Generic placeholder image

Current Vascular Pharmacology

Editor-in-Chief

ISSN (Print): 1570-1611
ISSN (Online): 1875-6212

Review Article

Intermittent Fasting as Possible Treatment for Heart Failure

Author(s): Salvador Garza-González, Bianca Nieblas, María M. Solbes-Gochicoa, Julio Altamirano and Noemí García*

Volume 20, Issue 3, 2022

Published on: 13 July, 2022

Page: [260 - 271] Pages: 12

DOI: 10.2174/1570161120666220610151915

Price: $65

Abstract

Western-style diet often leads to food overconsumption, which triggers the development of comorbidities, such as obesity, insulin resistance, hypercholesterolemia, hypertriglyceridemia, type 2 diabetes, and heart failure (HF). Several studies suggest that intermittent fasting (IF) protects against the development of those morbidities. This study presents evidence of the beneficial effects of IF on HF. Based on the current evidence, we discuss the potential molecular mechanisms by which IF works and where liver ketone bodies (KBs) play important roles. There is evidence that IF promotes a metabolic switch in highly metabolic organs, such as the heart, which increases the use of KBs during fasting. However, besides their role as energy substrates, KBs participate in the signaling pathways that control the expression of genes involved in oxidative stress protection and metabolism. Several molecular factors, such as adenosine monophosphate-activated protein kinase (AMPK), peroxisome proliferatoractivated receptor, fibroblast growth factor 21 (FGF21), sirtuins, and nuclear factor erythroid 2-related factor 2 (Nrf2) are involved. Furthermore, IF appears to maintain circadian rhythm, which is essential for highly metabolically active organs. Finally, we highlight the important research topics that need to be pursued to improve current knowledge and strengthen the potential of IF as a preventive and therapeutic approach to HF.

Keywords: Caloric restriction, cardiovascular diseases, insulin resistance, obesity, metabolic syndrome, oxidative stress, ketosis, intermittent fasting.

