The Intestinal Microbiota and Short-Chain Fatty Acids in Association with Advanced Metrics of Glycemia and Adiposity Among Young Adults with Type 1 Diabetes and Overweight or Obesity

Daria Igudesman, Jamie Crandell, Karen D Corbin, Franklin Muntis, Dessi P Zaharieva, Anna Casu, Joan M Thomas, Cynthia M Bulik, Ian M Carroll, Brian W Pence, Richard E Pratley, Michael R Kosorok, David M Maahs, Elizabeth J Mayer-Davis, Daria Igudesman, Jamie Crandell, Karen D Corbin, Franklin Muntis, Dessi P Zaharieva, Anna Casu, Joan M Thomas, Cynthia M Bulik, Ian M Carroll, Brian W Pence, Richard E Pratley, Michael R Kosorok, David M Maahs, Elizabeth J Mayer-Davis

Abstract

Background: Comanagement of glycemia and adiposity is the cornerstone of cardiometabolic risk reduction in type 1 diabetes (T1D), but targets are often not met. The intestinal microbiota and microbiota-derived short-chain fatty acids (SCFAs) influence glycemia and adiposity but have not been sufficiently investigated in longstanding T1D.

Objectives: We evaluated the hypothesis that an increased abundance of SCFA-producing gut microbes, fecal SCFAs, and intestinal microbial diversity were associated with improved glycemia but increased adiposity in young adults with longstanding T1D.

Methods: Participants provided stool samples at ≤4 time points (NCT03651622: https://ichgcp.net/clinical-trials-registry/NCT03651622). Sequencing of the 16S ribosomal RNA gene measured abundances of SCFA-producing intestinal microbes. GC-MS measured total and specific SCFAs (acetate, butyrate, propionate). DXA (body fat percentage and percentage lean mass) and anthropometrics (BMI) measured adiposity. Continuous glucose monitoring [percentage of time in range (70-180 mg/dL), above range (>180 mg/dL), and below range (54-69 mg/dL)] and glycated hemoglobin (i.e., HbA1c) assessed glycemia. Adjusted and Bonferroni-corrected generalized estimating equations modeled the associations of SCFA-producing gut microbes, fecal SCFAs, and intestinal microbial diversity with glycemia and adiposity. COVID-19 interrupted data collection, so models were repeated restricted to pre-COVID-19 visits.

Results: Data were available for ≤45 participants at 101 visits (including 40 participants at 54 visits pre-COVID-19). Abundance of Eubacterium hallii was associated inversely with BMI (all data). Pre-COVID-19, increased fecal propionate was associated with increased percentage of time above range and reduced percentage of time in target and below range; and abundances of 3 SCFA-producing taxa (Ruminococcus gnavus, Eubacterium ventriosum, and Lachnospira) were associated inversely with body fat percentage, of which two microbes were positively associated with percentage lean mass. Abundance of Anaerostipes was associated with reduced percentage of time in range (all data) and with increased body fat percentage and reduced percentage lean mass (pre-COVID-19).

Conclusions: Unexpectedly, fecal propionate was associated with detriment to glycemia, whereas most SCFA-producing intestinal microbes were associated with benefit to adiposity. Future studies should confirm these associations and determine their potential causal linkages in T1D.This study is registered at clinical.trials.gov (NCT03651622; https://ichgcp.net/clinical-trials-registry/NCT03651622).

Keywords: adiposity; body mass index; continuous glucose monitoring; dual-energy X-ray absorptiometry; glycemia; gut microbiota; hemoglobin A1c; short-chain fatty acids; type 1 diabetes.

© The Author(s) 2022. Published by Oxford University Press on behalf of the American Society for Nutrition.

