Short-term time-restricted feeding is safe and feasible in non-obese healthy midlife and older adults

Christopher R Martens, Matthew J Rossman, Melissa R Mazzo, Lindsey R Jankowski, Erzsebet E Nagy, Blair A Denman, James J Richey, Sarah A Johnson, Brian P Ziemba, Yang Wang, Courtney M Peterson, Michel Chonchol, Douglas R Seals, Christopher R Martens, Matthew J Rossman, Melissa R Mazzo, Lindsey R Jankowski, Erzsebet E Nagy, Blair A Denman, James J Richey, Sarah A Johnson, Brian P Ziemba, Yang Wang, Courtney M Peterson, Michel Chonchol, Douglas R Seals

Abstract

Chronic calorie restriction (CR) improves cardiovascular function and several other physiological markers of healthspan. However, CR is impractical in non-obese older humans due to potential loss of lean mass and bone density, poor adherence, and risk of malnutrition. Time-restricted feeding (TRF), which limits the daily feeding period without requiring a reduction in calorie intake, may be a promising alternative healthspan-extending strategy for midlife and older adults; however, there is limited evidence for its feasibility and efficacy in humans. We conducted a randomized, controlled pilot study to assess the safety, tolerability, and overall feasibility of short-term TRF (eating <8 h day-1 for 6 weeks) without weight loss in healthy non-obese midlife and older adults, while gaining initial insight into potential efficacy for improving cardiovascular function and other indicators of healthspan. TRF was safe and well-tolerated, associated with excellent adherence and reduced hunger, and did not influence lean mass, bone density, or nutrient intake. Cardiovascular function was not enhanced by short-term TRF in this healthy cohort, but functional (endurance) capacity and glucose tolerance were modestly improved. These results provide a foundation for conducting larger clinical studies of TRF in midlife and older adults, including trials with a longer treatment duration.

Keywords: Aging; Calorie restriction; Healthspan; Intermittent fasting.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Feasibility of time-restricted feeding. a Compared with normal feeding (NF, black bars), all subjects successfully reduced their feeding window to within the allowed feeding duration (red lines) during time-restricted feeding (TRF, gray bars) resulting in b a reduction in the average daily feeding window. c Subjects were able to maintain a reduction in their daily feeding period throughout the 6-week TRF period with d good overall adherence. There were no adverse changes in e overall diet quality as assessed by the healthy eating index, f energy intake, g estimated energy expenditure, or h body mass. N = 22. Boxes represent median and interquartile range with 95% confidence intervals as whiskers. *p < 0.05. See also Supplementary Table 1, Supplementary Table 2, Supplementary Table 3, and Supplementary Table 4
Fig. 2
Fig. 2
Subjective appetite. Six weeks of time-restricted feeding (TRF) vs. normal feeding (NF) reduced sensations of a hunger but did not alter b desire to eat, c capacity to eat, d fullness, or e stomach fullness despite expected fluctuations between morning (fasted) and evening (fed) conditions. N = 22. Boxes represent median and interquartile range with 95% confidence intervals as whiskers. Data are in millimeters (mm) drawn on a standardized visual-analog scale. *p values < 0.05 based on two-factor repeated measures ANOVA
Fig. 3
Fig. 3
Body composition and bone density. In comparison to normal feeding (NF), time-restricted feeding (TRF) was associated with a a small change in leg fat mass that did not affect total body composition, and b no change in bone mineral density compared with normal feeding (NF). N = 22. Data are mean ± standard error of mean (SEM). *p < 0.05
Fig. 4
Fig. 4
Vascular function and blood pressure at rest. Compared with normal feeding (NF), 6 weeks of TRF did not influence a flow-mediated dilation (FMD), b carotid-femoral pulse wave velocity (PWV), c carotid artery compliance, d carotid intima-media thickness (IMT), e resting systolic blood pressure (SBP), or f resting diastolic blood pressure (DBP). N = 22. Boxes represent median and interquartile range with 95% confidence intervals as whiskers
