Journal Home > Volume 9 , Issue 1

With improvements in healthcare and lifestyle, the proportion of the aging population is rising steadily across the world. Many physiological functions are altered during aging and resemble those occurring in disease conditions involving metabolic and mitochondrial disturbances. Thus, there is an urge to better develop dietary or medicinal interventions targeting the mechanisms underlying aging and aging-related diseases. Many reports indicate that in geroscience, dietary interventions such as bioactive peptides to slow aging are a matter of 'when' rather than 'if'. Leading targets for peptides include the metabolic-mitochondrial pathway accompanied by improved nutrient sensing. Modulation of these pathways diminish aging biomarkers in various model organisms and confers protection against a growing list of age-related pathophysiologies. Food derived bioactive peptides are characterized modulators of these pathways, while some verified in vivo and even clinically approved, and numerous others are under development. This brief review focuses on the latest scientific advances in understanding the anti-aging ability of bioactive peptides as well as delineates the possible future directions in this process.


menu
Abstract
Full text
Outline
About this article

Dietary peptides in aging: Evidence and prospects

Show Author's information Khushwant S. Bhullara,bJianping Wua( )
Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
Department of Pharmacology, University of Alberta, Edmonton, AB, Canada

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

With improvements in healthcare and lifestyle, the proportion of the aging population is rising steadily across the world. Many physiological functions are altered during aging and resemble those occurring in disease conditions involving metabolic and mitochondrial disturbances. Thus, there is an urge to better develop dietary or medicinal interventions targeting the mechanisms underlying aging and aging-related diseases. Many reports indicate that in geroscience, dietary interventions such as bioactive peptides to slow aging are a matter of 'when' rather than 'if'. Leading targets for peptides include the metabolic-mitochondrial pathway accompanied by improved nutrient sensing. Modulation of these pathways diminish aging biomarkers in various model organisms and confers protection against a growing list of age-related pathophysiologies. Food derived bioactive peptides are characterized modulators of these pathways, while some verified in vivo and even clinically approved, and numerous others are under development. This brief review focuses on the latest scientific advances in understanding the anti-aging ability of bioactive peptides as well as delineates the possible future directions in this process.

Keywords: Anti-aging, Dietary peptides, Aging biomarkers

References(114)

[1]

M. Serrano, Understanding aging, N. Engl. J. Med. 376 (11) (2017) 1083-1085.

[2]

A.D. Sniderman, C.D. Furberg, Age as a modifiable risk factor for cardiovascular disease, Lancet 371 (9623) (2008) 1547-1549.

[3]

E.M. Crimmins, Y.S. Zhang, Aging populations, mortality, and life expectancy, Annu. Rev. Sociol. 45 (2019).

[4]

H. Beltrán-Sánchez, S. Soneji, E.M. Crimmins, Past, present, and future of healthy life expectancy, Cold Spring Harb. Perspect. Med. 5 (11) (2015) a025957.

[5]

S.J. Olshansky, L. Hayflick, B.A. Carnes, No truth to the fountain of youth, Sci. Am. 286 (6) (2002) 92-95.

[6]

C. Haber, Anti-aging medicine: the history: life extension and history: the continual search for the fountain of youth, J. Gerontol. A Biol. Sci. Med. Sci. 59 (6) (2004) B515-B522.

[7]

R.A. Van de Ven, D. Santos, M.C. Haigis, Mitochondrial sirtuins and molecular mechanisms of aging, Trends Mol. Med. 23 (4) (2017) 320-331.

[8]

C.M. McCay, M.F. Crowell, L.A. Maynard, Retarded growth, life span, ultimate body size and age changes in the albino rat after feeding diets restricted in calories: four figures, J. Nutr. 18 (1) (1939) 1-13.

[9]

D. Omodei, L. Fontana, Calorie restriction and prevention of age‐associated chronic disease, FEBS Lett. 585 (11) (2011) 1537-1542.

[10]

P.B. Medawar, An unsolved problem of biology, College. (1952).