Graphical Abstract
[1]
Bozkurt B, Coats AJS, Tsutsui H, et al. Universal definition and classification of heart failure: A report of the heart failure society of amer-ica, heart failure association of the european society of cardiology, japanese heart failure society and writing committee of the universal definition of heart failure: Endorsed by the canadian heart failure society, heart failure association of india, cardiac society of australia and new zealand, and chinese heart failure association. Eur J Heart Fail 2021; 23(3): 352-80.
[http://dx.doi.org/10.1002/ejhf.2115] [PMID: 33605000]
[2]
Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management of heart failure: A report of the american college of cardiology foundation/american heart association task force on practice guidelines. J Am Coll Cardiol 2013; 62(16): e147-239.
[http://dx.doi.org/10.1016/j.jacc.2013.05.019] [PMID: 23747642]
[3]
Bloom MW, Greenberg B, Jaarsma T, et al. Heart failure with reduced ejection fraction. Nat Rev Dis Primers 2017; 3(1): 17058.
[http://dx.doi.org/10.1038/nrdp.2017.58] [PMID: 28836616]
[4]
Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2017; 14(10): 591-602.
[http://dx.doi.org/10.1038/nrcardio.2017.65] [PMID: 28492288]
[5]
Pfeffer MA, Shah AM, Borlaug BA. Heart failure with preserved ejection fraction in perspective. Circ Res 2019; 124(11): 1598-617.
[http://dx.doi.org/10.1161/CIRCRESAHA.119.313572] [PMID: 31120821]
[6]
Hunter WG, Kelly JP, McGarrah RW III, Kraus WE, Shah SH. Metabolic dysfunction in heart failure: Diagnostic, prognostic, and patho-physiologic insights from metabolomic profiling. Curr Heart Fail Rep 2016; 13(3): 119-31.
[http://dx.doi.org/10.1007/s11897-016-0289-5] [PMID: 27216948]
[7]
McCommis KS, Kovacs A, Weinheimer CJ, et al. Nutritional modulation of heart failure in mitochondrial pyruvate carrier-deficient mice. Nat Metab 2020; 2(11): 1232-47.
[http://dx.doi.org/10.1038/s42255-020-00296-1] [PMID: 33106690]
[8]
Dong TA, Sandesara PB, Dhindsa DS, et al. Intermittent fasting: A heart healthy dietary pattern? Am J Med 2020; 133(8): 901-7.
[http://dx.doi.org/10.1016/j.amjmed.2020.03.030] [PMID: 32330491]
[9]
Malinowski B, Zalewska K, Węsierska A, et al. Intermittent fasting in cardiovascular disorders-an overview. Nutrients 2019; 11(3): 673.
[http://dx.doi.org/10.3390/nu11030673] [PMID: 30897855]
[10]
Varady KA, Bhutani S, Church EC, Klempel MC. Short-term modified alternate-day fasting: A novel dietary strategy for weight loss and cardioprotection in obese adults. Am J Clin Nutr 2009; 90(5): 1138-43.
[http://dx.doi.org/10.3945/ajcn.2009.28380] [PMID: 19793855]
[11]
Patterson RE, Sears DD. Metabolic effects of intermittent fasting. Annu Rev Nutr 2017; 37(1): 371-93.
[http://dx.doi.org/10.1146/annurev-nutr-071816-064634] [PMID: 28715993]
[12]
Teng NIMF, Shahar S, Rajab NF, Manaf ZA, Johari MH, Ngah WZ. Improvement of metabolic parameters in healthy older adult men fol-lowing a fasting calorie restriction intervention. Aging Male 2013; 16(4): 177-83.
[http://dx.doi.org/10.3109/13685538.2013.832191] [PMID: 24044618]
[13]
Hoddy KK, Kroeger CM, Trepanowski JF, Barnosky A, Bhutani S, Varady KA. Meal timing during alternate day fasting: Impact on body weight and cardiovascular disease risk in obese adults. Obesity (Silver Spring) 2014; 22(12): 2524-31.
[http://dx.doi.org/10.1002/oby.20909] [PMID: 25251676]
[14]
Schroder JD, Falqueto H, Mânica A, et al. Effects of time-restricted feeding in weight loss, metabolic syndrome and cardiovascular risk in obese women. J Transl Med 2021; 19(1): 3.
[http://dx.doi.org/10.1186/s12967-020-02687-0] [PMID: 33407612]
[15]