Fig. 5
Fig. 5
Cardiorespiratory fitness. In comparison to normal feeding (NF), time-restricted feeding (TRF) was associated with a increased distance traveled during a 6-min walk test and b reduced heart rate during light and moderate intensity exercise. N = 22. Boxes represent median and interquartile range with 95% confidence intervals as whiskers. *p < 0.05 based on mixed effects model with repeated measures. See also Supplementary Fig. 2 and Supplementary Table 5
Fig. 6
Fig. 6
Glucose tolerance. Compared with normal feeding (NF), time-restricted feeding (TRF) was associated with a, b a reduction in glucose area-under-the-curve (AUC) during a 2-h OGTT, c, d no changes in insulin levels and the insulin AUC, and e no change in the insulinogenic index (a marker of pancreatic β-cell function). N = 16 participants. Boxes represent median and interquartile range with 95% confidence intervals as whiskers. *p < 0.05 based on mixed effects model with repeated measures
Fig. 7
Fig. 7
Mechanistic biomarkers. Compared with the normal feeding (NF) condition, time-restricted feeding (TRF) was not associated with any changes in a oxidized-LDL levels, b the oxidized-to-total LDL ratio, c interleukin-6 (IL-6), d C-reactive protein (CRP) or plasma ketones including e acetoacetate or f β-hydroxybutyrate. Boxes represent median and interquartile range with 95% confidence intervals as whiskers. See also Supplementary Table 6

References

    1. Ahmet I, Wan RQ, Mattson MP, Lakatta EG, Talan M. Cardioprotection by intermittent fasting in rats. Circulation. 2005;112:3115–3121. doi: 10.1161/CIRCULATIONAHA.105.563817.
    1. Anton SD, Han H, York E, Martin CK, Ravussin E, Williamson DA. Effect of calorie restriction on subjective ratings of appetite. J Hum Nutr Diet. 2009;22:141–147. doi: 10.1111/j.1365-277X.2008.00943.x.
    1. Anton SD, Lee SA, Donahoo WT, et al. The effects of time restricted feeding on overweight, older adults: a pilot study. Nutrients. 2019;11:1500. doi: 10.3390/nu11071500.
    1. Bak AM, Vendelbo MH, Christensen B, et al. Prolonged fasting-induced metabolic signatures in human skeletal muscle of lean and obese men. PLoS One. 2018;13:1–19. doi: 10.1371/journal.pone.0200817.
    1. Bell C, Jones PP, Seals DR. Oxidative stress does not modulate metabolic rate or skeletal muscle sympathetic activity with primary aging in adult humans. J Clin Endocrinol Metab. 2003;88:4950–4954. doi: 10.1210/jc.2003-030454.
    1. Bell C, Stob NR, Seals DR. Thermogenic responsiveness to beta-adrenergic stimulation is augmented in exercising versus sedentary adults: role of oxidative stress. J Physiol. 2006;570:629–635. doi: 10.1113/jphysiol.2005.098756.
    1. Berliner JA, Heinecke JW (1996) The role of oxidized lipoproteins in atherogenesis. Free Radic Biol Med 20:707–727
    1. Blomquist C, Saltin B. Cardiovascular adaptations to physical training. Annu Rev Physiol. 1977;39:221–231. doi: 10.1146/annurev.ph.39.030177.001253.
    1. Brandhorst S, Choi IY, Wei M, Cheng CW, Sedrakyan S, Navarrete G, Dubeau L, Yap LP, Park R, Vinciguerra M, di Biase S, Mirzaei H, Mirisola MG, Childress P, Ji L, Groshen S, Penna F, Odetti P, Perin L, Conti PS, Ikeno Y, Kennedy BK, Cohen P, Morgan TE, Dorff TB, Longo VD. A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab. 2015;22:86–99. doi: 10.1016/j.cmet.2015.05.012.
    1. Butler RN, Warner HR, Williams TF, et al. The aging factor in health and disease: the promise of basic research on aging. Aging Clin Exp Res. 2004;16:104–111. doi: 10.1007/BF03324538.
    1. Callahan D, Phillips E, Carabello R, et al. Assessment of lower extremity muscle power in functionally-limited elders. Aging Clin Exp Res. 2007;19:194–199. doi: 10.1007/BF03324689.
    1. Canto C, Jiang LQ, Deshmukh AS, et al. Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab. 2010;11:213–219. doi: 10.1016/j.cmet.2010.02.006.
