Time and Spice
On
As we pass through time, or as it passes through us, a series of changes accumulate which we call ageing.
Superficial changes such as the progressive symptoms of chronic degenerative diseases can in many cases be prevented, and partially or completely reversed using inexpensive pharmaco-nutritional tools.
Deeper delta such as the reduced expression of mRNA splicing factors, the loss of telomeric DNA, declining immune-surveillance, the lack of progenitor cells, the loss of proteostasis and the accumulation of senescent cells, seemed inaccessible.
Until quite recently.
For anyone who left college more than 30 years ago, RNA splicing might just be the least familiar of these deep signs. It is incredibly exotic but it is also, now, biology 101.
For non-scientists, it is a stage in the DNA-RNA-protein sequence where RNA transcribed from DNA is edited before translation into protein can begin. It involves the cutting out of introns, genetic hot-spots which do not code for proteins but are more like switch points or punctuation marks; and the re-assembling of exons, those sections which do code for proteins.
This editing process takes place in particles called spliceosomes, which are mostly located in the cell nucleus. It is essential for correct protein synthesis, and is aided and abetted by mRNA splicing factors which are tissue-specific, and influenced by the outside world via epigenetic factors (1). (Pending post ‘The Meth Lab’ goes into the epigenetic factors in more detail.)
Minor modifications in pre-mRNA splicing, facilitated by the different splicing factors in different tissues, allow the cell to build an extensive portfolio of proteins from a more limited number of genes. The best-known example of this is probably the single gene that codes for a structural protein (crystallin) in the lens and an enzymatic protein (LDH) in muscle (2).
Quality control of proteins in the cell (aka proteostasis) is critical for the functioning and survival of the cell. It involves the fine control of protein assembly, folding, monitoring and, when the protein has reached the end of its working life, dispatching it to a proteasome for breakdown and recycling.
Faulty RNA splicing contributes to the synthesis of incorrectly built proteins, and the quality and quantity of splicing factors decline with age. This causes a progressive increase in faulty protein synthesis, the loss of proteostatic control, growing endoplasmic reticular stress and the beginning of the end of the cell’s productive life (3, 4). (See previous post, Know When to Fold ‘em).
The vast majority of cells then activate one of several possible suicide sequences, sacrificing themselves for the collective good.
A few drift off into senescence, lingering on aimlessly but still consuming valuable resources, creating chronic inflammatory stress and contributing to inflammatory and degenerative pathology. A smaller number become cancerous, striking a Faustian bargain whereby the cell gains temporary immortality but the collective dies.
Most of these mini-Fausts fail. They are killed off by metabolic bottlenecks, immunosurveillance and/or the cancer-killing properties of an array of phytonutrients, aided and abetted by the omega 3 HUFA’s. Only a very small percentage of cancer cells survive and multiply to the point where they become clinically significant.
The fact that around 30% of us now die from cancer, as opposed to 3-5% in pre-transitional societies (5, 6) and 1-3% in non-domestic animals (7), testifies to our disastrous diet and lifestyle. (Pending post ‘Peto’s Pet Paradox’ focuses on diet and cancer in other species).
Back to the RNA splicing factors. Recent work stands received wisdom (and time) on its head, by showing that the age-related decline in splicing factors can be reversed by specific phytonutrients.
Polyphenols such as resveratrol and genistein can bind directly to DNA and RNA (8). At high doses they cause DNA damage and can induce apoptosis (ie 9), but at low doses they enhance DNA repair via hormetic mechanisms (10).
They also positively influence the splicing process (11-13). They appear to do so in part by restoring levels of specific RNA splicing factors (11-13), and have now been shown to rejuvenate senescent cells, with restoration of telomere length and the ability to replicate (13, 14). They also selectively kill those senescent cells that do not regenerate (14, 15).
These are profound anti-ageing and life-extending effects; but what causes senescence in the first place?