Cienfuegos S, Gabel K, Kalam F, et al. Effects of 4- and 6-h time-restricted feeding on weight and cardiometabolic health: A randomized controlled trial in adults with obesity. Cell Metab 2020; 32(3): 366-378.e3.
[http://dx.doi.org/10.1016/j.cmet.2020.06.018] [PMID: 32673591]
[16]
de Cabo R, Mattson MP. Effects of intermittent fasting on health, aging, and disease. N Engl J Med 2019; 381(26): 2541-51.
[http://dx.doi.org/10.1056/NEJMra1905136] [PMID: 31881139]
[17]
Anton SD, Moehl K, Donahoo WT, et al. Flipping the metabolic switch: Understanding and applying the health benefits of fasting. Obesity (Silver Spring) 2018; 26(2): 254-68.
[http://dx.doi.org/10.1002/oby.22065] [PMID: 29086496]
[18]
Newman JC, Verdin E. Ketone bodies as signaling metabolites. Trends Endocrinol Metab 2014; 25(1): 42-52.
[http://dx.doi.org/10.1016/j.tem.2013.09.002] [PMID: 24140022]
[19]
Higashida K, Fujimoto E, Higuchi M, Terada S. Effects of alternate-day fasting on high-fat diet-induced insulin resistance in rat skeletal muscle. Life Sci 2013; 93(5-6): 208-13.
[http://dx.doi.org/10.1016/j.lfs.2013.06.007] [PMID: 23782997]
[20]
Rohner M, Heiz R, Feldhaus S, Bornstein SR. Hepatic-metabolite-based intermittent fasting enables a sustained reduction in insulin re-sistance in type 2 diabetes and metabolic syndrome. Horm Metab Res 2021; 53(8): 529-40.
[http://dx.doi.org/10.1055/a-1510-8896] [PMID: 34192792]
[21]
Sutton EF, Beyl R, Early KS, Cefalu WT, Ravussin E, Peterson CM. Early time-restricted feeding improves insulin sensitivity, blood pres-sure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab 2018; 27(6): 1212-1221.e3.
[http://dx.doi.org/10.1016/j.cmet.2018.04.010] [PMID: 29754952]
[22]
Weng ML, Chen WK, Chen XY, et al. Fasting inhibits aerobic glycolysis and proliferation in colorectal cancer via the Fdft1-mediated AKT/mTOR/HIF1α pathway suppression. Nat Commun 2020; 11(1): 1869.
[http://dx.doi.org/10.1038/s41467-020-15795-8] [PMID: 32313017]
[23]
Singh R, Lakhanpal D, Kumar S, et al. Late-onset intermittent fasting dietary restriction as a potential intervention to retard age-associated brain function impairments in male rats. Age (Dordr) 2012; 34(4): 917-33.
[http://dx.doi.org/10.1007/s11357-011-9289-2] [PMID: 21861096]
[24]
Kaptoge S, Pennells L, de Bacquer D, et al. World Health Organization cardiovascular disease risk charts: Revised models to estimate risk in 21 global regions. Lancet Glob Health 2019; 7(10): e1332-45.
[http://dx.doi.org/10.1016/S2214-109X(19)30318-3] [PMID: 31488387]
[25]
Abiri B, Vafa M. Dietary restriction, cardiovascular aging and age-related cardiovascular diseases: a review of the evidence. Adv Exp Med Biol 2019; 1178: 113-27.
[26]
Harvie MN, Pegington M, Mattson MP, et al. 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(5): 714-27.
[http://dx.doi.org/10.1038/ijo.2010.171] [PMID: 20921964]
[27]
Guo Y, Luo S, Ye Y, Yin S, Fan J, Xia M. Intermittent fasting improves cardiometabolic risk factors and alters gut microbiota in metabolic syndrome patients. J Clin Endocrinol Metab 2021; 106(1): 64-79.
[http://dx.doi.org/10.1210/clinem/dgaa644] [PMID: 33017844]
[28]
Sundfør TM, Svendsen M, Tonstad S. Effect of intermittent versus continuous energy restriction on weight loss, maintenance and cardi-ometabolic risk: A randomized 1-year trial. Nutr Metab Cardiovasc Dis 2018; 28(7): 698-706.
[http://dx.doi.org/10.1016/j.numecd.2018.03.009] [PMID: 29778565]
[29]
Headland ML, Clifton PM, Keogh JB. Effect of intermittent compared to continuous energy restriction on weight loss and weight mainte-nance after 12 months in healthy overweight or obese adults. Int J Obes 2019; 43(10): 2028-36.
