Category: Moms

Boost insulin sensitivity and enhance brain function

Boost insulin sensitivity and enhance brain function

Stoica, L. Bedse, G. Jauch-Chara, K.

Boost insulin sensitivity and enhance brain function -

Over time, these reactions in the body may trigger chronic disorders especially in those already at a higher risk genetically that could have been prevented through simple blood sugar balancing habits. Symptoms of dysregulated glucose levels arise long before the onset of disorders.

These start with glucose spikes. In the short term, these spikes disrupt our hunger cues and lead to cravings, fatigue, migraines, mood swings, poor sleep, hormone imbalances and reduced cognitive function. In the long-term, chronic conditions such as insulin resistance, Alzheimer's disease, fatty liver disease, cancer, arthritis, depression, infertility, poly-cystic ovarian syndrome PCOS , type 2 diabetes, and heart disease may develop.

Moreover, some researchers are calling Alzheimer's 'type 3 diabetes' because of the effects of insulin resistance on the aging brain.

Insulin receptors are present in organs all over the body, including the brain. The brain uses glucose as its main energy source. With insulin resistance, neurons are not able to receive sufficient amounts of glucose to carry out basic tasks, including learning and memory.

A study published in Neuron found a gene responsible for disrupting how the brain processes insulin. In mice, a high-fat diet accelerated the activation of this gene, therefore further impairing insulin function in the brain and suggesting that lowering saturated fat intake may be preventative.

While genetics remains one of the contributors to developing Alzheimer's disease, scientists agree that it is a unique combination of genetic, environmental, and lifestyle factors that cause the associated changes in the brain.

These changes include a loss of neurons and their connections because of amyloid plaque formation and tau protein tangles. Through influencing the phosphorylation and formation of plaques and tangles, insulin resistance contributes to neurodegeneration.

Insulin resistance has also been associated with depression, Parkinson's disease, and cognitive decline. A study published in Frontiers in Neuroscience shows that insulin promotes synaptic plasticity in the hippocampus by modulating long-term potentiation LTP and long-term depression LTD.

These mechanisms allow for better learning and memory. With higher blood sugar and lower insulin sensitivity, the brain is unable to use these mechanisms, which may lead to a faster rate of cognitive decline. In a year longitudinal study , researchers found that those with higher blood sugar had worse cognitive performance and thinking over time.

These changes begin years before the first disease symptoms appear and could start after years of glucose spikes, fatigue after meals, and sugar crashes.

Additionally, brain insulin signaling helps regulate whole body metabolism. A recent Nature study has shown that brain insulin sensitivity is associated with one's body fat distribution. The more sensitive the brain is to insulin, the healthier one's body weight and visceral fat is.

Not only is regulating insulin healthy for your brain, but it is also automatically healthy for the rest of your body. J Neurochem 54 5 —5. Santos MS, Pereira EM, Carvaho AP.

Stimulation of immunoreactive insulin release by glucose in rat brain synaptosomes. Neurochem Res 24 1 —6. Brain insulin, energy and glucose homeostasis; genes, environment and metabolic pathologies. Eur J Pharmacol 1 — Burgos-Ramos E, Gonzalez-Rodriguez A, Canelles S, Baquedano E, Frago LM, Revuelta-Cervantes J, et al.

Endocrinology 3 — Miller DW, Keller BT, Borchardt RT. Identification and distribution of insulin receptors on cultured bovine brain microvessel endothelial cells: possible function in insulin processing in the blood-brain barrier.

J Cell Physiol 2 — Tagliamonte A, DeMontis MG, Olianas M, Onali PL, Gessa GL. Possible role of insulin in the transport of tyrosine and tryptophan from blood to brain.

Adv Exp Med Biol 69 — Ayre SG, Skaletski B, Mosnaim AD. Blood-brain barrier passage of azidothymidine in rats: effect of insulin. Res Commun Chem Pathol Pharmacol 63 1 — Kastin AJ, Akerstrom V. Glucose and insulin increase the transport of leptin through the blood-brain barrier in normal mice but not in streptozotocin-diabetic mice.

