Schellekens_BLongum_Obesity_Stress_EBIOM_103176 (003)

14

H. Schellekens et al. / EBioMedicine xxx (xxxx) 103176

showed the ability to modulate ghrelin signalling invitro [37], have sim- ilar effects in obese individuals. Furthermore, our data reveal that B. longum APC1472 decreased fasting corticosterone levels in HFD-induced obese mice, indicating the downregulation of the hypothalamic-pituitary-adrenal (HPA) axis. In line with these results, B. longum APC1472 reduced cortisol awakening responses in obese individuals. Dysregulation of the HPA axis, which is colloquially seen as the “ body's stress system ” , is a risk factor for obe- sity-related conditions such as cardiovascular disease, insulin resistance and type 2 diabetes [93]. Hence, the stress hormone cortisol (corticos- terone in rodents), which is central in the HPA axis, has been shown to promote the accumulation of fat cells and weight gain [93] and to regulate the function of pancreatic α and β cells affecting glucagon and insulin secretion [94]. As such, even though no changes were observed in insulin, the changes in cortisol awakening responses could indicate that the HPA axis has contributed to the B. longum APC1472-induced decrease in fasting blood glucose. Furthermore, the HPA axis is also af- fected by ghrelin, indicating that the observed changes in ghrelin could have also contributed to the changes in cortisol [95,99]. Finally, we investigated the effects of B. longum APC1472 treatment on gut microbiota composition. Overall, B. longum APC1472 treatment did not have a major impact on microbiota composition other than the partial restoration of Bifidobacterium levels in HFD-fed mice. These find- ings are in line with the effects of B. longum APC1472 on healthy human overweight and obese individuals and with other investigations on obe- sity using different probiotics strains, where major rearrangements on microbiota composition were also not observed [96,97]. Of note, while the modulation of ghrelin receptor signalling by B. longum APC1472 strain may have contributed to an improved metabolic profile, we cannot rule out other beneficial anti-obesity effects. As such, future studies are warranted further investigating the mechanisms and metabolites through which B. longum APC1472 modulates host glucose homeostasis, with a focus on the ghrelinergic system. In conclusion, we have demonstrated positive anti-obesity effects of the novel B. longum APC1472 strain in HFD-induced obese mice and a partial translation of these positive effects of B. longum APC1472 sup- plementation in otherwise healthy overweight and obese individuals. In particular, we show the promising potential of B. longum APC1472 to be developed as a valuable supplement in reducing specific markers of obe- sity, possibly via the ghrelinergic system. Most notably, the decrease in fasting plasma glucose induced by B. longum APC1472 may have clini- cally significant health implications for prediabetic and type 2 diabetes mellitus populations in particular. Data sharing Deidentified data and related documents will be made available upon request. Disclosure This research was funded in part by Science Foundation Ireland in the form of a Research Centre grant (SFI/12/RC/2273) to APC Micro- biome Ireland and by a research grant from Cremo S.A. J.F.C and T.G.D have research support from Mead Johnson, Cremo, 4D Pharma, Dupont, and Nutricia. J.F.C, T.G.D and P.D.C. have spoken at meetings sponsored by food and pharmaceutical companies. All other authors report no con-

