03894nas a2200649 4500000000100000008004100001653001100042653001100053653000900064653000900073653001600082653001700098653003800115653002500153653006100178653003700239653001200276653004900288653002700337653002500364653004600389653004900435653007400484653002100558653002500579653004200604653002400646653005800670653000900728653002300737653005400760653003400814653002600848653007400874653003800948653005000986653004001036100001601076700001601092700001101108700001601119700001501135700001201150700001801162700001101180700001301191700001201204700001601216700001401232700002301246245010201269250001501371300001101386490000801397520178801405020005103193 2014 d10aFemale10aHumans10aAged10aMale10aMiddle Aged10aTime Factors10aADAM Proteins/genetics/metabolism10aAdministration, Oral10aAmyloid Precursor Protein Secretases/genetics/metabolism10aAmyloid beta-Peptides/metabolism10aAnimals10aBrain/drug effects/pathology/physiopathology10aCognition/drug effects10aCytokines/metabolism10aDementia/blood/ pathology/physiopathology10aGliosis/metabolism/pathology/physiopathology10aGlycosylation End Products, Advanced/administration & dosage/ adverse10aeffects/toxicity10aInsulin/pharmacology10aMembrane Proteins/genetics/metabolism10aMemory/drug effects10aMetabolic Syndrome X/blood/ pathology/physiopathology10aMice10aMice, Inbred C57BL10aNicotinamide Phosphoribosyltransferase/metabolism10aOxidative Stress/drug effects10aPPAR gamma/metabolism10aPyruvaldehyde/administration & dosage/ adverse effects/blood/toxicity10aReceptors, Immunologic/metabolism10aSirtuin 1/antagonists & inhibitors/metabolism10aTranscription, Genetic/drug effects1 aVlassara H.1 aUribarri J.1 aCai W.1 aGrosjean F.1 aStriker G.1 aChen X.1 aWoodward Mark1 aZhu L.1 aSwamy S.1 aZhao Z.1 aSimonaro C.1 aKuchel G.1 aSchnaider-Beeri M.00aOral glycotoxins are a modifiable cause of dementia and the metabolic syndrome in mice and humans a2014/02/26 a4940-50 v1113 a

Age-associated dementia and Alzheimer's disease (AD) are currently epidemic. Neither their cause nor connection to the metabolic syndrome (MS) is clear. Suppression of deacetylase survival factor sirtuin 1 (SIRT1), a key host defense, is a central feature of AD. Age-related MS and diabetes are also causally associated with suppressed SIRT1 partly due to oxidant glycotoxins [advanced glycation end products (AGEs)]. Changes in the modern diet include excessive nutrient-bound AGEs, such as neurotoxic methyl-glyoxal derivatives (MG). To determine whether dietary AGEs promote AD, we evaluated WT mice pair-fed three diets throughout life: low-AGE (MG(-)), MG-supplemented low-AGE (MG(+)), and regular (Reg) chow. Older MG(+)-fed mice, similar to old Reg controls, developed MS, increased brain amyloid-beta42, deposits of AGEs, gliosis, and cognitive deficits, accompanied by suppressed SIRT1, nicotinamide phosphoribosyltransferase, AGE receptor 1, and PPARgamma. These changes were not due to aging or caloric intake, as neither these changes nor the MS were present in age-matched, pair-fed MG(-) mice. The mouse data were enhanced by significant temporal correlations between high circulating AGEs and impaired cognition, as well as insulin sensitivity in older humans, in whom dietary and serum MG levels strongly and inversely associated with SIRT1 gene expression. The data identify a specific AGE (MG) as a modifiable risk factor for AD and MS, possibly acting via suppressed SIRT1 and other host defenses, to promote chronic oxidant stress and inflammation. Because SIRT1 deficiency in humans is both preventable and reversible by AGE reduction, a therapeutic strategy that includes AGE reduction may offer a new strategy to combat the epidemics of AD and MS.

 a1091-6490 (Electronic)
0027-8424 (Linking)