[http://dx.doi.org/10.1038/s41366-018-0247-2] [PMID: 30470804]
[30]
Antoni R, Johnston KL, Collins AL, Robertson MD. Intermittent v. continuous energy restriction: Differential effects on postprandial glu-cose and lipid metabolism following matched weight loss in overweight/obese participants. Br J Nutr 2018; 119(5): 507-16.
[http://dx.doi.org/10.1017/S0007114517003890] [PMID: 29508693]
[31]
Liu B, Hutchison AT, Thompson CH, Lange K, Wittert GA, Heilbronn LK. Effects of intermittent fasting or calorie restriction on markers of lipid metabolism in human skeletal muscle. J Clin Endocrinol Metab 2021; 106(3): e1389-99.
[http://dx.doi.org/10.1210/clinem/dgaa707] [PMID: 33031557]
[32]
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(3): 462-476.e6.
[http://dx.doi.org/10.1016/j.cmet.2019.07.016] [PMID: 31471173]
[33]
Gabel K, Hoddy KK, Haggerty N, et al. Effects of 8-hour time restricted feeding on body weight and metabolic disease risk factors in obese adults: A pilot study. Nutr Healthy Aging 2018; 4(4): 345-53.
[http://dx.doi.org/10.3233/NHA-170036] [PMID: 29951594]
[34]
Liang B-J, Liao S-R, Huang W-X, Huang C, Liu HS, Shen WZ. Intermittent fasting therapy promotes insulin sensitivity by inhibiting NLRP3 inflammasome in rat model. Ann Palliat Med 2021; 10(5): 5299-309.
[http://dx.doi.org/10.21037/apm-20-2410] [PMID: 34107698]
[35]
Hołowko J, Michalczyk MM, Zając A, et al. Six weeks of calorie restriction improves body composition and lipid profile in obese and overweight former athletes. Nutrients 2019; 11(7): 1461.
[http://dx.doi.org/10.3390/nu11071461] [PMID: 31252598]
[36]
Catenacci VA, Pan Z, Ostendorf D, et al. A randomized pilot study comparing zero-calorie alternate-day fasting to daily caloric restriction in adults with obesity. Obesity (Silver Spring) 2016; 24(9): 1874-83.
[http://dx.doi.org/10.1002/oby.21581] [PMID: 27569118]
[37]
Klempel MC, Kroeger CM, Varady KA. Alternate day fasting (ADF) with a high-fat diet produces similar weight loss and cardio-protection as ADF with a low-fat diet. Metabolism 2013; 62(1): 137-43.
[http://dx.doi.org/10.1016/j.metabol.2012.07.002] [PMID: 22889512]
[38]
Varady KA, Bhutani S, Klempel MC, et al. Alternate day fasting for weight loss in normal weight and overweight subjects: A randomized controlled trial. Nutr J 2013; 12(1): 146.
[http://dx.doi.org/10.1186/1475-2891-12-146] [PMID: 24215592]
[39]
Fontana L, Meyer TE, Klein S, Holloszy JO. Long-term calorie restriction is highly effective in reducing the risk for atherosclerosis in humans. Proc Natl Acad Sci USA 2004; 101(17): 6659-63.
[http://dx.doi.org/10.1073/pnas.0308291101] [PMID: 15096581]
[40]
Cai H, Qin Y-L, Shi Z-Y, et al. Effects of alternate-day fasting on body weight and dyslipidaemia in patients with non-alcoholic fatty liver disease: A randomised controlled trial. BMC Gastroenterol 2019; 19(1): 219.
[http://dx.doi.org/10.1186/s12876-019-1132-8] [PMID: 31852444]
[41]
Hoddy KK, Gibbons C, Kroeger CM, et al. Changes in hunger and fullness in relation to gut peptides before and after 8 weeks of alternate day fasting. Clin Nutr 2016; 35(6): 1380-5.
[http://dx.doi.org/10.1016/j.clnu.2016.03.011] [PMID: 27062219]
[42]
Ibrahim M, Davies MJ, Ahmad E, et al. Recommendations for management of diabetes during Ramadan: Update 2020, applying the prin-ciples of the ADA/EASD consensus. BMJ Open Diabetes Res Care 2020; 8(1): e001248.
[http://dx.doi.org/10.1136/bmjdrc-2020-001248] [PMID: 32366501]
[43]
Hassanein M, Bashier A, Randeree H, et al. Use of SGLT2 inhibitors during Ramadan: An expert panel statement. Diabetes Res Clin Pract 2020; 169: 108465.
