The PERSonalized Glucose Optimization Through Nutritional Intervention (PERSON) Study: Rationale, Design and Preliminary Screening Results

Anouk Gijbels, Inez Trouwborst, Kelly M Jardon, Gabby B Hul, Els Siebelink, Suzanne M Bowser, Dilemin Yildiz, Lisa Wanders, Balázs Erdos, Dick H J Thijssen, Edith J M Feskens, Gijs H Goossens, Lydia A Afman, Ellen E Blaak, Anouk Gijbels, Inez Trouwborst, Kelly M Jardon, Gabby B Hul, Els Siebelink, Suzanne M Bowser, Dilemin Yildiz, Lisa Wanders, Balázs Erdos, Dick H J Thijssen, Edith J M Feskens, Gijs H Goossens, Lydia A Afman, Ellen E Blaak

Abstract

Background: It is well-established that the etiology of type 2 diabetes differs between individuals. Insulin resistance (IR) may develop in different tissues, but the severity of IR may differ in key metabolic organs such as the liver and skeletal muscle. Recent evidence suggests that these distinct tissue-specific IR phenotypes may also respond differentially to dietary macronutrient composition with respect to improvements in glucose metabolism. Objective: The main objective of the PERSON study is to investigate the effects of an optimal vs. suboptimal dietary macronutrient intervention according to tissue-specific IR phenotype on glucose metabolism and other health outcomes. Methods: In total, 240 overweight/obese (BMI 25 - 40 kg/m2) men and women (age 40 - 75 years) with either skeletal muscle insulin resistance (MIR) or liver insulin resistance (LIR) will participate in a two-center, randomized, double-blind, parallel, 12-week dietary intervention study. At screening, participants undergo a 7-point oral glucose tolerance test (OGTT) to determine the hepatic insulin resistance index (HIRI) and muscle insulin sensitivity index (MISI), classifying each participant as either "No MIR/LIR," "MIR," "LIR," or "combined MIR/LIR." Individuals with MIR or LIR are randomized to follow one of two isocaloric diets varying in macronutrient content and quality, that is hypothesized to be either an optimal or suboptimal diet, depending on their tissue-specific IR phenotype (MIR/LIR). Extensive measurements in a controlled laboratory setting as well as phenotyping in daily life are performed before and after the intervention. The primary study outcome is the difference in change in disposition index, which is the product of insulin sensitivity and first-phase insulin secretion, between participants who received their hypothesized optimal or suboptimal diet. Discussion: The PERSON study is one of the first randomized clinical trials in the field of precision nutrition to test effects of a more personalized dietary intervention based on IR phenotype. The results of the PERSON study will contribute knowledge on the effectiveness of targeted nutritional strategies to the emerging field of precision nutrition, and improve our understanding of the complex pathophysiology of whole body and tissue-specific IR. Clinical Trial Registration: https://ichgcp.net/clinical-trials-registry/NCT03708419, clinicaltrials.gov as NCT03708419.

Keywords: dietary intervention study; glucose homeostasis; insulin resistance; metabolic phenotype; obesity; personalized nutrition; precision nutrition; randomized clinical trial.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Gijbels, Trouwborst, Jardon, Hul, Siebelink, Bowser, Yildiz, Wanders, Erdos, Thijssen, Feskens, Goossens, Afman and Blaak.

Figures

Figure 1
Figure 1
Study design of the PERSON study. Tissue-specific insulin resistance (MIR, muscle insulin resistance; LIR, liver insulin resistance) is assessed at screening using a 7-point oral glucose tolerance test and eligible participants with MIR or LIR are randomized to follow either their hypothesized optimal (dark purple) or suboptimal (light purple) diet for 12 weeks. Before and after the intervention, participants are extensively phenotyped during a “characterization week” in a controlled laboratory setting as well as in daily life. BMI, body mass index; MUFA, monounsaturated fatty acid.