Neuroendocrinology 73 4 — Liu H, Yang H, Wang D, Liu Y, Liu X, Li Y, et al. Eur J Pharmacol 2—3 — Liu X, Jing XY, Jin S, Li Y, Liu L, Yu YL, et al. Insulin suppresses the expression and function of breast cancer resistance protein in primary cultures of rat brain microvessel endothelial cells.

Pharmacol Rep 63 2 — Catalan RE, Martinez AM, Aragones MD, Miguel BG, Robles A. Insulin action on brain microvessels; effect on alkaline phosphatase.

Biochem Biophys Res Commun 2 — Langston JW, Li W, Harrison L, Aw TY. Activation of promoter activity of the catalytic subunit of gamma-glutamylcysteine ligase GCL in brain endothelial cells by insulin requires antioxidant response element 4 and altered glycemic status: implication for GCL expression and GSH synthesis.

Free Radic Biol Med 51 9 — Hurley JH, Zhang S, Bye LS, Marshall MS, DePaoli-Roach AA, Guan K, et al. Insulin signaling inhibits the 5-HT2C receptor in choroid plexus via MAP kinase.

BMC Neurosci 4 Bernstein HG, Lendeckel U, Bukowska A, Ansorge S, Ernst T, Stauch R, et al. Regional and cellular distribution patterns of insulin-degrading enzyme in the adult human brain and pituitary.

J Chem Neuroanat 35 2 — Lynch JA, George AM, Eisenhauer PB, Conn K, Gao W, Carreras I, et al. Insulin degrading enzyme is localized predominantly at the cell surface of polarized and unpolarized human cerebrovascular endothelial cell cultures.

J Neurosci Res 83 7 — Seino S, Seino M, Nishi S, Bell GI. Structure of the human insulin receptor gene and characterization of its promoter. Proc Natl Acad Sci U S A 86 1 —8. Moller DE, Yokota A, Caro JF, Flier JS.

Tissue-specific expression of two alternatively spliced insulin receptor mRNAs in man. Mol Endocrinol 3 8 —9. Seino S, Bell GI. Alternative splicing of human insulin receptor messenger RNA. Biochem Biophys Res Commun 1 —6.

Vienberg SG, Bouman SD, Sorensen H, Stidsen CE, Kjeldsen T, Glendorf T, et al. Receptor-isoform-selective insulin analogues give tissue-preferential effects. Biochem J 3 —8. Schulingkamp RJ, Pagano TC, Hung D, Raffa RB. Insulin receptors and insulin action in the brain: review and clinical implications.

Neurosci Biobehav Rev 24 8 — Szabo O, Szabo AJ. Evidence for an insulin-sensitive receptor in the central nervous system. Am J Physiol 6 — Posner BI, Kelly PA, Shiu RP, Friesen HG. Studies of insulin, growth hormone and prolactin binding: tissue distribution, species variation and characterization.

Endocrinology 95 2 — Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature —9. Lowe WL Jr, Boyd FT, Clarke DW, Raizada MK, Hart C, LeRoith D. Development of brain insulin receptors: structural and functional studies of insulin receptors from whole brain and primary cell cultures.

Endocrinology 1 — Landau BR, Takaoka Y, Abrams MA, Genuth SM, van Houten M, Posner BI, et al. Binding of insulin by monkey and pig hypothalamus. Diabetes 32 3 — Hill JM, Lesniak MA, Pert CB, Roth J. Autoradiographic localization of insulin receptors in rat brain: prominence in olfactory and limbic areas.

Neuroscience 17 4 — Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, et al. Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry.

Endocrinology 4 — Marks JL, Porte D Jr, Stahl WL, Baskin DG. Localization of insulin receptor mRNA in rat brain by in situ hybridization. Endocrinology 6 —6. Amessou M, Tahiri K, Chauvet G, Desbuquois B. Age-related changes in insulin receptor mRNA and protein expression in genetically obese Zucker rats.