Figures

FIGURE 1
FIGURE 1
Heatmaps with standardized β estimates from covariate-adjusted GEE models for associations of SCFA-producing microbes, fecal SCFAs, and α-diversity with glycemia and adiposity. (A) Results using all available data; (B) Results restricted to pre-COVID-19 data.P values were Bonferroni corrected and statistically significant at P  < 0.1 (denoted by asterisks). Units are micromoles per gram for SCFAs, normalized abundance for gen us-level intestinal microbes, and the number of unique taxa from the phylum to the genus level for intestinal microbial diversity. DXA was only performed prior to COVID-19. BFP, body fat percentage; GEE, generalized estimating equation; HbA1c, glycated hemoglobin; PLM, percentage lean mass; TAB, percentage of time above target glucose range (>180 mg/dL); TBR, percentage of time below target glucose range (54–69 mg/dL); TIR, percentage of time in target glucose range (70–180 mg/dL).
FIGURE 2
FIGURE 2
Scatterplots of raw pre-COVID-19 data for associations of fecal SCFAs with measures of glycemia that were statistically significant after adjustment for potential confounders and correction for multiple hypothesis testing. (A, B) Increased total fecal SCFAs and fecal propionate were associated with reduced percentage of time in target glucose range. (C, D) Increased fecal propionate was associated with reduced percentage of time above target glucose range (C) and percentage of time in clinical hypoglycemia (D).