Cells have an array of responses to different degrees and types of stress ranging from positive adaptations at one end (generally induced by lower levels of stress), to death when stress is extreme. Just below the lethal threshold, very high levels of stress – such as those triggered by severe endoplasmic reticular stress, telomere loss, DNA damage and/or the beginning of cancerous change, can cause senescence (16).
Small numbers of senescent cells can be tolerated, but as their numbers increase the tissues and organs in which they are located become compromised. The cells express an array of pro-inflammatory compounds called the Senescence-Associated Secretory Profile or SASP. The SASP causes progressive tissue damage and is a significant driver of the ageing process (17, 18). Via SASP, senescent cells induce senescence in neighboring cells, and the spreading rot leads towards tissue and organ failure (19, 20).
Local injections of senescent cells into various tissues accelerate the age-related diseases (21, 22). Conversely, the elimination of senescent cells delays the onset of age-related pathology in neurodegenerative, cardiovascular and other pathologies, including cancers (23-24).
Cancer becomes more likely as we age, at least in societies consuming a post-transitional diet.
The accumulation of senescent cells in aged tissues or xenograft models correlates with the incidence of cancer (25-27); while senolytic drugs (compounds which selectively kill senescent cells) delay tumor formation and reduce metastasis (24, 28, 29).
Senolytic drugs are, therefore, starting to find many clinical applications. They target a number of well-defined key proteins involved in apoptosis such as Bcl-2, Bcl-XL, p21, PI3K, AKT, FOXO4 and p53. But do we really need these drugs? The polyphenols, which are a good deal safer (they are in our foods after all), act at all of these same points (30-36).
It has been suggested that they may not be safe for general consumption, because in some pre-cancerous cells a shift into senescence is protective against cancer; and some anti-cancer treatments work by inducing senescence in cancer cells. However, these senescent cells must be cleared to avoid a chronic pro-tumorigenic state (37).
Furthermore, the same polyphenols which rejuvenate or kill senescent cells are also good at killing cancer cells, and/or inducing cell cycle arrest and/or redifferentiation (ie 38-41). In this way they provide an umbrella seno- and chemo-protective effect; one of the reasons why increased intakes of polyphenols within the dietary range are associated with better health (42, 43).
These same polyphenols also damp chronic inflammation (44), and are involved in the fine control of many enzymatically controlled reactions (44, 45). They protect, in multiple ways, against metabolic, cardiovascular, neurodegenerative and oncological diseases (37, 46, 47). In short, these ‘anti-nutrients’ as they were once called, are as important to our short and long-term health as any established vitamin or trace element.
We are thoroughly adapted to the near-constant presence of polyphenols in our bodies as modifiers / modulators. Their polyfunctional profiles trace a map of our evolutionary and dietary origins, a map on which the borders between the animal and plant kingdoms are long and complex, with very many checkpoints.
Neglecting those borders incurs a heavy penalty.
Over the last few decades, as our understanding of the significance of the polyphenols and other phytonutrients has developed, Big Phood’s irresponsibility in removing them from our diet has imperceptibly morphed into criminal irresponsibility. Ultra-processed food is the new tobacco. Post-transitional public health illustrates the madness of the modern diet. The political classes take their money and drag their feet.
The harm done by removing polyphenols from our diet is compounded by the addition of huge amounts of empty, pro-inflammatory calories in plant oils and sugars, to provide cheap and infantilized flavour profiles (48, 49). These in turn drive the epidemics of obesity, non-alcoholic liver disease and all the other ‘diseases of civilization’.
In a final turn of the screw obesity, metabolic syndrome and diabetes increase the senescent cell load (50-53). This provides yet another mechanism linking the ultra-processed diet with rising BMI, accelerated ageing, declining health and life expectancy, and increased anxiety and unhappiness (54).
The utter idiocy of Covid lockdowns manifests inter alia in the near 100% rise in obesity and types 1 and 2 diabetes in children (55). There will be blood, because outside the Dumb and Dumberer world of politics, everything connects. Time may not be speeding up but one of the ways in which we experience it – the ageing process – certainly is.
What can a concerned person do?
