Anti-aging Interventions: Caloric Restriction and Beyond

Authors

  • Khing S. Ong Director Allergy and Immunology Division, Department of Pediatrics, University of California Irvine. 1978-1985, USA
  • Zack ST. Lim Retired RPH (Registered Pharmacist), USA
  • Boenjamin Setiawan Founder, Stem Cell Institute, Jakarta, Indonesia

DOI:

https://doi.org/10.55175/cdk.v50i9.726

Keywords:

Antiaging, caloric restriction

Abstract

Aging is the biggest risk factor for most of the age-related chronic diseases, which carried a heavy health-care costs burden worldwide. These age-related diseases, such as cardiovascular diseases, neurodegenerative diseases, cancers, type 2 diabetes, and osteoarthritis, shared common mechanisms that associate with aging, such as chronic sterile low grade inflammation, cellular senescence, DNA damages, and stem cells dysfunctions. In this review, we will first discussed the dietary manipulation, such as caloric restriction as the mean of delaying aging, and we will also review nutraceuticals or natural compounds, and pharmaceutical drugs as the mean for intervention of aging.

Proses menua dikaitkan dengan berbagai penyakit kronis terkait-usia, yang mengakibatkan beban biaya perawatan kesehatan yang besar di seluruh dunia. Penyakit-penyakit tersebut meliputi penyakit kardiovaskuler, penyakit neurodegeneratif, kanker, diabetes tipe 2, dan osteoartritis, yang mempunyai gambaran umum serupa yaitu inflamasi steril kronis kadar rendah, penuaan sel, kerusakan DNA, dan disfungsi sel punca. Tinjauan ini berkenaan dengan manipulasi diet berupa restriksi kalori, dan nutraceuticals serta senyawa alami, juga sediaan farmakologis untuk tujuan mengintervensi proses penuaan

Downloads

Download data is not yet available.

References

Barardo D, Thornton D, Thoppil H, Walsh M, Sharifi S, Ferreira S, et al. The DrugAge database of aging-related drugs. Aging Cell 2017;16:594-7.

Ong Khing S., Lim Zack ST, Setiawan B. Senolytics: A potiential fountain of youth. CDK in press.

Dong X, Milholland B, Vijg J. Evidence for a limit to human lifespan. Nature 2016;538: 257-9

Gan W, Liu XL, Yu T, Zou YG, Li TT, Wang S, et al. Urinary 8-oxo-7,8-dihydroguanosine as a potential biomarker of aging. Front Aging Neurosci. 2018;10:34. doi: 10.3389/fnagi.2018.00034.

Weindruch R, Walford. The retardation of aging and disease by dietary restriction. Springfield, IL.: Charles C. Thomas Publ.; 1988.

Mattison JA, Roth GS, Beasley TM, Tilmont EM, Handy AH, Herbert RL, et al. Impact of caloric restriction on health and survival in rhesus monkeys: the NIH study. Nature 2012;489(7415):10.1038/nature11432.

Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM, et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 2009;325(5937):201-4

Anderson RM. The caloric restriction paradigm. Biochemistr Society [Internet]. 2015. Available from: http://www.biochemist.org/bio/03704/0020/037040020.pdf

Maegawa S, Lu Y, Tahara T, Lee JT, Madzo J, Liang S, Jelinek J, et al. Caloric restriction delays age-related methylation drift. Nature Comm [Internet]. 2017. Available from: https://www.nature.com/articles/s41467-017-00607-3

Pifferi F, Terrien J, Marchal J, Dal-Pan A, Djelti F, Hardy I, et al. Caloric restriction increases lifespan but affects brain integrity in grey mouse lemur primate. Nature Comm Biol. 2018;1:30

Yin Z, Raj DD, Schaafsma W, van der Heijden RA, Kooistra SM, Reijne AC, et al. Low-fat diet with caloric restriction reduces white matter microglia activation during aging. Front Mol Neurosci. 2018;11:65.

