Exercise restores brain insulin sensitivity in sedentary adults who are overweight and obese

Stephanie Kullmann, Thomas Goj, Ralf Veit, Louise Fritsche, Lore Wagner, Patrick Schneeweiss, Miriam Hoene, Christoph Hoffmann, Jürgen Machann, Andreas Niess, Hubert Preissl, Andreas L Birkenfeld, Andreas Peter, Hans-Ulrich Häring, Andreas Fritsche, Anja Moller, Cora Weigert, Martin Heni, Stephanie Kullmann, Thomas Goj, Ralf Veit, Louise Fritsche, Lore Wagner, Patrick Schneeweiss, Miriam Hoene, Christoph Hoffmann, Jürgen Machann, Andreas Niess, Hubert Preissl, Andreas L Birkenfeld, Andreas Peter, Hans-Ulrich Häring, Andreas Fritsche, Anja Moller, Cora Weigert, Martin Heni

Abstract

BACKGROUNDInsulin resistance of the brain can unfavorably affect long-term weight maintenance and body fat distribution. Little is known if and how brain insulin sensitivity can be restored in humans. We aimed to evaluate the effects of an exercise intervention on insulin sensitivity of the brain and how this relates to exercise-induced changes in whole-body metabolism and behavior.METHODSIn this clinical trial, sedentary participants who were overweight and obese underwent an 8-week supervised aerobic training intervention. Brain insulin sensitivity was assessed in 21 participants (14 women, 7 men; age range 21-59 years; BMI range 27.5-45.5 kg/m2) using functional MRI, combined with intranasal administration of insulin, before and after the intervention.RESULTSThe exercise program resulted in enhanced brain insulin action to the level of a person of healthy weight, demonstrated by increased insulin-induced striatal activity and strengthened hippocampal functional connectivity. Improved brain insulin action correlated with increased mitochondrial respiration in skeletal muscle, reductions in visceral fat and hunger, as well as improved cognition. Mediation analyses suggest that improved brain insulin responsiveness helps mediate the peripheral exercise effects leading to healthier body fat distribution and reduced perception of hunger.CONCLUSIONOur study demonstrates that an 8-week exercise intervention in sedentary individuals can restore insulin action in the brain. Hence, the ameliorating benefits of exercise toward brain insulin resistance may provide an objective therapeutic target in humans in the challenge to reduce diabetes risk factors.TRIAL REGISTRATIONClinicalTrials.gov (NCT03151590).FUNDINGBMBF/DZD 01GI0925.

Keywords: Adipose tissue; Insulin signaling; Metabolism; Neuroimaging; Neuroscience.