Selectively killing senescent cells in obese mice restores metabolic parameters (56) – so here is one anti-ageing role for the polyphenols. Conversely, caloric restriction reduces the burden of senescent cells (57), and the polyphenols, by acting as caloric-restriction mimetics (58), are likely acting here also. As mentioned above, the polyphenols are also good at killing senescent cell (ie 59).
Increasing our intake of polyphenols provides a set of keys, therefore, to modifying deep delta.
With the exception of the few amphiphilic polyphenols (such as olive and marine algal polyphenols), these compounds are not stored in the body for long. They are regarded as foreign molecules and rapidly metabolized and excreted, so we should plan on consuming polyphenols on a daily basis.
Polyphenols are not, however, the whole story.
Senescent cells are antigenic, and are aggressively picked off by the immune system. As we get older the immune system tends to become less effective, which is another reason why senescent cells accumulate as the decades pass (60).
The most effective method known of enhancing immune-surveillance is exercise (61-64). Much or all of this effect is mediated via AMP-kinase (65, 66), and is therefore theoretically achieved with phytonutrients derived from ie Gynostemma pentaphyllum (67), marketed as ActivAmp. In support of this idea, there is emerging evidence that physical exercise reduces numbers of senescent cells (68).
Exercise and/or ActivAMP, plus frequent doses of a range of polyphenols, will help to reduce the burden of senescent cells and will also improve immune function. This should provide significant anti-ageing benefits. Adding an omega 3 / amphiphile polyphenol combination to reduce the inflammatory effects of SASP (69-71), and inflammageing in general, should push the clock even further back.
In practical terms cut out the ultra-processed foods, and add spice.
Spices are the most concentrated dietary sources of polyphenols (72), and the curcuminoids in turmeric are among the best documented up-regulators of RNA splicing factors (73-75). They are also senolytics (76, 77), and will do both of these things if you manage to achieve meaningful plasma levels of these actives. Currently, HydroCurc is one of the best ways of doing this (78).
Just one last slice of deep delta.
There is evidence that along with the accumulation of senescent cells, ageing of tissues and organs is also driven by a growing scarcity of new progenitor (or precursor) cells (ie 79, 80), which are needed to replace those which have died or become senescent.
We carry a reserve of precursor (stem) cells which act as a repair kit. These stem cells are stored in the marrow of our long bones but as we age they become more reluctant to leave their bunker, becoming less available just when we need them most. Fucoidans, polysulphated oligosaccharides from brown seaweeds, release them into the circulation and put them back to work (81).
It seems logical to combine fucoidans with the right polyphenols, and many of us are already in the throes of self-experimentation. Based on blue zone data, the optimal long-term dietary intake of polyphenols is approximately ten times higher than the average amounts consumed today (ie 40). If you intend to self-administer higher doses of polyphenols, this should be restricted in my view to a semi-acute window of no more than 2 weeks.
Finally, senolysis is not the only road ahead. Senescent cells can be re-programmed and partially rejuvenated using a variant of the Yamanaka approach (82-84). These exciting research findings are consistent with the reported benefits of heterochronic transfusion (ie 85), which may work in a broadly similar way.
Ask about deferred payment schemes at your local blood bank.
Next week: Old salt, and how the food industry fights dirty.
References:
- Zhang J, Zhang YZ, Jiang J, Duan CG. The Crosstalk Between Epigenetic Mechanisms and Alternative RNA Processing Regulation. Front Genet. 2020 Aug 20;11:998.
- Wistow G, Mulders J, de Jong W. The enzyme lactate dehydrogenase as a structural protein in avian and crocodilian lenses. Nature 326, 622–624 (1987).
- Holly AC, Melzer D, Pilling LC, Fellows AC, Tanaka T, Ferrucci L, Harries LW. Changes in splicing factor expression are associated with advancing age in man. Mech Ageing Dev. 2013 Sep;134(9):356-66.
- Deschênes, M., & Chabot, B. (2017). The emerging role of alternative splicing in senescence and aging. Aging cell, 16(5), 918–933.
