Ketogenic diet enhances neurovascular function with altered gut microbiome in young healthy mice

David Ma, Amy C Wang, Ishita Parikh, Stefan J Green, Jared D Hoffman, George Chlipala, M Paul Murphy, Brent S Sokola, Björn Bauer, Anika M S Hartz, Ai-Ling Lin, David Ma, Amy C Wang, Ishita Parikh, Stefan J Green, Jared D Hoffman, George Chlipala, M Paul Murphy, Brent S Sokola, Björn Bauer, Anika M S Hartz, Ai-Ling Lin

Abstract

Neurovascular integrity, including cerebral blood flow (CBF) and blood-brain barrier (BBB) function, plays a major role in determining cognitive capability. Recent studies suggest that neurovascular integrity could be regulated by the gut microbiome. The purpose of the study was to identify if ketogenic diet (KD) intervention would alter gut microbiome and enhance neurovascular functions, and thus reduce risk for neurodegeneration in young healthy mice (12-14 weeks old). Here we show that with 16 weeks of KD, mice had significant increases in CBF and P-glycoprotein transports on BBB to facilitate clearance of amyloid-beta, a hallmark of Alzheimer's disease (AD). These neurovascular enhancements were associated with reduced mechanistic target of rapamycin (mTOR) and increased endothelial nitric oxide synthase (eNOS) protein expressions. KD also increased the relative abundance of putatively beneficial gut microbiota (Akkermansia muciniphila and Lactobacillus), and reduced that of putatively pro-inflammatory taxa (Desulfovibrio and Turicibacter). We also observed that KD reduced blood glucose levels and body weight, and increased blood ketone levels, which might be associated with gut microbiome alteration. Our findings suggest that KD intervention started in the early stage may enhance brain vascular function, increase beneficial gut microbiota, improve metabolic profile, and reduce risk for AD.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Ketogenic diet enhances neurovascular functions. (a) Representative cerebral blood flow (CBF) maps superimposed on structural images; color code indicates level of CBF in a linear scale. KD mice exhibited significantly higher CBF in the (b) ventromedial hypothalamus. Data are presented as mean ± SEM, ***p < 0.001. (c) Western blot (WB) images for mTOR, P-gp, and eNOS from the cortical vasculature, β-Actin was used as loading control. (d) The corresponding values of the levels of protein expression. WB data from KD mice were normalized to β-Actin and compared to the control mice (100%), *p < 0.05, **p < 0.01, ***p < 0.001. (e) Representative confocal images showing increased luminal accumulation of NBD-CSA fluorescence in brain capillaries isolated from KD mice compared to control mice, indicating higher P-gp transport activity. Corresponding quantitative fluorescence data; images are shown in arbitrary fluorescence units (scale 0–255). Data are mean ± SEM for 10 capillaries from one preparation of 10 mice per group, ***p < 0.001. mTOR: mechanistic target of Rapamycin; P-gp: P-glycoprotein; eNOS; endothelial nitric oxide synthase.
Figure 2
Figure 2
Ketogenic diet alters gut microbial diversity and increases pro-vascular microbiota. (a) Microbial diversity (Shannon index) was significantly higher in fecal samples from control relative to KD mice (Mann-Whitney U < 0.02). (b) Genus-level metric multi-dimensional scaling (mMDS) plot of 16S rRNA gene amplicon microbiome data was generated with a Bray-Curtis resemblance matrix. Fecal microbial communities of control and KD mice were significantly different in terms of individual taxa (ANOSIM R = 0.473; p = 0.0002), as described in the text and Table 1. All samples were standardized and square root transformed. 2D stress = 0.16.
Figure 3
Figure 3
Ketogenic diet modulates blood ketone and glucose, and decreases body weight. (a) KD mice had significantly lower blood glucose and (b) significantly higher blood ketone levels than control mice. (c) A scatter plot of blood ketone and glucose showing an inverse linear relationship (Pearson’s r = −0.5761, p < 0.01) where each point represents a mouse (n = 19). (d) KD mice had a significant decrease in weight over the 16 weeks compared to control mice.

