Life Extension Magazine®

Cross section of healthy kidneys before kidney disease

Innovative Strategies to Combat Kidney Disease

Researchers have only recently uncovered the epidemic of chronic kidney disease striking aging Americans. In some cases, up to 60% of kidney function may be lost before standard testing reveals a problem. The encouraging news is that there are proven methods to diagnose kidney disease early, and natural interventions that protect against this widespread killer.

Scientifically reviewed by: Dr. Gary Gonzalez, MD, in August 2023. Written by: Julius Goepp, MD.

Innovative Strategies to Combat Kidney Disease

You may be surprised to learn that until 2002, no standard definition for chronic kidney disease (CKD) existed within the medical community.1 Before then, conflicting classifications had created a state of confusion as to how many Americans were afflicted with this progressive, life-threatening condition.

Once proper categorization of the various phases of CKD was established, the stark and daunting scale of this modern epidemic emerged.

We now know that as many as 26 million Americans currently suffer from some form of chronic kidney disease.2,3 Aging individuals are especially vulnerable.4

Yet public awareness of the threat remains low. When you consider that the risk of cardiovascular mortality in CKD sufferers is 30 times that of the general population,5 the steady increase in kidney disease rates seen today amounts to a public health disaster hidden in plain sight.

What you need to know

  • It is imperative that aging individuals receive regular blood tests to monitor kidney health. In addition to standard creatinine, albumin, and BUN/creatinine ratio testing, cystatin-C levels should also be measured, as this constitutes a far more accurate biomarker of renal function.
  • The high-pressure and toxin-rich environment involved in renal function renders these delicate, highly complex organs especially vulnerable to damage, dysfunction, and disease.
  • High blood pressure, elevated blood sugar¸ NSAIDs, certain medications, and high-protein diets are the most common threats to kidney health.
  • Nutrients such as pyridoxal-5-phosphate (P5P) fight AGEs and ALEs.
  • CoQ10, silymarin, resveratrol, and lipoic acid are also clinically supported, potent interventions.
  • Omega-3 fatty acids help quell inflammation, contributing to enhanced kidney health.
  • A host of additional nutrients complement these actions, including folic acid (folate) and vitamins C and E.

Life Extension® has long emphasized the need for vigilance through regular testing (at least once a year) to monitor kidney health. In addition to the standard tests for creatinine, albumin, and BUN/creatinine ratio, certain individuals should insist their doctor test for cystatin-C, a largely overlooked blood marker which provides a far more precise measure of renal function. Optimal levels are less than .91 mg/L.

Individuals should also keep a record of their test results. Once any sign of disease is detected (such as an increase in creatinine), it is imperative that immediate steps be taken to halt its progress, as kidney function can decline precipitously and may be irreversible. Fortunately, many Life Extension® members are already taking a variety of nutrients that support kidney health.

Pyridoxamine or Pyridoxal-5-Phosphate: Potent Kidney Defense

In this article, you will discover the most recent scientific advances in our understanding of how CKD unfolds, the specific risk factors that contribute to its progress, and how you can bring them under control.

You will also learn of safe, low-cost, natural interventions that have been shown to stop CKD in its tracks, long before end-stage renal disease (ESRD) renders dialysis or kidney transplant the only option.

Pyridoxamine or Pyridoxal-5-Phosphate: Potent Kidney Defense

Since the formation of advanced glycation end-products (AGEs) is such a well-established factor in the onset and progression of kidney disease, nutrients that have been conclusively shown to mitigate the effects of these lethal agents constitute a front line, low-cost intervention.

A formidable AGE antagonist is the vitamin B6 compound pyridoxamine. A plethora of research confirms its power to halt formation of AGEs.6-8 Evidence has also emerged that pyridoxamine drastically limits formation of equally deadly advanced lipoxidation end products (ALEs)—another deadly catalyst for kidney disease.9-11

A team of biochemists at the University of South Carolina were able to show that pyridoxamine traps the reactive molecules formed during lipid (fat) peroxidation and helps to “chaperone” them harmlessly into the urine.10,12

Their colleagues subsequently found that neutralizing AGEs and ALEs can prevent kidney disease and lipid profile abnormalities in diabetic rats.13 They found that rats supplemented with pyridoxamine had lower levels of albumin (protein) in their urine, lower plasma levels of the waste product creatinine, and less dramatically elevated blood lipids than the placebo treated animals, all directly related to the reduction of AGE/ALEs.

They subsequently examined whether similar results could be obtained in obese animals that had not yet developed diabetes.14 Three groups of animals were studied:

  1. lean (healthy) rats
  2. obese rats without treatment
  3. obese rats treated with pyridoxamine.
Four Complementary Kidney Protectors

As expected, AGE and ALE formation underwent a two- to threefold increase in obese untreated rats compared to lean animals. Conversely, those increases were entirely absent in obese animals treated with pyridoxamine. Treated animals also experienced a smaller increase in plasma triglycerides, cholesterol, and creatinine levels, compared with the obese untreated rats.14

In an equally compelling development, hypertension in animals treated with pyridoxamine also resolved, as did thickening of blood vessel walls. Untreated animals displayed urinary evidence of renal disease (albuminuria) that in contrast had been nearly normalized in supplemented animals. This provides powerful evidence of pyridoxamine’s multi-targeted protective effect against CKD.14

In 2004, the same research team made a landmark discovery: while studying the relative effects of pyridoxamine along with a variety of additional natural antioxidants on the progression of kidney disease in diabetic rats, they decided to examine how these natural compounds stacked up against enalapril, a standard pharmaceutical intervention used to prevent CKD.9 Enalapril is an ACE inhibitor, one of a class of drugs commonly used to control blood pressure and kidney disease.

They found that pyridoxamine therapy was the most effective at preventing progression of kidney disease, followed by vitamin E and lipoic acid. Enalapril, the prescription drug, proved to be the least effective intervention. Pyridoxamine also limited lipid profile abnormalities and formation of AGEs and ALEs, offering a far broader spectrum of preventive effects than enalapril.9

Researchers at the University of Miami advanced these findings by treating diabetic mice with both pyridoxamine and enalapril.15 Again they found that pyridoxamine alone provided substantial benefit, cutting albuminuria and damage to the glomeruli. Combining enalapril with pyridoxamine reduced kidney disease mortality in these animals as well, leading the researchers to suggest that the ACE-inhibitor (enalapril)/pyridoxamine combination might be useful.

