Ageing: Is there a role for arachidonic acid and other bioactive lipids? A review

Undurti N Das, Undurti N Das

Abstract

Ageing is inevitable. Recent studies suggest that it could be delayed. Low-grade systemic inflammation is seen in type 2 diabetes mellitus, hypertension and endothelial dysfunction that are common with increasing age. In all these conditions, an alteration in arachidonic acid (AA) metabolism is seen in the form of increased formation of pro-inflammatory eicosanoids and decreased production of anti-inflammatory lipoxins, resolvins, protectins and maresins and decreased activity of desaturases. Calorie restriction, exercise and parabiosis delay age-related changes that could be related to enhanced proliferation of stem cells, decrease in inflammation and transfer of GDF-11 (growth differentiation factor-11) and other related molecules from the young to the old, increase in the formation of lipoxin A4, resolvins, protectins and maresins, hydrogen sulfide (H2S) and nitric oxide (NO); inhibition of ageing-related hypothalamic or brain IKK-β and NF-kB activation, decreased gonadotropin-releasing hormone (GnRH) release resulting in increased neurogenesis and consequent decelerated ageing. This suggests that hypothalamus participates in ageing process. N-acylethanolamines (NAEs) and lipid-derived signalling molecules can be tuned favorably under dietary restriction to extend lifespan and/or prevent advanced age associated diseases in an mTOR dependent pathway manner. Sulfur amino acid (SAA) restriction increased hydrogen sulfide (H2S) production and protected tissues from hypoxia and tissue damage. Anti-inflammatory metabolites formed from AA such as LXA4, resolvins, protectins and maresins enhance production of NO, CO, H2S; suppress NF-kB expression and alter mTOR expression and thus, may aid in delaying ageing process. Dietary restriction and exercise enhance AA metabolism to form LXA4, resolvins, protectins and maresins that have anti-inflammatory actions. AA and their metabolites also influence stem cell biology, enhance neurogenesis to improve memory and augment autophagy to prolong life span. Thus, AA and other PUFAs and their anti-inflammatory metabolites inhibit inflammation, augment stem cell proliferation, restore to normal lipid-derived signaling molecules and NO and H2S production, enhance autophagy and prolong life span.

Keywords: Ageing; Arachidonic acid; Calorie restriction; GDF-11; Hydrogen sulfide; Hypothalamus; Inflammation; Lipids; Nitric oxide; Polyunsaturated fatty acids; Stem cells; Sulfur amino acid.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Scheme showing metabolism of essential fatty acids, their role in inflammation and cytoprotection of endothelial cells.
Fig. 2
Fig. 2
Scheme showing metabolism of arachidonic acid. AA can react with NO and NO2 and form vicinal nitrohydroxyeicosatrienoic acids that have vasodilator actions . Even EPA and DHA may give rise to similar metabolites that are formed from AA. Fatty acid hydroxy fatty acids (FAHFAs) are newly discovered and are also called as lipokines. They can be formed from all PUFAs. So far lipokines derived from DHA, LA, palmitic acid and stearic acid have been described. But are likely to be formed form other PUFAs as well. They are present in the plasma, adipose tissue and human breast milk. Some of the DHA derived FAHFAs include: 9- and 13-hydroxyoctadecadienoic acid (HLA) or 14-hydroxydocosahexaenoic acid (HDHA), termed 9-DHAHLA, 13-DHAHLA, and 14-DHAHDHA. FAHFAs have the potential to improve blood sugar, protect against diabetes, and reduce inflammation. PAHSA, the combination of palmitic acid and hydroxy stearic acid, was abundantly found in the fat of diabetes-resistant mice and was significantly reduced in humans with early stages of diabetes. When fed to obese diabetic mice, 9-PAHSA was reported to contribute to glucose-insulin homeostasis and to elicit anti-inflammatory effects. FAHFAs do exist at low levels within certain foods, but are mainly synthesized in the body. FHAFAs may also form from AA. Patients with type 2 diabetes have low plasma levels of FHAFAs, AA and LXA4, which have anti-inflammatory actions. This may imply that decreased formation of AA in the elderly may render them to develop low-grade systemic inflammation, partly, due to decreased formation of FHAFAs and LXA4 from AA. N-acylethanolamine (NAE) is a type of fatty acid amide formed when one of several types of acyl group is linked to the nitrogen atom of ethanolamine. These amides can be formed from a fatty acid and ethanolamine with the release of a molecule of water. NAE can be formed due to the action of phospholipase D that cleave the phospholipid unit from N-acylphosphatidylethanolamines. Examples of N-acylethanolamines include: (i) Anandamide (N-arachidonoylethanolamine; NAE 20:4) or arachidonlyethanolamine (AEA) is the amide of arachidonic acid (20:4 ω-6) and ethanolamine. It is the ligand of both cannabinoid receptors and vanilloid receptor that attenuates pain sensation. (ii) N-Palmitoylethanolamine is the amide of palmitic acid (16:0) and ethanolamine. It has anti-inflammatory activity and also attenuates pain sensation. N-Oleoylethanolamine is the amide of oleic acid (18:1) and ethanolamine. It has anorexic effects and enables fat breakdown by stimulating PPAR-alpha. (iii) N-Stearoylethanolamine is the amide of stearic acid (18:0) and ethanolamine. It has pro-apoptotic activity. It operates independently of the known cannabinoid and vanilloid receptors targeted by anandamide. (iv) N-Docosahexaenoylethanolamine is the amide of docosahexaenoic acid (22:6) and ethanolamine. It has anti-proliferative effects on prostate cancer cell lines and promotes synaptogenesis. Thus, NAEs may be formed from PUFAs that have important biological functions.
