Quicksilver Scientific

Free Shipping on Orders over $99 (Excludes )

0
0
Subtotal: 0.00

No products in the cart.

Bio-Age Reversal Program References

Dr. Shade’s Liver Sauce®  https://www.quicksilverscientific.com/liversaucereferences/

[1] Grant DM. Detoxification pathways in the liver. J Inherit Metab Dis. 1991;14(4):421-30.

[2] Leslie EM, Deeley RG et al. Multidrug resistance proteins: role of P-glycoprotein, MRP1, MRP2, and BCRP (ABCG2) in tissue defense. Toxicol Appl Pharmacol. 2005 May 1;204(3):216-37

[3] Zamek-Gliszczynski MJ, et al. Integration of hepatic drug transporters and phase II metabolizing enzymes: mechanisms of hepatic excretion of sulfate, glucuronide, and GSH metabolites. Eur J Pharm Sci. 2006 Apr;27(5):447-86

[4] Watanabe M, Houten SM et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 2006 Jan 26;439(7075):484-9.

[5] Trauner M, Boyer JL. Bile salt transporters: molecular characterization, function, and regulation. Physiol Rev. 2003 Apr;83(2):633-71

[6] Hellström PM, Nilsson I et al. Role of bile in regulation of gut motility. J Intern Med. 1995 Apr;237(4):395-402.

[7] Nguyen T, Nioi P et al. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem. 2009 May 15;284(20):13291-5

[8] Perricone C, et al. Glutathione: a key player in autoimmunity. Autoimmun Rev. 2009 Jul;8(8):697-701

[9] Lefebvre P, Cariou B et al. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev. 2009 Jan;89(1):147-91

[10] Gadaleta RM, Oldenburg B et al. Activation of bile salt nuclear receptor FXR is repressed by pro-inflammatory cytokines activating NF-κB signaling in the intestine. Biochim Biophys Acta. 2011 Aug;1812(8):851-8

[11] Sharma R, Long A et al. Advances in Bile Acid Medicinal Chemistry. Curr Med Chem. 2011;18(26):4029-52

[12] Islam KB, Fukiya S et al. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats.Gastroenterology. 2011 Nov;141(5):1773-81.

[13] Fernandesa CF, Shahani KM et al. Effect of Nutrient Media and Bile Salts on Growth and Antimicrobial Activity of Lactobacillus acidophilus. Journal of Dairy Science Volume 71, Issue 12, December 1988, Pages 3222-32299.

[14] D’Aldebert E, Biyeyeme Bi Mve MJ et al. Bile salts control the antimicrobial peptide cathelicidin through nuclear receptors in the human biliary epithelium. Gastroenterology. 2009 Apr;136(4):1435-43.

[15] Trauner M, Boyer JL. Bile salt transporters: molecular characterization, function, and regulation. Physiol Rev. 2003 Apr;83(2):633-71.

[16] Kalitsky-Szirtes J, Shayeganpour A et al. Suppression of drug-metabolizing enzymes and efflux transporters in the intestine of endotoxin-treated rats. Drug Metab Dispos. 2004 Jan;32(1):20-7.

[17] Awad WA, Hess C et al. Enteric Pathogens and Their Toxin-Induced Disruption of the Intestinal Barrier through Alteration of Tight Junctions in Chickens. Toxins (Basel). 2017 Feb 10;9(2).

[18] Islam KB, Fukiya  et al. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology. 2011 Nov;141(5):1773-81.

[19] Hellström PM, Nilsson I et al. Role of bile in regulation of gut motility. J Intern Med. 1995 Apr;237(4):395-402.

[20] Ahmad R, Sorrell MF et al. Gut permeability and mucosal inflammation: bad, good or context dependent. Mucosal Immunol. 2017 Mar;10(2):307-317

[21] Whiting JF, Green RM, et al. Tumor necrosis factor-alpha decreases hepatocyte bile salt uptake and mediates endotoxin-induced cholestasis. Hepatology. 1995 Oct;22(4 Pt 1):1273-8.

[22] Yu Y, Hao G et al. Berberine induces GLP-1 secretion through activation of bitter taste receptor pathways. Biochem Pharmacol. 2015 Sep 15;97(2):173-7.

[23] Shaik FA, Singh N et al. Bitter taste receptors: Extraoral roles in pathophysiology. Int J Biochem Cell Biol. 2016 Aug;77(Pt B):197-204.

[24] Lee RJ, Cohen NA. The emerging role of the bitter taste receptor T2R38 in upper respiratory infection and chronic rhinosinusitis. Am J Rhinol Allergy. 2013 Jul-Aug;27(4):283-6.

[25] Gaida MM, Dapunt U et al. Sensing developing biofilms: the bitter receptor T2R38 on myeloid cells. Pathog Dis. 2016 Apr;74(3).

