Dietary, Botanical, and Nutraceutical Treatments for Type 2 Diabetes


 

The Web diabetesmanager

 

 

Non-Pharmaceutical Interventions for Type 2 Diabetes: Nutrition, Botanicals, Antioxidants, Minerals, and Other Dietary Supplements

 

Joseph L Evans, PhD

 

 

Last Author Revision: March 2010

 


 

INTRODUCTION

Type 2 diabetes has reached epidemic proportions in the US and worldwide (>18 million and 160 million individuals, respectively), and is projected to increase dramatically [1]. Furthermore, the prevalence of insulin resistance, a major causative factor in the early development of type 2 diabetes and an independent risk factor for cardiovascular disease and the metabolic syndrome X, is even more widespread [2][3][4]. This situation is further exacerbated by obesity, a major risk for developing type 2 diabetes. The number of adults overweight or obese in the US is 125 million (65% of population) and 1.3 billion worldwide [5]. Since dietary modification and increased physical activity provide insufficient glucose control over the long-term course of the disease, the vast majority of patients require some type of pharmacological intervention [6].

 

Although pharmacological options for the management of type 2 diabetes have been increasing, and will continue to do so [7][8], not all patients benefit from them. In addition, the cost of prescription medications may exceed the financial capacity of an increasing number of older citizens, those without adequate health insurance, and those living in poverty [9][10]). Furthermore, certain ethnic groups who are at increased risk for developing diabetes (e.g. Asians, Hispanics, and Native Americans), come from cultures with a long history of use of traditional medicines, and are likely to employ one or more folk (botanical) treatments rather than prescription medications [11][12][13][14][15][16][17][18][19][20]. Though a majority of diabetic patients are being treated, many are unable to achieve the current American Diabetes Association-recommended goal of HbA1c < 7%. Thus, in patients with poorly controlled type 2 diabetes, especially those who are obese, there is a need to identify and evaluate adjunctive therapies that are safe, efficacious, and cost-effective.

 

Botanical extracts, vitamins, antioxidants, minerals, amino acids, and fatty acids (natural products collectively and interchangeably referred to here as dietary supplements / nutraceuticals) are an important source of new therapies for T2D and insulin resistance ( http://nccam.nih.gov/health/diabetes/). These agents are marketed in the US under the Dietary Supplement Health and Education Act (DSHEA), passed by Congress in 1994. DSHEA defined a new category of food for regulatory purposes, termed dietary supplements, one that also includes concentrates, metabolites, constituents, extracts, and combinations, and has resulted in major changes in the marketing and use of nutraceuticals in the US (see below).

 

From a regulatory perspective, nutraceuticals are treated differently compared to either over the counter (OTC) or prescription pharmaceuticals [21][22][23]. DSHEA strictly requires manufacturers of botanicals and nutraceuticals to clearly state on all product labels that it is not intended to diagnose, treat, cure, or prevent any disease. Statements can be made to suggest that a dietary supplement enhances general health, or refers to a documented biochemical or physiological mechanism whereby a supplement affects a structure or function. These claims are commonly referred to as structure/function claims (e.g. supports cardiovascular health, immune function, etc.). In contrast to the regulatory requirements for OTC and prescription pharmaceuticals, manufacturers of dietary supplements are not required, under DSHEA, to submit evidence of safety or efficacy prior to marketing. Although a dietary supplement manufacturer is ultimately responsible for the safety of its products, the Food and Drug Administration (FDA) bears the burden of proof to show that a product is unsafe. To remove a product from the market, the FDA must be convinced that it possesses an unreasonable risk of harm at the recommended dose [24].

 

For example, the increased risk for adverse events by ephredra-containing products compared with other botanicals and nutraceuticals has triggered increased pressure on the FDA to remove ephredra products from the market [25][26][27][28]. A detailed comparison of the regulatory requirements of dietary supplements, OTC, and prescription medications has been recently published [29].

 

Despite the caveat emptor atmosphere surrounding dietary supplements [30], they are used by approximately 60% of the US population on at least an occasional basis (Figure 1), and constitute a significant portion of the US healthcare market (Figure 2). In 2001, US sales of botanicals and nutraceuticals were estimated at $17.6 billion (Consumer Research in the Nutrition Industry II, March 2002, Nutrition Business Journal Report, New Hope Natural Media). This figure represents approximately 33% of the total US nutrition market in 2001 (~$53 billion). The US sales in 2001 of botanicals and nutraceuticals directed towards glucose control and the resultant micro- and macrovascular complications has been estimated at approximately 2.5% of the total supplement sales or $438 million, and is expected to grow at an annual rate of approximately 10% (Condition-Specific Supplement Markets I, November 2001, Nutrition Business Journal Report, New Hope Natural Media). A recent nationally (US) representative survey has reported that over 30% of patients with diabetes used complimentary and alternative medicine (CAM) to manage their condition [31]; in certain ethnic populations with diabetes (Navajo, Vietnamese, Hispanic), the percentages are even higher (40-66%)[32][33][34][35]. In response to the increasing use of CAM by the general public [36] and by patients with diabetes [37], the American Diabetes Association has issued a ‘position paper’ calling on health care providers to ask their patients about their use of CAM [38].

 

 

Figure 1. US consumer supplement use and sales, 2001. Left panel, Estimated supplement use as percent of total US population. Right panel, Estimated supplement sales according to purchasing frequency. US sales of dietary supplements has been estimated at $17.6 billion in 2001 Figure adapted from: Consumer Research in the Nutrition Industry II, March 2002, Nutrition Business Journal Report, New Hope Natural Media).

  

Figure 2. US healthcare market sales, 2000. Distribution of total sales of $184 billion according to category. Rx, prescription drugs; OTC, over-the-counter drugs; DS, dietary supplements. Figure adapted from: Condition-Specific Supplement Markets I, November 2001, Nutrition Business Journal Report, New Hope Natural Media).

 

 

 

Due to the increasing use of nutraceuticals by patients with diabetes and their potential for both positive and negative impact, it is important, therefore, that diabetes health care professionals increase their knowledge of these agents [39]. To this end, several recent comprehensive reviews have focused on the use of botanicals and nutraceuticals for diabetes [40][41][42][43][44]. The overall objective of this chapter therefore is to provide a concise, comparative overview of those botanicals and nutraceuticals that have received the most scientific attention.

 

 

NUTRITIONAL INTERVENTION

Dietary modification is universally recognized by caregivers as an initial intervention and mainstay for the treatment of overweight patients with T2D [45][46][47]. The other side of the non-pharmacological intervention ‘coin’ involves changes in lifestyle, usually consisting of increased physical activity [48], smoking cessation, and reduced intake of alcohol. The overall objectives of these approaches are 1) weight loss and exercise training both resulting in improved insulin action, 2) improved glycemic and lipid control (both short-term and chronic), 3) reduced likelihood of developing microvascular and macrovascular complications, and 4) improved quality of life. It is important to remember that patients do not have to achieve their ideal body weight to reap significant health benefit. It has been reported that a loss of 10-20 lb (4.5-9 kg) will be helpful, as long as the weight loss and exercise programs are maintained [49][50][51].