A convincing body of research on pyridoxamine therapy in humans with CKD has also emerged in recent years. In 2007, a team of researchers at Harvard set out to determine optimal interventions to halt the progression of kidney disease in diabetics.16 They conducted two 24-week multicenter placebo-controlled trials in patients with known diabetic nephropathy—treatment of which is known to delay the onset of end-stage renal disease in diabetics. Doses of pyridoxamine ranged from 50 to 250 mg twice daily.

Pyridoxamine significantly inhibited the rise in blood levels of the waste product creatinine, one of the key biomarkers of kidney dysfunction and a predictor of kidney failure. Urinary levels of inflammatory cytokines were also significantly lower in the treated group compared to controls.

Pyridoxamine has been firmly established as a front line, safe, low-cost intervention in CKD caused or exacerbated by AGEs and ALEs. Further, this natural vitamin B6 compound has been shown to significantly improve outcomes of experimental kidney transplants and other forms of kidney disease.17-19

It therefore borders on the criminal that in January of 2009, the FDA classified this potent, entirely safe CKD therapeutic as a drug, putting it out of reach for many Americans suffering from this deadly condition. No one should be forced to bear the outrageous burden of costly pharmaceuticals and their toxic side effects when a perfectly safe alternative exists.

Fortunately, there is another equally safe option available—another form of vitamin B6 known as pyridoxal-5-phosphate (P5P) that also exerts potent anti-AGE effects. It has been shown to prevent the progression of diabetic kidney disease in pre-clinical models.20 In fact, as far back as 1988, P5P was used by a German research group to reduce blood lipids in humans with chronic kidney disease.21

Four Complementary Kidney Protectors

Coenzyme Q10

Because of the tremendous blood flow and high concentration of metabolic toxins continuously circulating through the kidneys, they are the site of extraordinary oxidative stress, which is known to contribute to progressive kidney damage and its complications, such as high LDL and increased cardiovascular disease risk.22

Coenzyme Q10 (CoQ10) fortifies the body’s natural antioxidant capacity and reduces levels of oxygen free radicals, indicating its important defense against CKD. As it happens, CoQ10 has been used experimentally to control hypertension and kidney disease in laboratory animals since the early 1970s.23,24

Human studies have shown that CoQ10 levels substantially decline, while markers of oxidation such as malondialdehyde are dramatically elevated, in kidney disease patients with even mild renal dysfunction.41 These decreased CoQ10 levels also make circulating lipoproteins (such as LDL) more vulnerable to oxidative damage, which in turn increases risk for further cardiovascular damage, adding to the renal burden and substantially increasing the risk of kidney disease.25

A team of European researchers published compelling evidence in 2001 of how effective such a nutritional intervention can be, studying a group of patients with established kidney disease.26 Subjects received antioxidant therapy with vitamin C, E, and riboflavin (vitamin B2) for one month before the addition of 2 months of CoQ10 therapy. Prior to supplementation, CoQ10 values in blood were just one-quarter of normal levels; they increased to nearly four times the reference level following supplementation. The study was too brief to demonstrate any change in kidney function, but evidence from animal trials that same year showed that when CoQ10 levels were increased in tissues of diabetic rats, a reversal of markers of oxidative stress in kidney, heart, and liver resulted.27

By 2004, definitive demonstration of CoQ10 in human kidney disease patients was demonstrated by researchers working with transplant recipients.28 Such individuals undergo tremendous oxidative stress and typically have marked disturbances in lipid profiles as a result. The European group provided their patients with CoQ10 supplements of 30 mg three times per day for four weeks, and monitored levels of oxidation factors (such as malondialdehyde), levels of natural antioxidant enzymes in the body, and lipid profiles.28

Significant improvements were seen after just four weeks, with reduction in LDL, increase in beneficial HDL, and a decrease in presence of inflammatory cells. These results suggest a potentially dramatic improvement in both quality of life and survival rates for patients whose disease has progressed to the point of kidney failure requiring transplantation or dialysis. They also bode well for those with early-stage kidney disease.

Animal studies have also shown that CoQ10 can protect kidney tissue from numerous nephrotoxic drugs, including gentamicin, a powerful antibiotic with a notorious propensity for causing kidney damage.33,34 These findings are significant both because they offer protection in patients who might be exposed to such drugs, and because of what they teach us about CoQ10’s potent ability to combat the extreme oxidant stress that the kidney faces as it deals with a variety of foreign chemicals.

Understanding Kidney Disease

The kidney ranks among the most complex and delicately evolved of all the major organs, making it particularly vulnerable to damage and dysfunction. As the body’s primary filtration system, it must “process” roughly 200 quarts of blood per day, rendering about 2 quarts of waste products and water.29

The fundamental structural unit of the kidney is the nephron. These high-pressure filtering mechanisms govern the removal of waste products and toxins, control blood pressure and volume, and regulate levels of electrolytes and metabolites in the blood. A healthy kidney contains approximately 800,000 to 1 million nephrons.

Housed within each nephron is a front-line filtration element called the glomerulus, a miniscule capillary coil. (The two together resemble an incandescent light bulb containing a convoluted filament.) The endothelial cells of the glomerulic capillaries act as the direct physical exchange between the kidney and the bloodstream. Waste products and water are combined to form urine, while blood cells and protein remain in the circulatory system.

The kidney’s tight control of water and mineral flow, and its role in maintaining healthy blood pressure and mineral balance, rely on the optimal functioning of nephrons and glomeruli. For this reason, one of the primary markers of kidney function is the glomerular filtration rate (GFR), a measure of the volume of fluid the kidney is able to process at any given time.

The glomerular filtration rate; plasma concentrations of the waste substances creatinine, urea, and nitrogen (blood urea nitrogen or BUN); and levels of protein in the blood and urine are the most commonly used measures to determine the presence of CKD. Rapidly rising creatinine usually signals imminent kidney failure. There should be no protein in the urine if your kidneys are functioning optimally.

