References

Functional Med System - F.M.S

References

  • Cooper DS. Subclinical Hypothyroidism. NEJM. 2001 Jul 26;345: 260– 265.
  • Persky VW, Turyk ME, Wang L, Freels S, Chatterton R Jr, Barnes S, Erdman J Jr, Sepkovic DW, Bradlow HL, Potter S. Effect of soy protein on endogenous hormones in postmenopausal women. Am J Clin Nutr. 2002 Jan; 75( 1): 145– 153. Erratum in: Am J Clin Nutr. 2002 Sep; 76( 3): 695
  • Toscano V, Conti FG, Anastasi E, Mariani P, Tiberti C, Poggi M, Montuori M, Monti S, Laureti S, Cipolletta E, Gemme G, Caiola S, Di Mario U, Bonamico M. Importance of gluten in the induction of endocrine autoantibodies and organ dysfunction in adolescent celiac patients. Am J Gastroenterol. 2000 Jul; 95( 7): 1742–1748.
  • Ellingsen DG, Efskind J. Effects of low mercury vapour exposure on the thyroid function in chloralkali workers. J Appl Toxicol. 2000 Nov– Dec; 20( 6): 483– 489.
  • WJ, Pan Y; Johnson AR, et al. Reduction of chemical sensitivity by means of heat depuration, physical therapy and nutritional supplementation in a controlled environment. J Nutr Env Med. 1996;6: 141– 148.
  • Pelletier C, Imbeault P, Tremblay A. Energy balance and pollution by organochlorines and polychlorinated biphenyls. Obes Rev. 2003 Feb; 4( 1): 17– 24. Review.
  • Bland J. Nutritional Endocrinology, Normalizing Hypothalamus-Pituitary-Thyroid Axis Function, 2002 Seminar Series Syllabus.
  • Gaby AR. Sub-laboratory hypothyroidism and the empirical use of Armour thyroid. Altern Med Rev. 2004 Jun; 9( 2): 157– 179.
  • Goglia F. Biological effects of 3,5-diiodothyronine (T( 2)). Biochemistry (Moscow). 2005 Feb; 70( 2): 164– 172.
  • Trbojević B, Djurica S. Diagnosis of autoimmune thyroid disease. Srp Arh Celok Lek 2005;133 Suppl 1:25-33. [Abstract]
  • Duntas LH. Environmental factors and autoimmune thyroiditis. Nat Clin Pract Endocrinol Metab 2008;4(8):454-460. [Abstract]
  • Wiersinga WM. Clinical relevance of environmental factors in the pathogenesis of autoimmune thyroid disease. Endocrinol Metab (Seoul) 2016;31(2):213-222. [Full text]
  • Kawicka A, Regulska-Ilow B. Metabolic disorders and nutritional status in autoimmune thyroid diseases. Postepy Hig Med Dosw (Online) 2015;69:80-90. [Full text]
  • Wang L, Wang B, Chen SR, et al. Effect of selenium supplementation on recurrent hyperthyroidism caused by Graves’ disease: a prospective pilot study. Horm Metab Res 2016;[Epub ahead of print]. [Abstract]
  • Dharmasena A. Selenium supplementation in thyroid associated ophthalmopathy: an update. Int J Ophthalmol 2014;7(2):365-375. [Full text]
  • Marcocci C, Kahaly GJ, Krassas GE, et al. Selenium and the course of mild Graves’ orbitopathy. N Engl J Med 2011;364(20):1920-1931. [Full text]
  • Hwang S, Byun JW, Yoon JS, et al. Inhibitory effects of α-lipoic acid on oxidative stress-induced adipogenesis in orbital fibroblasts from patients with Graves ophthalmopathy. Medicine (Baltimore) 2016;95(2):e2497. [Full text]
  • Chen K, Yan B, Wang F, et al. Type 1 5′-deiodinase activity is inhibited by oxidative stress and restored by alpha-lipoic acid in HepG2 cells. Biochem Biophys Res Commun 2016;472(3):496-501. [Abstract]
  • Sharma BR, Joshi AS, Varthakavi PK, et al. Celiac autoimmunity in autoimmune thyroid disease is highly prevalent with a questionable impact. Indian J Endocrinol Metab 2016;20(1):97-100. [Full text]
  • Roy A, Laszkowska M, Sundström J, et al. Prevalence of celiac disease in patients with autoimmune thyroid disease: a meta-analysis. Thyroid 2016;26(7):880-890. [Abstract]
  • Drago S, El Asmar R, Di Pierro M, et al. Gliadin, zonulin and gut permeability: effects on celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand J Gastroenterol 2006;41(4):408-419. [Abstract]
  • Fasano A. Leaky gut and autoimmune diseases. Clin Rev Allergy Immunol 2012;42(1):71-78. [Abstract]
  • Park JH, Jeong SY, Choi AJ, et al. Lipopolysaccharide directly stimulates Th17 differentiation in vitro modulating phosphorylation of RelB and NF-κB1. Immunol Lett 2015;165(1):10-19. [Abstract]
  • Orlando A, Linsalata M, Notarnicola M, et al. Lactobacillus GGrestoration of the gliadin induced epithelial barrier disruption: the role of cellular polyamines. BMC Microbiol 2014;14:19. [Full text]
  • Anderson RC, Cookson AL, McNabb WC, et al. Lactobacillus plantarumMB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol 2010;10:316. [Full text]
  • Liu Z, Li C, Huang M, et al. Positive regulatory effects of perioperative probiotic treatments on postoperative liver complications after colorectal liver metastases surgery: a double-center and double-blind randomized clinical trial. BMC Gastroenterol 2015;15:34. [Full text]
  • Skrovanek S, DiGuilio K, Bailey R, et al. Zinc and gastrointestinal disease. World J Gastrointest Pathophysiol 2014;5(4):496-513. [Full text]
  • Finamore A, Massimi M, Conti Devirgiliis L, et al. Zinc deficiency induces membrane barrier damage and increases neutrophil transmigration in Caco-2 cells. J Nutr 2008;138(9):1664-1670. [Full text]
  • Wang J, Lv S, Chen G, et al. Meta-analysis of the association between vitamin D and autoimmune thyroid disease. Nutrients 2015;7(4):2485-2498. [Full text]
  • Kim D. Low vitamin D status is associated with hypothyroid Hashimoto’s thyroiditis. Hormones (Athens). 2016;[Epub ahead of print]. [Full text]
  • Arslan MS, Ekiz F, Deveci M, et al. The relationship between cytotoxin-associated gene A positive Helicobacter pyloriinfection and autoimmune thyroid disease. Endocr Res 2015;40(4):211-214. [Abstract]
  • Radić M. Role of Helicobacter pyloriinfection in autoimmune systemic rheumatic diseases. World J Gastroenterol 2014;20(36):12839-12846. [Full text]
  • Di Crescenzo V, D’Antonio A, Tonacchera M, et al. Human herpes virus associated with Hashimoto’s thyroiditis. Infez Med 2013;21(3):224-228. [Full text]
  • Draborg AH, Duus K, Houen G. Epstein-Barr virus in systemic autoimmune diseases. Clin Dev Immunol 2013;2013:535738. [Full text]
  • Gaberšček S, Zaletel K, Schwetz V, et al. Mechanisms in endocrinology: thyroid and polycystic ovary syndrome. Eur J Endocrinol 2015;172(1):R9-21. [Abstract]
  • Arduc A, Aycicek Dogan B, Bilmez S, et al. High prevalence of Hashimoto’s thyroiditis in patients with polycystic ovary syndrome: does the imbalance between estradiol and progesterone play a role? Endocr Res 2015;40(4):204-210. [Abstract]
  • Jara LJ, Medina G, Saavedra MA, et al. Prolactin and autoimmunity. Clin Rev Allergy Immunol 2011;40(1):50-59. [Abstract]
  • Nociti V, Frisullo G, Tartaglione T, et al. Multiple sclerosis attacks triggered by hyperprolactinemia. J Neurooncol 2010;98(3):407-409. [Full text]
  • Legorreta-Haquet MV, Chávez-Rueda K, Chávez-Sánchez L, et al. Function of Treg cells decreased in patients with systemic lupus erythematosus due to the effect of prolactin. Medicine (Baltimore) 2016;95(5):e2384. [Full text]
  • van Die MD, Burger HG, Teede HJ, et al. Vitex agnus-castusextracts for female reproductive disorders: a systematic review of clinical trials. Planta Med 2013;79(7):562-575. [Abstract]
  • Yu Q, Wang JB. Subclinical hypothyroidism in PCOS: impact on presentation, insulin resistance, and cardiovascular risk. Biomed Res Int 2016;2016:2067087. [Full text]
  • Tenazzani AD, Despini G, Santagni S, et al. Effects of a combination of alpha lipoic acid and myo-inositol on insulin dynamics in overweight/obese patients with PCOS. Endocrinol Metab Synd 2014;3:3. [Full text]
  • Morgante G, Cappelli V, Di Sabatino A, et al. Polycystic ovary syndrome (PCOS) and hyperandrogenism: the role of a new natural association. Minerva Ginecol 2015;67(5):457-463. [Abstract]
  • Mortimer RH. Thyroid function tests. Aust Prescr 2011;34:12-15. [Full text]
  • The Australian Thyroid Foundation Ltd. Thyroid conditions. Viewed 6 Spetember 2016, https://www.thyroidfoundation.org.au/page/58/thyroid-conditions
  • Act for Libraries. Difference between Hashimotos thyroiditis and Graves disease. Viewed 6 September, http://www.actforlibraries.org/difference-between-hashimotos-thyroiditis…
  • Lab Tests Online. Thyroid antibodies, 2015. Viewed 6 September 2016, https://labtestsonline.org/understanding/analytes/thyroid-antibodies/tab…
  • Trbojević B, Djurica S. Diagnosis of autoimmune thyroid disease. Srp Arh Celok Lek 2005;133 Suppl 1:25-33. [Abstract]
  • Duntas LH. Environmental factors and autoimmune thyroiditis. Nat Clin Pract Endocrinol Metab 2008;4(8):454-460. [Abstract]
  • Wiersinga WM. Clinical relevance of environmental factors in the pathogenesis of autoimmune thyroid disease. Endocrinol Metab (Seoul) 2016;31(2):213-222. [Full text]
  • Kawicka A, Regulska-Ilow B. Metabolic disorders and nutritional status in autoimmune thyroid diseases. Postepy Hig Med Dosw (Online) 2015;69:80-90. [Full text]
  • Wang L, Wang B, Chen SR, et al. Effect of selenium supplementation on recurrent hyperthyroidism caused by Graves’ disease: a prospective pilot study. Horm Metab Res 2016;[Epub ahead of print]. [Abstract]
  • Dharmasena A. Selenium supplementation in thyroid associated ophthalmopathy: an update. Int J Ophthalmol 2014;7(2):365-375. [Full text]
  • Marcocci C, Kahaly GJ, Krassas GE, et al. Selenium and the course of mild Graves’ orbitopathy. N Engl J Med 2011;364(20):1920-1931. [Full text]
  • Hwang S, Byun JW, Yoon JS, et al. Inhibitory effects of α-lipoic acid on oxidative stress-induced adipogenesis in orbital fibroblasts from patients with Graves ophthalmopathy. Medicine (Baltimore) 2016;95(2):e2497. [Full text]
  • Chen K, Yan B, Wang F, et al. Type 1 5′-deiodinase activity is inhibited by oxidative stress and restored by alpha-lipoic acid in HepG2 cells. Biochem Biophys Res Commun 2016;472(3):496-501. [Abstract]
  • Sharma BR, Joshi AS, Varthakavi PK, et al. Celiac autoimmunity in autoimmune thyroid disease is highly prevalent with a questionable impact. Indian J Endocrinol Metab 2016;20(1):97-100. [Full text]
  • Roy A, Laszkowska M, Sundström J, et al. Prevalence of celiac disease in patients with autoimmune thyroid disease: a meta-analysis. Thyroid 2016;26(7):880-890. [Abstract]
  • Drago S, El Asmar R, Di Pierro M, et al. Gliadin, zonulin and gut permeability: effects on celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand J Gastroenterol 2006;41(4):408-419. [Abstract]
  • Fasano A. Leaky gut and autoimmune diseases. Clin Rev Allergy Immunol 2012;42(1):71-78. [Abstract]
  • Park JH, Jeong SY, Choi AJ, et al. Lipopolysaccharide directly stimulates Th17 differentiation in vitro modulating phosphorylation of RelB and NF-κB1. Immunol Lett 2015;165(1):10-19. [Abstract]
  • Orlando A, Linsalata M, Notarnicola M, et al. Lactobacillus GGrestoration of the gliadin induced epithelial barrier disruption: the role of cellular polyamines. BMC Microbiol 2014;14:19. [Full text]
  • Anderson RC, Cookson AL, McNabb WC, et al. Lactobacillus plantarumMB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol 2010;10:316. [Full text]
  • Liu Z, Li C, Huang M, et al. Positive regulatory effects of perioperative probiotic treatments on postoperative liver complications after colorectal liver metastases surgery: a double-center and double-blind randomized clinical trial. BMC Gastroenterol 2015;15:34. [Full text]