Cheng CW, Adams GB, Perin L, Wei M, Zhou X, Lam BS, et al. Prolonged fasting reduces IGF-1/PKA to promote hematopoietic stem cell-based regeneration and reverse immunosuppression. Cell Stem Cell 2014;14(6): 810-23

Wei M, Brandhorst S, Shelehchi M, Mirzaei H, Cheng CW, Budniak J, et al. Fasting mimicking diet and markers/risk factors for aging, diabetes, cancers and cardiovascular disease. Sci Translati Med 2017;9(377). pii: eaai8700.

Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: Health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28 Suppl:500-16.

Luciano M, Corley J, Cox SR, Hernandez MCV, Craig LCA, Dickie DA, et al. Mediterranean-type diet and brain structural changes from 73 to 76 years in a Scotish cohort. Neurology 2017;88(5):449-55.

Ikuta T, Saito S, Tani H, Tatefuji T, Hashimoto K. Resveratrol derivative rich melinjo (Gnetum gnemon L.) seed extract improves obesity and survival of C57BL/6 mice fed a high-fat diet. Biosci Biotechnol Biochemistr 2015;79(12):2044-9

Hubbard BP, Gomes AP, Dai H, Li J, Case AW, Considine T, et al. Evidence for a common mechanism of SIRT1 regulation by allosteric activator. Science 2013;339(6124):1216-9

Mitchell SJ, Martin-Montalvo A, Mercken EM, Palacios HH, Ward TM, Abulwerdi G, et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Reports 2014;6:836-43.

Park SJ, Ahmad F, Philp A, Baar K, Williams T, Luo H, et al. Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell 2012;148(3):421-33

Kodali M, Parihar VK, Hattiangady B, Mishra V, Shuai B, Shetty AK. Resveratrol prevents age-related memory and mood dysfunction with increased hippocampal neurogenesis and microvasculature, and reduced glial activation. Scientific Rep. 2015;5: 8075

Feldman JL, Dittenhafer-Reed KE, Denu JM. Sirtuin catalysis and regulation. J Biol Chemistr. 2012;287:42419-127

Polito L, Kehoe PG, Forloni G, Albani D. The molecular genetics of sirtuins: association with human longevity and age-related diseases. Int J Epidemiol Genet. 2010; 1(3):214-25.

Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Biol. 2012;13(4): 225-38

Haigis MC, Guarente L. Mammalian sirtuins—e merging roles in physiology, aging, and caloric restriction. Genes & Development 2006;20:2913-21

Gomes AP, Price NL, Ling AJ, Moslehi JJ, Montgomery MK, Rajman L, et al. Declining NAD+ induces a pseudohypoxic state disrupting nuclear-mitochondria communication during aging. Cell 2013;155(7):1624-38.

Hwang ES, Hwang SY. Cellular NAD+ level: A key determinantof mitochondrial quality and health. Ann Geriartr Med Res. 2017;21(4):149-57

Martens CR, Denman BA, Mazzo MR, Armstrong ML, Reisdorph N, McQueen MB, et al. Chronic nicotinamide riboside supplementation is well-tokerated and elevates NAD+ in healthy middle-aged and older adults. Nature Comm. 2018;9:1286

Goldberg J, Currais A, Prior M, Fischer W, Chiruta C, Ratliff E, et al. The mitochondrial ATP synthase is shared drug target for aging and dementia. Aging Cell. 2018 Apr;17(2). doi: 10.1111/acel.12715.

Xu D, Chen H, Mak S, Hu S, Tsim KWK, Hu Y, et al. Neuroprotection against glutamate induced excitotoxicity and induction of neurite outgowth by T006, a novel multifunctional derivatove of tetramethypyrazine in neuronal cell models. Neurochem Int. 2016;99:194-205.

Everts S. Can we hit the snooze button on aging? C& EN; 2017 .p 31-5

Bian G, Gloor GB, Gong A, Jia C, Zhang W, Hu J, et al. The gut microbiota of healthy aged Chinese is similar to that of the healthy young. Amer Soc Microbiol. 2017;2(5):1-12.

