Life Extension Magazine®

Mitochondrial damage that is predicted before the pictured DNA damage

Reverse Mitochondrial Damage

Age-related deterioration of the mitochondria in our cells is associated with an array of deadly conditions, ranging from senility to diabetes and heart failure. Researchers have recently discovered that this deadly process can be reversed with cellular energizers that help restore and preserve mitochondrial function.

Scientifically reviewed by: Juanita Enogieru, MS, RD/N, in August 2023. Written by: Steven Ryan.

Cellular Aging and Mitochondrial Function

As we age, a decline in mitochondrial function is associated with various conditions that impact our overall health, ranging from diabetes to heart failure. However, researchers have discovered that a decline in mitochondrial health can be reversed with cellular energizers that can help maintain mitochondrial function.

Reverse Mitochondrial Damage

Progressive loss of function in the mitochondria—the cellular generators responsible for nearly all the body’s energy output—speeds aging and death.

Mitochondrial dysfunction has been linked to an array of degenerative illnesses, ranging from diabetes and neurological disorders to heart failure.1,2

In 2007, a group of researchers reported a major (but little-known) breakthrough in our understanding of how mitochondrial dysfunction unfolds—and what can be done to protect yourself against its lethal impact.3

They discovered that potentially deadly defects in human mitochondria, including molecular decay and membrane injury, begin to appear and can be detected nearly a decade before the onset of permanent damage to the DNA.3

More importantly, their analysis revealed that in its initial stages, mitochondrial dysfunction is reversible, enabling the life and health of cells to be prolonged at the molecular level. The key lies in early interventions to ensure optimal mitochondrial function before irreversible DNA damage occurs.

In this article, we review the latest research on a set of compounds that specifically target and enhance mitochondrial function through multiple modes of action.

What you need to know

  • Mitochondria are the cellular organelles that power every energy-requiring bodily process.
  • Progressive loss of function in the mitochondria—the cellular power generators responsible for nearly all energy output in the body—speeds cell aging and death.
  • Researchers recently discovered that signs of age-related mitochondrial damage appear nearly a decade before the onset of permanent DNA damage.
  • They also found that mitochondrial decay and dysfunction are reversible.
  • A handful of mitochondrial-energizing nutrients have been shown to offer powerful protection from mitochondrial damage and dysfunction.
  • CoQ10 speeds mitochondrial electron transport, increases energy production, and protects tissues from mitochondrial decline.
  • Shilajit, an ancient Indian adaptogen, enhances CoQ10’s mitochondrial benefits and supports levels of the active ubiquinol form.
  • R-alpha-lipoic acid further supports mitochondrial energy production.
  • Acetyl-L-carnitine “feeds” energy-releasing molecules to mitochondria, improving their efficiency and preventing damage.

The Cellular Death Spiral

Mitochondria are responsible for converting energy from the food you ingest into usable “currency.” Carbohydrates, fats, and proteins are broken down inside your cells into components that enter the cellular powerhouses known as mitochondria. Throughout this cellular journey, these “macronutrients” undergo a complex series of biochemical transformations that generate adenosine triphosphate (ATP), the molecular energy currency behind all biological functions. To give you an idea of ATP’s life-sustaining importance, your body converts a volume of ATP equal to your entire weight every day.

The Cellular Death Spiral

At the core of this energy conversion matrix lies the electron transport chain, a series of molecules embedded in the inner mitochondrial membrane. It serves as the “power line” through which needed chemical energy is released and transferred into vital ATP.

This energy-intensive process throws off an immense number of electrons within the mitochondria, resulting in constant exposure to free radicals—and rendering the mitochondria especially vulnerable to oxidative damage.4-6

The result is a cellular death spiral: the mitochondria gradually deteriorate, leading to a decrease in vital ATP production and a deadly increase in free radical generation. Over time, this continuous free-radical onslaught destroys the mitochondria through progressive membrane damage and molecular decay.

As levels of oxidative damage from mitochondrial dysfunction steadily rise with age,7,8 the body’s antioxidant defenses gradually weaken at the same time, accelerating cellular senescence and death.4,9,10

Left unchecked, this fatal cycle speeds the general decline in overall function that accompanies aging4,11 and contributes to the onset of degenerative disease.12-15

CoQ10’s Rejuvenating Power

Coenzyme Q10 (CoQ10) powerfully safeguards mitochondria from age-related decay and death through two principal pathways.

It plays an essential role in the electron transport chain, facilitating the efficient transfer of electrons into ATP for use in cellular function.16 CoQ10 resides primarily on the inner membranes of the mitochondria; 95% of all cellular energy production depends on it.

CoQ10 also acts as a powerful free radical scavenger, neutralizing their lethal action and dramatically reducing oxidative damage. The more available CoQ10 in the mitochondria, the less free radical damage.16 This is one of the reasons why the highest CoQ10 concentrations are found in the most energy-intensive organs: the brain, heart, liver, and kidneys.17

CoQ10 levels in our vital organs, like the heart, steadily rise after birth and peak at about 20 years of age. After that, they undergo a continuous decline.18 Fortunately, three decades of cutting-edge research have shown us how to restore CoQ10 levels in the mitochondria to slow and even reverse the effects of aging.19-22

In pre-clinical models, CoQ10 supplementation protects tissue from lethal DNA damage and increases lifespan.23 It boosts mitochondrial function and total energy output in heart muscle in aging animals.24 And in animal models, lifelong CoQ10 supplementation has been shown to decrease oxidative damage in skeletal muscle, increase native antioxidant enzymes, and favorably modify age-related changes in muscular energy metabolism.25

Until 2007, the only form of CoQ10 available was ubiquinone. Unfortunately, the ubiquinone form of CoQ10 has limited absorption.26 Another form of CoQ10, known as ubiquinol, remains up to eight times longer in the blood.27,28

The Heart Health Warrior

Dense with mitochondria, the heart requires more energy than any other organ—and the greatest concentration of CoQ10.29 This is especially true for aging individuals, even those with advanced chronic heart disease. Scores of studies show that chronic heart conditions, including congestive heart failure (CHF), are characterized by diminished levels of CoQ10 in heart tissue. Its therapeutic benefit has proven just as profound for these individuals.

In a 2008 study, standard CoQ10 supplements failed to improve either CoQ10 levels or cardiac performance in individuals suffering from CHF, while ubiquinol succeeded on both fronts.30

The study involved individuals with advanced CHF. Their hearts pumped less than half as well as normal, with low CoQ10 levels despite taking an average of 450 mg/day of standard CoQ10. When the same people took ubiquinol (580 mg/day on average), their CoQ10 blood levels vaulted into the therapeutic range—and their hearts’ pumping action improved by 77%.