- Rowbotham J, Clayton P. An unsuitable and degraded diet? Part three: Victorian consumption patterns and their health benefits. J R Soc Med. 2008 Sep;101(9):454-62.
- Gurven M, Kaplan H. Longevity among hunter-gatherers: a cross-cultural examination. Pop Dem Rev 2007; 33: 321– 365.
- Vincze O, Colchero F, Lemaître JF, Conde DA, Pavard S, Bieuville M, Urrutia AO, Ujvari B, Boddy AM, Maley CC, Thomas F, Giraudeau M. Cancer risk across mammals. Nature. 2022 Jan;601(7892):263-267.
- Usha S, Johnson IM, Malathi R. Interaction of resveratrol and genistein with nucleic acids. J Biochem Mol Biol. 2005;38:198–205.
- Tyagi A, Gu M, Takahata T, Frederick B, Agarwal C, Siriwardana S, Agarwal R, Sclafani RA. Resveratrol selectively induces DNA Damage, independent of Smad4 expression, in its efficacy against human head and neck squamous cell carcinoma. Clin Cancer Res. 2011 Aug 15;17(16):5402-11
- Uchiumi F, Shoji K, Sasaki Y, Sasaki M, Sasaki Y, Oyama T, Sugisawa K, Tanuma S. Characterization of the 5′-flanking region of the human TP53 gene and its response to the natural compound, Resveratrol. J Biochem. 2016 Apr;159(4):437-47.
- Sakla MS, Lorson CL. Induction of full-length survival motor neuron by polyphenol botanical compounds. Hum Genet. 2008;122:635–643.
- Markus MA, Marques FZ, Morris BJ. Resveratrol, by modulating RNA processing factor levels, can influence the alternative splicing of pre-mRNAs. PLoS One. 2011;6(12):e28926.
- Latorre E, Birar VC, Sheerin AN, Jeynes JCC, Hooper A, Dawe HR, Melzer D, Cox LS, Faragher RGA, Ostler EL, Harries LW. Small molecule modulation of splicing factor expression is associated with rescue from cellular senescence. BMC Cell Biol. 2017 Oct 17;18(1):31.
- Kim J.K., Park S.U. Quercetin and its role in biological functions: an updated review. EXCLI J. 2018;17:856–863.
- Yousefzadeh M.J., Zhu Y., McGowan S.J., Angelini L., Fuhrmann-Stroissnigg H., Xu M., Ling Y.Y., Melos K.I., Pirtskhalava T., Inman C.L. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018;36:18–28.
- He S, Sharpless NE. Senescence in Health and Disease. Cell. 2017; 169:1000–11.
- López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013; 153:1194–217.
- Stojanović SD, Fiedler J, Bauersachs J, Thum T, Sedding DG. Senescence-induced inflammation: an important player and key therapeutic target in atherosclerosis. Eur Heart J. 2020 Aug 14;41(31):2983-2996.
- Da Silva PF, Ogrodnik M, Kucheryavenko O, Glibert J, Miwa S, Cameron K, Ishaq A, Saretzki G, Nagaraja-Grellscheid S, Nelson G, von Zglinicki T. The bystander effect contributes to the accumulation of senescent cells in vivo. Aging Cell 2019; 18: e12848.
- Nelson G, Wordsworth J, Wang C, Jurk D, Lawless C, Martin-Ruiz C, von Zglinicki T. A senescent cell bystander effect: senescence-induced senescence. Aging Cell 2012; 11: 345–349.
- Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, Orr ME. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell. 2018; 17:e12840.
- Xu M, Bradley EW, Weivoda MM, Hwang SM, Pirtskhalava T, Decklever T, Curran GL, Ogrodnik M, Jurk D, Johnson KO, Lowe V, Tchkonia T, Westendorf JJ, Kirkland JL. Transplanted Senescent Cells Induce an Osteoarthritis-Like Condition in Mice. J Gerontol A Biol Sci Med Sci. 2017; 72:780–85.
- Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL, van Deursen JM. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011; 479:232–36.
- Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature. 2018; 562:578–82.
- de Magalhães JP. How ageing processes influence cancer. Nat Rev Cancer. 2013; 13:357–65.
- Liu D, Hornsby PJ. Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res. 2007; 67:3117–26.
- von Kobbe C. Targeting senescent cells: approaches, opportunities, challenges. Aging (Albany NY). 2019 Nov 30;11(24):12844-12861.
- Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, Inman CL, Ogrodnik MB, Hachfeld CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, Langhi LGP, Weigl M, Giorgadze N, LeBrasseur NK, Miller JD, Jurk D, Singh RJ, Allison DB, Ejima K, Hubbard GB, Ikeno Y, Cubro H, Garovic VD, Hou X, Weroha SJ, Robbins PD, Niedernhofer LJ, Khosla S, Tchkonia T, Kirkland JL. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018; 24:1246–56.
- Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, Saltness RA, Jeganathan KB, Verzosa GC, Pezeshki A, Khazaie K, Miller JD, van Deursen JM. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature. 2016; 530:184–89.
- Verma S, Singh A, Kumari A, Tyagi C, Goyal S, Jamal S, Grover A. Natural polyphenolic inhibitors against the antiapoptotic BCL-2. J Recept Signal Transduct Res. 2017 Aug;37(4):391-400.
- Lin W, Tongyi S. Role of Bax/Bcl-2 family members in green tea polyphenol induced necroptosis of p53-deficient Hep3B cells. Tumour Biol. 2014 Aug;35(8):8065-75.
- Kazi A, Smith DM, Zhong Q, Dou QP. Inhibition of bcl-x(l) phosphorylation by tea polyphenols or epigallocatechin-3-gallate is associated with prostate cancer cell apoptosis. Mol Pharmacol. 2002 Oct;62(4):765-71.
- Khan H, Reale M, Ullah H, Sureda A, Tejada S, Wang Y, Zhang ZJ, Xiao J. Anti-cancer effects of polyphenols via targeting p53 signaling pathway: updates and future directions. Biotechnol Adv. 2020 Jan-Feb;38:107385.
- D’Angelo S, Martino E, Ilisso CP, Bagarolo ML, Porcelli M, Cacciapuoti G. Pro-oxidant and pro-apoptotic activity of polyphenol extract from Annurca apple and its underlying mechanisms in human breast cancer cells. Int J Oncol. 2017 Sep;51(3):939-948.
- Jiramongkol, Y., Lam, E.WF. FOXO transcription factor family in cancer and metastasis. Cancer Metastasis Rev 39, 681–709 (2020).
- Mirza-Aghazadeh-Attari M, Ekrami EM, Aghdas SAM, Mihanfar A, Hallaj S, Yousefi B, Safa A, Majidinia M. Targeting PI3K/Akt/mTOR signaling pathway by polyphenols: Implication for cancer therapy. Life Sci. 2020 Aug 15;255:117481.
- Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99-118.
- Zhou Y, Zheng J, Li Y, Xu DP, Li S, Chen YM, Li HB. Natural Polyphenols for Prevention and Treatment of Cancer. Nutrients. 2016 Aug 22;8(8):515.
- Niedzwiecki A, Roomi MW, Kalinovsky T, Rath M. Anticancer Efficacy of Polyphenols and Their Combinations. Nutrients. 2016 Sep 9;8(9):552.
- Livingstone TL, Beasy G, Mills RD, Plumb J, Needs PW, Mithen R, Traka MH. Plant Bioactives and the Prevention of Prostate Cancer: Evidence from Human Studies. Nutrients. 2019 Sep 18;11(9):2245.
- Abdal Dayem A, Choi HY, Yang GM, Kim K, Saha SK, Cho SG. The Anti-Cancer Effect of Polyphenols against Breast Cancer and Cancer Stem Cells: Molecular Mechanisms. Nutrients. 2016 Sep 21;8(9):581.