References

    1. De Ferranti SD, De Boer IH, Fonseca Vet al. . Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Circulation. 2014;130(13):1110–30.
    1. Miller KM, Foster NC, Beck RWet al. . Current state of type 1 diabetes treatment in the US: updated data from the T1D Exchange clinic registry. Diabetes Care. 2015;38(6):971–8.
    1. Liu LL, Lawrence JM, Davis Cet al. . Prevalence of overweight and obesity in youth with diabetes in USA: the SEARCH for Diabetes in Youth study. Pediatr Diabetes. 2010;11(1):4–11.
    1. Malik FS, Sauder KA, Isom Set al. . Trends in glycemic control among youth and young adults with diabetes: the SEARCH for Diabetes in Youth study. Diabetes Care. 2022;45(2):285–94.
    1. Wallace AS, Chang AR, Shin J-Iet al. . Obesity and chronic kidney disease in US adults with type 1 and type 2 diabetes mellitus. J Clin Endocrinol Metab. 2022;107(5):1247–56.
    1. Tauschmann M, Hovorka R. Technology in the management of type 1 diabetes mellitus—current status and future prospects. Nat Rev Endocrinol. 2018;14(8):464–75.
    1. Chambers ES, Morrison DJ, Frost G. Control of appetite and energy intake by SCFA: what are the potential underlying mechanisms?. Proc Nutr Soc. 2015;74(3):328–36.
    1. Murri M, Leiva I, Gomez-Zumaquero JMet al. . Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. BMC Med. 2013;11(1):46.
    1. Brown CT, Davis-Richardson AG, Giongo Aet al. . Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One. 2011;6(10):e25792.
    1. de Goffau MC, Luopajärvi K, Knip Met al. . Fecal microbiota composition differs between children with β-cell autoimmunity and those without. Diabetes. 2013;62(4):1238–44.
    1. Canfora EE, Jocken JW, Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol. 2015;11(10):577.
    1. De Groot PF, Belzer C, Aydin Öet al. . Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study. PLoS One. 2017;12(12):e0188475.
    1. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–45.
    1. David LA, Maurice CF, Carmody RNet al. . Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63.
    1. Krajmalnik-Brown R, Ilhan ZE, Kang DW, DiBaise JK. Effects of gut microbes on nutrient absorption and energy regulation. Nutr Clin Pract. 2012;27(2):201–14.
    1. Samuel BS, Shaito A, Motoike Tet al. . Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, GPR41. Proc Natl Acad Sci U S A. 2008;105(43):16767–72.
    1. Kimura I, Ozawa K, Inoue Det al. . The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun. 2013;4(1):1–12.
    1. de Groot P, Nikolic T, Pellegrini Set al. . Faecal microbiota transplantation halts progression of human new-onset type 1 diabetes in a randomised controlled trial. Gut. 2021;70(1):92–105.
    1. den Besten G, Bleeker A, Gerding Aet al. . Short-chain fatty acids protect against high-fat diet–induced obesity via a PPARγ-dependent switch from lipogenesis to fat oxidation. Diabetes. 2015;64(7):2398–408.
    1. Kootte RS, Levin E, Salojärvi Jet al. . Improvement of insulin sensitivity after lean donor feces in metabolic syndrome is driven by baseline intestinal microbiota composition. Cell Metab. 2017;26(4):611–9.e6.
    1. Corbin K. D., Igudesman D., Addala A., Casu A., Crandell J., Kosorok M. al. . Design of the Advancing Care for Type 1 Diabetes and Obesity Network energy metabolism and sequential multiple assignment randomized trial nutrition pilot studies: An integrated approach to develop weight management solutions for individuals with type 1 diabetes. Contemp Clin Trials. 2022;117:106765.
    1. Kidwell KM. SMART designs in cancer research: past, present, and future. Clin Trials. 2014;11(4):445–56.
    1. Lavori PW, Dawson R. Dynamic treatment regimes: practical design considerations. Clin Trials. 2004;1(1):9–20.
    1. Murphy SA. An experimental design for the development of adaptive treatment strategies. Stat Med. 2005;24(10):1455–81.
    1. Battelino T, Danne T, Bergenstal RMet al. . Clinical targets for continuous glucose monitoring data interpretation: recommendations from the International Consensus on Time in Range. Diabetes Care. 2019;42(8):1593–603.
    1. Standiford DA, Morwessel N, Bishop FKet al. . Two-step recruitment process optimizes retention in FLEX clinical trial. Contemp Clin Trials Commun. 2018;12:68–75.
    1. Fouladi F, Glenny EM, Bulik-Sullivan ECet al. . Sequence variant analysis reveals poor cor in microbial taxonomic abundance between humans and mice after gnotobiotic transfer. ISME J. 2020;14(7):1809–20.
    1. Kleiman SC, Glenny EM, Bulik-Sullivan ECet al. . Daily changes in composition and diversity of the intestinal microbiota in patients with anorexia nervosa: a series of three cases. Eur Eat Disord Rev. 2017;25(5):423–7.
    1. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13(7):581.
    1. Lin H, Peddada SD. Analysis of microbial compositions: a review of normalization and differential abundance analysis. NPJ Biofilms Microbiomes. 2020;6:60.