  • Skrovanek S, DiGuilio K, Bailey R, et al. Zinc and gastrointestinal disease. World J Gastrointest Pathophysiol 2014;5(4):496-513. [Full text]
  • Finamore A, Massimi M, Conti Devirgiliis L, et al. Zinc deficiency induces membrane barrier damage and increases neutrophil transmigration in Caco-2 cells. J Nutr 2008;138(9):1664-1670. [Full text]
  • Wang J, Lv S, Chen G, et al. Meta-analysis of the association between vitamin D and autoimmune thyroid disease. Nutrients 2015;7(4):2485-2498. [Full text]
  • Kim D. Low vitamin D status is associated with hypothyroid Hashimoto’s thyroiditis. Hormones (Athens). 2016;[Epub ahead of print]. [Full text]
  • Arslan MS, Ekiz F, Deveci M, et al. The relationship between cytotoxin-associated gene A positive Helicobacter pyloriinfection and autoimmune thyroid disease. Endocr Res 2015;40(4):211-214. [Abstract]
  • Radić M. Role of Helicobacter pyloriinfection in autoimmune systemic rheumatic diseases. World J Gastroenterol 2014;20(36):12839-12846. [Full text]
  • Di Crescenzo V, D’Antonio A, Tonacchera M, et al. Human herpes virus associated with Hashimoto’s thyroiditis. Infez Med 2013;21(3):224-228. [Full text]
  • Draborg AH, Duus K, Houen G. Epstein-Barr virus in systemic autoimmune diseases. Clin Dev Immunol 2013;2013:535738. [Full text]
  • Gaberšček S, Zaletel K, Schwetz V, et al. Mechanisms in endocrinology: thyroid and polycystic ovary syndrome. Eur J Endocrinol 2015;172(1):R9-21. [Abstract]
  • Arduc A, Aycicek Dogan B, Bilmez S, et al. High prevalence of Hashimoto’s thyroiditis in patients with polycystic ovary syndrome: does the imbalance between estradiol and progesterone play a role? Endocr Res 2015;40(4):204-210. [Abstract]
  • Jara LJ, Medina G, Saavedra MA, et al. Prolactin and autoimmunity. Clin Rev Allergy Immunol 2011;40(1):50-59. [Abstract]
  • Nociti V, Frisullo G, Tartaglione T, et al. Multiple sclerosis attacks triggered by hyperprolactinemia. J Neurooncol 2010;98(3):407-409. [Full text]
  • Legorreta-Haquet MV, Chávez-Rueda K, Chávez-Sánchez L, et al. Function of Treg cells decreased in patients with systemic lupus erythematosus due to the effect of prolactin. Medicine (Baltimore) 2016;95(5):e2384. [Full text]
  • van Die MD, Burger HG, Teede HJ, et al. Vitex agnus-castusextracts for female reproductive disorders: a systematic review of clinical trials. Planta Med 2013;79(7):562-575. [Abstract]
  • Yu Q, Wang JB. Subclinical hypothyroidism in PCOS: impact on presentation, insulin resistance, and cardiovascular risk. Biomed Res Int 2016;2016:2067087. [Full text]
  • Tenazzani AD, Despini G, Santagni S, et al. Effects of a combination of alpha lipoic acid and myo-inositol on insulin dynamics in overweight/obese patients with PCOS. Endocrinol Metab Synd 2014;3:3. [Full text]
  • Morgante G, Cappelli V, Di Sabatino A, et al. Polycystic ovary syndrome (PCOS) and hyperandrogenism: the role of a new natural association. Minerva Ginecol 2015;67(5):457-463. [Abstract]
  • Mortimer RH. Thyroid function tests. Aust Prescr 2011;34:12-15. [Full text]
  • The Australian Thyroid Foundation Ltd. Thyroid conditions. Viewed 6 Spetember 2016, https://www.thyroidfoundation.org.au/page/58/thyroid-conditions
  • Act for Libraries. Difference between Hashimotos thyroiditis and Graves disease. Viewed 6 September, http://www.actforlibraries.org/difference-between-hashimotos-thyroiditis…
  • Lab Tests Online. Thyroid antibodies, 2015. Viewed 6 September 2016, https://labtestsonline.org/understanding/analytes/thyroid-antibodies/tab…

 

  • Nobuyuki Amino 4 Autoimmunity and hypothyroidism Bailiére’s Clinical Endocrinology and Metabolism Volume 2, Issue 3 August 1988. doi:10.1016/S0950-351X(88)80055-7
  • van Rijn LE, Pop VJ, Williams GR. Low bone mineral density is related to high physiological levels of free thyroxine in peri-menopausal women. Eur J Endocrinol. 2014;170(3):461‐468. Published 2014 Feb 7. doi:10.1530/EJE-13-0769
  • Yaylali O, Kirac S, Yilmaz M, et al. Does hypothyroidism affect gastrointestinal motility?. Gastroenterol Res Pract. 2009;2009:529802. doi:10.1155/2009/529802
  • NHS “Erectile dysfunction (impotence)” 16 August 2017 https://www.nhs.uk/conditions/erection-problems-erectile-dysfunction/
  • Kalra S, Unnikrishnan AG, Sahay R. The hypoglycemic side of hypothyroidism. Indian J Endocrinol Metab. 2014;18(1):1‐3. doi:10.4103/2230-8210.126517
  • Abrams JJ, Grundy SM. Cholesterol metabolism in hypothyroidism and hyperthyroidism in man. J Lipid Res. 1981;22(2):323‐338.
  • CJ Gardner, P Richardson, et al. Hypothyroidism in a patient with non-alcoholic fatty liver disease BMJ 2011; 342 doi: https://doi.org/10.1136/bmj.c7199
  • Sintzel F, Mallaret M, Bougerol T. Potentialisation par les hormones thyroïdiennes des traitements tricycliques et sérotoninergiques dans les dépressions résistantes [Potentializing of tricyclics and serotoninergics by thyroid hormones in resistant depressive disorders]. Encephale. 2004;30(3):267‐275. doi:10.1016/s0013-7006(04)95439-5
  • Berent D, Zboralski K, Orzechowska A, Gałecki P. Thyroid hormones associated with depression severity and clinical outcome in patients with major depressive disorder. Mol Biol Rep. 2014;41(4):2419‐2425. doi:10.1007/s11033-014-3097-6
  • Brănişteanu DE, Dimitriu A, Vieriu M, et al. Cutaneous manifestations associated with thyroid disease. Rev Med Chir Soc Med Nat Iasi. 2014;118(4):953‐958.
  • B. Winsa, MD, A. Karlsson, MD et al. Stressful life events and Graves’ disease The Lancet VOLUME 338, ISSUE 8781, P1475-1479, DECEMBER 14, 1991 doi: https://doi.org/10.1016/0140-6736(91)92298-G
  • Matos‐Santos, A., Nobre, E.L., Costa, J.G.E., Nogueira, P.J., Macedo, A., Galvão‐Teles, A. and De Castro, J.J. (2001), Relationship between the number and impact of stressful life events and the onset of Graves’ disease and toxic nodular goitre★. Clinical Endocrinology, 55: 15-19. doi:10.1046/j.1365-2265.2001.01332.x
  • Sategna-Guidetti C, Bruno M, Mazza E, et al. Autoimmune thyroid diseases and coeliac disease. Eur J Gastroenterol Hepatol. 1998;10(11):927‐931. doi:10.1097/00042737-199811000-00005
  • Mainardi E, Montanelli A, Dotti M, Nano R, Moscato G. Thyroid-related autoantibodies and celiac disease: a role for a gluten-free diet?. J Clin Gastroenterol. 2002;35(3):245‐248. doi:10.1097/00004836-200209000-00009
  • Roland Gärtner, Barbara C. H. Gasnier, Johannes W. Dietrich, Bjarne Krebs, Matthias W. A. Angstwurm, Selenium Supplementation in Patients with Autoimmune Thyroiditis Decreases Thyroid Peroxidase Antibodies Concentrations, The Journal of Clinical Endocrinology & Metabolism, Volume 87, Issue 4, 1 April 2002, Pages 1687–1691, https://doi.org/10.1210/jcem.87.4.8421
  • Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C. Vitamin D: modulator of the immune system. Curr Opin Pharmacol. 2010;10(4):482‐496. doi:10.1016/j.coph.2010.04.001
  • Arrieta MC, Bistritz L, Meddings JB. Alterations in intestinal permeability. Gut. 2006;55(10):1512‐1520. doi:10.1136/gut.2005.085373
  • Langer P, Kocan A, Tajtaková M, et al. Fish from industrially polluted freshwater as the main source of organochlorinated pollutants and increased frequency of thyroid disorders and dysglycemia. Chemosphere. 2007;67(9):S379‐S385. doi:10.1016/j.chemosphere.2006.05.132
  • Martin I. Surks, M.D., and Rubens Sievert, M.D. Drugs and Thyroid Function N Engl J Med 1995; 333:1688-1694 DOI: 10.1056/NEJM199512213332507
  • Boukis MA, Koutras DA, Souvatzoglou A, Evangelopoulou A, Vrontakis M, Moulopoulos SD. Thyroid hormone and immunological studies in endemic goiter. J Clin Endocrinol Metab. 1983;57(4):859‐862. doi:10.1210/jcem-57-4-859
  • Tomer Y, Huber A. The etiology of autoimmune thyroid disease: a story of genes and environment. J Autoimmun. 2009;32(3-4):231‐239. doi:10.1016/j.jaut.2009.02.007
  • Vestergaard P, Rejnmark L, Weeke J, et al. Smoking as a risk factor for Graves’ disease, toxic nodular goiter, and autoimmune hypothyroidism. Thyroid. 2002;12(1):69‐75. doi:10.1089/105072502753451995
  • Wajner SM, Goemann IM, Bueno AL, Larsen PR, Maia AL. IL-6 promotes nonthyroidal illness syndrome by blocking thyroxine activation while promoting thyroid hormone inactivation in human cells. J Clin Invest. 2011;121(5):1834‐1845. doi:10.1172/JCI44678
  • Farhangi MA, Keshavarz SA, Eshraghian M, Ostadrahimi A, Saboor-Yaraghi AA. The effect of vitamin A supplementation on thyroid function in premenopausal women. J Am Coll Nutr. 2012;31(4):268‐274. doi:10.1080/07315724.2012.10720431
  • Hedrén E, Diaz V, Svanberg U. Estimation of carotenoid accessibility from carrots determined by an in vitro digestion method. Eur J Clin Nutr. 2002;56(5):425‐430. doi:10.1038/sj.ejcn.1601329
  • Olivieri O, Girelli D, Stanzial AM, Rossi L, Bassi A, Corrocher R. Selenium, zinc, and thyroid hormones in healthy subjects: low T3/T4 ratio in the elderly is related to impaired selenium status. Biol Trace Elem Res. 1996;51(1):31‐41. doi:10.1007/BF02790145
  • Drutel A, Archambeaud F, Caron P. Selenium and the thyroid gland: more good news for clinicians. Clin Endocrinol (Oxf). 2013;78(2):155‐164. doi:10.1111/cen.12066
  • Zimmermann MB, Köhrle J. The impact of iron and selenium deficiencies on iodine and thyroid metabolism: biochemistry and relevance to public health. Thyroid. 2002;12(10):867‐878. doi:10.1089/105072502761016494
  • Jain RB. Thyroid function and serum copper, selenium, and zinc in general U.S. population. Biol Trace Elem Res. 2014;159(1-3):87‐98. doi:10.1007/s12011-014-9992-9
  • Abdel-Dayem, M.M., Elgendy, M.S. Effects of chronic estradiol treatment on the thyroid gland structure and function of ovariectomized rats. BMC Res Notes 2, 173 (2009). https://doi.org/10.1186/1756-0500-2-173
  • Understanding Local Control of Thyroid Hormones: (Deiodinases Function and Activity) NAHIS https://www.nahypothyroidism.org/deiodinases/
  • Nobuyuki Amino 4 Autoimmunity and hypothyroidism Baillière’s Clinical Endocrinology and Metabolism Volume 2, Issue 3, August 1988, Pages 591-617 doi: https://doi.org/10.1016/S0950-351X(88)80055-7
  • Yang, J., Yang, X. & Li, M. Baicalin, a natural compound, promotes regulatory T cell differentiation. BMC Complement Altern Med 12, 64 (2012). https://doi.org/10.1186/1472-6882-12-64
  • Asvold BO, Bjøro T, Nilsen TI, Gunnell D, Vatten LJ. Thyrotropin levels and risk of fatal coronary heart disease: the HUNT study. Arch Intern Med. 2008;168(8):855‐860. doi:10.1001/archinte.168.8.855
  • Anthony Martin Gerdes and Giorgio Iervasi Thyroid Replacement Therapy and Heart Failure Circulation 2010;122:385–393 https://doi.org/10.1161/CIRCULATIONAHA.109.917922
  • Hiroaki Kimura and Patrizio Caturegli Chemokine Orchestration of Autoimmune Thyroiditis Thyroid 2007 17:10, 1005-1011 https://doi.org/10.1089/thy.2007.0267
  • Molnár I, Balázs C, Szegedi G, Sipka S. Inhibition of type 2,5′-deiodinase by tumor necrosis factor alpha, interleukin-6 and interferon gamma in human thyroid tissue. Immunol Lett. 2002;80(1):3‐7. doi:10.1016/s0165-2478(01)00301-7
  • Kjellman BF, Ljunggren JG, Beck-Friis J, Wetterberg L. Reverse T3 levels in affective disorders. Psychiatry Res. 1983;10(1):1‐9. doi:10.1016/0165-1781(83)90022-7
  • Mebis L, van den Berghe G. The hypothalamus-pituitary-thyroid axis in critical illness. Neth J Med. 2009;67(10):332‐340.