Figure 2
Figure 2
Graphical overview of the pre- and post-intervention characterization week. The characterization week contains multiple clinical test days, during which participants are extensively phenotyped. In addition, blood glucose and physical activity are continuously monitored. At home, participants record their dietary intake and feelings of well-being, collect a fecal sample and 24-h urine, and consume a standardized breakfast on day 4, and on day 5, participants have a full day of standardized meals and snacks, including the standardized breakfast. In a subgroup of the study population, additional measurements are performed. DXA, dual-energy X-ray absorptiometry; MRI, magnetic resonance imaging; 1H-MRS, proton magnetic resonance spectroscopy; AGEs, advanced glycation endproducts; CAR, carotid artery reactivity; OGTT, oral glucose tolerance test; GI, gastrointestinal; scAT, subcutaneous adipose tissue; SM, skeletal muscle.
Figure 3
Figure 3
Overview of all measurements performed within the PERSON study. *Performed in a subgroup of the study population. SCR, screening visit; CW, characterization week; DIW, dietary intervention week.
Figure 4
Figure 4
Graphical overview of the oral glucose tolerance test and the high-fat mixed-meal (HFMM) test that are performed during the pre- and post-intervention characterization week. Participants are instructed to drink the glucose drink or HFMM within 5 min, and fasting and postprandial blood samples are drawn at the indicated timepoints for determination of the indicated metabolites. CHO, carbohydrates; HbA1c, hemoglobin A1c; FFA, free fatty acids; TAG, triglycerides; GLP-1, glucagon-like peptide 1; PYY, peptide YY; NMR, nuclear magnetic resonance; HDL, high-density lipoprotein; SCFA, short-chain fatty acids; PBMCs, peripheral blood mononuclear cells.
Figure 5
Figure 5
Flowchart of participant enrollment and eligibility from March 2018 to March 2020.
Figure 6
Figure 6
Plasma glucose (A–C) and insulin (D–F) concentrations during an oral glucose tolerance test according to insulin resistance phenotype. (A,D): data are geometric means with 95% confidence intervals; significant differences for MIR vs. LIR as analyzed using estimated marginal means from linear mixed-effects models with adjustment for sex and Bonferroni post-hoc pairwise comparisons are denoted with *(p < 0.05) or ***(p < 0.001). (B,C,E,F): data are adjusted geometric means with 95% confidence intervals. Different letters (a, b, c, d) indicate significant differences (p < 0.05) between IR phenotypes, as tested using ANCOVA with adjustment for sex and Bonferroni post-hoc pairwise comparisons.
Figure 7
Figure 7
HOMA-IR (A), HOMA-β (B), Matsuda index (C), disposition index (D), muscle insulin sensitivity index (E), and hepatic insulin resistance index (F) according to insulin resistance (IR) phenotype. Data are adjusted geometric means with 95% confidence intervals. Different letters (a, b, c, d) indicate significant differences (p < 0.05) between IR phenotypes, as tested using ANCOVA with adjustment for sex and Bonferroni post-hoc pairwise comparisons.

References

    1. Blaak EE, Antoine JM, Benton D, Bjorck I, Bozzetto L, Brouns F, et al. . Impact of postprandial glycaemia on health and prevention of disease. Obes Rev. (2012) 13:923–84. 10.1111/j.1467-789X.2012.01011.x
    1. Penn L, White M, Lindstrom J, den Boer AT, Blaak E, Eriksson JG, et al. . Importance of weight loss maintenance and risk prediction in the prevention of type 2 diabetes: analysis of European diabetes prevention study RCT. PLoS ONE. (2013) 8:e57143. 10.1371/journal.pone.0057143