[26] Lian LH, Wu YL, Song SZ, et al. Gentiana manshurica Kitagawa reverses acute alcohol-induced liver steatosis through blocking sterol regulatory element-binding protein-1 maturation. J Agric Food Chem. 2010 Dec 22;58(24):13013-9

[27] Wang AY, Lian LH, Jiang YZ, et al. Gentiana manshurica Kitagawa prevents acetaminophen-induced acute hepatic injury in mice via inhibiting JNK/ERK MAPK pathway. World J Gastroenterol. 2010 Jan 21;16(3):384-91.

[28] González-Castejón M, Visioli F. Diverse biological activities of dandelion. Nutr Rev. 2012 Sep;70(9):534-47

[29] Schütz K, Carle R, Schieber A. Taraxacum–a review on its phytochemical and pharmacological profile. J Ethnopharmacol. 2006 Oct 11;107(3):313-23.

[30] You Y, Yoo S, Yoon HG, et al. In vitro and in vivo hepatoprotective effects of the aqueous extract from Taraxacum officinale (dandelion) root against alcohol-induced oxidative stress. Food Chem Toxicol. 2010 Jun;48(6):1632-7.

[31] Melzig MF. [Goldenrod–a classical exponent in the urological phytotherapy]. Wien Med Wochenschr. 2004 Nov;154(21-22):523-7. View Abstract

[32] Apáti P, et al. In-vitro effect of flavonoids from Solidago canadensis extract on GSH S-transferase. J Pharm Pharmacol. 2006 Feb;58(2):251-6. View Abstract

[33] Dolara P, Corte B, Ghelardini C, et al. Local anaesthetic, antibacterial and antifungal properties of sesquiterpenes from myrrh. Planta Med. 2000 May;66(4):356-8.

[34] Sheir Z, Nasr AA, Massoud A, et al. A safe, effective, herbal antischistosomal therapy derived from myrrh. Am J Trop Med Hyg. 2001 Dec;65(6):700-4.

[35] Shen T, Li GH, Wang XN, Lou HX. The genus Commiphora: a review of its traditional uses, phytochemistry and pharmacology. J Ethnopharmacol. 2012 Jul 13;142(2):319-30.

[36] Shah R, Gulati V, Palombo EA. Pharmacological properties of guggulsterones, the major active components of gum guggul. Phytotherapy Research. 2012 Nov;26(11):1594-605.

[37] Cui J, et al. Guggulsterone is a farnesoid X receptor antagonist in coactivator association assays but acts to enhance transcription of bile salt export pump J Biol Chem. 2003 Mar 21;278(12):10214-20

[38] Crocenzi FA, Roma MG. Silymarin as a new hepatoprotective agent in experimental cholestasis: new possibilities for an ancient medication. Curr Med Chem. 2006;13(9):1055-74.

[39] Abenavoli L, Capasso R et al. Milk thistle in liver diseases: past, present, future. Phytother Res. 2010 Oct;24(10):1423-32.

[40] Gu M, Zhao P et al. Silymarin Ameliorates Metabolic Dysfunction Associated with Diet-Induced Obesity via Activation of Farnesyl X Receptor. Front Pharmacol. 2016;7:345

[41] Crocenzi FA, Sánchez et al. Beneficial effects of silymarin on estrogen-induced cholestasis in the rat: a study in vivo and in isolated hepatocyte couplets. Hepatology. 2001 Aug;34(2):329-39.

[42] Suh JH, et al. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid Proc Natl Acad Sci U S A. 2004 Mar 9;101(10):3381-6

[43] Arredondo F, Echeverry  et al. After cellular internalization, quercetin causes Nrf2 nuclear translocation, increases glutathione levels, and prevents neuronal death against an oxidative insult. Free Radic Biol Med. 2010 Sep 1;49(5):738-47

[44] Saw CL, Cintrón M, et al. Pharmacodynamics of dietary phytochemical indoles I3C and DIM: Induction of Nrf2-mediated phase II drug metabolizing and antioxidant genes and synergism with isothiocyanates Biopharm Drug Dispos. 2011 Jul;32(5):289-300. Biopharm Drug Dispos. 2011 Jul;32(5):289-300

[45] Micek J, Jurkova T et al. Quercetin and Its Anti-Allergic Immune Response. Molecules. 2016 May 12;21(5)

[46] Seelinger G, Merfort I. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med. 2008;74(14):1667-77

[47] Spector AA, Yorek MA. Membrane lipid composition and cellular function. J Lipid Res. 1985 Sep;26(9):1015-35

[48] Chang CY, Ke DS. Essential fatty acids and human brain. Acta Neurol Taiwan. 2009 Dec;18(4):231-41

[49] Porter CJ. Drug delivery to the lymphatic system. Crit Rev Ther Drug Carrier Syst. 1997;14(4):333-93

[50] Ahn H, Park JH. Liposomal delivery systems for intestinal lymphatic drug transport. Biomater Res. 2016 Nov 23;20:36

[51] Alyautdin R, Khlain I et al. Nanoscale drug delivery systems and the blood brain barrier.  Int J Nanomedicine. 2014 Feb 7;9:795-81

Ultra Binder® Stick Packs Universal Toxin Binder https://www.quicksilverscientific.com/ultrabinderreferences/

[1] Frolis VV, Nikolav VG et al. Effect of enteroabsorption on animal lifespan. Biomat. Art. 1989. 17(3): 341-351.