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1. Schematic overview of the study…
Figure 1. Schematic overview of the study and major test procedures.
Whole-body MRI and functional MRI (fMRI) of the brain and skeletal muscle biopsies were acquired on 3 separate days before and after the 8-week exercise intervention. fMRI was assessed following overnight fasting (10 hours minimum) to investigate brain insulin action using cerebral blood flow (CBF) and functional connectivity (FC) in the fasted state (fMRI-1) and 30 minutes after nasal insulin spray application (fMRI-2). To quantify brain insulin action, ΔCBF and ΔFC were calculated (fMRI-2 – fMRI-1).
Figure 2. Increased brain insulin action after…
Figure 2. Increased brain insulin action after an 8-week exercise intervention in the putamen.
(A) Image shows cluster in the putamen with an increase in cerebral blood flow from before to after the exercise intervention. Color map corresponds to T values (P < 0.001 uncorrected for display). (BD) Box plots show change in absolute cerebral blood flow in the right putamen from before to after insulin nasal spray (ΔCBF= fMRI-2 – fMRI-1). (B) Before and after 8-week exercise intervention in overweight and obese individuals (n = 18; PFWE < 0.05). C and D are based on previously published data sets serving as comparison groups. Before and after 8 weeks without exercise intervention, after oral placebo intake (n = 19) (50) (C), and cross-sectionally in healthy weight (n = 17) and overweight/obese (n = 17) individuals at a single time point (13) (D). In the plot, the box indicates the first and third quartile (25th and 75th percentile), the line in the box marks the median, and whiskers above and below indicate 1.5 × interquartile range. CBF, cerebral blood flow. *PFWE < 0.05 SVC.
Figure 3. Improved brain insulin action after…
Figure 3. Improved brain insulin action after an 8-week exercise intervention in the right hippocampus.
(A) Image shows cluster (in blue) in the right hippocampus with an increase in functional connectivity to the anterior medial prefrontal cortex (region in yellow) of the default mode network from before to after the exercise intervention (PFWE < 0.05 SVC). Color map in red to yellow corresponds to group averaged default mode network at fMRI-1 (t test, PFWE < 0.05). (B) Box plot shows change in functional connectivity between the right hippocampus and medial prefrontal cortex from before to after insulin nasal spray (fMRI-2 – fMRI-1) before and after 8-week exercise intervention (n = 21; PFWE < 0.05). (C) Change in brain insulin action after an 8-week exercise intervention associates with cognitive function. The y axis displays the change in insulin action from before to after the exercise intervention (ΔFCpost-8-week– ΔFCpre), and the x axis shows the TMT B score in seconds.
Figure 4. Change in brain insulin action…
Figure 4. Change in brain insulin action after an 8-week exercise intervention associates with hunger ratings and metabolic measures.
(A) The y axis displays the change in right putamen blood flow in response to intranasal insulin from before to after the exercise intervention (ΔCBFpost-8-week – ΔCBFpre). The x axis shows the change in hunger ratings in response to intranasal insulin (ΔVASpost-8-week – ΔVASpre). (B) The fold change of visceral adipose tissue from before to after the 8-week exercise intervention. (C) The fold change of maximal coupled skeletal muscle mitochondrial respiration in skeletal muscle fibers from before to after the 8-week exercise intervention. CBF, cerebral blood flow; VAS, visual analogue scale.
Figure 5. Model of exercise-promoted central insulin…
Figure 5. Model of exercise-promoted central insulin action as a mediator between changes in peripheral metabolism and central insulin modulated feeling of hunger from before to after an 8-week exercise intervention.
Path coefficients and CIs are shown next to arrows. All variables relate to changes from before to after the 8-week exercise intervention. Brain template at the top right of the graph shows region in the striatum (i.e., right putamen), revealing a significant exercise-induced increase in central insulin action (ΔCBFpost-8-week – ΔCBF pre). In the model on the top left, path ab indicates the indirect effect of the change in maximal coupled mitochondrial respiration in skeletal muscle fibers on the change in VAT via the exercise-induced change in putamen insulin action. In the model on the bottom left, path ab indicates the indirect effect of the change in maximal coupled mitochondrial respiration in skeletal muscle fibers on the change in hunger (ΔVASpost-8-week – ΔVASpre) via the exercise-induced change in right putamen insulin action. In the model on the bottom right, path ab indicates the indirect effect of the change in VAT on hunger via the exercise-induced change in putamen insulin action. CBF, cerebral blood flow; O2, oxygen flux for mitochondrial respiration; VAS, visual analogue scale for hunger ratings; VAT, visceral adipose tissue.