    1. Carter JB, Banister EW, Blaber AP. Effect of endurance exercise on autonomic control of heart rate. Sports Med. 2003;33:33–46. doi: 10.2165/00007256-200333010-00003.
    1. Chen D, Bruno J, Easlon E, Lin SJ, Cheng HL, Alt FW, Guarente L. Tissue-specific regulation of SIRT1 by calorie restriction. Genes Dev. 2008;22:1753–1757. doi: 10.1101/gad.1650608.
    1. Chung KW, Kim DH, Park MH, Choi YJ, Kim ND, Lee J, Yu BP, Chung HY. Recent advances in calorie restriction research on aging. Exp Gerontol. 2013;48:1049–1053. doi: 10.1016/j.exger.2012.11.007.
    1. Colman RJ, Anderson RM, Johnson SC, et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325(80):201–204. doi: 10.1126/science.1173635.
    1. Colman RJ, Beasley TM, Allison DB, Weindruch R. Attenuation of sarcopenia by dietary restriction in rhesus monkeys. J Gerontol Ser A-Biol Sci Med Sci. 2008;63:556–559. doi: 10.1093/gerona/63.6.556.
    1. Crandall JP, Shamoon H, Cohen HW, et al. Post-challenge hyperglycemia in older adults is associated with increased cardiovascular risk profile. J Clin Endocrinol Metab. 2009;94:1595–1601. doi: 10.1210/jc.2008-1829.
    1. Csipo T, Fulop GA, Lipecz A, Tarantini S, Kiss T, Balasubramanian P, Csiszar A, Ungvari Z, Yabluchanskiy A. Short-term weight loss reverses obesity-induced microvascular endothelial dysfunction. GeroScience. 2018;40:337–346. doi: 10.1007/s11357-018-0028-9.
    1. de Picciotto NE, Gano LB, Johnson LC, et al. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell. 2016;15:522. doi: 10.1111/acel1246115:522-530.
    1. Dean W. The retardation of aging and diseases of aging by dietary restriction - Weindruch,r, Walford,rl. J Am Geriatr Soc. 1990;38:736. doi: 10.1111/j.1532-5415.1990.tb04441.x.
    1. DeVan AE, Johnson LC, Brooks FA, Evans TD, Justice JN, Cruickshank-Quinn C, Reisdorph N, Bryan NS, McQueen M, Santos-Parker JR, Chonchol MB, Bassett CJ, Sindler AL, Giordano T, Seals DR. Effects of sodium nitrite supplementation on vascular function and related small metabolite signatures in middle-aged and older adults. J Appl Physiol. 2016;120:416–425. doi: 10.1152/japplphysiol.00879.2015.
    1. Donato AJ, Walker AE, Magerko KA, Bramwell RC, Black AD, Henson GD, Lawson BR, Lesniewski LA, Seals DR. Life-long caloric restriction reduces oxidative stress and preserves nitric oxide bioavailability and function in arteries of old mice. Aging Cell. 2013;12:772–783. doi: 10.1111/acel.12103.
    1. Ensrud KE, Fullman RL, Barrett-Connor E, Cauley JA, Stefanick ML, Fink HA, Lewis CE, Orwoll E, Osteoporotic Fractures in Men Study Research Group Voluntary weight reduction in older men increases hip bone loss: the osteoporotic fractures in men study. J Clin Endocrinol Metab. 2005;90:1998–2004. doi: 10.1210/jc.2004-1805.
    1. Eskurza I, Monahan KD, Robinson JA, Seals DR. Effect of acute and chronic ascorbic acid on flow-mediated dilatation with sedentary and physically active human ageing. J Physiol. 2004;556:315–324. doi: 10.1113/jphysiol.2003.057042.
    1. Fries JF. Aging, natural death, and the compression of morbidity. N Engl J Med. 1980;303:130–135. doi: 10.1056/NEJM198007173030304.
    1. Fries JF. Perspectives from masters in rheumatology and autoimmunity: perspectives on the conventional wisdom. Arthritis Rheum. 2015;67:2806–2812. doi: 10.1002/art.39207.
    1. Gardner EM. Caloric restriction decreases survival of aged mice in response to primary influenza infection. J Gerontol Ser A Biol Sci Med Sci. 2005;60:688–694. doi: 10.1093/gerona/60.6.688.