  • Rannem T, Ladefoged K, Hylander E, Hegnhøj J, Staun M. Selenium depletion in patients with gastrointestinal diseases: are there any predictive factors?. Scand J Gastroenterol. 1998;33(10):1057‐1061. doi:10.1080/003655298750026750
  • Vitamin B12 Health Sheet NIH March 30, 2020 https://ods.od.nih.gov/factsheets/VitaminB12-HealthProfessional/
  • Schroeder AC, Privalsky ML. Thyroid hormones, t3 and t4, in the brain. Front Endocrinol (Lausanne). 2014;5:40. Published 2014 Mar 31. doi:10.3389/fendo.2014.00040
  • Moncayo R, Kroiss A, Oberwinkler M, et al. The role of selenium, vitamin C, and zinc in benign thyroid diseases and of selenium in malignant thyroid diseases: Low selenium levels are found in subacute and silent thyroiditis and in papillary and follicular carcinoma. BMC Endocr Disord. 2008;8:2. Published 2008 Jan 25. doi:10.1186/1472-6823-8-2
  • Guy E. Abraham, M.D. (1)and David Brownstein, M.D. Evidence that the administration of Vitamin C improves a defective cellular transport mechanism for iodine: A Case Report Optimox https://www.optimox.com/iodine-study-11
  • Anna J Duffield, Christine D Thomson, Kristina E Hill, Sheila Williams, An estimation of selenium requirements for New Zealanders, The American Journal of Clinical Nutrition, Volume 70, Issue 5, November 1999, Pages 896–903, https://doi.org/10.1093/ajcn/70.5.896
  • Hossein-nezhad A, Spira A, Holick MF. Influence of vitamin D status and vitamin D3 supplementation on genome wide expression of white blood cells: a randomized double-blind clinical trial. PLoS One. 2013;8(3):e58725. doi:10.1371/journal.pone.0058725
  • Verma MK, Sobha K. Understanding the major risk factors in the beginning and the progression of rheumatoid arthritis: current scenario and future prospects. Inflamm Res. 2015;64(9):647-659. doi:1007/s00011-015-0843-8
  • Hootman JM, Helmick CG, Barbour KE, Theis KA, Boring MA. Updated projected prevalence of self-reported doctor-diagnosed arthritis and arthritis-attributable activity limitation among US adults, 2015-2040. Arthritis Rheumatol. 2016;68(7):1582-1587. doi:1002/art.39692
  • MacGregor AJ, Snieder H, Rigby AS, et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 2000;43(1):30-37. doi:1002/1529-0131(200001)43:1<30::AID-ANR5>3.0.CO;2-B
  • Araki Y, Mimura T. The mechanisms underlying chronic inflammation in rheumatoid arthritis from the perspective of the epigenetic landscape. J Immunol Res. 2016;2016:6290682. doi:1155/2016/6290682
  • Glant TT, Mikecz K, Rauch TA. Epigenetics in the pathogenesis of rheumatoid arthritis. BMC Med. 2014;12:35. doi:1186/1741-7015-12-35
  • Nielen MM, van Schaardenburg D, Reesink HW, et al. Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis Rheum. 2004;50(2):380-386. doi:1002/art.20018
  • Tracy A, Buckley CD, Raza K. Pre-symptomatic autoimmunity in rheumatoid arthritis: when does the disease start? Semin Immunopathol. 2017;39(4):423-435. doi:1007/s00281-017-0620-6
  • Rakieh C, Nam JL, Hunt L, et al. Predicting the development of clinical arthritis in anti-CCP positive individuals with non-specific musculoskeletal symptoms: a prospective observational cohort study. Ann Rheum Dis. 2015;74(9):1659-1666. doi:1136/annrheumdis-2014-205227
  • Ramos-Remus C, Castillo-Ortiz JD, Aguilar-Lozano L, et al. Autoantibodies in prediction of the development of rheumatoid arthritis among healthy relatives of patients with the disease. Arthritis Rheumatol. 2015;67(11):2837-2844. doi:1002/art.39297
  • Deane KD, Demoruelle MK, Kelmenson LB, Kuhn KA, Norris JM, Holers VM. Genetic and environmental risk factors for rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2017;31(1):3-18. doi:1016/j.berh.2017.08.003
  • Harrison M, Spooner L, Bansback N, et al. Preventing rheumatoid arthritis: preferences for and predicted uptake of preventive treatments among high risk individuals. PLoS One. 2019;14(4):e0216075. doi:1371/journal.pone.0216075
  • Hunt L, Emery P. Defining populations at risk of rheumatoid arthritis: the first steps to prevention. Nat Rev Rheumatol. 2014;10(9):521-530. doi:1038/nrrheum.2014.82
  • Paul BJ, Kandy HI, Krishnan V. Pre-rheumatoid arthritis and its prevention. Eur J Rheumatol. 2017;4(2):161-165. doi:5152/eurjrheum.2017.16006
  • Gerlag DM, Norris JM, Tak PP. Towards prevention of autoantibody-positive rheumatoid arthritis: from lifestyle modification to preventive treatment. Rheumatology (Oxford). 2016;55(4):607-614. doi:1093/rheumatology/kev347
  • Silman AJ, Newman J, MacGregor AJ. Cigarette smoking increases the risk of rheumatoid arthritis. Results from a nationwide study of disease-discordant twins. Arthritis Rheum. 1996;39(5):732-735. doi:1002/art.1780390504
  • Stolt P, Bengtsson C, Nordmark B, et al. Quantification of the influence of cigarette smoking on rheumatoid arthritis: results from a population based case-control study, using incident cases. Ann Rheum Dis. 2003;62(9):835-841. doi:1136/ard.62.9.835
  • Hutchinson D, Shepstone L, Moots R, Lear JT, Lynch MP. Heavy cigarette smoking is strongly associated with rheumatoid arthritis (RA), particularly in patients without a family history of RA. Ann Rheum Dis. 2001;60(3):223-227. doi:1136/ard.60.3.223
  • Anderson R, Meyer PW, Ally MM, Tikly M. Smoking and air pollution as pro-inflammatory triggers for the development of rheumatoid arthritis. Nicotine Tob Res. 2016;18(7):1556-1565. doi:1093/ntr/ntw030
  • Kharlamova N, Jiang X, Sherina N, et al. Antibodies to Porphyromonas gingivalis indicate interaction between oral infection, smoking, and risk genes in rheumatoid arthritis etiology. Arthritis Rheumatol. 2016;68(3):604-613. doi:1002/art.39491
  • Pretorius E, Akeredolu OO, Soma P, Kell DB. Major involvement of bacterial components in rheumatoid arthritis and its accompanying oxidative stress, systemic inflammation and hypercoagulability. Exp Biol Med (Maywood). 2017;242(4):355-373. doi:1177/1535370216681549
  • Lu B, Hiraki LT, Sparks JA, et al. Being overweight or obese and risk of developing rheumatoid arthritis among women: a prospective cohort study. Ann Rheum Dis. 2014;73(11):1914-1922. doi:1136/annrheumdis-2014-205459
  • Sparks JA, Chang SC, Nguyen US, et al. Weight change during the early rheumatoid arthritis period and risk of subsequent mortality in women with rheumatoid arthritis and matched comparators. Arthritis Rheumatol. 2018;70(1):18-29. doi:1002/art.40346
  • Baker JF, Von Feldt J, Mostoufi-Moab S, et al. Deficits in muscle mass, muscle density, and modified associations with fat in rheumatoid arthritis. Arthritis Care Res (Hoboken). 2014;66(11):1612-1618. doi:1002/acr.22328
  • Hanaoka BY, Ithurburn MP, Rigsbee CA, et al. Chronic inflammation in rheumatoid arthritis and mediators of skeletal muscle pathology and physical impairment: a review. Arthritis Care Res (Hoboken). 2019;71(2):173-177. doi:1002/acr.23775
  • Bärebring L, Winkvist A, Gjertsson I, Lindqvist HM. Poor dietary quality is associated with increased inflammation in Swedish patients with rheumatoid arthritis. Nutrients. 2018;10(10):E1535. doi:3390/nu10101535
  • Essouma M, Noubiap JJ. Is air pollution a risk factor for rheumatoid arthritis? J Inflamm (Lond). 2015;12:48. doi:1186/s12950-015-0092-1
  • Jung CR, Hsieh HY, Hwang BF. Air pollution as a potential determinant of rheumatoid arthritis: a population-based cohort study in Taiwan. 2017;28(Suppl 1):S54-S59. doi:10.1097/EDE.0000000000000732
  • Shin J, Lee J, Lee J, Ha EH. Association between exposure to ambient air pollution and rheumatoid arthritis in adults. Int J Environ Res Public Health. 2019;16(7):E1227. doi:3390/ijerph16071227
  • Sigaux J, Biton J, André E, Semerano L, Boissier MC. Air pollution as a determinant of rheumatoid arthritis. Joint Bone Spine. 2019;86(1):37-42. doi:1016/j.jbspin.2018.03.001
  • Araki Y, Mimura T. The mechanisms underlying chronic inflammation in rheumatoid arthritis from the perspective of the epigenetic landscape. J Immunol Res. 2016;2016:6290682. doi:1155/2016/6290682
  • Zaccardelli A, Friedlander HM, Ford JA, Sparks JA. Potential of lifestyle changes for reducing the risk of developing rheumatoid arthritis: is an ounce of prevention worth a pound of cure? Clin Ther. 2019;41(7):1323-1345. doi:1016/j.clinthera.2019.04.021
  • He J, Wang Y, Feng M, et al. Dietary intake and risk of rheumatoid arthritis—a cross section multicenter study. Clin Rheumatol. 2016;35(12):2901-2908. doi:1007/s10067-016-3383-x
  • Vadell AKE, Bärebring L, Hulander E, Gjertsson I, Lindqvist HM, Winkvist A. Anti-inflammatory diet in rheumatoid arthritis (ADIRA) – a randomized, controlled crossover trial indicating effects on disease activity. Am J Clin Nutri. 2020;111(6):1203-1213. doi:1093/ajcn/nqaa019
  • Philippou E, Nikiphorou E. Are we really what we eat? Nutrition and its role in the onset of rheumatoid arthritis. Autoimmun Rev. 2018;17(11):1074-1077. doi:1016/j.autrev.2018.05.009
  • Gioia C, Lucchino B, Tarsitano MG, Iannuccelli C, Di Franco M. Dietary habits and nutrition in rheumatoid arthritis: can diet influence disease development and clinical manifestations? Nutrients. 2020;12(5):1456. doi:3390/nu12051456
  • Asteriou E, Gkoutzourelas A, Mavropoulos A, Katsiari C, Sakkas LI, Bogdanos DP. Curcumin for the management of periodontitis and early ACPA-positive rheumatoid arthritis: killing two birds with one stone. 2018;10(7):E908. doi:10.3390/nu10070908
  • Schneider MR, Schmidt-Ullrich R, Paus R (2009) The hair follicle as a dynamic miniorgan. Curr Biol 19: R132-R142.
  • Almohanna HM, Ahmed AA, Tsatalis JP, Tosti A (2019) The Role of Vitamins and Minerals in Hair Loss: A Review. Dermatol Ther (Heidelb) 9: 51-70.
  • Guo EL, Katta R (2017) Diet and hair loss: Effects of nutrient deficiency and supplement use. Dermatol Pract Concept 7: 1-10.
  • Finner AM (2013) Nutrition and hair: Deficiencies and supplements. Dermatol Clin 31: 167-172.
  • Singh RK, Chang HW, Yan D, Lee KM, Ucmak D, et al. (2017) Influence of diet on the gut microbiome and implications for human health. J Transl Med 15: 73.
  • Scott KP, Gratz SW, Sheridan PO, Flint HJ, Duncan SH (2013) The influence of diet on the gut microbiota. Pharmacol Res 69: 52-60.
  • Vaughn AR, Notay M, Clark AK, Sivamani RK (2017) Skin-gut axis: The relationship between intestinal bacteria and skin health. World J Dermatol 6: 52-58.
  • Bowe WP, Joshi SS, Shalita AR (2010) Diet and acne. J Am Acad Dermatol 63: 124-141.
  • Dawber R (1989) Alopecia areata. Monogr Dermatol 2: 89-102.
  • Odom RB, Davidsohn IJ, William D, Henry JB, Berger TG (2006) Clinical diagnosis by laboratory methods. In: Elston Dirk M, Andrews’ Diseases of the Skin: Clinical Dermatology. Saunders Elsevier.
  • Brenner W, Diem E, Gschnait F (1979) Coincidence of vitiligo, alopecia areata, onychodystrophy, localized scleroderma and lichen planus. Dermatologica 159: 356-360.
  • Trink A, Sorbellini E, Bezzola P, Rodella L, Rezzani R, et al. (2013) A randomized, double-blind, placebo- and active-controlled, half-head study to evaluate the effects of platelet-rich plasma on alopecia areata. Br J Dermatol 169: 690-694.
  • Clavaud C, Jourdain R, Bar-Hen A, Magali Tichit, Christiane Bouchier, et al. (2013) Dandruff is associated with disequilibrium in the proportion of the major bacterial and fungal populations colonizing the scalp. PLoS One 8: e58203.