 

The recommendations of the American Diabetes Association [52] and the American Association of Clinical Endocrinologists [53] for the nutritional management of patients with T2D are provided elsewhere and will not be discussed in detail herein. The key to successful dietary intervention is to ensure that caloric intake is less than caloric output. For long-term success, this should be coupled with education designed to improve the patient’s understanding of the beneficial effects of dietary modification on blood glucose, lipids, blood pressure, and overall quality of life. Some successful dietary approaches have included those designed to limit intake of saturated fats (< 7% of total caloric intake), and spreading the nutrient load. Dietary factors that are able to spread the nutrient load include increased frequency of food intake, increased intake of soluble fiber, legumes, and increased intake of foods with a low glycemic index. The beneficial role that supplements and foodstuffs high in soluble fiber play in the conventional dietary management of patients with T2D is well documented [54][55][56][57].

 

According to the ADA, it is unlikely that there is an optimal mix of macronutrients for the ‘diabetic diet’. The best mix of carbohydrate, protein, and fat appears to vary depending on individual circumstances. The best mix should provide a total caloric intake that facilitates, at the very minimum, weight maintenance and ideally weight loss. The average daily intake of carbohydrates recommended for patients with diabetes is approximately 45-65% of total caloric intake; low or restricted carbohydrate diets are not recommended in the management of diabetes. Intake of fat should be limited to approximately 20-35%; and dietary cholesterol should be < 200 mg/day. In patients with dyslipidemia, a special effort should be made to limit saturated fat and to substitute unsaturated or monounsaturated fat, especially omega-3 fatty acids. To reduce the risk of stroke and cardiovascular disease, diets high (3-6 servings per day) in fruits and vegetables are highly recommended [58][59][60]. Emerging epidemiological data suggests that higher coffee intake may reduce the risk for the development of T2D [61]. Protein intake (in patients with normal renal function) should be limited to 10-20% of total caloric intake, and reduced to 10-15% at the onset of macroalbuminuria.

 

With regard to micronutrient supplementation, the ADA does not recommend routine supplementation with any vitamin, mineral, or antioxidant in patients with diabetes who do not have an underlying deficiency [62]. The basis for this position is the lack of clear evidence of efficacy and, in some cases, a concern related to long-term safety [63][64][65][66][67]. With regard to alcohol intake, patients with diabetes are advised to follow the same precautions as the general population. Daily intake should be limited to a moderate amount (one drink per day or less for women, and two drinks per day or less for men). In those patients using insulin or insulin secretagogs, alcohol should be consumed with food to reduce the risk of nocturnal hypoglycemia. Alcohol alone does not acutely affect blood glucose or insulin but, if co-ingested with carbohydrate, could raise glucose [68].

 

Mediterranean Diet

Perhaps the most widely recognized diet with health promoting benefits is the so-called Mediterranean Diet. Around 50 years ago, it was recognized by Keys and associates that very low incidences of cardiovascular disease in the areas around Naples, Italy, were associated with what was soon to be termed the Mediterranean Diet [69]. The heart of this diet is mainly vegetarian, and differs from North American and Western European diets in that it is much lower in meat and dairy products, and frequently contains fruits for dessert. This diet reflects a pattern of eating most highly associated with olive growing areas of the Mediterranean. Olive oil is a critical component of this diet both because of its inherent benefits (presence of mono unsaturated fat, i.e. oleic acid) along it allowing for the consumption of high quantities of fruits and vegetables in salads and other prepared foods. Other essential components of this diet are fish (a rich source of omega-3 fatty acids; see Polyunsaturated Fatty Acids in Fatty Acid section below), nuts, wheat, grapes, and derived products including wine [70]. Many studies have convincingly demonstrated that this dietary pattern plays an important role in the prevention of cardiovascular disease [71][72][73][74][75][76][77][78].

 

Chocolate — What’s for Dessert?

As discussed above, there is a growing body of epidemiological evidence supporting the idea that diets rich in fruits and vegetables reduce or delay the onset of many chronic diseases, including cardiovascular and related metabolic diseases [79]. A major class of compounds present in fruits and vegetables that is associated with cardio-protective effects is the flavanols [80]. Cocoa is also rich source of flavanols, a class of polyphenolic antioxidant compounds (e.g. (-)-epicatechin, (+)-catechin, procyanidins) found in plants [81]. In addition to their antioxidant activity, flavanols have also been associated with increased in nitric oxide bioavailability [82][83]. Consumption of chocolate can result in significant increases in plasma epicatechin concentrations and decreases in plasma baseline oxidation products [84]. Since nitric oxide bioavailability deeply influences insulin-stimulated glucose uptake and vascular tone, flavanols have been evaluated for potential positive metabolic and pressor effects. Flavanoid-rich dark chocolate (100g) improved endothelial function and was associated with an increase in plasma epicatechin concentrations in healthy adults [85][86].

 

The effects of either dark or white chocolate bars on blood pressure and glucose and insulin responses to an oral-glucose-tolerance test in healthy subjects was compared [87]. After a 7-day cocoa-free run-in phase, 15 healthy subjects were randomly assigned to receive for 15 days either 100 g dark chocolate bars, which contained approximately 500 mg polyphenols, or 90 g white chocolate bars, which contained no polyphenols. Successively, subjects entered a further cocoa-free washout phase of 7 days, and then were crossed over to the other condition. Oral glucose tolerance tests were performed at the end of each period to calculate the homeostasis model assessment of insulin resistance (HOMA-IR) and the quantitative insulin sensitivity check index (QUICKI); blood pressure was measured daily. HOMA-IR was significantly lower after dark than after white chocolate ingestion (0.94 ± 0.42 compared with 1.72 ± 0.62; P < 0.001), and QUICKI was significantly higher after dark than after white chocolate ingestion (0.398 ± 0.04 compared with 0.356 ± 0.02; P = 0.001). Although within normal values, systolic blood pressure was lower after dark than after white chocolate ingestion (107.5 ± 8.6 compared with 113.9 ± 8.4 mm Hg; P < 0.05). These results suggest that dark but not white chocolate decreases blood pressure and improves insulin sensitivity in healthy persons.

 

These same researchers have also evaluated the effects of dark chocolate (DC) consumption on blood pressure (BP), flow-mediated dilation (FMD), oral glucose tolerance (OGTT), and insulin sensitivity in patients with essential hypertension (EH) [88]. After a 7-day chocolate-free run-in phase, 20 never-treated, grade I patients with EH (10 males; 43.7 ± 7.8 years) were randomized to receive either 100 g per day DC (containing 88 mg flavanols) or 90 g per day flavanol-free white chocolate (WC) in an isocaloric manner for 15 days. After a second 7-day chocolate-free period, patients were crossed over to the other treatment. Noninvasive 24-hour ambulatory BP, FMD, OGTT, serum cholesterol, and markers of vascular inflammation were evaluated at the end of each treatment. The HOMA-IR, QUICKI, and insulin sensitivity index (SI) were calculated from OGTT values. Ambulatory BP decreased after DC (24-hour systolic BP -11.9 ± 7.7 mm Hg, P < 0.0001; 24-hour diastolic BP -8.5 ± 5.0 mm Hg, P < 0.0001), but not WC. DC but not WC decreased HOMA-IR (P < 0.0001), and it improved QUICKI, SI, and FMD. DC also decreased serum LDL cholesterol (from 3.4 ± 0.5 to 3.0 ± 0.6 mmol/L; P<0.05). In summary, DC decreased BP and serum LDL cholesterol, improved FMD, and enhanced insulin sensitivity in patients with EH. Additional studies in larger groups and in individuals with T2D will be needed to confirm these results.