    1. Gill S, Panda S. A smartphone app reveals erratic diurnal eating patterns in humans that can be modulated for health benefits. Cell Metab. 2015;22:789–798. doi: 10.1016/j.cmet.2015.09.005.
    1. Goodrick CL, Ingram DK, Reynolds MA, Freeman JR, Cider NL. Effects of intermittent feeding upon growth and life span in rats. Gerontology. 1982;28:233–241. doi: 10.1159/000212538.
    1. Harvie MN, Pegington M, Mattson MP, Frystyk J, Dillon B, Evans G, Cuzick J, Jebb SA, Martin B, Cutler RG, Son TG, Maudsley S, Carlson OD, Egan JM, Flyvbjerg A, Howell A. The effects of intermittent or continuous energy restriction on weight loss and metabolic disease risk markers: a randomized trial in young overweight women. Int J Obes. 2011;35:714–727. doi: 10.1038/ijo.2010.171.
    1. Hatori M, Vollmers C, Zarrinpar A, DiTacchio L, Bushong EA, Gill S, Leblanc M, Chaix A, Joens M, Fitzpatrick JA, Ellisman MH, Panda S. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 2012;15:848–860. doi: 10.1016/j.cmet.2012.04.019.
    1. Hunt ND, Li GD, Zhu M, Miller M, Levette A, Chachich ME, Spangler EL, Allard JS, Hyun DH, Ingram DK, de Cabo R. Effect of calorie restriction and refeeding on skin wound healing in the rat. Age (Omaha) 2012;34:1453–1458. doi: 10.1007/s11357-011-9321-6.
    1. Ingram DK, de Cabo R. Calorie restriction in rodents: caveats to consider. Ageing Res Rev. 2017;39:15–28. doi: 10.1016/j.arr.2017.05.008.
    1. Jablonski KL, Racine ML, Geolfos CJ, et al. Dietary sodium restriction reverses vascular endothelial dysfunction in middle-aged/older adults with moderately elevated systolic blood pressure. J Am Coll Cardiol. 2013;61:335–343. doi: 10.1016/j.jacc.2012.09.010.
    1. Jang YC, Liu Y, Hayworth CR, Bhattacharya A, Lustgarten MS, Muller FL, Chaudhuri A, Qi W, Li Y, Huang JY, Verdin E, Richardson A, van Remmen H. Dietary restriction attenuates age-associated muscle atrophy by lowering oxidative stress in mice even in complete absence of CuZnSOD. Aging Cell. 2012;11:770–782. doi: 10.1111/j.1474-9726.2012.00843.x.
    1. Johnson JB, Summer W, Cutler RG, Martin B, Hyun DH, Dixit VD, Pearson M, Nassar M, Telljohann R, Maudsley S, Carlson O, John S, Laub DR, Mattson MP. Alternate day calorie restriction improves clinical findings and reduces markers of oxidative stress and inflammation in overweight adults with moderate asthma. Free Radic Biol Med. 2007;42:665–674. doi: 10.1016/j.freeradbiomed.2006.12.005.
    1. Justice JN, Johnson LC, DeVan AE, et al. Improved motor and cognitive performance with sodium nitrite supplementation is related to small metabolite signatures: a pilot trial in middle-aged and older adults. Aging-Us. 2015;7:1004–1021. doi: 10.18632/aging.100842.
    1. Kalyani RR, Egan JM. Diabetes and altered glucose metabolism with aging. Endocrinol Metab Clin N Am. 2013;42:333–347. doi: 10.1016/j.ecl.2013.02.010.
    1. Kaplon RE, Hill SD, Bispham NZ, et al. Oral trehalose supplementation improves resistance artery endothelial function in healthy middle-aged and older adults. Aging (Albany NY) 2016;8:1167–1183. doi: 10.18632/aging.100962.
    1. Kiss T, Balasubramanian P, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Csipo T, Lipecz A, Reglodi D, Zhang XA, Bari F, Farkas E, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) treatment attenuates oxidative stress and rescues angiogenic capacity in aged cerebromicrovascular endothelial cells: a potential mechanism for the prevention of vascular cognitive impairment. GeroScience. 2019;41:619–630. doi: 10.1007/s11357-019-00074-2.