CoQ10’s Rejuvenating Power

At the outset of this study, every participant suffered from category IV CHF (the most severe form), presenting continuous symptoms—even at rest—with severely limited activity. By the end of the study, the average CHF score had fallen to category II, indicated by mild symptoms (such as slight shortness of breath and/or angina) and minimal limitations during ordinary activity.31

CoQ10 supplementation also increases heart muscle contractility—the strength of the heart’s squeezing action—enabling the heart to pump more blood more efficiently, even in patients with advanced CHF.32

Chronic CoQ10 deficiency has been linked to poor surgical outcomes in elderly patients compared to younger ones.21,33 By energizing cardiac mitochondria, CoQ10 exerts a powerful effect on cardiac performance in individuals with CHF. Supplementation with CoQ10 and other antioxidants and heart-energizing nutrients such as L-carnitine and taurine reduces distended heart volume in patients—a vital factor in reducing the risk of bypass surgery.34

Following a heart attack, cardiac tissue is at great risk for further injury, including a second attack. In patients recovering from recent heart attacks, just 120 mg of CoQ10 per day produced remarkable benefits.35 After one year, only 25% of supplemented patients suffered a cardiac event, compared with 45% in the placebo group, and cardiac deaths were significantly fewer compared with placebo. Supplemented patients also had increased high-density lipoprotein (HDL) and dramatically lower measures of oxidative stress.

CoQ10 also benefits people undergoing cardiac surgery, particularly older adults whose outcomes tend to be worse than younger people’s, owing to declining mitochondrial function and density in heart tissue.21

Treating heart tissue with CoQ10 improves its metabolic stress response and speeds recovery after ischemia (loss of blood flow)—two major concerns after cardiac surgery.21 Oral CoQ10 therapy for one week before surgery improves mitochondrial energy efficiency and post-operative heart function, while reducing heart muscle damage and shortening hospital stays.21 A 2008 study also showed significantly fewer arrhythmias (abnormal heart beats), less need for medications to boost cardiac strength, and less need for blood transfusion in patients who received CoQ10 supplementation prior to cardiac surgery, compared to patients who did not receive CoQ10.36

Potent Endothelial Defense

The lining of our blood vessels, or endothelium, regulates blood flow and pressure, and is easily damaged by oxidative stress and inflammation, which increases the risk of atherosclerosis and erectile dysfunction. CoQ10 powerfully protects endothelial function, an effect that is likely due to its uniquely beneficial effect on mitochondrial function.37

One study of the ubiquinol form of CoQ10 showed that it protects against hypertension, improves endothelial function, and reduces cardiac enlargement in stroke-prone rats.38 When humans with endothelial dysfunction took 300 mg/day of CoQ10 orally for one month, their blood vessels relaxed more readily and they moved more oxygenated blood into tissues compared with placebo recipients.39

People with type 2 diabetes are at particularly high risk for endothelial dysfunction, and have more heart attacks and strokes as a result.40 CoQ10 supplementation is especially effective at improving endothelial function in this population.41 Diabetics (and others) often need to take statin-type lipid-lowering medications to control their cholesterol levels. Unfortunately, these drugs (also known as HMG Co A reductase inhibitors) are known to deplete CoQ10 and can cause muscle pain that may be related to this depletion.42 CoQ10 overcomes this problem and has been shown to improve endothelial function in diabetic patients on statins.43

Mitochondrial dysfunction is linked to a broad range of degenerative illnesses, from diabetes and neurological disorders to heart disease.

Muscular Energy Enhancement

Exercise can boost longevity and even increase mitochondrial density in the short term; however, exercise can also damage the mitochondria in the long term.44,45 The high rate of oxygen and electron flow that exercise requires can lead to chronically low ATP levels, which may exert negative effects during vigorous exercise.46 CoQ10 supplementation can counteract such effects, enhancing the adaptive response of skeletal muscle following exercise.47

Muscular Energy Enhancement

CoQ10 supplementation before exercise increases muscle CoQ10 levels, reduces muscular oxidant stress, and may increase the amount of time you can exercise until exhaustion.48 To take one dramatic example, CoQ10 supplementation of 300 mg/day resulted in improved blood markers of exercise-induced muscle injury among elite Japanese Kendo athletes (a form of martial arts) practicing up to five-and-a-half hours per day.49

CoQ10 at just 100 mg/day even enhances performance of normally sedentary men during repeated bouts of exercising.50 Supplementation of 300 mg/day enabled adults to increase their velocity on a stationary bike compared with placebo, while reducing fatigue.51

CoQ10’s remarkable energy-boosting effects can also reduce adverse effects associated with statin therapy, including fatigue, muscle pain, shortness of breath, memory loss, and nerve pain in the extremities.52 Patients with statin-induced fatigue who stopped the drug and took 240 mg/day of CoQ10 saw a decrease in fatigue from 84% to just 15%; a drop in muscle pain from 64% to 6%, and a decline in shortness of breath from 58% to 12%. These are all manifestations of restored mitochondrial energy and function—and the study found no adverse consequences from discontinuing the statin drugs. (You should never abruptly discontinue any medication without discussing it with your doctor.)

System-Wide Protection

CoQ10 also has dramatic benefits in other tissues, particularly in the brain, eyes, and skin.

There’s growing recognition of the role of “brain energetics,” including mitochondrial health, in causing (or preventing) progressively fatal neurological conditions, including Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis (ALS or Lou Gehrig’s Disease).12,53 Animal studies show that CoQ10 supplementation increases brain levels of CoQ10, sustaining the brain’s tremendous energy needs. At the same time it reduces brain injury and increases life span in mice with a neurodegenerative disease similar to ALS.54

CoQ10 benefits peripheral nerves as well. People with diabetes often succumb to painful diabetic neuropathy and decreased ability to sense pressure, which can lead to disastrous injuries. Studies of diabetic rats with neuropathy show that CoQ10 improved nerve conduction velocity and strength of nerve impulses.55

Nerve cells in the eye are faced with enormous energy demands—they must convert light into electrical impulses, while protecting themselves from the damaging effects of both.56 Researchers now know that mitochondrial health is vital to sustaining the health of cells in the retina, where optical nerves are concentrated.57 Unfortunately, CoQ10 levels in the retina decline rapidly with age, leaving delicate cells vulnerable.58

In combination with acetyl-L-carnitine and omega-3 fatty acids, CoQ10 generated dramatic results in studies of individuals suffering from early age-related macular degeneration. Supplemented patients had a 10-fold lower risk of worsening over a 12-month period, compared with those who received placebo!59 Pre-clinical models suggest that CoQ10 may even protect retinal tissue from the effects of glaucoma.56