References

    1. Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nature reviews. Neuroscience. 2011;12:723–738. doi: 10.1038/nrn3114.
    1. Bangen KJ, et al. APOE genotype modifies the relationship between midlife vascular risk factors and later cognitive decline. J Stroke Cerebrovasc Dis. 2013;22:1361–1369. doi: 10.1016/j.jstrokecerebrovasdis.2013.03.013.
    1. Ebmeier KP, et al. Cerebral perfusion correlates of depressed mood. Br J Psychiatry. 1997;170:77–81. doi: 10.1192/bjp.170.1.77.
    1. Gur RC, et al. The effect of anxiety on cortical cerebral blood flow and metabolism. Journal of cerebral blood flow and metabolism: official journal of the International Society of Cerebral Blood Flow and Metabolism. 1987;7:173–177. doi: 10.1038/jcbfm.1987.40.
    1. Park, J. & Moghaddam, B. Impact of anxiety on prefrontal cortex encoding of cognitive flexibility. Neuroscience (2016).
    1. Bell RD, et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature. 2012;485:512–516. doi: 10.1038/nature11087.
    1. Kelly JR, et al. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci. 2015;9:392.
    1. Braniste V, et al. The gut microbiota influences blood-brain barrier permeability in mice. Science translational medicine. 2014;6:263ra158. doi: 10.1126/scitranslmed.3009759.
    1. Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol. 2004;54:1469–1476. doi: 10.1099/ijs.0.02873-0.
    1. Belzer C, de Vos WM. Microbes inside–from diversity to function: the case of Akkermansia. ISME J. 2012;6:1449–1458. doi: 10.1038/ismej.2012.6.
    1. Li J, Lin S, Vanhoutte PM, Woo CW, Xu A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe−/− Mice. Circulation. 2016;133:2434–2446. doi: 10.1161/CIRCULATIONAHA.115.019645.
    1. Baranano KW, Hartman AL. The ketogenic diet: uses in epilepsy and other neurologic illnesses. Curr Treat Options Neurol. 2008;10:410–419. doi: 10.1007/s11940-008-0043-8.
    1. Walczyk T, Wick JY. The Ketogenic Diet: Making a Comeback. Consult Pharm. 2017;32:388–396. doi: 10.4140/TCP.n.2017.388.
    1. Vanitallie TB, et al. Treatment of Parkinson disease with diet-induced hyperketonemia: a feasibility study. Neurology. 2005;64:728–730. doi: 10.1212/01.WNL.0000152046.11390.45.
    1. Evangeliou A, et al. Application of a ketogenic diet in children with autistic behavior: pilot study. J Child Neurol. 2003;18:113–118. doi: 10.1177/08830738030180020501.
    1. Van der Auwera I, Wera S, Van Leuven F, Henderson ST. A ketogenic diet reduces amyloid beta 40 and 42 in a mouse model of Alzheimer’s disease. Nutr Metab (Lond) 2005;2:28. doi: 10.1186/1743-7075-2-28.
    1. Prins ML, Fujima LS, Hovda DA. Age-dependent reduction of cortical contusion volume by ketones after traumatic brain injury. J Neurosci Res. 2005;82:413–420. doi: 10.1002/jnr.20633.
    1. Puchowicz MA, et al. Neuroprotection in diet-induced ketotic rat brain after focal ischemia. J Cereb Blood Flow Metab. 2008;28:1907–1916. doi: 10.1038/jcbfm.2008.79.
    1. Yang Q, et al. Ischemic preconditioning with a ketogenic diet improves brain ischemic tolerance through increased extracellular adenosine levels and hypoxia-inducible factors. Brain Res. 2017;1667:11–18. doi: 10.1016/j.brainres.2017.04.010.
    1. Newell C, et al. Ketogenic diet modifies the gut microbiota in a murine model of autism spectrum disorder. Mol Autism. 2016;7:37. doi: 10.1186/s13229-016-0099-3.
    1. Lin AL, et al. Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer’s disease. J Cereb Blood Flow Metab. 2013;33:1412–1421. doi: 10.1038/jcbfm.2013.82.
    1. Cheng C, et al. Rapamycin modulates the eNOS vs. shear stress relationship. Cardiovasc Res. 2008;78:123–129. doi: 10.1093/cvr/cvm103.
    1. Parikh I, et al. Caloric restriction preserves memory and reduces anxiety of aging mice with early enhancement of neurovascular functions. Aging. 2016;8:2814–2826. doi: 10.18632/aging.101094.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Amer, M. et al. Probiotics and Their Use in Inflammatory Bowel Disease. Altern Ther Health Med (2017).
    1. Woting A, Blaut M. The Intestinal Microbiota in Metabolic Disease. Nutrients. 2016;8:202. doi: 10.3390/nu8040202.