[2] Su W, Ding X. Methods of endotoxin detection. J Lab Autom. 2015 Aug;20(4):354-64

[3] Aitken AE, Richardson TA et al. Regulation of drug-metabolizing enzymes and transporters in inflammation. Annu Rev Pharmacol Toxicol 2006: 46:123–149

[4] Bolder U, Ton-Nu HT et al. Hepatocyte transport of bile acids and organic anions in endotoxemic rats: impaired uptake and secretion. Gastroenterology 1997: 112:214–225.

[5] Cherrington NJ, Slitt AL et al. Lipopolysaccharide-mediated regulation of hepatic transporter mRNA levels in rats. Drug MetabDispos2004: 32:734–741

[6] Tang W, Yi C et al. Endotoxin downregulates hepatic expression of P-glycoprotein and MRP2 in 2-acetylaminofluorene-treated rats. 2000 Mol Cell Biol Res Commun 4:90–97

[7] Maklad A, Emara A et al. Pediatric poisoning in Egypt. Journal of Applied Pharmaceutical Science, 2012 2 (2): 1-6.

[8] Khalid J, Zailaey A. Medical and environmental applications of activated charcoal: review article. European Scientific Journal January 2015 (11): 3: 50-56

[9] Neuvonen J, Olkkola KT. Oral activated charcoal in the treatment of intoxications: role of single and repeated doses. Med Toxicol, 1988: 3; 33-58

[10] Karnib M, Kabbani A et al. Heavy metals removal using activated carbon, silica and silica activated carbon composite. Energy Procedia 2014: 50, 113 – 120.

[11] Du XN, Niu Z et al. Effect of activated charcoal on endotoxin adsorption. Part I. An in vitro study. Biomater Artif Cells Artif Organs. 1987;15(1):229-35

[12] Dalefield R. Emergency care and stabilization of the poisoned patient. In: Veterinary Toxicology for Australia and New Zealand, 2017: 19-32

[13] Rodriguez-Reinoso. Activated carbon and adsorption. in Encyclopedia of Materials: Science and Technology, 2001: 22-24

[14] Krasopoulos JC, De Bari VA et al The adsorption of bile salts on lipids. 1980 May;15(5):365-70

[15] Neuvonen PJ, Kuusisto P. Activated charcoal in the treatment of hypercholesterolaemia: dose-response relationships and comparison with cholestyramine Eur J Clin Pharmacol. 1989;37(3):225-30

[16] Musso CG, Michelangelo H et al. Combination of oral activated charcoal plus low protein diet as a new alternative for handling in the old end-stage renal disease patients Saudi J Kidney Dis Transpl. 2010 Jan;21(1):102-4

[17] Koide, S. S. Chitin-chitosan: properties, benefits and risks. Nutrition Research 1998;8(6):1091-1101

[18] Macchi G. A new approach to the treatment of obesity: chitosan’s effects on body weight reduction and plasma cholesterol levels. Acta Toxicol Ther 1996;17:303-320

[19] Lütjohann D, Marinova M. Nutrients. Influence of Chitosan Treatment on Surrogate Serum Markers of Cholesterol Metabolism in Obese Subjects. 2018 Jan 11;10(1)

[20] Maezaki Y, Tsuji K et al. Hypocholesterolaemic effect of chitosan in adult males. Biosc Biochem Biotech 1993;57:1439-1444

[21] Shoemaker, RC. (2001) Desperation Medicine. Gateway Press: Baltimore. 2. Shoemaker, RC, Schaller J, Schmidt P. (2005) Mold Warriors: Fighting America’s Hidden Threat. Gateway Press: Baltimore

[22] Karunasena E, Larrañaga MD et al. Building-Associated neurological damage modeled in human cells: a mechanism of neurotoxic effects by exposure to mycotoxins in the indoor environment. Mycopathologia. 2010 Dec;170(6):377-90

[23] Carretero MI. Clay minerals and their beneficial effects upon human health. A review. Appl Clay Sci 2002; 21: 155–63.

[24] Herrera P, Burghardt RC et al. Adsorption of Salmonella enteritidis by cetylpyridinium-exchanged montmorillonite clays. Vet Microbiol 2000; 74: 259–72

[25] Haydel SE, Remenih CM. J Broad-spectrum in vitro antibacterial activities of clay minerals against antibiotic-susceptible and antibiotic-resistant bacterial pathogens Antimicrob Chemother. 2008 Feb;61(2):353-61

[26] Schaumberger S, Ladining A et al. Evaluation of the endotoxin binding efficiency of clay minerals using the Limulus Amebocyte lysate test: an in vitro study. MB Express. 2014; 4: 1

[27] Bland, Jeffrey. Effect of orally consumed Aloe vera Juice on Gastrointestinal Function in Normal Humans. Preventative Medicine, March-April 1985

[28] Marzorati M. In vitro modulation of the human gastrointestinal microbial community by plant-derived polysaccharide-rich dietary supplements. Int J Food Microbiol. 2010 May 15;139(3):168-76.

[29] Im SA, Lee YR et al. In vivo evidence of the immunomodulatory activity of orally administered Aloe vera gel. Arch Pharm Res. 2010 Mar;33(3):451-6.

[30] Visuthikosol V, Chowchuen B et al. Effect of aloe vera gel to healing of burn wound a clinical and histologic study. J Med Assoc Thai. 1995 Aug;78(8):403-9

[31] Im SA, Oh ST et al. Identification of optimal molecular size of modified Aloe polysaccharides with maximum immunomodulatory activity. International Immunopharmacology. 2005;5(2):271-279

[32] Hu Y, Xu J, Hu Q. Evaluation of antioxidant potential of aloe vera (Aloe barbadensis miller) extracts. J Agric Food Chem. 2003 Dec 17;51(26):7788-91

[33] Eshun, K., He, Q. Aloe vera: a valuable ingredient for the food, pharmaceutical and cosmetic industries—a review. Crit. Rev. Food Sci. Nutr. 2004: 44, 91–96

[34] Mohamed RE, Gadour MO. The lowering effect of Gum Arabic on hyperlipidemia in Sudanese patients. Front Physiol. 2015 May 18;6:160

[35] Crociani F, Alessandrini A et al. Degradation of complex carbohydrates by Bifidobacterium spp. Int.J Food Microbiol. 1994;24:199-210.

[36] Walter DJ, Eastwood MA et al. Fermentation of wheat bran and gum arabic in rats fed on an elemental diet. Br.J.Nutr. 1988;60:225-32

[37] Wyatt GM, Bayliss CE et al. A change in human faecal flora in response to inclusion of gum arabic in the diet. Br.J.Nutr. 1986;55:261-6.

Glutathione Complex  https://www.quicksilverscientific.com/glutathionecomplexreferences/

[1] Homma T et al. Application of glutathione as anti-oxidative and anti-aging drugs. Curr Drug Metab. 2015;16(7):560-71 View Abstract

[2] Balendiran GK et al. Cell Biochem Funct. The role of glutathione in cancer. 2004 Nov-Dec;22(6):343-52. View Abstract

[3] Mari M et al. Mitochondrial glutathione, a key survival antioxidant. Antioxid Redox Signal. 2009 Nov;11(11):2685-70 View Full Paper

[4] Perricone C et al. Glutathione: a key player in autoimmunity. Autoimmun Rev. 2009 Jul;8(8):697-701. View Abstract

[5] Dröge W et al. Glutathione and immune function. Proc Nutr Soc. 2000 Nov;59(4):595-600. Review. View Abstract

[6] Bajic VP et al. Glutathione “redox homeostasis” and its relation to cardiovascular disease. Oxidative Medicine and Cellular Longevity 2019 View Abstract

[7] Abenavoli L. et al. Milk thistle in liver diseases: past, present, future. Phytother Res. 2010 Oct;24(10):1423-32 View Abstract

[8] Day CR et al. Betaine chemistry, roles, and potential use in liver disease. Biochim Biophys Acta. 2016 Jun;1860(6):1098-106. View Abstract

[9] Khodayar MJ et al. Betaine protects mice against acetaminophen hepatotoxicity possibly via mitochondrial complex II and glutathione availability. Biomed Pharmacother. 2018 Jul;103:1436-1445 View Abstract

[10] Pizzorno J. Glutathione! Integrative Medicine 2014 (13):1:8-12 View Full Paper

[11] Forman HJ. Glutathione: overview of its protective roles, measurement, and biosynthesis. Mol Aspects Med. 2009 Feb-Apr;30(1-2):1-12. View Abstract

[12] Hodges RE et al. Modulation of metabolic detoxification pathways using foods and food-derived components: a scientific review with clinical application. J Nutr Metab. 2015;2015:760689 View Full Paper

[13] Keum YS. Regulation of Nrf2-mediated phase II detoxification and anti-oxidant genes. Biomol Ther. 2012;20(2):144-151. View Abstract

[14] Fraternale A et al. Glutathione and glutathione derivatives in immunotherapy. Biol Chem. 2017 Feb 1;398(2):261-275 View Abstract

[15] Kamide Y. Allergy. Intracellular glutathione redox status in human dendritic cells regulates IL-27 production and T-cell polarization. Allergy. 2011 Sep;66(9):1183-92. View Abstract

[16] Dröge W et al. Functions of glutathione and glutathione disulfide in immunology and immunopathology. FASEB J 1994;8:1131–8. View Abstract

[17] Gambhir JK et al. Correlation between blood antioxidant levels and lipid peroxidation in rheumatoid arthritis. Clin Biochem 1997;30:351–5. View Abstract

[18] Ortona E, Redox state, cell death and autoimmune diseases: a gender perspective. Autoimmun Rev 2008;7:579–84. View Abstract

[19] Griffiths HR. Is the generation of neo-antigenic determinants by free radicals central to the development of autoimmune rheumatoid disease? Autoimmun Rev 2008;7:544–9. View Abstract

[20] Burek CL, Rose NR. Autoimmune thyroiditis and ROS. Autoimmun Rev 2008;7:530–7. View Abstract

[21] Gheita TA et al.  Measurement of malondialdehyde, glutathione, and glutathione peroxidase in SLE patients. Methods Mol Biol. 2014;1134:193-9 View Abstract

[22] Kumar D et al. A link between maternal malnutrition and depletion of glutathione in the developing lens: a possible explanation for idiopathic childhood cataract? Clin Exp Optom. 2013 Nov;96(6):523-8 View Abstract

[23] Teskey G. Glutathione as a marker for human disease. Adv Clin Chem. 2018;87:141-159. View Abstract

[24] Jiang S et al. Glutathione protects against hepatic injury in a murine model of primary Sjögren’s syndrome. Bosn J Basic Med Sci. 2016 Aug 2;16(3):227-31 View Abstract

[25] Sinha R et al. Oral supplementation with liposomal glutathione elevates body stores of glutathione and markers of immune function. Eur J Clin Nutr. 2018 Jan;72(1):105-111 View Abstract

[26] Drisko JA. Chelation Therapy. In: Integrative Medicine (Fourth Edition) 2018: (107): 1004-1014.

[27] Lawley SD et al. Mathematical modeling of the effects of glutathione on arsenic methylation. Theor Biol Med Model. 2014 May 16;11:20. View Abstract

[28] Guildford FT et al. Deficient glutathione in the pathophysiology of mycotoxin-related illness. Toxins (Basel). 2014 Feb 10;6(2):608-23 View Full Paper

[29] Hope JH et al. A review of the diagnosis and treatment of ochratoxin a inhalational exposure associated with human illness and kidney disease including focal segmental glomerulosclerosis. J. Environ. Public Health 2012: 2012, 835059. View Abstract

[30] Damy T et al. Glutathione deficiency in cardiac patients is related to the functional status and structural cardiac abnormalities. PLoS One 2009. (4):3: e4781 vol. 4. View Abstract

[31] Biswas SK et al. Depressed glutathione synthesis precedes oxidative stress and atherogenesis in Apo-E−/− Biochemical and Biophysical Research Communications 2005 (338): 3: 1368–1373 View Abstract

[32] Shimizu H et al. Relationship between plasma glutathione levels and cardiovascular disease in a defined population: the Hisayama study.  Stroke. 2004 (35):9: 2072-2077 View Abstract

[33] de la Asuncion JG et al. Mitochondrial glutathione oxidation correlates with age-associated oxidative damage to mitochondrial DNA. The FASEB Journal. 1996;10(2):333–338. View Abstract

[34] Rae CD et al. Glutathione in the human brain: Review of its roles and measurement by magnetic resonance spectroscopy. Anal Biochem. 2017 Jul 15;529:127-143. View Abstract

[35] Saharan S et al. The emerging role of glutathione in Alzheimer’s disease J Alzheimers Dis. 2014;40(3):519-29. View Abstract

[36] Chen Y et al. Glutathione defense mechanism in liver injury: insights from animal models. Food Chem Toxicol. 2013 Oct;60:38-44 View Full Paper

[37] Crocenzi FA  et al. Silymarin as a new hepatoprotective agent in experimental cholestasis: new possibilities for an ancient medication. Curr Med Chem. 2006;13(9):1055-74. View Abstract

[38] Soto C. Effect of silymarin on kidneys of rats suffering from alloxan-induced diabetes mellitus Phytomedicine. 2010 Dec 1;17(14):1090-4. View Abstract

[39] Kiruthiga PV Silymarin protects PBMC against B(a)P induced toxicity by replenishing redox status and modulating glutathione metabolizing enzymes–an in vitro study. Toxicol Appl Pharmacol. 2010 Sep 1;247(2):116-28. View Abstract

[40] Day CR et al. Betaine chemistry, roles, and potential use in liver disease. Biochim Biophys Acta. 2016 Jun;1860(6):1098-106 View Abstract

[41] Kempson SA et al. Betaine transport in kidney and liver: use of betaine in liver injury. Cell Physiol Biochem. 2013;32(7):32-40 View Abstract

[42] Zhao G et al. Betaine in inflammation: mechanistic aspects and applications.  Front Immunol. 2018 May 24;9:1070. View Full Paper

[43] Craig SA. Betaine in human nutrition. Am J Clin Nutr. 2004 Sep;80(3):539-49. View Abstract

[44] Mikkelsen K et al. B Vitamins and Ageing. In: Biochemistry and Cell Biology of Ageing: Part I Biomedical Science, Subcellular Biochemistry 90: Chapter 15. View Abstract

[45] Hodges RE et al. Modulation of metabolic detoxification pathways using foods and food-derived components: a scientific review with clinical application. J Nutr Metab. 2015;2015:760689. View Abstract

[46] Gambhir JK et al. Correlation between blood antioxidant levels and lipid peroxidation in rheumatoid arthritis. Clin Biochem 1997;30:351–5. View Abstract

[47] Ortona E, Redox state, cell death and autoimmune diseases: a gender perspective. Autoimmun Rev 2008;7:579–84. View Abstract

[48] Griffiths HR. Is the generation of neo-antigenic determinants by free radicals central to the development of autoimmune rheumatoid disease? Autoimmun Rev 2008;7:544–9. View Abstract

[49] Burek CL, Rose NR. Autoimmune thyroiditis and ROS. Autoimmun Rev 2008;7:530–7. View Abstract

[50] Gheita TA et al.  Measurement of malondialdehyde, glutathione, and glutathione peroxidase in SLE patients. Methods Mol Biol. 2014;1134:193-9 View Abstract

[51] Kumar D et al. A link between maternal malnutrition and depletion of glutathione in the developing lens: a possible explanation for idiopathic childhood cataract? Clin Exp Optom. 2013 Nov;96(6):523-8 View Abstract

[52] Teskey G. Gluathione as a marker for human disease. Adv Clin Chem. 2018;87:141-159. View Abstract

[53] Jiang S et al. Glutathione protects against hepatic injury in a murine model of primary Sjögren’s syndrome. Bosn J Basic Med Sci. 2016 Aug 2;16(3):227-31 View Abstract

Cat’s Claw Elite® https://www.quicksilverscientific.com/catsclawelitereferences/

[1] Cat’s Claw. American Botanical Councilhttp://cms.herbalgram.org/ABCGuide/Monographs/CatsClaw.html?ts=1574449798&signature=699be2950b6b1525d87c4cc1e64b16a3.

[2] Navarro M, et al. Polyphenolic composition and antioxidant activity of Uncaria tomentosacommercial bark products. Antioxidants (Basel). 2019; 8(9): pii: E339.

[3] Aguilar JLH, et al. Anti-inflammatory activity of two different extracts of Uncaria tomentosa (Rubiaceae). J Ethnopharmacol. 2002; 81(2): 271-276.

[4] Herrera DR, et al. Antimicrobial activity and substantivity of Uncaria tomentosain infected root canal dentin. Braz Oral Res. 2016; 30(1): e61.

[5] Farias I, et al. Uncaria tomentosa stimulates the proliferation of myeloid progenitor cells. J Ethnopharmacol. 2011; 137(1): 856-863.

[6] Hardin SR. Cat’s Claw: an Amazonian vine decreases inflammation in osteoarthritis. Complement Ther Clin Pract. 2007; 13(1): 25-28.

[7] Mur E, et al. Randomized double-blind trial of an extract from the pentacyclic alkaloid-chemotype of Uncaria tomentosa for the treatment of rheumatoid arthritis. J Rheumatol. 2002; 29(4): 678-681.

[8] Schlievert PM, et al. Glycerol monolaurate contributes to the antimicrobial and anti-inflammatory activity of human milk. Sci Rep. 2019; 9: 14550.

[9] Da Silva Lima R, Block JM. Coconut oil: what do we really know about it so far? Food Qual Safety. 2019; 3(2): 61-72.

[10] Saleem D, et al. In vitro evaluation of antifungal activity of monolaurin against Candida albicans biofilms. Peer J. 2016; 4: e2148.

[11] Ham Y, Kim TJ. Inhibitory activity of monoacylglycerols on biofilm formation in Aeromonas hydrophilaStreptococcus mutansXanthomonas oryzae, and Yersinia enterocoliticaSpringerplus. 2016; 5(1): 1526.

[12] Chirumbolo S, et al. The role of vitamin D in the immune system as a pro-survival molecule. Clin Ther. 2017; 39(5): 894-916.

[13] Prietl B, et al. Vitamin D and immune function. Nutrients. 2013; 5(7): 2502-2521.

[14] Heaney RP, et al. Vitamin D3 is more potent than vitamin D2 in humans. JCEM. 2011; 96(3): E447-E452.

[15] Firenzuoli F, et al. Essential oils: New perspectives in human health and wellness. Evid Based Complement Alternat Med. 2014; 2014: 467363.

[16] Hussain AI, et al. Chemical composition, and antioxidant and antimicrobial activities of essential oil of spearmint (Mentha spicata L.) from Pakistan. J Essent Oil Res. 2010; 22(1): 78-84.

[17] Nayebi N, et al. A systematic review of the efficacy and safety of Rosa damascena Mill. with an overview on its phytopharmacological properties. Complement Ther Med. 2017; 34: 129-140.

[18] Miraj S, et al. Melissa officinalis L: A review study with an antioxidant perspective. J Evid Based Complementary Altern Med. 2017; 22(3): 385-394.

[19] Sherry M, et al. Essential oils encapsulated in liposomes: a review. J Liposome Res. 2013; 23(4): 268-275.

[20] Jackman JA, et al. Nanotechnology formulations for antibacterial free fatty acids and monoglycerides. Molecules. 2016; 21(3): 305.

[21] Mohammadi M, et al. Formulation of nanoliposomal vitamin D3 for potential application in beverage fortification. Adv Pharm Bull. 2014; 4(Suppl 2): 569-575.

[22] Ahn H, Park JH. Liposomal delivery systems for intestinal lymphatic drug transport. Biomater Res. 2016; 20: 36.

[23] Alyautdin R, et al. Nanoscale drug delivery systems and the blood-brain barrier. Int J Nanomedicine. 2014; 9: 795-811.

[24] Spector AA, Yorek MA. Membrane lipid composition and cellular function. J Lipid Res. 1985; 26(9): 1015-1035.

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

Glutathione https://www.quicksilverscientific.com/glutathionereferences/

[1] Homma T et al. Application of glutathione as anti-oxidative and anti-aging drugs. Curr Drug Metab. 2015;16(7):560-71 View Abstract

[2] Ighodaroab OM et al. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid.  Alexandria Journal of Medicine 2018 (54): 287-293 View Abstract

[3] Szarka A et al. The ascorbate-glutathione-α-tocopherol triad in abiotic stress response. Int J Mol Sci. 2012;13(4):4458-83View Full Paper

[4] Balendiran GK et al. Cell Biochem Funct. The role of glutathione in cancer. 2004 Nov-Dec;22(6):343-52. View Abstract

[5] Mari M et al. Mitochondrial glutathione, a key survival antioxidant. Antioxid Redox Signal. 2009 Nov;11(11):2685-70 View Full Paper

[6] Perricone C et al. Glutathione: a key player in autoimmunity. Autoimmun Rev. 2009 Jul;8(8):697-701. View Abstract

[7] Dröge W et al. Glutathione and immune function. Proc Nutr Soc. 2000 Nov;59(4):595-600. Review. View Abstract

[8] Bajic VP et al. Glutathione “redox homeostasis” and its relation to cardiovascular disease. Oxidative Medicine and Cellular Longevity 2019 View Abstract

[9] Pizzorno J. Glutathione! Integrative Medicine 2014 (13):1:8-12 View Full Paper

[10] Forman HJ. Glutathione: overview of its protective roles, measurement, and biosynthesis. Mol Aspects Med. 2009 Feb-Apr;30(1-2):1-12. View Abstract

[11] Hodges RE et al. Modulation of metabolic detoxification pathways using foods and food-derived components: a scientific review with clinical application. J Nutr Metab. 2015;2015:760689 View Full Paper

[12] Keum YS. Regulation of Nrf2-mediated phase II detoxification and anti-oxidant genes. Biomol Ther. 2012;20(2):144-151. View Abstract

[13] Fraternale A et al. Glutathione and glutathione derivatives in immunotherapy. Biol Chem. 2017 Feb 1;398(2):261-275 View Abstract

[14] Kamide Y. Allergy. Intracellular glutathione redox status in human dendritic cells regulates IL-27 production and T-cell polarization. Allergy. 2011 Sep;66(9):1183-92. View Abstract

[15] Dröge W et al. Functions of glutathione and glutathione disulfide in immunology and immunopathology. FASEB J 1994;8:1131–8. View Abstract

[16] Gambhir JK et al. Correlation between blood antioxidant levels and lipid peroxidation in rheumatoid arthritis. Clin Biochem 1997;30:351–5. View Abstract

[17] Ortona E, Redox state, cell death and autoimmune diseases: a gender perspective. Autoimmun Rev 2008;7:579–84. View Abstract