[11]

M. Rose, B. Charlesworth, A test of evolutionary theories of senescence, Nature 287 (5778) (1980) 141.

[12]

G. Bell, Evolutionary and nonevolutionary theories of senescence, Am. Nat. 124 (4) (1984) 600-603.

[13]

D.B. Friedman, T.E. Johnson, A mutation in the age-1 gene in Caenorhabditis elegans lengthens life and reduces hermaphrodite fertility, Genetics 118 (1) (1988) 75-86.

[14]

C. Kenyon, J. Chang, Gensch E, et al., A C. Elegans mutant that lives twice as long as wild type, Nature 366 (6454) (1993) 461.

[15]

J. Heitman, N.R. Movva, M.N. Hall, Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast, Science 253 (5022) (1991) 905-909.

[16]

B.K. Kennedy, , J. Zhang, et al., Mutation in the silencing gene S/R4 can delay aging in S. Cerevisiae, Cell 80 (3) (1995) 485-496.

[17]

J. Sun, D. Folk, T.J. Bradley, et al., Induced overexpression of mitochondrial Mn-superoxide dismutase extends the life span of adult Drosophila melanogaster, Genetics 161 (2) (2002) 661-672.

[18]

C. Franceschi, M. Bonafè, S. Valensin, et al., Inflamm‐aging: an evolutionary perspective on immunosenescence, Ann. N. Y. Acad. Sci. 908 (1) (2000) 244-254.

[19]

S.E. Ozanne, C.N. Hales, Lifespan: catch-up growth and obesity in male mice, Nature 427 (6973) (2004) 411.

[20]

D. Bouglé, S. Bouhallab, Dietary bioactive peptides: human studies, Crit. Rev. Food Sci. Nutr. 57 (2) (2017) 335-343.

[21]

S. Ribarič, Diet and aging, Oxid. Med. Cell. Longev.2012 (2012).

[22]

N.P. Möller, K.E. Scholz-Ahrens, N. Roos, et al., Bioactive peptides and proteins from foods: indication for health effects, Eur. J. Nutr. 47 (4) (2008) 171-182.

[23]

D.D. Kitts, K. Weiler, Bioactive proteins and peptides from food sources. Applications of bioprocesses used in isolation and recovery, Curr. Pharm. Des. 9 (16) (2003) 1309-1323.

[24]

A. Sánchez, A. Vázquez, Bioactive peptides: a review, Food Qual. Saf. 1 (1) (2017) 29-46.

[25]

K. Fields, T.J. Falla, K. Rodan, et al., Bioactive peptides: signaling the future, J. Cosmet. Dermatol. 8 (1) (2009) 8-13.

[26]

F. Gorouhi, H. Maibach, Role of topical peptides in preventing or treating aged skin, Int. J. Cosmet. Sci. 31 (5) (2009) 327-345.

[27]

E. Haque, R. Chand, S. Kapila, Biofunctional properties of bioactive peptides of milk origin, Food Rev. Int. 25 (1) (2008) 28-43.

[28]

T. Finkel, The metabolic regulation of aging, Nat. Med. 21 (12) (2015) 1416.

[29]

V. Pak, M.S. Koo, T.D. Kasymova, et al., Isolation and identification of peptides from soy 11S-globulin with hypocholesterolemic activity, Chem. Nat. Compd. 41 (6) (2005) 710-714.

[30]

C. Lammi, C. Zanoni, A. Arnoldi, IAVPGEVA, IAVPTGVA, and LPYP, three peptides from soy glycinin, modulate cholesterol metabolism in HepG2 cells through the activation of the LDLR-SREBP2 pathway, J. Funct. Foods 14 (2015) 469-478.

[31]

C. Zanoni, G. Aiello, A. Arnoldi, et al., Investigations on the hypocholesterolaemic activity of LILPKHSDAD and LTFPGSAED, two peptides from lupin β-conglutin: focus on LDLR and PCSK9 pathways, J. Funct. Foods 32 (2017) 1-8.