- Cory H, Passarelli S, Szeto J, Tamez M, Mattei J. The Role of Polyphenols in Human Health and Food Systems: A Mini-Review. Front Nutr. 2018 Sep 21;5:87.
- Clayton P, Rowbotham J. How the mid-Victorians worked, ate and died. Int J Environ Res Public Health. 2009 Mar;6(3):1235-53.
- Hussain T, Tan B, Yin Y, Blachier F, Tossou MC, Rahu N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxid Med Cell Longev. 2016;2016:7432797.
- Harikumar KB, Aggarwal BB. Resveratrol: a multitargeted agent for age-associated chronic diseases. Cell Cycle. 2008;7:1020–1035.
- Samodien E, Johnson R, Pheiffer C, Mabasa L, Erasmus M, Louw J, Chellan N. Diet-induced hypothalamic dysfunction and metabolic disease, and the therapeutic potential of polyphenols. Mol Metab. 2019 Sep;27:1-10.
- Colizzi C. The protective effects of polyphenols on Alzheimer’s disease: A systematic review. Alzheimers Dement (N Y). 2018 Nov 1;5:184-196.
- Soares S, Kohl S, Thalmann S, Mateus N, Meyerhof W, De Freitas V. Different phenolic compounds activate distinct human bitter taste receptors. J Agric Food Chem. 2013 Feb 20;61(7):1525-33.
- Ventura AK, Mennella JA. Innate and learned preferences for sweet taste during childhood. Curr Opin Clin Nutr Metab Care. 2011 Jul;14(4):379-84.
- Gustafson B, Nerstedt, A, Smith U. Reduced subcutaneous adipogenesis in human hypertrophic obesity is linked to senescent precursor cells. Nat Commun 10, 2757 (2019).
- Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, Victorelli S, Ruswhandi RA, Giorgadze N, Pirtskhalava T, Podgorni O, Enikolopov G, Johnson KO, Xu M, Inman C, Palmer AK, Schafer M, Weigl M, Ikeno Y, Burns TC, Passos JF, von Zglinicki T, Kirkland JL, Jurk D. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab. 2019 May 7;29(5):1061-1077.e8. Erratum in: Cell Metab. 2019 May 7;29(5):1233.
- Kuki S, Imanishi T, Kobayashi K, Matsuo Y, Obana M, Akasaka T. Hyperglycemia accelerated endothelial progenitor cell senescence via the activation of p38 mitogen-activated protein kinase. Circ J. 2006;70(8):1076–1081.
- Palmer AK, Tchkonia T, LeBrasseur NK, Chini EN, Xu M, Kirkland JL. Cellular senescence in type 2 diabetes: a therapeutic opportunity. Diabetes. 2015;64(7):2289–2298.
- Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, Victorelli S, Ruswhandi RA, Giorgadze N, Pirtskhalava T, Podgorni O, Enikolopov G, Johnson KO, Xu M, Inman C, Palmer AK, Schafer M, Weigl M, Ikeno Y, Burns TC, Passos JF, von Zglinicki T, Kirkland JL, Jurk D. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab. 2019 May 7;29(5):1061-1077.e8. Erratum in: Cell Metab. 2019 May 7;29(5):1233.
- Lawrence JM, Divers J, Isom S. Trends in Prevalence of Type 1 and Type 2 Diabetes in Children and Adolescents in the US, 2001-2017. JAMA. 2021;326(8):717-727.
- Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, Prata LG, van Dijk TH, Verkade E, Casaclang-Verzosa G, Johnson KO, Cubro H, Doornebal EJ, Ogrodnik M, Jurk D, Jensen MD, Chini EN, Miller JD, Matveyenko A, Stout MB, Schafer MJ, White TA, Hickson LJ, Demaria M, Garovic V, Grande J, Arriaga EA, Kuipers F, von Zglinicki T, LeBrasseur NK, Campisi J, Tchkonia T, Kirkland JL. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell. 2019 Jun;18(3):e12950.