  • Rinaldi F, Pinto D, Marzani B, Rucco M, Giuliani G, et al. (2018) Human microbiome: What’s new in scalp diseases. J Transl Sci 4: 1-4.
  • Pinto D, Sorbellini E, Marzani B, Rucco M, Giuliani G, et al. (2019) Scalp bacterial shift in Alopecia areata. PLoS One 14: e0215206.
  • Ho BS, Ho EXP, Chu CW, Ramasamy S, Bigliardi-Qi M, et al. (2019) Microbiome in the hair follicle of androgenetic alopecia patients. PLoS One 14: e0216330.
  • Polak-Witka K, Rudnicka L, Blume-Peytavi U, Vogt A (2019) The role of the microbiome in scalp hair follicle biology and disease. Exp Dermatol.
  • L Nair, Z Dai, AM Christiano (2017) 649 Gut microbiota plays a role in the development of alopecia areata. Journal of Investigative Dermatology 137: S112.
  • Olsen EA, Hordinsky MK, Price VH, Roberts JL, Shapiro J, et al. (2004) Alopecia areata investigational assessment guidelines–Part II. National Alopecia Areata Foundation. J Am Acad Dermatol 51: 440-447.
  • Grice EA, Kong HH, Conlan S, Deming CB, Davis J, et al. (2010) Topographical and temporal diversity of the human skin microbiome. Science 324: 1190-1192.
  • Paulino LC, Tseng CH, Strober BE, Blaser MJ (2006) Molecular analysis of fungal microbiota in samples from healthy human skin and psoriatic lesions. J Clin Microbiol 44: 2933-2941.
  • Gao Z, Perez-Perez GI, Chen Y, Blaser MJ (2010) Quantitation of major human cutaneous bacterial and fungal populations. J Clin Microbiol 48: 3575-3581.
  • Klindworth A, Pruesse E, Schweer T, Jörg Peplies, Christian Quast, et al. (2013) Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res 41: e1.
  • Takahashi S, Tomita J, Nishioka K, Hisada T, Nishijima M (2014) Development of a prokaryotic universal primer for simultaneous analysis of bacteria and archaea using next-generation sequencing. PLoS One 9: e105592.
  • Apprill A, McNally S, Parsons R, Weber L (2015) Minor revision to V4 region SSU rRNA 806R gene primer greatly increases detection of SAR11 bacterioplankton. Aquat Microb Ecol 75: 129-137.
  • Parada AE, Needham DM, Fuhrman JA (2016) Every base matters: assessing small subunit rRNA primers for marine microbiomes with mock communities, time series and global field samples. Environ Microbiol 18: 1403-1414.
  • Walters W, Hyde ER, Berg-Lyons D, Ackermann G, Humphrey G, et al. (2015) Improved bacterial 16S rRNA Gene (V4 and V4-5) and fungal internal transcribed spacer marker gene primers for microbial community surveys. mSystems 1.
  • Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, et al. (2011) Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci USA 108: 4516-4522.
  • Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD (2013) Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microbiol 79: 5112-5120.
  • Vigetti D, Viola M, Karousou E, Rizzi M , Moretto P, et al. (2008) Hyaluronan-CD44-ERK1/2 regulate human aortic smooth muscle cell motility during aging. J Biol Chem 283: 4448-4458.
  • Castro-Quezada I, Román-Viñas B, Serra-Majem L (2014) The mediterranean diet and nutritional adequacy: A review. Nutrients 6: 231-248.
  • Rushton DH (2002) Nutritional factors and hair loss. Clin Exp Dermatol 27: 396-404.
  • Mubki T, Rudnicka L, Olszewska M, Shapiro J (2014) Evaluation and diagnosis of the hair loss patient: Part I. History and clinical examination. J Am Acad Dermatol 71: 415.
  • Spivak JL, Jackson DL (1997) Pellagra: An analysis of 18 patients and a review of the literature. Johns Hopkins Med J 140: 295-309.
  • Goldberg LJ, Lenzy Y (2010) Nutrition and hair. Clin Dermatol 28: 412-419.
  • Kato I, Vasquez A, Moyerbrailean G, Land S, Djuric Z, et al. (2017) Nutritional correlates of human oral microbiome. J Am Coll Nutr 36: 88-98.
  • Manam S, Tsakok T, Till S, Flohr C (2014) The association between atopic dermatitis and food allergy in adults. Curr Opin Allergy Clin Immunol 14: 423-429.
  • Cordain L, Lindeberg S, Hurtado M, Hill K, Eaton SB, et al. (2002) Acne vulgaris: A disease of Western civilization. Arch Dermatol. 138: 1584-1590.
  • Grossi E, Cazzaniga S, Crotti S, Naldi L, Di Landro A, et al. (2016) The constellation of dietary factors in adolescent acne: A semantic connectivity map approach. J Eur Acad Dermatol Venereol 30: 96-100.
  • Zouboulis CC, Jourdan E, Picardo M (2014) Acne is an inflammatory disease and alterations of sebum composition initiate acne lesions. J Eur Acad Dermatol Venereol 28: 527-532.
  • Zákostelská Z, Málková J, Klimešová K, Pavel Rossmann, Michaela Hornová, et al. (2016) Intestinal microbiota promotes psoriasis-like skin inflammation by enhancing Th17 response. PLoS One 11: e0159539.
  • Zhang C, Zhang M, Wang S, Han R, Cao Y, et al. (2010) Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J 4: 232-241.
  • Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI (2008) Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 3: 213-223.
  • Mu Q, Kirby J, Reilly CM, Luo XM, (2017) Leaky gut as a danger signal for autoimmune diseases. Front Immunol 8: 598.
  • Sartor RB. Mechanisms of disease: pathogenesis of Crohn’s disease and ulcerative colitis. Nat Clin Pract Gastroenterol Hepatol. 2006;3(7): 390–407.
  • Itta M. Minderhoud, Bas Oldenburg, Marguerite E.I. Schipper, Jose – J.M. ter Linde, Melvin Samson. Serotonin Synthesis and Uptake in Symptomatic Patients with Crohn’s Disease in Remission. Clinical Gastroenterology and Hepatology. June 2007
  • R. Spiller. Recent advances in understanding the role of serotonin in gastrointestinal motility in functional bowel disorders: alterations in 5-HT signaling and metabolism in human disease. Neurogastroenterology and Motility. Jul. 2007
  • Jean-Eric Ghia, Nan Li, Huaqing Wang et al. Serotonin has a key role in pathogenesis of experimental colitis. Gastroenterology. Nov 2009
  • Stein, A., Hinz, M., & Uncini, T. (2010). Amino acid-responsive Crohn’s disease: a case study. Clinical and experimental gastroenterology, 3, 171.
  • Hinz, M., Stein, A., & Uncini, T. (2012). Relative nutritional deficiencies associated with centrally acting monoamines. Int J Gen Med, 5, 413-430.Loddo I, Romano C. Inflammatory bowel disease: genetics, epigenetics, and pathogenesis. Frontiers in immunology. 2015 Nov 2;6:551. Ni J, Wu GD, Albenberg L, Tomov VT. Gut microbiota and IBD: causation or correlation?. Nature reviews Gastroenterology & hepatology. 2017 Oct;14(10):573. 
  • Liu TC, Stappenbeck TS. Genetics and pathogenesis of inflammatory bowel disease. Annual Review of Pathology: Mechanisms of Disease. 2016 May 23;11:127-48. 
  • De Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nature reviews Gastroenterology & hepatology. 2016 Jan;13(1):13. 
  • Davies JM, Abreu MT. The innate immune system and inflammatory bowel disease. Scandinavian Journal of Gastroenterology. 2015 Jan 2;50(1):24-33. 
  • Zhao M, Burisch J. Impact of genes and the environment on the pathogenesis and disease course of inflammatory bowel disease. Digestive diseases and sciences. 2019 Jul 15;64(7):1759-69. 
  • Knight-Sepulveda K, Kais S, Santaolalla R, Abreu MT. Diet and inflammatory bowel disease. Gastroenterology & hepatology. 2015 Aug;11(8):511. 
  • Lewis JD, Abreu MT. Diet as a trigger or therapy for inflammatory bowel diseases. Gastroenterology. 2017 Jan 1;152(2):398-414. 
  • Bernstein CN. Psychological stress and depression: risk factors for IBD?. Digestive Diseases. 2016;34(1-2):58-63. 
  • Bernstein CN. The brain-gut axis and stress in inflammatory bowel disease. Gastroenterology Clinics. 2017 Dec 1;46(4):839-46. 
  • Tontini GE, Vecchi M, Pastorelli L, Neurath MF, Neumann H. Differential diagnosis in inflammatory bowel disease colitis: state of the art and future perspectives. World journal of gastroenterology: WJG. 2015 Jan 7;21(1):21.  Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nature reviews Gastroenterology & hepatology. 2015 Apr;12(4):205-17. 
  • Pituch-Zdanowska A, Banaszkiewicz A, Albrecht P. The role of dietary fibre in inflammatory bowel disease. Przeglad gastroenterologiczny. 2015;10(3):135. 
  1. Dharmage SC, Lowe AJ, Matheson MC, Burgess JA, Allen KJ, Abramson MJ. Atopic dermatitis and the atopic march revisited. Allergy. 2014;69(1):17-27. doi:10.1111/all.12268
  2. Kapoor R, Menon C, Hoffstad O, Bilker W, Leclerc P, Margolis DJ. The prevalence of atopic triad in children with physician-confirmed atopic dermatitis. J Am Acad Dermatol. 2008;58(1):68-73. doi:10.1016/j.jaad.2007.06.041
  3. Thyssen JP, Hamann CR, Linneberg A, et al. Atopic dermatitis is associated with anxiety, depression, and suicidal ideation, but not with psychiatric hospitalization or suicide. Allergy. 2018;73(1):214-220. doi:10.1111/all.13231
  4. Brunner PM, Silverberg JI, Guttman-Yassky E, et al. Increasing comorbidities suggest that atopic dermatitis is a systemic disorder. J Invest Dermatol. 2017;137(1):18-25. doi:10.1016/j.jid.2016.08.022
  5. Strom MA, Silverberg JI. Associations of physical activity and sedentary behavior with atopic disease in United States children. J Pediatr. 2016;174:247-253.e3. doi:10.1016/j.jpeds.2016.03.063
  6. Zheng T, Yu J, Oh MH, Zhu Z. The atopic march: progression from atopic dermatitis to allergic rhinitis and asthma. Allergy Asthma Immunol Res. 2011;3(2):67-73. doi:10.4168/aair.2011.3.2.67
  7. Huang R, Ning H, Shen M, Li J, Zhang J, Chen X. Probiotics for the treatment of atopic dermatitis in children: a systematic review and meta-analysis of randomized controlled trials. Front Cell Infect Microbiol. 2017;7:392. doi:10.3389/fcimb.2017.00392
  8. Rusu E, Enache G, Cursaru R, et al. Prebiotics and probiotics in atopic dermatitis. Exp Ther Med. 2019;18(2):926-931. doi:10.3892/etm.2019.7678
  9. Foolad N, Armstrong AW. Prebiotics and probiotics: the prevention and reduction in severity of atopic dermatitis in children. Benef Microbes. 2014;5(2):151-160. doi:10.3920/BM2013.0034
  10. Ahn K. The role of air pollutants in atopic dermatitis. J Allergy Clin Immunol. 2014;134(5):993-1000. doi:10.1016/j.jaci.2014.09.023
  11. Han H, Roan F, Ziegler SF. The atopic march: current insights into skin barrier dysfunction and epithelial cell-derived cytokines. Immunol Rev. 2017;278(1):116-130. doi:10.1111/imr.12546
  • Autoimmune diseases. National Institute of Allergy and Infectious Diseases. Last reviewed May 2, 2017. Accessed June 4, 2020. https://www.niaid.nih.gov/diseases-conditions/autoimmune-diseases
  • Hood E. Measuring autoimmunity in America. Environmental Factor. Published April 2018. Accessed June 4, 2020. https://factor.niehs.nih.gov/2018/4/science-highlights/autoimmunity/index.htm
  • Autoimmune diseases. National Institute of Environmental Health Sciences. Last Reviewed May 6, 2020. Accessed June 4, 2020. https://www.niehs.nih.gov/health/topics/conditions/autoimmune/index.cfm – footnote1
  • Marker of autoimmunity increases in the U.S. National Institutes of Health. Published April 21, 2020. Accessed August 19, 2020. https://www.nih.gov/news-events/nih-research-matters/marker-autoimmunity-increases-us
  • Roberts MH, Erdei E. Comparative United States autoimmune disease rates for 2010-2016 by sex, geographic region, and race. Autoimmun Rev. 2020;19(1):102423. doi:10.1016/j.autrev.2019.102423
  • The Autoimmune Diseases Coordinating Committee. Progress in Autoimmune Diseases Research: Report to Congress. National Institutes of Health; 2005. Accessed August 31, 2020. https://www.niaid.nih.gov/sites/default/files/adccfinal.pdf
  • Somers EC, Marder W, Cagnoli P, et al. Population-based incidence and prevalence of systemic lupus erythematosus: the Michigan Lupus Epidemiology and Surveillance program. Arthritis Rheumatol. 2014;66(2):369-378. doi:10.1002/art.38238
  • Ramos PS, Shedlock AM, Langefeld CD. Genetics of autoimmune diseases: insights from population genetics. J Hum Genet. 2015;60(11):657-664. doi:10.1038/jhg.2015.94
  • Dinse GE, Parks CG, Weinberg CR, et al. Increasing prevalence of antinuclear antibodies in the United States. Arthritis Rheumatol. 2020;72(6):1026-1035. doi:10.1002/art.41214
  • American College of Rheumatology. Antinuclear antibodies (ANA). Updated March 2019. Accessed August 20, 2020. https://www.rheumatology.org/I-Am-A/Patient-Caregiver/Diseases-Conditions/Antinuclear-Antibodies-ANA
  • Aringer M, Costenbader K, Daikh D, et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Ann Rheum Dis. 2019;78(9):1151-1159. doi:10.1136/annrheumdis-2018-214819
  • Rösken GHJ, van Beek AA, Bakker-Jonges LE, Schreurs MWJ. Antinuclear antibodies in systemic autoimmune disease. Ned Tijdschr Geneeskd. 2020;164:D4066.
