Introduction: Women, Aging, Genetics
1. Rappaport, S. M. “Implications of the exposome for exposure science.” Journal of Exposure Science and Environmental Epidemiology 21, no. 1 (2011): 5–9; Rappaport, S. M., et al. “Using the blood exposome to discover causes of disease.” Agilent Technologies, September 15, 2015, accessed February 9, 2015, www.agilent.com/cs/library/technicaloverviews/ Public/5991–3418EN.pdf; Harmon, K. “Sequencing the exposome: Researchers take a cue from genomics to decipher environmental exposure’s links to disease.” Scientific American, October 21, 2010, accessed February 2, 2016, www.scientificamerican.com/article/ environmental-exposure.
2. “Vital Statistics Rapid Release.” Centers for Disease Control and Prevention, accessed April 11, 2016, www.cdc.gov/nchs/products/vsrr/mortality-dashboard.htm; Ludwig, D. S. “Lifespan weighed down by diet.” JAMA (2016).
3. Vincent, G. K., et al. The Next Four Decades: The Older Population in the United States: 2010 to 2050 (U.S. Department of Commerce, Economics and Statistics Administration, U.S. Census Bureau, 2010).
4. Hebert, L. E., et al. “Annual incidence of Alzheimer disease in the United States projected to the years 2000 through 2050.” Alzheimer Disease and Associated Disorders 15, no. 4 (2001): 169–73; Alzheimer’s Association, “2015 Alzheimer’s disease facts and figures.” Alzheimer’s and Dementia: Journal of the Alzheimer’s Association 11, no. 3 (2015): 332.
5. “U.S. breast cancer cases expected to increase by as much as 50 percent by 2030.” American Association for Cancer Research, accessed April 30, 2015, www.aacr.org/Newsroom/Pages/News-Release-Detail.aspx?ItemID=708#.VUJpv61VhBc; Brown, E. “Breast cancer cases in U.S. projected to rise as much as 50% by 2030.” Los Angeles Times, April 20, 2015, accessed April 30, 2015, www.latimes.com/science/sciencenow/la-sci-sn-breast-cancer-cases-2030–20150420-story.html.
6. van Drielen, K., et al. “Disentangling the effects of circulating IGF-1, glucose, and cortisol on features of perceived age.” Age 37, no. 3 (2015): 1–10.
7. Krøll, J. “Correlations of plasma cortisol levels, chaperone expression and mammalian longevity: a review of published data.” Biogerontology 11, no. 4 (2010): 495–99; van Drielen, K., et al. “Disentangling the effects of circulating IGF-1, glucose, and cortisol”; Christensen, K, et al. “Perceived age as clinically useful biomarker of ageing: cohort study.” British Medical Journal 339 (2009): b5262; Noordam, R., et al. “Serum insulin‐like growth factor 1 and facial ageing: high levels associate with reduced skin wrinkling in a cross‐sectional study.” British Journal of Dermatology 168, no. 3 (2013): 533–38; Noordam, R., et al. “Cortisol serum levels in familial longevity and perceived age: the Leiden longevity study.” Psychoneuroendocrinology 37, no. 10 (2012): 1669–75.
Chapter 1: Unlock Your Genes
1. “Cleveland Clinic’s Center for Functional Medicine: A Test Kitchen for Healthcare’s Future,” Holistic Primary Care, https://holisticprimarycare.net/topics/topics-a-g/functional-medicine/1680-cleveland-clinic-s-center-for-functional-medicine-a-test-kitchen-for-healthcare-s-future.html; http://my.clevelandclinic.org/services/center-for-functional-medicine
2. Gifford, B. Spring Chicken: Stay Young Forever (or Die Trying) (New York: Grand Central, 2015), 37. Data are from 2012.
3. “Top five cosmetic surgical procedures of 2013.” PlasticSurgery.org, accessed April 10, 2015, www.plasticsurgery.org/news/plastic-surgery-statistics/2013/top-five-cosmetic-surgery-procedures.html.
4. Furnham, A., et al. “Factors that motivate people to undergo cosmetic surgery.” Canadian Journal of Plastic Surgery 20, no. 4 (2012): e47.
5. “Get your bolder on. We can all use a little motivation.” GrowingBolder.com, accessed June 25, 2015, www.growingbolder.com/quotes/#.
6. Goodstein, G. “Ida Keeling still setting records, examples at 100.” Bronx Times, May 8, 2015, accessed June 25, 3015, www.bxtimes.com/stories/2015/19/19-ida-2015–05–08-bx.html.
7. Arem, H., et al. “Leisure time physical activity and mortality: A detailed pooled analysis of the dose-response relationship.” JAMA Internal Medicine 175, no. 6 (2015): 959–67.
Chapter 2: The Gene/Lifestyle Conversation
1. Memisoglu, A., et al. “Interaction between a peroxisome proliferator-activated receptor γ gene polymorphism and dietary fat intake in relation to body mass.” Human Molecular Genetics 12, no. 22 (2003): 2923–29.
2. Walsh, T., et al. “Ten genes for inherited breast cancer.” Cancer Cell 11, no. 2 (2007): 103–5; Walsh, T., et al. “Spectrum of mutations in BRCA1, BRCA2, CHEK2, and TP53 in families at high risk of breast cancer.” JAMA 295, no. 12 (2006): 1379–88; Aloraifi, F., et al. “Gene analysis techniques and susceptibility gene discovery in non-BRCA1/BRCA2 familial breast cancer.” Surgical Oncology 24, no. 2 (2015): 100–109; Lee, D. S. C., et al. “Comparable frequency of BRCA1, BRCA2 and TP53 germline mutations in a multi-ethnic Asian cohort suggests TP53 screening should be offered together with BRCA½ screening to early-onset breast cancer patients.” Breast Cancer Research 14, no. 2 (2012): R66. For lay audiences, these citations may be helpful: “Genetics,” BreastCancer.org, accessed February 13, 2016, www.breastcancer.org/risk/factors/genetics; “Inherited gene mutations,” Komen.org, accessed February 13, 2016, http://ww5.komen.org/BreastCancer/InheritedGenetic Mutations.html; “Breast cancer genes,” Cancer Research UK, accessed February 13, 2016, www.cancerresearchuk.org/about-cancer/type/breast-cancer/about/risks/breast-cancer-genes
3. Winkler, T. W., et al. “The influence of age and sex on genetic associations with adult body size and shape: A large-scale genome-wide interaction study.” PLoS Genetics 11, no. 10 (2015): e1005378.
4. “Orientation,” SNPedia, August 15, 2015, accessed October 20, 2015, http://snpedia.com/index.php/Orientation.
5. Miller, J. W., et al. “Vitamin D status and rates of cognitive decline in a multiethnic cohort of older adults.” JAMA Neurology (2015); Wilson, V. K., et al. “Relationship between 25-hydroxyvitamin D and cognitive function in older adults: The health, aging and body composition study.” Journal of the American Geriatrics Society 62, no. 4 (2014): 636–41; Chei, C. L., et al. “Vitamin D levels and cognition in elderly adults in China.” Journal of the American Geriatrics Society 62, no. 11 (2014): 2125–29; Littlejohns, T. J., et al. “Vitamin D and the risk of dementia and Alzheimer disease.” Neurology 83, no. 10 (2014): 920–28; Annweiler, C., et al. “Vitamin D-mentia: randomized clinical trials should be the next step.” Neuroepidemiology 37, nos. 3–4 (2011): 249–58.
Chapter 3: Epigenetics: Turning Genes On and Off
1. Siddhartha Mukherjee, The Gene: An Intimate History (New York: Scribner: 2016), 400.
2. Audergon, P., et al. “Restricted epigenetic inheritance of H3K9 methylation.” Science 348, no. 6230 (2015): 132–35.
3. Shapira, I., et al. “Evolving concepts: How diet and the intestinal microbiome act as modulators of breast malignancy.” ISRN Oncology 2013 (2013); Xuan, C., et al. “Microbial dysbiosis is associated with human breast cancer.” PloS One 9, no. 1 (2014): e83744; Sheflin, A. M., et al. “Cancer-promoting effects of microbial dysbiosis.” Current Oncology Reports 16, no. 10 (2014): 1–9; Kwa, M., et al. “The intestinal microbiome and estrogen receptor— positive female breast cancer.” Journal of the National Cancer Institute 108, no. 8 (2016): djw029; Plottel, C. S., et al. “Microbiome and malignancy.” Cell Host & Microbe 10, no. 4 (2011): 324-35.
4. Cummings S. R., et al. “Prevention of breast cancer in postmenopausal women: Approaches to estimating and reducing risk.” Journal of the National Cancer Institute 101, no. 6 (2009): 384–98.
5. Jolie, A. “My medical choice,” New York Times, May 14, 2013, www.nytimes.com/2013/05/14/opinion/my-medical-choice.html?_r=0.
6. Jolie, A. “Diary of a surgery,” New York Times, March 24, 2015, www.nytimes.com/2015/03/24/opinion/angelina-jolie-pitt-diary-of-a-surgery.html.
7. “The human genome project completion: Frequently asked questions.” National Human Genome Research Institute, www.genome.gov/11006943; “Talking glossary of genetic terms,” National Human Genome Research Institute, www.genome.gov/Glossary.
8. Shamovsky, I., et al. “New insights into the mechanism of heat shock response activation,” Cellular and Molecular Life Sciences 65, no. 6 (2008): 855–61; Miozzo, F., et al. “HSFs, stress sensors and sculptors of transcription compartments and epigenetic landscapes,” Journal of Molecular Biology 427, no. 24 (2015): 3793–3816; and Santoro, M. G. “Heat shock factors and the control of the stress response,” Biochemical Pharmacology 59, no. 1 (2000): 55–63.
9. Yamashita, H., et al. “A glucose-responsive transcription factor that regulates carbohydrate metabolism in the liver,” Proceedings of the National Academy of Sciences 98, no. 16 (2001): 9116–21.
10. Osborne, C. K., et al. “Estrogen receptor: current understanding of its activation and modulation,” Cancer Research 7, no. 12 (2001): 4338s–42s; Halachmi, S., et al. “Estrogen receptor-associated proteins: possible mediators of hormone-induced transcription,” Science 264, no. 5164 (1994): 1455–58; and Marino, M., et al. “Estrogen signaling multiple pathways to impact gene transcription,” Current Genomics 7, no. 8 (2006): 497–508.
11. Audergon, P., et al. “Restricted epigenetic inheritance.”
12. Sun, C., et al. “Potential epigenetic mechanism in non-alcoholic fatty liver disease.” International Journal of Molecular Sciences 16, no. 3 (2015): 5161–79.
13. Er, T. K., et al. “Targeted next-generation sequencing for molecular diagnosis of endometriosis-associated ovarian cancer.” Journal of Molecular Medicine (2016): 1–13; Wiegand, K. C., et al. “ARID1A mutations in endometriosis-associated ovarian carcinomas.” New England Journal of Medicine 363, no. 16 (2010): 1532–43; Ayhan, A., et al. “Loss of ARID1A expression is an early molecular event in tumor progression from ovarian endometriotic cyst to clear cell and endometrioid carcinoma.” International Journal of Gynecological Cancer: Official Journal of the International Gynecological Cancer Society 22, no. 8 (2012): 1310; Takeda, T., et al. “ARID1A gene mutation in ovarian and endometrial cancers (Review).” Oncology Reports 35, no. 2 (2016): 607–13.
14. Cao-Lei, L., et al. “DNA methylation signatures triggered by prenatal maternal stress exposure to a natural disaster: Project ice storm.” PLoS One 9, no. 9 (2014).
Chapter 4: Get to the Root
1. Volpato, S., et al. “Cardiovascular disease, interleukin-6, and risk of mortality in older women the women’s health and aging study.” Circulation 103, no. 7 (2001): 947–53; Harris, T. B., et al. “Associations of elevated interleukin-6 and C-reactive protein levels with mortality in the elderly.” American Journal of Medicine 106, no. 5 (1999): 506–12; Ferrucci, L., et al. “Serum IL-6 level and the development of disability in older persons.” Journal of the American Geriatrics Society 47, no. 6 (1999): 639–46.
2. Lin, H., et al. “Whole blood gene expression and interleukin-6 levels.” Genomics 104, no. 6 (2014): 490–95.
3. Barron, E., et al. “Blood-borne biomarkers of mortality risk: systematic review of cohort studies.” PloS One 10, no. 6 (2015): e0127550.
4. www.nhlbi.nih.gov/health/educational/lose_wt/BMI/bmicalc.htm.
5. Curtis, B. M., et al. “Autonomic tone as a cardiovascular risk factor: the dangers of chronic fight or flight.” Mayo Clinic Proceedings, 77, no. 1 (2002): 45–54; Thayer, J. F., et al. “The role of vagal function in the risk for cardiovascular disease and mortality.” Biological Psychology 74, no. 2 (2007): 224–42.
6. Kleiger, R. E., et al. “Heart rate variability: measurement and clinical utility.” Annals of Noninvasive Electrocardiology 10, no. 1 (2005): 88–101; Dekker, J. M., et al. “Low heart rate variability in a 2-minute rhythm strip predicts risk of coronary heart disease and mortality from several causes The ARIC Study.” Circulation 102, no. 11 (2000): 1239–44; Galinier, M. A., et al. “Depressed low frequency power of heart rate variability as an independent predictor of sudden death in chronic heart failure.” European Heart Journal 21, no. 6 (2000): 475–82.
7. Buettner, D. “The island where people forget to die,” New York Times, October 24, 2012, accessed August 17, 2015, www.nytimes.com/2012/10/28/magazine/the-island-where-people-forget-to-die.html?_r=1.
8. Panagiotakos, D. B., et al. “Sociodemographic and lifestyle statistics of oldest old people (> 80 years) living in Ikaria island: the Ikaria study.” Cardiology Research and Practice (2011); Chrysohoou, C., et al. “Four-year (2009–2013) All cause and cardiovascular disease mortality and its determinants: The Ikaria study.” Journal of the American College of Cardiology 63, no. 12_S (2014); Stefanadis, C. I., “Aging, genes and environment: lessons from the Ikaria study.” Hellenic Journal of Cardiology 54, no. 3 (2013): 237–38; Trichopoulou, A., et al. “Anatomy of health effects of Mediterranean diet: Greek EPIC prospective cohort study.” British Medical Journal 338 (2009).
9. Buettner, D. The Blue Zones: 9 Lessons for Living Longer from the People Who’ve Lived the Longest, 2nd ed. (Washington, DC: National Geographic, 2012).
10. Chilton, S. N., et al. “Inclusion of fermented foods in food guides around the world.” Nutrients 7, no. 1 (2015): 390–404.
11. Timmers, S., et al. “Calorie restriction–like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans.” Cell Metabolism 14, no. 5 (2011): 612–22; Morselli, E., et al. “Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy.” Cell Death and Disease 1, no. 1 (2010): e10; Baur, J. A., et al. “Resveratrol improves health and survival of mice on a high-calorie diet.” Nature 444, no. 7117 (2006): 337–42.
12. Pérez-Rubio, K. G., et al. “Effect of berberine administration on metabolic syndrome, insulin sensitivity, and insulin secretion.” Metabolic Syndrome and Related Disorders 11, no. 5 (2013): 366–69; Pirillo, A., et al. “Berberine, a plant alkaloid with lipid-and glucose-lowering properties: From in vitro evidence to clinical studies.” Atherosclerosis 243, no. 2 (2015): 449–61; Pang, B., et al. “Application of berberine on treating type 2 diabetes mellitus.” International Journal of Endocrinology 2015 (2015).
13. Yarla, N. S., et al. “Targeting arachidonic acid pathway by natural products for cancer prevention and therapy.” Seminars in Cancer Biology (2016); Zarei, A., et al. “A quick overview on some aspects of endocrinological and therapeutic effects of Berberis vulgaris L.” Avicenna Journal of Phytomedicine 5, no. 6 (2015): 485; Caliceti, C., et al. “Potential benefits of berberine in the management of perimenopausal syndrome.” Oxidative Medicine and Cellular Longevity (2015); Yang, J., et al. “Berberine improves insulin sensitivity by inhibiting fat store and adjusting adipokines profile in human preadipocytes and metabolic syndrome patients,” Evidence-Based Complementary and Alternative Medicine 2012 (2012); and Hu, Y., et al. “Lipid-lowering effect of berberine in human subjects and rats,” Phytomedicine 19, no. 10 (2012): 861–67.
14. Guo, Y., et al. “Repeated administration of berberine inhibits cytochromes P450 in humans,” European Journal of Clinical Pharmacology 68, no. 2 (2012): 213–17.
Chapter 5: Feed—Week 1
1. Clark, M. “Still blazing trails,” New York Times, August 4, 2014, accessed October 7, 2015, www.nytimes.com/2014/08/06/dining/still-blazing-trails.html.
2. Fussell, B. “Earning her food,” New York Times, March 26, 2010, accessed September 20, 2015, www.nytimes.com/2010/03/28/magazine/28lives-t.html?_r=1.
3. Bergen, H. R., et al. “Myostatin as a mediator of sarcopenia versus homeostatic regulator of muscle mass: Insights using a new mass spectrometry-based assay.” Skeletal Muscle 5, no. 1 (2015): 1.
4. Willer, C. J., et al. “Six new loci associated with body mass index highlight a neuronal influence on body weight regulation.” Nature Genetics 41, no. 1 (2009): 25–34; Wang, J., et al. “Study of eight GWAS-identified common variants for association with obesity-related indices in Chinese children at puberty.” International Journal of Obesity 36, no. 4 (2012): 542–47; Speakman, J. R. “Functional analysis of seven genes linked to body mass index and adiposity by genome-wide association studies: a review.” Human Heredity 75, nos. 2–4 (2013): 57–79; Fawcett, K. A., et al. “The genetics of obesity: FTO leads the way.” Trends in Genetics 26, no. 6 (2010): 266–74.
5. Donaldson, C. M., et al. “Glycemic index and endurance performance.” International Journal of Sport Nutrition and Exercise Metabolism 20, no. 2 (2010): 154–65; Bornet, F. R. J., et al. “Glycaemic response to foods: impact on satiety and long-term weight regulation.” Appetite 49, no. 3 (2007): 535–53; Philippou, E., et al. “The influence of glycemic index on cognitive functioning: a systematic review of the evidence.” Advances in Nutrition: An International Review Journal 5, no. 2 (2014): 119–30; Vranešić Bender, D., et al. “Nutritional and behavioral modification therapies of obesity: facts and fiction.” Digestive Diseases 30, no. 2 (2012): 163–67; Mediano, M. F. F., et al. “Insulin Resistance Predicts the Effectiveness of Different Glycemic Index Diets on Weight Loss in Non-Obese Women.” Obesity Facts 5, no. 5 (2012): 641–47; Sichieri, R., et al. “An 18-mo randomized trial of a low-glycemic-index diet and weight change in Brazilian women.” American Journal of Clinical Nutrition 86, no. 3 (2007): 707–13.
6. Martins, M. L., et al. “Incidence of microflora and of ochratoxin A in green coffee beans (Coffea arabica).” Food Additives and Contaminants 20, no. 12 (2003): 1127–31; Studer-Rohr, I., et al. “The occurrence of ochratoxin A in coffee.” Food and Chemical Toxicology 33, no. 5 (1995): 341–55.
7. Frankenfeld, C.L., et al. “High-intensity sweetener consumption and gut microbiome content and predicted gene function in a cross-sectional study of adults in the United States.” Annals of Epidemiology 25, no. 10 (2015): 736–42; Burke, M. V., et al. “Physiological mechanisms by which non-nutritive sweeteners may impact body weight and metabolism.” Physiology and Behavior (2015); Pepino, M. Y., “Metabolic effects of non-nutritive sweeteners.” Physiology and Behavior (2015).
8. Chen, W. Y., et al. “Moderate alcohol consumption during adult life, drinking patterns, and breast cancer risk.” JAMA 306, no. 17 (2011): 1884–90.
9. Iwai, K., et al. “Identification of food-derived collagen peptides in human blood after oral ingestion of gelatin hydrolysates.” Journal of Agricultural and Food Chemistry 53, no. 16 (2005): 6531–36.
10. Choonpicharn, S., et al. “Antioxidant and antihypertensive activity of gelatin hydrolysate from Nile tilapia skin.” Journal of Food Science and Technology 52, no. 5 (2014): 3134–39; Ao, J., et al. “Amino acid composition and antioxidant activities of hydrolysates and peptide fractions from porcine collagen.” Food Science and Technology International 18, no. 5 (2012): 425–34.
11. Choonpicharn et al. “Antioxidant and antihypertensive activity”; Ngo, D. H., et al. “Angiotensin-I converting enzyme inhibitory peptides from antihypertensive skate (Okamejei kenojei) skin gelatin hydrolysate in spontaneously hypertensive rats.” Food Chemistry 174 (2015): 37–43.
12. Leem, K. H., et al. “Porcine skin gelatin hydrolysate promotes longitudinal bone growth in adolescent rats.” Journal of Medicinal Food 16, no. 5 (2013): 447–53.
13. Costanzo, S., et al. “Wine, beer or spirit drinking in relation to fatal and non-fatal cardiovascular events: a meta-analysis.” European Journal of Epidemiology 26, no. 11 (2011): 833–50.
14. Park, K., et al. “Acute and subacute toxicity of copper sulfate pentahydrate (CuSO45 H2O) in the guppy (Poecilia reticulata).” Journal of Veterinary Medical Science 71, no. 3 (2009): 333–36; Hébert, C. D., et al. “Subchronic toxicity of cupric sulfate administered in drinking water and feed to rats and mice.” Fundamental and Applied Toxicology 21, no. 4 (1993): 461–75; Sinkovič, A., et al. “Severe acute copper sulphate poisoning: a case report.” Archives of Industrial Hygiene and Toxicology (2008): 31–35.
15. “All 48 fruits and vegetables with a pesticide residue data,” Environmental Working Group, accessed June 15, 2015, www.ewg.org/foodnews/list.php.
16. Costanzo et al. “Wine, beer or spirit drinking in relation”; Streppel, M. T., et al. “Long-term wine consumption is related to cardiovascular mortality and life expectancy independently of moderate alcohol intake: the Zutphen Study.” Journal of Epidemiology and Community Health 63, no. 7 (2009): 534–40.
17. Chen et al. “Moderate alcohol consumption”; Strumylaite, L., et al. “The association of low-to-moderate alcohol consumption with breast cancer subtypes defined by hormone receptor status.” PloS One 10, no. 12 (2015): e0144680; Williams, L. A., et al. “Alcohol intake and invasive breast cancer risk by molecular subtype and race in the Carolina Breast Cancer Study.” Cancer Causes and Control 27, no. 2 (2016): 259–69; Cao, Y., et al. “Light to moderate intake of alcohol, drinking patterns, and risk of cancer: results from two prospective US cohort studies.” British Medical Journal (2015): h4238.
18. Goldberg, D. M., et al. “A global survey of trans-resveratrol concentrations in commercial wines.” American Journal of Enology and Viticulture 46, no. 2 (1995): 159–65; Crandall, J. P., et al. “Pilot study of resveratrol in older adults with impaired glucose tolerance.” Journals of Gerontology Series A: Biological Sciences and Medical Sciences (2012): glr235; Zamora-Ros, R., et al. “High urinary levels of resveratrol metabolites are associated with a reduction in the prevalence of cardiovascular risk factors in high-risk patients.” Pharmacological Research 65, no. 6 (2012): 615–20; Brasnyó, P., et al. “Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients.” British Journal of Nutrition 106, no. 3 (2011): 383–89; Marchal, J., et al. “Resveratrol in mammals: effects on aging biomarkers, age-related diseases, and life span.” Annals of the New York Academy of Sciences 1290, no. 1 (2013): 67–73.
19. Semba, R. D., et al. “Resveratrol levels and all-cause mortality in older community-dwelling adults.” JAMA Internal Medicine 174, no. 7 (2014): 1077–84; Yoshino, J., et al. “Resveratrol supplementation does not improve metabolic function in non-obese women with normal glucose tolerance.” Cell Metabolism 16 (2012): 658–64; Bitterman, J. L., et al. “Metabolic effects of resveratrol: addressing the controversies.” Cellular and Molecular Life Sciences 72, no. 8 (2015): 1473–88.
20. Timmers, S., et al. “Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans.” Cell Metabolism 14, no. 5 (2011): 612–22; Morselli, E., et al. “Caloric restriction and resveratrol promote longevity through the sirtuin-1-dependent induction of autophagy.” Cell Death and Disease 1, no. 1 (2010): e10; Baur, J. A., et al. “Resveratrol improves health and survival of mice on a high-calorie diet.” Nature 444, no. 7117 (2006): 337–42.
21. Friedlander, B. “New York red wines show higher levels of resveratrol, a Cornell University study finds.” Cornell Chronicle, February 2, 1998, accessed September 1, 2015. www.news.cornell.edu/stories/1998/02/ny-red-wines-show-more-resveratrol.
22. Paganini-Hill, A., et al. “Dental health behaviors, dentition, and mortality in the elderly: the leisure world cohort study.” Journal of Aging Research (2011).
23. Olsen, I. “Update on bacteraemia related to dental procedures.” Transfusion and Apheresis Science 39, no. 2 (2008): 173–78.
24. Akaji, E. A., et al. “Halitosis: a review of the literature on its prevalence, impact and control.” Oral Health and Preventative Dentistry 12 (2014): 297–304.
25. Desvarieux, M., et al. “Periodontal microbiota and carotid intima-media thickness the oral infections and vascular disease epidemiology study (INVEST).” Circulation 111, no. 5 (2005): 576–82.
26. Yaacob, M., et al. “Powered versus manual toothbrushing for oral health.” Cochrane Database of Systematic Reviews 6 (2014).
27. Desvarieux, M., et al. “Gender differences in the relationship between periodontal disease, tooth loss, and atherosclerosis.” Stroke 35, no. 9 (2004): 2029–35; Wu, T., et al. “Periodontal disease and risk of cerebrovascular disease: the first national health and nutrition examination survey and its follow-up study.” Archives of Internal Medicine 160, no. 18 (2000): 2749–55.
28. Peedikayil, F. C., et al. “Effect of coconut oil in plaque related gingivitis-A preliminary report.” Nigerian Medical Journal: Journal of the Nigeria Medical Association 56, no. 2 (2015): 143; Asokan, S., et al. “Effect of oil pulling on plaque induced gingivitis: A randomized, controlled, triple-blind study.” Indian Journal of Dental Research 20, no. 1 (2009): 47; Roldan, S., et al. “Biofilms and the tongue: therapeutical approaches for the control of halitosis.” Clinical Oral Investigations 7, no. 4 (2003): 189–97; Asokan, S., et al. “Effect of oil pulling on Streptococcus mutans count in plaque and saliva using Dentocult SM Strip mutans test: A randomized, controlled, triple-blind study.” Journal of Indian Society of Pedodontics and Preventive Dentistry 26, no. 1 (2008): 12.
29. Sambunjak, D., et al. “Flossing for the management of periodontal diseases and dental caries in adults.” Cochrane Database of Systematic Reviews 12 (2011).
Chapter 6: Sleep—Week 2
1. He, Y., et al. “The transcriptional repressor DEC2 regulates sleep length in mammals.” Science 325, no. 5942 (2009): 866–70.
2. Gooley, J. J. “Circadian regulation of lipid metabolism.” The Proceedings of the Nutrition Society (2016): 1–11; Gooley, J. J., et al. “Diurnal regulation of lipid metabolism and applications of circadian lipidomics.” Journal of Genetics and Genomics 41, no. 5 (2014): 231–50; Horne, J. “The end of sleep: ‘sleep debt’ versus biological adaptation of human sleep to waking needs.” Biological Psychology 87, no. 1 (2011): 1–14; Jackson, M. L., et al. “Cognitive components of simulated driving performance: sleep loss effects and predictors.” Accident Analysis & Prevention 50 (2013): 438–44; McGrath, E., et al. “Sleep to lower elevated blood pressure: A randomized controlled trial (Slept).” Journal of Hypertension 34 (2016): e48; Wehr, T. A. “The durations of human melatonin secretion and sleep respond to changes in daylength (photoperiod).” Journal of Clinical Endocrinology & Metabolism 73, no. 6 (1991): 1276–80; Weintraub, K. “Ask well: Catching up on lost sleep,” New York Times, July 24, 2015, accessed October 22, 2015, http://well.blogs.nytimes.com/2015/07/24/ask-well-catching-up-on-lost-sleep/?_r=0.
3. Archer, S. N., et al. “How sleep and wakefulness influence circadian rhythmicity: effects of insufficient and mistimed sleep on the animal and human transcriptome.” Journal of Sleep Research 24, no. 5 (2015): 476–93.
4. Archer, S. N., et al. “Mistimed sleep disrupts circadian regulation of the human transcriptome.” Proceedings of the National Academy of Sciences 111, no. 6 (2014): E682–91.
5. Archer et al. “How sleep and wakefulness influence circadian rhythmicity.”
6. Tworoger, S. S., et al. “The association of self-reported sleep duration, difficulty sleeping, and snoring with cognitive function in older women.” Alzheimer Disease and Associated Disorders 20, no. 1 (2006): 41–48.
7. Ferrie, J. E., et al. “Change in sleep duration and cognitive function: findings from the Whitehall II Study.” Sleep 34, no. 5 (2011): 565.
8. Horne, J. “The end of sleep: ‘sleep debt’ versus biological adaptation of human sleep to waking needs.” Biological Psychology 87, no. 1 (2011): 1–14.
9. Panagiotakos, D. B., et al. “Sociodemographic and lifestyle statistics of oldest old people (> 80 years) living in Ikaria island: the Ikaria study.” Cardiology Research and Practice 2011 (2011).
10. Spiegel, K., et al. “Brief communication: Sleep curtailment in healthy young men is associated with decreased leptin levels, elevated ghrelin levels, and increased hunger and appetite.” Annals of Internal Medicine 141, no. 11 (2004): 846–50; Taheri, S., et al. “Short sleep duration is associated with reduced leptin, elevated ghrelin, and increased body mass index.” PLoS Medicine 1, no. 3 (2004): 210; Nedeltcheva, A. V., et al. “Sleep curtailment is accompanied by increased intake of calories from snacks.” American Journal of Clinical Nutrition 89, no. 1 (2009): 126–33; Hart, C. N., et al. “Changes in children’s sleep duration on food intake, weight, and leptin.” Pediatrics 132, no. 6 (2013): e1473–80; Kjeldsen, J. S., et al. “Short sleep duration and large variability in sleep duration are independently associated with dietary risk factors for obesity in Danish school children.” International Journal of Obesity 38, no. 1 (2014): 32–39; Leger, D., et al. “The role of sleep in the regulation of body weight.” Molecular and Cellular Endocrinology (2015); Capers, P. L., et al. “A systemic review and meta-analysis of randomized controlled trials of the impact of sleep duration on adiposity and components of energy balance.” Obesity Reviews 16, no. 9 (2015): 771–82; Broussard, J. L., et al. “Elevated ghrelin predicts food intake during experimental sleep restriction.” Obesity (2015).
11. Kim, T. W., et al. “The impact of sleep and circadian disturbance on hormones and metabolism.” International Journal of Endocrinology (2015).
12. Spira, A. P., et al. “Self-reported sleep and β-amyloid deposition in community-dwelling older adults.” JAMA Neurology 70, no. 12 (2013): 1537–43; Lim, A.S.P., et al. “Modification of the relationship of the apolipoprotein E ε4 allele to the risk of Alzheimer disease and neurofibrillary tangle density by sleep.” JAMA Neurology 70, no. 12 (2013): 1544–51.
13. Kripke, D. F., et al. “Hypnotics’ association with mortality or cancer: a matched cohort study.” British Medical Journal Open 2, no. 1 (2012): e000850; Kripke, D. F. “Mortality risk of hypnotics: strengths and limits of evidence.” Drug Safety (2015): 1–15; Mallon, L., et al. “Is usage of hypnotics associated with mortality?” Sleep Medicine 10, no. 3 (2009): 279–86.
14. Huedo-Medina, T. B., et al. “Effectiveness of non-benzodiazepine hypnotics in treatment of adult insomnia: meta-analysis of data submitted to the Food and Drug Administration.” British Medical Journal 345 (2012): e8343.
15. Cedernaes, J., et al. “Acute sleep loss induces tissue-specific epigenetic and transcriptional alterations to circadian clock genes in men.” Journal of Clinical Endocrinology and Metabolism 100, no. 9 (2015): E1255–61.
16. Jackson, M. L., et al. “Cognitive components of simulated driving performance: sleep loss effects and predictors.” Accident Analysis and Prevention 50 (2013): 438–44.
17. Rattue, G. “Night shift working ‘A probable human carcinogen.’” Medical News Today, October 28, 2011, accessed February 2, 2016. www.medicalnewstoday.com/articles/236731.php; “IARC monographs programme finds cancer hazards associated with shiftwork, painting and firefighting,” International Agency for Research on Cancer December 5, 2007, accessed February 2, 2016, www.iarc.fr/en/media-centre/pr/2007/pr180. html; “Shiftwork,” IARC Monographs on the Evaluation of Carcinogenic Risks to Humans 98 (2010), http://monographs.iarc.fr/ENG/Monographs/vol98/mono98–8.pdf.
18. Becker-Krail, D., et al. “Implications of circadian rhythm and stress in addiction vulnerability.” F1000Research 5 (2016).
19. Roehrs, T., et al. “Caffeine: sleep and daytime sleepiness.” Sleep Medicine Reviews 12, no. 2 (2008): 153–62.
20. Filipski, E., et al. “Effects of chronic jet lag on tumor progression in mice.” Cancer Research 64, no. 21 (2004): 7879–85.
21. Wirz-Justice, A., et al. “Circadian disruption and psychiatric disorders: the importance of entrainment.” Sleep Medicine Clinics 4, no. 2 (2009): 273–84; Davies, G., et al. “A systematic review of the nature and correlates of sleep disturbance in early psychosis.” Sleep Medicine Reviews (2016).
22. Cajochen, C., et al. “Evening exposure to a light-emitting-diode (LED) backlit computer screen affects circadian physiology and cognitive performance.” Journal of Applied Physiology 110, no. 5 (2011): 1432–38; Gooley, J. J., et al. “Exposure to room light before bedtime suppresses melatonin onset and shortens melatonin duration in humans.” Journal of Clinical Endocrinology and Metabolism 96, no. 3 (2010): E463–72; Vinogradova, I. A., et al. “Circadian disruption induced by light-at-night accelerates aging and promotes tumorigenesis in rats.” Aging 1, no. 10 (2009): 855.
23. Mallis, M. M., et al. “Circadian rhythms, sleep, and performance in space.” Aviation, Space, and Environmental Medicine 76, no. Supplement 1 (2005): B94–B107.
24. Altevogt, B. M., et al., eds. Sleep Disorders and Sleep Deprivation: An Unmet Public Health Problem (Washington, DC: National Academies Press, 2006).
25. Copinschi, G. “Metabolic and endocrine effects of sleep deprivation.” Essential Psychopharmacology 6, no. 6 (2004): 341–47.
26. Cohen, S., et al. “Sleep habits and susceptibility to the common cold.” Archives of Internal Medicine 169, no. 1 (2009): 62–67; Opp, M. R. “Sleep and psychoneuroimmunology.” Immunology and Allergy Clinics of North America 29, no. 2 (2009): 295–307; Krueger, J. M., et al. “Sleep, microbes and cytokines.” Neuroimmunomodulation 1, no. 2 (1994): 100–109.
27. Hill, S. M., et al. “Melatonin: an inhibitor of breast cancer.” Endocrine-Related Cancer (2015): ERC-15.
28. Stevens, R. G., et al. “Breast cancer and circadian disruption from electric lighting in the modern world.” CA: A Cancer Journal for Clinicians 64, no. 3 (2014): 207–18; Hansen, J., et al. “Case–control study of shift-work and breast cancer risk in Danish nurses: impact of shift systems.” European Journal of Cancer 48, no. 11 (2012): 1722–29; Knutsson, A., et al. “Breast cancer among shift workers: results of the WOLF longitudinal cohort study.” Scandinavian Journal of Work, Environment and Health 39, no. 2 (2013): 170–77; Rabstein, S., et al. “Night work and breast cancer estrogen receptor status-results from the German GENICA study.” Scandinavian Journal of Work, Environment and Health 39, no. 5 (2013): 448; Megdal, S. P., et al. “Night work and breast cancer risk: a systematic review and meta-analysis.” European Journal of Cancer 41, no. 13 (2005): 2023–32.
29. Kamdar, B. B., et al. “Night-shift work and risk of breast cancer: a systematic review and meta-analysis.” Breast Cancer Research and Treatment 138, no. 1 (2013): 291–301.
30. Maltese, F., et al. “Night shift decreases cognitive performance of ICU physicians.” Intensive Care Medicine (2015): 1–8.
31. Bøggild, H., et al. “Shift work, risk factors and cardiovascular disease.” Scandinavian Journal of Work, Environment & Health (1999): 85–99; Kawachi, I., et al. “Prospective study of shift work and risk of coronary heart disease in women.” Circulation 92, no. 11 (1995): 3178–82; Vetter, C., et al. “Association Between Rotating Night Shift Work and Risk of Coronary Heart Disease Among Women.” Journal of the American Medical Association 315, no. 16 (2016): 1726–34; Wang, A., et al. “Shift work and 20-year incidence of acute myocardial infarction: results from the Kuopio Ischemic Heart Disease Risk Factor Study.” Occupational and Environmental Medicine (2016): oemed-2015.
32. Brown, D. L., et al. “Rotating night shift work and the risk of ischemic stroke.” American Journal of Epidemiology (2009): kwp056.
33. Knutsson, A., et al. “Shift work and diabetes-A systematic review.” Chronobiology International 31, no. 10 (2014): 1146–51; Pan, A., et al. “Rotating night shift work and risk of type 2 diabetes: two prospective cohort studies in women.” PLoS Medicine 8, no. 12 (2011): 1660.
34. Bhatti, P., et al. “Nightshift work and risk of ovarian cancer.” Occupational and Environmental Medicine 70, no. 4 (2013): 231–37; Hammer, G. P., et al. “Shift work and prostate cancer incidence in industrial workers: A historical cohort study in a German chemical company.” Deutsches Ärzteblatt International 112, no. 27–28 (2015): 463; Hansen, J., et al. “Nested case-control study of night shift work and breast cancer risk among women in the Danish military.” Occupational and Environmental Medicine 69, no. 8 (2012): 551–56; Heikkila, K., et al. “Long working hours and cancer risk: a multi-cohort study.” British Journal of Cancer 114, no. 7 (2016): 813–18; Lin, X., “Night-shift work increases morbidity of breast cancer and all-cause mortality: a meta-analysis of 16 prospective cohort studies.” Sleep Medicine 16, no. 11 (2015): 1381–87; Papantoniou, K., et al. “Increased and mistimed sex hormone production in night shift workers.” Cancer Epidemiology Biomarkers & Prevention 24, no. 5 (2015): 854–63; Rao, D., et al. “Does night-shift work increase the risk of prostate cancer? a systematic review and meta-analysis.” OncoTargets and Therapy 8 (2015): 2817; Reszka, E., et al. “Circadian genes in breast cancer.” Advances in Clinical Chemistry (2016).
35. Hansen et al. “Case–control study of shift-work and breast cancer risk.”
36. Phipps, A. I., et al. “Sleep duration and quality may impact cancer survival rate.” Sleep 38 (2015).
37. Guarnieri, B., et al. “Sleep and cognitive decline: A strong bidirectional relationship. It is time for specific recommendations on routine assessment and the management of sleep disorders in patients with mild cognitive impairment and dementia.” European Neurology 74, nos. 1–2 (2015): 43–48.
38. Blackwell, T., et al. “Poor sleep is associated with impaired cognitive function in older women: the study of osteoporotic fractures.” Journals of Gerontology Series A: Biological Sciences and Medical Sciences 61, no. 4 (2006): 405–10.
39. Mander, B. A., et al. “Beta-amyloid disrupts human NREM slow waves and related hippocampus-dependent memory consolidation.” Nature Neuroscience (2015).
40. Musiek, E. S., et al. “Sleep, circadian rhythms, and the pathogenesis of Alzheimer Disease.” Experimental and Molecular Medicine 47, no. 3 (2015): e148.
41. Adan, A., et al. “Gender differences in morningness–eveningness preference.” Chronobiology International 19, no. 4 (2002): 709–20.
42. Duffy, J. F., et al. “Sex difference in the near-24-hour intrinsic period of the human circadian timing system.” Proceedings of the National Academy of Sciences 108, no. Suppl. 3 (2011): 15602–08; Lim, A. S. P., et al. “Sex difference in daily rhythms of clock gene expression in the aged human cerebral cortex.” Journal of Biological Rhythms 28, no. 2 (2013): 117–29.
43. Roenneberg, T., et al “Epidemiology of the human circadian clock.” Sleep Medicine Reviews 11, no. 6 (2007): 429–38.
44. Gominak, S. C., et al. “The world epidemic of sleep disorders is linked to vitamin D deficiency.” Medical Hypotheses 79, no. 2 (2012): 132–35.
45. Gray, M. G., et al. “Multiple integrated complementary healing approaches: Energetics and light for bone.” Medical Hypotheses 86 (2016): 18–29.
46. Shiue, I. “Low vitamin D levels in adults with longer time to fall asleep: US NHANES, 2005–2006.” International Journal of Cardiology 41 (2013): 20–21.
47. Grandner, M. A., et al. “Relationships among dietary nutrients and subjective sleep, objective sleep, and napping in women.” Sleep Medicine 11, no. 2 (2010): 180–84.
48. Massa, J., et al. “Vitamin D and actigraphic sleep outcomes in older community-dwelling men: the MrOS sleep study.” Sleep 38, no. 2 (2014): 251–57.
49. Beydoun, M. A., et al. “Serum nutritional biomarkers and their associations with sleep among US adults in recent national surveys.” PloS One 9, no. 8 (2014): e103490; Grandner, M. A., et al. “Sleep symptoms associated with intake of specific dietary nutrients.” Journal of Sleep Research 23, no. 1 (2014): 22–34.
50. Shipton, E. A., et al. “Vitamin D and pain: Vitamin D and its role in the aetiology and maintenance of chronic pain states and associated comorbidities.” Pain Research and Treatment 2015 (2015).
51. Balaban, H., et al. “Serum 25-hydroxyvitamin D levels in restless legs syndrome patients.” Sleep Medicine 13, no. 7 (2012): 953–57; Wali, S. et al. “The effect of vitamin D supplements on the severity of restless legs syndrome.” Sleep and Breathing 19, no. 2 (2015): 579–83; Gupta, R., et al. “Restless legs syndrome and pregnancy: prevalence, possible pathophysiological mechanisms and treatment.” Acta Neurologica Scandinavica (2015).
52. Beydoun et al. “Serum nutritional biomarkers.”
53. Xie, L., et al. “Sleep drives metabolite clearance from the adult brain.” Science 342, no. 6156 (2013): 373–77; Jessen, N.A., et al. “The glymphatic system: A beginner’s guide.” Neurochemical Research (2015): 1–17; Tarasoff-Conway, J. M., et al. “Clearance systems in the brain—implications for Alzheimer disease.” Nature Reviews Neurology 11, no. 8 (2015): 457–70; Mendelsohn, A. R., et al. “Sleep facilitates clearance of metabolites from the brain: glymphatic function in aging and neurodegenerative diseases.” Rejuvenation Research 16, no. 6 (2013): 518–23.
54. Lee, H., et al. “The effect of body posture on brain glymphatic transport.” Journal of Neuroscience 35, no. 31 (2015): 11034–44.
55. Wang, T. J., et al. “Common genetic determinants of vitamin D insufficiency: a genome-wide association study.” Lancet 376, no. 9736 (2010): 180–88.
56. Ross, A. C., et al., eds. Dietary reference intakes for calcium and vitamin D (Washington, DC: National Academies Press, 2010).
57. Crowley, S. J., et al. “Increased sensitivity of the circadian system to light in early/midpuberty.” Journal of Clinical Endocrinology and Metabolism 100, no. 11 (2015): 4067–73.
58. Tamakoshi, A., et al. “Self-reported sleep duration as a predictor of all-cause mortality: results from the JACC study, Japan.” Sleep 27, no. 1 (2004): 51–54; Hublin, C., et al. “Sleep and mortality: a population-based 22-year follow-up study.” Sleep 30, no. 10 (2007): 1245; Gallicchio, L., et al. “Sleep duration and mortality: a systematic review and meta analysis.” Journal of Sleep Research 18, no. 2 (2009): 148–58.
59. Youngstedt, S. D., et al. “Long sleep and mortality: rationale for sleep restriction.” Sleep Medicine Reviews 8, no. 3 (2004): 159–74.
60. Sofer, S., et al. “Greater weight loss and hormonal changes after 6 months diet with carbohydrates eaten mostly at dinner.” Obesity 19, no. 10 (2011): 2006–14; Sofer, S., et al. “Changes in daily leptin, ghrelin and adiponectin profiles following a diet with carbohydrates eaten at dinner in obese subjects.” Nutrition, Metabolism and Cardiovascular Diseases 23, no. 8 (2013): 744–50.
61. Richards, J., et al. “Higher serum vitamin D concentrations are associated with longer leukocyte telomere length in women.” American Journal of Clinical Nutrition 86, no. 5 (2007): 1420–25; Liu, J. J., et al. “Plasma vitamin D biomarkers and leukocyte telomere length.” American Journal of Epidemiology (2013): kws435.
62. Satlin, A., et al. “Bright light treatment of behavioral and sleep disturbances in patients with Alzheimer’s disease.” American Journal of Psychiatry 149 (1992): 1028–32; Mishima, K., et al. “Morning bright light therapy for sleep and behavior disorders in elderly patients with dementia.” Acta Psychiatrica Scandinavica 89, no. 1 (1994): 1–7; Stewart, K. T., et al. “Light treatment for NASA shiftworkers.” Chronobiology International 12, no. 2 (1995): 141–151; Mishima, K., et al. “Randomized, dim light controlled, crossover test of morning bright light therapy for rest-activity rhythm disorders in patients with vascular dementia and dementia of Alzheimer’s type.” Chronobiology International 15, no. 6 (1998): 647–54; Lyketsos, C. G., et al. “A randomized, controlled trial of bright light therapy for agitated behaviors in dementia patients residing in long-term care.” International Journal of Geriatric Psychiatry 14, no. 7 (1999): 520–25; Yamadera, H., et al. “Effects of bright light on cognitive and sleep–wake (circadian) rhythm disturbances in Alzheimer-type dementia.” Psychiatry and Clinical Neurosciences 54, no. 3 (2000): 352–53; Ancoli-Israel, S., et al. “Increased light exposure consolidates sleep and strengthens circadian rhythms in severe Alzheimer’s disease patients.” Behavioral Sleep Medicine 1, no. 1 (2003): 22–36; Fetveit, A., et al. “Bright light treatment improves sleep in institutionalised elderly—an open trial.” International Journal of Geriatric Psychiatry 18, no. 6 (2003): 520–26.
63. Lockley, S. W., et al. “High sensitivity of the human circadian melatonin rhythm to resetting by short wavelength light.” Journal of Clinical Endocrinology and Metabolism 88, no. 9 (2003): 4502–05; Sasseville, A., et al. “Wearing blue-blockers in the morning could improve sleep of workers on a permanent night schedule: a pilot study.” Chronobiology International 26, no. 5 (2009): 913–25; Wood, B., et al. “Light level and duration of exposure determine the impact of self-luminous tablets on melatonin suppression.” Applied Ergonomics 44, no. 2 (2013): 237–40; van der Lely, S., et al. “Blue blocker glasses as a countermeasure for alerting effects of evening light-emitting diode screen exposure in male teenagers.” Journal of Adolescent Health 56, no. 1 (2015): 113–19.
64. Duffy, J. F., et al. “Sex difference in the near-24-hour intrinsic period of the human circadian timing system.” Proceedings of the National Academy of Sciences 108, no. Supplement 3 (2011): 15602–8.
Chapter 7: Move—Week 3
1. “Health risks of physically strenuous work,” European Observatory of Working Life, March 8, 2005, accessed November 2, 2015, www.eurofound.europa.eu/observatories/eurwork/articles/working-conditions/health-risks-of-physically-strenuous-work; Künzler, G., et al. “Arme sterben früher: soziale Schicht, mortalität und rentenalterspolitik in der Schweiz.” Vol. 11. Caritas-Verlag (2002); “National census of fatal occupational injuries in 2014,” Bureau of Labor Statistics, US Department of Labor, September 17, 2015, accessed November 2, 2015, www.bls.gov/news.release/pdf/cfoi.pdf; Raley, D. “New NFL goal: A longer life,” Seattle Pi, May 8, 2008, accessed November 2, 2015, www.seattlepi.com/news/article/New-NFL-goal-A-longer-life-1272886.php.
2. Matthews, C. E., et al. “Amount of time spent in sedentary behaviors in the United States, 2003–2004.” American Journal of Epidemiology 167, no. 7 (2008): 875–81.
3. Patel, A. V., et al. “Leisure-time spent sitting and site-specific cancer incidence in a large US cohort.” Cancer Epidemiology Biomarkers and Prevention 24, no. 9 (2015): 1350–59.
4. Shibata, A., et al. “Physical activity, television viewing time and 12-year changes in waist circumference.” Medicine and Science in Sports and Exercise (2015); Chastin, S.F.M., et al. “Combined effects of time spent in physical activity, sedentary behaviors and sleep on obesity and cardio-metabolic health markers: A novel compositional data analysis approach.” PloS One 10, no. 10 (2015): e0139984.
5. Chastin et al. “Combined effects of time spent in physical activity”; Lamb, M. J. E., et al. “Prospective associations between sedentary time, physical activity, fitness and cardiometabolic risk factors in people with type 2 diabetes.” Diabetologia 59, no. 1 (2016): 110–20.
6. Wilmot, E. G., et al. “Sedentary time in adults and the association with diabetes, cardiovascular disease and death: systematic review and meta-analysis.” Diabetologia 55 (2012): 2895–05; Warburton, D., et al. “Health benefits of physical activity: the evidence.” Canadian Medical Association Journal 174, no. 6 (2006): 801–09.
7. Owen, N., et al. “Too much sitting: a novel and important predictor of chronic disease risk?” British Journal of Sports Medicine 43, no. 2 (2009): 81–83; Bauman, A., et al. “Leisure‐time physical activity alone may not be a sufficient public health approach to prevent obesity—a focus on China.” Obesity Reviews 9, no. s1 (2008): 119–26.
8. Chastin et al. “Combined effects of time spent in physical activity.”
9. Samitz, G, et al. “Domains of physical activity and all-cause mortality: systematic review and dose–response meta-analysis of cohort studies.” International Journal of Epidemiology 40, no. 5 (2011): 1382–1400; Hu, G., et al. “The effects of physical activity and body mass index on cardiovascular, cancer and all-cause mortality among 47 212 middle-aged Finnish men and women.” International Journal of Obesity 29, no. 8 (2005): 894–902. Schnohr, P., et al. “Longevity in male and female joggers: the Copenhagen City Heart Study.” American Journal of Epidemiology 177, no. 7 (2013): 683–89.
10. Oguma, Y., et al. “Physical activity and all-cause mortality in women: a review of the evidence.” British Journal of Sports Medicine 36, no. 3 (2002): 162–72.
11. Gregg, E. W., et al. “Relationship of changes in physical activity and mortality among older women.” JAMA 289, no. 18 (2003): 2379–86.
12. Stanford, K. I., et al. “Exercise effects on white adipose tissue: Beiging and metabolic adaptations.” Diabetes 64, no. 7 (2015): 2361–68.
13. Levine, H. J. “Rest heart rate and life expectancy.” Journal of the American College of Cardiology 30, no. 4 (1997): 1104–6.
14. “Physical activity guidelines,” Health.Gov, accessed December 7, 2015, http://health.gov/paguidelines.
15. Schnohr, P., et al. “Dose of jogging and long-term mortality: the Copenhagen City Heart Study.” Journal of the American College of Cardiology 65, no. 5 (2015): 411–19.
16. Lavie, C. J., et al. “Effects of running on chronic diseases and cardiovascular and all-cause mortality.” Mayo Clinic Proceedings 90, no. 11 (2015): 1541–52.
17. Day, S. M., et al. “Cardiac risks associated with marathon running.” Sports Health: A Multidisciplinary Approach 2, no. 4 (2010): 301–6; Kim, J. H., et al. “Cardiac arrest during long-distance running races.” New England Journal of Medicine 366, no. 2 (2012): 130–40; Hart, L. “Marathon-related cardiac arrest.” Clinical Journal of Sport Medicine 23, no. 5 (2013): 409–10.
18. Du, M., et al. “Physical activity, sedentary behavior, and leukocyte telomere length in women.” American Journal of Epidemiology (2012): kwr330.
19. Ibid.
20. Ibid.; Krishna, B. H., et al. “Association of leukocyte telomere length with oxidative stress in yoga practitioners.” Journal of Clinical and Diagnostic Research: JCDR 9, no. 3 (2015): CC01.
21. Martyn–St. James, M., et al. “Meta-analysis of walking for preservation of bone mineral density in postmenopausal women.” Bone 43, no. 3 (2008): 521–31.
22. Zhao, R., et al. “The effects of differing resistance training modes on the preservation of bone mineral density in postmenopausal women: a meta-analysis.” Osteoporosis International 26, no. 5 (2015): 1605–18.
23. Patel, N. K., et al. “The effects of yoga on physical functioning and health related quality of life in older adults: a systematic review and meta-analysis.” Journal of Alternative and Complementary Medicine 18, no. 10 (2012): 902–17; Phoosuwan, M., et al. “The effects of weight bearing yoga training on the bone resorption markers of the postmenopausal women.” Chotmaihet Thangphaet [Journal of the Medical Association of Thailand] 92 (2009): S102–8.
24. Melov, S. et al. “Resistance exercise reverses aging in human skeletal muscle.” PLoS One 2, no. 5 (2007): e465.
25. Lee, J. A., et al. “Effects of yoga exercise on serum adiponectin and metabolic syndrome factors in obese postmenopausal women.” Menopause 19, no. 3 (2012): 296–301.
26. Watson, K., et al. “MTOR and the health benefits of exercise.” Seminars in Cell and Developmental Biology, no. 36, (2014): 130–39.
27. Markofski, M. M., et al. “Effect of age on basal muscle protein synthesis and mTORC1 signaling in a large cohort of young and older men and women.” Experimental Gerontology 65 (2015): 1–7.
28. Rönn, T., et al. “A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue.” PLoS Genetics 9, no. 6 (2013): e1003572.
29. Vimaleswaran, K. S., et al. “Physical activity attenuates the body mass index–increasing influence of genetic variation in the FTO gene.” American Journal of Clinical Nutrition 90, no. 2 (2009): 425–28; Kilpeläinen, T. O., et al. “Physical activity attenuates the influence of FTO variants on obesity risk: a meta-analysis of 218,166 adults and 19,268 children.” PLoS Medicine 8, no. 11 (2011): e1001116; Shengxu, L., et al. “Cumulative effects and predictive value of common obesity-susceptibility variants identified by genome-wide association studies.” American Journal of Clinical Nutrition 91, no. 1 (2010): 184–90.
30. Kilpeläinen, T. O., et al. “Genome-wide meta-analysis uncovers novel loci influencing circulating leptin levels.” Nature Communications 7 (2016).
31. Dupuis, J., et al. “New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk.” Nature Genetics 42, no. 2 (2010): 105–16; Strawbridge, R. J., et al. “Genome-wide association identifies nine common variants associated with fasting proinsulin levels and provides new insights into the pathophysiology of type 2 diabetes.” Diabetes 60, no. 10 (2011): 2624–34; Teran-Garcia, M., et al. “Hepatic lipase gene variant— 514C>T is associated with lipoprotein and insulin sensitivity response to regular exercise.” Diabetes 54, no. 7 (2005): 2251–55.
32. Ahmad, T., et al. “Physical activity modifies the effect of LPL, LIPC, and CETP polymorphisms on HDL-C levels and the risk of myocardial infarction in women of European ancestry.” Circulation: Cardiovascular Genetics 4, no. 1 (2011): 74–80.
33. Barres, R., et al. “Acute exercise remodels promoter methylation in human skeletal muscle.” Cell Metabolism 15, no. 3 (2012): 405–11.
34. Hargreaves, M. “Exercise and Gene Expression.” Progress in Molecular Biology and Translational Science 135 (2015): 457–69.
35. Bratman, G. N., et al. “The benefits of nature experience: Improved affect and cognition.” Landscape and Urban Planning 138 (2015): 41–50.
36. Bratman, G. N., et al. “Nature experience reduces rumination and subgenual prefrontal cortex activation.” Proceedings of the National Academy of Sciences 112, no. 28 (2015): 8567–72.
37. Song, C., et al. “Physiological and psychological responses of young males during springtime walks in urban parks.” Journal of Physiological Anthropology 33, no. 8 (2014); Song, C., et al. “Physiological and psychological effects of walking on young males in urban parks in winter.” Journal of Physiological Anthropology 32, no. 1 (2013): 18; Song, C., et al. “Effect of forest walking on autonomic nervous system activity in middle-aged hypertensive individuals: A pilot study.” International Journal of Environmental Research and Public Health 12, no. 3 (2015): 2687–99.
38. Gammon, M. D., et al. “Recreational and occupational physical activities and risk of breast cancer.” Journal of the National Cancer Institute 90, no. 2 (1998): 100–117.
39. Dorn, J., et al. “Lifetime physical activity and breast cancer risk in pre-and postmenopausal women.” Medicine and Science in Sports and Exercise 35, no. 2 (2003): 278–85.
40. Patel, A. V., et al. “Recreational physical activity and risk of postmenopausal breast cancer in a large cohort of US women.” Cancer Causes and Control 14, no. 6 (2003): 519–29.
41. Patel, A. V., et al. “Lifetime recreational exercise activity and risk of breast carcinoma in situ.” Cancer 98, no. 10 (2003): 2161–69; Lu, Y., et al. “History of recreational physical activity and survival after breast cancer: the California Breast Cancer Survivorship Consortium.” American Journal of Epidemiology (2015): kwu466; Warburton et al. “Health benefits of physical activity.”
42. Lek, M., et al. “Analysis of protein-coding genetic variation in 60,706 humans.” bioRxiv (2015): 030338.
43. Rankinen, T., et al. “Effect of endothelin 1 genotype on blood pressure is dependent on physical activity or fitness levels.” Hypertension 50, no. 6 (2007): 1120–25; Li, T. C., et al. “Associations of EDNRA and EDN1 polymorphisms with carotid intima media thickness through interactions with gender, regular exercise, and obesity in subjects in Taiwan: Taichung Community Health Study (TCHS).” BioMedicine 5, no. 2 (2015).
44. Yaffe, K., et al. “A prospective study of physical activity and cognitive decline in elderly women: women who walk.” Archives of Internal Medicine 161, no. 14 (2001): 1703–8.
45. Weuve, J., et al. “Physical activity, including walking, and cognitive function in older women.” JAMA 292, no. 12 (2004): 1454–61.
46. Zulfiqar, U., et al. “Relation of high heart rate variability to healthy longevity.” American Journal of Cardiology 105, no. 8 (2010): 1181–85.
47. Blomstrand, A., et al. “Effects of leisure time physical activity on well-being among women in a 32-year perspective.” Scandinavian Journal of Public Health (2009).
48. Crane, J. D., et al. “Exercise‐stimulated interleukin‐15 is controlled by AMPK and regulates skin metabolism and aging.” Aging Cell (2015).
49. Li, F., et al. “Tai chi and self‐rated quality of sleep and daytime sleepiness in older adults: A randomized controlled trial.” Journal of the American Geriatrics Society 52, no. 6 (2004): 892–900.
50. Irwin, M. R., et al. “Tai chi, cellular inflammation, and transcriptome dynamics in breast cancer survivors with insomnia: a randomized controlled trial.” Journal of the National Cancer Institute. Monographs 2014, no. 50 (2014): 295–301.
51. Buman, M. P., et al. “Does nighttime exercise really disturb sleep? Results from the 2013 National Sleep Foundation Sleep in America poll.” Sleep Medicine 15, no. 7 (2014): 755–61; Qian-Chun, Y., et al. “Impact of evening exercise on college students’ sleep quality.” Zhonghua Yu Fang Yi Xue Za Zhi [Chinese Journal of Preventive Medicine] 47, no. 6 (2013): 542–46; Brand, S., et al. “High self-perceived exercise exertion before bedtime is associated with greater objectively assessed sleep efficiency.” Sleep Medicine 15, no. 9 (2014): 1031–36. Alley, J. R., et al. “Effects of resistance exercise timing on sleep architecture and nocturnal blood pressure.” Journal of Strength and Conditioning Research 29, no. 5 (2015): 1378–85.
52. Yang, P. Y., et al. “Exercise training improves sleep quality in middle-aged and older adults with sleep problems: a systematic review.” Journal of Physiotherapy 58, no. 3 (2012): 157–63.
53. Fiatarone, M. A., et al. “High-intensity strength training in nonagenarians: effects on skeletal muscle.” Journal of the American Medical Association 263, no. 22 (1990): 3029–34.
54. 58 Poirier, P. “Exercise, heart rate variability, and longevity: the cocoon mystery?” Circulation 129 (2014): 2085–87; U.S. Department of Health and Human Services. Physical Activity Guidelines for Americans: Be Active, Healthy and Happy (Washington, DC: US Department of Health and Human Services, 2008); Myers, J., et al. “Exercise capacity and mortality among men referred for exercise testing.” New England Journal of Medicine 346, no. 11 (2002): 793–801; Gulati, M., et al. “The prognostic value of a nomogram for exercise capacity in women.” New England Journal of Medicine 353, no. 5 (2005): 468–75.
55. Reynolds, G. “Walk hard. Walk easy. Repeat,” New York Times, February 19, 2015, accessed September 20, 2015, http://well.blogs.nytimes.com/2015/02/19/walk-hard-walk-easy-repeat/?_r=0; Masuki, S., et al. “The factors affecting adherence to a long-term interval walking training program in middle-aged and older people.” Journal of Applied Physiology 118, no. 5 (2015): 595–603.
56. Esmarck, B., et al. “Timing of post exercise protein intake is important for muscle hypertrophy with resistance training in elderly humans.” Journal of Physiology 535, no. 1 (2001): 301–11.
57. Buchheit, M., et al. “Parasympathetic reactivation after repeated sprint exercise.” American Journal of Physiology-Heart and Circulatory Physiology 293, no. 1 (2007): H133-H141; Bishop, P. A., et al. “Recovery from training: a brief review: brief review.” Journal of Strength and Conditioning Research 22, no. 3 (2008): 1015–24; Gisselman, A. et al. “Musculoskeletal overuse injuries and heart rate variability: Is there a link?” Medical Hypotheses 87 (2016): 1–7; Mayo, X., et al. “Exercise type affects cardiac vagal autonomic recovery after a resistance training session.” Journal of Strength and Conditioning Research / National Strength and Conditioning Association (2016); Vernillo, G., et al. “Postexercise autonomic function after repeated-sprints training.” European Journal of Applied Physiology 115, no. 11 (2015): 2445–55.
58. Gupta, S., et al. “Cardiorespiratory fitness and classification of risk of cardiovascular disease mortality.” Circulation 123, no. 13 (2011): 1377–83; Berry, J. D., et al. “Lifetime risks for cardiovascular disease mortality by cardiorespiratory fitness levels measured at ages 45, 55, and 65 years in men: the Cooper Center Longitudinal Study.” Journal of the American College of Cardiology 57, no. 15 (2011): 1604–10.
59. Waygood, E., et al. “Active travel by built environment and lifecycle stage: case study of Osaka metropolitan area.” International Journal of Environmental Research and Public Health 12, no. 12 (2015): 15900–24; Oja, P., et al. “Health benefits of cycling: a systematic review.” Scandinavian Journal of Medicine and Science in Sports 21, no. 4 (2011): 496–509; Chiu, M, et al. “Moving to a Highly Walkable Neighborhood and Incidence of Hypertension: A Propensity-score Matched Cohort Study.” Circulation 132, no. Suppl. 3 (2015): A11545.
60. Martin, A., et al. “Does active commuting improve psychological wellbeing? Longitudinal evidence from eighteen waves of the British Household Panel Survey.” Preventive Medicine 69 (2014): 296–303; Gatersleben, B., et al. “Affective appraisals of the daily commute comparing perceptions of drivers, cyclists, walkers, and users of public transport.” Environment and Behavior 39, no. 3 (2007): 416–31.
Chapter 8: Release—Week 4
1. Krishna, B. H., et al. “Association of leukocyte telomere length with oxidative stress in yoga practitioners.” Journal of Clinical and Diagnostic Research: JCDR 9, no. 3 (2015): CC01; Balasubramanian, S., et al. “Induction of salivary nerve growth factor by Yogic breathing: a randomized controlled trial.” International Psychogeriatrics 27, no. 1 (2015): 168–70; Bower, J. E., et al. “Yoga reduces inflammatory signaling in fatigued breast cancer survivors: a randomized controlled trial.” Psychoneuroendocrinology 43 (2014): 20–29; Qu, S., et al. “Rapid gene expression changes in peripheral blood lymphocytes upon practice of a comprehensive yoga program.” PloS One 8, no. 4 (2013): e61910; Sharma, H., et al. “Sudarshan Kriya practitioners exhibit better antioxidant status and lower blood lactate levels.” Biological Psychology 63, no. 3 (2003): 281–91.
2. Inanir, A., et al. “Clinical symptoms in fibromyalgia are associated to catechol-O-methyltransferase (COMT) gene Val158Met polymorphism.” Xenobiotica 44, no. 10 (2014): 952–56.
3. Vossen, H., et al. “The genetic influence on the cortical processing of experimental pain and the moderating effect of pain status.” PLoS One 5, no. 10 (2010): e13641; Nijs, J., et al. “Brain-derived neurotrophic factor as a driving force behind neuroplasticity in neuropathic and central sensitization pain: a new therapeutic target?” Expert Opinion on Therapeutic Targets 19, no. 4 (2015): 565–76.
4. Forrest, A. Fierce Medicine (San Francisco: HarperOne, 2012).
5. For more information on Forrest yoga, visit www.forrestyoga.com.
6. Kogan, L. “Oprah’s new workout and what happened when we tried it).” Oprah.com, January 2016, accessed January 26, 2016, www.oprah.com/health/Oprahs-New-Workout-and-What-Happened-When-We-Tried-It.
7. Bougea, A. M., et al. “Effect of the emotional freedom technique on perceived stress, quality of life, and cortisol salivary levels in tension-type headache sufferers: a randomized controlled trial.” Explore: Journal of Science and Healing 9, no. 2 (2013): 91–99.
8. Church, D., et al. “The effect of emotional freedom techniques on stress biochemistry: a randomized controlled trial.” Journal of Nervous and Mental Disease 200, no. 10 (2012): 891–96.
9. Brattberg, G. “Self-administered EFT (Emotional Freedom Techniques) in individuals with fibromyalgia: a randomized trial.” Integrative Medicine 7, no. 4 (2008): 30–35.
10. Boath, E., et al. “A narrative systematic review of the effectiveness of Emotional Freedom Techniques (EFT).” Staffordshire University, CPSI Monograph, Centre for Practice and Service Improvement (2012).
11. Ibid.
12. Upledger, J. E., et al. CranioSacral Therapy: What It Is, How It Works (Berkeley: North Atlantic Books, 2008), 103.
13. Henschke, N., et al. “Stretching before or after exercise does not reduce delayed-onset muscle soreness.” British Journal of Sports Medicine 45, no. 15 (2011): 1249–50.
14. Simic, L., N. et al. “Does pre‐exercise static stretching inhibit maximal muscular performance? A meta‐analytical review.” Scandinavian Journal of Medicine and Science in Sports 23, no. 2 (2013): 131–48.
15. Cheatham, S. W., et al. “The effects of self-myofascial release using a foam roll or roller massager on joint range of motion, muscle recovery and performance: A systematic review.” International Journal of Sports Physical Therapy 10, no. 6 (2015): 827.
16. Beardsley, C., et al. “Effects of self-myofascial release: A systematic review.” Journal of Bodywork and Movement Therapies 19, no. 4 (2015): 747–58.
17. Chen, Y. H., et al. “Increased sliding of transverse abdominis during contraction after myofascial release in patients with chronic low back pain.” Manual Therapy (2015).
18. Bleakley, C., et al. “Cold-water immersion for preventing and treating muscle soreness after exercise.” Cochrane Library (2012); Costello, J. T., et al. “Whole-body cryotherapy (extreme cold air exposure) for preventing and treating muscle soreness after exercise in adults.” Cochrane Library (2015).
19. Bleakley, C. M., et al. “What is the biochemical and physiological rationale for using Cold Water Immersion in Sports Recovery? A Systematic Review.” British Journal of Sports Medicine (2009): bjsm-2009.
20. Van der Kolk, B. The Body Keeps the Score (New York: Viking, 2014), 356.
21. Ferris, T. “Relax like a pro: 5 steps to hacking your sleep,” The Tim Ferris Experiment, January 27, 2008, accessed February 1, 2016, http://fourhourworkweek.com/2008/01/27/relax-like-a-pro-5-steps-to-hacking-your-sleep.
22. Hamblin, J. “The benefits of being cold,” Atlantic, January 2015, accessed February 1, 2016, www.theatlantic.com/magazine/archive/2015/01/does-global-warming-make-me-look-fat/383509.
23. Zhornitsky, S., et al. “Cannabidiol in humans—the quest for therapeutic targets,” Pharmaceuticals 5, no. 5 (2012): 529–52; Welty, T. E., et al. “Cannabidiol: promise and pitfalls,” Epilepsy Currents 14, no. 5 (2014): 250–52; and Fasinu, P. S., et al. “Current status and prospects for cannabidiol preparations as new therapeutic agents,” Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy (2016).
Chapter 9: Expose—Week 5
1. Gore, A. C., et al. “Executive summary to EDC-2: The Endocrine Society’s second scientific statement on endocrine-disrupting chemicals.” Endocrine Reviews 36, no. 6 (2015): 593–602.
2. Christensen, K., et al. “Ageing populations: the challenges ahead.” Lancet 374, no. 9696 (2009): 1196–1208.
3. Wild, C. P. “Complementing the genome with an ‘exposome’: the outstanding challenge of environmental exposure measurement in molecular epidemiology.” Cancer Epidemiology Biomarkers and Prevention 14, no. 8 (2005): 1847–50.
4. Coughlin, S. S., et al. “The impact of the natural, social, built, and policy environments on breast cancer.” Journal of Environment and Health Sciences 1, no. 3 (2015); Land, C. E. “Studies of cancer and radiation dose among atomic bomb survivors: the example of breast cancer.” JAMA 274, no. 5 (1995): 402–7; Hancock, S. L., et al. “Breast cancer after treatment of Hodgkin’s disease.” Journal of the National Cancer Institute 85, no. 1 (1993): 25–31.
5. Inspired in part by the International Living Future Institute’s red list of building materials that contain harmful substances. “Materials red list,” International Living Future Institute, accessed February 6, 2016, http://living-future.org/topic/materials-red-list.
6. Kojima, H., et al. “In vitro endocrine disruption potential of organophosphate flame retardants via human nuclear receptors.” Toxicology 314, no. 1 (2013): 76–83.
7. Liu, X., et al. “Endocrine disruption potentials of organophosphate flame retardants and related mechanisms in H295R and MVLN cell lines and in zebrafish.” Aquatic Toxicology 114 (2012): 173–81.
8. Su, G., et al. “Rapid in vitro metabolism of the flame retardant triphenyl phosphate and effects on cytotoxicity and mRNA expression in chicken embryonic hepatocytes.” Environmental Science and Technology 48, no. 22 (2014): 13511–19.
9. Mendelsohn, E., et al. “Nail polish as a source of exposure to triphenyl phosphate.” Environment International 86 (2016): 45–51.
10. Calle, E. E., et al. “Organochlorines and breast cancer risk.” CA: A Cancer Journal for Clinicians 52, no. 5 (2002): 301–9; Hertz-Picciotto, I., ed. Breast Cancer and the Environment: A Life Course Approach (National Academies Press, 2012); Ekenga, C. C., et al. “Breast cancer risk after occupational solvent exposure: The influence of timing and setting.” Cancer Research 74, no. 11 (2014): 3076–83; Labrèche, F., et al. “Postmenopausal breast cancer and occupational exposures.” Occupational and Environmental Medicine 67, no. 4 (2010): 263–69; Millikan, R., et al. “Dichlorodiphenyldichloroethene, polychlorinated biphenyls, and breast cancer among African-American and white women in North Carolina.” Cancer Epidemiology Biomarkers and Prevention 9, no. 11 (2000): 1233–40; Krieger, N., et al. “Breast cancer and serum organochlorines: a prospective study among white, black, and Asian women.” Journal of the National Cancer Institute 86, no. 8 (1994): 589–99.
11. Kochan, D. Z., et al. “Circadian disruption and breast cancer: An epigenetic link?” Oncotarget 6, no. 19 (2015): 16866.
12. Heikkinen, S., et al. “Does hair dye use increase the risk of breast cancer? A population-based case-control study of Finnish women.” PloS One 10, no. 8 (2015): e0135190.
13. Rollison, D. E., et al. “Personal hair dye use and cancer: a systematic literature review and evaluation of exposure assessment in studies published since 1992.” Journal of Toxicology and Environmental Health, Part B 9, no. 5 (2006): 413–39.
14. Takkouche, B., et al. “Personal use of hair dyes and risk of cancer: a meta-analysis.” JAMA 293, no. 20 (2005): 2516–25.
15. Takkouche, B., et al. “Risk of cancer among hairdressers and related workers: a metaanalysis.” International Journal of Epidemiology 38, no. 6 (2009): 1512–31.
16. “Progress cleaning the air and improving people’s health,” United States Environmental Protection Agency, accessed February 7, 2016, www.epa.gov/clean-air-act-overview/progress-cleaning-air-and-improving-peoples-health.
17. “Southern California air regulators fail to make decision on methane gas leak,” Fox News, January 17, 2016, accessed February 7, 2016, www.foxnews.com/us/2016/01/17/southern-california-air-regulators-fail-to-make-decision-on-methane-gas-leak.html; “Methane from massive gas leak in Porter Ranch is boosting global warming: Experts,” Los Angeles Times, January 24, 2016, accessed February 7, 2016, http://ktla.com/2016/01/24/methane-from-massive-gas-leak-in-porter-ranch-is-boosting-global-warming-experts/; “As California methane leak displaces thousands, will U.S. regulate natural gas sites nationwide,” Democracy Now, January 14, 2016, accessed February 7, 2016, www.democracynow. org/2016/1/14/as_california_methane_leak_displaces_thousands.
18. “Erin Brockovich: California methane gas leak is worst U.S. environmental disaster since BP oil spill,” Democracy Now, December 30, 2015, accessed February 7, 2016, www.democracy now.org/2015/12/30/erin_brockovich_california_methane_gas_leak.
19. Bell, M. L., et al. “Ozone and short-term mortality in 95 US urban communities, 1987–2000.” JAMA 292, no. 19 (2004): 2372–78; Gryparis, A., et al. “Acute effects of ozone on mortality from the ‘air pollution and health: a European approach’ project.” American Journal of Respiratory and Critical Care Medicine 170, no. 10 (2004): 1080–87; Bell, M. L., et al. “A meta-analysis of time-series studies of ozone and mortality with comparison to the national morbidity, mortality, and air pollution study.” Epidemiology 16, no. 4 (2005): 436; Levy, J. I., et al. “Ozone exposure and mortality: an empiric bayes metaregression analysis.” Epidemiology 16, no. 4 (2005): 458–68; Ito, K., et al. “Associations between ozone and daily mortality: analysis and meta-analysis.” Epidemiology 16, no. 4 (2005): 446–57; Zanobetti, A., et al. “Mortality displacement in the association of ozone with mortality: an analysis of 48 cities in the United States.” American Journal of Respiratory and Critical Care Medicine 177, no. 2 (2008): 184–89; Katsouyanni, K., et al. “Air pollution and health: a European and North American approach (APHENA).” Research Report (Health Effects Institute) 142 (2009): 5–90; Samoli, E., et al. “The temporal pattern of mortality responses to ambient ozone in the APHEA project.” Journal of Epidemiology and Community Health 63, no. 12 (2009): 960–66.
20. Pope III, C. A., et al. “Health effects of fine particulate air pollution: lines that connect.” Journal of the Air and Waste Management Association 56, no. 6 (2006): 709–42; Seaton, A., et al. “Particulate air pollution and acute health effects.” Lancet 345, no. 8943 (1995): 176–78; Kim, J. J. “Ambient air pollution: health hazards to children.” Pediatrics 114, no. 6 (2004): 1699–1707; “Health effects of ozone and particle pollution,” State of the Air, accessed December 28, 2015, www.stateoftheair.org/2013/health-risks.
21. Calderón-Garcidueñas, L., et al. “Mexico City normal weight children exposed to high concentrations of ambient PM 2.5 show high blood leptin and endothelin-1, vitamin D deficiency, and food reward hormone dysregulation versus low pollution controls. Relevance for obesity and Alzheimer disease.” Environmental Research 140 (2015): 579–92.
22. Morris, B. J., et al. “FOXO3: A major gene for human longevity—a mini-review.” Gerontology (2015); Singh, R., et al. “Anti-inflammatory heat shock protein 70 genes are positively associated with human survival.” Current Pharmaceutical Design 16, no. 7 (2010): 796.
23. Laukkanen, T., et al. “Association between sauna bathing and fatal cardiovascular and all-cause mortality events.” JAMA Internal Medicine 175, no. 4 (2015): 542–48.
24. Kenttämies, A., et al. “Death in sauna.” Journal of Forensic Sciences 53, no. 3 (2008): 724–29.
25. Scoon, G. S., et al. “Effect of post-exercise sauna bathing on the endurance performance of competitive male runners.” Journal of Science and Medicine in Sport 10, no. 4 (2007): 259–62.
26. Stanley, J., et al. “Effect of sauna-based heat acclimation on plasma volume and heart rate variability.” European Journal of Applied Physiology 115, no. 4 (2015): 785–94.
27. Krause, M., et al. “Heat shock proteins and heat therapy for type 2 diabetes: pros and cons.” Current Opinion in Clinical Nutrition and Metabolic Care 18, no. 4 (2015): 374–80.
28. Kukkonen-Harjula, K., et al. “Haemodynamic and hormonal responses to heat exposure in a Finnish sauna bath.” European Journal of Applied Physiology and Occupational Physiology 58, no. 5 (1989): 543–50.
29. Gryka, D., et al. “The effect of sauna bathing on lipid profile in young, physically active, male subjects.” International Journal of Occupational Medicine and Environmental Health 27, no. 4 (2014): 608–18; Pilch, W., et al. “Changes in the lipid profile of blood serum in women taking sauna baths of various duration.” International Journal of Occupational Medicine and Environmental Health 23, no. 2 (2010): 167–74; van der Wall, E. E. “Sauna bathing: a warm heart proves beneficial.” Netherlands Heart Journal 23, no. 5 (2015): 247.
30. Tomiyama, C., et al. “The effect of repetitive mild hyperthermia on body temperature, the autonomic nervous system, and innate and adaptive immunity.” Biomedical Research 36, no. 2 (2015): 135–42.
31. Hooper, L. V. “You AhR what you eat: linking diet and immunity.” Cell 147, no. 3 (2011): 489–91.
32. Li, Y., et al. “Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation.” Cell 147, no. 3 (2011): 629–40.
33. Nishi, K., et al. “Immunostimulatory in vitro and in vivo effects of a water-soluble extract from kale.” Bioscience, Biotechnology, and Biochemistry 75, no. 1 (2011): 40–46.
34. Haddad, E. H., et al. “Effect of a walnut meal on postprandial oxidative stress and antioxidants in healthy individuals.” Nutrition Journal 13, no. 1 (2014): 1.
35. Cominetti, C., et al. “Associations between glutathione peroxidase-1 Pro198Leu polymorphism, selenium status, and DNA damage levels in obese women after consumption of Brazil nuts.” Nutrition 27, no. 9 (2011): 891–96.
36. Song, J. M., et al. “Antiviral effect of catechins in green tea on influenza virus.” Antiviral Research 68, no. 2 (2005): 66–74; Hsu, S. “Compounds derived from epigallocatechin-3-gallate (EGCG) as a novel approach to the prevention of viral infections.” Inflammation and Allergy-Drug Targets 14, no. 1 (2015): 13–18; Rowe, C. A., et al. “Specific formulation of Camellia Sinensis prevents cold and flu symptoms and enhances γδ T cell function: a randomized, double-blind, placebo-controlled study.” Journal of the American College of Nutrition 26, no. 5 (2007): 445–52.
37. Mak, J.C.W. “Potential role of green tea catechins in various disease therapies: progress and promise.” Clinical and Experimental Pharmacology and Physiology 39, no. 3 (2012): 265–73; Apetz, N., et al. “Natural compounds and plant extracts as therapeutics against chronic inflammation in Alzheimer’s disease-a translational perspective.” CNS and Neurological Disorders-Drug Targets (Formerly Current Drug Targets-CNS and Neurological Disorders) 13, no. 7 (2014): 1175–91; Chen, G., et al. “Nutraceuticals and functional foods in the management of hyperlipidemia.” Critical Reviews in Food Science and Nutrition 54, no. 9 (2014): 1180–1201; Johnson, R., et al. “Green tea and green tea catechin extracts: an overview of the clinical evidence.” Maturitas 73, no. 4 (2012): 280–87. “Green tea extract for external anogenital warts.” Drug and Therapeutics Bulletin 53, no. 10 (2015): 114–16; Gupta, A. K., et al. “Sinecatechins 10% ointment: A green tea extract for the treatment of external genital warts.” Pain 46 (2015): 14–15; Tatti, S., et al. “Sinecatechins, a defined green tea extract, in the treatment of external anogenital warts: a randomized controlled trial.” Obstetrics and Gynecology 111, no. 6 (2008): 1371–79.
38. Marinac, C. R., et al. “Frequency and circadian timing of eating may influence biomarkers of inflammation and insulin resistance associated with breast cancer risk.” PloS One 10, no. 8 (2015): e0136240; Marinac, C. R., et al. “Prolonged nightly fasting and breast cancer risk: Findings from NHANES (2009–2010).”Cancer Epidemiology Biomarkers and Prevention 24, no. 5 (2015): 783–89.
39. Beitner, H. “Randomized, placebo‐controlled, double blind study on the clinical efficacy of a cream containing 5% α‐lipoic acid related to photoageing of facial skin.” British Journal of Dermatology 149, no. 4 (2003): 841–49.
40. Moura, F. A., et al. “Lipoic acid: its antioxidant and anti-inflammatory role and clinical applications.” Current Topics in Medicinal Chemistry 15, no. 5 (2015): 458–83; Patel, M. K., et al. “Can α-lipoic acid mitigate progression of aging-related decline caused by oxidative stress?” Southern Medical Journal 107, no. 12 (2014): 780–87.
41. Maczurek, A., et al. “Lipoic acid as an anti-inflammatory and neuroprotective treatment for Alzheimer’s disease.” Advanced Drug Delivery Reviews 60, no. 13 (2008): 1463–70.
42. Roberts, J. L., et al. “Emerging role of alpha-lipoic acid in the prevention and treatment of bone loss.” Nutrition Reviews 73, no. 2 (2015): 116–25.
43. Kouzi, S. A., et al. “Natural supplements for improving insulin sensitivity and glucose uptake in skeletal muscle.” Frontiers in Bioscience 7 (2014): 94–106; Lee, T., et al. “Nutritional supplements and their effect on glucose control.” Advances in Experimental Medicine and Biology 771, (2012): 381–95.
44. Huerta, A. E., et al. “Effects of α‐lipoic acid and eicosapentaenoic acid in overweight and obese women during weight loss.” Obesity 23, no. 2 (2015): 313–21.
45. Carbonelli, M. G., et al. “α-Lipoic acid supplementation: a tool for obesity therapy?” Current Pharmaceutical Design 16, no. 7 (2010): 840–46.
46. Koh, E. H., et al. “Effects of alpha-lipoic acid on body weight in obese subjects.” American Journal of Medicine 124, no. 1 (2011): 85-e1.
47. Natural Resources Defense Council, “What’s on Tap?” last revised 2/6/2012, www.nrdc.org/water/drinking/uscities/pdf/chap04.pdf, accessed February 15, 2016. See also www.nrdc.org/health/safe-drinking-water.asp. You can also look up your drinking water on the Environmental Working Group website, www.ewg.org/tap-water/whats-in-yourwater.php, accessed February 15, 2016.
Chapter 10: Soothe—Week 6
1. Epel, E. S., et al. “Accelerated telomere shortening in response to life stress.” Proceedings of the National Academy of Sciences of the United States of America 101, no. 49 (2004): 17312–15.
2. Bonini, L. “The Extended Mirror Neuron Network Anatomy, Origin, and Functions.” Neuroscientist (2016): 1073858415626400; Caramazza, A., et al. “Embodied cognition and mirror neurons: a critical assessment.” Annual Review of Neuroscience 37 (2014): 1–15; Cook, R., et al. “Mirror neurons: from origin to function.” Behavioral and Brain Sciences 37, no. 2 (2014): 177–92.
3. Acharya, S., et al. “Mirror neurons: Enigma of the metaphysical modular brain.” Journal of Natural Science, Biology and Medicine 3, no. 2 (2012): 118.
4. Yehuda, R., et al. “Holocaust exposure induced intergenerational effects on FKBP5 methylation.” Biological Psychiatry (2015).
5. Yehuda, R., et al. “Gene expression patterns associated with posttraumatic stress disorder following exposure to the World Trade Center attacks.” Biological Psychiatry 66, no. 7 (2009): 708–11.
6. Dias, B. G., et al. “Parental olfactory experience influences behavior and neural structure in subsequent generations.” Nature Neuroscience 17, no. 1 (2014): 89–96.
7. McEwen, B. S., et al. “Protective and damaging effects of stress mediators.” New England Journal of Medicine 338, no. 3 (1998): 171–79; Jiang, W., et al. “Mental stress–induced myocardial ischemia and cardiac events.” JAMA 275, no. 21 (1996): 1651–56; Deanfield, J. E., et al. “Silent myocardial ischaemia due to mental stress.” Lancet 324, no. 8410 (1984): 1001–5; Rozanski, A., et al. “Mental stress and the induction of silent myocardial ischemia in patients with coronary artery disease.” New England Journal of Medicine 318, no. 16 (1988): 1005–12; Nabi, H., et al. “Increased risk of coronary heart disease among individuals reporting adverse impact of stress on their health: the Whitehall II prospective cohort study.” European Heart Journal (2013): eht216; Arnold, S. V., et al. “Perceived stress in myocardial infarction: long-term mortality and health status outcomes.” Journal of the American College of Cardiology 60, no. 18 (2012): 1756–63; Richardson, S., et al. “Meta-analysis of perceived stress and its association with incident coronary heart disease.” American Journal of Cardiology 110, no. 12 (2012): 1711–16; Deedwania, P. C. “Editorial comment: Mental stress, pain perception and risk of silent ischemia.” Journal of the American College of Cardiology 25, no. 7 (1995): 1504–6; Steptoe, A., et al. “Stress and cardiovascular disease.” Nature Reviews Cardiology 9, no. 6 (2012): 360–70; Alevizos, M., et al. “Stress triggers coronary mast cells leading to cardiac events.” Annals of Allergy, Asthma and Immunology 112, no. 4 (2014): 309–16; Chida, Y., et al. “A bidirectional relationship between psychosocial factors and atopic disorders: a systematic review and meta-analysis.” Psychosomatic Medicine 70, no. 1 (2008): 102–16; Theoharides, T. C., et al. “Critical role of mast cells in inflammatory diseases and the effect of acute stress.” Journal of Neuroimmunology 146, no. 1 (2004): 1–12; Wright, R., et al. “The impact of stress on the development and expression of atopy.” Current Opinion in Allergy and Clinical Immunology 5, no. 1 (2005): 23–29; Slattery, M. J. “Psychiatric comorbidity associated with atopic disorders in children and adolescents.” Immunology and Allergy Clinics of North America 25, no. 2 (2005): 407–20; Seiffert, K., et al. “Psychophysiological reactivity under mental stress in atopic dermatitis.” Dermatology 210, no. 4 (2005): 286293; Chen, E., et al. “Stress and inflammation in exacerbations of asthma.” Brain, Behavior, and Immunity 21, no. 8 (2007): 993–99; Theoharides, T. C., et al. “Contribution of stress to asthma worsening through mast cell activation.” Annals of Allergy, Asthma and Immunology 109, no. 1 (2012): 14–19.
8. Judge, T. A., et al. “Genetic influences on core self-evaluations, job satisfaction, and work stress: A behavioral genetics mediated model.” Organizational Behavior and Human Decision Processes 117, no. 1 (2012): 208–20.
9. Ising, M., et al. “Genetics of stress response and stress-related disorders.” Dialogues in Clinical Neuroscience 8, no. 4 (2006): 433.
10. Slominski, A. T., et al. “Key role of CRF in the skin stress response system.” Endocrine Reviews 34, no. 6 (2013): 827–84.
11. Ising, M., et al. “Polymorphisms in the FKBP5 gene region modulate recovery from psychosocial stress in healthy controls.” European Journal of Neuroscience 28, no. 2 (2008): 389–98.
12. Zuhl, M., et al. “Exercise regulation of intestinal tight junction proteins.” British Journal of Sports Medicine (2012): bjsports-2012.
13. Piacentini, M. F., et al. “Stress related changes during a half marathon in master endurance athletes.” Journal of Sports Medicine and Physical Fitness 55, no. 4 (2015): 329–36; Brisswalter, J., et al. “Neuromuscular factors associated with decline in long-distance running performance in master athletes.” Sports Medicine 43, no. 1 (2013): 51–63; Lac, G., et al. “Changes in cortisol and testosterone levels and T/C ratio during an endurance competition and recovery.” Journal of Sports Medicine and Physical Fitness 40, no. 2 (2000): 139.
14. Lamprecht, M., et al. “Exercise, intestinal barrier dysfunction and probiotic supplementation.” Medicine and Sport Science (2012): 47–56; Lamprecht, M., et al. “Probiotic supplementation affects markers of intestinal barrier, oxidation, and inflammation in trained men; a randomized, double-blinded, placebo-controlled trial.” Journal of the International Society of Sports Nutrition 9, no. 1 (2012): 45.
15. Delarue, J., et al. “Fish oil prevents the adrenal activation elicited by mental stress in healthy men.” Diabetes and Metabolism 29, no. 3 (2003): 289–95.
16. Peters, E. M., et al. “Vitamin C supplementation attenuates the increases in circulating cortisol, adrenaline and anti-inflammatory polypeptides following ultramarathon running.” International Journal of Sports Medicine 22, no. 7 (2001): 537–43; Peters, E. M., et al. “Attenuation of increase in circulating cortisol and enhancement of the acute phase protein response in vitamin C-supplemented ultramarathoners.” International Journal of Sports Medicine 22, no. 2 (2001): 120–26.
17. Bryant, E. F. The Yoga Sutras of Patanjali: A New Edition, Translation, and Commentary (New York: North Point Press, 2009).
18. Luders, E., et al. “The underlying anatomical correlates of long-term meditation: larger hippocampal and frontal volumes of gray matter.” Neuroimage 45, no. 3 (2009): 672–78.
19. Slagter, H. A., et al. “Theta phase synchrony and conscious target perception: impact of intensive mental training.” Journal of Cognitive Neuroscience 21, no. 8 (2009): 1536–49.
20. Albert, E. After Birth (Boston: Houghton Mifflin Harcourt, 2015), 112.
21. Porges, S. W. “The polyvagal theory: new insights into adaptive reactions of the autonomic nervous system.” Cleveland Clinic Journal of Medicine 76, Suppl. 2 (2009): S86; Danner, D. D., et al. “Positive emotions in early life and longevity: findings from the nun study.” Journal of Personality and Social Psychology 80, no. 5 (2001): 804.
22. Mäkinen, T. M., et al. “Autonomic nervous function during whole-body cold exposure before and after cold acclimation.” Aviation, Space, and Environmental Medicine 79, no. 9 (2008): 875–82.
23. Lu, W. A., et al. “Foot reflexology can increase vagal modulation, decrease sympathetic modulation, and lower blood pressure in healthy subjects and patients with coronary artery disease.” Alternative Therapies in Health and Medicine 17, no. 4 (2011): 8–14.
24. Yang, J. L., et al. “Comparison of effect of 5 recumbent positions on autonomic nervous modulation in patients with coronary artery disease.” Circulation Journal 72, no. 6 (2008): 902–08.
25. Vickhoff, B., et al. “Music structure determines heart rate variability of singers.” Frontiers in Psychology 4 (2013).
26. Richards, D., et al. “Stimulation of auricular acupuncture points in weight loss.” Australian Family Physician 27 (1998): S73–7; Yang, S. B., et al. “Efficacy comparison of different points combination in the treatment of menopausal insomnia: a randomized controlled trial.” Zhongguo Zhen Jiu [Chinese Acupuncture and Moxibustion] 34, no. 1 (2014): 3–8; da Silva, M.A.H., et al. “Neuroanatomic and clinical correspondences: acupuncture and vagus nerve stimulation.” Journal of Alternative and Complementary Medicine 20, no. 4 (2014): 233–40; He, W., et al. “Auricular acupuncture and vagal regulation.” Evidence-Based Complementary and Alternative Medicine (2012).
27. Girsberger, W., et al. “Heart rate variability and the influence of craniosacral therapy on autonomous nervous system regulation in persons with subjective discomforts: a pilot study.” Journal of Integrative Medicine 12, no. 3 (2014): 156–61.
28. Kabat‐Zinn, J. “Mindfulness‐based interventions in context: past, present, and future.” Clinical Psychology: Science and Practice 10, no. 2 (2003): 144–56.
29. Davidson, R. J. “Affective style, psychopathology, and resilience: brain mechanisms and plasticity.” American Psychologist 55, no. 11 (2000): 1196; Davidson, R. J., et al. “Alterations in brain and immune function produced by mindfulness meditation.” Psychosomatic Medicine 65, no. 4 (2003): 564–70.
30. Fredrickson, B. L., et al. “Open hearts build lives: positive emotions, induced through loving-kindness meditation, build consequential personal resources.” Journal of Personality and Social Psychology 95, no. 5 (2008): 1045; Kalyani, B. G., et al. “Neurohemodynamic correlates of ‘OM’ chanting: a pilot functional magnetic resonance imaging study.” International Journal of Yoga 4, no. 1 (2011): 3; Mason, H., et al. “Cardiovascular and respiratory effect of yogic slow breathing in the yoga beginner: what is the best approach?” Evidence-Based Complementary and Alternative Medicine (2013); Khalsa, D. S., et al. “Cerebral blood flow changes during chanting meditation.” Nuclear Medicine Communications 30, no. 12 (2009): 956–61; Chang, R. Y., et al. “The effect of t’ai chi exercise on autonomic nervous function of patients with coronary artery disease.” Journal of Alternative and Complementary Medicine 14, no. 9 (2008): 1107–13.
31. Leung, M.K., et al. “Increased gray matter volume in the right angular and posterior parahippocampal gyri in loving-kindness meditators.” Social Cognitive and Affective Neuroscience (2012): nss076; Kang, D. H., et al. “The effect of meditation on brain structure: cortical thickness mapping and diffusion tensor imaging.” Social Cognitive and Affective Neuroscience 8, no. 1 (2013): 27–33; Lazar, S. W., et al. “Meditation experience is associated with increased cortical thickness.” Neuroreport 16, no. 17 (2005): 1893.
32. Macartney, M. J., et al. “Intrinsic heart rate recovery after dynamic exercise is improved with an increased omega-3 index in healthy males.” British Journal of Nutrition 112, no. 12 (2014): 1984–92; Ninio, D. M., et al. “Docosahexaenoic acid-rich fish oil improves heart rate variability and heart rate responses to exercise in overweight adults.” British Journal of Nutrition 100, no. 05 (2008): 1097–1103; Sjoberg, N. J., et al. “Dose-dependent increases in heart rate variability and arterial compliance in overweight and obese adults with DHA-rich fish oil supplementation.” British Journal of Nutrition 103, no. 2 (2010): 243–48; Xin, W., et al. “Short-term effects of fish-oil supplementation on heart rate variability in humans: a meta-analysis of randomized controlled trials.” American Journal of Clinical Nutrition 97, no. 5 (2013): 926–35; Noreen, E. E., et al. “Effects of supplemental fish oil on resting metabolic rate, body composition, and salivary cortisol in healthy adults.” Journal of the International Society of Sports Nutrition 7, no. 31 (2010); Delarue et al. “Fish oil prevents the adrenal activation.”
33. Keating, T. Intimacy with God: An Introduction to Centering Prayer (Spring Valley, NY: Crossroad, 2009); Bourgeault, C. Centering Prayer and Inner Awakening (Lanham, MD: Rowman and Littlefield, 2004).
34. Hölzel, B. K., et al. “Stress reduction correlates with structural changes in the amygdala.” Social Cognitive and Affective Neuroscience (2009): nsp034.
Chapter 11: Think—Week 7
1. Stone, A. A., et al. “A snapshot of the age distribution of psychological well-being in the United States.” Proceedings of the National Academy of Sciences 107, no. 22 (2010): 9985–90.
2. O’Donohue, J. Beauty: The Invisible Embrace (New York: HarperCollins, 2004).
3. Brach, T. “Working with Difficulties: The Blessings of RAIN,” TaraBrach.com, accessed March 24, 2016. www.tarabrach.com/articles-interviews/rain-workingwithdifficulties.
4. Tippett, K. “John O’Donohue—the inner landscape of beauty,” On Being with Krista Tippett, August 6, 2015, accessed February 10, 2016. www.onbeing.org/program/john-o-donohue-the-inner-landscape-beauty/203.
5. Dadhania, V. P., et al. “Nutraceuticals against neurodegeneration: A mechanistic insight.” Current Neuropharmacology (2016).
6. Amor, S., et al. “Inflammation in neurodegenerative diseases.” Immunology 129, no. 2 (2010): 154–69.
7. Fischer, R., et al. “Interrelation of oxidative stress and inflammation in neurodegenerative disease: role of TNF.” Oxidative Medicine and Cellular Longevity (2015); Hooshmand, B., et al. “Homocysteine and holotranscobalamin and the risk of Alzheimer disease a longitudinal study.” Neurology 75, no. 16 (2010): 1408–14; Laurin, D., et al. “Midlife C-reactive protein and risk of cognitive decline: a 31-year follow-up.” Neurobiology of Aging 30, no. 11 (2009): 1724–27; Komulainen, P., et al. “Serum high sensitivity C-reactive protein and cognitive function in elderly women.” Age and Ageing 36, no. 4 (2007): 443–48; Alcolea, D., et al. “Amyloid precursor protein metabolism and inflammation markers in preclinical Alzheimer disease.” Neurology 85, no. 7 (2015): 626–33.
8. Duchen, M. R. “Mitochondria and calcium: from cell signalling to cell death.” Journal of Physiology 529, no. 1 (2000): 57–68; Marambaud, P., et al “Calcium signaling in neurodegeneration.” Molecular Neurodegeneration 4, no. 20 (2009): 6–5; Bezprozvanny, I. B. “Calcium signaling and neurodegeneration.” Acta Naturae 2, no. 1 (2010): 72.
9. Toescu, E. C., et al. “The importance of being subtle: small changes in calcium homeostasis control cognitive decline in normal aging.” Aging Cell 6, no. 3 (2007): 267–73.
10. Sun, A. Y., et al. “Oxidative stress and neurodegenerative disorders.” Journal of Biomedical Science 5, no. 6 (1998): 401–14; López-Armada, M. J., et al. “Mitochondrial dysfunction and the inflammatory response.” Mitochondrion 13, no. 2 (2013): 106–18.
11. Lane, R. K., et al. “The role of mitochondrial dysfunction in age-related diseases.” Biochimica et Biophysica Acta (BBA)-Bioenergetics 1847, no. 11 (2015): 1387–1400.
12. Lin, M. T., et al. “Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases.” Nature 443, no. 7113 (2006): 787–95; Petrozzi, L., et al. “Mitochondria and neurodegeneration.” Bioscience Reports 27 (2007): 87–104; Kaminsky, Y. G., et al. “Critical analysis of Alzheimer’s amyloid-beta toxicity to mitochondria.” Frontiers in Bioscience 20 (2015): 173–97; Lionaki, E., et al. “Mitochondria, autophagy and age-associated neurodegenerative diseases: New insights into a complex interplay.”Biochimica et Biophysica Acta (BBA)-Bioenergetics (2015).
13. Alzheimer’s Association. “2015 Alzheimer’s disease facts and figures.” Alzheimer’s and Dementia: Journal of the Alzheimer’s Association 11, no. 3 (2015): 332.
14. “What we know today about Alzheimer’s Disease,” Alzheimer’s Association, accessed January 9, 2016, www.alz.org/research/science/alzheimers_disease_causes.asp.
15. Kuro-o, M. “Klotho as a regulator of oxidative stress and senescence.” Biological Chemistry 389, no. 3 (2008): 233–41; Mitobe, M., et al. “Oxidative stress decreases klotho expression in a mouse kidney cell line.” Nephron Experimental Nephrology 101, no. 2 (2005): e67–e74; Yamamoto, M., et al. “Regulation of oxidative stress by the anti-aging hormone klotho.” Journal of Biological Chemistry 280, no. 45 (2005): 38029–34; Troyano-Suárez, N., et al. “Glucose oxidase induces cellular senescence in immortal renal cells through ILK by downregulating klotho gene expression.” Oxidative Medicine and Cellular Longevity (2015); Kim, J. H., et al. “Biological role of anti-aging protein klotho.” Journal of Lifestyle Medicine 5, no. 1 (2015): 1.
16. Kuro-o, M., et al. “Mutation of the mouse klotho gene leads to a syndrome resembling ageing.” Nature 390, no. 6655 (1997): 45–51.
17. “2015 Alzheimer’s disease facts and figures,” Alzheimer’s Association.
18. Mayeux, R., et al. “Epidemiology of Alzheimer disease.” Cold Spring Harbor Perspectives in Medicine 2, no. 8 (2012): a006239.
19. Blennow K., et al. “Alzheimer’s disease.” Lancet, 368, no. 9533 (2006): 387–403.
20. Loy, C. T., et al. “Genetics of dementia.” Lancet 383, no. 9919 (2014): 828–40; Holtzman, D. M., et al. “Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in Alzheimer disease.” Cold Spring Harbor Perspectives in Medicine 2, no. 3 (2012): a006312.
21. Theendakara, V., et al. “Neuroprotective sirtuin ratio reversed by ApoE4.” Proceedings of the National Academy of Sciences 110, no. 45 (2013): 18303–8; Tramutola, A., et al. “Alteration of mTOR signaling occurs early in the progression of Alzheimer disease (AD): analysis of brain from subjects with pre‐clinical AD, amnestic mild cognitive impairment and late‐stage AD.” Journal of Neurochemistry 133, no. 5 (2015): 739–49.
22. Bredesen, D. E. “Reversal of cognitive decline: a novel therapeutic program.” Aging 6, no. 9 (2014): 707; Bredesen, D. E., et al. “Reversal of cognitive decline in Alzheimer’s disease.” Aging 8, no. 6 (2016): 1250.
23. Bredesen, D. E. “Metabolic profiling distinguishes three subtypes of Alzheimer’s disease.” Aging 7, no. 8 (2015): 595; Bredesen, D. E. “Inhalational Alzheimer’s disease: an unrecognized—and treatable—epidemic.” Aging 8, no. 2 (2016): 304.
24. Gatz, M., et al. “Role of genes and environments for explaining Alzheimer disease.” Archives of General Psychiatry 63, no. 2 (2006): 168–74; Kamboh, M. I., et al. “Genome-wide association study of Alzheimer’s disease.” Translational Psychiatry 2, no. 5 (2012): e117.
25. Sleegers, K., et al. “The pursuit of susceptibility genes for Alzheimer’s disease: progress and prospects.” Trends in Genetics 26, no. 2 (2010): 84–93.
26. Fraga, M. F., et al. “Epigenetic differences arise during the lifetime of monozygotic twins.” Proceedings of the National Academy of Sciences of the United States of America 102, no. 30 (2005): 10604–9; Heyn, H., et al. “Distinct DNA methylomes of newborns and centenarians.” Proceedings of the National Academy of Sciences 109, no. 26 (2012): 10522–27.
27. Bredesen, D. E., et al. “Next generation therapeutics for Alzheimer’s disease.” EMBO Molecular Medicine 5, no. 6 (2013): 795–98; Yaffe, K., et al. “Estrogen use, APOE, and cognitive decline evidence of gene–environment interaction.” Neurology 54, no. 10 (2000): 1949–54.
28. “2005 adult sleep habits and styles,” National Sleep Foundation, accessed February 1, 2016, https://sleepfoundation.org/sleep-polls-data/sleep-in-america-poll/2005-adult-sleep-habits-and-styles.
29. Smith, G. E., et al. “A cognitive training program based on principles of brain plasticity: Results from the Improvement in Memory with Plasticity‐Based Adaptive Cognitive Training (IMPACT) Study.” Journal of the American Geriatrics Society 57, no. 4 (2009): 594–603.
30. Hatch, S. L., et al. “The continuing benefits of education: adult education and midlife cognitive ability in the British 1946 birth cohort.” Journals of Gerontology Series B: Psychological Sciences and Social Sciences 62, no. 6 (2007): S404–14.
31. Woods, B., et al. “Cognitive stimulation to improve cognitive functioning in people with dementia.” Cochrane Database of Systematic Reviews 2 (2012).
32. Schmidt, S. R. “Effects of humor on sentence memory.” Journal of Experimental Psychology: Learning, Memory, and Cognition 20, no. 4 (1994): 953.
33. Ybarra, O., et al. “Mental exercising through simple socializing: Social interaction promotes general cognitive functioning.” Personality and Social Psychology Bulletin 34, no. 2 (2008): 248–59.
34. Bassuk, S. S., et al. “Social disengagement and incident cognitive decline in community-dwelling elderly persons.” Annals of Internal Medicine 131, no. 3 (1999): 165–73.
35. Savignac, H. M., et al. “Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-D-aspartate receptor subunits and D-serine.” Neurochemistry International 63, no. 8 (2013): 756–64.
36. Beck, J. “Your gut bacteria want you to eat a cupcake,” Atlantic August 19, 2014, accessed February 1, 2016, www.theatlantic.com/health/archive/2014/08/your-gut-bacteriawant-you-to-eat-a-cupcake/378702.
37. Kivipelto, M., et al. “Alzheimer disease: To what extent can Alzheimer disease be prevented?” Nature Reviews Neurology 10, no. 10 (2014): 552–53.
38. Ibid.; Kawas, C., et al. “Age-specific incidence rates of Alzheimer’s disease: The Baltimore Longitudinal Study of Aging.” Neurology 54, no. 11 (2000): 2072–77.
39. Smith, J., et al. “Physical activity reduces hippocampal atrophy in elders at genetic risk for Alzheimer’s disease.” Frontiers in Aging Neuroscience 6 (2014).
40. Sui, X., et al. “A prospective study of cardiorespiratory fitness and risk of type 2 diabetes in women.” Diabetes Care 31, no. 3 (2008): 550–55; Blair, S. N., et al. “Physical fitness and all-cause mortality: a prospective study of healthy men and women.” JAMA 262, no. 17 (1989): 2395–2401; Hooker, S. P., et al. “Cardiorespiratory fitness as a predictor of fatal and nonfatal stroke in asymptomatic women and men.” Stroke 39, no. 11 (2008): 2950–57; Wei, M., et al. “The association between cardiorespiratory fitness and impaired fasting glucose and type 2 diabetes mellitus in men.” Annals of Internal Medicine 130, no. 2 (1999): 89–96.
41. DeFina, L. F., et al. “The association between midlife cardiorespiratory fitness levels and later-life dementia: a cohort study.” Annals of Internal Medicine 158, no. 3 (2013): 162–68.
42. Smith, G. E., et al. “A cognitive training program based on principles of brain plasticity: Results from the Improvement in Memory with Plasticity‐based Adaptive Cognitive Training (IMPACT) Study.” Journal of the American Geriatrics Society 57, no. 4 (2009): 594–603.
43. Cotman, C. W., et al. “Exercise builds brain health: key roles of growth factor cascades and inflammation.” Trends in Neurosciences 30, no. 9 (2007): 464–72; Aguiar, P., et al. “Rivastigmine transdermal patch and physical exercises for Alzheimer’s disease: a randomized clinical trial.” Current Alzheimer Research 11, no. 6 (2014): 532–37.
44. Etnier, J. L., et al. “A meta-regression to examine the relationship between aerobic fitness and cognitive performance.” Brain Research Reviews 52, no. 1 (2006): 119–30; Angevaren, M., et al. “Physical activity and enhanced fitness to improve cognitive function in older people without known cognitive impairment.” Cochrane Database System Review 3, no. 3 (2008); Erickson, K. I., et al. “Exercise training increases size of hippocampus and improves memory.” Proceedings of the National Academy of Sciences 108, no. 7 (2011): 3017–22; Woodard, J. L., et al. “Lifestyle and genetic contributions to cognitive decline and hippocampal integrity in healthy aging.” Current Alzheimer Research 9, no. 4 (2012): 436.
45. Eny, K. M., et al. “Genetic variant in the glucose transporter type 2 is associated with higher intakes of sugars in two distinct populations.” Physiological Genomics 33, no. 3 (2008): 355–60.
46. Matthews, G., et al. Personality Traits (Cambridge: Cambridge University Press, 2003).
47. Harvey, C. J., et al. “Who is predisposed to insomnia: a review of familial aggregation, stress-reactivity, personality and coping style.” Sleep Medicine Reviews 18, no. 3 (2014): 237–47; Smith, T. W., et al. “Hostility, anger, aggressiveness, and coronary heart disease: An interpersonal perspective on personality, emotion, and health.” Journal of Personality 72, no. 6 (2004): 1217–70; Suarez, E. C., et al. “The relation of aggression, hostility, and anger to lipopolysaccharide-stimulated tumor necrosis factor (TNF)-α by blood monocytes from normal men.” Brain, Behavior, and Immunity 16, no. 6 (2002): 675–84; Jylhä, P., et al. “The relationship of neuroticism and extraversion to symptoms of anxiety and depression in the general population.” Depression and Anxiety 23, no. 5 (2006): 281–89.
48. Akram, U., et al. “Anxiety mediates the relationship between perfectionism and insomnia symptoms: A longitudinal study.” PloS One 10, no. 10 (2015): e0138865; Vincent, N. K., et al. “Perfectionism and chronic insomnia.” Journal of Psychosomatic Research 49, no. 5 (2000): 349–54; de Azevedo, M. H. P., et al. “Perfectionism and sleep disturbance.” World Journal of Biological Psychiatry 10, no. 3 (2009): 225–33; Azevedo, M. H., et al. “Longitudinal study on perfectionism and sleep disturbance.” World Journal of Biological Psychiatry 11, no. 2 (2010): 476–85; van de Laar, M., et al. “The role of personality traits in insomnia.” Sleep Medicine Reviews 14, no. 1 (2010): 61–68; Schramm, E., et al. “Mental comorbidity of chronic insomnia in general practice attenders using DSM‐III‐R.” Acta Psychiatrica Scandinavica 91, no. 1 (1995): 10–17.
49. www.ncbi.nlm.nih.gov/pubmed/8893314; www.researchgate.net/profile/Laura_Richman/publication/7701075_Positive_emotion_and_health_Going_beyond_the_negative/links/0046351a8c8c404c29000000.pdf.
50. Gold, S. M., et al. “Basal serum levels and reactivity of nerve growth factor and brain-derived neurotrophic factor to standardized acute exercise in multiple sclerosis and controls.” Journal of Neuroimmunology 138, no. 1 (2003): 99–105; Rojas Vega, S., et al. “Acute BDNF and cortisol response to low intensity exercise and following ramp incremental exercise to exhaustion in humans.” Brain Research 1121, no. 1 (2006): 59–65; Ferris, L. T., et al. “The effect of acute exercise on serum brain-derived neurotrophic factor levels and cognitive function.” Medicine and Science in Sports and Exercise 39, no. 4 (2007): 728–34; Tang, S. W., et al. “Influence of exercise on serum brain-derived neurotrophic factor concentrations in healthy human subjects.” Neuroscience Letters 431, no. 1 (2008): 62–65; Gustafsson, G., et al. “The acute response of plasma brain-derived neurotrophic factor as a result of exercise in major depressive disorder.” Psychiatry Research 169, no. 3 (2009): 244–48; Schmolesky, M. T., et al. “The effects of aerobic exercise intensity and duration on levels of brain-derived neurotrophic factor in healthy men.” Journal of Sports Science and Medicine 12, no. 3 (2013): 502; de Melo Coelho, F. G., et al. “Acute aerobic exercise increases brain-derived neurotrophic factor levels in elderly with Alzheimer’s disease.” Journal of Alzheimer’s Disease 39, no. 2 (2014): 401; Saucedo-Marquez, C. M., et al. “High intensity interval training evokes larger serum BDNF levels compared to intense continuous exercise.” Journal of Applied Physiology (2015): jap-00126.
51. Fernando, W., et al. “The role of dietary coconut for the prevention and treatment of Alzheimer’s disease: potential mechanisms of action.” British Journal of Nutrition (2015): 1–14; Rebello, C. J., et al. “Pilot feasibility and safety study examining the effect of medium chain triglyceride supplementation in subjects with mild cognitive impairment: A randomized controlled trial.” BBA Clinical 3 (2015): 123–25.
52. Reger, M. A., et al. “Effects of beta-hydroxybutyrate on cognition in memory-impaired adults.” Neurobiology of Aging 25, no. 3 (2004): 311–14.
53. Zong, G., et al. “Frequent consumption of meals prepared at home and risk of Type 2 diabetes among American men and women.” Circulation 132, Suppl 3 (2015): A17285.
54. Haze, S., et al. “Effects of fragrance inhalation on sympathetic activity in normal adults.” Japanese Journal of Pharmacology 90, no. 3 (2002): 247253.
55. Lee, I. S., et al. “Effects of lavender aromatherapy on insomnia and depression in women college students.” Taehan Kanho Hakhoe Chi 36, no. 1 (2006): 136–43; Shiina, Y., et al. “Relaxation effects of lavender aromatherapy improve coronary flow velocity reserve in healthy men evaluated by transthoracic Doppler echocardiography.” International Journal of Cardiology 129, no. 2 (2008): 193–97; Lytle, J., et al. “Effect of lavender aromatherapy on vital signs and perceived quality of sleep in the intermediate care unit: a pilot study.” American Journal of Critical Care 23, no. 1 (2014): 24–29; Lillehei, A. S., et al. “A systematic review of the effect of inhaled essential oils on sleep.” Journal of Alternative and Complementary Medicine 20, no. 6 (2014): 441–51.
56. Igarashi, M., et al. “Effect of olfactory stimulation by fresh rose flowers on autonomic nervous activity.” Journal of Alternative and Complementary Medicine20, no. 9 (2014): 727–31.
57. World Health Organization, Nutritional Anemias: Report of a Scientific Group, World Health Organization Technical Report Series 405 (1968).
58. Tucker, K. L., et al. “Plasma vitamin B-12 concentrations relate to intake source in the Framingham Offspring Study,” American Journal of Clinical Nutrition 71 (2000): 514–22.
59. Pawlak, R., et al. “How prevalent is vitamin B(12) deficiency among vegetarians?” Nutritional Review 71, no. 2 (February 2013): 110–17.
60. Tucker, “Plasma vitamin B-12 concentrations,” and Tucker, K. L., et al. “Low plasma vitamin B12 is associated with lower BMD: The Framingham Osteoporosis Study,” Journal of Bone and Mineral Research 20, no. 1 (January 2005): 152–58.
61. Knott, V., et al. “Neurocognitive effects of acute choline supplementation in low, medium and high performer healthy volunteers.” Pharmacology Biochemistry and Behavior 131 (2015): 119–29.
62. Overgaard, K. “The effects of citicoline on acute ischemic stroke: A review.” Journal of Stroke and Cerebrovascular Diseases 23, no. 7 (2014): 1764–69.
63. “Horizon: How video games can change your brain,” BBC September 16, 2015, accessed January 21, 2016, www.bbc.com/news/technology-34255492; Churchland, P. “Videogames for seniors boost brainpower,” Wall Street Journal, September 20, 2015, accessed January 21, 2016, www.wsj.com/articles/videogames-for-seniors-boost-brainpower-1443623158.
Chapter 12: Integrate
1. Painter, R. C., et al. “Transgenerational effects of prenatal exposure to the Dutch famine on neonatal adiposity and health in later life.” BJOG: An International Journal of Obstetrics and Gynaecology 115, no. 10 (2008): 1243–49.
2. de Rooij, S. R., et al. “Prenatal undernutrition and cognitive function in late adulthood.” Proceedings of the National Academy of Sciences 107, no. 39 (2010): 16881–86.
3. www.ncbi.nlm.nih.gov/pubmed/27146370 Bleker 2016
4. Tobi, E. W., et al. “Early gestation as the critical time-window for changes in the prenatal environment to affect the adult human blood methylome.” International Journal of Epidemiology (2015): dyv043.
5. Veenendaal, M.V.E., et al. “Transgenerational effects of prenatal exposure to the 1944–45 Dutch famine.” BJOG: An International Journal of Obstetrics and Gynaecology 120, no. 5 (2013): 548–54.
6. Ekamper, P., et al. “Prenatal famine exposure and adult mortality from cancer, cardiovascular disease, and other causes through age 63 years.” American Journal of Epidemiology 181, no. 4 (2015): 271–79.
7. de Rooij, S. R., et al. “Prenatal undernutrition and leukocyte telomere length in late adulthood: the Dutch famine birth cohort study.” American Journal of Clinical Nutrition 102, no. 3 (2015): 655–60.
8. Halstead, R. “Study: Marin County men have the highest life expectancy in the nation, women rank no. 2,” Marin Independent Journal, April 19, 2012, accessed January 31, 2016, www.marinij.com/article/ZZ/20120419/NEWS/120418529.
9. Levy, B. R., et al. “Association between positive age stereotypes and recovery from disability in older persons.” JAMA 308, no. 19 (2012): 1972–73; Levy, B. R., et al. “Longitudinal benefit of positive self-perceptions of aging on functional health.” Journals of Gerontology Series B: Psychological Sciences and Social Sciences 57, no. 5 (2002): P409–17; Sargent-Cox, K. A., et al. “The relationship between change in self-perceptions of aging and physical functioning in older adults.” Psychology and Aging 27, no. 3 (2012): 750.
10. Levy, B.R., et al. “Subliminal strengthening improving older individuals’ physical function over time with an implicit-age-stereotype intervention.” Psychological Science 25, no. 12 (2014): 2127–35.
11. Levy, B. R., et al. “The stereotype-matching effect: greater influence on functioning when age stereotypes correspond to outcomes.” Psychology and Aging 24, no. 1 (2009): 230.
12. Murphy, S. T., et al. “Additivity of nonconscious affect: combined effects of priming and exposure.” Journal of Personality and Social Psychology 69, no. 4 (1995): 589.
13. Schneier, M. “Fashion’s gaze turned to Joan Didion in 2015,” New York Times, December 18, 2015, accessed December 26, 2015, www.nytimes.com/2015/12/20/fashion/joan-didionceline-fashion-gaze.html.
14. Cooke, R. “Joan Didion as the new face of Celine? That’s so smart,” Guardian January 11, 2015, accessed December 26, 2015, www.theguardian.com/commentisfree/2015/jan/11/ joan-didion-new-face-celine-smart.
15. Levy, B. R., et al. “Longevity increased by positive self-perceptions of aging.” Journal of Personality and Social Psychology 83, no. 2 (2002): 261.
Top Seven Genes: What to Do
1. Read more at www.snpedia.com/index.php/Orientation.