Diabetes Metab 36 2 —8. Pacold ST, Blackard WG. Central nervous system insulin receptors in normal and diabetic rats. Endocrinology 6 —7. Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Mendelsohn FA. J Neuroendocrinol 1 5 — Hami J, Sadr-Nabavi A, Sankian M, Haghir H.

Sex differences and left-right asymmetries in expression of insulin and insulin-like growth factor-1 receptors in developing rat hippocampus. Brain Struct Funct 2 — Hopkins DF, Williams G. Insulin receptors are widely distributed in human brain and bind human and porcine insulin with equal affinity.

Diabet Med 14 12 — Potau N, Escofet MA, Martinez MC. Ontogenesis of insulin receptors in human cerebral cortex. J Endocrinol Invest 14 1 —8.

Zahniser NR, Goens MB, Hanaway PJ, Vinych JV. Characterization and regulation of insulin receptors in rat brain. J Neurochem 42 5 — Heidenreich KA, Zahniser NR, Berhanu P, Brandenburg D, Olefsky JM. Structural differences between insulin receptors in the brain and peripheral target tissues.

J Biol Chem 14 — Joost HG. Structural and functional heterogeneity of insulin receptors. Cell Signal 7 2 — Kar S, Chabot JG, Quirion R. Quantitative autoradiographic localization of [I]insulin-like growth factor I, [I]insulin-like growth factor II, and [I]insulin receptor binding sites in developing and adult rat brain.

J Comp Neurol 3 — Shemer J, Adamo M, Raizada MK, Heffez D, Zick Y, LeRoith D. Insulin and IGF-I stimulate phosphorylation of their respective receptors in intact neuronal and glial cells in primary culture. J Mol Neurosci 1 1 :3—8.

Brummer T, Schmitz-Peiffer C, Daly RJ. Docking proteins. FEBS J 21 — Taguchi A, White MF. Insulin-like signaling, nutrient homeostasis, and life span. Annu Rev Physiol 70 — Saltiel AR, Pessin JE. Insulin signaling pathways in time and space. Trends Cell Biol 12 2 — Pilch PF, Shia MA, Benson RJ, Fine RE.

Coated vesicles participate in the receptor-mediated endocytosis of insulin. J Cell Biol 96 1 —8. Heffetz D, Zick Y. Receptor aggregation is necessary for activation of the soluble insulin receptor kinase.

J Biol Chem 2 — White MF. Insulin signaling in health and disease. Science —1. Myers MG Jr, Sun XJ, White MF. The IRS-1 signaling system. Trends Biochem Sci 19 7 — Aguirre V, Werner ED, Giraud J, Lee YH, Shoelson SE, White MF.

Phosphorylation of Ser in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. J Biol Chem 2 —7. Krebs DL, Hilton DJ. A new role for SOCS in insulin action.

Suppressor of cytokine signaling. Sci STKE :e6. Jhala US, Canettieri G, Screaton RA, Kulkarni RN, Krajewski S, Reed J, et al.

cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev 17 13 — Abbott MA, Wells DG, Fallon JR. J Neurosci 19 17 —8. Okamura-Oho Y, Miyashita T, Yamada M. Distinctive tissue distribution and phosphorylation of IRSp53 isoforms.

Biochem Biophys Res Commun 5 — Miyahara A, Okamura-Oho Y, Miyashita T, Hoshika A, Yamada M. Genomic structure and alternative splicing of the insulin receptor tyrosine kinase substrate of kDa protein.

J Hum Genet 48 8 —4. Choi J, Ko J, Racz B, Burette A, Lee JR, Kim S, et al. Regulation of dendritic spine morphogenesis by insulin receptor substrate 53, a downstream effector of Rac1 and Cdc42 small GTPases.

J Neurosci 25 4 — Mackie S, Aitken A. Novel brain interacting proteins involved in neurodegenerative disease. FEBS J 16 — Sawallisch C, Berhorster K, Disanza A, Mantoani S, Kintscher M, Stoenica L, et al.

