Free Shipping on Orders over $300 (Excludes , , )
Bio-Age Elevate References
AMPK Charge + https://www.quicksilverscientific.com/ampkchargereferences/
[1] Herzig S and Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018; 19(2): 121-135.
[2] Hardie DG, et al. Targeting an energy sensor to treat diabetes. Science. 2017; 357 (6350): 455-456.
[3] Foretz M and Viollet B. Activation of AMPK for a break in hepatic lipid accumulation and circulating cholesterol. EBio Medicine. 2018; 31: 15-16.
[4] Tamargo-Gomez I, et al. AMPK: Regulation of metabolic dynamics in the context of autophagy. Int J Mol Sci. 2018; 19(12): 3812.
[5] Furman D, et al. Chronic inflammation in the etiology of disease across the life span. Nature Medicine. 2019; 25: 1822-1832.
[6] Jeon SM, et al. Regulation and function of AMPK in physiology and diseases. Exp Mol Med. 2016; 48: e245.
[7] Shirwany NA and Zou MH. AMPK in cardiovascular health and disease. Acta Pharmacol Sin. 2010; 31(9): 1075-1084.
[8] Ruderman NB, et al. AMPK, insulin resistance, and the metabolic syndrome. J Clin Investig. 2013.
[9] Seabright AP, et al. AMPK activation induces mitophagy and promotes mitochondrial fission while activating TBK1 in a PINK1-Parkin independent manner. FASEB J. 2020; 34(5): 6284-6301.
[10] Ruderman NB, et al. AMPK and SIRT1: a long-standing partnership? Am J Physiol Endocrinol Metab. 2010; 298(4): E751-E760.
[11] Pan H and Finkel T. Key proteins and pathways that regulate lifespan. J Biol Chem. 2017; 292(16): 6452-6460.
[12] Connell NJ, et al. NAD+ metabolism as a target for metabolic health: have we found the silver bullet? Diabetologia. 2019; 62(6): 888-899.
[13] Anton SD, et al. Flipping the metabolic switch: Understanding and applying health benefits of fasting. Obesity (Silver Spring).
[14] Fan W and Evans RM. Exercise mimetics: Impact on health and performance. Cell Metab. 2017; 25(2): 242-247.
[15] Dolinksy VW, et al. Improvements in skeletal muscle strength and cardiac function induced by resveratrol during exercise training contribute to enhanced exercise performance in rats. J Physiol. 2012; 590(Pt 11): 2783-2799.
[16] Konrad M and Nieman DC. Evaluation of quercetin as a countermeasure to exercise-induced physiological stress. antioxidants in sports nutrition. 2015.
[17] Kaeberlein M, et al. Substrate-specific activation of sirtuins by resveratrol. J Biol Chem. 2005; 280(17): 17038-17045.
[18] Park D, et al. Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Sci Rep. 2016; 6: 21772.
[19] Grant R. Resveratrol increases intracellular NAD+ levels through the up-regulation of the NAD+ synthetic enzyme nicotinamide mononucleotide adenylyltransferase. Nature Precedings. 2010.
[20] Csiszar A, et al. Resveratrol induces mitochondrial biogenesis in endothelial cells. Am J Physiol Heart Circ Physiol.
[21] Sun H, et al. Berberine ameliorates blockade of autophagic flux in the liver by regulating cholesterol metabolism and inhibiting COX2-prostaglandin synthesis. Cell Death & Dis. 2018; 9: 824.
[22] Lee YS, et al. Berberine, a natural plant product, activates AMP-activated protein kinase with beneficial metabolic effects in diabetic and insulin-resistant states. Pharmacol & Ther. 2006; 55(8).
[23] Gomes AP, et al. Berberine protects against high fat diet-induced dysfunction in muscle mitochondria by inducing SIRT1-dependent mitochondrial biogenesis. Biochim Biophys Acta. 2012; 1822(2): 185-195.
[24] Rayamajhi N, et al. Quercetin induces mitochondrial biogenesis through activation of HO-1 in HepG2 Cells. Oxid Med Cell Longev. 2013; 2013: 154279.
[25] Li Y, et al. Quercetin, inflammation and immunity. Nutrients. 2016; 8(3): 167.
[26] Kim SG, et al. Quercetin-induced AMP-activated protein kinase activation attenuates vasoconstriction through LKB1-AMPK signaling pathway. J Med Food. 2018; 21(2): 146-153.
[27] Lewinska A, et al. AMPK-mediated senolytic and senostatic activity of quercetin surface functionalized Fe3O4 nanoparticles during oxidant-induced senescence in human fibroblasts. Redox Biol. 2020; 28: 101337.
[28] Van Deursen JM. Senolytic therapies for healthy longevity. Science. 2019; 364(6441): 636-637.
[29] Weng Z, et al. Quercetin Is more effective than Cromolyn in blocking human mast cell cytokine release and inhibits contact dermatitis and photosensitivity in humans. PLoS One. 2012; 7(3): e33805.
[30] Jiang K, et al. Silibinin, a natural flavonoid, induces autophagy via ROS-dependent mitochondrial dysfunction and loss of ATP involving BNIP3 in human MCF7 breast cancer cells. Oncol Rep. 2015; 33(6): 2711-2718.
[31] Lovelace ES, et al. Silymarin suppresses cellular inflammation by inducing reparative stress signaling. J Nat Prod. 2015; 78(8): 1990-2000.
[32] Ye Y, et al. 3,3′-Diindolylmethane induces anti-human gastric cancer cells by the miR-30e-ATG5 modulating autophagy. Biochem Pharmacol. 2016; 115: 77-84.
[33] Hornero RA, et al. The impact of dietary components on regulatory T cells and disease. Front Immunol. 2020; 11: 253.
[34] Shen Y, Honma N et al. Cinnamon extract enhances glucose uptake in 3T3-L1 adipocytes and C2C12 myocytes by inducing LKB1-AMPactivated protein kinase signaling. PLoS One. 2014 Feb 14;9(2):e8789
[35] Park KR, Nam D. β-Caryophyllene oxide inhibits growth and induces apoptosis through the suppression of PI3K/AKT/mTOR/S6K1 pathways and ROS-mediated MAPKs activation. Cancer Lett. 2011 Dec 22;312(2):178-88
[36] Mollazadeh H and Hosseinzadeh H. Cinnamon effects on metabolic syndrome: a review based on its mechanisms. Iran J Basic Med Sci. 2016; 19(12): 1258-1270.
NAD + Platinum https://www.quicksilverscientific.com/nadplatinumreferences/
[1] Longo VD et al. Interventions to Slow Aging in Humans: Are We Ready? Aging Cell 14 (4): 497-510.
[2] Fang EF et al. NAD (+) in aging: molecular mechanisms and translational implications. Trends Mol Med. 2017;23(10):899–916
[3] Keller K and Engelhardt M. Strength and muscle mass loss with the aging process. Age and strength loss. Muscles Ligaments Tendons J. 2013; 3(4): 346-350.
[4] Chang AM and Halter JB. Aging and insulin secretion. Am J Physiol Endocrinol Metab. 2003; 284(1): E7-12.
[5] Caito SW and Aschner M. NAD+ Supplementation attenuates methylmercury dopaminergic and mitochondrial toxicity in Caenorhabditis Elegans. Toxicol Sci. 2016; 151(1): 139-149.
[6] Gizem Kivrak E, et al. Effects of electromagnetic fields exposure on the antioxidant defense system. J Microsc Ultrastruct. 2017; 2017; 5(4): 167-176.
[7] Xie N, et al. NAD+ metabolism: pathophysiologic mechanisms and therapeutic potential. Signal Transduct Target Ther. 2020; 5: 227.
[8] Hong W, et al. Nicotinamide mononucleotide: A promising molecule for therapy of diverse diseases by targeting NAD+ metabolism. Front Cell Dev Biol. 2020.
[9] Wu, L et al. The elusive NMN transporter is found. Nat Metab 2019: 1; 8-9
[10] Yamaguchi S and Yoshino J. Adipose tissue NAD+ biology in obesity and insulin resistance: From mechanism to therapy. Bioessays. 2017; 39(5): 10.1002/bies.201600227.
[11] Guarente L, Franklin H. Epstein lecture: sirtuins, aging, and medicine. N Engl J Med. (2011) 364:2235–44.
[12] Kane AE, Sinclair DA. Sirtuins and NAD+ in the development and Treatment of Metabolic and Cardiovascular Diseases. Circ Res. 2018; 123:868-885.
[13] Mangerich A, et al. Pleiotropic cellular functions of PARP1 in longevity and aging: Genome maintenance meets inflammation. Oxid Med Cell Longev. 2012; 2012: 321653.
[14] Bonkowski MS and Sinclair D. Slowing aging by design: the rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol. 2016; 17(11): 679-690.
[15] Lewinska A, et al. AMPK-mediated senolytic and senostatic activity of quercetin surface functionalized Fe3O4 nanoparticles during oxidant-induced senescence in human fibroblasts. Redox Biol. 2020; 28: 101337.
[16] Jesko H, et al. Sirtuins and their roles in brain aging and neurodegenerative disorders. Neurochem Res. 2017; 42(3): 876-890.
[17] Warren JL, et al. Regulation of adaptive immune cells by sirtuins. Front Endocrinol (Lausanne). 2019; 10:466.
[18] Radak Z, et al. The systemic role of SIRT1 in exercise mediated adaptation. Redox Biol. 2020; 35: 101467.
[19] Vargas-Ortiz K, et al. Exercise and sirtuins: A way to mitochondrial health in skeletal muscle. Int J Mol Sci. 2019; 20(11): 2717.
[20] Asher G, et al. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell. 2008; 134(2): 317-328.
[21] Grabowska W, et al. Sirtuins, a promising target in slowing down the ageing process. Biogerontology. 2017; 18(4): 447-476.
[22] Schafer MJ, et al. Exercise prevents diet-induced cellular senescence in adipose tissue. Diabetes. 2016; 65(6): 1606-1615.
[23] Han YM, et al. β-Hydroxybutyrate prevents vascular senescence through hnRNP A1-mediated upregulation of Oct4.Mol Cell. 2018; 71(6): 1064-1078.
[24] Weng Z, et al. Quercetin is more effective than cromolyn in blocking human mast cell cytokine release and inhibits contact dermatitis and photosensitivity in humans. PLoS One. 2012; 7(3): e33805.
[25] Mohar DS and Malik S. The sirtuin system: The holy grail of resveratrol? J Clin Exp Cardiol. 2012; 3(11): 216.
[26] Hustad S, et al. Riboflavin and methylenetetrahydrofolate reductase. Madame Curie Bioscience Database. 2013.
[27] Ahn H, Park JH. Liposomal delivery systems for intestinal lymphatic drug transport.Biomater Res. 2016 Nov 23;20:36 View Full Paper
[28] Alyautdin R et al. Nanoscale drug delivery systems and the blood brain barrier. Int J Nanomedicine. 2014 Feb 7;9:795-811 View Full Paper
Membrane Mend™ https://www.quicksilverscientific.com/membranemendreferences/
[1] Casares D, et al. Membrane lipid composition: Effect on membrane and organelle structure, function and compartmentalization and therapeutic avenues. Int J Mol Sci. 2019; 20(9): 2167.
[2] Leekumjorn S, et al. The role of fatty acid unsaturation in minimizing biophysical changes on the structure and local effects of bilayer membranes. Biochim Biophys Acta. 2009; 1788(7): 1508-1516.
[3] Van Meer G, et al. Membrane lipids: where they are and how they behave. Nat Rev Mol Cell Biol. 2009; 9(2): 112-124.
[4] Zorova LD, et al. Mitochondrial membrane potential. Anal Biochem. 2018; 552: 50-59.
[5] Chew S, et al. Impairment of mitochondrial function by particulate matter: Implications for the brain. Neurochem Int. 2020; 135(104694).
[6] Zulkifli-Cunningham Z, et al. Clinical effects of chemical exposures on mitochondrial function. Toxicology. 2017; 391: 90-99.
[7] Lin JH, et al. Endoplasmic reticulum stress in disease pathogenesis. Annu Rev Pathol. 2008; 3: 399-425.
[8] Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 2010; 140(6): P900-P917.
[9] Kalghatgi S, et al. Bactericidal Antibiotics Induce Mitochondrial Dysfunction and Oxidative Damage in Mammalian Cells. Sci Transl Med. 2013; 5(192): 192ra85.
[10] Santini SJ, et al. Role of Mitochondria in the Oxidative Stress Induced by Electromagnetic Fields: Focus on Reproductive Systems. Oxid Med Cell Longev. 2018; 2018: 5076271.
[11] Zorova LD, et al. Mitochondrial membrane potential. Anal Biochem. 2018; 552: 50-59.
[12] Nicolson GL, et al. Clinical uses of membrane lipid replacement supplements in restoring membrane function and reducing fatigue in chronic diseases and cancer. Discoveries (Craiova). 2016; 4(1): e54.
[13] Na JY, et al. Hepatoprotective effect of phosphatidylcholine against carbon tetrachloride liver damage in mice. Biochem Biophys Res Commun. 2015; 460(2): 308-313.
[14] Maev IV, et al. Effectiveness of phosphatidylcholine in alleviating steatosis in patients with non-alcoholic fatty liver disease and cardiometabolic comorbidities (MANPOWER study). BMJ Open Gastroenterol. 2020; 7: e000341.
[15] Kennelly JP, et al. Intestinal de novo phosphatidylcholine synthesis is required for dietary lipid absorption and metabolic homeostasis. J Lipid Res. 2018; 59(9): 1695-1708.
[16] Schneider H, et al. Lipid-based therapy for ulcerative colitis—Modulation of intestinal mucus membrane phospholipids as a tool to influence inflammation. Int J Mol Sci. 2010; 11(10): 4149-4164.
[17] Chen M, et al. Oral phosphatidylcholine improves intestinal barrier function in drug-induced liver injury in rats. Gastroenterol Res Pract. 2019; Article ID 8723460.
[18] Lichtenberger LM. Role of phospholipids in protection of the GI mucosa. Digestive Dis Sci. 2013; 58: 891-893.
[19] Blusztajn JK, et al. Neuroprotective actions of dietary choline. Nutrients. 2017; 9(8): 815.
[20] Ojo JO, et al. Disruption in brain phospholipid content in a humanized tau transgenic model following repetitive mild traumatic brain injury. Front Neurosci. 2018; [online].
[21] Yu C, et al. HC diet inhibited testosterone synthesis by activating endoplasmic reticulum stress in testicular Leydig cells. J Cell Molec Med. 2019; 23(5): 3140-3150.
[22] Wen G, et al. Endoplasmic reticulum stress inhibits expression of genes involved in thyroid hormone synthesis and their key transcriptional regulators in FRTL-5 thyrocytes. PLoS One. 2017; [online].
[23] Lefort N, et al. Dietary Buglossoides Arvensisoil increases circulating n-3 polyunsaturated fatty acids in a dose-dependent manner and enhances lipopolysaccharide-stimulated whole blood interleukin-10—A randomized placebo-controlled trial. Nutrients. 2017; 9(3): 261.
[24] Lefort N, et al. Consumption of Buglossoides arvensis seed oil is safe and increases tissue long-chain n-3 fatty acid content more than flaxseed oil – results of a phase I randomised clinical trial. J Nutr Sci. 2016; 5: e2.
[25] Sztretye M, et al. Astaxanthin: A potential mitochondrial-targeted antioxidant treatment in diseases and with aging. Oxid Med Cell Longev. 2019; 2019: 3849692.
Ultra Vitamin® https://www.quicksilverscientific.com/ultravitaminreferences/
[1] Chouliaras L, et al. Peripheral DNA methylation, cognitive decline, and brain aging: pilot findings from the Whitehall II imaging study. Epigenomics. 2018; 10(5): 585-595.
[2] Liu G, et al. DNA methylation and the potential role of methyl-containing nutrients in cardiovascular diseases. Oxid Med Cell Longev. 2017; 2017: 1670815.
[3] Ulrich CM, et al. Metabolic, hormonal, and immunological associations with global DNA methylation among postmenopausal women. Epigenetics. 2012; 7(9): 1020-1028.
[4] Samodien E, et al. Diet‐induced DNA methylation within the hypothalamic arcuate nucleus and dysregulated leptin and insulin signaling in the pathophysiology of obesity.Food Sci Nutr. 2019; 7(10): 3131-3145.
[5] Anderson OS, et al. Nutrition and epigenetics: An interplay of dietary methyl donors, one-carbon metabolism, and DNA methylation. J Nutr Biochem. 2012; 23(8): 853-859.
[6] Zeisel S. Choline, other methyl-donors and epigenetics. Nutrients. 2017; 9(5).
[7] Bird JK, et al. Risk of deficiency in multiple concurrent micronutrients in children and adults in the United States. Nutrients. 2017; 9(7): 655.
[8] Depeint F, et al. Mitochondrial function and toxicity: Role of the B vitamin family on mitochondrial energy metabolism. Chem Biol Interact. 2006; 163(1-2): 94-112.
[9] Kennedy DO. B vitamins and the brain: Mechanisms, dose and efficacy—A review. Nutrients. 2016; 8(2): 68.
[10] Pehlivan FE. Vitamin C: An antioxidant agent. IntechOpen. 2016; [online].
[11] Carr AC, Maggini S. Vitamin C and immune function. Nutrients. 2017; 9(11): 1211.
[12] Young JI, et al. Regulation of the epigenome by vitamin C. Annu Rev Nutr. 2015; 35: 545-564.
[13] Lykkesfeldt J, Tveden-Nyborg P. The pharmacokinetics of vitamin C. Nutrients. 2019; 11(10): 2412.
[14] Sato T, et al. Comparison of menaquinone-4 and menaquinone-7 bioavailability in healthy women. Nutr J. 2012; 11: 93.
[15] Akbari S, et al. Vitamin K and bone metabolism: A review of the latest evidence in preclinical studies. Biomed Res Int. 2018; 2018: 4629383.
[16] Simes DC, et al. Vitamin K as a powerful micronutrient in aging and age-related diseases: Pros and cons from clinical studies. Int J Mol Sci. 2019; 20(17): 4150.
[17] Peh HY, et al. Vitamin E therapy beyond cancer: Tocopherol versus tocotrienol. Pharmacol Ther. 2016; 162: 152-169.
[18] Sen CK, et al. Tocotrienols: Vitamin E beyond tocopherols. Life Sci. 2006; 78(18): 2088-2098.
[19] Bell TD, et al. The biology and pathology of vitamin D control in bone. J Cell Biochem. 2010; 111(1): 7-13.
[20] Kheiri B, et al. Vitamin D deficiency and risk of cardiovascular diseases: a narrative review. Clin Hypertens. 2018; 24: 9.
[21] Khammissa RAG, et al. Vitamin D deficiency as it relates to oral immunity and chronic periodontitis. Int J Dent. 2018; 2018: 7315797.
[22] Shang M, Sun J. Vitamin D/VDR, probiotics, and gastrointestinal diseases. Curr Med Chem. 2017; 24(9): 876-887.
[23] Anjum I, et al. The role of vitamin D in brain health: A mini literature review. Cureus. 2018; 10(7): e2960.
[24] Parva NR, et al. Prevalence of vitamin D deficiency and associated risk factors in the US population (2011-2012). Cureus. 2018; 10(6): e2741.
[25] Tang G. Bioconversion of dietary provitamin A carotenoids to vitamin A in humans. Am J Clin Nutr. 2010; 91(5): 1468S-1473S.
[26] Czarnewski P, et al. Retinoic acid and its role in modulating intestinal innate immunity. Nutrients. 2017; 9(1): 68.
[27] Iyer N, Vaishnava S. Vitamin A at the interface of host–commensal–pathogen interactions. PLoS Pathog. 2019; 15(6): e1007750.
[28] Wolf G. The discovery of the visual function of vitamin A. J Nutr. 2001; 131(6): 1647-1650.
[29] Zasada M, Budzisz E. Retinoids: active molecules influencing skin structure formation in cosmetic and dermatological treatments. Postepy Dermatol Alergol. 2019; 36(4): 392-397.
[30] Long MD, et al. Vitamin D receptor and RXR in the post-genomic era. J Cell Physiol. 2015; 230(4): 758-766.
[31] Van Ballegooijen AJ, et al. The synergistic interplay between vitamins D and K for bone and cardiovascular health: A narrative review. Int J Endocrinol. 2017; 2017: 7454376.
[32] Li H, et al. Lutein suppresses oxidative stress and inflammation by Nrf2 activation in an osteoporosis rat model. Med Sci Monit. 2018; 24: 5071-5075.
[33] Zou X, et al. Zeaxanthin induces Nrf2-mediated phase II enzymes in protection of cell death. Cell Death and Dis. 2014; 5: e1218.
[34] Lian F, et al. Enzymatic metabolites of lycopene induce Nrf2-mediated expression of phase II detoxifying/antioxidant enzymes in human bronchial epithelial cells. Int J Cancer. 2008; 123(6): 1262-1268.
[35] Stringham JM, et al. Lutein across the lifespan: From childhood cognitive performance to the aging eye and brain. Curr Dev Nutr. 2019; 3(7): nzz066.
[36] Przybylska S. Lycopene – a bioactive carotenoid offering multiple health benefits: a review. Food Sci Technol. 2019; [early view online version].
[37] Alyautdin R, et al. Nanoscale drug delivery systems and the blood-brain barrier. Int J Nanomedicine.2014; 9: 795-811.
[38] Ahn H, et al. Liposomal delivery systems for intestinal lymphatic drug transport. Biomater Res. 2016; 20: 36.
[39] Spector AA, et al. Membrane lipid composition and cellular function. J Lipid Res. 1985; 26(9): 1015-1035.