 

 

BOTANICAL INTERVENTIONS

Botanicals have been used for medicinal purposes since the dawn of civilization [89]. It is well documented [90] that many pharmaceuticals commonly used today are structurally derived from natural compounds found in traditional medicinal plants. The development of the anti-hyperglycemic drug metformin (dimethlybiguanide; Glucophage®) can be traced to the traditional use of Galega officinalis to treat diabetes, and the subsequent search to identify active compounds with reduced toxicity [91][92][93]. G. officinalis is far from the only botanical to have been used as a treatment for diabetes. Chinese medical books written as early as 3000 B.C. spoke of diabetes and described therapies for this disease [94][95]. These historical accounts reveal that T2D existed long ago, and medicinal plants have been used for many millennia to treat this disease. To date, the anti-diabetic activities of well over 1200 traditional plants has been reported, although scant few have been subjected to rigorous scientific evaluation for safety and efficacy in humans [96][97][98][99][100][101]. This section will provide a brief overview of those botanicals used for diabetes that have received the most scientific attention, have been evaluated for their anti-diabetic effects in individuals with diabetes, and are deserving of additional evaluation. The botanicals are presented in the order of their relative (highest-to-lowest) ranking for safety and efficacy.

 

Ipomoea batas (Caiapo)

Caiapo is derived from the skin of a variety of white sweet potato of South American origin, Ipomoea batatas, which is cultivated in a mountainous region in Kagawa Prefecture, Japan. It has been eaten raw for centuries in the belief that it is effective for anaemia, hypertension, and diabetes. Caiapo is commercially available throughout Japan without prescription, and used for the prevention and treatment of T2D. In rodents, caiapo exhibits anti-diabetic activity, and the active component is thought to be a high-molecular weight acid glycoprotein [102][103].

 

A beneficial effect of caiapo on glycemic control has been reported in several clinical studies. In a pilot study, a total of 18 male patients with T2D (age: 58 ± 8 years; weight: 88 ± 3 kg; BMI: 27.7 ± 2.7 kg/m2; means ± SEM) treated by diet alone were randomized to receive placebo (n = 6) or 2 (low dose; n = 6) or 4 g (high dose; n = 6) caiapo (four tablets each containing 168 or 336 mg powdered white-skinned sweet potato, respectively) before breakfast, lunch, and dinner for 6 weeks [104]. At the end of treatment, no statistically significant changes in fasting glucose, insulin, or lipids occurred in either the low-dose caiapo or placebo groups. In the group treated with the high dose caiapo, fasting plasma glucose (-13%) as well as cholesterol [total (-10.5%) and LDL (-13%) cholesterol] were significantly decreased (P < 0.05, compared to baseline). Body weight and blood pressure remained unchanged in all three groups. In patients receiving low-dose caiapo, insulin sensitivity (SI) increased by 37% (2.02 ± 0.70 vs. 2.76 ± 0.89 104 min-1 · μU-1 · ml-1, P < 0.05); in those on high-dose caiapo, the FSIGT demonstrated an increase of SI by 42% (1.21 ± 0.32 vs. 1.73 ± 0.40 104 min-1 · μU-1 · ml-1, P < 0.03). No changes were seen for SI in patients receiving placebo (1.52 ± 0.28 vs. 1.35 ± 0.21 104 min-1 · μU-1 · ml-1). Glucose tolerance was significantly increased (~72%; P < 0.02) in the high-dose group [105].  No adverse events were reported.

 

The results of this pilot study have been confirmed in a randomized, double-blind placebo controlled trial [106]. A total of 61 patients with T2D treated by diet were given 4 g caiapo (n = 30; mean age 55.2 ± 2.1 years; BMI 28.0 ± 0.4 kg/m2) or placebo (n = 31; mean age 55.6 ± 1.5 years; BMI 27.6 ± 0.3 kg/m2) once daily for 12 weeks. Each subject underwent a 75-g oral glucose tolerance test (OGTT) at baseline and after 1, 2, and 3 months to assess 2-h glucose levels. Additionally, fasting blood glucose, HbA1C, total cholesterol, and triglyceride levels were measured. Fasting blood glucose levels decreased in the caiapo group (143.7 ± 1.9 vs. 128.5 ± 1.7 mg/dl; P < 0.001), and remained unchanged in the placebo group (144.3 ± 1.9 vs. 138.2 ± 2.1 mg/dl; P = 0.052). In the caiapo group, HbA1C decreased significantly (-0.53%; P < 0.001) from 7.21 ± 0.15 to 6.68 ± 0.14%, and remained unchanged in the placebo group (7.04 ± 0.17 vs. 7.10 ± 0.19%; P = 0.23). Two-hour glucose levels were significantly (P < 0.001) decreased in the caiapo group (193.3 ± 10.4 vs. 162.8 ± 8.2 mg/dl) compared with the placebo group (191.7 ± 9.2 vs. 181.0 ± 7.1 mg/dl). Mean cholesterol at the end of the treatment was significantly lower in the caiapo group (214.6 ± 11.2 mg/dl) than in the placebo group (248.7 ± 11.2 mg/dl; P < 0.05). A decrease in body weight was observed in both the placebo group (P = 0.0027), and in the caiapo group (P < 0.0001); in the caiapo group, body weight was related to the improvement in glucose control (r = 0.618; P < 0.0002). No significant changes in triglyceride levels or blood pressure were observed. Unfortunately, possible effects on fasting insulin, C-peptide, or insulin sensitivity were not reported in this study. Caiapo was well tolerated without significant adverse effects. This study confirms the results of the pilot with regard to the beneficial effects of caiapo on short-tem glycemic control (as well as cholesterol levels) and documents the efficacy of caiapo on long-term glycemic control in patients with T2D. The magnitude of the effect of caiapo at reducing HbA1c (-0.53% absolute decrease) is comparable with that of Acarbose, an approved oral anti-hyperglycemic medication [107], although it falls short of what has been suggested as the minimal acceptable level for a new anti-hyperglycemic medication (-0.7% absolute decrease) [108]. It will be of interest and important to see if the results of this very exciting outcome with caiapo can be confirmed in other well designed clinical trails, and to establish that caiapo does not exhibit any adverse interaction with existing anti-hyperglycemic medication.