It should be noted that BUN and creatinine may not increase above the normal range until 60% of total kidney function is lost. This is why certain aging individuals should ask their doctors to test for cystatin-C in the blood. Cystatin-C is a protein produced by virtually all cells and tissues in the body. Because it is formed freely and at a near-constant rate—as opposed to albumin, which may fluctuate with dietary protein intake—plasma cystatin-C serves as a more accurate biomarker of renal function.30

CKD may be categorized in one of 5 stages. Stage 1, the mildest, is defined only by the persistent presence of protein in urine (GFR may be normal); in each successively higher stage, GFR declines, until Stage 5 is reached, defining end-stage renal disease (ESRD), or kidney failure.31 ESRD is irreversible and results in death without dialysis or kidney transplant.32

silymarin complex

Silymarin

Silymarin is extracted from milk thistle (Silybum marianum), a plant rich in the flavonolignans silychristin, silydianin, silybin A, silybin B, isosilybin A and isosilybin B, which are collectively known as the silymarin complex.

This safe, natural compound has a long history as a traditional therapy for liver and kidney conditions.35,36 It has been used in Western medical practice for more than a quarter of a century as the treatment of choice for the serious kidney injury resulting from severe mushroom poisoning, owing to its potent antioxidant and nephron-protective effects.37 In fact, we’ve known since 1979 that kidney injury by mushroom poisoning in animals pre-treated with silymarin can be almost entirely prevented.38 These effects make it a natural choice for protection against drug-induced kidney damage, since so many drugs can act like poisons, exerting extreme oxidant stress on kidney tissue.

Mushroom poisons (mycotoxins) are among the most deadly natural toxins known. Their kidney toxicity is surpassed only by some of the most aggressive chemotherapy agents. Physicians have therefore looked to silymarin as a potential “renoprotective” agent for patients undergoing chemotherapy.

Silymarin is also protective against several classes of nephrotoxic drugs, in particular cisplatin and Adriamycin®, two of the most potent chemotherapeutic drugs—but also two of the most damaging to the kidney owing to oxidative damage and severe inflammation.39-41 Researchers around the world have found that silymarin and its components reduce and often entirely prevent the kidney damage caused by these drugs.42-45

Silymarin’s ability to protect against the oxidative stress produced by potent drugs suggests that it may be useful in protecting against more subtle, chronic injury by free radicals, particularly those generated by chronic blood glucose elevations. German researchers, for instance, have found that silymarin could entirely prevent injury to renal cells incubated with elevated glucose concentrations while blocking production of oxidative stress markers.46

Silymarin’s protective power also extends to ischemia/reperfusion injury (restoration of blood supply following restriction of blood flow). Turkish researchers demonstrated that they could completely prevent visible and functional damage to kidney structures exposed to this kind of injury by pre-treating animals with silymarin.47,48 Studies such as these have huge implications for the general population, because they suggest that by maintaining optimal antioxidant function through supplementation, we may be able to prevent much (if not most) of the chronic oxidative damage to which our kidneys are exposed daily.

Resveratrol

Resveratrol

The considerable advance in our understanding of the cyclical relationships between oxidative stress, endothelial dysfunction, inflammation, atherosclerosis, and chronic kidney disease points to resveratrol as an intervention in the chain of events that ultimately lead to renal failure.49

Italian researchers are among the leaders in resveratrol research, and early in this century one group published remarkable research demonstrating the impact of resveratrol on preserving kidney structure and function in rats exposed to ischemia/reperfusion injury.50,51

Japanese and Indian urologists followed that up in 2005 and 2006 with reports detailing the mechanisms by which resveratrol combats oxidative damage following reperfusion, markedly reducing kidney dysfunction.52-56 Overwhelming bacterial infections (sepsis) are a common cause of kidney failure in the intensive care unit and following surgery or trauma. Turkish physiologists demonstrated that resveratrol can reduce or prevent both kidney and lung injury in septic rats.57

Resveratrol’s unmatched antioxidant and anti-inflammatory potential has been tapped in studies of its ability to prevent drug-induced kidney damage as well. Nephrotoxicity in rats exposed to the antibiotic gentamicin was significantly reduced and more rapid healing of injured kidney tissue was attained using resveratrol, with dramatic reduction in markers of oxidant injury.58 A team of toxicologists in Brazil demonstrated its kidney protective power against cisplatin, the powerful chemotherapy agent responsible for so much drug-induced kidney damage.59 Finally, Indian pharmacologists were successful in protecting animal kidneys from damage caused by another common chemotherapy and immune suppressant drug cyclosporine A by pre-treating the animals with resveratrol.56

Since diabetes is the leading cause of kidney disease—and because the damage it inflicts is largely mediated by free radical production resulting from destructive alteration of proteins by glucose (glycation)—researchers have explored resveratrol as a preventive in diabetic kidney damage. Promising work has come from Indian pharmacologists, who’ve shown that they could significantly attenuate kidney damage in rats with experimentally induced diabetes—even 4 weeks after the diabetes was induced!60

In the researchers’ own words, “The present study reinforces the important role of oxidative stress in diabetic kidney disease and points towards the possible antioxidative mechanism being responsible for the renoprotective action of resveratrol.”

Resveratrol’s unmatched antioxidant and anti-inflammatory potential has been tapped in studies of its ability to prevent drug-induced kidney damage as well.

Lipoic Acid

Like resveratrol, lipoic acid is a powerful antioxidant with few known side effects.76 Lipoic acid has been successfully employed in the laboratory to block the oxidative damage caused by ischemia/reperfusion injury, thereby opening the door to another effective treatment for this common cause of acute kidney failure.77 For example, in 2008 researchers showed that they could reverse all adverse effects on renal function and lab abnormalities produced following experimental ischemia/reperfusion injury in animals.78

Lipoic acid has been comprehensively studied worldwide for its power to prevent or mitigate drug-induced kidney damage. We know that lipoic acid is an effective kidney-protective agent against damage inflicted by Adriamycin®,79,80 the immunosuppressive drug cyclosporine A,76,81,82 and even against acute toxic doses of the pain reliever acetaminophen.83 In studies of protection against cyclosporine toxicity, lipoic acid also helped to normalize blood lipid abnormalities.82

Lipoic Acid

Nephrologists at Georgetown University are examining lipoic acid in the context of diabetic kidney disease. Their results show it can improve renal function in diabetes by lowering sugar levels.84

They have also recently demonstrated that lipoic acid lowers protein loss in urine and improves kidney structure and function in diabetic laboratory animals by reducing oxidative stress.84

In yet another compelling study, Korean researchers recently showed that they could improve kidney patients’ responses to the vasodilator (blood vessel relaxer) nitric oxide (NO) by supplementing them with lipoic acid.85 Loss of endothelial responsiveness to NO is a cause of vascular disease in diabetics, and a chemical called asymmetric dimethylarginine (ADMA) is a sensitive marker and predictor of cardiovascular outcome in patients with end-stage renal disease. Fifty patients on hemodialysis were treated with lipoic acid 600 mg per day for 12 weeks. Levels of the marker ADMA remained unchanged in the control group, but fell significantly in the lipoic acid group, suggesting that lipoic acid may reduce the risk of cardiovascular complications in this group of patients.