Our skin shows the most immediate and visible signs of aging. Only recently have we learned how much this has to do with mitochondrial dysfunction in skin cells: skin biopsies from older people show substantially less mitochondrial function than those from younger people.60

Increased oxidative damage from diminished mitochondrial function has been shown to trigger inflammation and launch protein-destroying enzymes into action. Over time this leads to a weakening of the delicate matrix of skin tissue, spots, wrinkles, dryness—even cancer.61 Many studies show that topical CoQ10 treatment inhibits inflammatory cytokines, reduces wrinkling enzyme production, and improves the appearance and radiation-resistance of older skin.61-63 Boosting CoQ10 through oral supplementation also affords vital protection.64

Advanced Mitochondrial Threats: Glycation and Lipoxidation

The chemical reaction of glucose with proteins and fats that occurs over a lifetime produces advanced glycation end-products (AGEs) and advanced lipoxidation end-products (ALEs).104 These deadly molecules cause oxidative and inflammatory damage to mitochondria, hastening mitochondrial dysfunction and aging.105-109 Specific compounds have been shown to provide targeted mitochondrial defense against glycation and the inflammation it produces.

  • Carnosine is a nutrient comprised of two amino acids. It’s a natural antioxidant and anti-glycation molecule proven to reduce reactive oxygen and nitrogen species resulting from chronic glucose exposure, while also binding to potentially dangerous metal ions (chelation). These features make it attractive as an anti-aging, anti-Alzheimer’s agent.110-112
  • Luteolin is a flavonoid with potent anti-inflammatory effects.113,114 It directly inhibits AGE formation at early, middle, and late stages in their development—more powerfully than standard chemical AGE-inhibitors.115,116 It also directly counters the sugar-induced mitochondrial damage caused by reduction in a survival protein called Bcl-2.117
  • Benfotiamine is a fat-soluble form of thiamine (vitamin B1). Its higher bioavailability allows it to strongly increase glycation-fighting thiamine levels in blood and tissues in normal people and in people with either type 1 or 2 diabetes.118-120 Benfotiamine powerfully reduces AGE production and damage to vascular endothelial cells under high-glucose conditions.121,122 It blocks three distinct pathways of sugar-induced tissue damage to protect against retinal damage in diabetes.123
  • Pyridoxal-5’-phosphate (PLP) is the biologically active form of vitamin B6. It is a powerful inhibitor of both protein and fat glycation.124,125 Glycation reductions by PLP are credited with reducing sugar-induced blood vessel and kidney damage from diabetes.126,127

Each of these nutrients works through distinct pathways, acting as a “therapeutic cocktail” that provides maximum protection against glycation-induced toxicity and mitochondrial damage.128

Mitochondrial Protection with a Potent Adaptogen

Long known to Ayurvedic practitioners for its healing power, shilajit is an organic substance harvested from biomass high in the Himalayas.65,66 It acts as a powerful adaptogen, providing broad systemic defense against stress and illness. Cutting-edge scientific analysis has isolated humic substances as the principal active ingredients that enhance mitochondrial energy flow.67

Mitochondrial Protection with a Potent Adaptogen

In 2009, a series of landmark studies detailed for the first time how shilajit works on energy metabolism.

Mice subjected to strenuous exercise underwent expected ATP declines in muscle, blood, and brain tissue. When supplemented with shilajit, ATP loss was sharply reduced.68 Other biochemical markers of energy status also dramatically improved in the supplemented animals—including levels of CoQ10, which fell twice as fast in control mice as in supplemented animals. When given in combination, CoQ10 and shilajit displayed a powerful synergistic effect. Energy parameters such as CoQ10 levels increased significantly more than with either supplement alone.

Further analysis brought some of its key mechanisms of action to light. Shilajit contains two primary components, fulvic acid and DBPs (dibenzo-a-pyrones). Fulvic acid independently stimulates mitochondrial energy metabolism, protects mitochondrial membranes from oxidative damage, and helps channel electron-rich DBPs into the mitochondria to support the electron transfer chain.69,70 Fulvic acid works as an electron “shuttle,” augmenting CoQ10 to speed electron flow within mitochondria.71-73

The DBPs in shilajit serve as electron “reservoirs,” replenishing electrons lost by CoQ10 when it donates them to free radicals (thereby neutralizing them).70,74

When laboratory mice are supplemented with oral CoQ10 alone, CoQ10 levels rise in heart, liver, and kidney tissue, as might be expected.75 When DBPs from shilajit are added to the supplement, CoQ10 levels rise still further—as much as 29% in the liver.75

A recent study suggests that DBPs from shilajit preserve CoQ10 in its superior ubiquinol form.75

Preliminary findings suggest that shilajit protects human tissue from lost energy in the form of ATP, while maximizing benefits from CoQ10, with dramatic improvement in exercise performance.76 In an as-yet unpublished study, people who took shilajit 200 mg once daily for 15 days registered 14% higher post-exercise ATP levels in the blood—equivalent to levels in people who hadn’t exercised at all. The average number of steps they took on a standardized dynamic exercise test rose significantly, and their mean fitness scores increased by 15%—without any intervening exercise training.

In pre-clinical studies, shilajit has been shown to possess a number of additional benefits, allowing it to work in synergy with CoQ10 to protect and support mitochondrial health:

• Preliminary unpublished studies showed that shilajit (250 mg twice daily for 90 days) lowered fasting blood sugar and a measure of systemic inflammation called the ESR (erythrocyte sedimentation rate), while increasing hemoglobin levels and platelet counts.77,78

• Shilajit protected laboratory rats from developing chemically-induced diabetes through its free-radical scavenging properties.70

• Shilajit augmented learning acquisition and memory retrieval in laboratory rats while reducing manifestations of anxiety during maze experiments.70

• Shilajit reduced levels of the enzyme acetylcholinesterase that destroys the vital neurotransmitter acetylcholine. This effect may help to prevent or treat Alzheimer’s disease by maintaining levels of the neurotransmitter.70

• Shilajit increased levels of the neurotransmitter dopamine in rat brains, making it an attractive candidate for treatment of Parkinson’s disease and other movement disorders.70

• In pre-clinical studies, shilajit produced significant increases in the endogenous antioxidants superoxide dismutase (SOD), catalase, and glutathione peroxidase in brain tissue. Increased levels of these enzymes protect vulnerable brain cells against the oxidative damage that leads to brain aging and cognitive decline.70

A Complementary Coenzyme

Lipoic acid is a naturally occurring compound found in mitochondria. Like CoQ10, it is a coenzyme required for proper function of the mitochondrial energy chain.4 Lipoic acid directly increases ATP production in mitochondria.79 Clinical models indicate that lipoic acid may serve as a first-line defense for diseases involving impaired energy utilization, including diabetes and the nerve damage associated with it.80-82

