Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Physiology and Biochemistry

The visceral adipose tissue bacterial microbiota provides a signature of obesity based on inferred metagenomic functions

A Correction to this article was published on 08 September 2023

This article has been updated

Abstract

Background

Metabolic inflammation mediated obesity requires bacterial molecules to trigger immune and adipose cells leading to inflammation and adipose depot development. In addition to the well-established gut microbiota dysbiosis, a leaky gut has been identified in patients with obesity and animal models, characterized by the presence of a tissue microbiota in the adipose fat pads.

Methods

To determine its potential role, we sequenced the bacterial 16 S rRNA genes in the visceral adipose depot of patients with obesity. Taking great care (surgical, biochemical, and bioinformatic) to avoid environmental contaminants. We performed statistical discriminant analyses to identify specific signatures and constructed network of interactions between variables.

Results

The data showed that a specific 16SrRNA gene signature was composed of numerous bacterial families discriminating between lean versus patients with obesity and people with severe obesity. The main discriminant families were Burkholderiaceae, Yearsiniaceae, and Xanthomonadaceae, all of which were gram-negative. Interestingly, the Morganellaceae were totally absent from people without obesity while preponderant in all in patients with obesity. To generate hypotheses regarding their potential role, we inferred metabolic pathways from the 16SrRNA gene signatures. We identified several pathways associated with adenosyl-cobalamine previously described to be linked with adipose tissue development. We further identified chorismate biosynthesis, which is involved in aromatic amino-acid metabolism and could play a role in fat pad development. This innovative approach generates novel hypotheses regarding the gut to adipose tissue axis.

Conclusions

This innovative approach generates novel hypotheses regarding the gut to adipose tissue axis in obesity and notably the potential role of tissue microbiota.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Principal component analysis of the clinical features of the cohort of patients.
Fig. 2: Alpha and beta diversity indexes.
Fig. 3: Individual patient relative abundances and classification.
Fig. 4: Multivariate analyses.
Fig. 5: Summary of specific group signatures.
Fig. 6: Multivariate analyses of the Pathways inferred from OTUs.

Data availability

MiSeq 16 S rRNA gene sequences were deposited under the primary accession number PRJEB53362 and a secondary number ERP138154 on June 30th 2022.

Change history

References

  1. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–6.

    Article  PubMed  Google Scholar 

  2. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31.

    Article  PubMed  Google Scholar 

  3. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470–81.

    Article  CAS  PubMed  Google Scholar 

  4. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–72.

    Article  CAS  PubMed  Google Scholar 

  5. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177–85.

    Article  CAS  PubMed  Google Scholar 

  6. Luche E, Cousin B, Garidou L, Serino M, Waget A, Barreau C, et al. Metabolic endotoxemia directly increases the proliferation of adipocyte precursors at the onset of metabolic diseases through a CD14-dependent mechanism. Mol Metab. 2013;2:281–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bischoff SC, Barbara G, Buurman W, Ockhuizen T, Schulzke JD, Serino M, et al. Intestinal permeability-a new target for disease prevention and therapy. BMC Gastroenterol. 2014;14:189.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermudez-Humaran LG, et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol Med. 2011;3:559–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lluch J, Servant F, Paisse S, Valle C, Valiere S, Kuchly C, et al. The characterization of novel tissue microbiota using an optimized 16S metagenomic sequencing pipeline. PLoS One. 2015;10:e0142334.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Paisse S, Valle C, Servant F, Courtney M, Burcelin R, Amar J, et al. Comprehensive description of blood microbiome from healthy donors assessed by 16S targeted metagenomic sequencing. Transfusion. 2016;56:1138–47.

    Article  CAS  PubMed  Google Scholar 

  11. Burcelin R, Serino M, Chabo C, Garidou L, Pomie C, Courtney M, et al. Metagenome and metabolism: the tissue microbiota hypothesis. Diabetes Obes Metab. 2013;15:61–70.