[18] Griffiths HR. Is the generation of neo-antigenic determinants by free radicals central to the development of autoimmune rheumatoid disease? Autoimmun Rev 2008;7:544–9. View Abstract

[19] Burek CL, Rose NR. Autoimmune thyroiditis and ROS. Autoimmun Rev 2008;7:530–7. View Abstract

[20] Gheita TA et al.  Measurement of malondialdehyde, glutathione, and glutathione peroxidase in SLE patients. Methods Mol Biol. 2014;1134:193-9 View Abstract

[21] Kumar D et al. A link between maternal malnutrition and depletion of glutathione in the developing lens: a possible explanation for idiopathic childhood cataract? Clin Exp Optom. 2013 Nov;96(6):523-8 View Abstract

[22] Teskey G. Glutathione as a marker for human disease. Adv Clin Chem. 2018;87:141-159. View Abstract

[23] Jiang S et al. Glutathione protects against hepatic injury in a murine model of primary Sjögren’s syndrome. Bosn J Basic Med Sci. 2016 Aug 2;16(3):227-31 View Abstract

[24] Sinha R et al. Oral supplementation with liposomal glutathione elevates body stores of glutathione and markers of immune function. Eur J Clin Nutr. 2018 Jan;72(1):105-111 View Abstract

[25] Drisko JA. Chelation Therapy. In: Integrative Medicine (Fourth Edition) 2018: (107): 1004-1014.

[26] Lawley SD et al. Mathematical modeling of the effects of glutathione on arsenic methylation. Theor Biol Med Model. 2014 May 16;11:20. View Abstract

[27] Guildford FT et al. Deficient glutathione in the pathophysiology of mycotoxin-related illness. Toxins (Basel). 2014 Feb 10;6(2):608-23 View Full Paper

[28] Hope JH et al. A review of the diagnosis and treatment of ochratoxin a inhalational exposure associated with human illness and kidney disease including focal segmental glomerulosclerosis. J. Environ. Public Health 2012: 2012, 835059. View Abstract

[29] Damy T et al. Glutathione deficiency in cardiac patients is related to the functional status and structural cardiac abnormalities. PLoS One 2009. (4):3: e4781 vol. 4. View Abstract

[30] Biswas SK et al. Depressed glutathione synthesis precedes oxidative stress and atherogenesis in Apo-E−/− Biochemical and Biophysical Research Communications 2005 (338): 3: 1368–1373View Abstract

[31] Shimizu H et al. Relationship between plasma glutathione levels and cardiovascular disease in a defined population: the Hisayama study.  Stroke. 2004 (35):9: 2072-2077 View Abstract

[32] de la Asuncion JG et al. Mitochondrial glutathione oxidation correlates with age-associated oxidative damage to mitochondrial DNA. The FASEB Journal. 1996;10(2):333–338. View Abstract

[33] Rae CD et al. Glutathione in the human brain: Review of its roles and measurement by magnetic resonance spectroscopy. Anal Biochem. 2017 Jul 15;529:127-143. View Abstract

[34] Saharan S et al. The emerging role of glutathione in Alzheimer’s disease J Alzheimers Dis. 2014;40(3):519-29. View Abstract

[35] Gambhir JK et al. Correlation between blood antioxidant levels and lipid peroxidation in rheumatoid arthritis. Clin Biochem 1997;30:351–5. View Abstract

[36] Ortona E, Redox state, cell death and autoimmune diseases: a gender perspective. Autoimmun Rev 2008;7:579–84. View Abstract

[37] Griffiths HR. Is the generation of neo-antigenic determinants by free radicals central to the development of autoimmune rheumatoid disease? Autoimmun Rev 2008;7:544–9. View Abstract

[38] Burek CL, Rose NR. Autoimmune thyroiditis and ROS. Autoimmun Rev 2008;7:530–7. View Abstract

[39] Gheita TA et al.  Measurement of malondialdehyde, glutathione, and glutathione peroxidase in SLE patients. Methods Mol Biol. 2014;1134:193-9 View Abstract

[40] Kumar D et al. A link between maternal malnutrition and depletion of glutathione in the developing lens: a possible explanation for idiopathic childhood cataract? Clin Exp Optom. 2013 Nov;96(6):523-8 View Abstract

[41] Teskey G. Gluathione as a marker for human disease. Adv Clin Chem. 2018;87:141-159. View Abstract

[42] Jiang S et al. Glutathione protects against hepatic injury in a murine model of primary Sjögren’s syndrome. Bosn J Basic Med Sci. 2016 Aug 2;16(3):227-31 View Abstract

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.

0
    0
    Your Cart
    You're 99.00 away from free shipping.
    Your cart is empty
      Calculate Shipping
      Apply Coupon

      to your account for access to our

      Memorial Day

      Weekend Sale!

      Don’t have an account? Sign up here!