References

    1. DeFronzo RA, et al. Type 2 diabetes mellitus. Nat Rev Dis Primers. 2015;1:15019. doi: 10.1038/nrdp.2015.19.
    1. Kullmann S, et al. Central nervous pathways of insulin action in the control of metabolism and food intake. Lancet Diabetes Endocrinol. 2020;8(6):524–534. doi: 10.1016/S2213-8587(20)30113-3.
    1. Koch L, et al. Central insulin action regulates peripheral glucose and fat metabolism in mice. J Clin Invest. 2008;118(6):2132–2147. doi: 10.1172/JCI31073.
    1. Bruning JC, et al. Role of brain insulin receptor in control of body weight and reproduction. Science. 2000;289(5487):2122–2125. doi: 10.1126/science.289.5487.2122.
    1. Hill JM, et al. Autoradiographic localization of insulin receptors in rat brain: prominence in olfactory and limbic areas. Neuroscience. 1986;17(4):1127–1138. doi: 10.1016/0306-4522(86)90082-5.
    1. Garcia-Caceres C, et al. Astrocytic insulin signaling couples brain glucose uptake with nutrient availability. Cell. 2016;166(4):867–880. doi: 10.1016/j.cell.2016.07.028.
    1. Porniece Kumar M, et al. Insulin signalling in tanycytes gates hypothalamic insulin uptake and regulation of AgRP neuron activity. Nat Metab. 2021;3(12):1662–1679. doi: 10.1038/s42255-021-00499-0.
    1. Liu S, Borgland SL. Insulin actions in the mesolimbic dopamine system. Exp Neurol. 2019;320:113006. doi: 10.1016/j.expneurol.2019.113006.
    1. Okamoto H, et al. Transgenic rescue of insulin receptor-deficient mice. J Clin Invest. 2004;114(2):214–223. doi: 10.1172/JCI200421645.
    1. Kleinridders A, Pothos EN. Impact of brain insulin signaling on dopamine function, food intake, reward, and emotional behavior. Curr Nutr Rep. 2019;8(2):83–91. doi: 10.1007/s13668-019-0276-z.
    1. Schilling TM, et al. Intranasal insulin increases regional cerebral blood flow in the insular cortex in men independently of cortisol manipulation. Hum Brain Mapp. 2014;35(5):1944–1956. doi: 10.1002/hbm.22304.
    1. Novak V, et al. Enhancement of vasoreactivity and cognition by intranasal insulin in type 2 diabetes. Diabetes Care. 2014;37(3):751–759. doi: 10.2337/dc13-1672.
    1. Kullmann S, et al. Selective insulin resistance in homeostatic and cognitive control brain areas in overweight and obese adults. Diabetes Care. 2015;38(6):1044–1050. doi: 10.2337/dc14-2319.
    1. Kullmann S, et al. Dose-dependent effects of intranasal insulin on resting-state brain activity. J Clin Endocrinol Metab. 2018;103(1):253–262. doi: 10.1210/jc.2017-01976.
    1. Kullmann S, et al. Intranasal insulin enhances brain functional connectivity mediating the relationship between adiposity and subjective feeling of hunger. Sci Rep. 2017;7(1):1627. doi: 10.1038/s41598-017-01907-w.
    1. Heni M, et al. Hypothalamic and striatal insulin action suppresses endogenous glucose production and may stimulate glucose uptake during hyperinsulinemia in lean but not in overweight men. Diabetes. 2017;66(7):1797–1806. doi: 10.2337/db16-1380.
    1. Kullmann S, et al. Intranasal insulin modulates intrinsic reward and prefrontal circuitry of the human brain in lean women. Neuroendocrinology. 2013;97(2):176–182. doi: 10.1159/000341406.
    1. Wingrove JO, et al. Intranasal insulin administration decreases cerebral blood flow in cortico-limbic regions: a neuropharmacological imaging study in normal and overweight males. Diabetes Obes Metab. 2021;23(1):175–185. doi: 10.1111/dom.14213.
    1. Zhang H, et al. Intranasal insulin enhanced resting-state functional connectivity of hippocampal regions in type 2 diabetes. Diabetes. 2015;64(3):1025–1034. doi: 10.2337/db14-1000.
    1. Wagner L, et al. Sex differences in central insulin action: Effect of intranasal insulin on neural food cue reactivity in adults with normal weight and overweight. Int J Obes (Lond) doi: 10.1038/s41366-022-01167-3. doi: 10.1038/s41366-022-01167-3. [published online June 17, 2022].
    1. Schneider E, et al. The effect of intranasal insulin on appetite and mood in women with and without obesity: an experimental medicine study. Int J Obes (Lond) 2022;46(7):1319–1327. doi: 10.1038/s41366-022-01115-1.
    1. Guthoff M, et al. Insulin modulates food-related activity in the central nervous system. J Clin Endocrinol Metab. 2010;95(2):748–755. doi: 10.1210/jc.2009-1677.