[32]

C. Lammi, C. Zanoni, A. Arnoldi, Three peptides from soy glycinin modulate glucose metabolism in human hepatic HepG2 cells, Int. J. Mol. Sci. 16 (11) (2015) 27362-27370.

[33]

C. Zanoni, G. Aiello, A. Arnoldi, et al., Hempseed peptides exert hypocholesterolemic effects with a statin-like mechanism, J. Agric. Food Chem. 65 (40) (2017) 8829-8838.

[34]

R. Soares, S. Mendonça, L.Í. de Castro, et al., Major peptides from amaranth (Amaranthus cruentus) protein inhibit HMG-CoA reductase activity, Int. J. Mol. Sci. 16 (2) (2015) 4150-4160.

[35]

M.S. Coelho, R.A.M. Soares-Freitas, J.A.G. Arêas, et al., Peptides from Chia present antibacterial activity and inhibit cholesterol synthesis, Plant Foods Hum. Nutr. 73 (2) (2018) 101-107.

[36]

G. Aiello, S. Ferruzzab, G. Ranaldi, et al., Behavior of three hypocholesterolemic peptides from soy protein in an intestinal model based on differentiated Caco-2 cell, J. Funct. Foods 45 (2018) 363-370.

[37]

C. Lammi, C. Zanoni, A. Arnoldi, et al., Peptides derived from soy and lupin protein as dipeptidyl-peptidase Ⅳ inhibitors: in vitro biochemical screening and in silico molecular modeling study, J. Agric. Food Chem. 64 (51) (2016) 9601-9606.

[38]

C. Lammi, C. Zanoni, L. Calabresi, et al., Lupin protein exerts cholesterol-lowering effects targeting PCSK9: from clinical evidences to elucidation of the in vitro molecular mechanism using HepG2 cells, J. Funct. Foods 23 (2016) 230-240.

[39]

M. McDonagh, K. Peterson, B. Holzhammer, et al., A systematic review of PCSK9 inhibitors alirocumab and evolocumab, J. Manag. Care Spec. Pharm. 22 (6) (2016) 641-653q.

[40]

K. Majumder, J. Wu, A new approach for identification of novel antihypertensive peptides from egg proteins by QSAR and bioinformatics, Food Res. Int. 43 (5) (2010) 1371-1378.

[41]

K. Majumder, J. Wu, Purification and characterisation of angiotensin I converting enzyme (ACE) inhibitory peptides derived from enzymatic hydrolysate of ovotransferrin, Food Chem. 126 (4) (2011) 1614-1619.

[42]

M. Son, C.B. Chan, J. Wu, Egg white ovotransferrin‐derived ACE inhibitory peptide ameliorates angiotensin Ⅱ‐Stimulated insulin resistance in skeletal muscle cells, Mol. Nutr. Food Res. 62 (4) (2018) 1700602.

[43]

A. Zambrowicz, M. Pokora, B. Setner, et al., Multifunctional peptides derived from an egg yolk protein hydrolysate: isolation and characterization, Amino Acids 47 (2) (2015) 369-380.

[44]

M.R. Marques, R.A. Manólio, S. Freitasa, et al., Peptides from cowpea present antioxidant activity, inhibit cholesterol synthesis and its solubilisation into micelles, Food Chem. 168 (2015) 288-293.

[45]

Y. Kashima, S. Kanematsu, S. Asai, et al., Identification of a novel hypocholesterolemic protein, major royal jelly protein 1, derived from royal jelly, PLoS One 9 (8) (2014) e105073.

[46]
A. Arnoldi, L. Carmen, G. Aiello, Cholesterol-reducing foods: Proteins and peptides, 2019.
DOI
[47]

M. Shi, M.L. Mathaia, G. Xu, et al., The effects of supplementation with blueberry, cyanidin-3-O-β-glucoside, yoghurt and its peptides on obesity and related comorbidities in a diet-induced obese mouse model, J. Funct. Foods 56 (2019) 92-101.