Risks to Kidney Health

Given the toxic, high-pressure conditions involved in renal function and the delicacy of the kidney’s structural components, it comes as no surprise that an array of near-constant internal and external insults may take a severe toll on the glomeruli and other parts of the kidney. Their incremental damage and destruction leads to the progressive decline in renal function seen in aging humans.

These internal and external insults include:

  • Hypertension. Over time, chronic high blood pressure inflicts damage to the endothelial cells lining the kidney’s blood vessels, including those within the glomeruli. The result is a familiar cascade of events that leads to the thickening of blood vessel walls and reduction in blood flow seen in atherosclerosis. Reduced blood flow is in turn directly translated into lower GFR. Pressure damage to the glomeruli also diminishes their filtration capacity, permitting large protein molecules such as albumin to pass into urine instead of remaining in circulation. (This is why urine albumin levels are used to detect kidney disease.)
  • Elevated serum glucose. Diabetes is now the leading cause of CKD.61 Experts predict even greater increases in CKD if rates of diabetes incidence continue to rise steeply.62 It should be noted, however, that high blood sugar poses a threat to kidney health even in non-diabetic individuals. Chronic exposure to glucose degrades and destroys kidney cells through the formation of advanced glycation end products (AGEs)—molecules generated through the pathologic binding of glucose to proteins in the body. AGEs cause primary structural proteins in the cells to cross-link and become non-functional, increasing oxidative stress, inflammation, and directly damaging kidney tissue.63-66 It has been established that even early-stage insulin resistance is associated with CKD.67
  • Excess fatty tissue. Body fat contributes to the development of CKD through production of inflammatory cytokines specific to adipose (fatty) tissue called adipokines. Along with AGEs and oxidative stress, adipokines exacerbate the inflammation commonly found in people with CKD.67 For this reason, metabolic syndrome—co-occurring insulin resistance, hypertension, and abdominal obesity—represents a perfect storm for the development of CKD. A 2007 study found that metabolic syndrome occurs in 30.5% of individuals with stage 4 or stage 5 chronic kidney disease.68 Metabolic syndrome increases the risk of chronic kidney disease, even before diabetes manifests.69
  • Protein over-consumption. Ingesting an excessive amount of protein, particularly meat, may tax the kidneys to the point of distress. The extraordinary increase in individuals adhering misguidedly to high-protein diets in order to lose weight has had the unintended consequence of boosting rates of kidney damage and disease. Meat consumption also results in high AGE production and the consequent inflammatory injury to kidney tissue.69 A prudent approach to dietary protein is thus encouraged by most experts, particularly in people who already have some degree of CKD.
  • Drugs. The nephrotoxic side effects of many commonly used medications comprise another significant causative factor in CKD. Chief among the mechanisms by which drugs cause kidney damage are oxidative stress and adverse alterations in cellular energy management. So-called “analgesic nephropathy” involves destruction of the active regions of the kidney by overuse of pain relievers, usually used in combinations of two or more, including the common over-the-counter medication acetaminophen (Tylenol®) as well as non-steroidal anti-inflammatory medications (NSAIDs) including high-dose aspirin, and ibuprofen (Advil®, Motrin®).70-73 Chemotherapy agents have also been shown to significantly impair renal function.74,75 Please note that acetaminophen inflicts kidney damage via a different mechanism than pain relieving drugs like ibuprofen.

Overcoming CKD-Induced Fatigue

L-carnitine, an amino acid-derived nutrient crucial to cellular energy management, can play a vital role in kidney disease prevention and management.86,87 Carnitine deficiency is itself a known causative factor in the development of kidney disease. Conversely, patients with kidney disease frequently develop carnitine deficiency, especially those on dialysis. Carnitine therapy is known to lead to improvements in many kidney-disease-associated complications including cardiovascular disease, anemia, decreased exercise tolerance, weakness, and fatigue.87

As noted earlier, CKD sufferers are at very high risk for developing cardiovascular complications, including heart attacks and heart failure. This is thought to be in part related to massive oxidative stress induced by kidney disease, and partly to inadequate energy management in cardiac tissues induced by carnitine deficiency.88 The frequent result of these interrelated factors is a massive deterioration in energy, exercise tolerance, quality of life—and perhaps even longevity.89

As early as 1998 scientists in Kentucky discovered that supplementation with L-carnitine could improve patient-reported general health, vitality, and physical function in people on dialysis.90 In 2001, research by clinicians at Los Angeles Medical Center showed that L-carnitine, given intravenously to dialysis patients, could reduce fatigue and preserve exercise capacity.91 A literature review by nephrologists at Vanderbilt University in 2003 indicated that L-carnitine supplementation should be used to improve red blood cell count in dialysis patients whose anemia doesn’t respond to therapy with the hormone erythropoietin.92 Finally, more data from Italy demonstrate that L-carnitine supplements can help suppress levels of the inflammatory marker C-reactive protein, potentially reducing cardiovascular risk in dialysis patients.93

silymarin complex

Additional Nutrients that May Benefit CKD

Folic acid is well known for its capacity to reduce levels of the metabolite homocysteine, which is strongly associated with cardiovascular disease and dramatically elevated in individuals with kidney disease or kidney failure.94-97

Omega-3 fatty acids have been shown to help correct cardiovascular risk factors98-100 and to improve kidney function in patients with established kidney disease.101,102 Research published in 2009 suggests that diets rich in omega-3s may actually prevent kidney disease.103,104

Through powerful antioxidant effects, vitamin E may help prevent CKD onset, and vitamins E and C may mitigate development of cardiovascular and other complications in patients with chronic kidney disease.105-110

Summary

Chronic kidney disease (CKD) is rapidly approaching epidemic proportions, with up to 26 million Americans suffering from some form of kidney disease. Kidneys must filter 200 quarts of blood every day. The high-pressure and toxin-rich environment to surrounding renal function renders these delicate, highly complex organs especially vulnerable to damage, dysfunction, and disease.