Fig. 3
Fig. 3
Scheme showing possible relationship among PGE2, LXA4, and various PLA2 enzymes as seen in inflammation and inflammation resolution process. PGE2; LXA4; iPLA2; sPLA2; cPLA2; COX-2. All these concentrations and activities of enzymes as expected to behave during normal inflammatory process. PGE2 when inflammation persists; COX-2 when inflammation persists. LXA4 when resolution of inflammation is defective. Although possible changes in the activities of various PLA2 are not shown during persistance of inflammation or defective resolution of inflammation, they are expected to behave in tune with the concentrations of PGE2 and LXA4. It also need to be noted that despite the fact that LXA4, resolvins, protectins and maresins have anti-inflammatory actions, there could be subtle differences in their major and minor actions with some amount of overlap in their anti-inflammatory actions. Though the role of nitrolipids is not shown, it is expected to behave similar to LXA4. It is evident from the figure that there are two waves of release of AA (and other PUFAs), one in the early period of inflammation (within the first 24 h due the activation of iPLA2) that leads to the formation of PGE2 and other pro-inflammatory molecules. Once the concentrations of PGE2 reach the optimum level (say by the end of 24–48 h), a second wave of AA release occurs (due to the activation of sPLA2) that leads to the formation of LXA4 that initiates resolution of inflammation. The activation of cPLA2 occurs around 48–72 h in all probability to accelerate or continue the resolution of inflammation process. The activation of iPLA2 and formation of PGE2 are closely associated with the activation of COX-2. In this process of inflammation and resolution of inflammation there is a critical role for PGDH enzyme (see text for details). With regard to the actions of LXA4, resolvins, protectins and maresins, it is to be noted here that though all these are anti-inflammatory molecules they may have slightly but critically important differences in their actions to resolve the inflammation. For instance, LXA4 is needed to induce anti-inflammatory events (to suppress inflammation and this is not equal to resolution of inflammation. During the process of suppressing inflammation, LXA4 may inhibit leukocyte infiltration); while resolvins are needed for resolution of inflammation (such as removing the debris of wound, phagocytosis of dead leukocytes, etc.,); protectins protect normal cells/tissues from further damage); and maresins may act on stem cells for the repair process to proceed and restore homeostasis. Despite these different actions assigned to different molecules (LXA4, resolvins, protectins and maresins), all these bioactive lipids have all the enumerated actions except that the degree to which each action is brought about may be variable and it may vary from cell/tissues that are in the need of their action. It is also depicted in the figure how this sequence of orderly activation and deactivation of PLA2, COX-2 and formation of PGE2 and LXA4 are likely to get deranged in the face of failure of resolution of inflammation process. It is likely that in patients with hypertension, diabetes mellitus and ageing there is low-grade systemic inflammation as a result of sustained activation of COX2 and formation of PGE2 and failure of formation of adequate amounts of LXA4 and other anti-inflammatory compounds and corresponding activation of PLA2 at the most appropriate time. It is noteworthy that failure of the inflammation resolution process may lead to the onset of ageing associated osteoporosis, sarcopenia and when this inflammatory process is severe it can lead to sepsis and septic shock.
Fig. 4
Fig. 4
Scheme showing relationship among ageing and its associated diseases and their relationship to hypothalamus, oxidative stress, PUFAs, lipoxins, resolvins, protectins, maresins, eicosanoids, CO, NO, H2S and telomere length. High calorie diet stimulates ROS generation that may overwhelm antioxidant system protection in adipose and other tissues; augment the synthesis of pro-inflammatory cytokines, inhibit the formation of anti-inflammatory cytokines that ultimately results in low-grade systemic inflammation, enhance DNA damage and ageing. These events may lead to ageing of endothelial cells and telomere shortening, and alteration in p53 expression. These events cause endothelial dysfunction and insulin resistance leading to the development of hypertension, type 2 diabetes mellitus, atherosclerosis and ageing. High calorie diet and insulin resistance suppress Δ6 and Δ5 desaturases activity resulting in reduced formation of PUFAs, the precursors of lipoxins, resolvins, protectins and maresins. Decreased lipoxins, resolvins, protectins and maresins impair resolution of inflammation, DNA damage, telomere shortening, p53 dysfunction, and stem cell function leading to the onset and progression of ageing and age-associated diseases. These events may also decrease CO, NO and H2S production. PUFAs and their metabolites influence stem cell biology and thus, affect ageing process and ageing-associated diseases including Alzheimer’s disease (for further details see text). PUFAs can give rise to FAHFAs that have anti-inflammatory properties and may enhance NO, CO and H2S production, and mediate exercise-induced anti-inflammatory actions. PUFAs form precursors to anti-inflammatory lipoxins, resolvins, protectins and maresins that suppress production of pro-inflammatory IL-6, TNF-α and prostanoids. It is not yet known but possible that FAHFAs may suppress tumor cell growth, inhibit inflammatory events that occur in hypothalamus due to high fat diet. Though the role of p53 in ageing and diseases is not discussed in detail here, it may be noted that p53 is the guardian of the genome. PUFAs and their metabolites, cytokines, NO, CO, H2S, ROS, GDF-11, GnRH and NAE may modulate the action of p53. For instance, exercise reduces the incidence of cancer, possibly, by augmenting the production of IL-6 and TNF-α that are cytotoxic to tumor cells either by their direct action and/or enhancing the production of ROS that are tumoricidal. In addition, exercise may enhance the expression and action of p53 that leads to apoptosis of cancer cells. PUFAs have tumoricidal action and may bring about this action by augmenting free radical generation and formation of excess lipid peroxides selectively in tumor cells and augmenting the expression and action of p53.

References

    1. Vasto S., Carruba G., Lio D., Colonna-Romano G., Di Bona D., Candore G. Inflammation, ageing and cancer. Mech Ageing Dev. 2009;130(1–2):40–45.
    1. Adams A.A., Katepalli M.P., Kohler K., Reedy S.E., Stilz J.P., Vick M.M. Effect of body condition, body weight and adiposity on inflammatory cytokine responses in old horses. Vet Immunol Immunopathol. 2009;127(3–4):286–294.
    1. Guarner V., Rubio-Ruiz M.E. Low-grade systemic inflammation connects aging, metabolic syndrome and cardiovascular disease. Interdiscip Top Gerontol. 2015;40:99–106.
    1. Jurk D., Wilson C., Passos J.F., Oakley F., Correia-Melo C., Greaves L. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat Commun. 2014;2:4172.
    1. Counter C.M., Avilion A.A., LeFeuvre C.E., Stewart N.G., Greider C.W., Harley C.B. Telomere shortening associated with chromosome instability is arrested in immortal cells which express telomerase activity. EMBO J. 1992;11:1921–1929.
    1. Counter C.M., Botelho F.M., Wang P., Harley C.B., Bacchetti S. Stabilization of short telomeres and telomerase activity accompany immortalization of Epstein-Barr virus-transformed human B lymphocytes. J Virol. 1994;68:3410–3414.