[48]

W. Shi, T. Hou, D. Guo, et al., Evaluation of hypolipidemic peptide (Val-Phe-Val-Arg-Asn) virtual screened from chickpea peptides by pharmacophore model in high-fat diet-induced obese rat, J. Funct. Foods 54 (2019) 136-145.

[49]

P. Mudgil, H. Kamal, G.C. Yuen, et al., Characterization and identification of novel antidiabetic and anti-obesity peptides from camel milk protein hydrolysates, Food Chem. 259 (2018) 46-54.

[50]

T. Aoyama, K. Fukui, T. Nakamori, et al., Effect of soy and milk whey protein isolates and their hydrolysates on weight reduction in genetically obese mice, Biosci. Biotechnol. Biochem. 64 (12) (2000) 2594-2600.

[51]

D.P. Jones, Redox theory of aging, Redox Biol. 5 (2015) 71-79.

[52]

D. Harraan, Aging: a theory based on free radical and radiation chemistry. (1955).

[53]

E. Escudero, L. Mora, P.D. Fraser, et al., Identification of novel antioxidant peptides generated in Spanish dry-cured ham, Food Chem. 138 (2-3) (2013) 1282-1288.

[54]

M. Gallego, L. Mora, M. Reig, et al., Stability of the potent antioxidant peptide SNAAC identified from Spanish dry-cured ham, Food Res. Int. 105 (2018) 873-879.

[55]

M. Gallego, L. Mora, F. Toldrá, Characterisation of the antioxidant peptide AEEEYPDL and its quantification in Spanish dry-cured ham, Food Chem. 258 (2018) 8-15.

[56]

A.T. Girgih, R. He, S. Malomo, et al., Structural and functional characterization of hemp seed (Cannabis sativa L.) protein-derived antioxidant and antihypertensive peptides, J. Funct. Foods 6 (2014) 384-394.

[57]

I. Jemil, L. Mora, R. Nasri, et al., A peptidomic approach for the identification of antioxidant and ACE-inhibitory peptides in sardinelle protein hydrolysates fermented by Bacillus subtilis A26 and Bacillus amyloliquefaciens An6, Food Res. Int. 89 (2016) 347-358.

[58]

C.G. Rizzello, A. Lorusso, V. Russo, et al., Improving the antioxidant properties of quinoa flour through fermentation with selected autochthonous lactic acid bacteria, Int. J. Food Microbiol. 241 (2017) 252-261.

[59]

A.R. Corrochano, A. Ferraretto, E. Arranz, et al., Bovine whey peptides transit the intestinal barrier to reduce oxidative stress in muscle cells, Food Chem. 288 (2019) 306-314.

[60]

A. Moayedi, L. Mora, M.C. Aristoy, et al., ACE-inhibitory and antioxidant activities of peptide fragments obtained from tomato processing by-products fermented using Bacillus subtilis: effect of amino acid composition and peptides molecular mass distribution, Appl. Biochem. Biotechnol. 181 (1) (2017) 48-64.

[61]

P. García-Mora, M. Martín-Martínez, A.M. Bonache, et al., Identification, functional gastrointestinal stability and molecular docking studies of lentil peptides with dual antioxidant and angiotensin I converting enzyme inhibitory activities, Food Chem. 221 (2017) 464-472.

[62]

R. He, Y. Yang, Z. Wang, et al., Rapeseed protein-derived ACE inhibitory peptides LY, RALP and GHS show antioxidant and anti-inflammatory effects on spontaneously hypertensive rats, J. Funct. Foods 55 (2019) 211-219.

[63]

H.R. Warner, Superoxide dismutase, aging, and degenerative disease, Free Radic. Biol. Med. 17 (3) (1994) 249-258.

[64]

A. Paul, A. Belton, S. Nag, et al., Reduced mitochondrial SOD displays mortality characteristics reminiscent of natural aging, Mech. Ageing Dev. 128 (11-12) (2007) 706-716.