The insulin receptor substrate of 53 kDa IRSp53 limits hippocampal synaptic plasticity. Lizcano JM, Alessi DR. The insulin signalling pathway. Curr Biol 12 7 :R—8.

Jewell JL, Guan KL. Nutrient signaling to mTOR and cell growth. Trends Biochem Sci 38 5 — Kolch W, Kotwaliwale A, Vass K, Janosch P. The role of Raf kinases in malignant transformation. Expert Rev Mol Med 4 8 :1— Evans RM, Hui S, Perkins A, Lahiri DK, Poirier J, Farlow MR.

Cholesterol and APOE genotype interact to influence Alzheimer disease progression. Neurology 62 10 — Frodin M, Gammeltoft S. Role and regulation of 90 kDa ribosomal S6 kinase RSK in signal transduction. Mol Cell Endocrinol 1—2 — White H, Venkatesh B. Clinical review: ketones and brain injury.

Crit Care 15 2 Marty N, Dallaporta M, Thorens B. Brain glucose sensing, counterregulation, and energy homeostasis. Physiology Bethesda 22 — Alvarez E, Roncero I, Chowen JA, Thorens B, Blazquez E.

Expression of the glucagon-like peptide-1 receptor gene in rat brain. J Neurochem 66 3 —7. Navarro M, Rodriquez de Fonseca F, Alvarez E, Chowen JA, Zueco JA, Gomez R, et al. Colocalization of glucagon-like peptide-1 GLP-1 receptors, glucose transporter GLUT-2, and glucokinase mRNAs in rat hypothalamic cells: evidence for a role of GLP-1 receptor agonists as an inhibitory signal for food and water intake.

J Neurochem 67 5 — Roncero I, Alvarez E, Vazquez P, Blazquez E. Functional glucokinase isoforms are expressed in rat brain. J Neurochem 74 5 — Thorens B, Mueckler M. Glucose transporters in the 21st century. Am J Physiol Endocrinol Metab 2 :E—5.

Simpson IA, Appel NM, Hokari M, Oki J, Holman GD, Maher F, et al. Blood-brain barrier glucose transporter: effects of hypo- and hyper-glycemia revisited. J Neurochem 72 1 — Kang L, Routh VH, Kuzhikandathil EV, Gaspers LD, Levin BE. Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons.

Diabetes 53 3 — Levin BE, Routh VH, Kang L, Sanders NM, Dunn-Meynell AA. Neuronal glucosensing: what do we know after 50 years? Diabetes 53 10 —8. Li B, Xi X, Roane DS, Ryan DH, Martin RJ. Distribution of glucokinase, glucose transporter GLUT2, sulfonylurea receptor-1, glucagon-like peptide-1 receptor and neuropeptide Y messenger RNAs in rat brain by quantitative real time RT-PCR.

Brain Res Mol Brain Res 1—2 — Nagamatsu S, Kornhauser JM, Burant CF, Seino S, Mayo KE, Bell GI. Glucose transporter expression in brain. cDNA sequence of mouse GLUT3, the brain facilitative glucose transporter isoform, and identification of sites of expression by in situ hybridization.

J Biol Chem 1 — Gould GW, Brant AM, Kahn BB, Shepherd PR, McCoid SC, Gibbs EM. Expression of the brain-type glucose transporter is restricted to brain and neuronal cells in mice.

Diabetologia 35 4 —9. El Messari S, Leloup C, Quignon M, Brisorgueil MJ, Penicaud L, Arluison M. Immunocytochemical localization of the insulin-responsive glucose transporter 4 Glut4 in the rat central nervous system.

J Comp Neurol 4 — Vannucci SJ, Koehler-Stec EM, Li K, Reynolds TH, Clark R, Simpson IA. GLUT4 glucose transporter expression in rodent brain: effect of diabetes. Brain Res 1 :1— Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, et al.