 

Trigonella foenum-graecum (Fenugreek)

Trigonella foenum-graecum, also known as fenugreek, is an herb native to southeastern Europe, northern Africa, and western Asia, but is also widely cultivated in other parts of the world [109]. Fenugreek has a long history of traditional use in both Ayurvedic [holistic system of healing which originated among the Brahmin (Hindu priestly caste) sages of ancient India and Nepal approximately 3000 - 5000 years ago] and Chinese medicine [110], and has been widely used for the treatment of diabetes [111]. The defatted seeds of the fenugreek plant contain ~50% fiber (similar to guar gum), along with a variety of bioactive saponins, alkaloids, coumarins, and 4-hydroxyisoleucine, the principle bioactive compound [112][113]. This latter compound exhibits insulinotropic activity [114][115][116]; following a 6-day sub-chronic administration, 4-hydroxyisoleucine (50 mg/kg/day) reduced fasting hyperglycemia, insulinemia, and improved glucose tolerance in diabetic rats [117].

 

The clinical studies that have evaluated the efficacy of fenugreek in individuals with both type 1 and T2D have recently been reviewed [118]. In one study, 17 of 21 patients with T2D showed a reduction in 2-hour post-prandial glucose averaging 30 mg/dl following administration of 15 g of ground fenugreek seed [119]. In a crossover, placebo-controlled trial with 60 individuals with type 2 diabetes, the treatment group received 12.5 mg defatted fenugreek at lunch and dinner with isocaloric diets for 24 weeks [120]. Fasting blood glucose was 151 mg/dl at baseline, and reduced to 112 mg/dl after 24 weeks (P < 0.05). Fenugreek also caused a significant decrease in the area under the glucose curve by approximately 40%.

 

In a small study, the effects of fenugreek seeds on glycemic control and insulin resistance in mild-to-moderate T2D was performed using a double-blind, placebo-controlled design [121]. Twenty-five newly diagnosed patients with T2D (fasting glucose < 200 mg/dl) were randomly divided into two groups. Group I (n = 12) received 1 g/day of fenugreek seeds and Group II (n = 13) received standard care (dietary control, exercise) plus placebo capsules for two months. After two months, fasting blood glucose and two-hour post-glucose load blood glucose were not significantly different. However, following an oral glucose challenge, the area under the curve (AUC) for blood glucose (2375 ± 574 vs 27597 ± 274) and insulin (2492 ± 2536 vs. 5631 ± 2428) was significantly lower (P < 0.001), as was the HOMA-IR index (112.9 ± 67 vs 92.2 ± 57; P < 0.05) in the treatment group compared to control. Serum triglycerides were decreased and HDL cholesterol increased significantly (both P < 0.05) following fenugreek treatment. Significant reductions in total cholesterol and triglycerides have also been reported in other studies following fenugreek treatment [122].

 

In an open label pilot study, fenugreek seeds were evaluated in combination with M. charantia and jamun seeds (Syzigium cumini) for effects on glycemic control in patients with T2D [123]. The patients were divided into two groups of 30 each. The patients of group I were given the raw powdered mixture in the form of capsules; the patients of group II were given this mixture in the form of salty biscuits. Daily supplementation of 1 g of this powered mixture for a 1.5-month period and increased to 2 g for another 1.5 months significantly reduced the fasting as well as the postprandial glucose level of the diabetic patients. A significant decrease in oral hypoglycemic drug intake and decline in percentage of the subjects who were on hypoglycemic drugs were found after the 3-month feeding trial. The authors concluded that 2 g of this powdered mixture of traditional medicinal plants in either raw or cooked form can be successfully used for lowering blood glucose in diabetics.

 

The doses of fenugreek used in clinical studies have ranged from 2.5 grams to 15 grams daily of the crushed and defatted seeds. Crushing is important in order to release the viscous gel fiber, which presumably contributes to the efficacy of fenugreek. Typical doses of seeds are in the range of 1-3 grams mixed with food and taken at mealtime. The most common side effects are gastrointestinal upset (diarrhea and flatulence), which often can be alleviated by dose-titration. Since the fenugreek fiber might absorb other oral medications, fenugreek should be taken independently (e.g. 1-2 h) of other medications. Due to its ability to lower blood glucose, individuals should monitor their glucose levels carefully if used in combination with insulin or other glucose-lowering agents. Fenugreek can exhibit anti-coagulant activity; it should not be used with other anti-coagulating agents due to the increased risk for bleeding.

The potential genotoxicity of a fenugreek seed extract (THL), containing a minimum of 40% 4-hydroxyisoleucine, was evaluated using the panel of assays recommended by US Food and Drug Administration for food ingredients (i.e. reverse mutation assay; mouse lymphoma forward mutation assay; mouse micronucleus assay). THL was determined not to be genotoxic under the conditions of the tested genetic toxicity battery [124].

 

Cinnamomum cassia (Cinnamon)

Anderson and colleagues have previously reported that an ammonium hydroxide extract of cinnamon potentiated the effect of insulin on glucose oxidation in isolated rat adipocytes [125][126]. Close evaluation of these in vitro data reveals that the cinnamon extract actually possessed insulin mimetic activity, since the stimulatory effects were independent of insulin concentration and no added insulin was required to achieve maximal glucose oxidation [127]. A water-soluble polyphenolic polymer from cinnamon has been isolated, and shown to have insulin-like activity as well as antioxidant activity in vitro [128]. Additionally, enzyme inhibition studies done have shown that the bioactive compound(s) isolated from cinnamon can stimulate autophosphorylation of a truncated form of the insulin receptor and can inhibit PTP-1, the rat homolog of the tyrosine phosphatase PTP-1B that inactivates the insulin receptor [129][130].

Three small studies have evaluated the use of cinnamon in T2D. The effects of cinnamon on blood glucose, triglyceride, total cholesterol, HDL cholesterol, and LDL cholesterol levels was evaluated in patients with T2D [131]. A total of 60 people with T2D (30 men and 30 women aged 52.2 ± 6.32 years) were divided randomly into six groups. Groups 1, 2, and 3 consumed 1, 3, or 6 g of cinnamon daily, respectively, and groups 4, 5, and 6 were given placebo capsules corresponding to the number of capsules consumed for the three levels of cinnamon. The cinnamon was consumed for 40 days, followed by a 20-day washout period. After 40 days, all three doses of cinnamon significantly reduced the mean fasting serum glucose (18-29%), triglyceride (23-30%), LDL cholesterol (7-27%), and total cholesterol (12-26%) levels; no significant changes were noted in the placebo groups. Changes in HDL cholesterol were not significant, and no significant adverse events were reported. The results of this interesting study suggest that intake of 1, 3, or 6 g of cinnamon per day reduces serum glucose, triglyceride, LDL cholesterol, and total cholesterol.

 

A more recent trial has evaluated an aqueous-purified cinnamon extract on long-term glycemic control and lipids in patients with T2D with equivocal results [132]. A total of 79 patients on insulin therapy but treated with oral anti-diabetics or diet were randomly assigned to take either cinnamon extract or a placebo capsule three times a day for 4 months in a double-blind study. The amount of aqueous cinnamon extract corresponded to 3 g of cinnamon powder per day. The mean absolute and percentage differences between the pre- and post-intervention fasting plasma glucose level of the cinnamon and placebo groups were significantly different. There was a significantly higher reduction in the cinnamon group (10.3%) than in the placebo group (3.4%). No significant intra-group or inter-group differences were observed regarding HbA1c, lipid profiles or differences between the pre- and post-intervention levels of these parameters The decrease in plasma glucose correlated significantly with the baseline concentrations, indicating that subjects with a higher initial plasma glucose level exhibited a greater response more from cinnamon intake [133]. No adverse effects were observed.