    1. Allsopp R.C., Harley C.B. Evidence for a critical telomere length in senescent human fibroblasts. Exp Cell Res. 1995;219:130–136.
    1. MacDonald L., Radler M., Paolini A.G., Kent S. Calorie restriction attenuates LPS-induced sickness behavior and shifts hypothalamic signaling pathways to an anti-inflammatory bias. Am J Physiol Regul Integr Comp Physiol. 2011;301:R172–R184.
    1. MacDonald L., Hazi A., Paolini A.G., Kent S. Calorie restriction dose-dependently abates lipopolysaccharide-induced fever, sickness behavior, and circulating interleukin-6 while increasing corticosterone. Brain Behav Immun. 2014;40:18–26.
    1. Radler M.E., Wright B.J., Walker F.R., Hale M.W., Kent S. Calorie restriction increases lipopolysaccharide-induced neuropeptide Y immunolabeling and reduces microglial cell area in the arcuate hypothalamic nucleus. Neuroscience. 2015;285:236–247.
    1. Poorani R., Bhatt A.N., Dwarakanath B.S., Das U.N. COX-2, aspirin and metabolism of arachidonic, eicosapentaenoic and docosahexaenoic acids and their physiological and clinical significance. Eur J Pharmacol. 2016;785:116–132.
    1. Das U.N., Ells G., Begin M.E., Horrobin D.F. Clinical significance of essential fatty acids. Nutrition. 1988;4:337–342.
    1. Das U.N. Nutritional factors in the pathobiology of human essential hypertension. Nutrition. 2001;17:337–346.
    1. Das U.N. When less is adequate: protein and calorie restriction boosts immunity and possibly, longevity-but, how and why? Nutrition. 2009;25:892–895.
    1. Das U.N. A defect in the activity of Delta6 and Delta5 desaturases may be a factor predisposing to the development of insulin resistance syndrome. Prostagl Leukot Essent Fatty Acids. 2005;72:343–350.
    1. Valenzuela R., Barrera C., Espinosa A., Llanos P., Orellana P., Videla L.A. Reduction in the desaturation capacity of the liver in mice subjected to high fat diet: relation to LCPUFA depletion in liver and extrahepatic tissues. Prostagl Leukot Essent Fatty Acids. 2015;98:7–14.
    1. Dalvi P.S., Chalmers J.A., Luo V., Han D.Y., Wellhauser L., Liu Y. High fat induces acute and chronic inflammation in the hypothalamus: effect of high-fat diet, palmitate and TNF-α on appetite-regulating NPY neurons. Int J Obes (Lond) 2017;41:149–158.
    1. Yuan Y., Naito H., Jia X., Kitamori K., Nakajima T. Combination of hypertension along with a high fat and cholesterol diet induces severe hepatic inflammation in rats via a signaling network comprising. Nutrients. 2017;9(9):E1018.
    1. Fang Y., Wang S., Zhu T., Zhang Y., Lian X. Atherogenic high cholesterol/high fat diet induces TLRs-associated pulmonary inflammation in C57BL/6J mice. Inflamm Res. 2017;66:39–47.
    1. Brunetti L., Leone S., Chiavaroli A., Orlando G., Recinella L., Ferrante C. Cafeteria diet increases prostaglandin E2 levels in rat prostate, kidney and testis. Int J Immunopathol Pharmacol. 2010;23:1073–1078.
    1. Wang W., Yang J., Yang H., Sanidad K.Z., Hammock B.D., Kim D. Effects of high-fat diet on plasma profiles of eicosanoid metabolites in mice. Prostagl Other Lipid Mediat. 2016;127:9–13.
    1. Das U.N. Obesity: genes, brain, gut and environment. Nutrition. 2010;26:459–473.
    1. Das U.N. Wiley-Blackwell Publishers; Ames, IA, USA: 2010. Metabolic syndrome pathophysiology: the role of essential fatty acids and their metabolites.
    1. Jeanclos E., Krolewski A., Skurnick J., Kimura M., Aviv H., Warram J.H. Shortened telomere length in white blood cells of patients with IDDM. Diabetes. 1998;47:482–486.
    1. Adaikalakoteswari A., Balasubramanyam M., Mohan V. Telomere shortening occurs in Asian Indian Type 2 diabetic patients. Diabet Med. 2005;22:1151–1156.
    1. Sampson M.J., Winterbone M.S., Hughes J.C., Dozio N., Hughes D.A. Monocyte telomere shortening and oxidative DNA damage in type 2 diabetes. Diabetes Care. 2006;29:283–289.
    1. Adaikalakoteswari A., Balasubramanyam M., Ravikumar R., Deepa R., Mohan V. Association of telomere shortening with impaired glucose tolerance and diabetic macroangiopathy. Atherosclerosis. 2007;195:83–89.
    1. Tentolouris N., Nzietchueng R., Cattan V., Poitevin G., Lacolley P., Papazafiropoulou A. White blood cells telomere length is shorter in males with type 2 diabetes and microalbuminuria. Diabetes Care. 2007;30:2909–2915.
    1. Uziel O., Singer J.A., Danicek V., Sahar G., Berkov E., Luchansky M. Telomere dynamics in arteries and mononuclear cells of diabetic patients: effect of diabetes and of glycemic control. Exp Gerontol. 2007;42:971–978.
    1. Salpea K.D., Humphries S.E. Telomere length in atherosclerosis and diabetes. Atherosclerosis. 2010;209:35–38.
    1. Jeanclos E., Schork N.J., Kyvik K.O., Kimura M., Skurnick J.H., Aviv A. Telomere length inversely correlates with pulse pressure and is highly familial. Hypertension. 2000;36:195–200.
    1. Tristano A., Eugenia Chollet M., Willson M.L., Adjounian H., Fernanda Correa M., Borges A. Telomerase activity in peripheral blood leukocytes from patients with essential hypertension. Med Clin (Barc) 2003;120:365–369.
    1. Benetos A., Gardner J.P., Zureik M., Labat C., Xiaobin L., Adamopoulos C. Short telomeres are associated with increased carotid atherosclerosis in hypertensive subjects. Hypertension. 2004;43:182–185.
    1. Nakashima H., Ozono R., Suyama C., Sueda T., Kambe M., Oshima T. Telomere attrition in white blood cell correlating with cardiovascular damage. Hypertens Res. 2004;27:319–325.