[65]

L. You, M. Zhao, R.H. Liu, et al., In vitro antioxidant activity and in vivo anti-fatigue effect of loach (Misgurnus anguillicaudatus) peptides prepared by papain digestion, Food Chem. 124 (1) (2011) 188-194.

[66]

K. Takemori, E. Yamamoto, H. Ito, et al., Prophylactic effects of elastin peptide derived from the bulbus arteriosus of fish on vascular dysfunction in spontaneously hypertensive rats, Life Sci. 120 (2015) 48-53.

[67]

S.M. Craige, S. Kröller-Schön, C. Li, et al., PGC-1α dictates endothelial function through regulation of eNOS expression, Sci. Rep. 6 (2016) 38210.

[68]

M.K. Lee, J.W. Choi, Y.H. Choi, et al., Protective effect of Pyropia yezoensis peptide on dexamethasone-induced myotube atrophy in C2C12 myotubes, Mar. Drugs 17 (5) (2019) 284.

[69]

J.J. Song, J.J. Song, Q. Wang, M. Du, et al., Peptide IPPKKNQDKTE ameliorates insulin resistance in HepG2 cells via blocking ROS-mediated MAPK signaling, J. Funct. Foods 31 (2017) 287-294.

[70]

J.J. Song, J. Gao, M. Du, et al., Casein glycomacropeptide hydrolysates ameliorate hepatic insulin resistance of C57BL/6J mice challenged with high-fat diet, J. Funct. Foods 45 (2018) 190-198.

[71]

H.Y. Lo, C.C. Li, T.Y. Ho, et al., Identification of the bioactive and consensus peptide motif from Momordica charantia insulin receptor-binding protein, Food Chem. 204 (2016) 298-305.

[72]

M.E.O. Toledo, E.J. de Mejia, M. Sivaguru, et al., Common bean (Phaseolus vulgaris L.) protein-derived peptides increased insulin secretion, inhibited lipid accumulation, increased glucose uptake and reduced the phosphatase and tensin homologue activation in vitro, J. Funct. Foods 27 (2016) 160-177.

[73]

L. Mojica, E.J. de Mejía, M.Á. Granados-Silvestre, et al., Evaluation of the hypoglycemic potential of a black bean hydrolyzed protein isolate and its pure peptides using in silico, in vitro and in vivo approaches, J. Funct. Foods 31 (2017) 274-286.

[74]

L. Mojica, D.A. Luna‐Vital, E. González de Mejía, Characterization of peptides from common bean protein isolates and their potential to inhibit markers of type‐2 diabetes, hypertension and oxidative stress, J. Sci. Food Agric. 97 (8) (2017) 2401-2410.

[75]

M.E. Oseguera-Toledo, E.G. de Mejia, S.L. Amaya-Llano, Hard-to-cook bean (Phaseolus vulgaris L.) proteins hydrolyzed by alcalase and bromelain produced bioactive peptide fractions that inhibit targets of type-2 diabetes and oxidative stress, Food Res. Int. 76 (2015) 839-851.

[76]

A. Jakubczyk, M. Karaś, U. Złotek, et al., Identification of potential inhibitory peptides of enzymes involved in the metabolic syndrome obtained by simulated gastrointestinal digestion of fermented bean (Phaseolus vulgaris L.) seeds, Food Res. Int. 100 (2017) 489-496.

[77]

Y. Ren, K. Liang, Y. Jinet, et al., Identification and characterization of two novel α-glucosidase inhibitory oligopeptides from hemp (Cannabis sativa L.) seed protein, J. Funct. Foods 26 (2016) 439-450.

[78]

B.K. Han, H.J. Lee, H.S. Lee, et al., Hypoglycaemic effects of functional tri‐peptides from silk in differentiated adipocytes and streptozotocin‐induced diabetic mice, J. Sci. Food Agric. 96 (1) (2016) 116-121.