[http://dx.doi.org/10.1016/j.diabres.2020.108465] [PMID: 32971151]
[44]
Abdul Kadir A, Clarke K, Evans RD. Cardiac ketone body metabolism. Biochim Biophys Acta Mol Basis Dis 2020; 1866(6): 165739.
[http://dx.doi.org/10.1016/j.bbadis.2020.165739] [PMID: 32084511]
[45]
Cantó 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(3): 213-9.
[http://dx.doi.org/10.1016/j.cmet.2010.02.006] [PMID: 20197054]
[46]
Gälman C, Lundåsen T, Kharitonenkov A, et al. The circulating metabolic regulator FGF21 is induced by prolonged fasting and PPARal-pha activation in man. Cell Metab 2008; 8(2): 169-74.
[http://dx.doi.org/10.1016/j.cmet.2008.06.014] [PMID: 18680716]
[47]
Montagner A, Polizzi A, Fouché E, et al. Liver PPARα is crucial for whole-body fatty acid homeostasis and is protective against NAFLD. Gut 2016; 65(7): 1202-14.
[http://dx.doi.org/10.1136/gutjnl-2015-310798] [PMID: 26838599]
[48]
Lundåsen T, Hunt MC, Nilsson L-M, et al. PPARalpha is a key regulator of hepatic FGF21. Biochem Biophys Res Commun 2007; 360(2): 437-40.
[http://dx.doi.org/10.1016/j.bbrc.2007.06.068] [PMID: 17601491]
[49]
Inagaki T, Dutchak P, Zhao G, et al. Endocrine regulation of the fasting response by PPARalpha-mediated induction of fibroblast growth factor 21. Cell Metab 2007; 5(6): 415-25.
[http://dx.doi.org/10.1016/j.cmet.2007.05.003] [PMID: 17550777]
[50]
Badman MK, Pissios P, Kennedy AR, Koukos G, Flier JS, Maratos-Flier E. Hepatic fibroblast growth factor 21 is regulated by PPARalpha and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab 2007; 5(6): 426-37.
[http://dx.doi.org/10.1016/j.cmet.2007.05.002] [PMID: 17550778]
[51]
Newman JC, Verdin E. β-Hydroxybutyrate: A signaling metabolite. Annu Rev Nutr 2017; 37(1): 51-76.
[http://dx.doi.org/10.1146/annurev-nutr-071816-064916] [PMID: 28826372]
[52]
Offermanns S, Colletti SL, Lovenberg TW, Semple G, Wise A, IJzerman AP. International union of basic and clinical pharmacology. lxxxii: Nomenclature and classification of hydroxy-carboxylic acid receptors (GPR81, GPR109A, and GPR109B). Pharmacol Rev 2011; 63(2): 269-90.
[http://dx.doi.org/10.1124/pr.110.003301] [PMID: 21454438]
[53]
Boden G. Obesity, insulin resistance and free fatty acids. Curr Opin Endocrinol Diabetes Obes 2011; 18(2): 139-43.
[http://dx.doi.org/10.1097/MED.0b013e3283444b09] [PMID: 21297467]
[54]
Vela-Guajardo JE, Garza-González S, García N. Glucolipotoxicity-induced oxidative stress is related to mitochondrial dysfunction and apoptosis of pancreatic β-cell. Curr Diabetes Rev 2021; 17(5): e031120187541.
[http://dx.doi.org/10.2174/1573399816666201103142102] [PMID: 33143630]
[55]
Stein LR, Imai S. The dynamic regulation of NAD metabolism in mitochondria. Trends Endocrinol Metab 2012; 23(9): 420-8.
[http://dx.doi.org/10.1016/j.tem.2012.06.005] [PMID: 22819213]
[56]
Imai S, Guarente L. NAD+ and sirtuins in aging and disease. Trends Cell Biol 2014; 24(8): 464-71.
[http://dx.doi.org/10.1016/j.tcb.2014.04.002] [PMID: 24786309]
[57]
Dabke P, Das AM. Mechanism of action of ketogenic diet treatment: Impact of decanoic acid and beta-hydroxybutyrate on sirtuins and energy metabolism in hippocampal murine neurons. Nutrients 2020; 12(8): 2379.
[http://dx.doi.org/10.3390/nu12082379] [PMID: 32784510]
[58]
Lewis KN, Mele J, Hayes JD, Buffenstein R. Nrf2, a guardian of healthspan and gatekeeper of species longevity. Integr Comp Biol 2010; 50(5): 829-43.
[http://dx.doi.org/10.1093/icb/icq034] [PMID: 21031035]
[59]
Bellezza I, Giambanco I, Minelli A, Donato R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim Biophys Acta Mol Cell Res 2018; 1865(5): 721-33.
[http://dx.doi.org/10.1016/j.bbamcr.2018.02.010] [PMID: 29499228]