    1. Gardner J.P., Li S., Srinivasan S.R., Chen W., Kimura M., Lu X. Rise in insulin resistance is associated with escalated telomere attrition. Circulation. 2005;111:2171–2177.
    1. Aviv A., Valdes A., Gardner J.P., Swaminathan R., Kimura M., Spector T.D. Menopause modifies the association of leukocyte telomere length with insulin resistance and inflammation. J Clin Endocrinol Metab. 2006;91:635–640.
    1. Demissie S., Levy D., Benjamin E.J., Cupples L.A., Gardner J.P., Herbert A. Insulin resistance, oxidative stress, hypertension, and leukocyte telomere length in men from the Framingham Heart Study. Aging Cell. 2006;5:325–330.
    1. Rufer N., Brümmendorf T.H., Kolvraa S., Bischoff C., Christensen K., Bischoff L. Telomere fluorescence measurements in granulocytes and T lymphocyte subsets point to a high turnover of hematopoietic stem cells and memory T cells in early childhood. J Exp Med. 1999;190:157–167.
    1. Aviv A. Leukocyte telomere length, hypertension, and atherosclerosis: are there potential mechanistic explanations? Hypertension. 2009;53:590–591.
    1. Satoh M., Ishikawa Y., Takahashi Y., Itoh T., Minami Y., Nakamura M. Association between oxidative DNA damage and telomere shortening in circulating endothelial progenitor cells obtained from metabolic syndrome patients with coronary artery disease. Atherosclerosis. 2008;198:347–353.
    1. Das U.N. Essential fatty acids: biochemistry, physiology, and pathology. Biotech J. 2006;1:420–439.
    1. Das U.N. Lipoxins, resolvins, protectins, maresins and nitrolipids and their clinical implications with specific reference to cancer: part I. Clin Lipidol. 2013;8:437–463.
    1. Das U.N. Lipoxins, resolvins, protectins, maresins and nitrolipids and their clinical implications with specific reference to diabetes mellitus and other diseases: part II. Clin Lipidol. 2013;8:465–480.
    1. Lima E.S., Di Mascio P., Abdalla D.S.P. Cholesteryl nitrolinoleate, a nitrated lipid present in human blood plasma and lipoproteins. J Lipid Res. 2003;44:1660–1666.
    1. Ferreira A.M., Ferrari M.I., Trostchansky A. Cholesteryl nitrolinoleate, a nitrated lipid present in human blood plasma and lipoproteins. J Lipid Res. 2003;44:1660–1666.
    1. Lima E.S., Bonini M.G., Augusto O., Barbeiro H.V., Souza H.P., Abdalla D.S. Nitrated lipids decompose to nitric oxide and lipid radicals and cause vasorelaxation. Free Radic Biol Med. 2005;39:532–539.
    1. Coles B., Bloodsworth A., Clark S.R., Lewis M.J., Cross A.R., Freeman B.A. Nitrolinoleate inhibits superoxide generation, degranulation, and integrin expression by human neutrophils: novel antiinflammatory properties of nitric oxide-derived reactive species in vascular cells. Circ Res. 2002;91:375–381.
    1. Coles B., Bloodsworth A., Eiserich J.P., Coffey M.J., McLoughlin R.M., Giddings J.C. Nitrolinoleate inhibits platelet activation by attenuating calcium mobilization and inducing phosphorylation of vasodilator-stimulated phosphoprotein through elevation of cAMP. J Biol Chem. 2002;277:5832–5840.
    1. Tain Y.L., Lee W.C., Wu K.L.H., Leu S., Chan J.Y.H. Targeting arachidonic acid pathway to prevent programmed hypertension in maternal fructose-fed male adult rat offspring. J Nutr Biochem. 2016;38:86–92.
    1. Tain Y.L., Leu S., Wu K.L., Lee W.C., Chan J.Y. Melatonin prevents maternal fructose intake-induced programmed hypertension in the offspring: roles of nitric oxide and arachidonic acid metabolites. J Pineal Res. 2014;57:80–89.
    1. Prabha P.S., Das U.N., Koratkar R., Sagar P.S., Ramesh G. Free radical generation, lipid peroxidation and essential fatty acids in uncontrolled essential hypertension. Prostagl Leukot Essent Fatty Acids. 1990;41:27–33.
    1. Singer P., Jaeger W., Voigt S., Thiel H. Defective desaturation and elongation of n-6 and n-3 fatty acids in hypertensive patients. Prostagl Leukot Med. 1984;15:159–165.
    1. Das U.N. Essential fatty acid metabolism in patients with essential hypertension, diabetes mellitus and coronary heart disease. Prostagl Leukot Essent Fatty Acids. 1995;52:387–391.
    1. Mtabaji J.P., Manku M.S., Horrobin D.F. Abnormalities in dihomo-gamma-linolenic acid release in the pathogenesis of hypertension. Am J Hypertens. 1993;6(6 Pt 1):458–462.
    1. Yagi S., Aihara K., Fukuda D., Takashima A., Bando M., Hara T. Reduced ratio of eicosapentaenoic acid and docosahexaenoic acid to arachidonic acid is associated with early onset of acute coronary syndrome. Nutr J. 2015;14:111.
    1. Gromovsky A.D., Schugar R.C., Brown A.L., Helsley R.N., Burrows A.C., Ferguson D. Δ-5 Fatty acid desaturase FADS1 impacts metabolic disease by balancing proinflammatory and proresolving lipid mediators. Arterioscler Thromb Vasc Biol. 2018;38:218–231.
    1. Yary T., Voutilainen S., Tuomainen T.P., Ruusunen A., Nurmi T., Virtanen J.K. Serum n-6 polyunsaturated fatty acids, Δ5- and Δ6-desaturase activities, and risk of incident type 2 diabetes in men: the Kuopio Ischaemic Heart Disease Risk Factor Study. Am J Clin Nutr. 2016;103:1337–1343.
    1. Jacobs S., Schiller K., Jansen E.H., Boeing H., Schulze M.B., Kröger J. Evaluation of various biomarkers as potential mediators of the association between Δ5 desaturase, Δ6 desaturase, and stearoyl-CoA desaturase activity and incident type 2 diabetes in the European Prospective Investigation into Cancer and Nutrition-Potsdam Study. Am J Clin Nutr. 2015;102:155–164.