[79]

Y.B. Henda, M. Laamaria, I. Lanneluc, et al., Di and tripeptides from marine sources can target adipogenic process and contribute to decrease adipocyte number and functions, J. Funct. Foods 17 (2015) 1-10.

[80]

T. de Souza Rocha, L.M.R. Hernandez, L. Mojica, et al., Germination of Phaseolus vulgaris and alcalase hydrolysis of its proteins produced bioactive peptides capable of improving markers related to type-2 diabetes in vitro, Food Res. Int. 76 (2015) 150-159.

[81]

J. Lu, Y. Zeng, W. Hou, et al., The soybean peptide aglycin regulates glucose homeostasis in type 2 diabetic mice via IR/IRS1 pathway, J. Nutr. Biochem. 23 (11) (2012) 1449-1457.

[82]

J.B. Mannick, G. Del Giudice, M. Lattanzi, et al., mTOR inhibition improves immune function in the elderly, Sci. Transl. Med.6 (268) (2014), p.268ra179-268ra179.

[83]

F. Gong, M.F. Chen, Y.Y. Zhang, et al., A novel peptide from abalone (Haliotis discus hannai) to suppress metastasis and vasculogenic mimicry of tumor cells and enhance anti-tumor effect in vitro, Mar. Drugs 17 (4) (2019) 244.

[84]

F. Gong, M.F. Chen, J. Chen, et al., Boiled abalone byproduct peptide exhibits anti-tumor activity in HT1080 cells and HUVECs by suppressing the metastasis and angiogenesis in vitro, J. Agric. Food Chem. 67 (32) (2019) 8855-8867.

[85]

X. Li, Y. Tang, F. Yu, et al., Inhibition of prostate cancer DU-145 cells proliferation by anthopleura anjunae oligopeptide (YVPGP) via PI3K/AKT/mTOR signaling pathway, Mar. Drugs 16 (9) (2018) 325.

[86]

S.J. Park, J. Ryu, I.H. Kim, et al., Activation of the mTOR signaling pathway in breast cancer MCF‑7 cells by a peptide derived from Porphyra yezoensis, Oncol. Rep. 33 (1) (2015) 19-24.

[87]

A. Salminen, K. Kaarniranta, AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network, Ageing Res. Rev. 11 (2) (2012) 230-241.

[88]

S. Zhao, S. Zhang, W. Zhang, et al., First demonstration of protective effects of purified mushroom polysaccharide-peptides against fatty liver injury and the mechanisms involved, Sci. Rep. 9 (1) (2019) 1-13.

[89]

R. Zhang, J. Chen, X. Mao, et al., Separation and lipid inhibition effects of a novel decapeptide from Chlorella pyenoidose, Molecules 24 (19) (2019) 3527.

[90]

N. Tachibana, Y. Yamashita, M. Nagata, et al., Soy β-conglycinin improves glucose uptake in skeletal muscle and ameliorates hepatic insulin resistance in Goto-Kakizaki rats, Nutr. Res. 34 (2) (2014) 160-167.

[91]

C. Roblet, A. Doyen, J. Amiot, et al., Enhancement of glucose uptake in muscular cell by soybean charged peptides isolated by electrodialysis with ultrafiltration membranes (EDUF): activation of the AMPK pathway, Food Chem. 147 (2014) 124-130.

[92]

R.P. Talens, K. Christensen, H. Putter, et al., Epigenetic variation during the adult lifespan: cross‐sectional and longitudinal data on monozygotic twin pairs, Aging Cell 11 (4) (2012) 694-703.

[93]

M.F. Fraga, M. Esteller, Epigenetics and aging: the targets and the marks, Trends Genet. 23 (8) (2007) 413-418.