    1. Tiedemann LJ, et al. Central insulin modulates food valuation via mesolimbic pathways. Nat Commun. 2017;8:16052. doi: 10.1038/ncomms16052.
    1. Edwin Thanarajah S, et al. Modulation of midbrain neurocircuitry by intranasal insulin. Neuroimage. 2019;194:120–127. doi: 10.1016/j.neuroimage.2019.03.050.
    1. Heni M, et al. Central insulin administration improves whole-body insulin sensitivity via hypothalamus and parasympathetic outputs in men. Diabetes. 2014;63(12):4083–4038. doi: 10.2337/db14-0477.
    1. Kishore P, et al. Activation of K(ATP) channels suppresses glucose production in humans. J Clin Invest. 2011;121(12):4916–4920. doi: 10.1172/JCI58035.
    1. Dash S, et al. Intranasal insulin suppresses endogenous glucose production in humans compared with placebo in the presence of similar venous insulin concentrations. Diabetes. 2015;64(3):766–774. doi: 10.2337/db14-0685.
    1. Heni M, et al. Insulin action in the hypothalamus increases second-phase insulin secretion in humans. Neuroendocrinology. 2020;110(11–12):929–937. doi: 10.1159/000504551.
    1. Iwen KA, et al. Intranasal insulin suppresses systemic but not subcutaneous lipolysis in healthy humans. J Clin Endocrinol Metab. 2014;99(2):E246–E251. doi: 10.1210/jc.2013-3169.
    1. Kellar D, Craft S. Brain insulin resistance in Alzheimer’s disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 2020;19(9):758–766. doi: 10.1016/S1474-4422(20)30231-3.
    1. Scherer T, et al. Brain insulin signalling in metabolic homeostasis and disease. Nat Rev Endocrinol. 2021;17(8):468–483. doi: 10.1038/s41574-021-00498-x.
    1. Kullmann S, et al. Brain insulin sensitivity is linked to adiposity and body fat distribution. Nat Commun. 2020;11(1):1841. doi: 10.1038/s41467-020-15686-y.
    1. Tschritter O, et al. The insulin effect on cerebrocortical theta activity is associated with serum concentrations of saturated nonesterified fatty acids. J Clin Endocrinol Metab. 2009;94(11):4600–4607. doi: 10.1210/jc.2009-0469.
    1. Tschritter O, et al. High cerebral insulin sensitivity is associated with loss of body fat during lifestyle intervention. Diabetologia. 2012;55(1):175–182. doi: 10.1007/s00125-011-2309-z.
    1. Arnold SE, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol. 2018;14(3):168–181. doi: 10.1038/nrneurol.2017.185.
    1. Kullmann S, et al. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. 2016;96(4):1169–1209. doi: 10.1152/physrev.00032.2015.
    1. Hawley JA, Lessard SJ. Exercise training-induced improvements in insulin action. Acta Physiol (Oxf) 2008;192(1):127–135. doi: 10.1111/j.1748-1716.2007.01783.x.
    1. Savikj M, Zierath JR. Train like an athlete: applying exercise interventions to manage type 2 diabetes. Diabetologia. 2020;63(8):1491–1499. doi: 10.1007/s00125-020-05166-9.
    1. Jakicic JM. The effect of physical activity on body weight. Obesity (Silver Spring) 2009;17 Suppl 3:S34–S38. doi: 10.1038/oby.2009.386.
    1. Jakicic JM. The role of physical activity in prevention and treatment of body weight gain in adults. J Nutr. 2002;132(12):3826S–3829S. doi: 10.1093/jn/132.12.3826S.
    1. Wing RR, Hill JO. Successful weight loss maintenance. Annu Rev Nutr. 2001;21:323–341. doi: 10.1146/annurev.nutr.21.1.323.
    1. Boule NG, et al. Effects of exercise training on glucose homeostasis: the HERITAGE Family Study. Diabetes Care. 2005;28(1):108–114. doi: 10.2337/diacare.28.1.108.
    1. Praet SF, van Loon LJ. Exercise: the brittle cornerstone of type 2 diabetes treatment. Diabetologia. 2008;51(3):398–401. doi: 10.1007/s00125-007-0910-y.
    1. Richter EA, et al. Metabolic responses to exercise. Effects of endurance training and implications for diabetes. Diabetes Care. 1992;15(11):1767–1776. doi: 10.2337/diacare.15.11.1767.
    1. Mattson MP. Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab. 2012;16(6):706–722. doi: 10.1016/j.cmet.2012.08.012.
    1. Bliss ES, et al. Benefits of exercise training on cerebrovascular and cognitive function in ageing. J Cereb Blood Flow Metab. 2021;41(3):447–470. doi: 10.1177/0271678X20957807.