  • Pérez D, Gilburd B, Cabrera-Marante Ó, et al. Predictive autoimmunity using autoantibodies: screening for anti-nuclear antibodies. Clin Chem Lab Med. 2018;56(10):1771-1777. doi:10.1515/cclm-2017-0241
  • Bloch DB. Patient education: antinuclear antibodies (ANA) (beyond the basics). UpToDate. Updated December 18, 2019. Accessed August 28, 2020. https://www.uptodate.com/contents/antinuclear-antibodies-ana-beyond-the-basics
  • Tan EM, Feltkamp TE, Smolen JS, et al. Range of antinuclear antibodies in “healthy” individuals. Arthritis Rheum. 1997;40(9):1601-1611. doi:10.1002/art.1780400909
  • Lyons R, Narain S, Nichols C, Satoh M, Reeves WH. Effective use of autoantibody tests in the diagnosis of systemic autoimmune disease. Ann N Y Acad Sci. 2005;1050:217-228. doi:10.1196/annals.1313.023
  • Nancy AL, Yehuda S. Prediction and prevention of autoimmune skin disorders. Arch Dermatol Res. 2009;301(1):57-64. doi:10.1007/s00403-008-0889-3
  • Ramos-Remus C, Castillo-Ortiz JD, Aguilar-Lozano L, et al. Autoantibodies in prediction of the development of rheumatoid arthritis among healthy relatives of patients with the disease. Arthritis Rheumatol. 2015;67(11):2837-2844. doi:10.1002/art.39297
  • Rakieh C, Nam JL, Hunt L, et al. Predicting the development of clinical arthritis in anti-CCP positive individuals with non-specific musculoskeletal symptoms: a prospective observational cohort study. Ann Rheum Dis. 2015;74(9):1659-1666. doi:10.1136/annrheumdis-2014-205227
  • Rose NR. Prediction and prevention of autoimmune disease in the 21st century: a review and preview. Am J Epidemiol. 2016;183(5):403-406. doi:10.1093/aje/kwv292
  • Choi MY, Fritzler MJ. Autoantibodies in SLE: prediction and the pvalue matrix. Lupus. 2019;28(11):1285-1293. doi:10.1177/0961203319868531
  • Mu Q, Kirby J, Reilly CM, Luo XM. Leaky gut as a danger signal for autoimmune diseases. Front Immunol. 2017;8:598. doi:10.3389/fimmu.2017.00598
  • Kharaeva Z1, Gostova E, De Luca C, Raskovic D, Korkina L.Clinical and biochemical effects of coenzyme Q10, vitamin E, and selenium supplementation to psoriasis patients.1.Nutrition. 2009 Mar;25(3):295-302. 
  • EFFECTIVENESS OF PROBiotics APPLICATION IN COMPLEX TREATMENT OF PSORIASIS.2 Millsop JW1, Bhatia BK2, Debbaneh M3, Koo J4, Liao W5.Diet and Psoriasis: Part 3. Role of Nutritional Supplements.J Am Acad Dermatol. 2014 Sep;71(3):561-9.
  •  Bhatia BK1, Millsop JW2, Debbaneh M3, Koo J4, Linos E4, Liao W5.Diet and Psoriasis: Part 2. Celiac Disease and Role of a Gluten-Free Diet.J Am Acad Dermatol. 2014 Aug;71(2):350-8.
  •  Brazzelli V1, Grasso V, Fornara L, Moggio E, Gamba G, Villani S, Borroni G.Homocysteine, vitamin B12 and folic acid levels in psoriatic patients and correlation with disease severity.Int J Immunopathol Pharmacol. 2010 Jul-Sep;23(3):911-6.
  • Sardu C, Cocco E, Mereu A, et al. Population based study of 12 autoimmune diseases in Sardinia, Italy: prevalence and comorbidity. PLoS One. 2012;7(3):e32487. doi:10.1371/journal.pone.0032487.
  • Wu CH, Chung PI, Wu CY, et al. Comorbid autoimmune diseases in patients with sarcoidosis: a nationwide case-control study in Taiwan. J Dermatol. 2017;44(4):423-430. doi:10.1111/1346-8138.13654.
  • Ruffilli I, Ragusa F, Benvenga S, et al. Psoriasis, psoriatic arthritis, and thyroid autoimmunity. Front Endocrinol. 2017;8:139. doi:10.3389/fendo.2017.00139.
  • Antonelli A, Delle Sedie A, Fallahi P, et al. High prevalence of thyroid autoimmunity and hypothyroidism in patients with psoriatic arthritis. J Rheumatol. 2006;33(10):2026-2028.
  • Furue M, Kadono T. “Inflammatory skin march” in atopic dermatitis and psoriasis [published online June 15, 2017]. Inflamm Res. doi:10.1007/s00011-017-1065-z.
  • Binus AM, Han J, Qamar AA, Mody EA, Holt EW, Qureshi AA. Associated comorbidities in psoriasis and inflammatory bowel disease. J Eur Acad Dermatol Venereol. 2012;26(5):644-650. doi:10.1111/j.1468-3083.2011.04153.x.
  • Hsu LN, Armstrong AW. Psoriasis and autoimmune disorders: a review of the literature. J Am Acad Dermatol. 2012;67(5):1076-1079. doi:10.1016/j.jaad.2012.01.029.
  • https://www.mayoclinic.org/diseases-conditions/psoriasis/symptoms-causes/syc-20355840.
  • https://www.aad.org/public/diseases/scaly-skin/psoriasis/who-gets-psoriasis-and-what-causes-it.
  • https://www.aad.org/public/diseases/scaly-skin/psoriasis/what-is-psoriasis.
  • https://www.mayoclinic.org/diseases-conditions/psoriasis/symptoms-causes/syc-20355840.
  • https://www.aad.org/public/diseases/scaly-skin/psoriasis/who-gets-psoriasis-and-what-causes-it.
  • https://www.niams.nih.gov/health-topics/psoriasis#tab-symptoms.
  • https://www.aad.org/public/diseases/scaly-skin/psoriasis/psoriasis-signs-and-symptoms/psoriasis-signs-and-symptoms.
  • https://www.mayoclinic.org/diseases-conditions/psoriasis/symptoms-causes/syc-20355840.
  • https://www.sciencedirect.com/science/article/pii/S019096229970112X.
  • https://www.psoriasis.org/about-psoriasis/specific-locations/scalp.
  • https://www.niams.nih.gov/health-topics/psoriasis#tab-types.
  • https://www.mayoclinic.org/diseases-conditions/psoriasis/symptoms-causes/syc-20355840.
  • https://www.aad.org/public/diseases/scaly-skin/psoriasis/diagnosis-and-treatment-of-psoriasis.
  • https://www.mayoclinic.org/diseases-conditions/psoriasis/diagnosis-treatment/drc-20355845.
  • https://www.niams.nih.gov/health-topics/psoriasis#tab-treatment.
  • https://www.aad.org/public/diseases/scaly-skin/psoriasis/diagnosis-and-treatment-of-psoriasis.
  • https://www.mayoclinic.org/diseases-conditions/psoriasis/diagnosis-treatment/drc-20355845.
  • https://www.mayoclinic.org/diseases-conditions/psoriasis/diagnosis-treatment/drc-20355845.
  • https://www.ncbi.nlm.nih.gov/pubmed/22109896.
  • https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4290643/.
  • https://www.ncbi.nlm.nih.gov/pubmed/12505286.
  • Moutsopoulos HM. Sjögren’s syndrome: autoimmune epithelitis. Clin Immunol Immunopathol (1994) 72(2):162–5.10.1006/clin.1994.1123 [PubMed] [CrossRef] [Google Scholar]
  • Ramos-Casals M, Brito-Zerón P, Kostov B, Sisó-Almirall A, Bosch X, Buss D, et al. Google-driven search for big data in autoimmune geoepidemiology: analysis of 394,827 patients with systemic autoimmune diseases. Autoimmun Rev (2015) 14(8):670–9.10.1016/j.autrev.2015.03.008 [PubMed] [CrossRef] [Google Scholar]
  • Humphreys-Beher MG, Brinkley L, Purushotham KR, Wang PL, Dusek D, Nakagawa Y, et al. Characterization of antinuclear autoantibodies present in the serum from nonobese diabetic (NOD) mice. Clin Immunol Immunopathol (1993) 68:350–6.10.1006/clin.1993.1137 [PubMed] [CrossRef] [Google Scholar]
  • Humphreys-Beher MG, Hu Y, Nakagawa Y, Wang PL, Purushotham KR. Utilization of the non-obese diabetic (NOD) mouse as an animal model for the study of secondary Sjogren’s syndrome. Adv Exp Med Biol (1994) 350:631–6.10.1007/978-1-4615-2417-5_105 [PubMed] [CrossRef] [Google Scholar]
  • Robinson CP, Yamamoto H, Peck AB, Humphreys-Beher MG. Genetically programmed development of salivary gland abnormalities in the NOD (nonobese diabetic)-scid mouse in the absence of detectable lymphocytic infiltration: a potential trigger for sialoadenitis of NOD mice. Clin Immunol Immunopathol (1996) 79:50–9.10.1006/clin.1996.0050 [PubMed] [CrossRef] [Google Scholar]
  • de Paiva CS, Jones DB, Stern ME, Bian F, Moore QL, Corbiere S, et al. Altered mucosal microbiome diversity and disease severity in Sjögren syndrome. Sci Rep (2016) 6:23561.10.1038/srep23561 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Spachidou MP, Bourazopoulou E, Maratheftis CI, Kapsogeorgou EK, Moutsopoulos HM, Tzioufas AG. Expression of functional toll-like receptors by salivary gland epithelial cells: increased mRNA expression in cells derived from patients with primary Sjogren’s syndrome. Clin Exp Immunol (2007) 147:497–503.10.1111/j.1365-2249.2006.03311.x [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Garcia-Carrasco M, Fuentes-Alexandro S, Escarcega RO, Salgado G, Riebeling C, Cervera R. Pathophysiology of Sjogren’s syndrome. Arch Med Res (2006) 37:921–32.10.1016/j.arcmed.2006.08.002 [PubMed] [CrossRef] [Google Scholar]
  • Daridon C, Devauchelle V, Hutin P, Berre RL, Martins-Carvalho C, Bendaoud B. Aberrant expression of BAFF by B lymphocytes infiltrating the salivary glands of patients with primary Sjögren’s syndrome. Arthritis Rheum (2007) 56:1134–44.10.1002/art.22458 [PubMed] [CrossRef] [Google Scholar]
  • Groom J, Kalled SL, Cutler AH, Olson C, Woodcock SA, Schneider P. Association of BAFF/BLyS overexpression and altered B cell differentiation with Sjogren’s syndrome. J Clin Invest (2002) 109:59–68.10.1172/JCI0214121 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Vlachogiannis NI, Nezos A, Tzioufas AG, Koutsilieris M, Moutsopoulos HM, Mavragani CP. Increased frequency of the PTPN22W* variant in primary Sjogren’s Syndrome: association with low type I IFN scores. Clin Immunol (2016) 173:157–60.10.1016/j.clim.2016.10.015 [PubMed] [CrossRef] [Google Scholar]
  • Johnsen SJ, Gudlaugsson E, Skaland I, Janssen EA, Jonsson MV, Helgeland L, et al. Low protein A20 in minor salivary glands is associated with lymphoma in primary Sjögren’s Syndrome. Scand J Immunol (2016) 83(3):181–7.10.1111/sji.12405 [PubMed] [CrossRef] [Google Scholar]
  • Williams AEG, Choi K, Chan AL, Lee YJ, Reeves WH, Bubb MR, et al. Sjögren’s syndrome-associated microRNAs in CD14+ monocytes unveils targeted TGFβ signaling. Arthritis Res Ther (2016) 18:95.10.1186/s13075-016-0987-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cole MB, Quach H, Quach D, Baker A, Taylor KE, Barcellos LF, et al. Epigenetic signatures of salivary gland inflammation in Sjögren’s syndrome. Arthritis Rheumatol (2016) 68(12):2936–44.10.1002/art.39792 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang J, Peng H, Tian J, Ma J, Tang X, Rui K, et al. Upregulation of long noncoding RNA TMEVPG1 enhances T helper type 1 cell response in patients with Sjögren syndrome. Immunol Res (2016) 64(2):489.10.1007/s12026-015-8715-4 [PubMed] [CrossRef] [Google Scholar]
  • Nair JJ, Singh TP. Sjogren’s syndrome: review of the aetiology, pathophysiology & potential therapeutic interventions. J Clin Exp Dent (2017) 9(4):e584–9.10.4317/jced.53605 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sardu C, Cocco E, Mereu A, et al. Population based study of 12 autoimmune diseases in Sardinia, Italy: prevalence and comorbidity. PLoS One. 2012;7(3):e32487. doi:10.1371/journal.pone.0032487.