[94]

S. Han, A. Brunet, Histone methylation makes its mark on longevity, Trends Cell Biol. 22 (1) (2012) 42-49.

[95]

B. Richardson, Impact of aging on DNA methylation, Ageing Res. Rev. 2 (3) (2003) 245-261.

[96]

S. Maegawa, G. Hinkal, H.S. Kim, et al., Widespread and tissue specific age-related DNA methylation changes in mice, Genome Res. 20 (3) (2010) 332-340.

[97]

S. Pal, J.K. Tyler, Epigenetics and aging, Sci. Adv. 2 (7) (2016) e1600584.

[98]

A.F. Galvez, Identification of lunasin as the active component in soy protein responsible for reducing LDL cholesterol and risk of cardiovascular disease, Am Heart Assoc. 2012.

[99]

Q. Jin, L.R. Yu, L. Wang, et al., Distinct roles of GCN5/PCAF‐mediated H3K9ac and CBP/p300‐mediated H3K18/27ac in nuclear receptor transactivation, EMBO J. 30 (2) (2011) 249-262.

[100]

C. Lammi, C. Zanoni, A. Arnoldi, et al., Two peptides from soy β-conglycinin induce a hypocholesterolemic effect in HepG2 cells by a statin-like mechanism: comparative in vitro and in silico modeling studies, J. Agric. Food Chem. 63 (36) (2015) 7945-7951.

[101]

M. Kaeberlein, M. McVey, L. Guarente, The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms, Genes Dev. 13 (19) (1999) 2570-2580.

[102]

S. Imai, C.M. Armstrong, M. Kaeberlein, et al., Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase, Nature 403 (6771) (2000) 795.

[103]

S.J. Lin, P.A. Defossez, L. Guarente, Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae, Science 289 (5487) (2000) 2126-2128.

[104]

B.J. North, E. Verdin, Sirtuins: Sir2-related NAD-dependent protein deacetylases, Genome Biol. 5 (5) (2004) 224.

[105]

C. Carrico, J.G. Meyer, W. He, et al., The mitochondrial acylome emerges: proteomics, regulation by sirtuins, and metabolic and disease implications, Cell Metab. 27 (3) (2018) 497-512.

[106]

Y. Kanfi, S. Naiman, G. Amir, et al., The sirtuin SIRT6 regulates lifespan in male mice, Nature 483 (7388) (2012) 218.

[107]

S.I. Imai, L. Guarente, NAD+ and sirtuins in aging and disease, Trends Cell Biol. 24 (8) (2014) 464-471.

[108]

X.Y. Yuan, M.Y. Liu, H.H. Cheng, et al., Replacing fish meal with cottonseed meal protein hydrolysate affects amino acid metabolism via AMPK/SIRT1 and TOR signaling pathway of Megalobrama amblycephala, Aquaculture 510 (2019) 225-233.

[109]

G. Aubert, P.M. Lansdorp, Telomeres and aging, Physiol. Rev. 88 (2) (2008) 557-579.

[110]

V.K. Khavinson, B.I. Kuznik, S.I. Tarnovskaya, et al., Short peptides and telomere length regulator hormone irisin, Bull. Exp. Biol. Med. 160 (3) (2016) 347-349.

[111]

D. Nesic35, I. Pantic, S. Mazic, The theories of aging: yesterday, today, tomorrow, Aging and Human Rights (2018) 82.

[112]

W. Shen, T. Matsui, Current knowledge of intestinal absorption of bioactive peptides, Food Funct. 8 (12) (2017) 4306-4314.

[113]

S. Li, L. Liu, G. He, et al., Molecular targets and mechanisms of bioactive peptides against metabolic syndromes, Food Funct. 9 (1) (2018) 42-52.

[114]

H. Pan, T. Finkel, Key proteins and pathways that regulate lifespan, J. Biol. Chem. 292 (16) (2017) 6452-6460.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 03 November 2019
Accepted: 06 January 2020
Published: 07 January 2020
Issue date: March 2020

Copyright

© 2020 "Society information".

Acknowledgements

Acknowledgements

This work is funded by Natural Sciences and Engineering Research Council of Canada (NSERC).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return