microbiota in determining propensity to obesity, T2D and metabolic syndrome.1 2 In experimental models, the gut microbiota has been shown to increase intestinal permeability, favouring the translocation of microbiome-derived lipopolysaccharide (LPS) to the bloodstream. This leads to metabolic endotoxaemia, which initiates obesity and insulin resistance.3 4 While some research has explored the links between gut microbiota and insulin sensitivity in humans, most studies have used single-omic approaches focussing on individual tissues and are often limited to a singular human cohort. Consequently, our understanding remains fragmented, lacking a holistic understanding of the intricate interplay among the gut microbiota, intestine, adipose tissue and liver influencing systemic insulin action.5 The limitations of prior studies underscore the need for a more comprehensive and integrative approach to grasp the complexities of these interactions.
In this work, we conducted an unparalleled investigation of the gut-adipose-liver axis in humans by applying a pioneering comprehensive multiomics, multitissue, cross-cohort integrative approach encompassing transcriptomics in the intestine (jejunum, ileum and colon), liver and adipose tissue (both visceral and subcutaneous). In addition, we incorporated plasma metabolomics, lipidomics of the intestine and plasma, and faecal, ileal and colon metagenomics, coupled with the gold standard assessment of insulin sensitivity using the hyperinsulinaemic-euglycaemic clamp, complemented by indirect measurements of insulin sensitivity such as fasting glucose and glycated haemoglobin (Hb1Ac) levels. This investigation was performed across six independent studies (ADIPOINST, n=29; IRONMET, n=121; SIMMUNIDIA, n=55; FLOROMIDIA, n=37; FLORINASH, n=80 and ADIPOMIT, n=740, online supplemental table 1) and validations in Drosophila melanogaster. Our unique approach highlighted an intricate crosstalk between the intestine, liver and adipose tissue and revealed a robust and reproducible microbial-metabolic-transcriptomics signature involving Proteobacteria, bile acids and T cell-related genes linked to insulin sensitivity.
Supplemental material
Results
Proteobacteria and Erysipelotrichaceae species are negatively associated with insulin sensitivity
Few studies have investigated the relationship between gut microbiota and insulin sensitivity using the hyperinsulinaemic-euglycaemic clamp.6–11 Importantly, all these studies assessed microbial composition using 16S rRNA sequencing, which did not provide information on microbial composition at the species levels or microbial function and did not take into account the compositional nature of the metagenomics datasets. To overcome these limitations, we analysed the faecal microbiota of three independent studies (ADIPOINST, n=15; IRONMET, n=121 and FLORINASH, n=67) by shotgun metagenomics. We applied linear regression models in the log-scale taking into account the underlying compositional structure of the metagenomics data using the analysis of compositions of microbiome with bias correction (ANCOM-BC)12 adjusting for age, sex and body mass index (BMI). We found a consistent negative association of insulin sensitivity with several genera and species (figure 1A-E and online supplemental tables 2–7) from the Erysipelotrichaceae family and the Proteobacteria phylum, specifically from the Enterobacteriaceae family, across three independent studies. We further validated these results on a subset of patients from the FLOROMIDIA cohort with available hyperinsulinaemic-euglycaemic clamps (n=24) using faecal 16S rRNA sequencing. Once again, the genus Desulfovibrio from the Proteobacteria phylum was strongly negatively associated with insulin sensitivity (online supplemental figure 1 and online supplemental table 8).
Supplemental material
Conversely, insulin sensitivity was positively associated with microbial species from the Bifidobacterium and Prevotella genera. In the ADIPOINST and FLORINASH cohort, we also found a positive association between short-chain fatty acid (SCFA)-producing species from the Blautia and Faecalibacterium genus and insulin sensitivity (figure 1D-E). Recent studies also demonstrate the causal role of gut microbiota in insulin sensitivity regulation. Faecal microbiota transplantation (FMT) from lean donors has been shown to enhance peripheral insulin sensitivity,7 9 whereas FMT from metabolic syndrome donors has had the opposite effect.6 Consistent with our results, FMT from lean male donors to male subjects with obesity increased SCFA-producing bacteria like Bifidobacterium pseudolongum, while reducing Escherichia coli levels compared with the autologous group.9 Additionally, increases in Desulfovibrio spp. predicted a ≥10% decrease in insulin sensitivity, whereas Prevotellaceae spp predicted non-deterioration.6 Similarly, treatment with vancomycin in male patients with obesity decreased peripheral insulin sensitivity and increased gram-negative bacteria, mainly Proteobacteria.8 Elevated Proteobacteria levels have also been observed in individuals with T2D,13 14 although these studies often did not account for medication use. Conversely, other studies found no clear link between increased Proteobacteria and insulin sensitivity after vancomycin treatment in obese individuals,15 and under certain conditions, a decrease in Proteobacteria has been associated with insulin sensitivity or with improved metabolic outcomes, such as after gastric bypass16 17 or in toll-like receptor 2 knockout mice.18
Medication is known to impact the gut microbiota.19 20 Therefore, to rule out the confounding effects of medication on our current associations, we conducted additional analyses controlling for hypertension and dyslipidaemia medication, as well as the consumption of proton pump inhibitors, which were the primary medications in our studies. Additionally, although the ADIPOINST and IRONMET studies did not include patients with T2D (HbA1c <6.5% and fasting glucose <126 mg/dL), nine patients (13%) in the FLORINASH cohort had T2D. To further eliminate the confounding effect of diabetes, we controlled the analyses for the presence of T2D in the FLORINASH cohort. Remarkably, after controlling for these additional covariates, not only did most of the associations remain significant, but genera from the Proteobacteria phylum also exhibited the strongest negative fold changes and most significant (padj) associations with insulin sensitivity (online supplemental figure 2 and online supplemental tables 9–11).