    1. Chen W, et al. Aerobic exercise improves food reward systems in obese rats via insulin signaling regulation of dopamine levels in the nucleus accumbens. ACS Chem Neurosci. 2019;10(6):2801–2808. doi: 10.1021/acschemneuro.9b00022.
    1. Hoffmann C, et al. Response of mitochondrial respiration in adipose tissue and muscle to 8 weeks of endurance exercise in obese subjects. J Clin Endocrinol Metab. 2020;105(11):dgaa571. doi: 10.1210/clinem/dgaa571.
    1. Bohm A, et al. TGF-β contributes to impaired exercise response by suppression of mitochondrial key regulators in skeletal muscle. Diabetes. 2016;65(10):2849–2861. doi: 10.2337/db15-1723.
    1. Kullmann S, et al. Empagliflozin improves insulin sensitivity of the hypothalamus in humans with prediabetes: a randomized, double-blind, placebo-controlled, phase 2 trial. Diabetes Care. 2022;45(2):398–406. doi: 10.2337/dc21-1136.
    1. Heni M, et al. Nasal insulin changes peripheral insulin sensitivity simultaneously with altered activity in homeostatic and reward-related human brain regions. Diabetologia. 2012;55(6):1773–1782. doi: 10.1007/s00125-012-2528-y.
    1. Kleinridders A, et al. Insulin resistance in brain alters dopamine turnover and causes behavioral disorders. Proc Natl Acad Sci U S A. 2015;112(11):3463–3468. doi: 10.1073/pnas.1500877112.
    1. Dunn JP, et al. Relationship of dopamine type 2 receptor binding potential with fasting neuroendocrine hormones and insulin sensitivity in human obesity. Diabetes Care. 2012;35(5):1105–1111. doi: 10.2337/dc11-2250.
    1. Kullmann S, et al. Central insulin modulates dopamine signaling in the human striatum. J Clin Endocrinol Metab. 2021;106(10):2949–2961. doi: 10.1210/clinem/dgab410.
    1. Bastioli G, et al. Voluntary exercise boosts striatal dopamine release: evidence for the necessary and sufficient role of BDNF. J Neurosci. 2022;42(23):4725–4736. doi: 10.1523/JNEUROSCI.2273-21.2022.
    1. Cabral DF, et al. Exercise for brain health: an investigation into the underlying mechanisms guided by dose. Neurotherapeutics. 2019;16(3):580–599. doi: 10.1007/s13311-019-00749-w.
    1. Park HS, et al. Exercise alleviates cognitive functions by enhancing hippocampal insulin signaling and neuroplasticity in high-fat diet-induced obesity. Nutrients. 2019;11(7):1603. doi: 10.3390/nu11071603.
    1. Brown C, et al. Insulin blood-brain barrier transport and interactions are greater following exercise in mice. J Appl Physiol (1985) 2022;132(3):824–834. doi: 10.1152/japplphysiol.00866.2021.
    1. Oberste M, et al. Acute exercise-induced set shifting benefits in healthy adults and its moderators: a systematic review and meta-analysis. Front Psychol. 2021;12:528352. doi: 10.3389/fpsyg.2021.528352.
    1. Lundby C, Jacobs RA. Adaptations of skeletal muscle mitochondria to exercise training. Exp Physiol. 2016;101(1):17–22. doi: 10.1113/EP085319.
    1. Ruegsegger GN, et al. Exercise and metformin counteract altered mitochondrial function in the insulin-resistant brain. JCI Insight. 2019;4(18):130681. doi: 10.1172/jci.insight.130681.
    1. Pedersen BK. Physical activity and muscle-brain crosstalk. Nat Rev Endocrinol. 2019;15(7):383–392. doi: 10.1038/s41574-019-0174-x.
    1. Hallschmid M. Intranasal insulin. J Neuroendocrinol. 2021;33(4):12934. doi: 10.1111/jne.12934.
    1. Heni M, et al. Impaired insulin action in the human brain: causes and metabolic consequences. Nat Rev Endocrinol. 2015;11(12):701–711. doi: 10.1038/nrendo.2015.173.
    1. Born J, et al. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5(6):514–516. doi: 10.1038/nn0602-849.
    1. Gancheva S, et al. Effects of intranasal insulin on hepatic fat accumulation and energy metabolism in humans. Diabetes. 2015;64(6):1966–1975. doi: 10.2337/db14-0892.
    1. Ssali T, et al. Mapping long-term functional changes in cerebral blood flow by arterial spin labeling. PLoS One. 2016;11(10):e0164112. doi: 10.1371/journal.pone.0164112.
    1. Chao-Gan Y, Yu-Feng Z. DPARSF: a MATLAB toolbox for “pipeline” data analysis of resting-state fMRI. Front Syst Neurosci. 2010;4:13. doi: 10.3389/fnsys.2010.00013.
    1. Wurslin C, et al. Topography mapping of whole body adipose tissue using a fully automated and standardized procedure. J Magn Reson Imaging. 2010;31(2):430–439. doi: 10.1002/jmri.22036.

Source: PubMed

3
Abonnieren