R-alpha-lipoic acid is the most bioactive form of lipoic acid—and a powerful activator of mitochondrial energy complexes.83,84 Studies in aging animals support the use of R-alpha-lipoic acid to improve mitochondrial function, decrease oxidative damage, and increase metabolic rate, all of which otherwise become impaired with aging.4

A Complementary Coenzyme

R-alpha-lipoic acid has been proven effective in reducing symptoms of diabetic neuropathy, without significant adverse reactions.81,85 It also increases nerve conduction velocity in people with diabetic neuropathy, crucial to improved nerve signaling.86 Experts attribute these effects to diminished fat oxidation in nerve cell membranes and improvements in local blood supply around nerves resulting from improved mitochondrial functioning.87,88

R-alpha-lipoic acid displays many protective effects. It reverses the age-related increase in liver cell damage caused by exogenous toxins, helping to protect liver function.89 It prevents brain cells from becoming depleted of the natural antioxidant reduced glutathione, an important intracellular antioxidant in the body. Deficiency of reduced glutathione can predispose people to liver failure, Parkinson’s disease, and other neurodegenerative conditions.90,91 A therapeutic dose of 600 mg/day even helped relieve migraine attack rates—an observation that may support the theory that migraines may be partially caused by impaired mitochondrial function.92

As you might expect of a mitochondrial energy booster, lipoic acid may also play a role in helping to ward off cardiovascular disease. Three months of lipoic acid supplementation provided pain relief to patients with peripheral vascular disease (PVD), extending the time they could walk before pain occurred.93 Combined therapy with acetyl-L-carnitine improved blood vessel relaxation and blood flow, while reducing blood pressure, in patients with coronary artery disease.94 And combined supplementation is a very good idea, as we’ll see next.

Your Mitochondrial Fat-Burner

L-carnitine is a molecule required for helping transport fatty acids into the mitochondria, where they can be burned as fuel. Acetyl-L-carnitine (ALC) is the form of carnitine optimally absorbed through oral delivery. It has also been shown to boost mitochondrial health, facilitating fuel delivery to the electron transport chain, where supplements like CoQ10, shilajit, and lipoic acid take over.

Your Mitochondrial Fat-Burner

Total carnitine levels diminish with age, a decline that may also be accelerated by overeating and diabetes.95 As with other mitochondrial energy optimizers, ALC supplementation possesses distinct benefits across numerous physiological systems.96

A review of clinical studies shows that ALC may slow the natural course of Alzheimer’s disease.97 It has substantially increased Alzheimer’s disease patients’ responses to drug treatment, from 38% to 50% in one study.98 ALC also protects brain tissue against destructive effects of hypoxia (low oxygen), by supporting cellular metabolism.99 ALC and lipoic acid supplementation partially restored depleted brain mitochondrial activity in aged rats to that of young adults.100

The combination of ALC with lipoic acid improved cognitive function in a mouse model of Alzheimer’s disease.101 ALC alone has exhibited powerful effects, restoring aging animals’ cardiac energy metabolism to that of young adults.102 In combination with lipoic acid, ALC helps maintain heart muscle function in aging animals as well.103

Summary

Mitochondrial dysfunction is linked to a broad range of degenerative illnesses, from diabetes and neurological disorders to heart disease. Researchers have discovered that age-related mitochondrial dysfunction—which can ultimately lead to DNA damage and cell death—may be prevented and even reversed. The key lies in early and sustained interventions that support optimal mitochondrial health and function. CoQ10 in its superior form as ubiquinol may restore mitochondrial function. The organic adaptogen shilajit acts in synergy with ubiquinol, further enhancing mitochondrial function. R-alpha-lipoic acid and acetyl-L-carnitine have been shown in clinical studies to provide additional mitochondrial support.

Be Proactive About Mitochondrial Function

Mitochondrial dysfunction, a decline in the health and function of mitochondria, is associated with degenerative diseases such as heart disease, neurological disorders and more. Nutrients such as coenzyme Q10, R-alpha-lipoic acid and adaptogens like shilajit can act as cellular energizers that can help restore and preserve mitochondrial function.

If you have any questions on the scientific content of this article, please call a Life Extension® Wellness Specialist at 1-866-864-3027.