J Clin Invest 4 — Livingstone C, Lyall H, Gould GW. Hypothalamic GLUT 4 expression: a glucose- and insulin-sensing mechanism? Mol Cell Endocrinol 1 — Wang R, Liu X, Hentges ST, Dunn-Meynell AA, Levin BE, Wang W, et al.

The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides. Diabetes 53 8 — Reagan LP, Gorovits N, Hoskin EK, Alves SE, Katz EB, Grillo CA, et al.

Localization and regulation of GLUTx1 glucose transporter in the hippocampus of streptozotocin diabetic rats. Proc Natl Acad Sci U S A 98 5 —5. Ibberson M, Riederer BM, Uldry M, Guhl B, Roth J, Thorens B. Immunolocalization of GLUTX1 in the testis and to specific brain areas and vasopressin-containing neurons.

Reagan LP, Rosell DR, Alves SE, Hoskin EK, McCall AL, Charron MJ, et al. GLUT8 glucose transporter is localized to excitatory and inhibitory neurons in the rat hippocampus. Brain Res 1—2 — Sankar R, Thamotharan S, Shin D, Moley KH, Devaskar SU.

Insulin-responsive glucose transporters-GLUT8 and GLUT4 are expressed in the developing mammalian brain. Brain Res Mol Brain Res 2 — McEwen BS, Reagan LP. Glucose transporter expression in the central nervous system: relationship to synaptic function.

Eur J Pharmacol 1—3 — Piroli GG, Grillo CA, Hoskin EK, Znamensky V, Katz EB, Milner TA, et al. Peripheral glucose administration stimulates the translocation of GLUT8 glucose transporter to the endoplasmic reticulum in the rat hippocampus.

J Comp Neurol 2 — Obici S, Feng Z, Karkanias G, Baskin DG, Rossetti L. Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat Neurosci 5 6 — Obici S, Zhang BB, Karkanias G, Rossetti L. Hypothalamic insulin signaling is required for inhibition of glucose production.

Nat Med 8 12 — Demuro G, Obici S. Central nervous system and control of endogenous glucose production. Curr Diab Rep 6 3 — Girard J. The inhibitory effects of insulin on hepatic glucose production are both direct and indirect.

Diabetes 55 Suppl 2 :S65—9. Pocai A, Lam TK, Gutierrez-Juarez R, Obici S, Schwartz GJ, Bryan J, et al. Hypothalamic K ATP channels control hepatic glucose production.

Nature — Cotero VE, Routh VH. Insulin blunts the response of glucose-excited neurons in the ventrolateral-ventromedial hypothalamic nucleus to decreased glucose.

Am J Physiol Endocrinol Metab 5 :E—9. Bernard C. Leçons de Physiologie Expérimentale Appliquée à la Médecine. Paris: Baillére Niswender KD, Morrison CD, Clegg DJ, Olson R, Baskin DG, Myers MG Jr, et al. Insulin activation of phosphatidylinositol 3-kinase in the hypothalamic arcuate nucleus: a key mediator of insulin-induced anorexia.

Diabetes 52 2 — Kim YB, Uotani S, Pierroz DD, Flier JS, Kahn BB. In vivo administration of leptin activates signal transduction directly in insulin-sensitive tissues: overlapping but distinct pathways from insulin. Endocrinology 7 — Castellano JM, Roa J, Luque RM, Dieguez C, Aguilar E, Pinilla L, et al.

Peptides 30 1 — Fernandez-Fernandez R, Martini AC, Navarro VM, Castellano JM, Dieguez C, Aguilar E, et al. Novel signals for the integration of energy balance and reproduction. Mol Cell Endocrinol 25 4— — Arias P, Rodriguez M, Szwarcfarb B, Sinay IR, Moguilevsky JA.

Effect of insulin on LHRH release by perifused hypothalamic fragments. Neuroendocrinology 56 3 —8. Miller DW, Blache D, Martin GB.

The role of intracerebral insulin in the effect of nutrition on gonadotrophin secretion in mature male sheep. J Endocrinol 2 —9. Dong Q, Lazarus RM, Wong LS, Vellios M, Handelsman DJ. Pulsatile LH secretion in streptozotocin-induced diabetes in the rat.

