Major microbiota dysbiosis in severe obesity: fate after bariatric surgery ========================================================================== * Judith Aron-Wisnewsky * Edi Prifti * Eugeni Belda * Farid Ichou * Brandon D Kayser * Maria Carlota Dao * Eric O Verger * Lyamine Hedjazi * Jean-Luc Bouillot * Jean-Marc Chevallier * Nicolas Pons * Emmanuelle Le Chatelier * Florence Levenez * Stanislav Dusko Ehrlich * Joel Dore * Jean-Daniel Zucker * Karine Clément ## Abstract **Objectives** Decreased gut microbial gene richness (MGR) and compositional changes are associated with adverse metabolism in overweight or moderate obesity, but lack characterisation in severe obesity. Bariatric surgery (BS) improves metabolism and inflammation in severe obesity and is associated with gut microbiota modifications. Here, we characterised severe obesity-associated dysbiosis (ie, MGR, microbiota composition and functional characteristics) and assessed whether BS would rescue these changes. **Design** Sixty-one severely obese subjects, candidates for adjustable gastric banding (AGB, n=20) or Roux-en-Y-gastric bypass (RYGB, n=41), were enrolled. Twenty-four subjects were followed at 1, 3 and 12 months post-BS. Gut microbiota and serum metabolome were analysed using shotgun metagenomics and liquid chromatography mass spectrometry (LC-MS). Confirmation groups were included. **Results** Low gene richness (LGC) was present in 75% of patients and correlated with increased trunk-fat mass and comorbidities (type 2 diabetes, hypertension and severity). Seventy-eight metagenomic species were altered with LGC, among which 50% were associated with adverse body composition and metabolic phenotypes. Nine serum metabolites (including *glutarate*, *3-methoxyphenylacetic acid* and *L-histidine*) and functional modules containing protein families involved in their metabolism were strongly associated with low MGR. BS increased MGR 1 year postsurgery, but most RYGB patients remained with low MGR 1 year post-BS, despite greater metabolic improvement than AGB patients. **Conclusions** We identified major gut microbiota alterations in severe obesity, which include decreased MGR and related functional pathways linked with metabolic deteriorations. The lack of full rescue post-BS calls for additional strategies to improve the gut microbiota ecosystem and microbiome–host interactions in severe obesity. **Trial registration number** [NCT01454232](http://gut.bmj.com/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01454232&atom=%2Fgutjnl%2F68%2F1%2F70.atom). * intestinal tract * intestinal bacteria * gastric surgery * obesity * obesity surgery ### Significance of this study #### What is already known on this subject? * Moderate obesity is characterised by decreased microbial gene richness (MGR) (20%–40% of the patients) associated with altered metabolic risk and a shift in metagenomic species (MGS) signature. * This has not been explored in severe obesity. * Some studies with limited number of subjects showed changes in the gut microbiota, but none explored precisely MGR and combined related metagenomics and metabolomics signatures after 6 months of follow-up. #### What are the new findings? * This is the first comprehensive study performed in severe obesity demonstrating a very high prevalence of patients (75%) with decreased MGR, which associates with overt metabolic complications. * We describe novel metabolomic and MGS signatures that are specific to decreased MGR found in severe obesity. * Bariatric surgery (both adjustable gastric banding and Roux-en-Y-gastric bypass (RYGB)) improves MGR, but it is partially restored in most patients, and most remain with low MGR despite major metabolic improvement and weight loss in all patients. * Clusters of metabolites (such as glycine, acetylglycine and methylmalonate) increasing post-RYGB were linked with improved body composition. * Importantly, even longer periods post-RYGB (ie, 5 years) do not further increase MGR. #### How might it impact on clinical practice in the foreseeable future? * Our results question whether specific interventions (specialised diets, prebiotics/probiotics or gut microbiota transfers) may be useful to consider prior or post bariatric surgery in severely obese individuals