 

In a third study, a total of 25 postmenopausal patients with T2D (aged 62.9 ± 1.5 y, BMI 30.4 ± 0.9 kg/m2) participated in a 6-wk intervention, during which they were supplemented with either cinnamon (1.5 g/d) or a placebo [134]. Before and after 2 and 6 wk of supplementation, arterialized blood samples were obtained and oral glucose tolerance tests were performed. Blood lipid profiles and multiple indices of whole-body insulin sensitivity were determined. There were no time x treatment interactions for whole-body insulin sensitivity or oral glucose tolerance. The blood lipid profile of fasting subjects did not change after cinnamon supplementation. These results showed that cinnamon supplementation (1.5 g/d) did not improve whole-body insulin sensitivity or oral glucose tolerance, and did not modulate blood lipid profile in postmenopausal patients with T2D. Thus, the ability of cinnamon to improve long-term glycemic control has yet to be confirmed.

 

Mormordica charantia (Bitter melon)

Mormordica charantia is reported to be the most popular plant used worldwide to treat diabetes [135][136]. It has many names depending on the geographic location of origin: in India, where it is widely used for diabetes [137], M. charantia is known as karela, bitter melon, and bitter gourd. In other parts of the world, it is also known as wild cucumber, ampalaya, and cundeamor [138]. The glucose-lowering activity of M. charantia (administered as both fresh juice and unripe fruit) has been well documented in animal models of diabetes [139][140]. Compounds possessing anti-diabetic activity include charantin and vicine [141][142][143]. In addition, other bioactive components of bitter melon extract appear to have structural similarities to animal insulin (e.g. polypeptide-p) [144][145]. Several modes of action have been proposed to account for the anti-diabetic activity of M. charantia including inhibition of glucose absorption in the gut, stimulation of insulin secretion, and the stimulation of hepatic glycogen synthesis [146][147]. Four clinical trials have reported bitter melon juice, fruit, and dried powder to have a moderate hypoglycemic effect [148][149][150]. However, these studies were small and were not randomized or double-blind, and of insufficient quality to recommend the use of M. charantia without careful monitoring. When used at typical doses, 300-600 mg of juice extract or 1-2 g of powdered leaf daily, M. charantia is generally well tolerated, although it should not be used by children or pregnant women [151][152].

 

Gymnema sylvestre (Gurmar)

Gymnema sylvestre (also called gurmar), a woody plant that grows wild in India, has a long history of use in Ayurvedic medicine [153]. The leaves of G. sylvestre contain glycosides and the peptide gurmarin [154][155]. Other plant constituents include resins, gymnemic acids, saponins, stigmasterol, quercitol, and several amino acid derivatives. A water-soluble acidic fraction called GS4 has been used in most clinical studies (see below). Several modes of action have been proposed to account for the anti-diabetic activity of G. sylvestre including increased glucose uptake and utilization, increased insulin secretion, and increased β-cell number [156].

There have been several small clinical studies in individuals with type 1 and T2D (reviewed in [157]). These studies have reported that G. sylvestre decreases fasting glucose and HbA1c, lowered insulin requirements in individuals with type 1 diabetes, and lowered the dose of anti-hyperglycemic medications in individuals with T2D. G. sylvestre also appears to facilitate endogenous insulin secretion, but it is not a substitute for insulin. There are no data from double-blind, placebo-controlled studies in humans that validate the efficacy of G. sylvestre in type 1 or type 2 diabetes. The extract (~400 mg daily) of G. sylvestre is well tolerated, and no significant side effects have been reported.

 

Opuntia (fuliginosa, streptacantha) (Prickly pear cactus; Nopal)

Nopal, a member of the Opuntia genus, is widely used in Mexico as a treatment for glucose control [158]. It grows in arid regions throughout the Western hemisphere, and is know in the United States as prickly pear cactus. This plant produces both a vegetable, called nopal, and a red egg-shaped fruit called tuna. Nopal is a common component of every day foods including soups, salads, sandwiches, and blended in drinks. When used for glucose control, nopal is prepared as a food, and is available as in bulk, dried powder form or in capsules. The glucose-lowering activity of nopal is likely due to its very high soluble fiber and pectin content [159][160][161], although its ability to reduce fasting glucose is suggestive of additional modes of action [162]

 

The results of most human studies of this plant have been reported in Spanish-language journals [163][164]; two studies evaluating the acute effects of nopal have been published in English by Frati et al [165][166]. Both studies used Opuntia streptacantha Lemaire, and reported improved glycemic control (decreased serum glucose) and improved insulin sensitivity (decreased serum insulin) following a single-dose (500 g of broiled or grilled nopal stems) in patients with type 2 diabetes (n= 14 and n = 22). No effect was observed in healthy individuals [167], nor were any adverse effects reported. The potential clinical efficacy of this plant warrants further study.

 

Coccinia indica

Coccinia indica is a creeper plant (one that spreads by means of stems that creep) that grows wildly in Bangladesh and in many other parts of the Indian sub-continent. Although C. indica has a long history of use as an antidiabetic treatment in Ayurvedic medicine [168], it has not been subjected to the number of clinical trials that have evaluated M. charantia, fenugreek, or G. sylvestre (based on published reports). Neither the bioactive compounds nor mode of action C. indica have been well-characterized, but there is some suggestion that a component(s) of the plant possesses insulin-mimetic activity [169].

 

A double blind, placebo-controlled trial in which a preparation from the leaves of the plant was administered to patients with uncontrolled T2D for 6 weeks [170]. Of the 16 patients who received the experimental preparations, 10 showed significant improvement in their glucose tolerance (P < 0.001), while none out of the 16 patients in the placebo group showed improvement. Several other studies also offered supporting evidence of the beneficial effect of this treatment (reviewed in [171]). No adverse effects were reported. The potential clinical efficacy of this plant warrants further study.

 

Panax (ginseng, japoncicus, quinquefolius, eleutherococcus)

Ginseng, a member of the plant family Aaraliaceae, has been used in traditional Chinese medicine for thousands of years [172][173][174]. The botanical names for Asian ginseng (Chinese or Korean) is Panax ginseng; Japanese ginseng is known as Panax japonicus; American ginseng is Panax quinquefolius. Siberian ginseng belongs to the genus Eleutherococcus. Many therapeutic claims have been ascribed to the use of ginseng root extract including improved vitality and immune function, along with beneficial effects on cancer, diabetes, cardiovascular disease, and sexual function [175]. Bioactive compounds that have been identified in ginseng species include ginsenosides, polysaccharides, peptides, and fatty acids [176][177]. Most pharmacological actions are attributed to the ginsenosides, a family of steroids named saponins [178][179]. A comprehensive review of the randomized controlled trials (RCTs) evaluating ginseng extracts (mostly Panax ginseng and Panax quinquefolius) has been performed, and it was concluded by the authors that there was insufficient evidence to support efficacy for any of the above indications [180]. However, in light of the extreme compositional variability of ginsenoside concentrations reported in a sampling of 32 studies, it is possible that the anti-hyperglycemic efficacy might be as variable as its ginsenoside content [181][182].