Supplemental material
Previous studies have overlooked microbial functionality analyses, which are essential to accurately capture the host-microbiome interactions due to microbial functional redundancy.21 Hence, we next performed functional analyses based on Kyoto Encyclopaedia of Genes and Genomes (KEGG) orthologs in the three studies (online supplemental tables 12–14) and a pathway enrichment analysis using those microbial genes associated with insulin sensitivity (padj<0.05). In the ADIPOINST, IRONMET and FLORINASH studies, we found a consistent over-representation of pathways involved in energy metabolism such as the tricarboxylic acid cycle, oxidative phosphorylation, glycolysis/gluconeogenesis, pyruvate metabolism and the pentose phosphate pathway (figure 1G-I and online supplemental tables 15–17). Notably, both the IRONMET and FLORINASH studies showed a strong association with lipoic acid and histidine metabolism. Alpha-lipoic acid has been demonstrated to increase insulin sensitivity measured with hyperinsulinaemic-euglycaemic clamp in patients with T2D.22–24 A functional analysis based on KEGG modules identified histidine degradation as one of the most significant pathways (online supplemental figure 3 and online supplemental tables 18–20). Notably, imidazole propionate, a microbially produced histidine metabolite, is elevated in subjects with prediabetes and diabetes and has been shown to impair insulin signalling.25 26
Supplemental material
A T cell-related jejunal transcriptomic signature is positively associated with systemic insulin sensitivity
Recent evidence suggests the gut is a key tissue in obesity-related insulin resistance through the gut microbiota-induced inflammation27 by disrupting intestinal permeability and enhancing absorption of microbial products such as LPS.3 4 Most of the digestion of lipids and sugars takes place in the jejunum and is regulated by insulin.28 However, little is known about the jejunal transcriptome in relation to insulin sensitivity. Considering this, we performed an RNAseq of jejunal samples from the ADIPOINST cohort (n=26) and applied robust linear regression models adjusted for age, BMI and sex, to identify jejunal transcriptome signatures associated with the hyperinsulinaemic-euglycaemic clamp (figure 2A and online supplemental table 21). A pathway over-representation analysis highlighted that most of the upregulated genes played a key role in both the CD4 and CD8 helper T cell receptor signalling (TCR) and differentiation (Th1 and Th2), innate immunity, modulation of the inflammatory response, the RHO GTPase cycle and maintenance of the intestinal epithelium integrity (figure 2B,C and online supplemental table 22). CR2, involved in the complement cascade, and CCL11, encoding for an eosinophil-specific chemokine, were the two jejunal genes that had the highest effect size. These findings hint at the preservation of intestinal immunity and integrity with increased insulin sensitivity.
As the TCR signalling pathway had the strongest association with insulin sensitivity in the jejunum, we sought to measure the content of T cells in the ileum of another independent cohort (SIMMUNIDIA, n=42). We found that both the numbers and proportion of ileal T cells were negatively associated with fasting glucose and HbA1c (figure 2D-G). While interpreting these results, it is important to consider that the ileum has a slightly distinct immune microenvironment than the jejunum.29 Overall, these findings indicate that the presence of T cells seems to exercise a protective function in the small intestine. Similarly, the frequency of Th2 cells has also been negatively correlated with insulin resistance in mice and humans in other tissues like VAT.30 31
Ileal Proteobacteria and deoxycholic acid (DCA) exhibit a negative association with insulin sensitivity
The gut microbiota and derived metabolites are key in intestinal barrier disruption and local immune responses that contribute to inflammation and lead to metabolic disease.32 However, only a limited number of studies have described the composition of the small intestine microbiota, and there is even little evidence about its relationship with obesity and insulin resistance.33 Thus, we analysed the microbiota of the ileum mucosa in the SIMMUNIDIA cohort (n=42) using 16S rRNA gene sequencing and identified bacterial genera associated with HbA1c as an indirect marker of insulin resistance (figure 2H). In agreement with our findings in faeces, bacterial genera from the Proteobacteria phylum (Escherichia, Shigella and Enterobacter) were associated with insulin resistance (figure 2H and online supplemental table 23) and negatively with the total number of T cells (figure 2I and online supplemental table 24), while the Bifidobacterium genus and SCFA-producing genera such as Faecalibacterium were negatively associated. In line with our results, a high-fat diet (HFD) resulted in alterations in the ileum microbiota that impaired the immune system of the small intestine by decreasing the number of interleukin (IL)-17-producing CD4 T cells (Th17) and induced T2D in mice.34
We next profiled the lipidome and metabolome of the jejunum in the ADIPOINST cohort (n=26) to elucidate the small intestine biochemical pathways linked to insulin sensitivity. We applied a random forest-based variable selection machine learning algorithm to identify lipids and metabolites predictive of the hyperinsulinaemic-euglycaemic clamp. DCA, a microbial-derived secondary bile acid, was identified as the most important metabolite negatively associated with insulin sensitivity (figure 2J and online supplemental table 25). Valine and isoleucine were also strongly negatively associated with insulin sensitivity, whereas taurine had a positive association.
Consistently, jejunal levels of DCA were positively associated with the presence of proinflammatory bacterial species in faeces such as Gammaproteobacteria (E. cloacae, E. coli and Citrobacter) and negatively associated with species from the Bifidobacterium and Faecalibacterium genera (online supplemental figure 4 and online supplemental table 26). In line with our results, FMT from donors with metabolic syndrome resulted in an increase in faecal levels of DCA and other secondary bile acids while decreasing the insulin sensitivity of recipient patients.6 DCA is known to induce intestinal inflammation, disrupting the epithelial barrier and increasing gut permeability.35–37 Elevated levels of intestinal DCA have been linked to HFDs and western diets38 and increased abundance of the genus Clostridium, the main producer of DCA, promoting colonic inflammation.38 Plasma levels of DCA were associated with insulin resistance and significantly elevated in patients with T2D.39
Supplemental material
Proteobacteria and a T cell-related transcriptomic signature in the colon are also associated with insulin sensitivity
We also performed a colon transcriptomics analysis in the FLOROMIDIA cohort to identify genes associated with insulin sensitivity (figure 3A and online supplemental table 27). Consistent with our findings in the jejunum, pathways involved in the CD4/CD8 helper T cell lineage commitment and the Th17 type immune response were enriched (figure 3B, online supplemental table 28). We found that insulin sensitivity was positively associated with interleukin 17 receptor E (IL17RE) and interleukin 6 receptor (IL6R), but negatively with IL-23 subunit alpha (IL23A) (figure 3A,C). Similarly, the fasting glucose levels in the SIMMUNIDIA cohort were associated with the expression of IL-17 receptor B (IL17RB) and tumour necrosis factor receptor (CD40) (figure 3D and online supplemental table 29). We then analysed the microbiota of the colon in the SIMMUNIDIA cohort (n=55) using 16S rRNA gene sequencing. In agreement with our results at the ileum, we found that genera from the Proteobacteria phylum (Escherichia, Shigella and Gammaproteobacteria) were positively associated with insulin resistance assessed with the HbA1c and negatively associated with the number of T cells in the colon, whereas Faecalibacterium were again negatively associated (figure 3E,F and online supplemental tables 30,31).
An adipose tissue signature involved in T cell and Rho GTPase signalling is associated with systemic insulin sensitivity and resistance
Chronic inflammation in the adipose tissue has become a hallmark of obesity-related insulin resistance. Hence, we performed an RNA-seq analysis of the subcutaneous adipose tissue (SAT) of patients with and without obesity from the ADIPOMIT cohort (n=740). After fitting robust linear regression models controlling for age, BMI and sex, we identified several genes associated with HbA1c (figure 4A and online supplemental table 32). An enrichment analysis highlighted that genes negatively associated with HbA1c were involved in pathways that included insulin signalling, insulin resistance, IRS activation or leptin signalling, whereas genes positively associated with HbA1c were mainly involved in the TCR signalling, the immune system and the signalling by Rho GTPases (figure 4B,C and online supplemental tables 33,34), consistent with our findings in the jejunum and colon. However, while higher expression of the genes associated with these pathways in the intestine appears to be linked to greater insulin sensitivity, in adipose tissue, it seems to be associated with increased insulin resistance. This disparity might indicate different stages of development and/or asynchronous responses to environmental stimuli. In the intestine, higher insulin sensitivity could correspond to a stronger immune response, suggesting that an adequate intestinal immune response might prevent gut dysbiosis-induced insulin resistance. Conversely, in adipose tissue, an increased immune response could indicate insulin resistance-associated adipose tissue dysfunction. Chronic activation of this response in adipose tissue can become detrimental. However, it should be noted that these interpretations remain speculative.