  • Blackwell D, Clarke TC. QuickStats: percentage of adults who often felt very tired or exhausted in the past 3 months, by sex and age group – National Health Interview Survey, United States, 2010-2011. MMWR Morb Mortal Wkly Rep. 2013;62(14);275. https://www.cdc.gov/mmwr/preview/mmwrhtml/mm6214a5.htm
  • Moore P. Two-fifths of Americans are tired most of the week. YouGov. Published June 2, 2015. Accessed May 30, 2019. https://today.yougov.com/topics/lifestyle/articles-reports/2015/06/02/sleep-and-dreams
  • 43 percent of Americans admit they’re too tired to function at work. Occupational Health & Safety. Published July 27, 2017. Accessed May 31, 2019. https://ohsonline.com/articles/2017/07/27/43-percent-of-americans-admit-they-are-too-tired-to-function-at-work.aspx
  • Rosenthal TC, Majeroni BA, Pretorius R, Malik K. Fatigue: an overview. Am Fam Physician. 2008;78(10):1173-1179.
  • Gormon GS, Elson JL, Newman J, et al. Perceived fatigue is highly prevalent and debilitating in patients with mitochondrial disease. Neuromuscul Disord. 2015;25(7):563-566. doi:1016/j.nmd.2015.03.001
  • Gellerich FN, Gizatullina Z, Gainutdinov T, et al. The control of brain mitochondrial energization by cytosolic calcium: the mitochondrial gas pedal. IUBMB Life. 2013;65(3):180-190. doi:1002/iub.1131
  • Filler K, Lyon D, Bennett J, et al. Association of mitochondrial dysfunction and fatigue: a review of the literature. BBA Clin. 2014;1:12-23. doi:1016/j.bbacli.2014.04.001
  • Gimenes AC, Bravo DM, Nápolis LM, et al. Effect of L-carnitine on exercise performance in patients with mitochondrial myopathy. Braz J Med Biol Res. 2015;48(4):354-362. doi:1590/1414-431X20143467
  • Morris G, Berk M, Walder K, Maes M. Central pathways causing fatigue in neuro-inflammatory and autoimmune illnesses. BMC Med. 2015;13:28. doi:1186/s12916-014-0259-2
  • Morris G, Berk M. The many roads to mitochondrial dysfunction in neuroimmune and neuropsychiatric disorders. BMC Med. 2015;13:68. doi:1186/s12916-015-0310-y
  • Mizuno K, Tanaka M, Nozaki S, et al. Antifatigue effects of coenzyme Q10 during physical fatigue. Nutrition. 2008;24(4):293-299. doi:1016/j.nut.2007.12.007
  • Faraut B, Boudjeltia KZ, Vanhamme L, Kerkhofs M. Immune, inflammatory and cardiovascular consequences of sleep restriction and recovery. Sleep Med Rev. 2012;16(2):137-149. doi:1016/j.smrv.2011.05.001
  • Lasselin J, Layé S, Dexpert S, et al. Fatigue symptoms relate to systemic inflammation in patients with type 2 diabetes. Brain Behav Immun. 2012;26(8):1211-1219. doi:1016/j.bbi.2012.03.003
  • Schneider MR, Schmidt-Ullrich R, Paus R (2009) The hair follicle as a dynamic miniorgan. Curr Biol 19: R132-R142.
  • Almohanna HM, Ahmed AA, Tsatalis JP, Tosti A (2019) The Role of Vitamins and Minerals in Hair Loss: A Review. Dermatol Ther (Heidelb) 9: 51-70.
  • Guo EL, Katta R (2017) Diet and hair loss: Effects of nutrient deficiency and supplement use. Dermatol Pract Concept 7: 1-10.
  • Finner AM (2013) Nutrition and hair: Deficiencies and supplements. Dermatol Clin 31: 167-172.
  • Singh RK, Chang HW, Yan D, Lee KM, Ucmak D, et al. (2017) Influence of diet on the gut microbiome and implications for human health. J Transl Med 15: 73.
  • Scott KP, Gratz SW, Sheridan PO, Flint HJ, Duncan SH (2013) The influence of diet on the gut microbiota. Pharmacol Res 69: 52-60.
  • Vaughn AR, Notay M, Clark AK, Sivamani RK (2017) Skin-gut axis: The relationship between intestinal bacteria and skin health. World J Dermatol 6: 52-58.
  • Bowe WP, Joshi SS, Shalita AR (2010) Diet and acne. J Am Acad Dermatol 63: 124-141.
  • Dawber R (1989) Alopecia areata. Monogr Dermatol 2: 89-102.
  • Odom RB, Davidsohn IJ, William D, Henry JB, Berger TG (2006) Clinical diagnosis by laboratory methods. In: Elston Dirk M, Andrews’ Diseases of the Skin: Clinical Dermatology. Saunders Elsevier.
  • Brenner W, Diem E, Gschnait F (1979) Coincidence of vitiligo, alopecia areata, onychodystrophy, localized scleroderma and lichen planus. Dermatologica 159: 356-360.
  • Trink A, Sorbellini E, Bezzola P, Rodella L, Rezzani R, et al. (2013) A randomized, double-blind, placebo- and active-controlled, half-head study to evaluate the effects of platelet-rich plasma on alopecia areata. Br J Dermatol 169: 690-694.
  • Clavaud C, Jourdain R, Bar-Hen A, Magali Tichit, Christiane Bouchier, et al. (2013) Dandruff is associated with disequilibrium in the proportion of the major bacterial and fungal populations colonizing the scalp. PLoS One 8: e58203.
  • Rinaldi F, Pinto D, Marzani B, Rucco M, Giuliani G, et al. (2018) Human microbiome: What’s new in scalp diseases. J Transl Sci 4: 1-4.
  • Pinto D, Sorbellini E, Marzani B, Rucco M, Giuliani G, et al. (2019) Scalp bacterial shift in Alopecia areata. PLoS One 14: e0215206.
  • Ho BS, Ho EXP, Chu CW, Ramasamy S, Bigliardi-Qi M, et al. (2019) Microbiome in the hair follicle of androgenetic alopecia patients. PLoS One 14: e0216330.
  • Polak-Witka K, Rudnicka L, Blume-Peytavi U, Vogt A (2019) The role of the microbiome in scalp hair follicle biology and disease. Exp Dermatol.
  • L Nair, Z Dai, AM Christiano (2017) 649 Gut microbiota plays a role in the development of alopecia areata. Journal of Investigative Dermatology 137: S112.
  • Olsen EA, Hordinsky MK, Price VH, Roberts JL, Shapiro J, et al. (2004) Alopecia areata investigational assessment guidelines–Part II. National Alopecia Areata Foundation. J Am Acad Dermatol 51: 440-447.
  • Grice EA, Kong HH, Conlan S, Deming CB, Davis J, et al. (2010) Topographical and temporal diversity of the human skin microbiome. Science 324: 1190-1192.
  • Paulino LC, Tseng CH, Strober BE, Blaser MJ (2006) Molecular analysis of fungal microbiota in samples from healthy human skin and psoriatic lesions. J Clin Microbiol 44: 2933-2941.
  • Gao Z, Perez-Perez GI, Chen Y, Blaser MJ (2010) Quantitation of major human cutaneous bacterial and fungal populations. J Clin Microbiol 48: 3575-3581.
  • Klindworth A, Pruesse E, Schweer T, Jörg Peplies, Christian Quast, et al. (2013) Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res 41: e1.
  • Takahashi S, Tomita J, Nishioka K, Hisada T, Nishijima M (2014) Development of a prokaryotic universal primer for simultaneous analysis of bacteria and archaea using next-generation sequencing. PLoS One 9: e105592.
  • Apprill A, McNally S, Parsons R, Weber L (2015) Minor revision to V4 region SSU rRNA 806R gene primer greatly increases detection of SAR11 bacterioplankton. Aquat Microb Ecol 75: 129-137.
  • Parada AE, Needham DM, Fuhrman JA (2016) Every base matters: assessing small subunit rRNA primers for marine microbiomes with mock communities, time series and global field samples. Environ Microbiol 18: 1403-1414.
  • Walters W, Hyde ER, Berg-Lyons D, Ackermann G, Humphrey G, et al. (2015) Improved bacterial 16S rRNA Gene (V4 and V4-5) and fungal internal transcribed spacer marker gene primers for microbial community surveys. mSystems 1.
  • Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, et al. (2011) Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci USA 108: 4516-4522.
  • Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD (2013) Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microbiol 79: 5112-5120.
  • Vigetti D, Viola M, Karousou E, Rizzi M , Moretto P, et al. (2008) Hyaluronan-CD44-ERK1/2 regulate human aortic smooth muscle cell motility during aging. J Biol Chem 283: 4448-4458.
  • Castro-Quezada I, Román-Viñas B, Serra-Majem L (2014) The mediterranean diet and nutritional adequacy: A review. Nutrients 6: 231-248.
  • Rushton DH (2002) Nutritional factors and hair loss. Clin Exp Dermatol 27: 396-404.
  • Mubki T, Rudnicka L, Olszewska M, Shapiro J (2014) Evaluation and diagnosis of the hair loss patient: Part I. History and clinical examination. J Am Acad Dermatol 71: 415.
  • Spivak JL, Jackson DL (1997) Pellagra: An analysis of 18 patients and a review of the literature. Johns Hopkins Med J 140: 295-309.
  • Goldberg LJ, Lenzy Y (2010) Nutrition and hair. Clin Dermatol 28: 412-419.
  • Kato I, Vasquez A, Moyerbrailean G, Land S, Djuric Z, et al. (2017) Nutritional correlates of human oral microbiome. J Am Coll Nutr 36: 88-98.
  • Manam S, Tsakok T, Till S, Flohr C (2014) The association between atopic dermatitis and food allergy in adults. Curr Opin Allergy Clin Immunol 14: 423-429.
  • Cordain L, Lindeberg S, Hurtado M, Hill K, Eaton SB, et al. (2002) Acne vulgaris: A disease of Western civilization. Arch Dermatol. 138: 1584-1590.
  • Grossi E, Cazzaniga S, Crotti S, Naldi L, Di Landro A, et al. (2016) The constellation of dietary factors in adolescent acne: A semantic connectivity map approach. J Eur Acad Dermatol Venereol 30: 96-100.
  • Zouboulis CC, Jourdan E, Picardo M (2014) Acne is an inflammatory disease and alterations of sebum composition initiate acne lesions. J Eur Acad Dermatol Venereol 28: 527-532.
  • Zákostelská Z, Málková J, Klimešová K, Pavel Rossmann, Michaela Hornová, et al. (2016) Intestinal microbiota promotes psoriasis-like skin inflammation by enhancing Th17 response. PLoS One 11: e0159539.
  • Zhang C, Zhang M, Wang S, Han R, Cao Y, et al. (2010) Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J 4: 232-241.
  • Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI (2008) Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 3: 213-223.
  • Mu Q, Kirby J, Reilly CM, Luo XM, (2017) Leaky gut as a danger signal for autoimmune diseases. Front Immunol 8: 598.
  • Sachdeva A, Cannon CP, Deedwania PC, et al. Lipid levels in patients hospitalized with coronary artery disease: an analysis of 136,905 hospitalizations in get with the guidelines. Am Heart J. 2009;157(1):111–117.
  • Nishikura T, Koba S, Yokota Y, et al. Elevated small dense low-density lipoprotein cholesterol as a predictor for future cardiovascular events in patients with stable coronary artery disease. J Atheroscler Thromb. 2014;21:755–767. doi:10.5551/jat.23465
  • Hoogeveen RC, Gaubatz JW, Sun W, et al. Small dense low-density lipoprotein-cholesterol concentrations predict risk for coronary heart disease: the atherosclerosis risk in communities (aric) study. Arterioscler Thromb Vasc Biol. 2014;34:1069–1077. doi:10.1161/ATVBAHA.114.303284
  • Austin MA. Small, dense low-density-lipoprotein as a risk factor for coronary heart-disease. Int J Clin Lab Res. 1994;24:187–192. doi:10.1007/BF02592460
  • Blake GJ. Low-density lipoprotein particle concentration and size as determined by nuclear magnetic resonance spectroscopy as predictors of cardiovascular disease in women. Circulation. 2002;106:1930–1937. doi:10.1161/01.cir.0000033222.75187.b97
  • Prado KB, Shugg S, Backstrand JR. Low-density lipoprotein particle number predicts coronary artery calcification in asymptomatic adults at intermediate risk of cardiovascular disease. J Clin Lipidol. 2011;5:408–413. doi:10.1016/j.jacl.2011.07.00124
  • Sacks FM. Low-density lipoprotein size and cardiovascular disease: a reappraisal. J Clin Endocrinol Metab. 2003;88:4525–4532. doi:10.1210/jc.2003-030636
  • Austin MA, Breslow JL, Hennekens CH, Buring JE, Willett WC, Krauss RM. Low-density lipoprotein subclass patterns and risk of myocardial infarction. JAMA. 1988;260:1917–1921.
  • Lindgren FT, Elliott HA, Gofman JW. The ultracentrifugal characterization and isolation of human blood lipids and lipoproteins, with applications to the study of atherosclerosis. J Phys Colloid Chem. 1951;55:80–93.