    1. Van den Berge K, Perraudeau F, Soneson Cet al. . Observation weights unlock bulk RNA-seq tools for zero inflation and single-cell applications. Genome Biol. 2018;19(1):24.
    1. Noble EE, Hsu TM, Jones RB, Fodor AA, Goran MI, Kanoski SE. Early-life sugar consumption affects the rat microbiome independently of obesity. J Nutr. 2017;147(1):20–8.
    1. Quast C, Pruesse E, Yilmaz Pet al. . The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2012;41(D1):D590–6.
    1. Willis AD. Rarefaction, alpha diversity, and statistics. Front Microbiol. 2019;10:2407.
    1. Noble EE, Hsu TM, Jones RB, Fodor AA, Goran MI, Kanoski SE. Early-life sugar consumption affects the rat microbiome independently of obesity. J Nutr. 2017;147(1):20–8.
    1. Wang Y, Wang H, Howard AGet al. . Circulating short-chain fatty acids are positively associated with adiposity measures in Chinese adults. Nutrients. 2020;12(7):2127.
    1. Remely M, Aumueller E, Merold Cet al. . Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity. Gene. 2014;537(1):85–92.
    1. Louis P, Young P, Holtrop G, Flint HJ. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene. Environ Microbiol. 2010;12(2):304–14.
    1. van der Lelie D, Oka A, Taghavi Set al. . Rationally designed bacterial consortia to treat chronic immune-mediated colitis and restore intestinal homeostasis. Nat Commun. 2021;12(1):1–17.
    1. Nathan DM, Krause-Steinrauf H, Braffett BHet al. . Comparison of central laboratory HbA1c measurements obtained from a capillary collection versus a standard venous whole blood collection in the GRADE and EDIC studies. PLoS One. 2021;16(11):e0257154.
    1. Posner BM, Smigelski C, Duggal A, Morgan J, Cobb J, Cupples L. Validation of two-dimensional models for estimation of portion size in nutrition research. J Am Diet Assoc. 1992;92((6):):738–41.
    1. Beaton GH, Milner J, Corey Pet al. . Sources of variance in 24-hour dietary recall data: implications for nutrition study design and interpretation. Am J Clin Nutr. 1979;32(12):2546–59.
    1. University of Minnesota . Nutrition Data System for Research. 2014.
    1. Tukey JW. The future of data analysis. Ann Math Stat. 1962;33(1):1–67.
    1. Dewulf EM, Cani PD, Claus SPet al. . Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut. 2013;62(8):1112–21.
    1. Nadal I, Santacruz A, Marcos Aet al. . Shifts in clostridia, bacteroides and immunoglobulin-coating fecal bacteria associated with weight loss in obese adolescents. Int J Obes. 2009;33(7):758–67.
    1. Weiss S, Xu ZZ, Peddada Set al. . Normalization and microbial differential abundance strategies depend upon data characteristics. Microbiome. 2017;5(1):27.
    1. Turnbaugh PJ, Hamady M, Yatsunenko Tet al. . A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480–4.
    1. Faith JJ, Guruge JL, Charbonneau Met al. . The long-term stability of the human gut microbiota. Science. 2013;341(6141):1237439.
    1. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. 2009;326(5960):1694–7.
    1. Hanley JA, Negassa A, MDd E, Forrester JE. Statistical analysis of correlated data using generalized estimating equations: an orientation. Am J Epidemiol. 2003;157(4):364–75.
    1. Mantel N. Why stepdown procedures in variable selection. Technometrics. 1970;12(3):621–5.
    1. Bland JM, Altman DG. Multiple significance tests: the Bonferroni method. BMJ. 1995;310(6973):170.
    1. Robertson MD, Bickerton AS, Dennis AL, Vidal H, Frayn KN. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am J Clin Nutr. 2005;82(3):559–67.
    1. De Groot P, Scheithauer T, Bakker GJet al. . Donor metabolic characteristics drive effects of faecal microbiota transplantation on recipient insulin sensitivity, energy expenditure and intestinal transit time. Gut. 2020;69(3):502–12.
    1. Canfora EE, van der Beek CM, Jocken JWet al. . Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci Rep. 2017;7(1):1–12.
    1. Vrieze A, Van Nood E, Holleman Fet al. . Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143(4):913–6.e7.
    1. Singh N, Gurav A, Sivaprakasam Set al. . Activation of GPR109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40(1):128–39.
    1. Tedelind S, Westberg F, Kjerrulf M, Vidal A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J Gastroenterol. 2007;13(20):2826.
    1. Yoshida H, Ishii M, Akagawa M. Propionate suppresses hepatic gluconeogenesis via GPR43/AMPK signaling pathway. Arch Biochem Biophys. 2019;672:108057.
    1. De Vadder F, Kovatcheva-Datchary P, Goncalves Det al. . Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell. 2014;156(1-2):84–96.
    1. Gerich JE, Langlois M, Noacco C, Karam JH, Forsham PH. Lack of glucagon response to hypoglycemia in diabetes: evidence for an intrinsic pancreatic alpha cell defect. Science. 1973;182(4108):171–3.
    1. Darzi J, Frost GS, Robertson MD. Effects of a novel propionate-rich sourdough bread on appetite and food intake. Eur J Clin Nutr. 2012;66(7):789–94.