    1. Gannon MC, Nuttall FQ. Control of blood glucose in type 2 diabetes without weight loss by modification of diet composition. Nutr Metab. (2006) 3:16. 10.1186/1743-7075-3-16
    1. Bray GA, Krauss RM, Sacks FM, Qi L. Lessons learned from the POUNDS lost study: genetic, metabolic, and behavioral factors affecting changes in body weight, body composition, and cardiometabolic risk. Curr Obes Rep. (2019) 8:262–83. 10.1007/s13679-019-00353-1
    1. Bashiardes S, Godneva A, Elinav E, Segal E. Towards utilization of the human genome and microbiome for personalized nutrition. Curr Opin Biotechnol. (2018) 51:57–63. 10.1016/j.copbio.2017.11.013
    1. Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, et al. . Personalized nutrition by prediction of glycemic responses. Cell. (2015) 163:1079–94. 10.1016/j.cell.2015.11.001
    1. Berry SE, Valdes AM, Drew DA, Asnicar F, Mazidi M, Wolf J, et al. . Human postprandial responses to food and potential for precision nutrition. Nat Med. (2020) 26:964–73. 10.1038/s41591-020-0934-0
    1. Konstantinidou V, Daimiel L, Ordovas JM. Personalized nutrition and cardiovascular disease prevention: from Framingham to PREDIMED. Adv Nutr. (2014) 5:368S−71S. 10.3945/an.113.005686
    1. Mendes-Soares H, Raveh-Sadka T, Azulay S, Edens K, Ben-Shlomo Y, Cohen Y, et al. . Assessment of a personalized approach to predicting postprandial glycemic responses to food among individuals without diabetes. JAMA Netw Open. (2019) 2:e188102. 10.1001/jamanetworkopen.2018.8102
    1. Abdul-Ghani MA, Matsuda M, Balas B, DeFronzo RA. Muscle and liver insulin resistance indexes derived from the oral glucose tolerance test. Diabetes Care. (2007) 30:89–94. 10.2337/dc06-1519
    1. DeFronzo RA, Simonson D, Ferrannini E. Hepatic and peripheral insulin resistance: a common feature of type 2 (non-insulin-dependent) and type 1 (insulin-dependent) diabetes mellitus. Diabetologia. (1982) 23:313–9. 10.1007/BF00253736
    1. O'Donovan SD, Lenz M, Goossens GH, van der Kallen CJH, Eussen S, Stehouwer CDA, et al. . Improved quantification of muscle insulin sensitivity using oral glucose tolerance test data: the MISI calculator. Sci Rep. (2019) 9:9388. 10.1038/s41598-019-45858-w
    1. van der Kolk BW, Vogelzangs N, Jocken JWE, Valsesia A, Hankemeier T, Astrup A, et al. . Plasma lipid profiling of tissue-specific insulin resistance in human obesity. Int J Obes. (2019) 43:989–98. 10.1038/s41366-018-0189-8
    1. Vogelzangs N, van der Kallen CJH, van Greevenbroek MMJ, van der Kolk BW, Jocken JWE, Goossens GH, et al. . Metabolic profiling of tissue-specific insulin resistance in human obesity: results from the diogenes study and the Maastricht study. Int J Obes. (2020) 44:1376–86. 10.1038/s41366-020-0565-z
    1. Goossens GH, Moors CC, Jocken JW, van der Zijl NJ, Jans A, Konings E, et al. . Altered skeletal muscle fatty acid handling in subjects with impaired glucose tolerance as compared to impaired fasting glucose. Nutrients. (2016) 8:164. 10.3390/nu8030164
    1. van der Kolk BW, Kalafati M, Adriaens M, van Greevenbroek MMJ, Vogelzangs N, Saris WHM, et al. . Subcutaneous adipose tissue and systemic inflammation are associated with peripheral but not hepatic insulin resistance in humans. Diabetes. (2019) 68:2247–58. 10.2337/db19-0560
    1. Trouwborst I, Bowser SM, Goossens GH, Blaak EE. Ectopic fat accumulation in distinct insulin resistant phenotypes; targets for personalized nutritional interventions. Front Nutr. (2018) 5:77. 10.3389/fnut.2018.00077