- Maduro AT, Luís C, Soares R. Ageing, cellular senescence and the impact of diet: an overview. Porto Biomed J. 2021 Feb 11;6(1):e120.
- Yessenkyzy A, Saliev T, Zhanaliyeva M, Masoud AR, Umbayev B, Sergazy S, Krivykh E, Gulyayev A, Nurgozhin T. Polyphenols as Caloric-Restriction Mimetics and Autophagy Inducers in Aging Research. Nutrients. 2020 May 8;12(5):1344.
- Varela-Eirín M, Carpintero-Fernández P, Sánchez-Temprano A, Varela-Vázquez A, Paíno CL, Casado-Díaz A, Continente AC, Mato V, Fonseca E, Kandouz M, Blanco A, Caeiro JR, Mayán MD. Senolytic activity of small molecular polyphenols from olive restores chondrocyte redifferentiation and promotes a pro-regenerative environment in osteoarthritis. Aging (Albany NY). 2020 Aug 3;12(16):15882-15905.
- Ovadya Y, Landsberger T, Leins, H, Vadai E, Gal H, Biran A, Yosef R, Sagiv A, Agrawal A, Shapira A, Windheim J, Tsoory M, Schirmbeck R, Amit I, Geiger H, Krizhanovsky V. Impaired immune surveillance accelerates accumulation of senescent cells and aging. Nat Commun 9, 5435 (2018).
- Fiuza-Luces C, Valenzuela PL, Castillo-García A, Lucia A. Exercise Benefits Meet Cancer Immunosurveillance: Implications for Immunotherapy. Trends Cancer. 2021 Feb;7(2):91-93.
- Nieman DC, Wentz LM. The compelling link between physical activity and the body’s defense system. J Sport Health Sci. 2019 May;8(3):201-217.
- Sitlinger A, Brander DM, Bartlett DB. Impact of exercise on the immune system and outcomes in hematologic malignancies. Blood Adv. 2020 Apr 28;4(8):1801-1811.
- Tylutka A, Morawin B, Gramacki A, Zembron-Lacny A. Lifestyle exercise attenuates immunosenescence; flow cytometry analysis. BMC Geriatr. 2021 Mar 22;21(1):200.
- Salminen A, Kauppinen A, Kaarniranta K. AMPK activation inhibits the functions of myeloid-derived suppressor cells (MDSC): impact on cancer and aging. J Mol Med (Berl). 2019 Aug;97(8):1049-1064.
- Prantner D, Perkins DJ, Vogel SN. AMP-activated Kinase (AMPK) Promotes Innate Immunity and Antiviral Defense through Modulation of Stimulator of Interferon Genes (STING) Signaling. J Biol Chem. 2017 Jan 6;292(1):292-304.
- Nguyen PH, Gauhar R, Hwang SL, Dao TT, Park DC, Kim JE, Song H, Huh TL, Oh WK. New dammarane-type glucosides as potential activators of AMP-activated protein kinase (AMPK) from Gynostemma pentaphyllum. Bioorg Med Chem. 2011 Nov 1;19(21):6254-60.
- Garatachea N, Pareja-Galeano H, Sanchis-Gomar F, Santos-Lozano A, Fiuza-Luces C, Morán M, Emanuele E, Joyner MJ, Lucia A. Exercise attenuates the major hallmarks of aging. Rejuvenation Res. 2015 Feb;18(1):57-89.
- Serino A, Salazar G. Protective Role of Polyphenols against Vascular Inflammation, Aging and Cardiovascular Disease. Nutrients. 2018 Dec 28;11(1):53.
- Qureshi A.W. Ageing enhances the shedding of splenocyte microvesicles with endothelial pro-senescent effect that is prevented by a short-term intake of omega-3 PUFA EPA:DHA 6:1. Biochem. Pharmacol. 2020;173
- Mária J, Ingrid Ž. Effects of bioactive compounds on senescence and components of senescence associated secretory phenotypes in vitro. Food Funct. 2017 Jul 19;8(7):2394-2418.