J Endocrinol 1 — Tanaka T, Nagatani S, Bucholtz DC, Ohkura S, Tsukamura H, Maeda K, et al. Central action of insulin regulates pulsatile luteinizing hormone secretion in the diabetic sheep model. Biol Reprod 62 5 — Bucholtz DC, Chiesa A, Pappano WN, Nagatani S, Tsukamura H, Maeda KI, et al.

Regulation of pulsatile luteinizing hormone secretion by insulin in the diabetic male lamb. Roger LJ, Fellows RE. Stimulation of ornithine decarboxylase activity by insulin in developing rat brain.

Endocrinology 2 — Wozniak M, Rydzewski B, Baker SP, Raizada MK. The cellular and physiological actions of insulin in the central nervous system.

Neurochem Int 22 1 :1— Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, et al. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci 23 18 — Xu QG, Li XQ, Kotecha SA, Cheng C, Sun HS, Zochodne DW. Insulin as an in vivo growth factor. Exp Neurol 1 — Raizada MK, Yang JW, Fellows RE.

Binding of [I]insulin to specific receptors and stimulation of nucleotide incorporation in cells cultured from rat brain. Brain Res 2 — Heidenreich KA, de Vellis G, Gilmore PR.

Functional properties of the subtype of insulin receptor found on neurons. J Neurochem 51 3 — Clarke DW, Boyd FT Jr, Kappy MS, Raizada MK. Insulin stimulates macromolecular synthesis in cultured glial cells from rat brain.

Am J Physiol 5 Pt 1 :C—9. Heidenreich KA, Toledo SP. Insulin receptors mediate growth effects in cultured fetal neurons.

Activation of a protein kinase that phosphorylates ribosomal protein S6. Recio-Pinto E, Lang FF, Ishii DN. Insulin and insulin-like growth factor II permit nerve growth factor binding and the neurite formation response in cultured human neuroblastoma cells.

Proc Natl Acad Sci U S A 81 8 —6. Ang LC, Bhaumick B, Juurlink BH. Neurite promoting activity of insulin, insulin-like growth factor I and nerve growth factor on spinal motoneurons is astrocyte dependent. Brain Res Dev Brain Res 74 1 —8.

Velazquez E, Blazquez E, Ruiz-Albusac JM. Synergistic effect of glucagon-like peptide 2 GLP-2 and of key growth factors on the proliferation of cultured rat astrocytes.

Evidence for reciprocal upregulation of the mRNAs for GLP-2 and IGF-I receptors. Mol Neurobiol 40 2 — Heni M, Hennige AM, Peter A, Siegel-Axel D, Ordelheide AM, Krebs N, et al. Insulin promotes glycogen storage and cell proliferation in primary human astrocytes. PLoS One 6 6 :e Heidenreich KA, Toledo SP, Brunton LL, Watson MJ, Daniel-Issakani S, Strulovici B.

Insulin stimulates the activity of a novel protein kinase C, PKC-epsilon, in cultured fetal chick neurons. J Biol Chem 25 — Vanhems E, Delbos M, Girardie J. Insulin and neuroparsin promote neurite outgrowth in cultured locust CNS.

Eur J Neurosci 2 9 — Heidenreich KA, Toledo SP, Kenner KA. Regulation of protein phosphorylation by insulin and insulin-like growth factors in cultured fetal neurons.

Adv Exp Med Biol — This fat depot specific effects of brain insulin action could further be determined by differential autonomic innervation of subcutaneous and visceral fat 26 , Both fat depots are innervated by distinct sympathetic and parasympathetic motor neurons with functional consequences of autonomic balance on adipocyte insulin sensitivity and energy storage The proximal regulatory neurons that project into the adipose tissue appear to reside in the hypothalamus 27 , Thus, changes in autonomic nervous system balance that are induced by brain insulin action 8 , 13 , 14 , 24 , 25 could exert differentially effects on subcutaneous and visceral adipocytes and thereby contribute to our current findings.