  • Griffin BA, Caslake MJ, Yip B, Tait GW, Packard CJ, Shepherd J. Rapid isolation of low density lipoprotein (ldl) subfractions from plasma by density gradient ultracentrifugation. Atherosclerosis. 1990;83:59–67. doi:10.1016/0021-9150(90)90131-2
  • MM KR S, Lindgren FT, Forte TM. Heterogeneity of serum low density lipoproteins in normal humans. J Lipid Res. 1981;22:236–244.
  • Davies IG. Rapid separation of LDL subclasses by iodixanol gradient ultracentrifugation. Clin Chem. 2003;49:1865–1872.
  • Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep. 2018;20(2):12. doi:10.1007/s11906-018-0812-z
  • Kelli HM, Kassas I, Lattouf OM. Cardio metabolic syndrome: a global epidemic. J Diabetes Metab. 2015;6(3):1-14. doi:10.4172/2155-6156.1000513
  • Wilson PW, D’Agostino RB, Parise H, Sullivan L, Meigs JB. Metabolic syndrome as a precursor of cardiovascular disease and type 2 diabetes mellitus. Circulation. 2005;112(20):3066-3072. doi:10.1161/CIRCULATIONAHA.105.539528
  • Santos AE, Araújo LF, Griep RH, et al. Shift work, job strain, and metabolic syndrome: cross-sectional analysis of ELSA-Brasil. Am J Ind Med. 2018;61(11):911-918. doi:10.1002/ajim.22910
  • He Y, Wu W, Wu S, et al. Linking gut microbiota, metabolic syndrome and economic status based on a population-level analysis. Microbiome. 2018;6(1):172. doi:10.1186/s40168-018-0557-6
  • Shuster A, Patlas M, Pinthus JH, Mourtzakis M. The clinical importance of visceral adiposity: a critical review of methods for visceral adipose tissue analysis. Br J Radiol. 2012;85(1009):1-10. doi:10.1259/bjr/38447238
  • Hurt L, Pinto CD, Watson J, Grant M, Gielner J; CDC. Diagnosis and screening for obesity-related conditions among children and teens receiving Medicaid—Maryland, 2005-2010. MMWR Morb Mortal Wkly Rep. 2014;63(14):305-308. https://www.cdc.gov/mmwr/preview/mmwrhtml/mm6314a2.htm
  • Hesse MB, Young G, Murray RD. Evaluating health risk using a continuous metabolic syndrome score in obese children. J Pediatr Endocrinol Metab. 2016;29(4):451-458. doi:10.1515/jpem-2015-0271
  • Löffler-Wirth H, Willscher E, Ahnert P, et al. Novel anthropometry based on 3D-bodyscans applied to a large population based cohort. PLoS One. 2016;11(7):e0159887. doi:10.1371/journal.pone.0159887
  • Apple and pear body shapes. Mayo Clinic. Accessed September 27, 2018. http://www.mayoclinic.org/diseases-conditions/metabolic-syndrome/multimedia/apple-and-pear-body-shapes/img-20006114
  • Kärkkäinen O, Lankinen MA, Vitale M, et al. Diets rich in whole grains increase betainized compounds associated with glucose metabolism. Am J Clin Nutr. 2018;108(5):971-979. doi:10.1093/ajcn/nqy169
  • Phillips CM, Harrington JM, Perry IJ. Relationship between dietary quality, determined by DASH score, and cardiometabolic health biomarkers: a cross-sectional analysis in adults. Clin Nutr. 2019;38(4):1620-1628. doi:10.1016/j.clnu.2018.08.028
  • Mathew AV, Li L, Byun J, et al. Therapeutic lifestyle changes improve HDL function by inhibiting myeloperoxidase-mediated oxidation in patients with metabolic syndrome. Diabetes Care. 2018;41(11):2431-2437. doi:10.2337/dc18-0049
  • Lackland DT, Voeks JH. Metabolic syndrome and hypertension: regular exercise as part of lifestyle management. Curr Hypertens Rep. 2014;16(11):492. doi:10.1007/s11906-014-0492-2
  • 1.Leasher JL, Bourne RR, Flaxman SR, et al. Global estimates on the number of people blind or visually impaired by diabetic retinopathy: a meta-analysis from 1990 to 2010. Diabetes Care. 2016;39(9):1643-1649. doi:2337/dc15-2171
  • 2.Rodríguez-Gutiérrez R, Montori VM. Glycemic control for patients with type 2 diabetes mellitus: our evolving faith in the face of evidence. Circ Cardiovasc Qual Outcomes. 2016;9(5):504-512. doi:1161/CIRCOUTCOMES.116.002901
  • 3.Forouhi NG, Misra A, Mohan V, Taylor R, Yancy W. Dietary and nutritional approaches for prevention and management of type 2 diabetes. BMJ. 2018;361:k2234. doi:1136/bmj.k2234
  • 4.Boulin M, Diaby V, Tannenbaum C. Preventing unnecessary costs of drug-induced hypoglycemia in older adults with type 2 diabetes in the United States and Canada. PLoS One. 2016;11(9):e0162951. doi:1371/journal.pone.0162951
  • 5.Abdelhafiz AH, McNicholas E, Sinclair AJ. Hypoglycemia, frailty and dementia in older people with diabetes: reciprocal relations and clinical implications. J Diabetes Complications. 2016;30(8):1548-1554. doi:1016/j.jdiacomp.2016.07.027
  • 6.Sala-Vila A, Díaz-López A, Valls-Pedret C, et al. Dietary marine omega-3 fatty acids and incident sight-threatening retinopathy in middle-aged and older individuals with type 2 diabetes: prospective investigation from the PREDIMED trial. JAMA Ophthalmol. 2016;134(10):1142-1149. doi:1001/jamaophthalmol.2016.2906
  • 7.Taheri S, Zaghloul H, Chagoury O, et al. Effect of intensive lifestyle intervention on bodyweight and glycaemia in early type 2 diabetes (DIADEM-I): an open-label, parallel group, randomized controlled trial. Lancet Diabetes Endocrinol. 2020;8(6):477-498. doi:1016/S2213-8587(20)30117-0
  • 8.Johansen MY, MacDonald CS, Hansen KB, et al. Effect of an intensive lifestyle intervention on glycemic control in patients with type 2 diabetes: a randomized clinical trial. 2017;318(7):637-646. doi:10.1001/jama.2017.10169
  • 9.Sbroma Tomaro E, Pippi R, Reginato E, et al. Intensive lifestyle intervention is particularly advantageous in poorly controlled type 2 diabetes. Nutr Metab Cardiovasc Dis. 2017;27(8):688-694. doi:1016/j.numecd.2017.06.009
  • 10.Markesbery, W. Damage to Lipids, Proteins, DNA, and RNA in Mild Cognitive Impairment. Arch Neurol. 64(7):954-956; July, 2007.
    Mecocci, P., et al. Arch Neurol 59:794-798; May, 2002.
  • 11.Geroldi, C., et al. Insulin Resistance and Cognitive Impairment, The InCHIANTI Study. Arch Neurol 62; July, 2005:, 1067-72.
  • 12.Ohara; T., et al. Glucose tolerance status and risk of dementia in the community, The Hisayama Study. Neurology September 20, 2011 vol. 77 no. 12 1126-1134.
  • 13.Ott, A., et al. Diabetes mellitus and the risk of dementia, The Rotterdam Study.  Neurology 53:1937-42, December, 1999.
  • 14.K. Yaffe. MD, et al. Advanced glycation and product level, diabetes, and accelerated cognitive aging. Neurology October 4, 2011 77:1351-56.
  • 15.Enzinger, C., et al. Annual brain volume change.  Neurology 64:1704-11; May 24, 2005.
  • 16.Wild RA. Obesity, lipids, cardiovascular risk and androgen excess. Am J Med 1995;98:27S-32S.
  • 17.Singh A, et al. Effect of insulin-like growth factor-type I (IFG-1) and insulin on the sectretion of sex-hormone binding globulin and IGF binding protein (IBP-1) by human hepatoma cells. J Endocrinol 1990;124:R1-3.
  • 18.Zakrzewska KE, et al. Glucocorticoids as counterregulatory hormones of leptin: toward an understanding of leptin resistance. Diabetes 1997;46:717-719.
  • 19.Zakrzewska, et al. Induction of obesity and hyperleptinemia by central glucocorticoid infusion in the rat. Diabetes 1999;48:365-370.
    Bermeis K, Vosmeer S, Kelly U. Effects of glucocorticoids and of growtu hormone on serum leptin concentratons in man. Eur J Endocrinol 1996;663-665.
  • 20.Weiderpass E, et al. Diabetes mellitus and risk of large bowel cancer. J Natl Cancer Inst 1997;89:660-661.
  • 21.Corpet D, et al. Insulin injections promote the growth of aberrant crypt foci in the colon of rats. Nutr Cancer 1997;27:316-320.
    22.Storlien LH, et al. Fatty acids, triglycerides and syndromes of insulin resistance. Prostaglandin Leukot Essent Fatty Acids 1997;57(4-5):379385.
  • 23.Brenner R. Nutrition and hormonal factors influencing desaturation of essential fatty acids. Prog Lipid Res 1982;20;41-48.
  • 24.Tuomi T, Groop LC, Zimmet PZ, Rowley MJ, Knowles W, Mackay IR. Antibodies to glutamic acid decarboxylase reveal latent autoimmune diabetes mellitus in adults with a non-insulin-dependent onset of disease. Diabetes 1993 Feb;42(2);359-362.
  • 25.Matejkova-Behanova M. Latent autoimmune diabetes in adults (LADA) and autoimmune thyroiditis. Endocr Regul. 2001 Sep;35(3):167-172.
    Unnikrishnan AG, Singh SK, Sanjeevi CB. Prevalence of GAD65 antibodies in leansubjects with type 2 diabetes. Ann N Y Acad Sci. 2004 Dec;1037:118-121.
  • 26.Appel SJ, Wadas TM, Rosenthal RS, Ovalle F. Latent autoimmune diabetes of adulthood (LADA): an often misdiagnosed type of diabetes mellitus. J Am Acad Nurse Pract. 2009 Mar;21(3)156-159.
  • 27.Nambam B, Aggarwal S, Jain A. Latent autoimmune diabetes in adults: A distinct but heterogeneous clinical entity. World J Diabetes. 2010 Sep 15;1(4)111-115.
  • 28.Zhou J, Ma XJ, BaoYQ, Pan XP, Lu W, Hu C, Xiang KS, Jia Wp. [Study on prevalence of latent autoimmune diabetes in adults and its relationship with metabolic syndrome]. Zhonghua Yi Xue Za Zhi. 2009 May 12;89(18):1250-4.
  • 29.Zhonghua, Yi Xue Za Zhi. 2009 May 12;89(18):1250-1254.
  • 30.Brooks-Worrell B, Palmer JP. Immunology in the Clinic Review Series; focus on metabolic diseases: development of islet autoimmune disease in type 2 diabetes patients: potential sequelae of chronic inflammation. Clin Exp Immunol. 2012 Jan;167(1):40-6.
  • 31.Li X, Zhou ZG, Yang L, Huang G, Yan X. [Metabolic syndrome and latent autoimmune diabetes in adults]. Zhongguo Yi Xue Yuan Xue Bao. 2003 Dec;25(6)676-9.
  • 32.Schernthaner G, Hink S, Kopp HP, Muzyka B, Streit G, Krosis A. Progress in the characterization of slowly progressive autoimmune diabetes in adult patients (LADA or type 1.5 diabetes). Exp Clin Endocrinol Diabetes. 2001;109 Suppl 2:S94-108.
  • Centers for Disease Control and Prevention. Second National Report on Human
    Exposure to Environmental Chemicals, NCEH Publication No. 03-0022. Atlanta,
    GA: Centers for Disease Control. Available at: http://www.cdc.gov/exposurereport/2nd/
  • Pelletier C, Imbeault P, Tremblay A. Energy balance and pollution by organochlorines and polychlorinated biphenyls. Obes Rev. 2003;4(1):17-24.
  • Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Oxidative stress and stress-activated signaling pathways: a unifying hypothesis of type 2 diabetes. Endocrine Reviews
    23(5):599–622.
  • Girard D. Activation of human polymorphonuclear neutrophils by environmental
    contaminants. Rev Environ Health 2003;18(2):75-89.
  • Sonnenberg GE, Krakower GR, Kissebah AH. A novel pathway to the manifestations
    of metabolic syndrome. Obes Res. 2004;12(2):180-186.
  • Wang Z, Zhou YT, Kakuma T, et al. Leptin resistance of adipocytes in obesity: role of
    suppressors of cytokine signaling. Biochem Biophys Res Commun. 2000;277 (1):20-26.
  • Beattie JH, Wood AM, Newman AM, Bremner I, et al. Obesity and hyperleptinemia in metallothionein (-I and -II) null mice. Proc Natl Acad Sci U S A. 1998;95(1):358-363.
  • Goto M, Arima H, Watanabe M, et al. Ghrelin increases neuropeptide y and agoutirelated peptide gene expression in the arcuate nucleus in rat hypothalamic organotypic] cultures. Endocrinol. 2006.;147(11):5102-5109.
  • Wynne K, Stanley S, McGowan B, Bloom S. Appetite control. J Endocrinol.
    2005;184(2):291-318.