 

Two small studies have evaluated the use of ginseng in diabetes. In a double-blind, placebo controlled study, 36 patients with T2D were treated for 8 weeks with ginseng extract (species not specified) at 100 (n =12) or 200 (n = 12) mg/day, or with a placebo (n =12) [183]. Ginseng (100 and 200 mg/day) but not placebo lowered fasting blood glucose by approximately 0.5-1 mmol/l (P < 0.05). Eight subjects who were given ginseng and two who were given placebo achieved normal fasting blood glucose. In response to an oral glucose challenge, the area under the 2-h blood glucose curve was reduced approximately 16% (P < 0.001) in the eight ginseng-treated patients who had normalized fasting blood glucose, without any concomitant change in immunoreactive insulin or C-peptide. The 200 mg dose improved HbA1c (~0.5% decrease; P < 0.05) and physical activity compared to placebo. Ginseng had no effect on plasma lipids. Another small study reported the acute effects of Panax quinquefolius administration (single dose on four separate occasions; 3 g/treatment) or placebo on glucose tolerance in patients with T2D (n = 9) and in non-diabetic subjects (n = 10) [184]. In both groups, ginseng caused a significant reduction (P < 0.05) in the area under the blood glucose curve by approximately 20% compared to placebo. The potential clinical efficacy of this plant warrants further study using a standardized extract.

 

Aloe vera

The dried sap of the aloe plant (aloes) is a traditional botanical remedy frequently used to treat dermatitis, burns and to enhance wound healing [185], and one of a variety of plants used for diabetes in India [186] and the Arabian peninsula [187]. Its ability to lower the blood glucose was studied in 5 patients with type 2 diabetes [188]. Following the ingestion of aloe (one-half a teaspoonful daily for 4-14 weeks), fasting serum glucose level decreased in every patient from a mean of 273 ± 25 (SE) to 151 ± 23 mg/dl (P < 0.05) with no change in body weight. This glucose-lowering activity has been confirmed in two other studies which reported that oral administration of the aloes juice (1 tablespoon twice daily) reduced fasting glucose and triglycerides in subjects with type 2 diabetes both in the absence and presence of concomitant sulfonylurea therapy [189][190]. No adverse effects were reported in these studies. Aloe gel also holds the potential for glucose-lowering activity, as it contains glucomannan, a water soluble fiber that reportedly has glucose-lowering and insulin sensitizing activities [191][192]. The potential clinical efficacy of this plant warrants further study.

 

Allium (sativum and cepa) (Garlic)

Allium sativum (garlic) has been used as a medicinal herb by the ancient Sumarians, Egyptians, Greeks, Chinese, Indians, and later the Italians and English [193]. The leading Indian ancient medical text, Charaka-Samhita recommended garlic for the treatment of heart disease and arthritis for over many centuries [194]. Compounds present in aqueous garlic extract or raw garlic homogenate though to be the principle bioactive components include allicin (allyl 2-propenethiosulfonate or diallyl thiosulfonate), allyl methyl thiosulfonate, 1-propenyl allyl thiosulfonate, and γ-L-glutamyl-S-alkyl-L-cysteine.

 

In modern times, garlic preparations have been widely recognized as agents for prevention and treatment of cardiovascular and other metabolic diseases, atherosclerosis, hyperlipidemia, thrombosis, hypertension and diabetes [195][196][197]. Epidemiological evidence indicates an inverse correlation between garlic consumption and the reduced risk of the development of cardiovascular disease [198][199][200]. The efficacy of garlic in cardiovascular diseases has been more evident in non-clinical models, thus prompting a variety of clinical trials [201]. Many of these trials have reported beneficial effects of garlic on almost all cardiovascular conditions mentioned above [202]; however, a number of studies have reported no beneficial effects, casting doubt on the reputed health benefits of garlic. These differences could have arisen as a result of methodological shortcomings, the use of different formulations/preparations of garlic, heterogeneity of patient populations, dietary inconsistencies, and different time scales of the studies. The glucose-lowering activity of garlic in humans with type 2 diabetes is not well studied. In the studies that have evaluated garlic, the data have been conflicting [203][204]. Thus, the role of garlic in glucose control has yet to be confirmed.

 

Resveratrol

It has long been known that the French consume a much greater amount of saturated fats in their diet, yet suffer from a lower incidence of cardiovascular disease. This phenomenon has been termed ‘The French Paradox’. It has been proposed that the greater amount of red wine consumed by the French protects them against the development of cardiovascular disease[205][206][207]. The intensive search for the actual molecule(s) that could be responsible for this almost miraculous activity led researchers to a compound identified as resveratrol. Resveratrol is a polyphenolic compound (a stillbenol) found in plants, especially red grapes and peanuts[208]. Red wine, produced from red grapes, contains the highest amount, on a percentage basis, of resveratrol ranging from 0.1 – 14.3 mg per liter. The more resveratrol is investigated, the more its diverse health benefits emerge. In animals, resveratrol has been associated with anti-cancer activity, cardioprotective activity, antioxidant and glutathione-sparing activities, anti-inflammatory activity, anti-viral activity, and anti-neurodegenerative activity[209][210][211]. But without a doubt, the activities that affords resveratrol the most notoriety, is its ability to increase lifespan and delay age-related deterioration in a variety of experimental models[212][213]. In experiments using yeast, nematodes (roundworms), fruit flies, and fish, resveratrol has been shown to increase mean lifespan by 18-70%, and maximum lifespan by 15-66%. While resveratrol has not yet been shown to increase lifespan in mammals, it has been consistently shown in mice to delay age-related diseases, improve metabolic efficiency, including a reduction in body weight and blood glucose, and largely mimic the biochemical and physiologic effects of caloric restriction, qualifying it, to a degree, as a caloric restriction mimetic[214][215].   

 

What does resveratrol do at the mechanistic level that has led to its being the most widely recognized polyphenolic? The striking similarities of the benefits of caloric restriction and resveratrol led researchers to evaluate the effects of resveratrol on SIRT1 activation. These experiments have conclusively shown that resveratrol does, indeed, activate SIRT1 and its signaling partner, PGC-1a[216][217][218]. While there might be (and are) other important molecular targets of resveratrol including AMPK[219], none are more important than SIRT1 and PGC-1a. In light of the critical roles played by SIRT1 and PGC-1a in mitochondrial biogenesis, the obvious question is what does resveratrol do, in this regard? Treatment of mice with resveratrol significantly increased their aerobic capacity, judged by their increased running time and oxygen consumption in their muscles[220]. Resveratrol induced the genes for oxidative phosphorylation and mitochondrial biogenesis, effects largely explained by the resveratrol-mediated decrease in PGC-1a acetylation and an increase in PGC-1a activity. This mechanism is consistent with resveratrol being a known activator of SIRT1, and by the lack of effect of resveratrol in cells lacking SIRT1. Importantly, resveratrol treatment protected mice against diet-induced-obesity and insulin resistance[221][222][223]. If there was any doubt, these results obtained with resveratrol clearly implicate SIRT1 and PGC-1a as key regulators of energy and metabolic homeostasis. In fact, resveratrol and several related resveratrol analogs are now being developed as potential new therapies for type 2 diabetes[224].

   

TRADITIONAL  CHINESE MEDICINE

A discussion of botanical interventions for T2D would not be complete without the thoughtful consideration of Traditional Chinese Medicine. Over the centuries, Chinese herbal ‘drugs’ have served as a primary source for the prevention and treatment of many diseases including diabetes. Xiao-ke is a term used to refer to wasting and thirsting syndrome (T1D); the more modern term, Tang-niao-bing, means ‘sugar urine illness’ (T2D)[225]. Unfortunately, a review of Traditional Chinese Medicine in the context of T2D is beyond the scope of this chapter. However, a number of excellent reviews on this subject are available, and will provide the reader with a more definitive evaluation of this subject than what could be accomplished in this presentation [226][227][228][229].

 

 

NON-BOTANICAL INTERVENTIONS

Oxidative stress, resulting primarily from chronic hyperglycemia, is a major cause of the complications of diabetes [230]. More recently, there is a growing appreciation of the role of oxidative stress as a mediator of insulin resistance and β dysfunction [231][232][233]. In this context, there are a growing number of studies in humans that have reported beneficial effects of antioxidants on various measures of abnormalities of diabetes [234][235][236][237]. In addition, it is often reported that individuals with diabetes are deficient in one or more essential micronutrients, and that supplementation often provides a significant improvement. This section will provide a concise overview of those antioxidants, vitamins, minerals, and other dietary supplements (collectively referred to here as nutraceuticals) that have received the most attention as potential adjunct treatments for diabetes.

 

Antioxidants and Vitamins

 

α-Lipoic Acid

α-Lipoic acid (LA) is an eight-carbon fatty acid that is synthesized in trace quantities in organisms ranging from bacteria to man [238][239][240]. LA functions naturally as a cofactor in several mitochondrial enzyme complexes responsible for oxidative glucose metabolism and cellular energy production [241][242]. LA has been prescribed in Germany for over thirty years for the treatment of diabetes-induced neuropathy [243][244][245]. Results from several controlled clinical studies indicate that this compound is safe, well tolerated, and efficacious when administered intravenously [246][247][248][249]. Results from a recently published study indicate that LA is equally efficacious at improving symptoms of diabetic neuropathy when administered orally [250]. New data obtained in animal models suggest that LA might be useful as an anti-obesity agent [251]. LA is commercially available in the US as a nutraceutical (dietary supplement).

 

In addition to the beneficial effects of LA on diabetes-induced neuropathy, several clinical studies have reported an improvement in insulin sensitivity and whole-body glucose metabolism in patients with type 2 diabetes after continuous intravenous (iv) infusion of LA [252][253][254][255]. Investigators have reported that a continuous infusion iv of LA substantially increases insulin-mediated glucose disposal (~30-50%) [256][257]. Oral administration of LA (enteric-coated tablet) exerts a smaller (~20%) but nonetheless significant effect on insulin sensitivity [258][259]. To overcome the abbreviated half-life of LA in plasma, a controlled release formulation of LA (CRLA) has been recently developed [260]. The pharmacokinetics, safety, and tolerability of CRLA were evaluated in healthy individuals and in patients with type 2 diabetes, and this agent was found to be safe, well-tolerated, and significantly reduced plasma fructosamine in patients with type 2 diabetes [261]. Also, non-controlled release LA recently has been reported to increase insulin mediated glucose disposal in patients with type 2 diabetes [262].

 

Although the exact mechanism of action of LA is unknown, in vitro data from the laboratories of Rudich and others have indicated that LA pretreatment maintains the intracellular level of reduced glutathione (the major intracellular antioxidant) in the presence of oxidative stress, and blocks the activation of serine kinases that could potentially mediate insulin resistance [263][264][265][266]. Thus, one potential explanation for the protective effects of LA might be related to its ability to preserve the intracellular redox balance (acting either directly or through other endogenous antioxidants such as glutathione), thereby blocking the activation of inhibitory stress-sensitive serine kinases including IKKβ [267]. This stress-sensitive kinase is a crucial regulator of the transcription factor nuclear factor-κB (NF-κB), a major target of hyperglycemia, cytokines, reactive oxygen species, and oxidative stress [268][269][270]. The aberrant regulation of NF-κB is associated with a number of chronic diseases including diabetes and atherosclerosis [271][272]. The ability of LA to block the activation of NF-κB is well established in vitro and in vivo [273][274][275][276][277].

 

Recent evidence has linked the activation of NF-κB with insulin resistance [278][279]. Activation of IKKβ inhibits insulin action. Salicylates, which inhibit IKKβ activity and block NF-κB activation [280], restore insulin sensitivity both in vitro and in vivo [281][282]. Treatment of nine patients with type 2 diabetes for two weeks with high-dose aspirin (7 g/day) resulted in a significant reduction in hepatic glucose production and fasting hyperglycemia, and increased insulin sensitivity [283]. The potential for toxicity associated with such a high dose of salicylate administered chronically precludes consideration of this agent for therapy, but the results support the rationale that IKKβ inhibition could be a useful pharmacological approach to increase insulin sensitivity. Furthermore, LA and other agents that interfere with the persistent activation of the NF-κB pathway appear to be promising approaches to increase insulin sensitivity, and perhaps even as treatments for complications of diabetes in which NF-κB activation has been implicated [284][285].

 

L-Arginine

L-Arginine is classified as a ‘semi-essential’ amino acid utilized by all cells [286][287][288]. It plays a critical role in cytoplasmic and nuclear protein synthesis, biosynthesis of other amino acids and derivatives, and in the urea cycle. In this essential biochemical pathway, urea is synthesized from arginine to enable the body to remove excess ammonia, which is toxic to cells. L-arginine is classified a glucogenic amino acid because it can be metabolized into α-ketoglutarate, and enter the citric acid cycle (Kreb’s Cycle). In one of its most important roles, L-arginine serves as a direct precursor for the biosynthesis of NO [289]. Although this reaction was originally discovered to occur in endothelial cells, the generation of NO from L-arginine occurs in a variety of other cell types including skeletal muscle [290][291][292]. NO is produced endogenously from L-arginine in a complex reaction that is catalyzed by the enzyme nitric oxide synthase (NOS). The other product that is formed in this reaction is citrulline. NO serves as a second messenger to trigger blood vessel dilation and increase blood flow. L-arginine is the only physiological substrate that the NOS enzymes use as a nitrogen donor. Thus, under certain conditions, it may be rate limiting for NO production.