High blood pressure, elevated blood sugar¸ NSAIDs (such as ibuprofen), certain medications, and high-protein diets are the most common threats to kidney health. The potentially lethal insults they inflict include oxidative stress, production of advanced glycation and lipoxidation end products (AGEs and ALEs), inflammation, and an excessive filtration burden that taxes renal function over time.

Nutrients such as pyridoxal-5-phosphate (P5P) fight AGEs and ALEs. CoQ10, silymarin, resveratrol, and lipoic acid are also clinically supported, potent interventions. Omega-3 fatty acids help quell inflammation, contributing to enhanced kidney health. A host of additional nutrients complement these actions, including folic acid (folate) and vitamins C and E.

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. Available at: http://emedicine.medscape.com/article/238798-overview. Accessed February 18, 2010.

2. Available at: http://www.usrds.org/adr.htm. Accessed March 30, 2009.

3. Available at: http://www.kidney.org/kidneydisease/ckd/index.cfm. Accessed April 4, 2009.

4. Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis. 2003 Jan;41(1):1-12.

5. Galil AG, Pinheiro HS, Chaoubah A, Costa DM, Bastos MG. Chronic kidney disease increases cardiovascular unfavourable outcomes in outpatients with heart failure. BMC Nephrol. 2009 Oct 21;10:31.

6. Voziyan PA, Hudson BG. Pyridoxamine: the many virtues of a maillard reaction inhibitor. Ann N Y Acad Sci. 2005 Jun;1043:807-16.

7. Ahmed N, Thornalley PJ. Advanced glycation endproducts: what is their relevance to diabetic complications? Diabetes Obes Metab. 2007 May;9(3):233-45.

8. Williams ME. New potential agents in treating diabetic kidney disease: the fourth act. Drugs. 2006;66(18):2287-98.

9. Alderson NL, Chachich ME, Frizzell N, et al. Effect of antioxidants and ACE inhibition on chemical modification of proteins and progression of nephropathy in the streptozotocin diabetic rat. Diabetologia. 2004 Aug;47(8):1385-95.

10. Metz TO, Alderson NL, Chachich ME, Thorpe SR, Baynes JW. Pyridoxamine traps intermediates in lipid peroxidation reactions in vivo: evidence on the role of lipids in chemical modification of protein and development of diabetic complications. J Biol Chem. 2003 Oct 24;278(43):42012-9.

11. Metz TO, Alderson NL, Thorpe SR, Baynes JW. Pyridoxamine, an inhibitor of advanced glycation and lipoxidation reactions: a novel therapy for treatment of diabetic complications. Arch Biochem Biophys. 2003 Nov 1;419(1):41-9.

12. Onorato JM, Jenkins AJ, Thorpe SR, Baynes JW. Pyridoxamine, an inhibitor of advanced glycation reactions, also inhibits advanced lipoxidation reactions. Mechanism of action of pyridoxamine. J Biol Chem. 2000 Jul 14;275(28):21177-84.

13. Degenhardt TP, Alderson NL, Arrington DD, et al. Pyridoxamine inhibits early renal disease and dyslipidemia in the streptozotocin-diabetic rat. Kidney Int. 2002 Mar;61(3):939-50.

14. Alderson NL, Chachich ME, Youssef NN, et al. The AGE inhibitor pyridoxamine inhibits lipemia and development of renal and vascular disease in Zucker obese rats. Kidney Int. 2003 Jun;63(6):2123-33.

15. Zheng F, Zeng YJ, Plati AR, et al. Combined AGE inhibition and ACEi decreases the progression of established diabetic nephropathy in B6 db/db mice. Kidney Int. 2006 Aug;70(3):507-14.

16. Williams ME, Bolton WK, Khalifah RG, Degenhardt TP, Schotzinger RJ, McGill JB. Effects of pyridoxamine in combined phase 2 studies of patients with type 1 and type 2 diabetes and overt nephropathy. Am J Nephrol. 2007;27(6):605-14.

17. Murakoshi M, Tanimoto M, Gohda T, et al. Pleiotropic effect of pyridoxamine on diabetic complications via CD36 expression in KK-Ay/Ta mice. Diabetes Res Clin Pract. 2009 Feb;83(2):183-9.

18. Tanimoto M, Gohda T, Kaneko S, et al. Effect of pyridoxamine (K-163), an inhibitor of advanced glycation end products, on type 2 diabetic nephropathy in KK-A(y)/Ta mice. Metabolism.2007 Feb;56(2):160-7.

19. Waanders F, van den Berg E, Nagai R, van Veen I, Navis G, van Goor H. Renoprotective effects of the AGE-inhibitor pyridoxamine in experimental chronic allograft nephropathy in rats. Nephrol Dial Transplant. 2008 Feb;23(2):518-24.

20. Nakamura S, Li H, Adijiang A, Pischetsrieder M, Niwa T. Pyridoxal phosphate prevents progression of diabetic nephropathy. Nephrol Dial Transplant. 2007 Aug;22(8):2165-74.

21. Kirsten R, Heintz B, Nelson K, et al. Magnesium pyridoxal 5-phosphate glutamate reduces hyperlipidaemia in patients with chronic renal insufficiency. Eur J Clin Pharmacol. 1988;34(2):133-7.

22. Gazdikova K, Gvozdjakova A, Kucharska J, Spustova V, Braunova Z, Dzurik R. Malondialdehyde and selected antioxidant plasma levels in conservatively treated patients with kidney diseases. Bratisl Lek Listy. 2000;101(9):490-4.

23. Igarashi T, Nakajima Y, Tanaka M, Otake S. Effect of coenzyme Q10 on experimental hypertension in rats and dogs. J Pharmacol Exp Ther. 1974 Apr;189(1):149-56.

24. Morotomi Y, Oniki H, Onoyama K, Fukiyama K, Omae T. Effects of coenzyme Q10 on the blood pressure and renal renin content in spontaneously hypertensive rats. Igaku Kenkyu. 1975 Nov;45(5):303-8.

25. Lippa S, Colacicco L, Bondanini F, et al. Plasma levels of coenzyme Q(10), vitamin E and lipids in uremic patients on conservative therapy and hemodialysis treatment: some possible biochemical and clinical implications. Clin Chim Acta. 2000 Feb 25;292(1-2):81-91.

26. Gazdikova K, Gvozdjakova A, Kucharska J, Spustova V, Braunova Z, Dzurik R. Effect of coenzyme Q10 in patients with kidney diseases. Cas Lek Cesk. 2001 May 24;140(10):307-10.

27. Rauscher FM, Sanders RA, Watkins JB, 3rd. Effects of coenzyme Q10 treatment on antioxidant pathways in normal and streptozotocin-induced diabetic rats. J Biochem Mol Toxicol. 2001;15(1):41-6.

28. Dlugosz A, Kuzniar J, Sawicka E, et al. Oxidative stress and coenzyme Q10 supplementation in renal transplant recipients. Int Urol Nephrol. 2004;36(2):253-8.

29. Available at: http://kidney.niddk.nih.gov/Kudiseases/pubs/yourkidneys/. Accessed February 17, 2010.

30. Shlipak MG, Sarnak MJ, Katz R, et al. Cystatin C and the risk of death and cardiovascular events among elderly persons. N Engl J Med. 2005 May 19;352(20):2049-60.

31. Levey AS, Stevens LA, Coresh J. Conceptual model of CKD: applications and implications. Am J Kidney Dis. 2009 Mar;53(3 Suppl 3):S4-16.

32. Available at:http://clinicalevidence.bmj.com/ceweb/conditions/knd/2002/2002_background.jsp. Accessed February 17, 2010.

33. Farswan M, Rathod SP, Upaganlawar AB, Semwal A. Protective effect of coenzyme Q10 in simvastatin and gemfibrozil induced rhabdomyolysis in rats. Indian J Exp Biol. 2005 Oct 2005;43(10):845-8.

34. Upaganlawar A, Farswan M, Rathod S, Balaraman R. Modification of biochemical parameters of gentamicin nephrotoxicity by coenzyme Q10 and green tea in rats. Indian J Exp Biol. 2006 May;44(5):416-8.

35. Post-White J, Ladas EJ, Kelly KM. Advances in the use of milk thistle (Silybum marianum). Integr Cancer Ther. 2007 Jun;6(2):104-9.

36. Wojcikowski K, Stevenson L, Leach D, Wohlmuth H, Gobe G. Antioxidant capacity of 55 medicinal herbs traditionally used to treat the urinary system: a comparison using a sequential three-solvent extraction process. J Altern Complement Med. 2007 Jan-Feb;13(1):103-9.

37. Floersheim GL. Experimental basis for the therapy of Amanita phalloides poisoning. Schweiz Med Wochenschr. 1978 Feb 11;108(6):185-97.

38. Vogel G, Braatz R, Mengs U. On the nephrotoxicity of alpha-amanitin and the antagonistic effects of silymarin in rats. Agents Actions. 1979 Jun;9(2):221-6.

39. Launay-Vacher V, Rey JB, Isnard-Bagnis C, Deray G, Daouphars M, European Society of Clinical Pharmacy Special Interest Group on Cancer C. Prevention of cisplatin nephrotoxicity: state of the art and recommendations from the European Society of Clinical Pharmacy Special Interest Group on Cancer Care. Cancer Chemother Pharmacol. 2008 May;61(6):903-9.

40. Machado V, Cabral A, Monteiro P, Goncalves L, Providencia LA. Carvedilol as a protector against the cardiotoxicity induced by anthracyclines (doxorubicin). Rev Port Cardiol. 2008 Oct;27(10):1277-96.

41. Yao X, Panichpisal K, Kurtzman N, Nugent K. Cisplatin nephrotoxicity: a review. Am J Med Sci. 2007Aug;334(2):115-24.

42. Bokemeyer C, Fels LM, Dunn T, et al. Silibinin protects against cisplatin-induced nephrotoxicity without compromising cisplatin or ifosfamide anti-tumour activity. Br J Cancer. 1996 Dec;74(12):2036-41.

43. Gaedeke J, Fels LM, Bokemeyer C, Mengs U, Stolte H, Lentzen H. Cisplatin nephrotoxicity and protection by silibinin. Nephrol Dial Transplant. 1996 Jan;11(1):55-62.

44. Karimi G, Ramezani M, Tahoonian Z. Cisplatin nephrotoxicity and protection by milk thistle extract in rats. Evid Based Complement Alternat Med. 2005 Sep;2(3):383-6.

45. El-Shitany NA, El-Haggar S, El-desoky K. Silymarin prevents adriamycin-induced cardiotoxicity and nephrotoxicity in rats. Food Chem Toxicol. 2008 Jul;46(7):2422-8.

46. Wenzel S, Stolte H, Soose M. Effects of silibinin and antioxidants on high glucose-induced alterations of fibronectin turnover in human mesangial cell cultures. J Pharmacol Exp Ther. 1996 Dec;279(3):1520-6.

47. Senturk H, Kabay S, Bayramoglu G, et al. Silymarin attenuates the renal ischemia/reperfusion injury-induced morphological changes in the rat kidney. World J Urol. 2008 Aug;26(4):401-7.

48. Turgut F, Bayrak O, Catal F, et al. Antioxidant and protective effects of silymarin on ischemia and reperfusion injury in the kidney tissues of rats. Int Urol Nephrol. 2008;40(2):453-60.

49. Caimi G, Carollo C, Lo Presti R. Chronic renal failure: oxidative stress, endothelial dysfunction and wine. Clin Nephrol. 2004 Nov;62(5):331-5.

50. Bertelli AA, Migliori M, Panichi V, et al. Resveratrol, a component of wine and grapes, in the prevention of kidney disease. Ann N Y Acad Sci. 2002 May;957:230-8.

51. Giovannini L, Migliori M, Longoni BM, et al. Resveratrol, a polyphenol found in wine, reduces ischemia reperfusion injury in rat kidneys. J Cardiovasc Pharmacol. 2001 Mar;37(3):262-70.

52. Saito M, Satoh S, Kojima N, et al. Effects of a phenolic compound, resveratrol, on the renal function and costimulatory adhesion molecule CD86 expression in rat kidneys with ischemia/reperfusion injury. Arch Histol Cytol. 2005;68(1):41-9.

53. Chander V, Chopra K. Protective effect of resveratrol, a polyphenolic phytoalexin on glycerol-induced acute renal failure in rat kidney. Ren Fail. 2006;28(2):161-9.

54. Chander V, Chopra K. Protective effect of nitric oxide pathway in resveratrol renal ischemia-reperfusion injury in rats. Arch Med Res. Jan 2006;37(1):19-26.

55. Chander V, Chopra K. Role of nitric oxide in resveratrol-induced renal protective effects of ischemic preconditioning. J Vasc Surg. 2005 Dec;42(6):1198-205.

56. Chander V, Tirkey N, Chopra K. Resveratrol, a polyphenolic phytoalexin protects against cyclosporine-induced nephrotoxicity through nitric oxide dependent mechanism. Toxicology. 2005 May 15;210(1):55-64.

57. Kolgazi M, Sener G, Cetinel S, Gedik N, Alican I. Resveratrol reduces renal and lung injury caused by sepsis in rats. J Surg Res. 2006 Aug;134(2):315-21.

58. Silan C, Uzun O, Comunoglu NU, Gokcen S, Bedirhan S, Cengiz M. Gentamicin-induced nephrotoxicity in rats ameliorated and healing effects of resveratrol. Biol Pharm Bull. 2007 Jan;30(1):79-83.

59. Do Amaral CL, Francescato HD, Coimbra TM, et al. Resveratrol attenuates cisplatin-induced nephrotoxicity in rats. Arch Toxicol. 2008 Jun;82(6):363-70.

60. Sharma S, Anjaneyulu M, Kulkarni SK, Chopra K. Resveratrol, a polyphenolic phytoalexin, attenuates diabetic nephropathy in rats. Pharmacology. 2006;76(2):69-75.

61. Whaley-Connell AT, Sowers JR, McFarlane SI, et al. Diabetes mellitus in CKD: Kidney Early Evaluation Program (KEEP) and National Health and Nutrition and Examination Survey (NHANES) 1999-2004. Am J Kidney Dis. 2008 Apr;51(4 Suppl 2):S21-9.

62. Fox CS, Muntner P. Trends in diabetes, high cholesterol, and hypertension in chronic kidney disease among U.S. adults: 1988-1994 to 1999-2004. Diabetes Care. 2008 Jul;31(7):1337-42.

63. Orasanu G, Plutzky J. The pathologic continuum of diabetic vascular disease. J Am Coll Cardiol. 2009 Feb 3;53(5 Suppl):S35-42.

64. Bohlender JM, Franke S, Stein G, Wolf G. Advanced glycation end products and the kidney. Am J Physiol Renal Physiol. 2005 Oct;289(4):F645-59.

65. Uribarri J, Cai W, Sandu O, Peppa M, Goldberg T, Vlassara H. Diet-derived advanced glycation end products are major contributors to the body’s AGE pool and induce inflammation in healthy subjects. Ann N Y Acad Sci. 2005 Jun;1043:461-6.

66. Linden E, Cai W, He JC, et al. Endothelial dysfunction in patients with chronic kidney disease results from advanced glycation end products (AGE)-mediated inhibition of endothelial nitric oxide synthase through RAGE activation. Clin J Am Soc Nephrol. 2008 May;3(3):691-8.

67. de Vinuesa SG, Goicoechea M, Kanter J, et al. Insulin resistance, inflammatory biomarkers, and adipokines in patients with chronic kidney disease: effects of angiotensin II blockade. J Am Soc Nephrol. 2006 Dec;17(12 Suppl 3):S206-12.

68. Johnson DW, Armstrong K, Campbell SB, et al. Metabolic syndrome in severe chronic kidney disease: Prevalence, predictors, prognostic significance and effects of risk factor modification. Nephrology (Carlton). 2007 Aug;12(4):391-8.

69. Lucove J, Vupputuri S, Heiss G, North K, Russell M. Metabolic syndrome and the development of CKD in American Indians: the Strong Heart Study. Am J Kidney Dis. 2008 Jan;51(1):21-8.

70. Uribarri J, Tuttle KR. Advanced glycation end products and nephrotoxicity of high-protein diets. Clin J Am Soc Nephrol. 2006 Nov;1(6):1293-9.

71. Gault MH, Barrett BJ. Analgesic nephropathy. Am J Kidney Dis. 1998 Sep;32(3):351-60.

72. Elseviers MM, De Broe ME. Analgesic nephropathy: is it caused by multi-analgesic abuse or single substance use? Drug Saf. 1999 Jan;20(1):15-24.

73. Mann JF, Goerig M, Brune K, Luft FC. Ibuprofen as an over-the-counter drug: is there a risk for renal injury? Clin Nephrol. 1993 Jan;39(1):1-6.

74. Naughton CA. Drug-induced nephrotoxicity. Am Fam Physician. 2008 Sep 15;78(6):743-50.

75. Zager RA. Pathogenetic mechanisms in nephrotoxic acute renal failure. Semin Nephrol. 1997 Jan;17(1):3-14.

76. Amudha G, Josephine A, Varalakshmi P. Role of lipoic acid in reducing the oxidative stress induced by cyclosporine A. Clin Chim Acta. Oct 2006;372(1-2):134-9.

77. Takaoka M, Ohkita M, Kobayashi Y, Yuba M, Matsumura Y. Protective effect of alpha-lipoic acid against ischaemic acute renal failure in rats. Clin Exp Pharmacol Physiol. 2002 Mar;29(3):189-94.

78. Sehirli O, Sener E, Cetinel S, Yuksel M, Gedik N, Sener G. Alpha-lipoic acid protects against renal ischaemia-reperfusion injury in rats. Clin Exp Pharmacol Physiol. 2008 Mar ;35(3):249-55.

79. Malarkodi KP, Balachandar AV, Varalakshmi P. Protective effect of lipoic acid on adriamycin induced lipid peroxidation in rat kidney. Mol Cell Biochem. 2003 May;247(1-2):9-13.

80. Malarkodi KP, Balachandar AV, Varlakshmi P. The influence of lipoic acid on adriamycin-induced hyperlipidemic nephrotoxicity in rats. Mol Cell Biochem. 2003 May;247(1-2):139-45.

81. Amudha G, Josephine A, Mythili Y, Sundarapandiyan R, Varalakshmi P. Therapeutic efficacy of DL-alpha-lipoic acid on cyclosporine A induced renal alterations. Eur J Pharmacol. 2007 Oct 1;571(2-3):209-14.

82. Amudha G, Josephine A, Varalakshmi P. Beneficial effect of DL-alpha-lipoic acid on cyclosporine A induced hyperlipidemic nephropathy in rats. Mol Cell Biochem. 2007 Jul;301(1-2):165-71.

83. Abdel-Zaher AO, Abdel-Hady RH, Mahmoud MM, Farrag MM. The potential protective role of alpha-lipoic acid against acetaminophen-induced hepatic and renal damage. Toxicology. 2008 Jan 20;243(3):261-70.