In line with this hypothesis, induction of brain insulin action was found to modulate systemic but not subcutaneous lipolysis in humans Our current findings are of particular importance, given that the enlarged visceral fat content not only poses a high risk factor for the subsequent development of diabetes, but is also robustly linked to the risk of cardiovascular disease and the development of cancer Brain insulin resistance therefore seems to be involved in the pathogenesis of obesity in general.

More importantly, it appears to be a determinant of healthy and unhealthy obesity. Unfortunately, only a limited sample size was available in our longitudinal cohort. MEG and fMRI most likely capture different aspects of brain insulin sensitivity and their comparability in this regard has not been tested so far.

In conclusion, we showed that high brain insulin sensitivity was linked to weight loss during lifestyle intervention and associates with a favorable body fat distribution. Our current results underline the importance of brain insulin action for the development of body weight and body fat distribution.

As visceral fat is strongly linked to diabetes, cardiovascular risk, and cancer, these findings have implications beyond metabolic diseases and indicate the necessity of strategies to resolve insulin resistance of the human brain.

Details on the TULIP lifestyle intervention study, including primary and secondary outcomes as well as inclusion and exclusion criteria, have been reported previously The study was conducted within the Deutsche Forschungsgemeinschaft DFG project KFO Three hundred participants at high risk for type 2 diabetes completed the intervention.

As reported in ref. In a subgroup of 28 participants, brain insulin sensitivity was assessed by MEG before lifestyle intervention. Of these, 15 individuals were followed-up after 9.

Total dietary energy intake was assessed in 10 of these participants at three time periods during the lifestyle intervention before, during the first 9 months of lifestyle intervention, and during month 9—24 of lifestyle intervention by the mean values of several 3-day food diaries obtained at each visit In all participants, body fat distribution was assessed by whole-body MRI as part of the baseline examination of clinical trials clinicaltrials.

gov: NCT, NCT, NCT, NCT, NCT, NCT For patients characteristics, see Supplementary Table 2. Whole-body MRI was performed in the early morning after overnight fasting on a 1. Detailed information is given in ref. Before lifestyle intervention, participants underwent two hyperinsulinaemic—euglycaemic glucose clamps with insulin or placebo saline infusion on two different days for details see Tschritter et al.

Cerebrocortical activity was assessed by magnetoencephalography MEG before and during the clamp experiment. The power spectrum for the spontaneous activity of the participants was analyzed by a standard statistical mapping procedure taking into account multiple comparison correction for the different frequency bands.

On the basis of earlier findings 20 , assessment of the cerebrocortical insulin effect as changes in theta activity during the insulin experiment corrected for the placebo experiment were calculated Participants underwent whole-brain fMRI at a 3.

Experiments were conducted after an overnight fast and started under basal condition to quantify cerebral blood flow CBF with a pulsed arterial spin labeling PASL measurement using a PICORE-Q2TIPS sequence proximal inversion with control for off-resonance effects—quantitative imaging of perfusion using a single subtraction.

Change in CBF was extracted from the hypothalamus based on recent findings All relevant ethical regulations were complied with and informed written consent was obtained from all participants.

The local ethics committee approved the study protocols Ethics Committee of the Medical Faculty of the Eberhard-Karls-Universität and the University Hopsital Tübingen. In the longitudinal study, changes in body weight, body fat depots, and further metabolic variables and their association with baseline brain insulin sensitivity theta activity were analyzed by MANOVA.

Continuous variables were used for analyses and stratified variables were used solely for better illustration of the results. For the cross-sectional study, correlations between body fat compartments and hypothalamic cerebral blood flow fMRI measurements were analyzed by linear regression models unadjusted and adjusted for sex and age as well as BMI.

Further information on research design is available in the Nature Research Reporting Summary linked to this article. The data that support the findings of this study are available on reasonable request from the corresponding author M.