    1. Krebs-Smith SM, Pannucci TE, Subar AF, et al. Update of the healthy eating index: HEI-2015. J Acad Nutr Diet. 2018;118:1591–1602. doi: 10.1016/j.jand.2012.12.016.
    1. LaRoche DP, Melanson EL, Baumgartner MP, Bozzuto BM, Libby VM, Marshall BN. Physiological determinants of walking effort in older adults: should they be targets for physical activity intervention? GeroScience. 2018;40:305–315. doi: 10.1007/s11357-018-0032-0.
    1. Laurent S, Boutouyrie P, Asmar R, et al. Aortic stiffness is an independent predictor of all-cause and cardiovascular mortality in hypertensive patients. Hypertens. 2001;37:1236–1241. doi: 10.1161/01.HYP.37.5.1236.
    1. Laurent S, Cockcroft J, Van Bortel L, et al. Expert consensus document on arterial stiffness: methodological issues and clinical applications. Eur Heart J. 2006;27:2588–2605. doi: 10.1093/eurheartj/ehl254.
    1. Longo VD, Mattson MP. Fasting: molecular mechanisms and clinical applications. Cell Metab. 2014;19:181–192. doi: 10.1016/j.cmet.2013.12.008.
    1. Luna-Moreno D, Aguilar-Roblero R, Díaz-Muñoz M. Restricted feeding entrains rhythms of inflammation-related factors without promoting an acute-phase response. Chronobiol Int. 2009;26:1409–1429. doi: 10.3109/07420520903417003.
    1. Martens CR, Denman BA, Mazzo MR et al (2018) Chronic nicotinamide riboside supplementation is well-tolerated and effectively elevates NAD+ in healthy middle-aged and older adults. Nat Commun 9. 10.1038/s41467-018-03421-7
    1. Martens CR, Seals DR. Practical alternatives to chronic caloric restriction for optimizing vascular function with ageing. J Physiol. 2016;594:7177–7195. doi: 10.1113/JP272348.
    1. Masoro EJ. Caloric restriction and aging: an update. Exp Gerontol. 2000;35:299–305. doi: 10.1016/S0531-5565(00)00084-X.
    1. Matyi S, Jackson J, Garrett K, Deepa SS, Unnikrishnan A. The effect of different levels of dietary restriction on glucose homeostasis and metabolic memory. GeroScience. 2018;40:139–149. doi: 10.1007/s11357-018-0011-5.
    1. McCrum C, Karamanidis K, Grevendonk L, Zijlstra W, Meijer K (2019) Older adults demonstrate interlimb transfer of reactive gait adaptations to repeated unpredictable gait perturbations. GeroScience.:1–11. 10.1007/s11357-019-00130-x
    1. Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, Redpath P, Migaud ME, Apte RS, Uchida K, Yoshino J, Imai SI. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab. 2016;24:795–806. doi: 10.1016/j.cmet.2016.09.013.
    1. Mitchell SJ, Bernier M, Mattison JA et al (2018) Daily fasting improves health and survival in male mice independent of diet composition and calories. Cell Metab:1–8. 10.1111/j.1096-0031.2010.00328.x
    1. Moreau KL, Donato AJ, Seals DR, DeSouza C, Tanaka H. Regular exercise, hormone replacement therapy and the age-related decline in carotid arterial compliance in healthy women. Cardiovasc Res. 2003;57:861–868. doi: 10.1016/S0008-6363(02)00777-0.
    1. Moreira EAM, Most M, Howard J, Ravussin E. Dietary adherence to long-term controlled feeding in a calorie-restriction study in overweight men and women. Nutr Clin Pract. 2011;26:309–315. doi: 10.1177/0884533611405992.
    1. Normandin E, Houston DK, Nicklas BJ. Caloric restriction for treatment of geriatric obesity: do the benefits outweigh the risks? Curr Nutr Rep. 2015;4(2):143–155. doi: 10.1007/s13668-015-0123-9.
    1. Olshansky SJ. From lifespan to healthspan from lifespan to HealthspanFrom lifespan to Healthspan. JAMA. 2018;320:1323–1324. doi: 10.1001/jama.2018.12621.