84. Bhatti F, Mankhey RW, Asico L, Quinn MT, Welch WJ, Maric C. Mechanisms of antioxidant and pro-oxidant effects of alpha-lipoic acid in the diabetic and nondiabetic kidney. Kidney Int. 2005 Apr;67(4):1371-80.

85. Chang JW, Lee EK, Kim TH, et al. Effects of alpha-lipoic acid on the plasma levels of asymmetric dimethylarginine in diabetic end-stage renal disease patients on hemodialysis: a pilot study. Am J Nephrol. 2007;27(1):70-4.

86. Kendler BS. Carnitine: an overview of its role in preventive medicine. Prev Med.1986 Jul;15(4):373-90.

87. Matera M, Bellinghieri G, Costantino G, Santoro D, Calvani M, Savica V. History of L-carnitine: implications for renal disease. J Ren Nutr. 2003 Jan;13(1):2-14.

88. Calo LA, Bertipaglia L, Pagnin E. Antioxidants, carnitine and erythropoietin. G Ital Nefrol. 2006 Jan-Feb;23 Suppl 34:S47-50.

89. Schreiber BD. Debate forum: levocarnitine therapy is rational and justified in selected dialysis patients. Blood Purif. 2006;24(1):128-39.

90. Sloan RS, Kastan B, Rice SI, et al. Quality of life during and between hemodialysis treatments: role of L-carnitine supplementation. Am J Kidney Dis. 1998 Aug;32(2):265-72.

91. Brass EP, Adler S, Sietsema KE, et al. Intravenous L-carnitine increases plasma carnitine, reduces fatigue, and may preserve exercise capacity in hemodialysis patients. Am J Kidney Dis. 2001 May;37(5):1018-28.

92. Golper TA, Goral S, Becker BN, Langman CB. L-carnitine treatment of anemia. Am J Kidney Dis. 2003 Apr;41(4 Suppl 4):S27-34.

93. Savica V, Santoro D, Mazzaglia G, et al. L-carnitine infusions may suppress serum C-reactive protein and improve nutritional status in maintenance hemodialysis patients. J Ren Nutr. 2005 Apr;15(2):225-30.

94. Alvares Delfino VD, de Andrade Vianna AC, Mocelin AJ, Barbosa DS, Mise RA, Matsuo T. Folic acid therapy reduces plasma homocysteine levels and improves plasma antioxidant capacity in hemodialysis patients. Nutrition. 2007 Mar;23(3):242-7.

95. Bostom AG, Carpenter MA, Kusek JW, et al. Rationale and design of the Folic Acid for Vascular Outcome Reduction In Transplantation (FAVORIT) trial. Am Heart J. 2006 Sep;152(3):448. e1-7.

96. Menon V, Wang X, Greene T, et al. Homocysteine in chronic kidney disease: Effect of low protein diet and repletion with B vitamins. Kidney Int. 2005 Apr;67(4):1539-46.

97. Nanayakkara PW, van Guldener C, ter Wee PM, et al. Effect of a treatment strategy consisting of pravastatin, vitamin E, and homocysteine lowering on carotid intima-media thickness, endothelial function, and renal function in patients with mild to moderate chronic kidney disease: results from the Anti-Oxidant Therapy in Chronic Renal Insufficiency (ATIC) Study. Arch Intern Med. 2007 Jun 25;167(12):1262-70.

98. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev. 2001(3):CD003205.

99. Hartweg J, Farmer AJ, Holman RR, Neil A. Potential impact of omega-3 treatment on cardiovascular disease in type 2 diabetes. Curr Opin Lipidol. 2009 Feb;20(1):30-8.

100. Moreira AC, Gaspar A, Serra MA, Simoes J, Lopes da Cruz J, Amaral TF. Effect of a sardine supplement on C-reactive protein in patients receiving hemodialysis. J Ren Nutr. 2007 May;17(3):205-13.

101. Miller ER 3rd, Juraschek SP, Appel LJ, et al. The effect of n-3 long-chain polyunsaturated fatty acid supplementation on urine protein excretion and kidney function: meta-analysis of clinical trials. Am J Clin Nutr. 2009 Jun;89(6):1937-45.

102. Parinyasiri U, Ong-Ajyooth L, Parichatikanond P, Ong-Ajyooth S, Liammongkolkul S, Kanyog S. Effect of fish oil on oxidative stress, lipid profile and renal function in IgA nephropathy. J Med Assoc Thai. 2004 Feb;87(2):143-9.

103. Bell S, Cooney J, Packard CJ, Caslake M, Deighan CJ. Omega-3 fatty acids improve postprandial lipaemia in patients with nephrotic range proteinuria. Atherosclerosis. 2009 Jul;205(1):296-301.

104. Garman JH, Mulroney S, Manigrasso M, Flynn E, Maric C. Omega-3 fatty acid rich diet prevents diabetic renal disease. Am J Physiol Renal Physiol. 2009 Feb;296(2):F306-16.

105. Abdel-Naim AB, Abdel-Wahab MH, Attia FF. Protective effects of vitamin e and probucol against gentamicin-induced nephrotoxicity in rats. Pharmacol Res.1999 Aug;40(2):183-7.

106. Boaz M, Smetana S, Weinstein T, et al. Secondary prevention with antioxidants of cardiovascular disease in endstage renal disease (SPACE): randomised placebo-controlled trial. Lancet. 2000 Oct 7;356(9237):1213-18.

107. Khajehdehi P, Mojerlou M, Behzadi S, Rais-Jalali GA. A randomized, double-blind, placebo-controlled trial of supplementary vitamins E, C and their combination for treatment of haemodialysis cramps. Nephrol Dial Transplant. 2001 Jul;16(7):1448-51.

108. Mune M, Yukawa S, Kishino M, et al. Effect of vitamin E on lipid metabolism and atherosclerosis in ESRD patients. Kidney Int Suppl. 1999 Jul;71:S126-9.

109. Ramos R, Gomez-Gerique N, Martinez-Castelao A. Lipoprotein oxidation profile in end stage renal disease patients. Role of vitamin C supplementation. Nefrologia. 2005;25(2):178-84.

110. Tain YL, Freshour G, Dikalova A, Griendling K, Baylis C. Vitamin E reduces glomerulosclerosis, restores renal neuronal NOS, and suppresses oxidative stress in the 5/6 nephrectomized rat. Am J Physiol Renal Physiol. 2007 May;292(5):F1404-10.