    1. Akter S., Kurotani K., Sato M., Hayashi T., Kuwahara K., Matsushita Y. High serum phospholipid dihomo-γ-linoleic acid concentration and low Δ5-desaturase activity are associated with increased risk of type 2 diabetes among japanese adults in the hitachi health study. J Nutr. 2017;147:1558–1566.
    1. Kim H.W., Cheon Y., Modi H.R., Rapoport S.I., Rao J.S. Effects of chronic clozapine administration on markers of arachidonic acid cascade and synaptic integrity in rat brain. Psychopharmacology. 2012;222:663–674.
    1. Lu Y., Vaarhorst A., Merry A.H., Dollé M.E., Hovenier R., Imholz S. Markers of endogenous desaturase activity and risk of coronary heart disease in the CAREMA cohort study. PLoS ONE. 2012;7:e41681.
    1. Vijay Kumar K., Das U.N. Lipid peroxides and essential fatty acids in patients with coronary heart disease. J Nutritional Med. 1994;4:33–37.
    1. Das U.N. The lipids that matter from infant nutrition to insulin resistance. Prostagl Leukot Essen Fatty Acids. 2002;67:1–12.
    1. Das U.N. A defect in the activities of Δ6 and Δ5 desaturases and pro-resolution bioactive lipids in the pathobiology of non-alcoholic fatty liver disease. World J Diabetes. 2011;2:176–188.
    1. Das U.N. A defect in the activity of Δ6 and Δ5 desaturases may be a factor in the initiation and progression of atherosclerosis. Prostagl Leukot Essen Fatty Acids. 2007;76:251–268.
    1. Das U.N. A defect in Δ6 and Δ5 desaturases may be a factor in the initiation and progression of insulin resistance, the metabolic syndrome and ischemic heart disease in South Asians. Lipid Health Dis. 2010;9:130.
    1. Das U.N. Springer-Verlag; 2011. Molecular basis of health and disease.
    1. Balazy M., Iesaki T., Park J.L., Jiang H., Kaminski P.M., Wolin M.S. Vicinal nitrohydroxyeicosatrienoic acids: vasodilator lipids formed by reaction of nitrogen dioxide with arachidonic acid. J Pharmacol Exp Ther. 2001;299:611–619.
    1. Matsuzaka T., Shimano H., Yahagi N. Dual regulation of mouse Delta(5)- and Delta(6)-desaturase gene expression by SREBP-1 and PPARalpha. J Lipid Res. 2002;43:107–114.
    1. Farzaneh-Far R., Lin J., Epel E.S., Harris W.S., Blackburn E.H., Whooley M.A. Association of marine omega-3 fatty acid levels with telomeric aging in patients with coronary heart disease. JAMA. 2010;303:250–257.
    1. Das U.N. Telomere length and polyunsaturated fatty acids. Nutrition. 2014;30:1218–1221.
    1. Richards J.B., Valdes A.M., Gardner J.P., Paximadas D., Kimura M., Nessa A. Higher serum vitamin D concentrations are associated with longer leukocyte telomere length in women. Am J Clin Nutr. 2007;86:1420–1425.
    1. Xu Q., Parks C.G., DeRoo L.A., Cawthon R.M., Sandler D.P., Chen H. Multivitamin use and telomere length in women. Am J Clin Nutr. 2009;89:1857–1863.
    1. Furumoto K., Inoue E., Nagao N., Hiyama E., Miwa N. Age-dependent telomere shortening is slowed down by enrichment of intracellular vitamin C via suppression of oxidative stress. Life Sci. 1998;63:935–948.
    1. Tanaka Y., Moritoh Y., Miwa N. Age-dependent telomere-shortening is repressed by phosphorylated alpha-tocopherol together with cellular longevity and intracellular oxidative-stress reduction in human brain microvascular endotheliocytes. J Cell Biochem. 2007;102:689–703.
    1. Paul L., Cattaneo M., D’Angelo A. Telomere length in peripheral blood mononuclear cells is associated with folate status in men. J Nutr. 2009;139:1273–1278.
    1. Das U.N. Exploring the actions of vitamin C. Canadian Med Assoc J. 2001;165:13–14.
    1. Srivastava K.C. Ascorbic acid enhances the formation of prostaglandin E1 in washed human platelets and prostacyclin in rat aortic rings. Prostagl Leukot Med. 1985;18:227–233.
    1. Das U.N. Folic acid says NO to vascular diseases. Nutrition. 2003;19:686–692.
    1. Wadhwani N.S., Manglekar R.R., Dangat K.D., Kulkarni A.V., Joshi S.R. Effect of maternal micronutrients (folic acid, vitamin B12) and omega 3 fatty acids on liver fatty acid desaturases and transport proteins in Wistar rats. Prostagl Leukot Essent Fatty Acids. 2012;86:21–27.
    1. Richards J.B., Valdes A.M., Gardner J.P., Kato B.S., Siva A., Kimura M. Homocysteine levels and leukocyte telomere length. Atherosclerosis. 2008;200:271–277.
    1. Matsui-Hirai H., Hayashi T., Yamamoto S., Ina K., Maeda M., Kotani H. Dose-dependent modulatory effects of insulin on glucose-induced endothelial senescence in vitro and in vivo: a relationship between telomeres and nitric oxide. J Pharmacol Exp Ther. 2011;337:591–599.
    1. Makino N., Maeda T., Oyama J., Higuchi Y., Mimori K. Improving insulin sensitivity via activation of PPAR-gamma increases telomerase activity in the heart of OLETF rats. Am J Physiol Heart Circ Physiol. 2009;297:H2188–H2195.
    1. Das U.N. Is insulin an anti-inflammatory molecule? Nutrition. 2001;17:409–413.
    1. Dandona P., Ghanim H., Green K., Sia C.L., Abuaysheh S., Kuhadiya N. Insulin infusion suppresses while glucose infusion induces Toll-like receptors and high-mobility group-B1 protein expression in mononuclear cells of type 1 diabetes patients. Am J Physiol Endocrinol Metab. 2013;304:E810–E818.
    1. Das U.N. Is insulin an endogenous cardioprotector? Crit Care. 2002;6:389–393.
    1. Laight D.W., Kaw A.V., Carrier M.J., Anggard E.E. Interaction between superoxide anion and nitric oxide in the regulation of vascular endothelial function. Br J Pharmacol. 1998;124:238–244.