References
  1. Conley KE, Amara CE, Jubrias SA, Marcinek DJ. Mitochondrial function, fibre types and ageing: new insights from human muscle in vivo. Exp Physiol. 2007 Mar;92(2):333-9.
  2. Lesnefsky EJ, Moghaddas S, Tandler B, Kerner J, Hoppel CL. Mitochondrial dysfunction in cardiac disease: ischemia—reperfusion, aging, and heart failure. J Mol Cell Cardiol. 2001 Jun;33(6):1065-89.
  3. Conley KE, Marcinek DJ, Villarin J. Mitochondrial dysfunction and age. Curr Opin Clin Nutr Metab Care. 2007 Nov;10(6):688-92.
  4. Hagen TM, Ingersoll RT, Lykkesfeldt J, et al. (R)-alpha-lipoic acid-supplemented old rats have improved mitochondrial function, decreased oxidative damage, and increased metabolic rate. FASEB J. 1999 Feb;13(2):411-8.
  5. Shigenaga MK, Hagen TM, Ames BN. Oxidative damage and mitochondrial decay in aging. Proc Natl Acad Sci U S A. 1994 Nov 8;91(23):10771-8.
  6. Genova ML, Pich MM, Bernacchia A, et al. The mitochondrial production of reactive oxygen species in relation to aging and pathology. Ann N Y Acad Sci. 2004 Apr;1011:86-100.
  7. Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging. Science. 1996 Jul 5;273(5271):59-63.
  8. Hagen TM, Yowe DL, Bartholomew JC, et al. Mitochondrial decay in hepatocytes from old rats: membrane potential declines, heterogeneity and oxidants increase. Proc Natl Acad Sci U S A. 1997 Apr 1;94(7):3064-9.
  9. Sanz N, Diez-Fernandez C, Alvarez A, Cascales M. Age-dependent modifications in rat hepatocyte antioxidant defense systems. J Hepatol. 1997 Sep;27(3):525-34.
  10. Erdincler DS, Seven A, Inci F, Beger T, Candan G. Lipid peroxidation and antioxidant status in experimental animals: effects of aging and hypercholesterolemic diet. Clin Chim Acta. 1997 Sep 8;265(1):77-84.
  11. DiMauro S, Tanji K, Bonilla E, Pallotti F, Schon EA. Mitochondrial abnormalities in muscle and other aging cells: classification, causes, and effects. Muscle Nerve. 2002 Nov;26(5):597-607.
  12. Sullivan PG, Brown MR. Mitochondrial aging and dysfunction in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2005 Mar;29(3):407-10.
  13. Choksi KB, Nuss JE, Boylston WH, Rabek JP, Papaconstantinou J. Age-related increases in oxidatively damaged proteins of mouse kidney mitochondrial electron transport chain complexes. Free Radic Biol Med. 2007 Nov 15;43(10):1423-38.
  14. Baines CP. The mitochondrial permeability transition pore and ischemia-reperfusion injury. Basic Res Cardiol. 2009 Mar;104(2):181-8.
  15. Di Lisa F, Kaludercic N, Carpi A, Menabo R, Giorgio M. Mitochondria and vascular pathology. Pharmacol Rep. 2009 Jan-Feb;61(1):123-30.
  16. Sohal RS, Forster MJ. Coenzyme Q, oxidative stress and aging. Mitochondrion. 2007 Jun;7 Suppl:S103-11.
  17. Aberg F, Appelkvist EL, Dallner G, Ernster L. Distribution and redox state of ubiquinones in rat and human tissues. Arch Biochem Biophys. 1992 Jun;295(2):230-4.
  18. Kalen A, Appelkvist EL, Dallner G. Age-related changes in the lipid compositions of rat and human tissues. Lipids.1989 Jul;24(7):579-84.
  19. Bliznakov EG. Immunological senescence in mice and its reversal by coenzyme Q10. Mech Ageing Dev. 1978 Mar;7(3):189-97.
  20. Rosenfeldt FL, Pepe S, Linnane A, et al. The effects of ageing on the response to cardiac surgery: protective strategies for the ageing myocardium. Biogerontology. 2002;3(1-2):37-40.
  21. Rosenfeldt FL, Pepe S, Linnane A, et al. Coenzyme Q10 protects the aging heart against stress: studies in rats, human tissues, and patients. Ann N Y Acad Sci. 2002 Apr;959:355-39; discussion 463-35.
  22. Aejmelaeus R, Metsa-Ketela T, Laippala P, Solakivi T, Alho H. Ubiquinol-10 and total peroxyl radical trapping capacity of LDL lipoproteins during aging: the effects of Q-10 supplementation. Mol Aspects Med. 1997;18 Suppl:S11320.
  23. Quiles JL, Ochoa JJ, Huertas JR, Mataix J. Coenzyme Q supplementation protects from age-related DNA double-strand breaks and increases lifespan in rats fed on a PUFA-rich diet. Exp Gerontol. 2004 Feb;39(2):189-94.
  24. Ochoa JJ, Quiles JL, Huertas JR, Mataix J. Coenzyme Q10 protects from aging-related oxidative stress and improves mitochondrial function in heart of rats fed a polyunsaturated fatty acid (PUFA)-rich diet. J Gerontol A Biol Sci Med Sci. 2005 Aug;60(8):970-5.
  25. Ochoa JJ, Quiles JL, Lopez-Frias M, Huertas JR, Mataix J. Effect of lifelong coenzyme Q10 supplementation on age-related oxidative stress and mitochondrial function in liver and skeletal muscle of rats fed on a polyunsaturated fatty acid (PUFA)-rich diet. J Gerontol A Biol Sci Med Sci. 2007 Nov;62(11):1211-8.
  26. Kaikkonen J, Tuomainen TP, Nyyssonen K, Salonen JT. Coenzyme Q10: absorption, antioxidative properties, determinants, and plasma levels. Free Radic Res. 2002 Apr;36(4):389-97.
  27. Hosoe K, Kitano M, Kishida H, Kubo H, Fujii K, Kitahara M. Study on safety and bioavailability of ubiquinol (Kaneka QH) after single and 4-week multiple oral administration to healthy volunteers. Regul Toxicol Pharmacol. 2007 Feb;47(1):19-28.
  28. Shults CW, Flint Beal M, Song D, Fontaine D. Pilot trial of high dosages of coenzyme Q10 in patients with Parkinson’s disease. Exp Neurol. 2004 Aug;188(2):491-4.
  29. Soukoulis V, Dihu JB, Sole M, et al. Micronutrient deficiencies an unmet need in heart failure. J Am Coll Cardiol. 2009 Oct 27;54(18):1660-73.
  30. Langsjoen PH, Langsjoen AM. Supplemental ubiquinol in patients with advanced congestive heart failure. Biofactors. 2008;32(1-4):119-28.
  31. Available at: http://www.abouthf.org/questions_stages.htm. Accessed October 9, 2009.
  32. Belardinelli R, Mucaj A, Lacalaprice F, et al. Coenzyme Q10 improves contractility of dysfunctional myocardium in chronic heart failure. Biofactors. 2005;25(1-4):137-45.
  33. Morisco C, Trimarco B, Condorelli M. Effect of coenzyme Q10 therapy in patients with congestive heart failure: a long-term multicenter randomized study. Clin Investig. 1993;71(8 Suppl):S134-6.