    1. Phillips DIW, Clark PM, Hales CN, Osmond C. Understanding oral glucose tolerance: comparison of glucose or insulin measurements during the oral glucose tolerance test with specific measurements of insulin resistance and insulin secretion. Diabet Med. 1994;11:286–292. doi: 10.1111/j.1464-5491.1994.tb00273.x.
    1. Pierce GL, Beske SD, Lawson BR, Southall KL, Benay FJ, Donato AJ, Seals DR. Weight loss alone improves conduit and resistance artery endothelial function in young and older overweight/obese adults. Hypertension. 2008;52:72–79. doi: 10.1161/HYPERTENSIONAHA.108.111427.
    1. Pierce GL, Jablonski KL, Walker AE, Seibert SM, DeVan A, Black SM, Sharma S, Seals DR. Tetrahydrobiopterin supplementation enhances carotid artery compliance in healthy older men: a pilot study. Am J Hypertens. 2012;25:1050–1054. doi: 10.1038/ajh.2012.70.
    1. Racette SB, Rochon J, Uhrich ML, Villareal DT, DAS S, Fontana L, Bhapkar M, Martin CK, Redman LM, Fuss PJ, Roberts SB, Kraus WE. Effects of two years of calorie restriction on aerobic capacity and muscle strength. Med Sci Sports Exerc. 2017;49:2240–2249. doi: 10.1249/MSS.0000000000001353.
    1. Ravussin E, Beyl RA, Poggiogalle E, Hsia DS, Peterson CM. Early time-restricted feeding reduces appetite and increases fat oxidation but does not affect energy expenditure in humans. Obesity. 2019;27:1244–1254. doi: 10.1002/oby.22518.
    1. Ricci TA, Heymsfield SB, Pierson RN, Stahl T, Chowdhury HA, Shapses SA (2001) Moderate energy restriction increases bone resorption in obese postmenopausal women. Am J Clin Nutr 73:347-52. 10.1093/ajcn/73.2.347
    1. Rippe C, Lesniewski LA, Connell ML, LaRocca T, Donato A, Seals D. Short-term calorie restriction reverses vascular endothelial dysfunction in old mice by increasing nitric oxide and reducing oxidative stress. Aging Cell. 2010;9:304–312. doi: 10.1111/j.1474-9726.2010.00557.x.
    1. Rivera-Zavala JB, Molina-Aguilar C, Pérez-Mendoza M, et al. Daytime restricted feeding modifies the daily regulation of fatty acid β-oxidation and the lipoprotein profile in rats. Br J Nutr. 2017;117:930–941. doi: 10.1017/S0007114517000800.
    1. Rochon J, Bales CW, Ravussin E, Redman LM, Holloszy JO, Racette SB, Roberts SB, Das SK, Romashkan S, Galan KM, Hadley EC, Kraus WE, CALERIE Study Group Design and conduct of the CALERIE study: comprehensive assessment of the long-term effects of reducing intake of energy. J Gerontol Ser A Biol Sci Med Sci. 2011;66(A):97–108. doi: 10.1093/gerona/glq168.
    1. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Nature. 2005;434:113–118. doi: 10.1038/nature03354.
    1. Rothschild J, Hoddy KK, Jambazian P, Varady KA. Time-restricted feeding and risk of metabolic disease: a review of human and animal studies. Nutr Rev. 2014;72:308–318. doi: 10.1111/nure.12104.
    1. Santos-Parker JR, Lubieniecki KL, Rossman MJ, van Ark H, Bassett CJ, Strahler TR, Chonchol MB, Justice JN, Seals DR. Curcumin supplementation and motor-cognitive function in healthy middle-aged and older adults. Nutr Heal Aging. 2018;4:323–333. doi: 10.3233/NHA-170029.
    1. Santos-Parker JR, Strahler TR, Bassett CJ, et al. Curcumin supplementation improves vascular endothelial function in healthy middle-aged and older adults by increasing nitric oxide bioavailability and reducing oxidative stress. Aging (Albany NY) 2017;9:187–208. doi: 10.18632/aging.101149.
    1. Saydah SH, Miret M, Sung J, et al. Postchallenge hyperglycemia and mortality in a national sample of US adults. Diabetes Care. 2001;24:1397. doi: 10.2337/diacare.24.8.1397.