    1. Hoyles L, Snelling T, Umlai U-Ket al. . Microbiome–host systems interactions: protective effects of propionate upon the blood–brain barrier. Microbiome. 2018;6(1):1–13.
    1. Yu EW, Gao L, Stastka Pet al. . Fecal microbiota transplantation for the improvement of metabolism in obesity: the FMT-TRIM double-blind placebo-controlled pilot trial. PLoS Med. 2020;17(3):e1003051.
    1. Engels C, Ruscheweyh H-J, Beerenwinkel N, Lacroix C, Schwab C. The common gut microbe Eubacterium hallii also contributes to intestinal propionate formation. Front Microbiol. 2016;7:713.
    1. Roshanravan N, Mahdavi R, Alizadeh Eet al. . Effect of butyrate and inulin supplementation on glycemic status, lipid profile and glucagon-like peptide 1 level in patients with type 2 diabetes: a randomized double-blind, placebo-controlled trial. Horm Metab Res. 2017;49(11):886–91.
    1. Jia L, Li D, Feng Net al. . Anti-diabetic effects of Clostridium butyricum CGMCC0313.1 through promoting the growth of gut butyrate-producing bacteria in type 2 diabetic mice. Sci Rep. 2017;7(1):7046.
    1. Lin HV, Frassetto A, Kowalik EJ Jret al. . Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One. 2012;7(4):e35240.
    1. De Groot PF, Nikolic T, Imangaliyev Set al. . Oral butyrate does not affect innate immunity and islet autoimmunity in individuals with longstanding type 1 diabetes: a randomised controlled trial. Diabetologia. 2020;63(3):597–610.
    1. Petriz BA, Castro AP, Almeida JAet al. . Exercise induction of gut microbiota modifications in obese, non-obese and hypertensive rats. BMC Genomics. 2014;15(1):511.
    1. Palmas V, Pisanu S, Madau Vet al. . Gut microbiota markers associated with obesity and overweight in Italian adults. Sci Rep. 2021;11(1):5532.
    1. Blanton LV, Charbonneau MR, Salih Tet al. . Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children. Science. 2016;351(6275):aad3311.
    1. Kasai C, Sugimoto K, Moritani Iet al. . Comparison of the gut microbiota composition between obese and non-obese individuals in a Japanese population, as analyzed by terminal restriction fragment length polymorphism and next-generation sequencing. BMC Gastroenterol. 2015;15(1):100.
    1. Mayengbam S, Lambert JE, Parnell JAet al. . Impact of dietary fiber supplementation on modulating microbiota–host–metabolic axes in obesity. J Nutr Biochem. 2019;64:228–36.
    1. Tolhurst G, Heffron H, Lam YSet al. . Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein–coupled receptor FFAR2. Diabetes. 2012;61(2):364–71.
    1. Ceriello A, Novials A, Ortega Eet al. . Glucagon-like peptide 1 reduces endothelial dysfunction, inflammation, and oxidative stress induced by both hyperglycemia and hypoglycemia in type 1 diabetes. Diabetes Care. 2013;36(8):2346–50.
    1. Frost G, Sleeth ML, Sahuri-arisoylu Met al. . The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun. 2014;5:3611.
    1. Ma Z, Li L, Gotelli NJ. Diversity-disease relationships and shared species analyses for human microbiome-associated diseases. ISME J. 2019;13(8):1911–9.
    1. Suez J, Zmora N, Zilberman-Schapira Get al. . Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174(6):1406–23.e16.
    1. Finlay BB, Amato KR, Azad Met al. . The hygiene hypothesis, the COVID pandemic, and consequences for the human microbiome. Proc Natl Acad Sci U S A. 2021;118(6):e2010217118.
    1. Tornese G, Ceconi V, Monasta L, Carletti C, Faleschini E, Barbi E. Glycemic control in type 1 diabetes mellitus during COVID-19 quarantine and the role of in-home physical activity. Diabetes Technol Ther. 2020;22(6):462–7.
    1. De la Cuesta-Zuluaga J, Mueller NT, Álvarez-Quintero Ret al. . Higher fecal short-chain fatty acid levels are associated with gut microbiome dysbiosis, obesity, hypertension and cardiometabolic disease risk factors. Nutrients. 2019;11(1):51.
    1. Kim KN, Yao Y, Ju SY. Short chain fatty acids and fecal microbiota abundance in humans with obesity: a systematic review and meta-analysis. Nutrients. 2019;11(10):2512.
    1. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987;28(10):1221–7.
    1. Boets E, Gomand SV, Deroover Let al. . Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: a stable isotope study. J Physiol. 2017;595(2):541–55.
    1. Beyond A1C Writing Group . Need for regulatory change to incorporate beyond A1C glycemic metrics. Diabetes Care. 2018;41(6):e92–4.
    1. Wang Y, Rimm EB, Stampfer MJ, Willett WC, Hu FB. Comparison of abdominal adiposity and overall obesity in predicting risk of type 2 diabetes among men. Am J Clin Nutr. 2005;81(3):555–63.
    1. DuBose SN, Hermann JM, Tamborlane WVet al. . Obesity in youth with type 1 diabetes in Germany, Austria, and the United States. J Pediatr. 2015;167(3):627–32.e4.
    1. Conway B, Miller RG, Costacou Tet al. . Temporal patterns in overweight and obesity in type 1 diabetes. Diabetes Med. 2010;27(4):398–404.

Source: PubMed

Подписаться