signed the human intervention study and interpreted the data and con- tributed to the writing of the manuscript. Avery M. performed DNA iso- lations, sequenced the DNA and assisted with the SCFA quantifications of the human intervention study. Amy M. assisted with the DNA isola- tions of the human intervention study. C.S. performed the SCFA quan- tifications of the human intervention study. K.B. interpreted the nutri- tional data. T.F.S.B. and F.F. performed the bioinformatics for the mi- crobiota for the human intervention study. K.R. interpreted the data and contributed to writing the manuscript. A.G. assisted with running the preclinical study. S.A. performed library preparation and SCFA quantifi- cations of the preclinical study. K.M. performed the bioinformatics of the preclinical study. M.V.P assisted with running the preclinical study. M.M.P. performed the corticosterone quantifications. P.R., B.L.R., C.S., T.G.D. and J.F.C. contributed to the design of the study, interpreted the data and contributed to the manuscript. Finally, T. G. D was Chief Inves- tigator on the clinical part of the study. Uncited references [98 – 101]. Acknowledgements The authors would like to thank Gillian DunnGalvin, Kelly Seamans, Ann-Marie Crowley, Andrea Doolan and Barry Skillington at Atlantia Food Clinical Trials (Cork, Ireland) for the execution of the human study (study reference: AFRCO-088). The authors would like to thank Lorenzo Miorelli from Sacco (Sacco SRI, Italy) for coordinating B. longum fer- mentation and initial stability investigations. Furthermore, the authors would like to thank Tara Foley, Naomi Hoevenaars and Patrick Fitzger- ald at University College Cork and APC Microbiome Ireland, for their help throughout the preclinical study, Fiona Crispie (Teagasc), Fiona Fouhy (Teagasc) and Brian Healy (APC Microbiome Ireland) for assis- tance with the sequencing of human samples, and Susan Rafferty-McAr- dle (APC Microbiome Ireland) for her help in the experimental design of the human intervention study. Supplementary materials Supplementary material associated with this article can be found, in the online version, at doi:10.1016/j.ebiom.2020.103176. Footnotes Article Footnotes For submission to EBioMedicine References [1] WHO. Obesity and overweight - key facts. 2018. Available from: https://www. who.int/en/news-room/fact-sheets/detail/obesity-and-overweight. [2] A. Tchernof, J.P Despres Pathophysiology of human visceral obesity: an update. Physiol Rev 2013;93(1):359 – 404. [3] V. Narayanaswami, L.P Dwoskin Obesity: current and potential pharmacotherapeutics and targets. Pharmacol Ther 2017;170:116 – 147. [4] C. Torres-Fuentes, H. Schellekens, T.G. Dinan, J.F Cryan A natural solution for obesity: bioactives for the prevention and treatment of weight gain. A review. Nutr Neurosci 2014. [5] S.R. Bloom, F.P. Kuhajda, I. Laher, X. Pi-Sunyer, G.V. Ronnett, T.M. Tan, et al. The obesity epidemic: pharmacological challenges. Mol Interv 2008;8(2):82 – 98. [6] K.M. Fock, J Khoo Diet and exercise in management of obesity and overweight. J Gastroenterol Hepatol 2013;28(Suppl 4):59 – 63. [7] J.K. DiBaise, D.N. Frank, R Mathur Impact of the gut microbiota on the development of obesity: current concepts. Am J Gastroenterol Suppl 2012;1(1):22 – 27. [8] E. Patterson, P.M. Ryan, J.F. Cryan, T.G. Dinan, R.P. Ross, G.F. Fitzgerald, et al. Gut microbiota, obesity and diabetes. Postgrad Med J 2016;92(1087):286 – 300. [9] C. Torres-Fuentes, H. Schellekens, T.G. Dinan, J.F Cryan The microbiota-gut-brain axis in obesity. Lancet Gastroenterol Hepatol 2017. [10] M. van de Wouw, H. Schellekens, T.G. Dinan, J.F Cryan Microbiota-gut-brain axis: modulator of host metabolism and appetite. J Nutr 2017;147(5):727 – 745. [11] P.D. Cani, M. Van Hul, C. Lefort, C. Depommier, M. Rastelli, A Everard Microbial regulation of organismal energy homeostasis. Nat Metab 2019. [12] M. Rosenbaum, R. Knight, R.L Leibel The gut microbiota in human energy homeostasis and obesity. Trends Endocrinol Metab 2015;26(9):493 – 501.

flicts of interest. Contributions

H.S. contributed to the design of the study, interpreted the data and ledthe writing of the manuscript. C.T.F. contributed to the design of the preclinical study, performed the preclinical study and contributed to the writing of the manuscript of the preclinical study. M.vdW. per- formed ELISAs, MSD assays, interpreted the data and contributed to the writing of the manuscript of the human intervention study. C.M.L.S. de

Powered by