[60]
Mutter FE, Park BK, Copple IM. Value of monitoring Nrf2 activity for the detection of chemical and oxidative stress. Biochem Soc Trans 2015; 43(4): 657-62.
[http://dx.doi.org/10.1042/BST20150044] [PMID: 26551708]
[61]
Evans RD, Clarke K. Myocardial substrate metabolism in heart disease. Front Biosci (Schol Ed) 2012; 4(2): 556-80.
[http://dx.doi.org/10.2741/s285] [PMID: 22202077]
[62]
Ho KL, Zhang L, Wagg C, et al. Increased ketone body oxidation provides additional energy for the failing heart without improving cardi-ac efficiency. Cardiovasc Res 2019; 115(11): 1606-16.
[http://dx.doi.org/10.1093/cvr/cvz045] [PMID: 30778524]
[63]
Quigley AF, Kapsa RMI, Esmore D, Hale G, Byrne E. Mitochondrial respiratory chain activity in idiopathic dilated cardiomyopathy. J Card Fail 2000; 6(1): 47-55.
[http://dx.doi.org/10.1016/S1071-9164(00)00011-7] [PMID: 10746819]
[64]
Sheeran FL, Pepe S. Posttranslational modifications and dysfunction of mitochondrial enzymes in human heart failure. Am J Physiol Endocrinol Metab 2016; 311(2): E449-60.
[http://dx.doi.org/10.1152/ajpendo.00127.2016] [PMID: 27406740]
[65]
Karwi QG, Uddin GM, Ho KL, Lopaschuk GD. Loss of metabolic flexibility in the failing heart. Front Cardiovasc Med 2018; 5: 68.
[http://dx.doi.org/10.3389/fcvm.2018.00068] [PMID: 29928647]
[66]
Bedi KC Jr, Snyder NW, Brandimarto J, et al. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ke-tone utilization in advanced human heart failure. Circulation 2016; 133(8): 706-16.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.115.017545] [PMID: 26819374]
[67]
Aubert G, Martin OJ, Horton JL, et al. The failing heart relies on ketone bodies as a fuel. Circulation 2016; 133(8): 698-705.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.115.017355] [PMID: 26819376]
[68]
Horton JL, Davidson MT, Kurishima C, et al. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 2019; 4(4): 1-19.
[http://dx.doi.org/10.1172/jci.insight.124079] [PMID: 30668551]
[69]
Uchihashi M, Hoshino A, Okawa Y, et al. Cardiac-specific bdh1 overexpression ameliorates oxidative stress and cardiac remodeling in pressure overload-induced heart failure. Circ Heart Fail 2017; 10(12): 1-11.
[http://dx.doi.org/10.1161/CIRCHEARTFAILURE.117.004417] [PMID: 29242353]
[70]
Snorek M, Hodyc D, Sedivý V, et al. Short-term fasting reduces the extent of myocardial infarction and incidence of reperfusion arrhyth-mias in rats. Physiol Res 2012; 61(6): 567-74.
[http://dx.doi.org/10.33549/physiolres.932338] [PMID: 23098657]
[71]
Planavila A, Redondo-Angulo I, Villarroya F. FGF21 and cardiac physiopathology. Front Endocrinol (Lausanne) 2015; 6: 133.
[http://dx.doi.org/10.3389/fendo.2015.00133] [PMID: 26379627]
[72]
Brahma MK, Adam RC, Pollak NM, et al. Fibroblast growth factor 21 is induced upon cardiac stress and alters cardiac lipid homeostasis. J Lipid Res 2014; 55(11): 2229-41.
[http://dx.doi.org/10.1194/jlr.M044784] [PMID: 25176985]
[73]
Planavila A, Redondo I, Hondares E, et al. Fibroblast growth factor 21 protects against cardiac hypertrophy in mice. Nat Commun 2013; 4(1): 2019.
[http://dx.doi.org/10.1038/ncomms3019] [PMID: 23771152]
[74]
Planavila A, Redondo-Angulo I, Ribas F, et al. Fibroblast growth factor 21 protects the heart from oxidative stress. Cardiovasc Res 2015; 106(1): 19-31.
[http://dx.doi.org/10.1093/cvr/cvu263] [PMID: 25538153]
[75]
Li S, Zhu Z, Xue M, et al. Fibroblast growth factor 21 protects the heart from angiotensin II-induced cardiac hypertrophy and dysfunction via SIRT1. Biochim Biophys Acta Mol Basis Dis 2019; 1865(6): 1241-52.
[http://dx.doi.org/10.1016/j.bbadis.2019.01.019] [PMID: 30677512]