  34. Jeejeebhoy F, Keith M, Freeman M, et al. Nutritional supplementation with MyoVive repletes essential cardiac myocyte nutrients and reduces left ventricular size in patients with left ventricular dysfunction. Am Heart J. 2002 Jun;143(6):1092-100.
  35. Singh RB, Neki NS, Kartikey K, et al. Effect of coenzyme Q10 on risk of atherosclerosis in patients with recent myocardial infarction. Mol Cell Biochem. 2003 Apr;246(1-2):75-82.
  36. Makhija N, Sendasgupta C, Kiran U, et al. The role of oral coenzyme Q10 in patients undergoing coronary artery bypass graft surgery. J Cardiothorac Vasc Anesth. 2008 Dec;22(6):832-9.
  37. Kuettner A, Pieper A, Koch J, Enzmann F, Schroeder S. Influence of coenzyme Q(10) and cerivastatin on the flow-mediated vasodilation of the brachial artery: results of the ENDOTACT study. Int J Cardiol. 2005 Feb 28;98(3):413-9.
  38. Graham D, Huynh NN, Hamilton CA, et al. Mitochondria-targeted antioxidant MitoQ10 improves endothelial function and attenuates cardiac hypertrophy. Hypertension. Aug 2009;54(2):322-8.
  39. Tiano L, Belardinelli R, Carnevali P, Principi F, Seddaiu G, Littarru GP. Effect of coenzyme Q10 administration on endothelial function and extracellular superoxide dismutase in patients with ischaemic heart disease: a double-blind, randomized controlled study. Eur Heart J. 2007 Sep;28(18):2249-55.
  40. Beckman JA, Creager MA, Libby P. Diabetes and atherosclerosis: epidemiology, pathophysiology, and management. JAMA. 2002 May 15;287(19):2570-81.
  41. Watts GF, Playford DA, Croft KD, Ward NC, Mori TA, Burke V. Coenzyme Q(10) improves endothelial dysfunction of the brachial artery in Type II diabetes mellitus. Diabetologia. 2002 Mar;45(3):420-6.
  42. Marcoff L, Thompson PD. The role of coenzyme Q10 in statin-associated myopathy: a systematic review. J Am Coll Cardiol. 2007 Jun 12;49(23):2231-7.
  43. Hamilton SJ, Chew GT, Watts GF. Coenzyme Q10 improves endothelial dysfunction in statin-treated type 2 diabetic patients. Diabetes Care. 2009 May;32(5):810-2.
  44. Di Meo S, Venditti P. Mitochondria in exercise-induced oxidative stress. Biol Signals Recept. 2001 Jan-Apr;10(1-2):125-40.
  45. Powers SK, Jackson MJ. Exercise-induced oxidative stress: cellular mechanisms and impact on muscle force production. Physiol Rev. 2008 Oct;88(4):1243-76.
  46. Yegutkin GG, Samburski SS, Mortensen SP, Jalkanen S, Gonzalez-Alonso J. Intravascular ADP and soluble nucleotidases contribute to acute prothrombotic state during vigorous exercise in humans. J Physiol. 2007 Mar 1;579(Pt 2):553-64.
  47. Hellsten Y, Nielsen JJ, Lykkesfeldt J, et al. Antioxidant supplementation enhances the exercise-induced increase in mitochondrial uncoupling protein 3 and endothelial nitric oxide synthase mRNA content in human skeletal muscle. Free Radic Biol Med. 2007 Aug 1;43(3):353-61.
  48. Cooke M, Iosia M, Buford T, et al. Effects of acute and 14-day coenzyme Q10 supplementation on exercise performance in both trained and untrained individuals. J Int Soc Sports Nutr.2008;5:8.
  49. Kon M, Tanabe K, Akimoto T, et al. Reducing exercise-induced muscular injury in kendo athletes with supplementation of coenzyme Q10. Br J Nutr. 2008 Oct;100(4):903-9.
  50. Gokbel H, Gul I, Belviranl M, Okudan N. The Effects Of Coenzyme Q10 Supplementation on Performance During Repeated Bouts of Supramaximal Exercise in Sedentary Men. J Strength Cond Res. 2009 Jul 28.
  51. Mizuno K, Tanaka M, Nozaki S, et al. Antifatigue effects of coenzyme Q10 during physical fatigue. Nutrition. 2008 Apr;24(4):293-9.
  52. Langsjoen PH, Langsjoen JO, Langsjoen AM, Lucas LA. Treatment of statin adverse effects with supplemental Coenzyme Q10 and statin drug discontinuation. Biofactors. 2005;25(1-4):147-52.
  53. Kidd PM. Neurodegeneration from mitochondrial insufficiency: nutrients, stem cells, growth factors, and prospects for brain rebuilding using integrative management. Altern Med Rev. 2005 Dec;10(4):268-93.
  54. Matthews RT, Yang L, Browne S, Baik M, Beal MF. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci U S A. 1998 Jul 21;95(15):8892-7.
  55. Ayaz M, Tuncer S, Okudan N, Gokbel H. Coenzyme Q(10) and alpha-lipoic acid supplementation in diabetic rats: conduction velocity distributions. Methods Find Exp Clin Pharmacol. 2008 Jun;30(5):367-74.
  56. Russo R, Cavaliere F, Rombola L, et al. Rational basis for the development of coenzyme Q10 as a neurotherapeutic agent for retinal protection. Prog Brain Res. 2008;173:575-82.
  57. Feher J, Papale A, Mannino G, Gualdi L, Balacco Gabrieli C. Mitotropic compounds for the treatment of age-related macular degeneration. The metabolic approach and a pilot study. Ophthalmologica. 2003 Sep-Oct;217(5):351-7.
  58. Qu J, Kaufman Y, Washington I. Coenzyme Q10 in the human retina. Invest Ophthalmol Vis Sci. 2009 Apr;50(4):1814-8.
  59. Feher J, Kovacs B, Kovacs I, et al. Improvement of visual functions and fundus alterations in early age-related macular degeneration treated with a combination of acetyl-L-carnitine, n-3 fatty acids, and coenzyme Q10. Ophthalmologica. 2005 May-Jun;219(3):154-66.
  60. Prahl S, Kueper T, Biernoth T, et al. Aging skin is functionally anaerobic: importance of coenzyme Q10 for anti aging skin care. Biofactors. 2008;32(1-4):245-55.
  61. Inui M, Ooe M, Fujii K, Matsunaka H, Yoshida M, Ichihashi M. Mechanisms of inhibitory effects of CoQ10 on UVB-induced wrinkle formation in vitro and in vivo. Biofactors. 2008;32(1-4):237-43.
  62. Hoppe U, Bergemann J, Diembeck W, et al. Coenzyme Q10, a cutaneous antioxidant and energizer. Biofactors. 1999;9(2-4):371-8.
  63. Blatt T, Lenz H, Koop U, et al. Stimulation of skin’s energy metabolism provides multiple benefits for mature human skin. Biofactors. 2005;25(1-4):179-85.
  64. Passi S, De Pità O, Grandinetti M, Simotti C, Littarru GP. The combined use of oral and topical lipophilic antioxidants increases their levels both in sebum and stratum corneum. Biofactors. 2003;18(1-4):289-97.