    Article  CAS  PubMed  Google Scholar 

  12. Massier L, Chakaroun R, Tabei S, Crane A, Didt KD, Fallmann J, et al. Adipose tissue derived bacteria are associated with inflammation in obesity and type 2 diabetes. Gut. 2020;69:1796–806.

    Article  CAS  PubMed  Google Scholar 

  13. Anhe FF, Jensen BAH, Varin TV, Servant F, Van Blerk S, Richard D, et al. Type 2 diabetes influences bacterial tissue compartmentalisation in human obesity. Nat Metab. 2020;2:233–42.

    Article  PubMed  Google Scholar 

  14. Escudie F, Auer L, Bernard M, Mariadassou M, Cauquil L, Vidal K, et al. FROGS: find, rapidly, OTUs with galaxy solution. Bioinformatics (Oxford, England). 2018;34:1287–94.

    CAS  PubMed  Google Scholar 

  15. Schierwagen R, Alvarez-Silva C, Madsen MSA, Kolbe CC, Meyer C, Thomas D, et al. Circulating microbiome in blood of different circulatory compartments. Gut. 2019;68:578–80.

    Article  CAS  PubMed  Google Scholar 

  16. Schierwagen R, Alvarez-Silva C, Servant F, Trebicka J, Lelouvier B, Arumugam M. Trust is good, control is better: technical considerations in blood microbiome analysis. Gut. 2020;69:1362–3.

    Article  PubMed  Google Scholar 

  17. Lelouvier B, Servant F, Delobel P, Courtney M, Elbaz M, Amar J. Identification by highly sensitive 16S metagenomic sequencing of an unusual case of polymicrobial bacteremia. J Infect. 2017;75:278–80.

    Article  PubMed  Google Scholar 

  18. Lelouvier B, Servant F, Paisse S, Brunet AC, Benyahya S, Serino M, et al. Changes in blood microbiota profiles associated with liver fibrosis in obese patients: a pilot analysis. Hepatology. 2017;65:1776–7.

    Article  PubMed  Google Scholar 

  19. Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO, Moffatt MF, et al. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 2014;12:87.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Laurence M, Hatzis C, Brash DE. Common contaminants in next-generation sequencing that hinder discovery of low-abundance microbes. PLoS One. 2014;9:e97876.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Fernandes AD, Reid JN, Macklaim JM, McMurrough TA, Edgell DR, Gloor GB. Unifying the analysis of high-throughput sequencing datasets: characterizing RNA-seq, 16S rRNA gene sequencing and selective growth experiments by compositional data analysis. Microbiome. 2014;2:15.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Denou E, Lolmede K, Garidou L, Pomie C, Chabo C, Lau TC, et al. Defective NOD2 peptidoglycan sensing promotes diet-induced inflammation, dysbiosis, and insulin resistance. EMBO Mol Med. 2015;7:259–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fu A, Yao B, Dong T, Chen Y, Yao J, Liu Y, et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell. 2022;185:1356–72.

    Article  CAS  PubMed  Google Scholar 

  24. Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018;8:403–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jervis-Bardy J, Leong LE, Marri S, Smith RJ, Choo JM, Smith-Vaughan HC, et al. Deriving accurate microbiota profiles from human samples with low bacterial content through post-sequencing processing of Illumina MiSeq data. Microbiome. 2015;3:19.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357:1156–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Geller LT, Straussman R. Intratumoral bacteria may elicit chemoresistance by metabolizing anticancer agents. Mol Cell Oncol. 2018;5:e1405139.

    Article  PubMed  Google Scholar 

  28. Costantini L, Magno S, Albanese D, Donati C, Molinari R, Filippone A, et al. Characterization of human breast tissue microbiota from core needle biopsies through the analysis of multi hypervariable 16S-rRNA gene regions. Sci Rep. 2018;8:16893.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Amar J, Serino M, Lange C, Chabo C, Iacovoni J, Mondot S, et al. Involvement of tissue bacteria in the onset of diabetes in humans: evidence for a concept. Diabetologia. 2011;54:3055–61.

    Article  CAS  PubMed  Google Scholar 

  30. Massier L, Bluher M, Kovacs P, Chakaroun RM. Impaired intestinal barrier and tissue bacteria: pathomechanisms for metabolic diseases. Front Endocrinol (Lausanne). 2021;12:616506.

    Article  PubMed  Google Scholar 

  31. Rus M, Licker M, Musuroi C, Muntean D, Vulpie S, Magiar O, et al. Association of proteus mirabilis and providencia stuartii infections with diabetes. Medicina (Kaunas). 2022;58:4751–61.

    Google Scholar 

  32. Koutoukidis DA, Jebb SA, Zimmerman M, Otunla A, Henry JA, Ferrey A, et al. The association of weight loss with changes in the gut microbiota diversity, composition, and intestinal permeability: a systematic review and meta-analysis. Gut Microbes. 2022;14:2020068.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Moreira AP, Texeira TF, Ferreira AB, Peluzio Mdo C, Alfenas, Rde C. Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br J Nutr. 2012;108:801–9.

    Article  CAS  PubMed  Google Scholar 

  34. Fouts DE, Torralba M, Nelson KE, Brenner DA, Schnabl B. Bacterial translocation and changes in the intestinal microbiome in mouse models of liver disease. J Hepatol. 2012;56:1283–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Stoidis CN, Misiakos EP, Patapis P, Fotiadis CI, Spyropoulos BG. Potential benefits of pro- and prebiotics on intestinal mucosal immunity and intestinal barrier in short bowel syndrome. Nutr Res Rev. 2010;24:1–9.

    Google Scholar 

  36. Muccioli GG, Naslain D, Backhed F, Reigstad CS, Lambert DM, Delzenne NM, et al. The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol. 2010;6:392.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Garidou L, Pomie C, Klopp P, Waget A, Charpentier J, Aloulou M, et al. The gut microbiota regulates intestinal CD4 T cells expressing rorgammat and controls metabolic disease. Cell Metab. 2015;22:100–12.

    Article  CAS  PubMed  Google Scholar 

  38. Carlos D, Perez MM, Leite JA, Rocha FA, Martins LMS, Pereira CA, et al. NOD2 deficiency promotes intestinal CD4+T lymphocyte imbalance, metainflammation, and aggravates type 2 diabetes in murine model. Front Immunol. 2020;11:1265.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ho-Plagaro A, Santiago-Fernandez C, Garcia-Serrano S, Rodriguez C, Garrido-Sanchez L, Escamilla A, et al. A lower duodenal immune response is associated with an increase of insulin resistance in patients with morbid obesity. Int J Obes (Lond). 2020;44:340–52.

    Article  CAS  PubMed  Google Scholar 

  40. Kim J, Choi JH, Oh T, Ahn B, Unno T. Codium fragile ameliorates high-fat diet-induced metabolism by modulating the gut microbiota in mice. Nutrients. 2020;12:1848.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Saben J, Sims C, Piccolo B, Andres A. Maternal adiposity alters the human milk metabolome: associations between nonglucose monosaccharides and infant adiposity. Am J Clin Nutr. 2020;11:1228–39.

    Article  Google Scholar 

  42. Schoppel K, Trachtmann N, Mittermeier F, Sprenger GA, Weuster-Botz D. Metabolic control analysis of L-tryptophan producing Escherichia coli applying targeted perturbation with shikimate. Bioprocess Biosyst Eng. 2021;44:2591–613.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Laurans L, Venteclef N, Haddad Y, Chajadine M, Alzaid F, Metghalchi S, et al. Genetic deficiency of indoleamine 2,3-dioxygenase promotes gut microbiota-mediated metabolic health. Nat Med. 2018;8:1113–20.

    Article  Google Scholar 

  44. Ratshikombo T, Goedecke JH, Soboyisi M, Kufe C, Makura-Kankwende CBT, Masemola M, et al. Sex differences in the associations of nutrient patterns with total and regional adiposity: a study of middle-aged black South African men and women. Nutrients. 2021;13:e2815.

    Article  Google Scholar 

  45. Mardinoglu A, Wu H, Bjornson E, Zhang C, Hakkarainen A, Rasanen SM, et al. An integrated understanding of the rapid metabolic benefits of a carbohydrate-restricted diet on hepatic steatosis in humans. Cell Metab. 2018;27:559–571.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sun WL, Hua S, Li XY, Shen L, Wu H, Ji HF. Microbially produced vitamin B12 contributes to the lipid-lowering effect of silymarin. Nat Commun. 2023;14:477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Oliai Araghi S, Braun KVE, van der Velde N, van Dijk SC, van Schoor NM, Zillikens MC, et al. B-vitamins and body composition: integrating observational and experimental evidence from the B-PROOF study. Eur J Nutr. 2020;59:1253–62.

    Article  CAS  PubMed  Google Scholar 

  48. Rubini E, Schenkelaars N, Rousian M, Sinclair KD, Wekema L, Faas MM, et al. Maternal obesity during pregnancy leads to derangements in one-carbon metabolism and the gut microbiota: implications for fetal development and offspring wellbeing. Am J Obstet Gynecol. 2022;227:392–400.

    Article  CAS  PubMed  Google Scholar 

  49. Ferrer M, Ruiz A, Lanza F, Haange SB, Oberbach A, Till H, et al. Microbiota from the distal guts of lean and obese adolescents exhibit partial functional redundancy besides clear differences in community structure. Environ Microbiol. 2013;15:211–26.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We wish to extend our gratitude to the study participants, investigators, monitors and study nurses who enabled this study. We are grateful to Bogdana Dorcioman MD and the corresponding team from the Laboratory Department and Emergency of the Mures County Hospital, who performed blood analysis and who provided some technical help.

Funding

This study was supported by subsides from the Agence Nationale de la Recherche, the Région Midi Pyrénées to R.B. This research program was partly funded as well by VAIOMER SAS (project no. 6869/12.06.2014) through the University of Medicine and Pharmacy Tirgu Mures. This work was supported by EU-FP7 FLORINASH (Health-F2-2009-241913) to RB, MF, JMFR. “This work was supported by the University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș Research Grant number 294/4/14.01.2020”. A subside was allocated to Jiuwen Sun and Alberic Germain from the Fondation Française de Recherche en Diabétologie (FFRD) and from the Société Francophone du Diabète (SFD) and European Foundation for the Study of Diabetes (EF).

Author information

Authors and Affiliations

Authors

Contributions

The study was designed by RB, JMFR, RN, and MF. The experiments and analyses were done by JS, GK, and FS. The manuscript was written and data analyzed by JS, AG, RB, FS, BL, MF, JMFR, and AB.

Corresponding author

Correspondence to Rémy Burcelin.

Ethics declarations

Competing interests

RB received honorarium from Vaiomer and had shares. BL and FS are employees of Vaiomer. The other authors have no competing interests.

Ethics approval and consent to participate

A monocentric observational study was conducted in the Second Department of Surgery, Emergency Mureş County Hospital of Romania. All research procedures performed in this study were in strict accordance with a pre-defined protocol and adhered to the Good Clinical Practice guidelines and the Declaration of Helsinki. The study was approved by the Coordinating Ethics Committee of the Emergency Mureş County Hospital, Romania (registration 4065/2014): TirguMECCH ROLIVER Prospective Cohort for the Identification of Liver and Adipose Tissue Microbiota, registration 4065/2014. Registered 01 01 2014. All participants provided informed consent prior to participation. The patients who gave their consent to perform an adipose tissue biopsy during the procedure were eligible.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, J., Germain, A., Kaglan, G. et al. The visceral adipose tissue bacterial microbiota provides a signature of obesity based on inferred metagenomic functions. Int J Obes 47, 1008–1022 (2023). https://doi.org/10.1038/s41366-023-01341-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41366-023-01341-1

Search

Quick links