We next performed an RNA-seq analysis of the VAT of patients from the ADIPOINST cohort (n=29). After applying robust linear regression models adjusted for age, BMI and sex, we identified a large number of VAT genes associated with insulin sensitivity (figure 5A and online supplemental table 35). Remarkably, a pathway enrichment analysis based on the genes negatively associated with insulin sensitivity revealed associations with innate immunity, specifically an over-representation of the TCR signalling pathway; the signalling by Rho GTPases; the IL4-, IL8-, IL3-, IL-18-, IL2-mediated signalling events and anticipated metabolic pathways such as leptin, adiponectin, glucagon and phosphatidylinositol 3-kinase (PI3K)/protein kinase B signalling (figure 5B and online supplemental table 36).
A multiomics and multitissue integration links Proteobacteria with jejunal DCA, jejunal and VAT genes involved in actin cytoskeleton, insulin and T cell signalling
Single-omics analyses only provide information about one layer of biological regulation. Therefore, we integrated the metagenomic, metabolomic and transcriptomic (jejunal and VAT) profiles linked to insulin sensitivity in the ADIPOINST cohort to obtain a holistic picture of the mechanisms underlying the gut microbiome-host interactions related to insulin sensitivity. Due to the large number of VAT genes associated with insulin sensitivity, we applied a multivariate method based on the multiblock-sparse projection to latent structures (MB-sPLS) using an L1 regularisation to select the 50 most relevant VAT genes and improve interpretation.
Cluster 1 depicted a strong negative association with insulin sensitivity and jejunal metabolites and lipids (DCA, valine, isoleucine, dehydroepiandrosterone sulfate (DHEAS)), microbial genera from the Proteobacteria phylum (Escherichia, Shigella and Legionella) and the Erysipelotrichaceae family (Allobaculum and Solobacterium) and transcriptomic signatures in the jejunum and VAT (figure 5C). Notably, this cluster was strongly negatively associated with cluster 2, with a strong correlation among metabolites (taurine), microbial genera (Faecalibacterium), jejunal and VAT genes strongly positively associated with insulin sensitivity.
DHEAS), microbial genera from the Proteobacteria phylum (Escherichia, Shigella and Legionella) and the Erysipelotrichaceae family (Allobaculum and Solobacterium) and transcriptomic signatures in the jejunum and VAT (figure 5C). Notably, this cluster was strongly negatively associated with cluster 2, with a strong correlation among metabolites (taurine), microbial genera (Faecalibacterium), jejunal and VAT genes strongly positively associated with insulin sensitivity.
To explore the role of the genes of both clusters in relation to insulin sensitivity, we conducted gene ontology-biological process (GO-BP) and pathway enrichment analyses. The most significant BP from jejunal genes in cluster 2 (positively associated with insulin sensitivity) involved lymphocyte migration and the regulation of the immune system (figure 5D). Consistently, the most significant jejunal pathways from this cluster included modulators of TCR signalling and T cell activation, the complement cascade, the innate immune system and the Rho GTPase cycle (figure 5E), whereas the most significant VAT pathway was precisely involved in the regulation of insulin-like growth factor (IGF) transport and uptake by IGFBPs (q value=0.0008).
Additionally, supramolecular fibre organisation (CCL1, CHRFAM7A, CYFIP2 and RHOH) and actin cytoskeleton organisation (CCL11 and RHOH) were among the GO-BP over-represented in cluster 2. Actin disassembly in both adipose and muscle cells has been shown to inhibit insulin-induced events such as glucose transporter recruitment to the cell surface and enhanced glucose transport40; actin is indispensable for the insulin-stimulated translocation of the glucose transporter GLUT4.41 Plasma CCL11, has been positively associated with steatosis severity42 and CCL11 knockout mice fed a high-fat, high-calorie diet improved insulin sensitivity.43 Additionally, RHOH is a key factor for the development and proper function of T cells.44 45
Of note, the GO-BP and pathways from the VAT most strongly associated with cluster 1 (negatively associated with insulin sensitivity) comprised again the regulation of the actin cytoskeleton organisation and regulation but also the insulin signalling (figure 5F,G) Similarly, the over-represented BP from these cluster in the jejunum also included the musculoskeletal movement and the supramolecular fibre organisation (figure 5H).
Finally, to assess whether the negative associations between faecal Proteobacteria and insulin sensitivity translated to the adipose tissue, we performed 16S rRNA sequencing of the VAT in a subset of patients from the IRONMET cohort (n=12). In line with our results, we found that genera from the Proteobacteria phylum (Bradyrhizobium and Moraxellaceae families) in the VAT were negatively associated with insulin sensitivity (figure 5I and online supplemental table 37).