[76]
Dwaib HS, AlZaim I, Eid AH, Obeid O, El-Yazbi AF. Modulatory effect of intermittent fasting on adipose tissue inflammation: Ameliora-tion of cardiovascular dysfunction in early metabolic impairment. Front Pharmacol 2021; 12: 626313.
[http://dx.doi.org/10.3389/fphar.2021.626313] [PMID: 33897419]
[77]
Ahmet I, Wan R, Mattson MP, Lakatta EG, Talan M. Cardioprotection by intermittent fasting in rats. Circulation 2005; 112(20): 3115-21.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.105.563817] [PMID: 16275865]
[78]
Castello L, Froio T, Maina M, et al. Alternate-day fasting protects the rat heart against age-induced inflammation and fibrosis by inhibiting oxidative damage and NF-kB activation. Free Radic Biol Med 2010; 48(1): 47-54.
[http://dx.doi.org/10.1016/j.freeradbiomed.2009.10.003] [PMID: 19818847]
[79]
Katare RG, Kakinuma Y, Arikawa M, Yamasaki F, Sato T. Chronic intermittent fasting improves the survival following large myocardial ischemia by activation of BDNF/VEGF/PI3K signaling pathway. J Mol Cell Cardiol 2009; 46(3): 405-12.
[http://dx.doi.org/10.1016/j.yjmcc.2008.10.027] [PMID: 19059263]
[80]
Khurana R, Simons M, Martin JF, Zachary IC. Role of angiogenesis in cardiovascular disease: A critical appraisal. Circulation 2005; 112(12): 1813-24.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.105.535294] [PMID: 16172288]
[81]
Ahmet I, Wan R, Mattson MP, Lakatta EG, Talan MI. Chronic alternate-day fasting results in reduced diastolic compliance and diminished systolic reserve in rats. J Card Fail 2010; 16(10): 843-53.
[http://dx.doi.org/10.1016/j.cardfail.2010.05.007] [PMID: 20932467]
[82]
Okoshi K, Cezar MDM, Polin MAM, et al. Influence of intermittent fasting on myocardial infarction-induced cardiac remodeling. BMC Cardiovasc Disord 2019; 19(1): 126.
[http://dx.doi.org/10.1186/s12872-019-1113-4] [PMID: 31138145]
[83]
Rodríguez-Colón S, He F, Bixler EO, et al. The circadian pattern of cardiac autonomic modulation and obesity in adolescents. Clin Auton Res 2014; 24(6): 265-73.
[http://dx.doi.org/10.1007/s10286-014-0257-7] [PMID: 25358502]
[84]
Steinberger J, Daniels SR, Eckel RH, et al. Progress and challenges in metabolic syndrome in children and adolescents: A scientific state-ment from the american heart association atherosclerosis, hypertension, and obesity in the young committee of the council on cardiovas-cular disease in the young; council on cardiovascular nursing; and council on nutrition, physical activity, and metabolism. Circulation 2009; 119(4): 628-47.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.108.191394] [PMID: 19139390]
[85]
Pagidipati NJ, Zheng Y, Green JB, et al. Association of obesity with cardiovascular outcomes in patients with type 2 diabetes and cardio-vascular disease: Insights from TECOS. Am Heart J 2020; 219: 47-57.
[http://dx.doi.org/10.1016/j.ahj.2019.09.016] [PMID: 31707324]
[86]
Morris CJ, Purvis TE, Hu K, Scheer FA. Circadian misalignment increases cardiovascular disease risk factors in humans. Proc Natl Acad Sci USA 2016; 113(10): E1402-11.
[http://dx.doi.org/10.1073/pnas.1516953113] [PMID: 26858430]
[87]
Durgan DJ, Young ME. The cardiomyocyte circadian clock: Emerging roles in health and disease. Circ Res 2010; 106(4): 647-58.
[http://dx.doi.org/10.1161/CIRCRESAHA.109.209957] [PMID: 20203314]
[88]
Azadbakht L, Kelishadi R, Khodarahmi M, et al. The association of sleep duration and cardiometabolic risk factors in a national sample of children and adolescents: The CASPIAN III study. Nutrition 2013; 29(9): 1133-41.
[http://dx.doi.org/10.1016/j.nut.2013.03.006] [PMID: 23927946]
[89]