in order to further improve MGR and metabolic health postbariatric interventions. ## Introduction Among the complex obesity causes1 and its related diseases2 (type 2 diabetes (T2D) and cardiometabolic diseases), the gut microbiota appears to be a relevant contributor and is likely a pivotal factor between changes in lifestyle and host biology (for review, see ref 3). Gut dysbiosis was identified in overweight and moderate obesity,4 5 as evidenced by substantial modifications in the gut microbiota composition (with enrichment or decrease in specific bacterial groups) and low microbial gene richness (MGR),4 5 which are associated with metabolic alterations (insulin resistance, low-grade inflammation and adipocyte hypertrophy).4 5 However, gut microbiota characteristics have been scarcely explored in extreme forms of obesity, although severe (body mass index (BMI) >35 kg/m²) and morbid obesity (BMI >40 kg/m²) have progressed worldwide, reaching 2.3% and 5% in men and women, respectively. While some severely obese patients remain metabolically healthy,6 in general, reaching a BMI >35 kg/m² induces a significant rise in chronic disorders.1 Furthermore, healthy obese individuals often develop metabolic alterations and comorbidities with time.6 Furthermore, severe/morbid obese individuals represent the only eligible candidates for bariatric surgery (BS), a treatment which has dramatically increased worldwide7 as it reduces cardiovascular risks and improves metabolic conditions.8 BS represents a good model to understand the intestinal contribution to health improvements by comparing adjustable gastric banding (AGB), a procedure solely inducing caloric restriction due to gastric volume reduction (equivalent to a successful diet intervention), and Roux-en-Y-Gastric bypass (RYGB), which by contrast drastically rearranges the digestive tract architecture and adds intestinal malabsorption to food intake reduction.9 MGR is modulated by dietary interventions and increased by 30% after a short-term dietary restriction (with fibre enrichment) in overweight/moderately obese individuals.5 Few studies addressed microbiota evolution using whole shotgun metagenomics (WGS)10–12 in paired subjects followed at several time points post-BS. Particularly, MGR evolution post-BS as well as its relation with other characteristics (clinical improvements or systemic metabolomics) have been scarcely assessed. Importantly, 50% of these beneficial associations depend on post-BS dietary modifications,13 therefore confirming the need to compare microbiota modifications after different BS techniques, namely AGB and RYGB, where food reduction in terms of total calorie does not differ.14 Herein, we used WGS and aimed to examine (1) whether MGR worsens in severe obesity and how it relates to aggravation of comorbidities, and (2) whether different BS types could differentially correct severe obesity-related gut microbial characteristics, including changes in MGR, composition and function. ## Materials and methods ### Clinical cohorts We prospectively included 61 severely obese women (Microbaria (MB) at Pitié-Salpêtrière Hospital Obesity Unit, Paris) (figure 1A), as described.15 Patients were assigned for AGB or RYGB following international BS guidelines (ie, BMI ≥40 kg/m² or ≥35 kg/m² with at least one severe obesity-related comorbidity) and patients’ preferences, a decision subsequently validated by a multidisciplinary panel. RYGB was frequently chosen for more severely diseased individuals. ![Figure 1](http://gut.bmj.com/https://gut.bmj.com/content/gutjnl/68/1/70/F1.medium.gif) [Figure 1](http://gut.bmj.com/content/68/1/70/F1) Figure 1 MGR in severe obesity. (A) Study flow chart: baseline (MB or MB+MO) and MB follow-up cohorts. Two independent confirmation cohorts (EROIC and ATOX) were used for data confirmation. (B) MGR bimodal distribution in the MB baseline cohort. (C) Baseline MGR in AGB and RYGB patients, including four enterotype characteristics in each surgery group. AGB, adjustable gastric banding; HGC, high gene count; LGC, low gene count; MB, Microbaria; MGR, microbial gene richness; MO, MICRO-Obes; RYGB, Roux-en-Y-gastric bypass. Clinical, anthropometric and biological evaluations were obtained at baseline (T0) and during follow-up at 1 