A multiomics and multitissue integration links proteobacteria, liver DCA and liver genes involved in actin cytoskeleton, insulin secretion and the immune system
The liver plays a central role in modulating glucose and insulin. Therefore, we next profiled the liver transcriptome of obese patients from the FLORINASH cohort (n=80). After fitting robust linear regression models adjusted for age, BMI and sex, we identified 1356 genes associated with the hyperinsulinaemic-euglycaemic clamp (figure 6A and online supplemental table 38). Consistent with our findings in the jejunum and the VAT, these genes were involved in the TCR signalling, the complement system, the Ras and Rap1 GTPase signalling and the cytokine–cytokine receptor interaction (figure 6B and online supplemental table 39). We also profiled the lipidome of the liver of a subset of these patients. We found several lysophosphatidylcholines and lysophosphatidylethanolamines associated with insulin sensitivity (figure 6C, online supplemental figure 5A and online supplemental table 40). Importantly, we found again a negative association of DCA with insulin sensitivity (figure 6C and online supplemental figure 5A). We also analysed the microbiota of the liver of a subset of participants from Spain (n=17) and Italy (n=20) in the FLORINASH study. Consistent with our previous results in faeces, ileum, jejunum and colon, we found that genera from the Proteobacteria phylum in the liver exhibited the strongest and/or most significant negative associations with insulin sensitivity, after controlling for age, BMI, sex, dyslipidaemia and hypertension medication and diabetes both in Spain (figure 6D and online supplemental table 41) and Italy (online supplemental figure 5B and online supplemental table 42). A MB-sPLS-based multiomics integration of the faecal microbiota and liver lipids and genes (only the 50 most relevant using an L1 regularisation) associated with insulin sensitivity revealed the presence of a cluster of highly interconnected features including genera from the Proteobacteria phylum (Deltaproteobacteria_uc, Desulfovibrionaceae_uc, Bilophila and Desulfovibrionales_uc) and the Erysipelotrichaceae family, primary (taurocholic acid) and secondary (DCA) bile acids and a transcriptomic signature negatively associated with insulin sensitivity (figure 6E).
Supplemental material
Consistent with our previous results in the jejunum and VAT, a pathway enrichment analysis on this transcriptome identified an over-representation of the Rap1 signalling pathway, the leucocyte transendothelial migration, the regulation of the actin cytoskeleton, the chemokine signalling pathway, the immune system, the cytokine signalling in the immune system and the secretion of insulin, among others (figure 6F,G and online supplemental table 43). Again, a GO-BP analysis highlighted an enrichment of pathways mainly involved in lymphocyte activation, leucocyte cell–cell adhesion, regulation of leucocyte migration and T cell activation (online supplemental table 44).
HFD and mono associations with Enterobacter cloacae induce transcriptional changes associated with insulin resistance in D. melanogaster intestine and fat body
To further explore the multiorgan transcriptional changes associated with insulin sensitivity, we used the model organism D. melanogaster. To model insulin resistance in Drosophila, we administer a HFD that causes an obese-like phenotype in flies, including increased circulating glucose levels and insulin-like resistance.46 To further assess the role of Proteobacteria in insulin resistance, E. cloacae was selected for monocolonisation experiments in D. melanogaster due to its dual relevance as both a commensal of the Proteobacteria phylum in Drosophila47 48 and its established association with obesity-related phenotypes in humans and animal models (figure 7A).49–51 We depicted the Drosophila orthologues of the jejunal genes identified in cluster 2 involved in the regulation of the immune system CR2 (hig), DOCK8 (Zir), GRAP2 (drk), CYFIP2 (Cyflip), SPN (Deaf) and RHOH (Rac1) (figure 5D) and assessed relative gene expression by qRT-PCR in the fly’s intestine fed with standard diet (SD) or HFD either raised in axenic conditions or mono-associated with E. cloacae. HFD-fed flies exhibited a significant decrease in the expression of hig, Rac1, Cyflip and Deaf, which aligns with our findings in humans, as these genes were positively associated with insulin sensitivity in the jejunum (figure 7B-G). Similarly, SD-fed flies mono-associated with E. cloacae showed a significant reduction in the intestinal expression of hig, Rac1 and a tendency for Deaf (figure 7B-G). These findings collectively suggest that both HFD and mono-associations with E. cloacae lead to alterations in intestinal gene expression patterns reminiscent of those observed in insulin-resistant humans.
Next, we conducted gene expression analysis in the Drosophila fat body, which serves as the equivalent of human adipose tissue. We assessed the expression of Cluster 1 genes, Past1, ssh, siz, cta, Dsor1 and Col4a1, which correspond to the human homologues of EHD2, SSH1, IQSEC1, GNA12, MAP2K1 and COL4A2, respectively (figure 5F,G). HFD-fed flies displayed a significant increase in the expression of Col4a1, siz and Past1 compared with SD-fed flies (figure 7H-M). In humans, these genes were negatively associated with insulin sensitivity in the VAT (figure 5F,G). Mono-associations with E. cloacae did not affect FB expression of the selected genes.