    1. Blanco-Rojo R, Alcala-Diaz JF, Wopereis S, Perez-Martinez P, Quintana-Navarro GM, Marin C, et al. . The insulin resistance phenotype (muscle or liver) interacts with the type of diet to determine changes in disposition index after 2 years of intervention: the CORDIOPREV-DIAB randomised clinical trial. Diabetologia. (2016) 59:67–76. 10.1007/s00125-015-3776-4
    1. Benton D. Carbohydrate ingestion, blood glucose and mood. Neurosci Biobehav Rev. (2002) 26:293–308. 10.1016/S0149-7634(02)00004-0
    1. Gailliot MT, Baumeister RF. The physiology of willpower: linking blood glucose to self-control. Pers Soc Psychol Rev. (2007) 11:303–27. 10.1177/1088868307303030
    1. Penckofer S, Quinn L, Byrn M, Ferrans C, Miller M, Strange P. Does glycemic variability impact mood and quality of life? Diabetes Technol Ther. (2012) 14:303–10. 10.1089/dia.2011.0191
    1. Schram MT, Sep SJ, van der Kallen CJ, Dagnelie PC, Koster A, Schaper N, et al. . The Maastricht study: an extensive phenotyping study on determinants of type 2 diabetes, its complications and its comorbidities. Eur J Epidemiol. (2014) 29:439–51. 10.1007/s10654-014-9889-0
    1. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. (1985) 28:412–9. 10.1007/BF00280883
    1. Matsuda M, DeFronzo RA. Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care. (1999) 22:1462–70. 10.2337/diacare.22.9.1462
    1. Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med. (1998) 15:539–53. 10.1002/(SICI)1096-9136(199807)15:7&lt;539::AID-DIA668&gt;;2-S
    1. Streppel MT, de Vries JH, Meijboom S, Beekman M, de Craen AJ, Slagboom PE, et al. . Relative validity of the food frequency questionnaire used to assess dietary intake in the Leiden longevity study. Nutr J. (2013) 12:75. 10.1186/1475-2891-12-75
    1. Goldberg GR, Black AE, Jebb SA, Cole TJ, Murgatroyd PR, Coward WA, et al. . Critical evaluation of energy intake data using fundamental principles of energy physiology: 1. Derivation of cut-off limits to identify under-recording. Eur J Clin Nutr. (1991) 45:569–81.
    1. Black AE. Critical evaluation of energy intake using the goldberg cut-off for energy intake:basal metabolic rate. A practical guide to its calculation, use and limitations. Int J Obes Relat Metab Disord. (2000) 24:1119–30. 10.1038/sj.ijo.0801376
    1. Hendriks AAJ, Ormel J, van de Willige G. Long-term difficulties measured by a self-report questionnaire and semi-structured interview: a comparison of methods [in Dutch]. Gedrag Gezondheid. (1990) 18:273–83. 10.1037/t30127-000
    1. VanderZee KI, Sanderman R, Heyink JW, de Haes H. Psychometric qualities of the RAND 36-Item health survey 1.0: a multidimensional measure of general health status. Int J Behav Med. (1996) 3:104–22. 10.1207/s15327558ijbm0302_2
    1. Nieboer A, Lindenberg S, Boomsma A, van Bruggen AC. Dimensions of wellBeing and their measurement: the spf-Il scale. Soc Indic Res. (2005) 73:313–53. 10.1007/s11205-004-0988-2
    1. Brown S, Thorpe H, Hawkins K, Brown J. Minimization–reducing predictability for multi-centre trials whilst retaining balance within centre. Stat Med. (2005) 24:3715–27. 10.1002/sim.2391
    1. Saghaei M, Saghaei S. Implementation of an open-source customizable minimization program for allocation of patients to parallel groups in clinical trials. J Biomed Sci Eng. (2011) 4:734–9. 10.4236/jbise.2011.411090
    1. Altman DG, Bland JM. Treatment allocation by minimisation. BMJ. (2005) 330:843. 10.1136/bmj.330.7495.843
    1. Schofield WN. Predicting basal metabolic rate, new standards and review of previous work. Hum Nutr Clin Nutr. (1985) 39(Suppl 1):5–41.
    1. Kromhout D, Spaaij CJ, de Goede J, Weggemans RM. The 2015 Dutch food-based dietary guidelines. Eur J Clin Nutr. (2016) 70:869–78. 10.1038/ejcn.2016.52
    1. Lucassen DA, Brouwer-Brolsma EM, van de Wiel AM, Siebelink E, Feskens EJM. Iterative development of an innovative smartphone-Based dietary assessment tool: traqq. J Vis Exp. (2021) 169. 10.3791/62032
    1. Soininen P, Kangas AJ, Wurtz P, Suna T, Ala-Korpela M. Quantitative serum nuclear magnetic resonance metabolomics in cardiovascular epidemiology and genetics. Circ Cardiovasc Genet. (2015) 8:192–206. 10.1161/CIRCGENETICS.114.000216
    1. van Mil AC, Hartman Y, van Oorschot F, Heemels A, Bax N, Dawson EA, et al. . Correlation of carotid artery reactivity with cardiovascular risk factors and coronary artery vasodilator responses in asymptomatic, healthy volunteers. J Hypertens. (2017) 35:1026–34. 10.1097/HJH.0000000000001274
    1. Linge J, Whitcher B, Borga M, Dahlqvist Leinhard O. Sub-phenotyping metabolic disorders using body composition: an individualized, nonparametric approach utilizing large data sets. Obesity. (2019) 27:1190–9. 10.1002/oby.22510
    1. Demerath EW, Sun SS, Rogers N, Lee M, Reed D, Choh AC, et al. . Anatomical patterning of visceral adipose tissue: race, sex, and age variation. Obesity. (2007) 15:2984–93. 10.1038/oby.2007.356
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol. (1997) 32:920–4. 10.3109/00365529709011203
    1. Surono IS, Wardana AA, Waspodo P, Saksono B, Verhoeven J, Venema K. Effect of functional food ingredients on gut microbiota in a rodent diabetes model. Nutr Metab. (2020) 17:77. 10.1186/s12986-020-00496-2
    1. Muller M, Hernandez MAG, Goossens GH, Reijnders D, Holst JJ, Jocken JWE, et al. . Circulating but not faecal short-chain fatty acids are related to insulin sensitivity, lipolysis and GLP-1 concentrations in humans. Sci Rep. (2019) 9:12515. 10.1038/s41598-019-48775-0
    1. Drossman DA. The functional gastrointestinal disorders and the rome III process. Gastroenterology. (2006) 130:1377–90. 10.1053/j.gastro.2006.03.008
    1. Laheij A, Raber-Durlacher JE, Koppelmans RGA, Huysmans M, Potting C, van Leeuwen SJM, et al. . Microbial changes in relation to oral mucositis in autologous hematopoietic stem cell transplantation recipients. Sci Rep. (2019) 9:16929. 10.1038/s41598-019-53073-w
    1. Wetzels S, Bijnen M, Wijnands E, Biessen EAL, Schalkwijk CG, Wouters K. Characterization of immune cells in human adipose tissue by using flow cytometry. J Vis Exp. (2018) 133:57319. 10.3791/57319
    1. Shanely RA, Zwetsloot KA, Triplett NT, Meaney MP, Farris GE, Nieman DC. Human skeletal muscle biopsy procedures using the modified bergstrom technique. J Vis Exp. (2014) 91:51812. 10.3791/51812
    1. DeFronzo RA, Tobin JD, Andres R. Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol. (1979) 237:E214–23. 10.1152/ajpendo.1979.237.3.E214
    1. Weir JB. New methods for calculating metabolic rate with special reference to protein metabolism. J Physiol. (1949) 109:1–9. 10.1113/jphysiol.1949.sp004363
    1. Frayn KN. Calculation of substrate oxidation rates in vivo from gaseous exchange. J Appl Physiol Respir Environ Exerc Physiol. (1983) 55:628–34. 10.1152/jappl.1983.55.2.628
    1. Winkler EA, Bodicoat DH, Healy GN, Bakrania K, Yates T, Owen N, et al. . Identifying adults' valid waking wear time by automated estimation in activPAL data collected with a 24 h wear protocol. Physiol Meas. (2016) 37:1653–68. 10.1088/0967-3334/37/10/1653
    1. Wilhelm PSD. Assessing mood in daily life: structural validity, sensitivity to change, and reliability of a short-scale to measure three basic dimensions of mood. Eur J Psychol Assessment. (2007) 23:258–67. 10.1027/1015-5759.23.4.258
    1. Akerstedt T, Gillberg M. Subjective and objective sleepiness in the active individual. Int J Neurosci. (1990) 52:29–37. 10.3109/00207459008994241
    1. Baulk SD, Reyner LA, Horne JA. Driver sleepiness–evaluation of reaction time measurement as a secondary task. Sleep. (2001) 24:695–8. 10.1093/sleep/24.6.695
    1. Cambridge Cognition Ltd .(2020). Available online at: (accessed April 8, 2021).
    1. de Bruijn SEM, de Varies YC, de Graaf C, Boesveldt S, Jager G. The reliability and validity of the macronutrient and taste preference ranking task: a new method to measure food preferences. Food Qual Prefer. (2017) 57:32–40. 10.1016/j.foodqual.2016.11.003
    1. Cohen S, Kamarck T, Mermelstein R. A global measure of perceived stress. J Health Soc Behav. (1983) 24:385–96. 10.2307/2136404
    1. Chalder T, Berelowitz G, Pawlikowska T, Watts L, Wessely S, Wright D, et al. . Development of a fatigue scale. J Psychosom Res. (1993) 37:147–53. 10.1016/0022-3999(93)90081-P
    1. Buysse DJ, Reynolds CF 3rd, Monk TH, Berman SR, Kupfer DJ. The pittsburgh sleep quality index: a new instrument for psychiatric practice and research. Psychiatry Res. (1989) 28:193–213. 10.1016/0165-1781(89)90047-4
    1. Roenneberg T, Wirz-Justice A, Merrow M. Life between clocks: daily temporal patterns of human chronotypes. J Biol Rhythms. (2003) 18:80–90. 10.1177/0748730402239679
    1. Johns MW. A new method for measuring daytime sleepiness: the Epworth sleepiness scale. Sleep. (1991) 14:540–5. 10.1093/sleep/14.6.540
    1. Baecke JA, Burema J, Frijters JE. A short questionnaire for the measurement of habitual physical activity in epidemiological studies. Am J Clin Nutr. (1982) 36:936–42. 10.1093/ajcn/36.5.936
    1. Chinapaw MJ, Slootmaker SM, Schuit AJ, van Zuidam M, van Mechelen W. Reliability and validity of the activity questionnaire for adults and adolescents (AQuAA). BMC Med Res Methodol. (2009) 9:58. 10.1186/1471-2288-9-58
    1. Schwarzer R, Renner B. Health-Specific Self-Efficacy Scales. Berlin: Freie Universität Berlin. (2009).
    1. Hausenblas HA, Nigg CR, dannecker EA, Downs DS, Gardner RE, Fallon EA, et al. . A missing piece of the transtheoretical model applied to exercise: development and validation of the temptation to not exercise scale. Psychol Health. (2001) 16:381–90. 10.1080/08870440108405514
    1. Orskov C, Rabenhoj L, Wettergren A, Kofod H, Holst JJ. Tissue and plasma concentrations of amidated and glycine-extended glucagon-like peptide I in humans. Diabetes. (1994) 43:535–9. 10.2337/diabetes.43.4.535
    1. Castor Electronic Data Capture: Castor EDC. (2021). Available online at: (accessed April 8, 2021).
    1. Wilkinson MD, Dumontier M, Aalbersberg IJ, Appleton G, Axton M, Baak A, et al. . The FAIR guiding principles for scientific data management and stewardship. Sci Data. (2016) 3:160018. 10.1038/sdata.2016.18
    1. Leon-Acuna A, Alcala-Diaz JF, Delgado-Lista J, Torres-Pena JD, Lopez-Moreno J, Camargo A, et al. . Hepatic insulin resistance both in prediabetic and diabetic patients determines postprandial lipoprotein metabolism: from the CORDIOPREV study. Cardiovasc Diabetol. (2016) 15:68. 10.1186/s12933-016-0380-y
    1. Lopez S, Bermudez B, Pacheco YM, Villar J, Abia R, Muriana FJ. Distinctive postprandial modulation of beta cell function and insulin sensitivity by dietary fats: monounsaturated compared with saturated fatty acids. Am J Clin Nutr. (2008) 88:638–44. 10.1093/ajcn/88.3.638
    1. Markova M, Pivovarova O, Hornemann S, Sucher S, Frahnow T, Wegner K, et al. . Isocaloric diets high in animal or plant protein reduce liver fat and inflammation in individuals with type 2 diabetes. Gastroenterology. (2017) 152:571–85.e8. 10.1053/j.gastro.2016.10.007
    1. Bortolotti M, Kreis R, Debard C, Cariou B, Faeh D, Chetiveaux M, et al. . High protein intake reduces intrahepatocellular lipid deposition in humans. Am J Clin Nutr. (2009) 90:1002–10. 10.3945/ajcn.2008.27296
    1. Guess ND, Dornhorst A, Oliver N, Bell JD, Thomas EL, Frost GS. A randomized controlled trial: the effect of inulin on weight management and ectopic fat in subjects with prediabetes. Nutr Metab. (2015) 12:36. 10.1186/s12986-015-0033-2
    1. Skytte MJ, Samkani A, Petersen AD, Thomsen MN, Astrup A, Chabanova E, et al. . A carbohydrate-reduced high-protein diet improves HbA1c and liver fat content in weight stable participants with type 2 diabetes: a randomised controlled trial. Diabetologia. (2019) 62:2066–78. 10.1007/s00125-019-4956-4
    1. Seppala-Lindroos A, Vehkavaara S, Hakkinen AM, Goto T, Westerbacka J, Sovijarvi A, et al. . Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J Clin Endocrinol Metab. (2002) 87:3023–8. 10.1210/jcem.87.7.8638
    1. Guess ND, Dornhorst A, Oliver N, Frost GS. A randomised crossover trial: the effect of inulin on glucose homeostasis in subtypes of prediabetes. Ann Nutr Metab. (2016) 68:26–34. 10.1159/000441626
    1. Bock G, Chittilapilly E, Basu R, Toffolo G, Cobelli C, Chandramouli V, et al. . Contribution of hepatic and extrahepatic insulin resistance to the pathogenesis of impaired fasting glucose: role of increased rates of gluconeogenesis. Diabetes. (2007) 56:1703–11. 10.2337/db06-1776
    1. Meyer C, Pimenta W, Woerle HJ, Van Haeften T, Szoke E, Mitrakou A, et al. . Different mechanisms for impaired fasting glucose and impaired postprandial glucose tolerance in humans. Diabetes Care. (2006) 29:1909–14. 10.2337/dc06-0438
    1. Jans A, Konings E, Goossens GH, Bouwman FG, Moors CC, Boekschoten MV, et al. . PUFAs acutely affect triacylglycerol-derived skeletal muscle fatty acid uptake and increase postprandial insulin sensitivity. Am J Clin Nutr. (2012) 95:825–36. 10.3945/ajcn.111.028787
    1. Clarke R. Long-term weight loss and prevention of cardiovascular disease. Circulation. (2011) 124:2801–2. 10.1161/CIRCULATIONAHA.111.071852
    1. Sommerfield AJ, Deary IJ, Frier BM. Acute hyperglycemia alters mood state and impairs cognitive performance in people with type 2 diabetes. Diabetes Care. (2004) 27:2335–40. 10.2337/diacare.27.10.2335
    1. Breymeyer KL, Lampe JW, McGregor BA, Neuhouser ML. Subjective mood and energy levels of healthy weight and overweight/obese healthy adults on high-and low-glycemic load experimental diets. Appetite. (2016) 107:253–9. 10.1016/j.appet.2016.08.008
    1. Tooley KL. Effects of the human gut microbiota on cognitive performance, brain structure and function: a narrative review. Nutrients. (2020) 12:3009. 10.3390/nu12103009
    1. Proctor C, Thiennimitr P, Chattipakorn N, Chattipakorn SC. Diet, gut microbiota and cognition. Metab Brain Dis. (2017) 32:1–17. 10.1007/s11011-016-9917-8
    1. Erdos B, van Sloun B, Adriaens ME, O'Donovan SD, Langin D, Astrup A, et al. . Personalized computational model quantifies heterogeneity in postprandial responses to oral glucose challenge. PLoS Comput Biol. (2021) 17:e1008852. 10.1371/journal.pcbi.1008852
    1. Goossens GH, Jocken JWE, Blaak EE. Sexual dimorphism in cardiometabolic health: the role of adipose tissue, muscle and liver. Nat Rev Endocrinol. (2020) 17:47–66. 10.1038/s41574-020-00431-8
    1. Hall H, Perelman D, Breschi A, Limcaoco P, Kellogg R, McLaughlin T, et al. . Glucotypes reveal new patterns of glucose dysregulation. PLoS Biol. (2018) 16:e2005143. 10.1371/journal.pbio.2005143
    1. Borg R, Kuenen JC, Carstensen B, Zheng H, Nathan DM, Heine RJ, et al. . Real-life glycaemic profiles in non-diabetic individuals with low fasting glucose and normal HbA1c: the A1C-derived average glucose (ADAG) study. Diabetologia. (2010) 53:1608–11. 10.1007/s00125-010-1741-9

Source: PubMed

3
Iratkozz fel