The source data underlying Figs. Kullmann, S. et al. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Article CAS Google Scholar. Brüning, J. Role of brain insulin receptor in control of body weight and reproduction. Science , — Article ADS Google Scholar.

Jauch-Chara, K. Intranasal Insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes 61 , — Krug, R. Insulin and estrogen independently and differentially reduce macronutrient intake in healthy men.

Article Google Scholar. Hallschmid, M. Postprandial administration of intranasal insulin intensifies satiety and reduces intake of palatable snacks in women. Heni, M. Impaired insulin action in the human brain: causes and metabolic consequences.

Intranasal insulin enhances brain functional connectivity mediating the relationship between adiposity and subjective feeling of hunger. Ruud, J. Neuronal control of peripheral insulin sensitivity and glucose metabolism.

Kleinridders, A. Insulin action in brain regulates systemic metabolism and brain function. Diabetes 63 , — Iwen, K. Intranasal insulin suppresses systemic but not subcutaneous lipolysis in healthy humans.

Gancheva, S. Effects of intranasal insulin on hepatic fat accumulation and energy metabolism in humans. Diabetes 64 , — Nasal insulin changes peripheral insulin sensitivity simultaneously with altered activity in homeostatic and reward-related human brain regions.

Diabetologia 55 , — Central insulin administration improves whole-body insulin sensitivity via hypothalamus and parasympathetic outputs in men. Hypothalamic and striatal insulin action suppresses endogenous glucose production and may stimulate glucose uptake during hyperinsulinemia in lean but not in overweight men.

Diabetes 66 , — Dash, S. Intranasal insulin suppresses endogenous glucose production in humans compared with placebo in the presence of similar venous insulin concentrations. Pocai, A.

Hypothalamic K ATP channels control hepatic glucose production. Nature , — Article ADS CAS Google Scholar. Häring, H. Novel phenotypes of prediabetes? Diabetologia 59 , — Stefan, N.

Metabolically healthy obesity: the low-hanging fruit in obesity treatment? Lancet Diabetes Endocrinol. Causes, characteristics, and consequences of metabolically unhealthy normal weight in humans.

Cell Metab. Tschritter, O. High cerebral insulin sensitivity is associated with loss of body fat during lifestyle intervention.

Matsuda, M. Altered hypothalamic function in response to glucose ingestion in obese humans. Diabetes 48 , — Selective insulin resistance in homeostatic and cognitive control brain areas in overweight and obese adults.

Diabetes Care 38 , — Guthoff, M. The insulin-mediated modulation of visually evoked magnetic fields is reduced in obese subjects. PLoS ONE 6 , e Hypothalamic insulin responsiveness is associated with pancreatic insulin secretion in humans.

Benedict, C. Intranasal insulin enhances postprandial thermogenesis and lowers postprandial serum insulin levels in healthy Men.

Diabetes 60 , — Frühbeck, G. Regulation of adipocyte lipolysis. Kreier, F.

An association between sensitivihy Boost insulin sensitivity and enhance brain function is ssensitivity blood glucose levels and cognitive dysfunction have been found in relatively sensitivify older individuals Eenhance Nutraceutical potential of plant compounds, et. al, Insulin resistance which happens functtion the insulin sensitivity enbance your tissue decreases was found to Boozt associated with Healthy habits impairment and the individuals with diabetes were worse. The research makes it clear that if you want to improve your memory or prevent it from getting worse, you need to implement strategies to improve insulin sensitivity. When you eat a meal consisting of high glycemic index carbohydrates, your blood glucose levels will increase too much. While you may tolerate that occasionally, eating that way every day will affect your insulin sensitivity, and it will not be good for your memory. Diets high in trans-fat and saturated fat adversely affect cognition. Boost insulin sensitivity and enhance brain function

Author: JoJogar

5 thoughts on “Boost insulin sensitivity and enhance brain function

  1. Ich meine, dass Sie nicht recht sind. Geben Sie wir werden es besprechen. Schreiben Sie mir in PM, wir werden reden.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com