    1. Schafer AL. Decline in bone mass during weight loss: a cause for concern? J Bone Miner Res. 2016;31:36–39. doi: 10.1002/jbmr.2701.
    1. Schulz KF, Grimes DA. Series epidemiology 4 multiplicity in randomised trials I : endpoints and treatments. Lancet. 2005;365:1591–1595. doi: 10.1016/S0140-6736(05)66461-6.
    1. Sperandio E, Matheus A, Lauria V et al (2014) Intensity and physiological responses to the six-minute walk test in middle-aged and older adults: a comparison with cardiopulmonary exercise testing. Eur Respir J:44
    1. Stekovic S, Hofer SJ, Tripolt N, et al. Alternate day fasting improves physiological and molecular markers of aging in healthy, non-obese humans. Cell Metab. 2019;30:462–476.e5. doi: 10.1016/j.cmet.2019.07.016.
    1. Stewart TM, Bhapkar M, Das S, Galan K, Martin CK, McAdams L, Pieper C, Redman L, Roberts S, Stein RI, Rochon J, Williamson DA, CALERIE Study Group Comprehensive assessment of long-term effects of reducing intake of energy phase 2 (CALERIE phase 2) screening and recruitment: methods and results. Contemp Clin Trials. 2013;34:10–20. doi: 10.1016/j.cct.2012.08.011.
    1. Stote K, Baer D, Spears K, et al. A controlled trial of reduced meal frequency without caloric. Am J Clin Nutr. 2007;85:1–8. doi: 10.1093/ajcn/85.4.981.
    1. Sutton EF, Beyl R, Early KS, et al. Early time-restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab. 2018;27:1212–1221.e3. doi: 10.1016/j.cmet.2018.04.010.
    1. Tanaka H, DeSouza CA, Seals DR. Arterial stiffness and hormone replacement use in healthy postmenopausal women. J Gerontol Ser A Biol Sci Med Sci. 1998;53:M344–M346. doi: 10.1093/gerona/53A.5.M344.
    1. Tanaka H, Dinenno FA, Monahan KD, et al. Carotid artery wall hypertrophy with age is related to local systolic blood pressure in healthy men. Arterioscler Thromb Vasc Biol. 2001;21:82–87. doi: 10.1161/01.ATV.21.1.82.
    1. Trepanowski JF, Kroeger CM, Barnosky A, Klempel MC, Bhutani S, Hoddy KK, Gabel K, Freels S, Rigdon J, Rood J, Ravussin E, Varady KA. Effect of alternate-day fasting on weight loss, weight maintenance, and cardioprotection among metabolically healthy obese adults: a randomized clinical trial. JAMA Intern Med. 2017;177:930–938. doi: 10.1001/jamainternmed.2017.0936.
    1. Van Der Wal AMG, Bakker O, Wiersinga WM. The decrease of liver LDL receptor mRNA during fasting is related to the decrease in serum T3. Int J Biochem Cell Biol. 1998;30:209–215. doi: 10.1016/S1357-2725(97)00120-9.
    1. Varady KA, Hellerstein MK. Alternate-day fasting and chronic disease prevention: a review of human and animal trials. Am J Clin Nutr. 2007;86:7–13. doi: 10.1093/ajcn/86.1.7.
    1. Villareal DT, Fontana L, Das SK, Redman L, Smith SR, Saltzman E, Bales C, Rochon J, Pieper C, Huang M, Lewis M, Schwartz AV, CALERIE Study Group Effect of two-year caloric restriction on bone metabolism and bone mineral density in non-obese younger adults: a randomized clinical trial. J Bone Miner Res. 2016;31:40–51. doi: 10.1002/jbmr.2701.
    1. Walker AE, Eskurza I, Pierce GL, Gates PE, Seals DR. Modulation of vascular endothelial function by low-density lipoprotein cholesterol with aging: influence of habitual exercise. Am J Hypertens. 2009;22:250–256. doi: 10.1038/ajh.2008.353.
    1. Walker AE, Kaplon RE, Pierce GL, Nowlan MJ, Seals DR. Prevention of age-related endothelial dysfunction by habitual aerobic exercise in healthy humans: possible role of nuclear factor kappa B. Clin Sci. 2014;127:645–654. doi: 10.1042/CS20140030.

Source: PubMed

3
Abonnere