    1. Mohanty P., Hamouda W., Garg R., Aljada A., Ghanim H., Dandona P. Glucose challenge stimulates reactive oxygen species (ROS) generation by leukocytes. J Clin Endocrinol Metab. 2000;85:2970–2973.
    1. Booth G., Stalker T.J., Lefer A.M., Scalia R. Elevated ambient glucose induces acute inflammatory events in the microvasculature: effects of insulin. Am J Physiol. 2001;280:E848–E856.
    1. Das U.N. Insulin and the critically ill. Crit Care. 2002;6:262–263.
    1. Dandona P., Aljada A., Mohanty P., Ghanim H., Hamouda W., Assian E. Insulin inhibits intranuclear nuclear factor kappaB and stimulates IkappaB in mononuclear cells in obese subjects: evidence for an inflammatory effect? J Clin Endocrinol Metab. 2001;86:3257–3265.
    1. Aljada A., Saadesh R., Assian E., Ghanim H., Dandona P. Insulin inhibits the expression of intercellular adhesion molecule-1 by human aortic endothelial cells through stimulation of nitric oxide. J Clin Endocrinol Metab. 2000;85:2572–2575.
    1. Aljada A., Ghanim H., Saadeh R., Dandona P. Insulin inhibits NFκB and MCP-1 expression in human aortic endothelial cells. J Clin Endocrinol Metab. 2001;86:450–453.
    1. Schambelan M., Blake S., Sraer J., Bens M., Nivez M.P. Wahbe F Increased prostaglandin production by glomeruli isolated from rats with streptozotocin-induced diabetes mellitus. J Clin Invest. 1985;75:404–412.
    1. Tesfamariam B., Brown M.L., Deykin D., Cohen R.A. Elevated glucose promotes generation of endothelium-derived vasoconstrictor prostanoids in rabbit aorta. J Clin Invest. 1990;85:929–932.
    1. Valentovic M.A., Lubawy W.C. Elevated glucose in vivo and in vitro adversely alters prostaglandin generation in rat aortas and platelets. Prostagl Leukot Med. 1985;19:271–277.
    1. Yatomi A., Iguchi A., Yanagisawa S., Matsunaga H., Niki I., Sakamoto N. Prostaglandins affect the central nervous system to produce hyperglycemia in rats. Endocrinology. 1987;121:36–41.
    1. Nonogaki K., Iguchi A., Zhu L.X., Kunoh Y., Tamagawa T., Sakamoto N. Involvement of central cholinergic muscarinic receptors and histamine H1 receptors in hyperglycemia induced by prostaglandin F2 alpha in rats. Neuroendocrinology. 1993;57:146–151.
    1. Cosentino F., Eto M., De Paolis P., van der Loo B., Bachschmid M., Ullrich V. High glucose causes upregulation of cyclooxygenase-2 and alters prostanoid profile in human endothelial cells: role of protein kinase C and reactive oxygen species. Circulation. 2003;107:1017–1023.
    1. Morita I., Murota S. Effect of aging on the prostaglandin-synthesizing system in rat liver. Prostagl Med. 1980;4:45–52.
    1. Ignatowska-Switalska H., Januszewicz P. Urinary prostaglandins E2 and F2 alpha in healthy newborns, infants, children, and adults. Prostagl Med. 1980;5:289–296.
    1. Goodwin J.S. Increased sensitivity to prostaglandin E2 in old people. Prostagl Med. 1979;3:395–400.
    1. Sampson M.J., Gopaul N., Davies I.R., Hughes D.A., Carrier M.J. Plasma F2 isoprostanes: direct evidence of increased free radical damage during acute hyperglycemia in type 2 diabetes. Diabetes Care. 2002;25:537–541.
    1. Tang E.H., Vanhoutte P.M. Gene expression changes of prostanoid synthases in endothelial cells and prostanoid receptors in vascular smooth muscle cells caused by aging and hypertension. Physiol Genomics. 2008;32:409–418.
    1. Stowe R.P., Peek M.K., Cutchin M.P., Goodwin J.S. Plasma cytokine levels in a population-based study: relation to age and ethnicity. J Gerontol A Biol Sci Med Sci. 2010;65:429–433.
    1. Valiathan R., Ashman M., Asthana D. Effects of ageing on the immune system: infants to elderly. Scand J Immunol. 2016;83:255–266.
    1. Pinti M., Cevenini E., Nasi M., De Biasi S., Salvioli S., Monti D. Circulating mitochondrial DNA increases with age and is a familiar trait: implications for “inflamm-aging”. Eur J Immunol. 2014;44:1552–1562.
    1. FitzGerald G.A. Bringing PGE2 in from the cold. Science. 2015;348:1208–1209.
    1. Zhang Y., Desai A., Yang S.Y., Bae K.B., Antczak M.I., Fink S.P. Inhibition of the prostaglandin-degrading enzyme 15-PGDH potentiates tissue regeneration. Science. 2015;348:1223.
    1. Gilroy D.W., Newson J., Sawmynaden P., Willoughby D.A., Croxtall J.D. A novel role for phospholipase A2 isoforms in the checkpoint control of acute inflammation. FASEB J. 2004;18:489–498.
    1. Cominelli F., Nast C.C., Llerena R., Dinarello C.A., Zipser R.D. Interleukin 1 suppresses inflammation in rabbit colitis. Mediation by endogenous prostaglandins. J Clin Invest. 1990;85:582–586.
    1. Schwab J.H., Anderle S.K., Brown R.R., Dalldorf F.G., Thompson R.C. Pro- and antiinflammatory roles of interleukin-1 in recurrence of bacterial cell wall induced arthritis in rats. Infect Immun. 1991;59:4436–4442.
    1. Ramos F.J., Kaeberlein M. A healthy diet for stem cells. Nature. 2012;486:477–478.
    1. Zhang G., Li J., Purkayastha S., Tang Y., Zhang H., Yin Y. Hypothalamic programming of systemic ageing involving IKK-β, NF-kB and GnRH. Nature. 2013;497:211–218.
    1. Lucanic M., Held J.M., Vantipalli M.C., Klang I.M., Graham J.B., Gibson B.W. N-acylethanolamine signalling mediates the effect of diet on lifespan in Caenorhabditis elegans. Nature. 2011;473:226–231.