Fruit flies do not contain an organ equivalent to the liver; its functions are assumed by the fat body.52 53 Therefore, we assessed fat body gene expression of vib, Moe, Ac76E and mys (figure 7N-Q), the Drosophila orthologues of PITPNA, MSN, ADCY7, and ITGB2, all of them associated with the hyperinsulinaemic-euglycaemic clamp in liver transcriptomics (figure 6G). HFD-fed flies mono-associated with E. cloacae exhibited a significant increase of vib expression in the FB (figure 7N). Remarkably, we observed that PITPNA, its corresponding human orthologue, was depicted as one of the genes associated with insulin resistance together with species from the Proteobacteria phylum (figure 6E). Furthermore, Moe, the Drosophila orthologue of MSN, which displayed an inverse association with insulin sensitivity in the human liver, exhibited a significant upregulation in HFD-fed flies (figure 7O).
Finally, in order to investigate the role of Proteobacteria in insulin resistance and validate our model, we assessed the expression of dilp2 (Drosophila insulin-like peptide 2), one of the Drosophila orthologues of human insulin. We observed increased expression of dilp2 in HFD-fed flies compared with SD-fed flies, resembling insulin resistance as previously described.46 Mono-association of flies with E. cloacae further elevated the levels of dilp2 expression of SD-fed flies, indicating that this bacterium exacerbates insulin resistance also in D. melanogaster (figure 7R). Additionally, flies monocolonised with E. cloacae showed a significant decrease in the expression of hydroxyphenylpyruvate dehydrogenase (Hpd) compared with axenic flies (figure 7S). Hpd expression is regulated by the transcription factor dFOXO, which is activated downstream of PI3K in response to insulin.54 In conditions of insulin resistance, there is a decrease in dFOXO signalling and the expression of its downstream genes55 (figure 7U). Finally, when challenged for glucose tolerance,56 larvae monocolonised with E. cloacae did not clear glucose from haemolymph as efficiently as sterile larvae. Ninety min after ingesting a glucose solution, the E. cloacae-colonised larvae had significantly higher levels of glucose in the haemolymph (figure 7T). Given that Drosophila larvae primarily use trehalose as a circulating sugar,56 we also measured the levels of circulating trehalose in addition to glucose. Our findings indicated that larvae monocolonised with E. cloacae consistently exhibited higher levels of trehalose and glucose in the haemolymph compared with sterile larvae (online supplemental figure 6).
Supplemental material
Overall, these experiments align with the transcriptional changes observed in the human intestine, VAT and liver, correlating with insulin sensitivity and further supporting the role of the Proteobacteria in the exacerbation of insulin resistance pathophysiology when present in the intestine.
Fasting glucose levels are linked to antiviral response genes
Our results identified consistent associations between insulin sensitivity and glycaemic state with specific microbiota and T cell numbers and signalling in different tissues. Given the importance of T cells in responding to viral infections, and our observation of intriguing associations between insulin sensitivity and viral infections such as Epstein-Barr virus infection, viral myocarditis and human cytomegalovirus infection (figure 2B and figure 6F), we also analysed the associations of fasting glucose with the transcriptome in the jejunum, ileum, VAT and liver (figure 8A-C,G and online supplemental tables 45–48). Strikingly, a GP-BP analysis revealed the defence response to the virus (figure 8D,H and online supplemental table 49) and viral transcription (figure 8E,H and online supplemental table 50) as the most over-represented processes in the jejunum and ileum. It also highlighted the type I interferon (IFN) production and the cytoplasmic pattern recognition receptor pathway in response to the virus (figure 8D). A KEGG over-representation analysis in the colon and VAT identified a significant enrichment of the viral protein interaction with cytokine and cytokine receptor (figure 8F,H and online supplemental table 51), and the viral infection, including the hepatitis C, influenza A and cytomegalovirus infection, which have been associated with an increased risk for the development of prediabetes and diabetes.57 A pathway over-representation analysis in the jejunum also highlighted an enrichment of the IFN signalling, the antiviral mechanism by IFN-stimulated genes and the oligoadenylate synthetase antiviral response (online supplemental table 52), while the type II IFN signalling was also over-represented in the colon (figure 8H and online supplemental table 53). A recent study also identified that downregulation of glucose metabolism promoted retinoic acid-inducible gene I-like receptor (RLR)-induced type I IFN production along with a reduction in virus replication and identified elevated lactate dehydrogenase and/or glycolysis as a potential mechanism for viruses to evade host defence by inhibiting RLR-triggered type I IFN production.58 Type I IFNs play a crucial role in protecting cells from viral pathogens. A screening of over >380 human IFN-stimulated genes revealed eight broadly acting effectors, including IRF7 and IFIH1,59 which we also found associated with glucose levels in the ileum (figure 8H). Genes identified in the jejunum included OASL, a gene involved in targeted antiviral specificity and well-known antiviral effectors such as EIF2AK2, or the IFN-induced genes, OAS1, OAS2 and OAS3, which play a key role in innate cellular antiviral response and have recently been identified as associated with critical illness caused by COVID-19.60
Conclusions
Our study sought to uncover the complex interactions among the gut microbiota, intestine, adipose tissue and liver and their effects on systemic insulin action. Using a multiomics (metagenomics, metabolomics, transcriptomics and lipidomics), multitissue (jejunum, ileum, adipose tissue and liver) approach across six different studies, combined with gold-standard measurements of insulin sensitivity using the hyperglycaemic-euglycaemic clamp and other measurements of glucose metabolism and insulin resistance, we provide the first comprehensive analysis of its kind. This integrative approach offers an unprecedented comprehensive view into the intricate microbiome-gut-adipose-liver crosstalk influencing insulin sensitivity (figure 9).
However, our study has several limitations. Although it includes multiomics measurements in multiple tissues across multiple studies, its nature is mainly observational. In addition, some studies had a small sample size, which is a limitation for the analysis of high-dimensional omics data sets and their integration. Thus, data from large-scale longitudinal studies would have further supported the current findings. Finally, not all omics data were available in all tissues and all studies. For instance, faecal shotgun metagenomics data was only available in three studies, while gold standard measurements of insulin sensitivity were only performed in four studies.
Despite this, it is the first study to offer comprehensive insights into the microbiome-gut-adipose-liver axis and its impact on systemic insulin action in humans. Our approach was crucial in underscoring the pivotal role of Proteobacteria, bile acids and T cell-related genes in influencing insulin sensitivity and identified a positive association between insulin sensitivity and SCFA-producing species from the Blautia and Faecalibacterium genera. Thus, our single- and integrated-omics analyses unveiled faecal microbial signatures comprising taxa from the Proteobacteria phylum and DCA in the jejunum negatively associated with insulin sensitivity, along with intestine (jejunum, ileum and colon) T cell-related signatures that were linked to better insulin sensitivity. Transcriptomics analyses of the SAT and VAT also revealed upregulated and downregulated T cell-related signatures linked to HbA1c and insulin sensitivity, respectively, while Proteobacteria in the VAT were negatively associated with insulin sensitivity. This opposite association of T cell signatures in the intestine and VAT in relation to insulin sensitivity might indicate asynchronous responses to environmental stimuli. Increased intestinal immune response might prevent gut dysbiosis-induced insulin resistance, while increased adipose immune response could indicate insulin resistance-associated adipose tissue dysfunction, for which chronic activation could be detrimental. Multiomics analyses in the liver revealed again negative associations of liver deoxycholic acid with insulin sensitivity as well as a liver transcriptomic signature involved in insulin and T cell signalling. Notably, the involvement of Enterobacteria through several of the identified genes in the jejunum, VAT and liver was validated in the intestine and fat body of D. melanogaster models. Lastly, we identified a consistent link between glucose metabolism and IFN-induced genes involved in the antiviral response. In fact, recent evidence supports a bidirectional relationship between metabolic disease and viral infection.57 Hence, the immune system may induce transient changes in glucose metabolism as a strategy against viral infection.61 Our study emphasises the significance of considering microbial functionality and adopting a multiomics integration approach to thoroughly understand the mechanisms driving the complex interplay among tissues and the gut microbiota within the context of obesity and insulin resistance. The novo insights gained from our findings may lead to the development of innovative therapeutic strategies targeting the gut microbiota and the intestine-adipose-liver axis to enhance insulin sensitivity and improve metabolic health.
Supplemental material
Data availability statement
Data are available upon reasonable request.
Ethics statements
Patient consent for publication
Ethics approval
This study involves human participants. All participants gave written informed consentto participate in the study before taking part and all procedures were performed in accordance with the Declaration of Helsinki. The studies IRONMET (2015.111; 2022.184 and 2023.239), ADIPOMIT (2019.062) and ADIPOINST (2016.051 and 2019.143) were approved by Ethics Committee of the Hospital Dr Josep Trueta. FLORINASH was approved by the Ethics Committee of the Hospital Dr Josep Trueta (200904, 2022.187, 2022.066) and the ethical committee of Policlinico Tor Vergata University of Rome (49/09). FLOROMIDIA was validated and approved by the ethical committee of Policlinico Tor Vergata University of Rome (Comitato Etico Indipendente, approval number 113/15). SIMMUNIDIA was approved by the Ethics Committee of the Rangueil Hospital, France (RC31/16/8217). Registration number IDRCB/EudraCT (reference CPP-ANSM): 2016-A01213-48. ClinicalTrials.gov Identifier: NCT02811575.