Zarrinpar A, Chaix A, Panda S. Daily eating patterns and their impact on health and disease. Trends Endocrinol Metab 2016; 27(2): 69-83.
[http://dx.doi.org/10.1016/j.tem.2015.11.007] [PMID: 26706567]
[90]
Panda S. Circadian physiology of metabolism. Science 2016; 354(6315): 1008-15.
[http://dx.doi.org/10.1126/science.aah4967] [PMID: 27885007]
[91]
Chaix A, Zarrinpar A, Panda S. The circadian coordination of cell biology. J Cell Biol 2016; 215(1): 15-25.
[http://dx.doi.org/10.1083/jcb.201603076] [PMID: 27738003]
[92]
Sherman H, Genzer Y, Cohen R, Chapnik N, Madar Z, Froy O. Timed high-fat diet resets circadian metabolism and prevents obesity. FASEB J 2012; 26(8): 3493-502.
[http://dx.doi.org/10.1096/fj.12-208868] [PMID: 22593546]
[93]
Hatori M, Vollmers C, Zarrinpar A, et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab 2012; 15(6): 848-60.
[http://dx.doi.org/10.1016/j.cmet.2012.04.019] [PMID: 22608008]
[94]
St-Onge M-P, Grandner MA, Brown D, et al. Sleep duration and quality: Impact on lifestyle behaviors and cardiometabolic health: A sci-entific statement from the american heart association. Circulation 2016; 134(18): e367-86.
[http://dx.doi.org/10.1161/CIR.0000000000000444] [PMID: 27647451]
[95]
Melkani GC, Panda S. Time-restricted feeding for prevention and treatment of cardiometabolic disorders. J Physiol 2017; 595(12): 3691-700.
[http://dx.doi.org/10.1113/JP273094] [PMID: 28295377]
[96]
Rudic RD, McNamara P, Reilly D, et al. Bioinformatic analysis of circadian gene oscillation in mouse aorta. Circulation 2005; 112(17): 2716-24.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.105.568626] [PMID: 16230482]
[97]
Koike N, Yoo S-H, Huang H-C, et al. Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science 2012; 338(6105): 349-54.
[http://dx.doi.org/10.1126/science.1226339] [PMID: 22936566]
[98]
McGinnis GR, Tang Y, Brewer RA, et al. Genetic disruption of the cardiomyocyte circadian clock differentially influences insulin-mediated processes in the heart. J Mol Cell Cardiol 2017; 110: 80-95.
[http://dx.doi.org/10.1016/j.yjmcc.2017.07.005] [PMID: 28736261]
[99]
Bray MS, Shaw CA, Moore MWS, et al. Disruption of the circadian clock within the cardiomyocyte influences myocardial contractile function, metabolism, and gene expression. Am J Physiol Heart Circ Physiol 2008; 294(2): H1036-47.
[http://dx.doi.org/10.1152/ajpheart.01291.2007] [PMID: 18156197]
[100]
Scheer FAJL, Hilton MF, Mantzoros CS, Shea SA. Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci USA 2009; 106(11): 4453-8.
[http://dx.doi.org/10.1073/pnas.0808180106] [PMID: 19255424]
[101]
Chaix A, Zarrinpar A, Miu P, Panda S. Time-restricted feeding is a preventative and therapeutic intervention against diverse nutritional challenges. Cell Metab 2014; 20(6): 991-1005.
[http://dx.doi.org/10.1016/j.cmet.2014.11.001] [PMID: 25470547]
[102]
Gill S, Le HD, Melkani GC, Panda S. Time-restricted feeding attenuates age-related cardiac decline in Drosophila. Science 2015; 347(6227): 1265-9.
[http://dx.doi.org/10.1126/science.1256682] [PMID: 25766238]
[103]
Melkani GC, Trujillo AS, Ramos R, Bodmer R, Bernstein SI, Ocorr K. Huntington’s disease induced cardiac amyloidosis is reversed by modulating protein folding and oxidative stress pathways in the Drosophila heart. PLoS Genet 2013; 9(12): e1004024.
[http://dx.doi.org/10.1371/journal.pgen.1004024] [PMID: 24367279]
[104]
Birse RT, Choi J, Reardon K, et al. High-fat-diet-induced obesity and heart dysfunction are regulated by the TOR pathway in Drosophila. Cell Metab 2010; 12(5): 533-44.
[http://dx.doi.org/10.1016/j.cmet.2010.09.014] [PMID: 21035763]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy