15 May 2020

Reversal of CVD with diet and lifestyle

Among the ocean of research, I’m very interested in natural interventions which have reversed common diseases, since they help reveal basic ecology. Here’s a mini-review based around my initial reading of such CVD trials.

Cardiovascular disease (CVD) is the leading cause of mortality worldwide, accounting for 31% of all deaths (WHO). The most common forms are coronary artery and cerebrovascular diseases (affecting heart and brain, respectively) driven by atherosclerosis (i.e. plaque build-up) 1,2. Further, CVD and intracranial atherosclerosis are also linked to dementia 3,4, another leading cause of death (e.g. Dementia Hub).

Atherosclerosis is a highly prevalent and systemic disease, affecting both ancient 5 and modern humans 6–8. For instance, a recent Scottish study using whole-body MR angiography on low-med risk adults (n=1528, >40yrs old) found half had some artery narrowing (stenosis), a quarter in multiple vessels, with specific sites distributed throughout the body 7,9. The early stages (e.g. fatty streaks) can start in childhood, as shown in US autopsy studies 10. However, prevalence is varied and notably low in some populations 11–13. For instance, the lowest prevalence of coronary artery disease (CAD, based on CAC) was recently reported in the Tsimane 13 (i.e. forager-horticulturists of the Bolivian Amazon 14), despite their high inflammatory burden.

CVD is associated with many risk factors, which particularly implicate lifestyle 1,2,15,16, while many related trials have improved and even reversed CVD (for reviews see 17,18). For instance, a 2015 meta-analysis of 14 RCTs found that intensive lifestyle changes decreased coronary and carotid atherosclerotic burden 17. Indeed, since 1990 19, several trials of diet/lifestyle changes have induced regression of CAD, as measured via coronary angiography 19–26 (Table 1), while lowering symptoms and further cardiac events 19,21–25 (except some cases of over-exercise 19,20). In addition, many other trials on various populations (at risk of CVD) have induced regression of carotid atherosclerosis, as measured non-invasively via carotid artery thickening (which can correlate coronary disease 27), with diet/lifestyle changes 28–33 (Table 2) and even single extracts/nutrients 34–41, providing further insight into mechanisms.


Reversal—how?

CAD regression trials generally used lipid-lowering diets (i.e. plant-based 19,21,22,24–26 or others 20,23), either alone 23,26, or with exercise 20 and psychotherapies (i.e. stress reduction 19,22 or yoga 21,24,25). They lowered various other biomarkers, including weight/BMI 19,21,22,24–26, insulin 26, blood pressure 21,24 and cholesterol 19–26; some of which correlated regression (e.g. weight/TGs 21, blood pressure 23 and LDL/HDL 22,23). Similarly, carotid regression in various populations was achieved with various diets (i.e. Mediterranean 30–33 (w/nuts 30,31) and low calorie/carb/fat 28,29,32,33), either alone 30–33, or with exercise 28 and psychotherapy 29; as well as in blinded/placebo-controlled trials with specific extracts/nutrients (e.g. pomegranate juice 34, plant oils 35, omega-3 36, magnesium 37, folate 38, vit-B12 39, carnitine 40 and artichoke extract 41). These trials altered many related biomarkers, including weight 28,29,32,33,40,41, blood pressure 32–34, lipids 28,29,32,33,35–38,41, immune 28,35,37,38, glucose 28,29,32,37,38,41, FMD/NO 38–41, redox 34,37 and homocysteine 33,39; some of which correlated regression (e.g. blood pressure 33, adiponectin/leptin 29, TMAO 28 and EPCs 32). The following discussion will consider some underlying mechanisms in relation to diet, although they may be modulated by other aspects of lifestyle.

Interestingly, some CAD reports note rapid improvement in symptoms 19,21,42 and heart perfusion 43, suggesting improved vascular function and blood flow 11. Of relevance, foods can acutely alter cardio-autonomic 44 and vascular function 45–47. For instance, in people with CAD/CHF, high fat meals can acutely suppress vasodilation (i.e. brachial artery at 4hrs 45), while polyphenol-rich plant foods can boost vasodilation (e.g. at 1 46 and 2hrs 47). These and other effects may involve redox modulation of vascular (e.g. endothelium, nitric oxide 11), neuroendocrine (e.g. hypothalamus, sympathetic activity 48,49) and heart tissues (e.g. cardiomyocytes 15, edema 42). Another target is bone marrow-derived stem cells (e.g. Med diet 32 and exercise 50), which can increase within 30 days (e.g. cocoa flavanols in CAD 51), and may support repair and function of the cardiovascular system.

Regarding atherosclerosis, regression characteristics will depend on population and intervention. The shortest trials tend to be those with extracts/nutrients, which have induced significant carotid regression at 3 months 34,35,38–40 (e.g. max: –19% with flax oil in obese elderly 35), 6 months 34,36,37,41 (e.g. max: –35% with artichoke extract in MetS 41) and 1 year 34,39 (e.g. max: –35% with pomegranate juice in severe CAS 34). Diet/lifestyle trials have induced carotid regression at 9 months to >1 year (e.g. max: –10% with Med diet + nuts in high CVD risk 30), which has depended on baseline severity 30 (e.g. cIMT ≥0.9mm 31) and dietary change (i.e. –12% with mod/high sat-fat reduction in obese adolescents 29). Similarly, in CAD trials, coronary regression was most marked in the most severe lesions 19,23,24, related to programme adherence 19,21,22 (e.g. max: –18% with most adherence at 2yrs 21) and accompanied by improved stenosis geometry 52. Of further note, some additional case/clinical reports, using diet/lifestyle and drugs, even report reversal of extreme coronary stenosis (e.g. 90–95% obstruction) and heart failure (e.g. ejection fraction 25%) 15,42,53,54, and extremely low recurrence of events (0.6%) 43.

Regarding biological pathways, cholesterol is commonly implicated. In general populations (>40yrs old, no known CVD), LDL-c and non-HDL-c correlate the prevalence of subclinical atherosclerosis (e.g. peripheral 6,7 and intracranial 8; LDL-c >60 mg/dl 6); while in CVD trials, intensive lipid-lowering with drugs (e.g. LDL-c <70/80 mg/dl) can induce regression of coronary, carotid and intracranial atherosclerosis 55–58 (in relation to achieved LDL-c 55 and HDL-c 59,60). Similarly, diet/lifestyle trials reversing CAD also lowered LDL-c 19–26, but varied HDL-c (e.g. –9.6% 22 to +5.6% 21) and TGs, with some reporting associations 21–23 (and diet 24). And of those inducing carotid regression, some altered cholesterol 29,36–38,41, while some implicate other factors 28,33,34,39 (see above). For instance, TMAO correlated carotid regression independent of traditional markers (e.g. cholesterol, insulin resistance and obesity) 28. Drugs to control postprandial glucose have also induced some regression in diabetics (n=175 T2D, 1yr) 61. However, another controlled trial (n=349 T2D, 50% statins, 1yr) with nutritional ketosis did not induce carotid regression, despite favourably modulating many biomarkers (e.g. weight, BP, CRP, HbA1c, TGs, HDL-c, etc.), but also increasing LDL-c (i.e. 100 to 111 mg/dl) 62.

Plaque composition and development have been extensively studied (e.g. review). Briefly, progression involves ApoB lipoproteins (e.g. native or modified LDL 63) and inflammation (e.g. M1 activity and foam cell expansion) 63–67; while regression may involve essentially opposite pathways (e.g. HDL and M2 cells), allowing removal of damage/lipids and lesion/plaque shrinking 68,69. These processes are affected by many factors. For instance, dietary sat-fat can increase LDL/HDL-c levels and oxidative/inflammatory modifications (e.g. vs low 70 or unsat-fat 71–74); whereas low fat, plant-based diets can decrease LDL-c and less so HDL-c, but may increase HDL anti-inflammatory activity (e.g. diet + exercise 75). While HDL-c is inversely related to CVD, HDL function seems more important 68,76 and related to other qualities (e.g. fluidity and redox) 77–79. In particular, HDL plays a central role in reverse cholesterol transport (RCT; macrophage–liver), although may not always reflect the complete pathway (incl. liver–faeces) 80. In general, HDL/RCT may be modulated by many factors (e.g. ↑: Med diet 78, orange juice 81, flavonoids 82, olive oil 77, omega-3s 83,84, GSH 85, fibre/SCFAs 86,87; ↓: sat-fat 74, immune/LPS 83,88, oxidative stress 79, FMO3/TMAO 89). Dysregulation of lipid and lipoprotein metabolism is also related to hepatic insulin resistance 74,90 and homocysteine 91–95.

Plant-based?

The fact that CVD can be reversed with lifestyle, suggests it may not be an inevitable consequence of our genes or ageing, but environment. Further, while CVD may be accelerated by all kinds of contemporary risk factors (e.g. processed foods, inactivity, pollution, etc.), the fact that atherosclerosis was also prevalent in ancient populations (incl. hunter-gatherers) suggests an even more basic origin 5. Interestingly, compared to other great apes, we get more atherosclerosis and Alzheimer’s dementia 96. So perhaps something lies within our divergence from great ape to ancient human?

The other great apes are largely plant-based, while our diet evolved to include more (vertebrate/meat-based) animal foods, with some putative benefits 97. However, since plant-based diets appear to support reversal of CVD (see Tables 1/2 and cases 15,42,43,53,54), perhaps there was also (delayed) compromise? Accordingly, while animal foods can support environmental adaptability and essential nutrition in the short-term, many have also been linked to CVD in the long-term, by various mechanisms (Table 3). For instance, we may have relatively high cholesterol levels compared to other mammals 12,98, which correlate atherosclerosis (as above; and brain amyloid burden 99,100), while plant-based diets typically lower our cholesterol. Further, the human species-specific loss of CMAH gene function (~2–3mya) may make us uniquely susceptible to Neu5Gc in red meat, which is present in human vascular tissues and plaques 101, and can promote inflammation and atherosclerosis 102. We also uniquely cook foods, where high/dry heat may especially increase the formation of cardiometabolic toxins in animal foods (e.g. AGEs) 103.

On the other hand, some animal foods have more neutral or inverse associations; especially fish, in relation to long-chain omega-3s (i.e. EPA/DHA) 36,104–107. However, some fish–health associations are U-curved (e.g. mortality in Western studies 104) or offset by toxins (e.g. LDL 108, T2D 109 and CVD 107). Fish oils are also prone to oxidation (e.g. supplements 110, gastric models 111,112 and in vivo 71,113,114), which may affect bioactivity 115,116; although oxidation and Hg might be inhibited by polyphenol-rich plant foods 111,112,117. Moreover, do they have benefits beyond ALA, the plant-based precursor? In trials, ALA (18:3) supplementation can increase blood EPA (20:5) and DPA (22:5), but not DHA (22:6) 118; although still might meet brain requirements 119. A 2014 review comparing ALA to EPA/DHA suggests they may have similar CVD benefits 120, although optimal doses and contexts seem unclear. Also, a recent 2020 systematic review of 86 RCTs for CVD prevention found only slight benefits for both, partly offset by slightly increased prostate cancer 121; although confounding may include supplement quality and background diet 105,110,122 (e.g. 2019 VITAL trial supp. data 123).

Animal foods also supply vit-B12, which is obviously essential 124,125, and deficiency could undermine cardiovascular benefits of plant-based diets 126. Note, incorporation of omega-3s into phospholipids is also related to (B12-dependent) methylation, as discussed in a previous post. Nowadays B12 is acquired mainly via meat and dairy (eggs = v low bioavailability) 125; although B12 is ultimately produced by microbes and various sources exist lower down the food chain (e.g. some plants/fungi 124,125, insects 127, molluscs and small fish). In studies with strict plant-based diets, B12 is often supplemented 15,19,43,54.

Most recently, our diet has evolved to include ultra-processed foods. Consequently, increased consumption of both animal and processed/refined plant foods are associated with risk of common diseases, including T2D and CVD 128–132, while whole plant foods appear protective (paralleled by carb quality: GI/sugar vs fibre). Accordingly, several lifestyle interventions emphasised a whole food, plant-based approach 21,22,24,25 (strictest: 15,42,43). Note however, CAD regression was achieved despite an increase in TGs 19 related to simple carbs 22, while in another trial correlated lower TGs 21. Another aspect worth considering is fat quantity vs quality. Some CAD trials employed a very low 10% fat diet 22,24,26,43, which may be difficult to follow 20,21,23 (e.g. UK avg. ~35%). Is this necessary 133? Greater CAD regression occurred with adherence to a lifestyle with 15% fat (no statins) 21, while recent case reports of CAD/CHF reversal had higher levels (e.g. ~19 42 and 38% 133), as did trials with carotid regression (e.g. 24% 29) which even added fats (e.g. ~38 35 and 41% 30). So perhaps fat quality is most important (e.g. low sat-fat 29,133, high unsat-fat 35,36 and whole food 30,42,43).

Resources

References

1.           Mendis, S., Puska, P. & Norrving, B. Global Atlas on Cardiovascular Disease Prevention and Control. WHO (2011).

2.           Thomas, H. et al. Global Atlas of Cardiovascular Disease 2000-2016: The Path to Prevention and Control. Glob. Heart 13, 143–163 (2018).

3.           Lathe, R., Sapronova, A. & Kotelevtsev, Y. Atherosclerosis and Alzheimer--diseases with a common cause? Inflammation, oxysterols, vasculature. BMC Geriatr. 14, 36 (2014).

4.           Shabir, O., Berwick, J. & Francis, S. E. Neurovascular dysfunction in vascular dementia, Alzheimer’s and atherosclerosis. BMC Neurosci. 19, 1–16 (2018).

5.           Thompson, R. C. et al. Atherosclerosis across 4000 years of human history: the Horus study of four ancient populations. Lancet 381, 1211–22 (2013).

6.           Fernández-Friera, L. et al. Normal LDL-Cholesterol Levels Are Associated With Subclinical Atherosclerosis in the Absence of Risk Factors. J. Am. Coll. Cardiol. 70, 2979–2991 (2017).

7.           Lambert, M. A. et al. Prevalence and Distribution of Atherosclerosis in a Low- to Intermediate-Risk Population: Assessment with Whole-Body MR Angiography. Radiology 287, 795–804 (2018).

8.           Wu, J., Wang, A., Li, X., Wu, S. & Zhao, X. Non-High-Density Lipoprotein Cholesterol Levels on the Risk of Asymptomatic Intracranial Arterial Stenosis: A Result from the APAC Study. Sci. Rep. 6, 37410 (2016).

9.           Leiner, T. Whole-Body MR Angiography: Assessing the Global Burden of Cardiovascular Disease. Radiology 287, 805–807 (2018).

10.        Gooding, H. & Johnson, H. M. The Unchartered Frontier: Preventive Cardiology Between the Ages of 15 and 35 Years. Curr. Cardiovasc. Risk Rep. 10, 29 (2016).

11.        Esselstyn, C. B. A plant-based diet and coronary artery disease: a mandate for effective therapy. J. Geriatr. Cardiol. 14, 317–320 (2017).

12.        O’Keefe, J. H., Cordain, L., Harris, W. H., Moe, R. M. & Vogel, R. Optimal low-density lipoprotein is 50 to 70 mg/dl: lower is better and physiologically normal. J. Am. Coll. Cardiol. 43, 2142–6 (2004).

13.        Kaplan, H. et al. Coronary atherosclerosis in indigenous South American Tsimane: a cross-sectional cohort study. Lancet (London, England) 389, 1730–1739 (2017).

14.        Kraft, T. S. et al. Nutrition transition in 2 lowland Bolivian subsistence populations. Am. J. Clin. Nutr. 108, 1183–1195 (2018).

15.        Allen, K. E., Gumber, D. & Ostfeld, R. J. Heart Failure and a Plant-Based Diet. A Case-Report and Literature Review. Front. Nutr. 6, 82 (2019).

16.        Tuso, P., Stoll, S. R. & Li, W. W. A plant-based diet, atherogenesis, and coronary artery disease prevention. Perm. J. 19, 62–7 (2015).

17.        Jhamnani, S. et al. Meta-analysis of the effects of lifestyle modifications on coronary and carotid atherosclerotic burden. Am. J. Cardiol. 115, 268–75 (2015).

18.        Parsons, C., Agasthi, P., Mookadam, F. & Arsanjani, R. Reversal of coronary atherosclerosis: Role of life style and medical management. Trends Cardiovasc. Med. 28, 524–531 (2018).

19.        Ornish, D. et al. Can lifestyle changes reverse coronary heart disease? The Lifestyle Heart Trial. Lancet (London, England) 336, 129–33 (1990).

20.        Schuler, G. et al. Regular physical exercise and low-fat diet. Effects on progression of coronary artery disease. Circulation 86, 1–11 (1992).

21.        Gupta, S. K. et al. Regression of coronary atherosclerosis through healthy lifestyle in coronary artery disease patients--Mount Abu Open Heart Trial. Indian Heart J. 63, 461–9 (2011).

22.        Ornish, D. et al. Intensive lifestyle changes for reversal of coronary heart disease. JAMA 280, 2001–7 (1998).

23.        Watts, G. F. et al. Effects on coronary artery disease of lipid-lowering diet, or diet plus cholestyramine, in the St Thomas’ Atherosclerosis Regression Study (STARS). Lancet (London, England) 339, 563–9 (1992).

24.        Yogendra, J. et al. Beneficial effects of yoga lifestyle on reversibility of ischaemic heart disease: caring heart project of International Board of Yoga. J. Assoc. Physicians India 52, 283–9 (2004).

25.        Manchanda, S. C. et al. Retardation of coronary atherosclerosis with yoga lifestyle intervention. J. Assoc. Physicians India 48, 687–94 (2000).

26.        Huh, K. B., Lee, H. C., Cho, S. Y., Lee, J. H. & Song, Y. D. The role of insulin resistance in Korean patients with coronary atherosclerosis. Diabetes 45 Suppl 3, S59-61 (1996).

27.        Coskun, U. et al. Relationship between carotid intima-media thickness and coronary angiographic findings: a prospective study. Cardiovasc. Ultrasound 7, 59 (2009).

28.        Randrianarisoa, E. et al. Relationship of Serum Trimethylamine N-Oxide (TMAO) Levels with early Atherosclerosis in Humans. Sci. Rep. 6, 26745 (2016).

29.        Masquio, D. C. L. et al. Reduction in saturated fat intake improves cardiovascular risks in obese adolescents during interdisciplinary therapy. Int. J. Clin. Pract. 69, 560–70 (2015).

30.        Sala-Vila, A. et al. Changes in ultrasound-assessed carotid intima-media thickness and plaque with a Mediterranean diet: a substudy of the PREDIMED trial. Arterioscler. Thromb. Vasc. Biol. 34, 439–45 (2014).

31.        Murie-Fernandez, M. et al. Carotid intima-media thickness changes with Mediterranean diet: a randomized trial (PREDIMED-Navarra). Atherosclerosis 219, 158–62 (2011).

32.        Maiorino, M. I. et al. Effect of a Mediterranean diet on endothelial progenitor cells and carotid intima-media thickness in type 2 diabetes: Follow-up of a randomized trial. Eur. J. Prev. Cardiol. 24, 399–408 (2017).

33.        Shai, I. et al. Dietary intervention to reverse carotid atherosclerosis. Circulation 121, 1200–8 (2010).

34.        Aviram, M. et al. Pomegranate juice consumption for 3 years by patients with carotid artery stenosis reduces common carotid intima-media thickness, blood pressure and LDL oxidation. Clin. Nutr. 23, 423–33 (2004).

35.        de Oliveira, P. A. et al. Unsaturated Fatty Acids Improve Atherosclerosis Markers in Obese and Overweight Non-diabetic Elderly Patients. Obes. Surg. 27, 2663–2671 (2017).

36.        Kajbaf, M. H. et al. Does Omega-3 supplementation decrease carotid intima-media thickening in hemodialysis patients? J. Res. Pharm. Pract. 5, 252–256 (2016).

37.        Talari, H. R. et al. Effects of magnesium supplementation on carotid intima-media thickness and metabolic profiles in diabetic haemodialysis patients: a randomised, double-blind, placebo-controlled trial. Br. J. Nutr. 121, 809–817 (2019).

38.        Talari, H. R. et al. The Effects of Folate Supplementation on Carotid Intima-Media Thickness and Metabolic Status in Patients with Metabolic Syndrome. Ann. Nutr. Metab. 69, 41–50 (2016).

39.        Kwok, T. et al. Vitamin B-12 supplementation improves arterial function in vegetarians with subnormal vitamin B-12 status. J. Nutr. Health Aging 16, 569–73 (2012).

40.        Talari, H. R. et al. Effects of Carnitine Administration on Carotid Intima-media Thickness and Inflammatory Factors in Patients with Polycystic Ovary Syndrome: A Randomized, Double-blind, Placebo-controlled Trial. Int. J. Prev. Med. 10, 89 (2019).

41.        Castellino, G. et al. Altilix® Supplement Containing Chlorogenic Acid and Luteolin Improved Hepatic and Cardiometabolic Parameters in Subjects with Metabolic Syndrome: A 6 Month Randomized, Double-Blind, Placebo-Controlled Study. Nutrients 11, (2019).

42.        Najjar, R. S. & Montgomery, B. D. A defined, plant-based diet as a potential therapeutic approach in the treatment of heart failure: A clinical case series. Complement. Ther. Med. 45, 211–214 (2019).

43.        Esselstyn, C. B., Gendy, G., Doyle, J., Golubic, M. & Roizen, M. F. A way to reverse CAD? J. Fam. Pract. 63, 356-364b (2014).

44.        Prasertsri, P., Booranasuksakul, U., Naravoratham, K. & Trongtosak, P. Acute Effects of Passion Fruit Juice Supplementation on Cardiac Autonomic Function and Blood Glucose in Healthy Subjects. Prev. Nutr. food Sci. 24, 245–253 (2019).

45.        Ling, L. et al. Vitamin C preserves endothelial function in patients with coronary heart disease after a high-fat meal. Clin. Cardiol. 25, 219–24 (2002).

46.        Lekakis, J. et al. Polyphenolic compounds from red grapes acutely improve endothelial function in patients with coronary heart disease. Eur. J. Cardiovasc. Prev. Rehabil. 12, 596–600 (2005).

47.        Flammer, A. J. et al. Cardiovascular effects of flavanol-rich chocolate in patients with heart failure. Eur. Heart J. 33, 2172–80 (2012).

48.        Cruz, J. C., Flôr, A. F. L., França-Silva, M. S., Balarini, C. M. & Braga, V. A. Reactive Oxygen Species in the Paraventricular Nucleus of the Hypothalamus Alter Sympathetic Activity During Metabolic Syndrome. Front. Physiol. 6, 384 (2015).

49.        Sun, W. et al. Pomegranate extract decreases oxidative stress and alleviates mitochondrial impairment by activating AMPK-Nrf2 in hypothalamic paraventricular nucleus of spontaneously hypertensive rats. Sci. Rep. 6, 34246 (2016).

50.        Wahl, P., Brixius, K. & Bloch, W. Exercise-induced stem cell activation and its implication for cardiovascular and skeletal muscle regeneration. Minim. Invasive Ther. Allied Technol. 17, 91–99 (2008).

51.        Heiss, C. et al. Improvement of endothelial function with dietary flavanols is associated with mobilization of circulating angiogenic cells in patients with coronary artery disease. J. Am. Coll. Cardiol. 56, 218–24 (2010).

52.        Gould, K. L. et al. Improved stenosis geometry by quantitative coronary arteriography after vigorous risk factor modification. Am. J. Cardiol. 69, 845–53 (1992).

53.        Chockalingam, P., Vinayagam, N. S., Chockalingam, V. & Chockalingam, A. Remarkable regression of coronary atherosclerosis: An interplay of pharmacotherapeutic and lifestyle factors. Indian Heart J. 68, 188–9 (2016).

54.        Esselstyn, C. B., Ellis, S. G., Medendorp, S. V & Crowe, T. D. A strategy to arrest and reverse coronary artery disease: a 5-year longitudinal study of a single physician’s practice. J. Fam. Pract. 41, 560–8 (1995).

55.        Gragnano, F. & Calabrò, P. Role of dual lipid-lowering therapy in coronary atherosclerosis regression: Evidence from recent studies. Atherosclerosis 269, 219–228 (2018).

56.        Biondi-Zoccai, G. et al. What We Have Learned from the Recent Meta-analyses on Diagnostic Methods for Atherosclerotic Plaque Regression. Curr. Atheroscler. Rep. 20, 2 (2018).

57.        Miao, H. et al. Intensive Lipid-Lowering Therapy Ameliorates Asymptomatic Intracranial Atherosclerosis. Aging Dis. 10, 258–266 (2019).

58.        Zhang, Q. et al. Achieving low density lipoprotein-cholesterol<70mg/dL may be associated with a trend of reduced progression of carotid artery atherosclerosis in ischemic stroke patients. J. Neurol. Sci. 378, 26–29 (2017).

59.        Okumura, K. et al. High HDL cholesterol level after treatment with pitavastatin is an important factor for regression in carotid intima-media thickness. Heart Vessels 30, 154–61 (2015).

60.        Puri, R. et al. The beneficial effects of raising high-density lipoprotein cholesterol depends upon achieved levels of low-density lipoprotein cholesterol during statin therapy: Implications for coronary atheroma progression and cardiovascular events. Eur. J. Prev. Cardiol. 23, 474–485 (2016).

61.        Esposito, K., Giugliano, D., Nappo, F., Marfella, R. & Campanian Postprandial Hyperglycemia Study Group. Regression of carotid atherosclerosis by control of postprandial hyperglycemia in type 2 diabetes mellitus. Circulation 110, 214–9 (2004).

62.        Bhanpuri, N. H. et al. Cardiovascular disease risk factor responses to a type 2 diabetes care model including nutritional ketosis induced by sustained carbohydrate restriction at 1 year: an open label, non-randomized, controlled study. Cardiovasc. Diabetol. 17, 56 (2018).

63.        Kruth, H. S. Fluid-phase pinocytosis of LDL by macrophages: a novel target to reduce macrophage cholesterol accumulation in atherosclerotic lesions. Curr. Pharm. Des. 19, 5865–72 (2013).

64.        Steven, S. et al. Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease. Oxid. Med. Cell. Longev. 2019, 1–26 (2019).

65.        Le, N.-A. Lipoprotein-associated oxidative stress: a new twist to the postprandial hypothesis. Int. J. Mol. Sci. 16, 401–19 (2015).

66.        Ishigaki, Y., Oka, Y. & Katagiri, H. Circulating oxidized LDL: a biomarker and a pathogenic factor. Curr. Opin. Lipidol. 20, 363–9 (2009).

67.        Ivanova, E. A., Myasoedova, V. A., Melnichenko, A. A., Grechko, A. V & Orekhov, A. N. Small Dense Low-Density Lipoprotein as Biomarker for Atherosclerotic Diseases. Oxid. Med. Cell. Longev. 2017, 1273042 (2017).

68.        Feig, J. E., Feig, J. L. & Dangas, G. D. The role of HDL on plaque stabilization and regression: basic mechanisms and clinical implications. Coron. Artery Dis. 27, 592 (2016).

69.        Chistiakov, D. A., Myasoedova, V. A., Revin, V. V., Orekhov, A. N. & Bobryshev, Y. V. The phenomenon of atherosclerosis reversal and regression: Lessons from animal models. Exp. Mol. Pathol. 102, 138–145 (2017).

70.        Yu-Poth, S. et al. Lowering dietary saturated fat and total fat reduces the oxidative susceptibility of LDL in healthy men and women. J. Nutr. 130, 2228–37 (2000).

71.        Mata, P. et al. Effect of dietary fat saturation on LDL oxidation and monocyte adhesion to human endothelial cells in vitro. Arterioscler. Thromb. Vasc. Biol. 16, 1347–55 (1996).

72.        Nicholls, S. J. et al. Consumption of saturated fat impairs the anti-inflammatory properties of high-density lipoproteins and endothelial function. J. Am. Coll. Cardiol. 48, 715–20 (2006).

73.        Cedó, L. et al. Consumption of polyunsaturated fat improves the saturated fatty acid-mediated impairment of HDL antioxidant potential. Mol. Nutr. Food Res. 59, 1987–96 (2015).

74.        O’Reilly, M. et al. High-Density Lipoprotein Proteomic Composition, and not Efflux Capacity, Reflects Differential Modulation of Reverse Cholesterol Transport by Saturated and Monounsaturated Fat Diets. Circulation 133, 1838–50 (2016).

75.        Roberts, C. K., Ng, C., Hama, S., Eliseo, A. J. & Barnard, R. J. Effect of a short-term diet and exercise intervention on inflammatory/anti-inflammatory properties of HDL in overweight/obese men with cardiovascular risk factors. J. Appl. Physiol. 101, 1727–32 (2006).

76.        Rosenson, R. S. et al. Dysfunctional HDL and atherosclerotic cardiovascular disease. Nat. Rev. Cardiol. 13, 48–60 (2016).

77.        Fernández-Castillejo, S. et al. Determinants of HDL Cholesterol Efflux Capacity after Virgin Olive Oil Ingestion: Interrelationships with Fluidity of HDL Monolayer. Mol. Nutr. Food Res. 61, (2017).

78.        Hernáez, Á. et al. Mediterranean Diet Improves High-Density Lipoprotein Function in High-Cardiovascular-Risk Individuals: A Randomized Controlled Trial. Circulation 135, 633–643 (2017).

79.        Anderson, J. L. C. et al. High density lipoprotein (HDL) particles from end-stage renal disease patients are defective in promoting reverse cholesterol transport. Sci. Rep. 7, 41481 (2017).

80.        Thyagarajan, N., Brannan, A. M. & Brown, R. J. Can we make high-density lipoproteins great again? Circulation 133, 1833–1835 (2016).

81.        Cesar, T. B., Aptekmann, N. P., Araujo, M. P., Vinagre, C. C. & Maranhão, R. C. Orange juice decreases low-density lipoprotein cholesterol in hypercholesterolemic subjects and improves lipid transfer to high-density lipoprotein in normal and hypercholesterolemic subjects. Nutr. Res. 30, 689–94 (2010).

82.        Millar, C. L., Duclos, Q. & Blesso, C. N. Effects of Dietary Flavonoids on Reverse Cholesterol Transport, HDL Metabolism, and HDL Function. Adv. Nutr. 8, 226–239 (2017).

83.        Zhang, J. et al. Walnut oil increases cholesterol efflux through inhibition of stearoyl CoA desaturase 1 in THP-1 macrophage-derived foam cells. Nutr. Metab. (Lond). 8, 61 (2011).

84.        Pizzini, A. et al. The role of omega-3 fatty acids in reverse cholesterol transport: A review. Nutrients 9, (2017).

85.        Rosenblat, M., Volkova, N., Coleman, R. & Aviram, M. Anti-oxidant and anti-atherogenic properties of liposomal glutathione: studies in vitro, and in the atherosclerotic apolipoprotein E-deficient mice. Atherosclerosis 195, e61-8 (2007).

86.        Han, S. et al. Cereal fiber improves blood cholesterol profiles and modulates intestinal cholesterol metabolism in C57BL/6 mice fed a high-fat, high-cholesterol diet. Food Nutr. Res. 63, (2019).

87.        Soliman, G. A. Dietary Fiber, Atherosclerosis, and Cardiovascular Disease. Nutrients 11, (2019).

88.        Feingold, K. R. & Grunfeld, C. The acute phase response inhibits reverse cholesterol transport. J. Lipid Res. 51, 682–684 (2010).

89.        Canyelles, M. et al. Trimethylamine N-oxide: A link among diet, gut microbiota, gene regulation of liver and intestine cholesterol homeostasis and HDL function. Int. J. Mol. Sci. 19, (2018).

90.        Biddinger, S. B. et al. Hepatic insulin resistance is sufficient to produce dyslipidemia and susceptibility to atherosclerosis. Cell Metab. 7, 125–34 (2008).

91.        Julve, J. et al. Methionine-induced hyperhomocysteinemia impairs the antioxidant ability of high-density lipoproteins without reducing in vivo macrophage-specific reverse cholesterol transport. Mol. Nutr. Food Res. 57, 1814–24 (2013).

92.        Yang, N. et al. Homocysteine diminishes apolipoprotein A-I function and expression in patients with hypothyroidism: a cross-sectional study. Lipids Health Dis. 15, 123 (2016).

93.        Obeid, R. & Herrmann, W. Homocysteine and lipids: S-Adenosyl methionine as a key intermediate. FEBS Lett. 583, 1215–1225 (2009).

94.        Momin, M. et al. Relationship between plasma homocysteine level and lipid profiles in a community-based Chinese population. Lipids Health Dis. 16, 54 (2017).

95.        Tehlivets, O. Homocysteine as a risk factor for atherosclerosis: is its conversion to s-adenosyl-L-homocysteine the key to deregulated lipid metabolism? J. Lipids 2011, 702853 (2011).

96.        Lowenstine, L. J., McManamon, R. & Terio, K. A. Comparative Pathology of Aging Great Apes: Bonobos, Chimpanzees, Gorillas, and Orangutans. Vet. Pathol. 53, 250–276 (2016).

97.        Milton, K. The critical role played by animal source foods in human (Homo) evolution. J. Nutr. 133, 3886S-3892S (2003).

98.        Xiangdong, L. et al. Animal models for the atherosclerosis research: a review. Protein Cell 2, 189–201 (2011).

99.        Lee, S. et al. Low-Density Lipoprotein Particle Size Subfractions and Cerebral Amyloidosis. J. Alzheimers. Dis. 68, 983–990 (2019).

100.      Szoeke, C. et al. Apolipoprotein E4 Mediates the Association Between Midlife Dyslipidemia and Cerebral Amyloid in Aging Women. J. Alzheimers. Dis. 68, 105–114 (2019).

101.      Pham, T. et al. Evidence for a novel human-specific xeno-auto-antibody response against vascular endothelium. Blood 114, 5225–35 (2009).

102.      Kawanishi, K. et al. Human species-specific loss of CMP-N-acetylneuraminic acid hydroxylase enhances atherosclerosis via intrinsic and extrinsic mechanisms. Proc. Natl. Acad. Sci. U. S. A. 116, 16036–16045 (2019).

103.      Uribarri, J. et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J. Am. Diet. Assoc. 110, 911–16.e12 (2010).

104.      Jayedi, A., Shab-Bidar, S., Eimeri, S. & Djafarian, K. Fish consumption and risk of all-cause and cardiovascular mortality: a dose-response meta-analysis of prospective observational studies. Public Health Nutr. 21, 1297–1306 (2018).

105.      Hall, W. L. The future for long chain n-3 PUFA in the prevention of coronary heart disease: do we need to target non-fish-eaters? Proc. Nutr. Soc. 76, 408–418 (2017).

106.      Zhang, Y. et al. Association of fish and long-chain omega-3 fatty acids intakes with total and cause-specific mortality: prospective analysis of 421 309 individuals. J. Intern. Med. 284, 399–417 (2018).

107.      Donat-Vargas, C. et al. Cardiovascular and cancer mortality in relation to dietary polychlorinated biphenyls and marine polyunsaturated fatty acids: a nutritional-toxicological aspect of fish consumption. J. Intern. Med. 287, 197–209 (2020).

108.      Aranda, N. et al. Consumption of seafood and its estimated heavy metals are associated with lipid profile and oxidative lipid damage on healthy adults from a Spanish Mediterranean area: A cross-sectional study. Environ. Res. 156, 644–651 (2017).

109.      Shi, L. et al. Joint Analysis of Metabolite Markers of Fish Intake and Persistent Organic Pollutants in Relation to Type 2 Diabetes Risk in Swedish Adults. J. Nutr. 149, 1413–1423 (2019).

110.      Cameron-Smith, D., Albert, B. B. & Cutfield, W. S. Fishing for answers: is oxidation of fish oil supplements a problem? J. Nutr. Sci. 4, e36 (2015).

111.      Tirosh, O., Shpaizer, A. & Kanner, J. Lipid Peroxidation in a Stomach Medium Is Affected by Dietary Oils (Olive/Fish) and Antioxidants: The Mediterranean versus Western Diet. J. Agric. Food Chem. 63, 7016–23 (2015).

112.      Kanner, J. et al. Redox homeostasis in stomach medium by foods: The Postprandial Oxidative Stress Index (POSI) for balancing nutrition and human health. Redox Biol. 12, 929–936 (2017).

113.      Purcell, R., Latham, S. H., Botham, K. M., Hall, W. L. & Wheeler-Jones, C. P. D. High-fat meals rich in EPA plus DHA compared with DHA only have differential effects on postprandial lipemia and plasma 8-isoprostane F2α concentrations relative to a control high-oleic acid meal: a randomized controlled trial. Am. J. Clin. Nutr. 100, 1019–28 (2014).

114.      Finnegan, Y. E. et al. Plant- and marine-derived n-3 polyunsaturated fatty acids have differential effects on fasting and postprandial blood lipid concentrations and on the susceptibility of LDL to oxidative modification in moderately hyperlipidemic subjects. Am. J. Clin. Nutr. 77, 783–95 (2003).

115.      Mason, R. P. & Sherratt, S. C. R. Omega-3 fatty acid fish oil dietary supplements contain saturated fats and oxidized lipids that may interfere with their intended biological benefits. Biochem. Biophys. Res. Commun. 483, 425–429 (2017).

116.      Rundblad, A., Holven, K. B., Ottestad, I., Myhrstad, M. C. & Ulven, S. M. High-quality fish oil has a more favourable effect than oxidised fish oil on intermediate-density lipoprotein and LDL subclasses: a randomised controlled trial. Br. J. Nutr. 117, 1291–1298 (2017).

117.      Girard, C., Charette, T., Leclerc, M., Shapiro, B. J. & Amyot, M. Cooking and co-ingested polyphenols reduce in vitro methylmercury bioaccessibility from fish and may alter exposure in humans. Sci. Total Environ. 616617, 863–874 (2018).

118.      Barceló-Coblijn, G. et al. Flaxseed oil and fish-oil capsule consumption alters human red blood cell n-3 fatty acid composition: a multiple-dosing trial comparing 2 sources of n-3 fatty acid. Am. J. Clin. Nutr. 88, 801–9 (2008).

119.      Domenichiello, A. F., Kitson, A. P. & Bazinet, R. P. Is docosahexaenoic acid synthesis from α-linolenic acid sufficient to supply the adult brain? Prog. Lipid Res. 59, 54–66 (2015).

120.      Fleming, J. A. & Kris-Etherton, P. M. The evidence for α-linolenic acid and cardiovascular disease benefits: Comparisons with eicosapentaenoic acid and docosahexaenoic acid. Adv. Nutr. 5, 863S–76S (2014).

121.      Abdelhamid, A. S. et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst. Rev. (2020). doi:10.1002/14651858.CD003177.pub5

122.      Kris-Etherton, P. M. et al. Recent Clinical Trials Shed New Light on the Cardiovascular Benefits of Omega-3 Fatty Acids. Methodist Debakey Cardiovasc. J. 15, 171–178 (2019).

123.      Manson, J. E. et al. Marine n-3 Fatty Acids and Prevention of Cardiovascular Disease and Cancer. N. Engl. J. Med. 380, 23–32 (2019).

124.      Rizzo, G. et al. Vitamin B12 among vegetarians: Status, assessment and supplementation. Nutrients 8, 1–23 (2016).

125.      Watanabe, F. & Bito, T. Vitamin B12 sources and microbial interaction. Exp. Biol. Med. (Maywood). 243, 148–158 (2018).

126.      Woo, K. S., Kwok, T. C. Y. & Celermajer, D. S. Vegan diet, subnormal vitamin B-12 status and cardiovascular health. Nutrients 6, 3259–3273 (2014).

127.      Schmidt, A., Call, L.-M., Macheiner, L. & Mayer, H. K. Determination of vitamin B12 in four edible insect species by immunoaffinity and ultra-high performance liquid chromatography. Food Chem. 281, 124–129 (2019).

128.      Satija, A. et al. Plant-Based Dietary Patterns and Incidence of Type 2 Diabetes in US Men and Women: Results from Three Prospective Cohort Studies. PLoS Med. 13, e1002039 (2016).

129.      Satija, A. et al. Healthful and Unhealthful Plant-Based Diets and the Risk of Coronary Heart Disease in U.S. Adults. J. Am. Coll. Cardiol. 70, 411–422 (2017).

130.      Acosta-Navarro, J. C. et al. Consumption of animal-based and processed food associated with cardiovascular risk factors and subclinical atherosclerosis biomarkers in men. Rev. Assoc. Med. Bras. 65, 43–50 (2019).

131.      Jakobsen, M. U. et al. Intake of carbohydrates compared with intake of saturated fatty acids and risk of myocardial infarction: importance of the glycemic index. Am. J. Clin. Nutr. 91, 1764–8 (2010).

132.      Li, Y. et al. Saturated Fats Compared With Unsaturated Fats and Sources of Carbohydrates in Relation to Risk of Coronary Heart Disease: A Prospective Cohort Study. J. Am. Coll. Cardiol. 66, 1538–1548 (2015).

133.      Sanchez, A. et al. Diets with customary levels of fat from plant origin may reverse coronary artery disease. Med. Hypotheses 122, 103–105 (2019).

134.      Agarwal, A. & Ioannidis, J. P. A. PREDIMED trial of Mediterranean diet: Retracted, republished, still trusted? BMJ 364, 1–5 (2019).

135.      Chen, Z. et al. Dietary protein intake and all-cause and cause-specific mortality: results from the Rotterdam Study and a meta-analysis of prospective cohort studies. Eur. J. Epidemiol. 35, 411–429 (2020).

136.      Tobias, D. K. et al. Circulating Branched-Chain Amino Acids and Incident Cardiovascular Disease in a Prospective Cohort of US Women. Circ. Genomic Precis. Med. 11, e002157 (2018).

137.      Dong, Z. et al. Association of sulfur amino acid consumption with cardiometabolic risk factors: Cross-sectional findings from NHANES III. EClinicalMedicine 0, 100248 (2020).

138.      Zhang, X. et al. High-protein diets increase cardiovascular risk by activating macrophage mTOR to suppress mitophagy. Nat. Metab. 2, 110–125 (2020).

139.      Selhub, J. & Troen, A. M. Sulfur amino acids and atherosclerosis: a role for excess dietary methionine. Ann. N. Y. Acad. Sci. 1363, 18–25 (2016).

140.      Cai, W. et al. High levels of dietary advanced glycation end products transform low-density lipoprotein into a potent redox-sensitive mitogen-activated protein kinase stimulant in diabetic patients. Circulation 110, 285–91 (2004).

141.      Di Pino, A. et al. Low advanced glycation end product diet improves the lipid and inflammatory profiles of prediabetic subjects. J. Clin. Lipidol. 10, 1098–108 (2016).

142.      Stirban, A. & Tschöpe, D. Vascular Effects of Dietary Advanced Glycation End Products. Int. J. Endocrinol. 2015, 836498 (2015).

143.      Etemadi, A. et al. Mortality from different causes associated with meat, heme iron, nitrates, and nitrites in the NIH-AARP Diet and Health Study: population based cohort study. BMJ 357, j1957 (2017).

144.      Kim, K. et al. Role of Total, Red, Processed, and White Meat Consumption in Stroke Incidence and Mortality: A Systematic Review and Meta‐Analysis of Prospective Cohort Studies. J. Am. Heart Assoc. 6, (2017).

145.      Bergeron, N., Chiu, S., Williams, P. T., M King, S. & Krauss, R. M. Effects of red meat, white meat, and nonmeat protein sources on atherogenic lipoprotein measures in the context of low compared with high saturated fat intake: a randomized controlled trial. Am. J. Clin. Nutr. 110, 24–33 (2019).

146.      Wang, Z. et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 40, 583–594 (2019).

147.      Fang, X. et al. Dietary intake of heme iron and risk of cardiovascular disease: a dose-response meta-analysis of prospective cohort studies. Nutr. Metab. Cardiovasc. Dis. 25, 24–35 (2015).

148.      Koeth, R. A. et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 19, 576–85 (2013).

149.      Shang, R., Sun, Z. & Li, H. Effective dosing of L-carnitine in the secondary prevention of cardiovascular disease: a systematic review and meta-analysis. BMC Cardiovasc. Disord. 14, 88 (2014).

150.      DiNicolantonio, J. J., McCarty, M. & OKeefe, J. Association of moderately elevated trimethylamine N-oxide with cardiovascular risk: is TMAO serving as a marker for hepatic insulin resistance. Open Hear. 6, e000890 (2019).

151.      Zhong, V. W. et al. Associations of Dietary Cholesterol or Egg Consumption With Incident Cardiovascular Disease and Mortality. JAMA 321, 1081–1095 (2019).

152.      Shapira, N. & Pinchasov, J. Modified egg composition to reduce low-density lipoprotein oxidizability: high monounsaturated fatty acids and antioxidants versus regular high n-6 polyunsaturated fatty acids. J. Agric. Food Chem. 56, 3688–93 (2008).

153.      Spence, J. D., Jenkins, D. J. a & Davignon, J. Dietary cholesterol and egg yolks: not for patients at risk of vascular disease. Can. J. Cardiol. 26, e336-9 (2010).

154.      Fontecha, J., Calvo, M. V., Juarez, M., Gil, A. & Martínez-Vizcaino, V. Milk and Dairy Product Consumption and Cardiovascular Diseases: An Overview of Systematic Reviews and Meta-Analyses. Adv. Nutr. 10, S164–S189 (2019).

155.      Yu, E. & Hu, F. B. Dairy Products, Dairy Fatty Acids, and the Prevention of Cardiometabolic Disease: a Review of Recent Evidence. Curr. Atheroscler. Rep. 20, (2018).

156.      Zhang, K., Chen, X., Zhang, L. & Deng, Z. Fermented dairy foods intake and risk of cardiovascular diseases: A meta-analysis of cohort studies. Crit. Rev. Food Sci. Nutr. 60, 1189–1194 (2020).

157.      Chen, M. et al. Dairy fat and risk of cardiovascular disease in 3 cohorts of US adults. Am. J. Clin. Nutr. 104, 1209–1217 (2016).

158.      Hooper, L. et al. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst. Rev. (2020). doi:10.1002/14651858.CD011737.PUB2


No comments:

Post a Comment