References
Supplementary materials
Supplementary Data
This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.
Supplementary Data
This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.
Footnotes
X @moyasimarro1
AC-N and JMM-N contributed equally.
Contributors AC-N performed part of statistical analysis and wrote the manuscript. JMM-N researched the data and partially wrote the manuscript. MA-R contributed to data collection for the IRONMET and ADIPOINST studies. AC-N, LdlV-C and IP performed Drosophila experiments. RB, LG-P, PG and LC contributed to the SIMMUNIDIA cohort. GF, LMS, JL-M, FT and CD contributed to the ADIPOMIT cohort. MF contributed to the FLOROMIDIA cohort. RB, M-ED, MF and JMF-R contributed to the FLORINASH cohort. VP-B and AM contributed to the shotgun metagenomics analysis of faecal samples. All authors contributed to the discussion and reviewed the manuscript. JMF-R and JM-P carried out the conception and coordination of the study, performed statistical analysis and wrote the manuscript. JIR-H and EC contributed to the collection of adipose and intestine tissue samples. JMF-R and JM-P are the guarantors.
Funding This work was partly supported by EU-FP7 FLORINASH (Health-F2-2009-241913) to RB, MF and JMFR, by Instituto de Salud Carlos III (Madrid, Spain) through the projects PI15/01934, PI18/01022, PI21/01361 and PMP21/00072 (co-funded by the European Union/Plan de Recuperación, Transformación y Resiliencia) to JMFR, PI19/01712 to JMM-N, and PI20/01090 and PI23/00575 to JM-P (co-funded by the European Union under the European Regional Development Fund (FEDER). 'A way to make Europe'), the project CNS2023-144218 funded by MCIN/AEI/10.13039/501100011033 and the European Union NextGenerationEU/PRTR to JM-P and Generalitat de Catalunya 2021 SGR 01263. This work has also been co-financed by the Spanish Ministry of Science, Innovation and Universities with funds from the European Union NextGenerationEU, from the Recovery, Transformation and Resilience Plan (PRTR-C17.I1) and from the Autonomous Community of Catalonia within the framework of the Biotechnology Plan Applied to Health through the project PPCC-ADGUT to JM-P. JM-P and ACN are funded by Instituto de Salud Carlos III (Madrid, Spain) through the Miguel Servet Program CP18/00009 and Sara Borrell Program CD20/00051 (co-funded by the European Union under the European Social Fund 'Investing in your future'), respectively. IDIBGI is a CERCA centre from the 'CERCA Programme/Generalitat de Catalunya'. The Cohort SIMMUNIDIA has been promoted by the Rangueil Hospital of Toulouse, France and received an essential financial support from the Federation Française de Recherche en Diabetologie, technical support from the departments of Gastroenterology and Diabetes. This study has been granted from the Fondation Francophone pour la Recherche sur le Diabète, sponsored by the FFD, Abbott, AstraZeneca, Lilly, MSD, Novo Nordisk and Sanofi. We thank Emeline Lhuilier at GeT-Santé facility (I2MC, Inserm, Génome et Transcriptome, GenoToul, Toulouse, France) for its contribution to the sequencing data (US1426, INRAE, Génome et Transcriptome, GenoToul, Toulouse, France) and Vaiomer SAS (Labège innopole France) for 16SrRNA sequencing. We thank Vincent Azalbert for then immune cell preparation. We thank Professor Louis Buscail, Professor Barbara Bournet and Mrs Emilie Bergereau (Department of Gastroenterology, CHU de Toulouse, Toulouse, France) for their contribution to the SIMMUNIDIA study. FLORINASH is also funded by GutsUK and Diabetes UK grants to M-ED and supported by the NIHR Imperial Biomedical Research Centre and by grants from the French National Research Agency (ANR-10-LABX-46 (European Genomics Institute for Diabetes)), the National Center for Precision Diabetic Medicine, which is jointly supported by the French National Agency for Research (ANR-18-IBHU-0001), the European Union (FEDER), the Hauts-de-France Regional Council (Agreement 20001891/NP0025517), the European Metropolis of Lille (agreement 2019_ESR_11) and site ULNE (R-00220-TALENT-DUMAS), which is also jointly funded by ANR (ANR-16-IDEX0004-ULNE), the Hauts-de-France Regional Council (20002845) and the European Metropolis of Lille. This research was conducted within the context of the CNRS–ULille–Imperial International Research Project in Integrative Metabolism 'METABO-LIC'.
Competing interests None declared.
Provenance and peer review Not commissioned; externally peer reviewed.
Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.