- Pérez-Jiménez J, Neveu V, Vos F, Scalbert A. Identification of the 100 richest dietary sources of polyphenols: an application of the Phenol-Explorer database. Eur J Clin Nutr. 2010 Nov;64 Suppl 3:S112-20.
- Feng D, Cheng Y, Meng Y, Zou L, Huang S, Xie J. Multiple effects of curcumin on promoting expression of the exon 7-containing SMN2 transcript. Genes Nutr. 2015 Nov;10(6):40.
- Verma AK, Khan E, Mishra SK, Mishra A, Charlet-Berguerand N, Kumar A. Curcumin Regulates the r(CGG)exp RNA Hairpin Structure and Ameliorate Defects in Fragile X-Associated Tremor Ataxia Syndrome. Front Neurosci. 2020 Apr 7;14:295.
- Bielak-Zmijewska A, Grabowska W, Ciolko A, Bojko A, Mosieniak G, Bijoch Ł, Sikora E. The Role of Curcumin in the Modulation of Ageing. Int J Mol Sci. 2019 Mar 12;20(5):1239.
- Cherif H, Bisson D, Kocabas S, Haglund L. Senolytic and anti-inflammatory effects of curcumin and O-vanillin to reduce intervertebral disc degeneration and low back pain. Orthopaedic ProceedingsVol. 102-B, No. Supp_8, The Canadian Orthopaedic Association (COA) and The International Combined Orthopaedic Research Societies (ICORS) Meeting, Montreal, Canada, June 2019. Part 3.
https://online.boneandjoint.org.uk/doi/abs/10.1302/1358-992X.2020.8.053
- Malavolta M, Bracci M, Santarelli L, Sayeed MA, Pierpaoli E, Giacconi R, Costarelli L, Piacenza F, Basso A, Cardelli M, Provinciali M. Inducers of Senescence, Toxic Compounds, and Senolytics: The Multiple Faces of Nrf2-Activating Phytochemicals in Cancer Adjuvant Therapy. Mediators Inflamm. 2018 Feb 12;2018:4159013.
- Briskey D, Sax A, Mallard AR, Rao A. Increased bioavailability of curcumin using a novel dispersion technology system (LipiSperse®). Eur J Nutr. 2019 Aug;58(5):2087-2097.
- Hammarstedt A, Gogg S, Hedjazifar S, Nerstedt A, Smith U. Impaired adipogenesis and dysfunctional adipose tissue in human hypertrophic obesity. Physiol Rev. 2018;98(4):1911–1941.
- Kirkland JL, Hollenberg CH, Gillon WS. Age, anatomic site, and the replication and differentiation of adipocyte precursors. Am J Phys. 1990;258(2):C206–C210.
- Irhimeh MR, Fitton JH, Lowenthal RM. Fucoidan ingestion increases the expression of CXCR4 on human CD34+ cells. Exp. Hematol. 2007;35:989–994.
- Lapasset L, Milhavet O, Prieur A, Besnard E, Babled A, Aït-Hamou N, Leschik J, Pellestor F, Ramirez JM, De Vos J, Lehmann S, Lemaitre JM. Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes Dev. 2011 Nov 1;25(21):2248-53.
- Cheng Z, Peng HL, Zhang R, Fu XM, Zhang GS. Rejuvenation of Cardiac Tissue Developed from Reprogrammed Aged Somatic Cells. Rejuvenation Res. 2017 Oct;20(5):389-400
- Sarkar TJ, Quarta M, Mukherjee S, Colville A, Paine P, Doan L, Tran CM, Chu CR, Horvath S, Qi LS, Bhutani N, Rando TA, Sebastiano V. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat Commun. 2020 Mar 24;11(1):1545.
- Villeda SA, Plambeck KE, Middeldorp J, Castellano JM, Mosher KI, Luo J, Smith LK, Bieri G, Lin K, Berdnik D, Wabl R, Udeochu J, Wheatley EG, Zou B, Simmons DA, Xie XS, Longo FM, Wyss-Coray T. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat Med. 2014 Jun;20(6):659-63.