Steppan J, Tran H, Benjo AM, Pellakuru L, Barodka V, Ryoo S, et al. Alagebrium in combination with exercise ameliorate age associated ventricular and vascular stiffness. Exp Gerontol. 2012;47(8) 565-72.

Harley CB, Liu W, Blasco M, Vera E, Andrews WH, Briggs LA, et al. A natural product Telomerase activator as part of a health maintenance program. Rejuvenation Res. 2011;14(1):45-56.

Molgora B, Bateman R, Sweeney G, Finger D, Dimler T, Effros RB, et al. Functional assessment of pharmacological telomerase activation in human T cells. Cells 2013 ;2:57-66

Xu Y, Sun Z .Molecular basis of klotho: From gene to function in aging. Endocr Rev. 2015; 36)(2):174-93.

Bannister CA, Holden SE,Jenkins-Jones S, Morgan CL, Halcox JP, Schernthaner G, et al. Can people with type 2 diabetes live longer than those without? A comparison of mortality in people initiated with metformin or sulphonylurea monotherapy and matched non diabetic controls. Diabetes Obes Metab. 2014;16(11):1165-73.

Onken B, Driscoll M. Metformin induces a dietary restriction-like state and oxidative stress response to extend C. elegans healthspan via AMPK, LKB1, and SKN-1. Plos One 2010;5(1):8758.

Pryor R, Cabreiro F. Repurposing metformin: An old drug with new tricks in its binding pockets. Biochem J. 2015;471:307-32.

Leslie M. A putative antiaging drug takes a step from mice to men. Science 2013;342:789.

Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, et al. Rapamycin fed late in life extend lifespan in genetically heterogeneous mice. Nature 2009;460(7253):392-5.

Anisimov VN, Zabezhinski MA, Popovich IG, Piskunova TS, Semenchenko AV, Tyndyk ML, et al. Rapamycin increases lifespan and inhibit spontaneous tumorigenesis in inbred female mice. Cell Cycle 2011;10:4230-6. 42. Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, et al. mTOR inhibition improves immune function in the elderly. Sci Transl Med. 2014;6(268):268ra179.

Eisenberg T, Knauer H, Schauer A, Büttner S, Ruckenstuhl C, Carmona-Gutierrez D, et al. Induction of autophagy by spermidine promote longevity. Nat Cell Biol. 2009;11:1305-14.

Poggioli T, Vujic A, Yang P, Macias-Trevino C, Uygur A, Loffredo FS, et al. Circulating growth differential factor 11/8 levels declined with age. Circ Res. 2016;118(1):29-37.

Egerman MA, Cadena SM, Gilbert JA, Meyer A, Nelson HN, Swalley SE, et al. GDF11 increase with age and inhibit skeletal muscle regeneration. Cell Metab. 2015;22(1):164-74.

Olson KA, Beatty AL,Heidecker B, Regan MC, Brody EN, Foreman T, et al. Association of growth differentiation factor 11/8 putative antiaging factor, with cardiovascular outcomes and overall mortality in human: Analysis of the heart and soul and HUNT3 cohort. Eur Heart J. 2015;36(48):3426-34

Cobb LJ, Lee C, Xiao J, Yen K, Wong RG, Nakamura HK, et al. Naturally occurring mitochondria-derived peptides are age-dependent regulators of apoptosis, insulin sensitivity, and inflammatory markers. Aging 2016;8(4):796-808

Singh A, Andreux P, Blanco-Bose W, Ryu D, Aebischer P, Auwerx J, et al. Orally administered urolithin A is safe and modulates muscle and mitochondrial biomarkers in elderly. Innovation in Aging 2017;1:1223-4

Downloads

Published

03-09-2018

How to Cite

Ong, K. S., Lim, Z. S., & Setiawan, B. (2018). Anti-aging Interventions: Caloric Restriction and Beyond. Cermin Dunia Kedokteran, 45(9), 686–690. https://doi.org/10.55175/cdk.v50i9.726

Issue

Section

Articles