    1. Villeda S.A., Luo J., Kira I., Mosher K.I. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477:90–96.
    1. Ransohoff R.M. Blood ties. Nature. 2011;477:41–42.
    1. Kaiser J. ‘Rejuvenation Factor’ in blood turns back the clock in old mice. Science. 2014;344:570–571.
    1. Das U.N. Ageing, telomere, stem cells biology and inflammation and their relationship to polyunsaturated fatty acids. Agro FOOD Ind Hi Tech. 2015;26:38–41.
    1. Das U.N. Essential fatty acids and their metabolites as modulators of stem cell biology. Agro FOOD Ind Hi-tech. 2010;21:2–3.
    1. Das U.N. Influence of polyunsaturated fatty acids and their metabolites on stem cell Biology. Nutrition. 2011;27:21–25.
    1. Das U.N. N-3 fatty acids, γ-linolenic acid, and antioxidants in sepsis. Crit Care. 2013;17:312.
    1. Faghiri Z., Bazan N.G. PI3K/Akt and mTOR/p70S6K pathways mediate neuroprotectin D1-induced retinal pigment epithelial cell survival during oxidative stress-induced apoptosis. Exp Eye Res. 2010;90:718–725.
    1. Soumya S.J., Binu S., Helen A., Reddanna P., Sudhakaran P.R. 15(S)-HETE-induced angiogenesis in adipose tissue is mediated through activation of PI3K/Akt/mTOR signaling pathway. Biochem Cell Biol. 2013;91:498–505.
    1. Neeli I., Yellaturu C.R., Rao G.N. Arachidonic acid activation of translation initiation signaling in vascular smooth muscle cells. Biochem Biophys Res Commun. 2003;309:755–761.
    1. Lee H.H., Kim H., Lee J.W., Kim Y.S., Yang H.Y., Chang H.K. Maternal swimming during pregnancy enhances short-term memory and neurogenesis in the hippocampus of rat pups. Brain Dev. 2006;28:147–154.
    1. Gangemi S., Luciotti G., D'Urbano E., Mallamace A., Santoro D., Bellinghieri G. Physical exercise increases urinary excretion of lipoxin A4 and related compounds. J Appl Physiol. 2003;94:2237–2240.
    1. Gangemi S., Pescara L., D'Urbano E., Basile G., Nicita-Mauro V., Davì G. Aging is characterized by a profound reduction in anti-inflammatory lipoxin A4 levels. Exp Gerontol. 2005;40:612–614.
    1. Naveen K.V.G., Naidu V.G.M., Das U.N. Arachidonic acid and lipoxin A4 attenuate alloxan-induced cytotoxicity to RIN5F cells in vitro and type 1 diabetes mellitus in vivo. BioFactors. 2017;43:251–271.
    1. Naveen K.V.G., Naidu V.G.M., Das U.N. Arachidonic acid and lipoxin A4 attenuate streptozotocin-induced cytotoxicity to RIN5F cells in vitro and type 1 and type 2 diabetes mellitus in vivo. Nutrition. 2017;35:61–80.
    1. Yong Q.C., Hu L.F., Wang S., Huang D., Bian J.S. Hydrogen sulfide interacts with nitric oxide in the heart: possible involvement of nitroxyl. Cardiovasc Res. 2010;88:482–491.
    1. Pong W.W., Eldred W.D. Interactions of the gaseous neuromodulators nitric oxide, carbon monoxide, and hydrogen sulfide in the salamander retina. J Neurosci Res. 2009;87:2356–2364.
    1. Chiang N., Shinohara M., Dalli J., Mirakaj V., Kibi M., Choi A.M.K. Inhaled carbon monoxide accelerates resolution of inflammation via unique proresolving mediator–heme oxygenase-1 circuits. J Immunol. 2013;190:6378–6388.
    1. Das U.N. Essential fatty acid metabolism in patients with essential hypertension, diabetes mellitus and coronary heart disease. Prostagl Leukot Essen Fatty Acids. 1995;52:387–391.
    1. Das U.N. Cross talk among leukocytes, platelets, and endothelial cells and its relevance to atherosclerosis and coronary heart disease. Current Nutr Food Sci. 2009;5:75–93.
    1. Birnbaum Y., Ye Y., Lin Y., Freeberg S.Y., Nishi S.P., Martinez J.D. Augmentation of myocardial production of 15-epi-lipoxin-a4 by pioglitazone and atorvastatin in the rat. Circulation. 2006;114:929–935.
    1. Merched A.J., Ko K., Gotlinger K.H., Serhan C.N., Chan L. Atherosclerosis: evidence for impairment of resolution of vascular inflammation governed by specific lipid mediators. FASEB J. 2008;22:3595–3606.
    1. Das U.N., Devi G.R., Rao K.P., Rao M.S. Benzo (a) pyrene and gamma-radiation-induced genetic damage in mice can be prevented by gamma-linolenic acid but not by arachidonic acid. Nutr Res. 1985;5:101.
    1. Das U.N., Devi G.R., Rao K.P., Rao M.S. Prostaglandins and their precursors can modify genetic damage induced by benzo (a,) pyrene and gamma-radiation. Prostaglandins. 1985;29:911–916.
    1. Das U.N., Devi G.R., Rao K.P., Rao M.S. Benzo (a) pyrene-induced genetic damage in mice can be prevented by evening primrose oil. IRCS Med Sci. 1985;13:316.
    1. Das U.N., Rao K.P. Effect of γ-linolenic acid and prostaglandins E1 on gamma-radiation and chemical-induced genetic damage to the bone marrow cells of mice. Prostagl Leukot Essen Fatty Acids. 2006;74:165–173.
    1. Shivani P., Rao K.P., Chaudhury J.R., Ahmed J., Rao B.R., Kanjilal S. Effect of polyunsaturated fatty acids on diphenyl hydantoin-induced genetic damage in vitro and in vivo. Prostagl Leukot Essen Fatty Acids. 2009;80:43–50.
    1. Franceschi C. Inflammaging as a major characteristic of old people: can it be prevented or cured? Nutr Rev. 2007;65(12 Pt 2):S173–S176.
    1. Laganiere S., Yu B.P. Modulation of membrane phospholipid fatty acid composition by age and food restriction. Gerontology. 1993;39:7–18.
    1. Kaviarasan K., Mohanlal J., Mohammad Mulla M.A., Shanmugam S., Sharma T., Das U.N. Low blood and vitreal BDNF, LXA4 and altered Th1/Th2 cytokine balance as potential risk factors for diabetic retinopathy. Metabolism. 2015;64:958–966.