  65. Schepetkin IA, Xie G, Jutila MA, Quinn MT. Complement-fixing activity of fulvic acid from Shilajit and other natural sources. Phytother Res. 2009 Mar;23(3):373-84.
  66. Goel RK, Banerjee RS, Acharya SB. Antiulcerogenic and antiinflammatory studies with shilajit. J Ethnopharmacol. 1990 Apr;29(1):95-103.
  67. Agarwal SP, Khanna R, Karmarkar R, Anwer MK, Khar RK. Shilajit: a review. Phytother Res. 2007 May;21(5):401-5.
  68. Bhattacharyya S, Pal D, Gupta AK, Ganguly P, Majumder UK, Ghosal S. Beneficial effect of processed shilajit on swimming exercise induced impaired energy status of mice. Pharmacologyonline. 2009;1:817-25.
  69. Piotrowska D, Dlugosz A, Witkiewicz K, Pajak J. The research on antioxidative properties of TOLPA Peat Preparation and its fractions. Acta Pol Pharm. 2000 Nov;57 Suppl:127-9.
  70. Ghosal S. Shilajit in Perspective. Oxford, U.K.: Narosa Publishing House; 2006.
  71. Visser SA. Effect of humic substances on mitochondrial respiration and oxidative phosphorylation. Sci Total Environ. 1987 Apr;62:347-54.
  72. Royer RA, Burgos WD, Fisher AS, Unz RF, Dempsey BA. Enhancement of biological reduction of hematite by electron shuttling and Fe(II) complexation. Environ Sci Technol. 2002 May 1;36(9):1939-46.
  73. Kang SH, Choi W. Oxidative degradation of organic compounds using zero-valent iron in the presence of natural organic matter serving as an electron shuttle. Environ Sci Technol. 2009 Feb 1;43(3):878-83.
  74. Islam A, Ghosh R, Banerjee D, Nath P, Mazumder U, Ghosal S. Biotransformation of 3-hydroxydibenzo—pyrone into 3,8 dihydroxydibenzo—pyrone and aminoacyl conjugates by Aspergillus niger isolated from native“shilajit.” Electronic Journal of Biotechnology. 2008 Jul 15;11(3):2-10.
  75. Bhattacharyya S, Pal D, Banerjee D, et al. Shilajit dibenzo—pyrones: Mitochondria targeted antioxidants. Pharmacologyonline. 2009; 2:690-8.
  76. Pal D, Bhattacharya S. Pilot Study on the Improvement of Human Performance with ReVitalETTM as Energy Booster: Part-IV. 2006. Data on file. Natreon, Inc.
  77. Clinical study for evaluation of safe use in purified and standardized shilajit in normal volunteers. J. B. Roy State Ayurvedic Medical College and Hospital, Kolkata. 2007. Data on file. Natreon, Inc.
  78. Clinical study for evaluation of plasma antioxidant capacity and safe use of purified and standardized Shilajit (ReVitalET) in normal volunteers. J. B. Roy State Ayurvedic Medical College and Hospital, Kolkata. 2007. Data on file. Natreon, Inc.
  79. Zimmer G, Mainka L, Kruger E. Dihydrolipoic acid activates oligomycin-sensitive thiol groups and increases ATP synthesis in mitochondria. Arch Biochem Biophys. 1991 Aug 1;288(2):609-13.
  80. Jacob S, Henriksen EJ, Schiemann AL, et al. Enhancement of glucose disposal in patients with type 2 diabetes by alpha-lipoic acid. Arzneimittelforschung. 1995 Aug;45(8):872-4.
  81. Ziegler D, Hanefeld M, Ruhnau KJ, et al. Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia. 1995 Dec;38(12):1425-33.
  82. Sachse G, Willms B. Efficacy of thioctic acid in the therapy of peripheral diabetic neuropathy. Horm Metab Res Suppl. 1980;9:105-7.
  83. Loffelhardt S, Bonaventura C, Locher M, Borbe HO, Bisswanger H. Interaction of alpha-lipoic acid enantiomers and homologues with the enzyme components of the mammalian pyruvate dehydrogenase complex. Biochem Pharmacol. 1995 Aug 25;50(5):637-46.
  84. Carlson DA, Smith AR, Fischer SJ, Young KL, Packer L. The plasma pharmacokinetics of R-(+)-lipoic acid administered as sodium R-(+)-lipoate to healthy human subjects. Altern Med Rev. 2007 Dec;12(4):343-51.
  85. Tankova T, Koev D, Dakovska L. Alpha-lipoic acid in the treatment of autonomic diabetic neuropathy (controlled, randomized, open-label study). Rom J Intern Med. 2004;42(2):457-64.
  86. Negrisanu G, Rosu M, Bolte B, Lefter D, Dabelea D. Effects of 3-month treatment with the antioxidant alpha-lipoic acid in diabetic peripheral neuropathy. Rom J Intern Med. 1999 Jul-Sep;37(3):297-306.
  87. Androne L, Gavan NA, Veresiu IA, Orasan R. In vivo effect of lipoic acid on lipid peroxidation in patients with diabetic neuropathy. In Vivo. 2000 Mar-Apr;14(2):327-30.
  88. Haak E, Usadel KH, Kusterer K, et al. Effects of alpha-lipoic acid on microcirculation in patients with peripheral diabetic neuropathy. Exp Clin Endocrinol Diabetes. 2000;108(3):168-74.
  89. Hagen TM, Vinarsky V, Wehr CM, Ames BN. (R)-alpha-lipoic acid reverses the age-associated increase in susceptibility of hepatocytes to tert-butylhydroperoxide both in vitro and in vivo. Antioxid Redox Signal. 2000 Fall;2(3):473-83.
  90. Bharat S, Cochran BC, Hsu M, Liu J, Ames BN, Andersen JK. Pre-treatment with R-lipoic acid alleviates the effects of GSH depletion in PC12 cells: implications for Parkinson’s disease therapy. Neurotoxicology. 2002 Oct;23(4-5):479-86.
  91. Suh JH, Wang H, Liu RM, Liu J, Hagen TM. (R)-alpha-lipoic acid reverses the age-related loss in GSH redox status in post-mitotic tissues: evidence for increased cysteine requirement for GSH synthesis. Arch Biochem Biophys. 2004 Mar 1;423(1):126-35.
  92. Magis D, Ambrosini A, Sandor P, Jacquy J, Laloux P, Schoenen J. A randomized double-blind placebo-controlled trial of thioctic acid in migraine prophylaxis. Headache. 2007 Jan;47(1):52-7.
  93. Vincent HK, Bourguignon CM, Vincent KR, Taylor AG. Effects of alpha-lipoic acid supplementation in peripheral arterial disease: a pilot study. J Altern Complement Med. 2007 Jun;13(5):577-84.
  94. McMackin CJ, Widlansky ME, Hamburg NM, et al. Effect of combined treatment with alpha-Lipoic acid and acetyl-L-carnitine on vascular function and blood pressure in patients with coronary artery disease. J Clin Hypertens (Greenwich). 2007 Apr;9(4):249-55.
  95. Noland RC, Koves TR, Seiler SE, et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009 Aug 21;284(34):22840-52.
  96. Rosca MG, Lemieux H, Hoppel CL. Mitochondria in the elderly: Is acetylcarnitine a rejuvenator? Adv Drug Deliv Rev. 2009 Aug 29.