  • Dube MG, Pu S, Kalra SP, et al. Melanocortin signaling is decreased during neurotoxin-induced transient hyperphagia and increased body-weight gain. Peptides.
    2000;21(6):793-801.
  • Shoemaker RC, House DE. A time-series study of sick building syndrome: chronic,
    biotoxin-associated illness from exposure to water-damaged buildings. Neurotoxicol
    Teratol. 2005;27(1):29-46.
  • Catania A, Delgado R, Airaghi L, et. al. alpha-MSH in systemic inflammation.
    Central and peripheral actions. Ann N Y Acad Sci. 1999;885:183-187.
  • Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin resistance and
    type 2 diabetes. Trends Endocrinol Metab. 2000;;11(6):212-217.
  • Dube MG, Pu S, Kalra SP, et al. Melanocortin signaling is decreased during neurotoxin-induced transient hyperphagia and increased body-weight gain. Peptides.
    2000;21(6):793-801.
  • Shoemaker RC, House DE. A time-series study of sick building syndrome: chronic,
    biotoxin-associated illness from exposure to water-damaged buildings. Neurotoxicol
    Teratol. 2005;27(1):29-46.
  • Gladen BC, Ragan NB, Rogan WJ. Pubertal growth and development and prenatal
    and lactational exposure to polychlorinated biphenyls and dichlorodiphenyl
    dichloroethene. J Pediatr. 2000;136(4):490-496.
  • Tremblay A, Pelletier C, Doucet E, et al. Thermogenesis and weight loss in obese
    individuals: a primary association with organochlorine pollution. Int J Obes Relat
    Metab Disord. 2004;28(7):936-939.
  • Imbeault P, Tremblay A, Simoneau JA, et al. Weight loss-induced rise in plasma pollutant is associated with reduced skeletal muscle oxidative capacity. Am J Physiol
    Endocrinol Metab. 2002;282(3):E574-E579.
  • Nicolau GY, Pesticide effects upon the circadian time structure in the thyroid, adrenal and testis in rats. Endocrinologie. 1982;20(2):73-90.
  • Duke University Integrated Toxicology Program, National Institute of
    Environmental Health Sciences/NIH/DHHS, Obesity: Developmental Origins and
    Environmental Influences. Available at: http://www.niehs.nih.gov/multimedia/
    qt/dert/obesity/agenda.htm.
  • Moskaug JO, Carlsen H, Myhrstad MD, et al. Polyphenols and glutathione synthesis
    regulation. Am J Clin Nutr. 2005;81(1 Suppl):277S-283S.
  • Masuda A, Miyata M, Kihara T, et al. Repeated sauna therapy reduces urinary 8-epiprostaglandin F(2alpha). Jpn Heart J. 2004;45(2):297-303.
  • Biro S, Masuda A, Kihara T, Tei C. Clinical implications of thermal therapy in
    lifestyle-related diseases. Exp Biol Med (Maywood). 2003;228(10):1245-1249.
  • Hyman M. Ultrametabolism. New York: Scribner; 2006.
  • Hyman MA. Systems biology: the gut-brain-fat cell connection and obesity. Altern
    Ther Health Med. 2006;12(1):10-16
  • Hyman MA. Refrigerator rights–the missing link in health, disease, and obesity,
    Altern Ther Health Med. 2005;11(4):10-12.
  • Lyon M, Bland J, Jones DS. Chapter 31: Clinical approaches to detoxification and
    biotransformation. In: Jones, DS (Ed). Textbook of Functional Medicine. Gig Harbor,
    WA: Institute for Functional Medicine; 2006.
  • Ammini A.C. et al. 2017. Andropause. In: Kumar A., Sharma M. Basics of Human Andrology.
  • Samaras, N. et al. 2012. Andropause: A review of the definition and treatment. European Geriatric Medicine.
  • NHS. 2019. The ‘male menopause’. Available: https://www.nhs.uk/conditions/male-menopause/. [Accessed September 2019].
  • Travison, T.G. et al. 2007. The relative contributions of aging, health, and lifestyle factors to serum testosterone decline in men. Journal of Clinical Endocrinology & Metabolism.
  • Khosravi, S. et al. 2015. Are andropause symptoms related to depression? Aging and Clinical Experimental Research.
  • Anaissie, J. et al. 2017. Testosterone deficiency in adults and corresponding treatment patterns across the globe. Translational Andrology and Urology.
  • Anderson, J.K et al. 2002. Andropause: Knowledge and perceptions among the general public and health care professionals. The Journals of Gerontology.
  • Snyder, P.J. et al. 2018. Lessons From The Testosterone Trials. Endocrine Reviews.
  • Guth, M.A.S. 2016. Bioidentical Hormone Replacement Therapy for Men in the Primary Care Setting. Quality in Primary Care.
  • Guth M.A. 2015. Compounded testosterone troches to optimize health and the testosterone controversy. International Journal of Pharmaceutical Compounding.
  • Tworek, D. 2019. Bioidentical Hormone Replacement Therapy for Men. Available: https://www.bodylogicmd.com/bioidentical-hormone-therapy-for-men. [Accessed September 2019].
  • Haring, R. et al. 2010. Prevalence, incidence and risk factors of testosterone deficiency in a population-based cohort of men: results from the study of health in Pomerania. Aging Male.
  • Hisause, S. 2015. Contemporary perspective and management of testosterone deficiency: Modifiable factors and variable management. International Journal of Urology.
  • Wu, F.C.W. et al. 2008. Hypothalamic-Pituitary-Testicular Axis Disruptions in Older Men Are Differentially Linked to Age and Modifiable Risk Factors: The European Male Aging Study. The Journal of Clinical Endocrinology & Metabolism.
  • Hirokawa, K. et al. 2012. Job demands as a potential modifier of the association between testosterone deficiency and andropause symptoms in Japanese middle-aged workers: A cross-sectional study. Maturitas.
  • Mawer, R. 2016. 8 Proven Ways to Increase Testosterone Levels Naturally. Available: https://www.healthline.com/nutrition/8-ways-to-boost-testosterone. [Accessed September 2019].
  • Harrison, J. 2011. ‘Talking about my generation’: a state-of-the-art review of health information for men in the andropause. Health Information and Libraries Journal.
  • National Institute of Environmental Health Sciences. Neurodegenerative diseases. Reviewed September 10, 2019. Accessed May 18, 2020. https://www.niehs.nih.gov/research/supported/health/neurodegenerative/index.cfm
  • Swerdlow RH. Mitochondria and mitochondrial cascades in Alzheimer’s disease. J Alzheimers Dis.2018;62(3):1403-1416. doi:10.3233/JAD-170585
  • García S, Martín Giménez VM, Mocayar Marón FJ, Reiter RJ, Manucha W. Melatonin and cannabinoids: mitochondrial-targeted molecules that may reduce inflammaging in neurodegenerative diseases. Histol Histopathol. 2020:18212. doi:10.14670/HH-18-212
  • Alzheimer’s Association. Alternative treatments. Accessed May 18, 2020. https://www.alz.org/alzheimers-dementia/treatments/alternative-treatments
  • Parkinson’s Foundation. Over the counter and complementary therapies. Accessed May 18, 2020. https://www.parkinson.org/Understanding-Parkinsons/Treatment/Over-the-Counter-and-Complementary-Therapies
  • National Multiple Sclerosis Society. Complementary and alternative medicines. Accessed May 18, 2020. https://www.nationalmssociety.org/Treating-MS/Complementary-Alternative-Medicines
  • Wesselman LMP, Doorduijn AS, de Leeuw FA, et al. Dietary patterns are related to clinical characteristics in memory clinic patients with subjective cognitive decline: the SCIENCe project. Nutrients. 2019;11(5):1057.doi:10.3390/nu11051057
  • Fieldhouse JLP, Doorduijn AS, de Leeuw FA, et al. A suboptimal diet is associated with poorer cognition: the NUDAD project. Nutrients. 2020;12(3):703. doi:10.3390/nu12030703
  • Nolan JM, Mulcahy R, Power R, Moran R, Howard AN. Nutritional intervention to prevent Alzheimer’s disease: potential benefits of xanthophyll carotenoids and omega-3 fatty acids combined. J Alzheimers Dis. 2018;64(2):367-378. doi:10.3233/JAD-180160
  • Calil SRB, Brucki SMD, Nitrini R, Yassuda MS. Adherence to the Mediterranean and MIND diets is associated with better cognition in healthy seniors but not in MCI or AD. Clin Nutr ESPEN. 2018;28:201-207. doi:10.1016/j.clnesp.2018.08.001
  • Power R, Prado-Cabrero A, Mulcahy R, Howard A, Nolan JM. The role of nutrition for the aging population: implications for cognition and Alzheimer’s disease. Annu Rev Food Sci Technol. 2019;10:619-639.doi:1146/annurev-food-030216-030125
  • Chai B, Gao F, Wu R, et al. Vitamin D deficiency as a risk factor for dementia and Alzheimer’s disease: anupdated meta-analysis. BMC Neurol. 2019;19(1):284. doi:10.1186/s12883-019-1500-6
  • Bianchi VE, Herrera PF, Laura R. Effect of nutrition on neurodegenerative diseases. A systematic review.Nutr Neurosci. Published online November 4, 2019. doi:10.1080/1028415X.2019.1681088
  • Grodzicki W, Dziendzikowska K. The role of selected bioactive compounds in the prevention of Alzheimer’s disease. Antioxidants. 2020;9(3):229. doi:10.3390/antiox9030229
  • AlAmmar WA, Albeesh FH, Ibrahim LM, Algindan YY, Yamani LZ, Khattab RY. Effect of omega-3 fatty acids and fish oil supplementation on multiple sclerosis: a systematic review. Nutr Neurosci. Published online August 28, 2019. doi:10.1080/1028415X.2019.1659560
  • Wlodarek D. Role of ketogenic diets in neurodegenerative diseases (Alzheimer’s disease and Parkinson’s disease). Nutrients. 2019;11(1):169. doi:10.3390/nu11010169
  • Chauhan A, Chauhan V. Beneficial effects of walnuts on cognition and brain health. Nutrients. 2020;12(2):550. doi:10.3390/nu12020550
  • Valls-Pedret C, Sala-Vila A, Serra-Mir M, et al. Mediterranean diet and age-related cognitive decline: a randomized clinical trial. JAMA Intern Med. 2015;175(7):1094-1103. doi:10.1001/jamainternmed.2015.1668
  • Meng X, Zhou J, Zhao CN, Gan RY, Li HB. Health benefits and molecular mechanisms of resveratrol: a narrative review. Foods. 2020;9(3):340. doi:10.3390/foods9030340
  • Huhn S, Beyer F, Zhang R, et al. Effects of resveratrol on memory performance, hippocampus connectivity and microstructure in older adults – a randomized controlled trial. Neuroimage. 2018;174:177-190. doi:10.1016/j.neuroimage.2018.03.023
  • Evans HM, Howe PR, Wong RH. Effects of resveratrol on cognitive performance, mood and cerebrovascular function in post-menopausal women: a 14-week randomised placebo-controlled intervention trial. Nutrients. 2017;9(1):27. doi:10.3390/nu9010027
  • Liu H, Ye M, Guo H. An updated review of randomized clinical trials testing the improvement of cognitive function of Ginkgo biloba extract in healthy people and Alzheimer’s patients. Front Pharmacol. 2020;10:1688. doi:10.3389/fphar.2019.01688
  • Power R, Coen RF, Beatty S, et al. Supplemental retinal carotenoids enhance memory in healthy individuals with low levels of macular pigment in a randomized, double-blind, placebo-controlled clinical trial. J Alzheimers Dis. 2018;61(3):947-961. doi:10.3233/JAD-170713
  • Lee DH, Chon J, Kim Y, et al. Association between vitamin D deficiency and cognitive function in the elderly Korean population: a Korean frailty and aging cohort study. Medicine (Baltimore). 2020;99(8):e19293. doi:10.1097/MD.0000000000019293
  • Feige J, Moser T, Bieler L, Schwenker K, Hauer L, Sellner J. Vitamin D supplementation in multiple sclerosis: a critical analysis of potentials and threats. Nutrients. 2020;12(3):783. doi:10.3390/nu12030783
  • Kouchaki E, Afarini M, Abolhassani J, et al. High-dose ?-3 fatty acid plus vitamin D3 supplementation affects clinical symptoms and metabolic status of patients with multiple sclerosis: a randomized controlled clinical trial. J Nutr. 2018;148(8):1380-1386. doi:10.1093/jn/nxy116
  • Martínez-Lapiscina EH, Clavero P, Toledo E, et al. Mediterranean diet improves cognition: the PREDIMED-NAVARRA randomised trial. J Neurol Neurosurg Psychiatry. 2013;84(12):1318-1325. doi:10.1136/jnnp-2012-304792
  • Lee JE, Bisht B, Hall MJ, et al. A multimodal, nonpharmacologic intervention improves mood and cognitive function in people with multiple sclerosis. J Am Coll Nutr. 2017;36(3):150-168. doi:10.1080/07315724.2016.1255160
  • Ota M, Matsuo J, Ishida I, et al. Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in patients with mild-to-moderate Alzheimer’s disease. Neurosci Lett. 2019;690:232-236. doi: 10.1016/j.neulet.2018.10.048

Schedule an Online Medical Consultation

1. Create your Personal Profile
2. Fill your Medical History Online
3. Schedule an Appointment

Latest News