    1. Merry B.J. Molecular mechanisms linking calorie restriction and longevity. Int J Biochem Cell Biol. 2002;34:1340–1354.
    1. Ford J.H. Saturated fatty acid metabolism is key link between cell division, cancer, and senescence in cellular and whole organism aging. Age (Dordr) 2010;32:231–237.
    1. Hine C., Harputlugil E., Zhang Y. Endogenous hydrogen sulfide production is essential for dietary restriction benefits. Cell. 2015;160:1–13.
    1. Tateishi N., Kakutani S., Kawashima H., Shibata H., Morita I. Lipids Health Dis. 2014;13:30.
    1. Tateishi N., Kaneda Y., Kakutani S., Kawashima H., Shibata H., Morita I. Dietary supplementation with arachidonic acid increases arachidonic acid content in paw, but does not affect arthritis severity or prostaglandin E2content in rat adjuvant-induced arthritis model. Lipids Health Dis. 2015;14:3.
    1. Kakutani S., Ishikura Y., Tateishi N., Horikawa C., Tokuda H., Kontani M. Supplementation of arachidonic acid-enriched oil increases arachidonic acid contents in plasma phospholipids, but does not increase their metabolites and clinical parameters in Japanese healthy elderly individuals: a randomized controlled study. Lipids Health Dis. 2011;10:241.
    1. Demetz E., Schroll A., Auer K., Heim C., Patsch J.R., Eller P. The arachidonic acid metabolome serves as a conserved regulator of cholesterol metabolism. Cell Metab. 2014;20:787–798.
    1. Fan Y.-Y., Monk J.M., Hou T.Y., Callway E., Vincent L., Weeks B. Characterization of an arachidonic acid-deficient (Fads1 knockout) mouse model. J Lipid Res. 2012;53:1287–1295.
    1. O’Rourke E.J., Kuballa P., Xavier R., Ruvkun G. V-6 Polyunsaturated fatty acids extend life span through the activation of autophagy. Genes Dev. 2013;27:429–440.
    1. Raisz L.G. Pathogenesis of osteoporosis: concepts, conflicts, and prospects. J Clin Invest. 2005;115:3318–3325.
    1. Waters D.J., Caywood D.D., Trachte G.J., Turner R.T., Hodgson S.F. Immobilization increases bone prostaglandin E. Effect of acetylsalicylic acid on disuse osteoporosis studied in dogs. Acta Orthop Scand. 1991;62:238–243.
    1. Sugiyama T. Involvement of interleukin-6 and prostaglandin E2 in periarticular osteoporosis of postmenopausal women with rheumatoid arthritis. J Bone Miner Metab. 2001;19:89–96.
    1. Horiuchi T., Yoshida T., Koshihara Y., Sakamoto H., Kanai H., Yamamoto S. The increase of parathyroid hormone-related peptide and cytokine levels in synovial fluid of elderly rheumatoid arthritis and osteoarthritis. Endocr J. 1999;46:643–649.
    1. Das U.N. Oestrogen, statins and essential fatty acids: similarity in their actions and benefits-is there a common link? Nutrition. 2002;18:178–188.
    1. Naik A.A., Xie C., Zuscik M.J., Kingsley P., Schwarz E.M., Awad H. Reduced COX-2 expression in aged mice is associated with impaired fracture healing. J Bone Miner Res. 2009;24:251–264.
    1. Xie C., Liang B., Xue M., Lin A.S., Loiselle A., Schwarz E.M. Rescue of impaired fracture healing in COX-2−/− mice via activation of prostaglandin E2 receptor subtype 4. Am J Pathol. 2009;175:772–785.
    1. Chang K., Chang W.H. Pulsed electromagnetic fields prevent osteoporosis in an ovariectomized female rat model: a prostaglandin E2-associated process. Bioelectromagnetics. 2003;24:189–198.
    1. Murakami K., Kobayashi Y., Uehara S., Suzuki T., Koide M., Yamashita T. A Jak1/2 inhibitor, baricitinib, inhibits osteoclastogenesis by suppressing RANKL expression in osteoblasts in vitro. PLoS ONE. 2017;12:e0181126.
    1. Standley R.A., Liu S.Z., Jemiolo B., Trappe S.W., Trappe T.A. Prostaglandin E2 induces transcription of skeletal muscle mass regulators interleukin-6 and muscle RING finger-1 in humans. Prostagl Leukot Essent Fatty Acids. 2013;88:361–364.
    1. Liu S.Z., Jemiolo B., Lavin K.M., Lester B.E., Trappe S.W., Trappe T.A. Prostaglandin E2/cyclooxygenase pathway in human skeletal muscle: influence of muscle fiber type and age. J Appl Physiol. 2016;120:546–551.
    1. Guo Y., Dong S.S., Chen X.F., Jing Y.A., Yang M., Yan H. Integrating epigenomic elements and GWASs identifies BDNF gene affecting bone mineral density and osteoporotic fracture risk. Sci Rep. 2016 Jul;28(6):30558.
    1. Liu C., Guan H., Cai C., Li F., Xiao J. Lipoxin A4 suppresses osteoclastogenesis in RAW264.7 cells and prevents ovariectomy-induced bone loss. Exp Cell Res. 2017;352:293–303.
    1. Pennisi P., D'Alcamo M.A., Leonetti C., Clementi A., Cutuli V.M., Riccobene S. Supplementation of L-arginine prevents glucocorticoid-induced reduction of bone growth and bone turnover abnormalities in a growing rat model. J Bone Miner Metab. 2005;23:134–139.
    1. van't Hof R.J., Ralston S.H. Nitric oxide and bone. Immunology. 2001;103:255–261.
    1. Wang Z., Bian L., Mo C., Kukula M., Schug K.A., Brotto M. Targeted quantification of lipid mediators in skeletal muscles using restricted access media-based trap-and-elute liquid chromatography-mass spectrometry. Anal Chim Acta. 2017 Sep;1(984):151–161.
    1. Thakare R., Chhonker Y.S., Gautam N., Nelson A., Casaburi R., Criner G. Simultaneous LC-MS/MS analysis of eicosanoids and related metabolites in human serum, sputum, and BALF. Biomed Chromatogr. 2017 Oct 4

Source: PubMed

3
Sottoscrivi