  97. Carta A, Calvani M. Acetyl-L-carnitine: a drug able to slow the progress of Alzheimer’s disease? Ann N Y Acad Sci. 1991;640:228-32.
  98. Bianchetti A, Rozzini R, Trabucchi M. Effects of acetyl-L-carnitine in Alzheimer’s disease patients unresponsive to acetylcholinesterase inhibitors. Curr Med Res Opin. 2003;19(4):350-3.
  99. Corbucci GG, Melis A, Piga M, Marchionni A, Calvani M. Influence of acetyl-carnitine on some mitochondrial enzymic activities in the human cerebral tissue in conditions of acute hypoxia. Int J Tissue React. 1992;14(4):183-94.
  100. Long J, Gao F, Tong L, Cotman CW, Ames BN, Liu J. Mitochondrial decay in the brains of old rats: ameliorating effect of alpha-lipoic acid and acetyl-L-carnitine. Neurochem Res. Apr 2009 Apr;34(4):755-63.
  101. Shenk JC, Liu J, Fischbach K, et al. The effect of acetyl-L-carnitine and R-alpha-lipoic acid treatment in ApoE4 mouse as a model of human Alzheimer’s disease. J Neurol Sci. 2009 Aug 15;283(1-2):199-206.
  102. Lesnefsky EJ, He D, Moghaddas S, Hoppel CL. Reversal of mitochondrial defects before ischemia protects the aged heart. FASEB J. 2006 Jul;20(9):1543-5.
  103. Hagen TM, Moreau R, Suh JH, Visioli F. Mitochondrial decay in the aging rat heart: evidence for improvement by dietary supplementation with acetyl-L-carnitine and/or lipoic acid. Ann N Y Acad Sci. 2002 Apr;959:491-507.
  104. Rahbar S. Novel inhibitors of glycation and AGE formation. Cell Biochem Biophys. 2007;48(2-3):147-57.
  105. Adeghate E. Molecular and cellular basis of the aetiology and management of diabetic cardiomyopathy: a short review. Mol Cell Biochem. 2004 Jun;261(1-2):187-91.
  106. Schleicher E, Friess U. Oxidative stress, AGE, and atherosclerosis. Kidney Int Suppl. 2007 Aug;(106):S17-26.
  107. Tan AL, Forbes JM, Cooper ME. AGE, RAGE, and ROS in diabetic nephropathy. Semin Nephrol. 2007 Mar;27(2):130-43.
  108. Gasser A, Forbes JM. Advanced glycation: implications in tissue damage and disease. Protein Pept Lett. 2008;15(4):385-91.
  109. Peppa M, Uribarri J, Vlassara H. Aging and glycoxidant stress. Hormones (Athens). 2008 Apr-Jun;7(2):123-32.
  110. Reddy VP, Garrett MR, Perry G, Smith MA. Carnosine: a versatile antioxidant and antiglycating agent. Sci Aging Knowledge Environ. 2005 May 4;2005(18):pe12.
  111. Hipkiss AR. Would carnosine or a carnivorous diet help suppress aging and associated pathologies? Ann N Y Acad Sci. 2006 May;1067:369-74.
  112. Hipkiss AR. Could carnosine or related structures suppress Alzheimer’s disease? J Alzheimers Dis. 2007 May;11(2):229-40.
  113. Kotanidou A, Xagorari A, Bagli E, et al. Luteolin reduces lipopolysaccharide-induced lethal toxicity and expression of proinflammatory molecules in mice. Am J Respir Crit Care Med. 2002 Mar 15;165(6):818-23.
  114. Kim JS, Jobin C. The flavonoid luteolin prevents lipopolysaccharide-induced NF-kappaB signalling and gene expression by blocking IkappaB kinase activity in intestinal epithelial cells and bone-marrow derived dendritic cells. Immunology. 2005 Jul;115(3):375-87.
  115. Wu CH, Yen GC. Inhibitory effect of naturally occurring flavonoids on the formation of advanced glycation endproducts. J Agric Food Chem. 2005 Apr 20;53(8):3167-73.
  116. Psotova J, Chlopcikova S, Miketova P, Hrbac J, Simanek V. Chemoprotective effect of plant phenolics against anthracycline-induced toxicity on rat cardiomyocytes. Part III. Apigenin, baicalelin, kaempherol, luteolin and quercetin. Phytother Res. 2004 Jul;18(7):516-21.
  117. Wu CH, Wu CF, Huang HW, Jao YC, Yen GC. Naturally occurring flavonoids attenuate high glucose-induced expression of proinflammatory cytokines in human monocytic THP-1 cells. Mol Nutr Food Res. 2009 Aug;53(8):984-95.
  118. Volvert ML, Seyen S, Piette M, et al. Benfotiamine, a synthetic S-acyl thiamine derivative, has different mechanisms of action and a different pharmacological profile than lipid-soluble thiamine disulfide derivatives. BMC Pharmacol. 2008;8:10.
  119. Du X, Edelstein D, Brownlee M. Oral benfotiamine plus alpha-lipoic acid normalises complication-causing pathways in type 1 diabetes. Diabetologia. 2008 Oct;51(10):1930-2.
  120. Stirban A, Negrean M, Stratmann B, et al. Benfotiamine prevents macro- and microvascular endothelial dysfunction and oxidative stress following a meal rich in advanced glycation end products in individuals with type 2 diabetes. Diabetes Care. 2006 Sep;29(9):2064-71.
  121. Pomero F, Molinar Min A, La Selva M, Allione A, Molinatti GM, Porta M. Benfotiamine is similar to thiamine in correcting endothelial cell defects induced by high glucose. Acta Diabetol. 2001;38(3):135-8.
  122. Marchetti V, Menghini R, Rizza S, et al. Benfotiamine counteracts glucose toxicity effects on endothelial progenitor cell differentiation via Akt/FoxO signaling. Diabetes. 2006 Aug;55(8):2231-7.
  123. Hammes HP, Du X, Edelstein D, et al. Benfotiamine blocks three major pathways of hyperglycemic damage and prevents experimental diabetic retinopathy. Nat Med. 2003 Mar;9(3):294-9.
  124. Khatami M, Suldan Z, David I, Li W, Rockey JH. Inhibitory effects of pyridoxal phosphate, ascorbate and aminoguanidine on nonenzymatic glycosylation. Life Sci.1988;43(21):1725-31.
  125. Higuchi O, Nakagawa K, Tsuzuki T, Suzuki T, Oikawa S, Miyazawa T. Aminophospholipid glycation and its inhibitor screening system: a new role of pyridoxal 5’-phosphate as the inhibitor. J Lipid Res. 2006 May;47(5):964-74.
  126. Nakamura S, Niwa T. Pyridoxal phosphate and hepatocyte growth factor prevent dialysate-induced peritoneal damage. J Am Soc Nephrol. 2005 Jan;16(1):144-50.
  127. Nakamura S, Li H, Adijiang A, Pischetsrieder M, Niwa T. Pyridoxal phosphate prevents progression of diabetic nephropathy. Nephrol Dial Transplant. 2007Aug;22(8):2165-74.
  128. Mehta R, Shangari N, O’Brien PJ. Preventing cell death induced by carbonyl stress, oxidative stress or mitochondrial toxins with vitamin B anti-AGE agents. Mol Nutr Food Res. 2008 Mar;52(3):379-85.