    1. Vieira AT, Teixeira MM, Martins FS. The role of probiotics and prebiotics in inducing gut immunity. Front Immunol. 2013;4:445. doi: 10.3389/fimmu.2013.00445.
    1. Duncan SH, Louis P, Flint HJ. Cultivable bacterial diversity from the human colon. Lett Appl Microbiol. 2007;44:343–350. doi: 10.1111/j.1472-765X.2007.02129.x.
    1. Tagliabue A, et al. Short-term impact of a classical ketogenic diet on gut microbiota in GLUT1 Deficiency Syndrome: A 3-month prospective observational study. Clin Nutr ESPEN. 2017;17:33–37. doi: 10.1016/j.clnesp.2016.11.003.
    1. Liu W, et al. Diet- and Genetically-induced Obesity Produces Alterations in the Microbiome, Inflammation and Wnt Pathway in the Intestine of Apc+/1638N Mice: Comparisons and Contrasts. J Cancer. 2016;7:1780–1790. doi: 10.7150/jca.15792.
    1. Dao MC, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut. 2016;65:426–436. doi: 10.1136/gutjnl-2014-308778.
    1. Escobar JS, Klotz B, Valdes BE, Agudelo GM. The gut microbiota of Colombians differs from that of Americans, Europeans and Asians. BMC Microbiol. 2014;14:311. doi: 10.1186/s12866-014-0311-6.
    1. Courchesne-Loyer, A. et al. Inverse relationship between brain glucose and ketone metabolism in adults during short-term moderate dietary ketosis: A dual tracer quantitative positron emission tomography study. J Cereb Blood Flow Metab (2016).
    1. Merra G, et al. Very-low-calorie ketogenic diet with aminoacid supplement versus very low restricted-calorie diet for preserving muscle mass during weight loss: a pilot double-blind study. Eur Rev Med Pharmacol Sci. 2016;20:2613–2621.
    1. Shimazu T, et al. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013;339:211–214. doi: 10.1126/science.1227166.
    1. Hasselbalch SG, et al. Changes in cerebral blood flow and carbohydrate metabolism during acute hyperketonemia. Am J Physiol. 1996;270:E746–751.
    1. Linde R, Hasselbalch SG, Topp S, Paulson OB, Madsen PL. Global cerebral blood flow and metabolism during acute hyperketonemia in the awake and anesthetized rat. J Cereb Blood Flow Metab. 2006;26:170–180. doi: 10.1038/sj.jcbfm.9600177.
    1. Veech RL. The therapeutic implications of ketone bodies: the effects of ketone bodies in pathological conditions: ketosis, ketogenic diet, redox states, insulin resistance, and mitochondrial metabolism. Prostaglandins, leukotrienes, and essential fatty acids. 2004;70:309–319. doi: 10.1016/j.plefa.2003.09.007.
    1. Lin AL, et al. Rapamycin rescues vascular, metabolic and learning deficits in apolipoprotein E4 transgenic mice with pre-symptomatic Alzheimer’s disease. J Cereb Blood Flow Metab. 2017;37:217–226. doi: 10.1177/0271678X15621575.
    1. Sengupta S, Peterson TR, Laplante M, Oh S, Sabatini DM. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing. Nature. 2010;468:1100–1104. doi: 10.1038/nature09584.
    1. Lin AL, Coman D, Jiang L, Rothman DL, Hyder F. Caloric restriction impedes age-related decline of mitochondrial function and neuronal activity. J Cereb Blood Flow Metab. 2014;34:1440–1443. doi: 10.1038/jcbfm.2014.114.
    1. Lin AL, Zhang W, Gao X, Watts L. Caloric restriction increases ketone bodies metabolism and preserves blood flow in aging brain. Neurobiology of aging. 2015;36:2296–2303. doi: 10.1016/j.neurobiolaging.2015.03.012.
    1. Guo J, Bakshi V, Lin AL. Early Shifts of Brain Metabolism by Caloric Restriction Preserve White Matter Integrity and Long-Term Memory in Aging Mice. Front Aging Neurosci. 2015;7:213.
    1. Vijay N, Morris ME. Role of monocarboxylate transporters in drug delivery to the brain. Curr Pharm Des. 2014;20:1487–1498. doi: 10.2174/13816128113199990462.
    1. Steele RD. Blood-brain barrier transport of the alpha-keto acid analogs of amino acids. Fed Proc. 1986;45:2060–2064.
    1. Lam YY, et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obesity (Silver Spring) 2015;23:1429–1439. doi: 10.1002/oby.21122.
    1. Janssen AW, Kersten S. The role of the gut microbiota in metabolic health. FASEB J. 2015;29:3111–3123. doi: 10.1096/fj.14-269514.
    1. Ley RE. Obesity and the human microbiome. Curr Opin Gastroenterol. 2010;26:5–11. doi: 10.1097/MOG.0b013e328333d751.
    1. Hur KY, Lee MSG. Microbiota and Metabolic Disorders. Diabetes Metab J. 2015;39:198–203. doi: 10.4093/dmj.2015.39.3.198.
    1. Schlender L, et al. Efficacy and safety of metformin in the management of type 2 diabetes mellitus in older adults: a systematic review for the development of recommendations to reduce potentially inappropriate prescribing. BMC Geriatr. 2017;17:227. doi: 10.1186/s12877-017-0574-5.
    1. Crovesy, L., Ostrowski, M., Ferreira, D., Rosado, E. L. & Soares-Mota, M. Effect of Lactobacillus on body weight and body fat in overweight subjects: a systematic review of randomized controlled clinical trials. Int J Obes (Lond) (2017).
    1. Neth BJ, Suzanne C. Insulin Resistance and Alzheimer’s Disease: Bioenergetic Linkages Frontiers in Aging. Neuroscience. 2017;9:345.
    1. Arnold, S. E. et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nature reviews. Neurology (2018).
    1. Bluher M. Adipose tissue dysfunction in obesity. Exp Clin Endocrinol Diabetes. 2009;117:241–250. doi: 10.1055/s-0029-1192044.
    1. Mohamed HE, El-Swefy SE, Rashed LA, Abd El-Latif SK. Biochemical effect of a ketogenic diet on the brains of obese adult rats. J Clin Neurosci. 2010;17:899–904. doi: 10.1016/j.jocn.2009.11.005.
    1. Pawlosky RJ, et al. Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer’s disease. Journal of neurochemistry. 2017;141:195–207. doi: 10.1111/jnc.13958.
    1. Augustin K, et al. Mechanisms of action for the medium-chain triglyceride ketogenic diet in neurological and metabolic disorders. Lancet Neurol. 2018;17:84–93. doi: 10.1016/S1474-4422(17)30408-8.
    1. Boraxbekk CJ, et al. Diet-Induced Weight Loss Alters Functional Brain Responses during an Episodic Memory Task. Obes Facts. 2015;8:261–272. doi: 10.1159/000437157.
    1. Gibas MK, Gibas KJ. Induced and controlled dietary ketosis as a regulator of obesity and metabolic syndrome pathologies. Diabetes Metab Syndr. 2017;11(1):S385–S390. doi: 10.1016/j.dsx.2017.03.022.
    1. Abbasi J. Interest in the Ketogenic Diet Grows for Weight Loss and Type 2 Diabetes. JAMA. 2018;319:215–217. doi: 10.1001/jama.2017.20639.
    1. Sun Y, Su Y, Zhu W. Microbiome-Metabolome Responses in the Cecum and Colon of Pig to a High Resistant Starch Diet. Front Microbiol. 2016;7:779.
    1. Laukens D, Brinkman BM, Raes J, De Vos M, Vandenabeele P. Heterogeneity of the gut microbiome in mice: guidelines for optimizing experimental design. FEMS Microbiol Rev. 2016;40:117–132. doi: 10.1093/femsre/fuv036.
    1. Kilkenny C, et al. Animal research: reporting in vivo experiments–the ARRIVE guidelines. J Cereb Blood Flow Metab. 2011;31:991–993. doi: 10.1038/jcbfm.2010.220.
    1. Hoffman JD, et al. Age Drives Distortion of BrainMetabolic, Vascular and Cognitive Functions, and the Gut Microbiome. Front Aging Neurosci. 2017;9:298. doi: 10.3389/fnagi.2017.00298.
    1. Walters, W. et al. Improved Bacterial 16S rRNA Gene (V4 and V4-5) and Fungal Internal Transcribed Spacer Marker Gene Primers for Microbial Community Surveys. mSystems1 (2016).
    1. Bybee SM, et al. Targeted amplicon sequencing (TAS): a scalable next-gen approach to multilocus, multitaxa phylogenetics. Genome Biol Evol. 2011;3:1312–1323. doi: 10.1093/gbe/evr106.
    1. Moonsamy PV, et al. High throughput HLA genotyping using 454 sequencing and the Fluidigm Access Array System for simplified amplicon library preparation. Tissue Antigens. 2013;81:141–149. doi: 10.1111/tan.12071.
    1. Zhang J, Kobert K, Flouri T, Stamatakis A. Bioinformatics. 2014. PEAR: a fast and accurate Illumina Paired-End reAd mergeR; pp. 614–620.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460–2461. doi: 10.1093/bioinformatics/btq461.
    1. McDonald D, et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J. 2012;6:610–618. doi: 10.1038/ismej.2011.139.
    1. Clarke, K. R. & Warwick, R. M. An approach to statistical analysis and interpretation. Change in marine communities2 (1994).

Source: PubMed

3
Iratkozz fel