month (T1), 3 months (T3) and 12 months (T12) post-surgery.16 T2D, glucose intolerant status and dyslipidaemia status (definitions in the online supplementary materials and methods) were acknowledged. Dual X-ray absorptiometry estimated the body composition (Hologic Discovery W, V.12.6 software, 2; Bedford, Massachusetts),16 which included total fat-free mass, total-fat mass, trunk-fat mass (all in kilogram or %) and gynoid-fat partitioning. Patients filled in the questionnaires to record general health, medications, birth mode and the Bristol Stool Scale (BSS). Blood samples were collected after a 12-hour overnight fast at all described time points to measure biochemical parameters using routine techniques for glucose homeostasis and lipid profiles. An oral glucose tolerance test (OGTT) was performed in a subset of patients (n=21, 34%) at T0 to assess glucose and insulin area under the curve (AUC), and the Stumvoll Index was used to characterise glucose tolerance.17 All patients undergo the same preparation pathway that lasts on average 6–12 months, where they initially are advised to have an equilibrate diet. On average their T0 visit was performed 3 months prior to their surgery. We do not advise them to modify their diet or undergo weight loss, but rather stabilise their weight. ### Supplementary data [[SP1.pdf]](pending:yes) Faeces were collected at each visit using a standardised method.18 No patients had received antibiotic treatment for 3 months pre-BS, nor had any history of acute or chronic GI diseases. All subjects signed the informed written consent and the protocol was registered at ClinicalTrial.gov ([NCT01454232](http://gut.bmj.com/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT01454232&atom=%2Fgutjnl%2F68%2F1%2F70.atom)). To examine gut microbiota across a broader range of BMI and metabolic complications, we used the previously described5 MICRO-Obes (MO) cohort (figure 1A), similar in age and composed mainly of women (84%), who are overweight/moderately obese patients without T2D or any medical treatment. Two confirmation and independent cohorts were added, including patients with severe obesity undergoing RYGB. A group of 10 severely obese individuals (ie, ATOX cohort) who underwent RYGB were further followed at T0, 3, 12 months and 5 years after their surgery, for whom we sampled faeces and performed WGS to analyse MGR and metagenomic species (MGS)-associated signatures. This cohort was initially designed for characterisation of longer term microbiota evolution. Therefore, we had access to only four patients at 1 year and the whole group at 5 years. Another independent confirmation cohort of 147 severely obese individuals (ie, EROIC cohort: 64 patients with T2D and 83 obese non-T2D patients) were followed at T0 and 12 months post-surgery and for whom we sampled blood and performed metabolomics analysis (figure 1A). MB, ATOX and MO gut microbiota were sequenced using the same WGS methodology and bioinformatics processing. Similar clinical phenotyping were also acquired in both cohorts. ### Gut microbiota analysis by quantitative metagenomics Participants collected faecal samples in two 20 mL tubes within 24 hours before each visit. Samples were either stored immediately at −80°C or briefly conserved in home freezers, in anaerobic conditions, before transport to the laboratory where they were immediately frozen at −80°C following guidelines.18 The total faecal DNA from 182 MB samples was extracted, sequenced and analysed. DNA extraction used quenching solutions to protect DNA from degradation by DNases and a bead-beating step that ensures the lysis of particularly robust cells.19 20 A barcoded fragment library was prepared for each sample and DNA sequencing data were generated using SOLiD 5500xl sequencers. An average of 68.72 million 35-base-long single reads (SD 26M) were obtained for the samples. The same methodology also applies for ATOX. Primary analysis, from reads quality cleaning to read mapping over a 3.9 million gene reference catalogue,21 was performed using Meteor Studio.5 Secondary analyses, from gene abundance normalisation to MGS projection,21 and statistical analysis were performed using the *momr* R package. The online supplementary materials and methods describe in detail the bioinformatics processing. ### Serum metabolomics Serum metabolomics were performed for 58 MB patients at baseline (figure 1A). Serum samples were extracted using cold acetonitrile containing labelled mix of 16 amino acids at 12.5 µg/mL and processed as described in the online supplementary materials and methods. LC-MS analysis was carried out on a UPLC Waters Acquity (Waters, Saint-Quentin-en-Yvelines, France) coupled to a Q Exactive (Thermo Fisher Scientific, Illkirch, France). Chromatographic conditions were adapted to screen microbiota-derived metabolites as described.22 Data were curated, normalised and annotated, yielding 242 different metabolites. Details about preprocessing and processing steps are reported in the online supplementary materials and methods. ### Statistical analyses Statistical analyses were performed on R using public and inhouse packages. Non-parametric statistics were performed when variables displayed non-normality. All tests were corrected for multiple testing using Benjamini-Hochberg. Results were considered significant at an false delivery rate (FDR) <5% (unless specified otherwise). Paired testing was performed for comparing samples across time. Graphics were built using R core and *ggplot* plots. ## Results ### Gut microbiota richness and clinical phenotypes in severe obesity In 61 obese women (MB) with BMI >35 kg/m², MGR exhibited a bimodal distribution.4 5 Using the same methodology and gene cut-off (480 000) as previously4 5 to create low gene count (LGC)/high gene count (HGC) classes, the vast majority of patients (75%) belonged to the LGC group (figure 1B), a dramatic increase compared with overweight/moderate obesity.4 5 At baseline, RYGB and AGB patients had similar overall clinical characteristics and BMI, except for increased DEXA-quantified trunk-fat mass and prevalence of obstructive sleep apnoea in RYGB patients (online supplementary table 1). MGR was significantly higher in AGB patients compared with RYGB patients (p=0.013) (figure 1C). ### Supplementary data [[SP2.pdf]](pending:yes) LGC inversely correlated with metabolic alterations: triglycerides (p=0.049), uricaemia (an indirect insulin resistance marker, p=0.038), and systemic inflammation markers fibrinogen (p=0.048) and neutrophil count (p=0.042) (online supplementary table 2).4 5 Beyond previous findings, MGR was identified to be inversely correlated with detrimental body composition (ie, trunk-fat mass (r=−0.27 p=0.04)) and was significantly decreased in patients with T2D (p=0.014), as compared with normoglycaemic patients. The MB cohort included nine patients with T2D (among the 61) and eight of them were on metformin treatment, the first treatment given to control glucose homeostasis, as recommended by the international recommendations for T2D therapy.23 The only one without metformin did not need any antidiabetic drugs to remain below the target of HbA1c of 6.5%. MGR decreased in patients with elevated blood pressure (p=0.05), and we observed a trend (p=0.058) towards more features of metabolic syndrome (International Diabetes Federation (IDF) definition) in LGC patients (online supplementary table 2). The BSS revealed that softer stools were associated with decreased MGR (p=0.005, r=−0.42) (online supplementary table 2). We did not observe any effect of birth mode (vaginal or C section), proton pump inhibitor (PPI) use or smoking on MGR. ### Supplementary data [[SP3.pdf]](pending:yes) To gain further insight into the microbiome composition in addition to MGR, we used the dirichlet multinomial mixtures (DMM) approach of Holmes *et al* 24 to characterise the enterotype composition of the MB cohort. We herein showed the presence of the same four enterotypes described recently25 explaining 40% of the variation in microbiome composition (R2=0.4; permutational multivariate analysis of variance (PERMANOVA) tests) (online supplementary figure 1), yet there was no significant association of enterotype composition and BSS at baseline (Fisher’s test p=0.97 (AGB) and p=0.57 (RYGB)). Interestingly, we observed that patients with the B2 enterotype were those with the lower MGR, whereas those with the Ruminococcaceae enterotype (although few in numbers) were indeed those with the higher MGR. B2 enterotype was mostly observed in patients with T2D at baseline. ### Supplementary data [[SP4.pdf]](pending:yes) ### Gene richness worsens with aggravated obesity We further aimed to gain insight of MGR in a broader BMI range spanning overweight to morbid obesity by examining its distribution and bioclinical relationships. For this, we pooled clinical information from the MB and MO (overweight/moderate obesity) cohorts examined in our centre5 (figure 1A). Compared with MO patients, the MB patients were younger, all women who displayed worse body composition (ie, increased total-fat and trunk-fat mass) and had more frequent cardiometabolic complications: T2D, hypertension (HTA), increased insulin resistance (at fasting and during the OGTT), and increased C reactive protein (CRP) and interleukin-6 (IL6) (table 1). View this table: [Table 1](http://gut.bmj.com/content/68/1/70/T1) Table 1 MB and MO patients’ baseline clinical characteristics In the entire population (MB+MO, n=110 subjects, BMI (26–61 kg/m²), fat-mass (16.5–81 kg)), MGR was inversely correlated with fat-mass (p=0.0002), leptin (p=0.0072), fasting insulin (p=0.00019), homeostatic model assessment insulin resistance (HOMA-IR) (p=0.00005), triglyceride levels (p=0.0024) and systemic inflammation (IL6 and CRP (p=0.019 and p=0.038)). MGR decreased from MO to AGB to RYGB patients, respectively, and was significantly and inversely correlated with BMI, total-fat mass and trunk-fat mass (Figure 2A), and positively associated with gynoid-fat distribution (p=0.037). MGR decreased with HTA and its severity (evidenced by drugs number to achieve normal blood pressure), glucose intolerance and T2D (Figure 2B). MGR negatively correlated with glucose intolerance-related parameters (OGTT glucose AUC and OGTT Stumvoll Index) and subcutaneous adipocyte volume, and positively associated with adiponectin (Figure 2C). There was no gender effect on MGR, confirming previous observations.4 5 ![Figure 2](http://gut.bmj.com/https://gut.bmj.com/content/gutjnl/68/1/70/F2.medium.gif) [Figure 2](http://gut.bmj.com/content/68/1/70/F2) Figure 2 Links between MGR and bioclinical characteristics in MO+MB subjects. (A) MGR relationships with anthropometric parameters. (B) MGR relation with metabolic comorbidities (hypertension and hypertension treatments and diabetes); N, non-diabetes; IG, glucose intolerance; D, diabetes. (C) MGR relation with OGTT-derived glucose tolerance parameters (AUC of glucose after OGTT with 75 g glucose and Stumvoll Index), adiponectin and adipocyte volume. Pearson’s correlations are performed (p=p value, q=FDR and r2; statistics include linear models (lm), Pearson’s and Spearman’s correlations, t-test and Kruskal-Wallis when appropriate. AGB, adjustable gastric banding; AUC, area under the curve; DXA, X-ray absorptiometry; MB, Microbaria; MGR, microbial gene richness; MO, MICRO-Obes; OGTT, oral glucose tolerance test; RYGB, Roux-en-Y-gastric bypass.  Among these subjects, we considered all 786 MGS, which represent coabundant groups of genes with at least 500 genes as described.21 The association between corpulence and the gut microbiome was also observed at the level of MGS abundance with a principal coordinates analysis (PCoA) analysis. The first two principal components described 23% of the total variance and the second component mostly associated with MGR (online supplementary figure 2A,B), which demonstrates important ecosystem differences according to the degree of obesity and richness. From overweight to morbid obesity, the loss of MGR is linked with adverse body composition, adipocyte hypertrophy and overt metabolic complications. ### Supplementary data [[SP5.pdf]](pending:yes) ### Richness-linked metagenomics species associate with metabolic deteriorations in severe obesity We examined whether some of the 786 MGS21 were specifically associated with parameters linked to clinical phenotypes in severe obesity. Out of this list, about 29% (n=226) significantly associated with MGR (FDR<0.05). We focused on the most MGR-correlated MGS (n=78 with FDR<0.001; figure 3A and online supplementary table 3), of which only 18 were previously found associated with LGC4 in less obese individuals. Whereas the vast majority of these 78 MGS associated positively with MGR (r>0.47), three correlated negatively (r<−0.51); 19 of them were annotated at the species level. Enrichment analysis (Fisher’s test), compared with the overall MGS catalogue (n=786), indicates Firmicutes (FDR<6.7e-05) as the most prevalent phylum associated with MGR and Clostridiales most prevalent at the order level (FDR<5.5e-06). Importantly, we confirmed this MGS signature of low MGR in the ATOX independent cohort composed of severely obese individuals who underwent RYGB (figure 1A). Most of these MGS (50/78) associated with low MGR (online supplementary figure 3). We also confirmed the significant association between low MGR and increasing BMI, and trunk-fat mass. Likewise, MGR was significantly lower in patients with comorbidities (T2D, HTA, use of antihypertensive drugs) (data not shown). ### Supplementary data [[SP6.pdf]](pending:yes) ### Supplementary data [[SP7.pdf]](pending:yes) ![Figure 3](http://gut.bmj.com/https://gut.bmj.com/content/gutjnl/68/1/70/F3.medium.gif) [Figure 3](http://gut.bmj.com/content/68/1/70/F3) Figure 3 MGR-associated MGS at baseline. (A) Heatmap of Spearman’s pairwise correlation coefficients between MGR-associated MGS abundance and metabolic variables (body composition and corpulence and metabolic traits) and MGR-associated serum metabolites. (B) Venn diagram of metabolic parameters associated with MGR-related MGS. (C) Heatmap of Spearman’s pairwise correlation coefficients between metabolic phenotypes and targeted serum metabolites. P value significance denoted by * and FDR significance by #. BMI, body mass index; HbA1c, haemoglobin A1c; HTA, hypertension; MGR, microbial gene richness; MGS, metagenomic species.  We found relevant associations between these 78 MGR-linked MGS and clinical variables (online supplementary table 3): five and seven MGS were significantly associated with BMI and fat-mass, respectively (two of them resisted multiple testing: *GU:373 Coprococcus_sp5* and *GU:115 Eubacterium\_sp_CAG_115*). Sixteen MGS were associated with trunk-fat distribution, including the two BMI-associated MGS, *GU:373 Coprococcus_sp5* and *GU:115 Eubacterium_sp_CAG_115* (figure 3A), and a few were associated with either T2D, HTA or metabolic syndrome, as shown in figure 3B. Looking at the overall patterns of MGS–phenotype associations, we observed relevant MGS subset positively associated with metabolic parameters and corpulence traits, such as total and trunk-fat mass, triglyceride and haemoglobin A1c (Hba1c) (which included *GU:6 Bacteroides vulgatus*, *GU:183 Erysipelatoclostridium ramosum* and *GU:373 Coprococcus_sp5*, the latter was also associated with a cluster of clinical comorbidities (metabolic syndrome, HTA, T2D)), and one cluster negatively associated with BMI, total-fat and trunk-fat, triglycerides and Hba1c (*GU:115 Eubacterium_sp_CAG_115*, *GU:121 Ruminococcaceae bacterium* and *GU:82 Faecalibacterium_6*) (figure 3A). Five MGS from the Firmicutes phylum were associated with three distinct metabolic alterations (T2D, metabolic syndrome and HTA), namely *GU:373 Coprococcus_sp5*, *GU:195 Faecalibacterium 1*, *GU:66 Lachnospiraceae*, *GU:82 Faecalibacterium 6*, *GU:86 Eubacterium sp CAG:86* and *GU:163 Clostridiales* (online supplementary table 3). Despite its smaller sample size, we observed similar trends of associations, in the confirmation cohort (ATOX group), between MGR-linked MGS and metabolic clinical variables: *GU:6 Bacteroides vulgatus* was positively associated with Hba1c (online supplementary figure 4). ### Supplementary data [[SP8.pdf]](pending:yes) In severe obesity, among the most MGR-linked MGS, 50% are associated with metabolic variables, among which 20.5% are associated with both adverse body composition and metabolic alterations. ### Metabolites associated with MGR and related bacterial functions At baseline, we identified nine serum metabolites (4% of the measured metabolites) significantly associated with MGR (Spearman’s correlations, FDR<0.05) (online supplementary table 4 and online supplementary figure 5). One metabolite (*glutarate*) correlated negatively (r=−0.4; p<0.0017) while eight metabolites (*3-methoxyphenylacetic acid*, *phloretate*, *hippurate*, *3-hydroxyphenylacetate*, *L-histidine* and three unknown) correlated positively with MGR (0.4