 

It is well established that aging leads to the deterioration of the vasculature and increased risk for cardiovascular disease [293][294][295]. Circulatory diseases account for considerable morbidity and almost half of all deaths in people over the age of 75 years. A major abnormality of the vasculature present in individuals with type 2 diabetes is endothelial dysfunction, or reduced blood flow capacity. As discussed above, NO is a major regulator of the blood flow. Basal release of NO from the vascular endothelium maintains a constant vasodilating tone. Impaired NO-mediated vasodilatation has been described in hypertension, diabetes, cardiovascular disease, and aging [296][297].

 

In atherosclerosis, the endothelium has a reduced capacity to produce NO and target cells are relatively insensitive to it [298]. The ability of NO to cause vasodilation provides an explanation for the mechanism of action of nitroglycerin, which has been used for over 100 years to treat patients with angina (pain due to inadequate blood flow to the heart) [299]. NO is produced following administration of nitroglycerin and other NO donors, such as L-arginine [300][301]. In particular, L-arginine is a substrate for NOS, which is responsible for the endothelial production of NO.

 

Therefore, many investigators have evaluated the usefulness of L-arginine supplementation in animals and in humans in increasing NO production and improving cardiovascular health. The results of these studies have been summarized in several recent books [302][303] and a review [304]. Results of oral L-arginine supplementation in hypercholesterolemic animals have consistently shown beneficial effects. L-arginine appears to inhibit the progression of atherosclerotic plaques and preserve endothelial function [305]. In addition, L-arginine affects other mediators of atherosclerosis, including circulating inflammatory cells and platelets [306].

 

On balance, the data in humans have also been positive, although more variable [307][308][309][310][311][312][313][314][315][316][317][318]. This variability is likely due to heterogeneous subject populations with a variety of non-standardized clinical symptoms, small sample sizes, abbreviated duration of treatment, and sub-therapeutic treatment doses. Five of the 17 studies showed no cardiovascular health benefit from oral L-arginine supplementation [319]. The remaining 12 studies demonstrated beneficial effects as evidenced by decreased platelet aggregation and adhesion, decreased monocyte adhesion, or improved endothelium-dependent vasodilation [320]. Taken together, these studies provide supporting evidence for the idea that treatment with an exogenous NO donor could have a beneficial effect on cardiovascular health.

 

Vitamin C

Epidemiological evidence suggests that a high dietary intake of vitamin C, a marker of fruit and vegetable intake, is associated with a reduced risk for the development of cardiovascular disease [321][322]. Plasma vitamin C, fruit intake, and dietary vitamin C intake is significantly and inversely associated with mean concentrations of C-reactive protein and tissue plasminogen activator (t-PA) antigen, a marker of endothelial dysfunction, suggesting that vitamin C has anti-inflammatory effects and is associated with lower endothelial dysfunction in men with no history of cardiovascular disease or diabetes [323][324]. However, a high vitamin C intake from supplements has been reported to be associated with an increased risk of cardiovascular disease mortality in postmenopausal women with diabetes [325].

The normal functions of vascular endothelial tissue include regulation of vasomotor tone, inhibition of platelet activity, and regulation of recruitment of inflammatory cells into the vasculature [326]. A damaged endothelium (‘endothelial dysfunction’) is a key event in the development of diabetic macroangiopathy, and is associated with the oxidative stress-mediated blunting of nitric oxide action [327][328][329]. Endothelial dysfunction has been documented in individuals who are insulin resistant, and in those at risk for developing T2D [330][331][332].

 

The data as to whether vitamin C treatment (either acute or sub-chronic) exerts a beneficial effect on endothelial dysfunction in individuals with T2D is conflicting. Acute treatment with vitamin C improved endothelial function in obese subjects [333], in patients with type 1 and T2D, and in women with gestational diabetes [334][335][336].

 

In patients with cardiovascular disease including endothelial dysfunction, both acute (single dose, 2 g) and chronic treatment with vitamin C (30 days, 500 mg/d) reverses the vasomotor defect, as judged by increased flow-mediated dilation of the brachial artery [337][338]. All of the above studies involved relatively small populations (< 75) and used acute treatment except one, which was for 30 days [339]. Nonetheless, the persistent finding of a beneficial effect of antioxidant treatment on endothelial function (flow-mediated dilation) in individuals with demonstrated endothelial dysfunction is encouraging. It is likely that these results will stimulate additional clinical studies of larger size and longer duration to evaluate the efficacy of vitamin C and perhaps other antioxidants.

 

In addition to playing a major role in the etiology of diabetic macroangiopathy, endothelial dysfunction could promote insulin resistance [340]. It is possible that oxidative stress-mediated blunting of nitric oxide action indirectly affects insulin sensitivity (e.g. reduced peripheral blood flow, increased peroxynitrite formation, others) consequently reducing insulin-stimulated glucose transport in skeletal muscle.

 

Cigarette smoking impairs endothelial function, and is one of the major risk factors for hypertension, atherosclerosis, and coronary heart disease. The effects of vitamin C (infusion) on insulin sensitivity and endothelial function (measured by flow-mediated dilation of brachial artery; FMD) were evaluated in smokers, non-smokers with impaired glucose tolerance, and non-smokers with normal glucose tolerance [341]. Both insulin sensitivity and FMD were blunted in smokers and nonsmokers with IGT, compared with controls. In smokers and in non-smokers with impaired glucose tolerance, vitamin C significantly improved FMD, increased insulin sensitivity, and decreased plasma thiobarbituric acid-reactive substances, an index of oxidative stress. In contrast, vitamin C had no effect on these parameters in non-smokers with normal glucose tolerance. In patients with coronary spastic angina and endothelial dysfunction, vitamin C infusion augmented FMD and increased insulin sensitivity [342]. In contrast, vitamin C had no effect in healthy controls.

 

More recent studies have reported that vitamin C treatment significantly increased forearm vasodilatory response to reactive hyperemia only in patients with combined T2D and cardiovascular disease [343], and improved endothelial dysfunction and attenuated post-prandiallipemia-induced oxidative stress in subjects with T2D [344].

 

To test the hypothesis that the vitamin C influences microcirculatory function in patients with T2D, subjects were treated with 1 g of vitamin C three times a day for 2 weeks in a randomized placebo-controlled double-blind cross-over design. Microvascular reactivity was assessed by vital capillaroscopy and post-occlusive reactive hyperaemia. hs-CRP (high-sensitivity C-reactive protein), IL-6 (interleukin-6), IL-1ra (interleukin-1 receptor antagonist), and ox-LDL (oxidized low-density lipoprotein) were analyzed. The results showed no significant change in microvascular reactivity assessed after 2 weeks of vitamin C treatment. IL-1ra, IL-6, hs-CRP and ox-LDL did not change significantly, neither as absolute or relative values. In conclusion, in contrast with some studies reported previously, this study did not demonstrate an effect of continuous oral treatment with vitamin C on microvascular reactivity assessed at the level of individual capillaries, nor any indication of an effect on inflammatory cytokines or ox-LDL.

 

Using an excellent study design, the effects of high-dose oral vitamin C to alter endothelial dysfunction and insulin resistance in T2D was investigated [345]. Thirty-two diabetic subjects with low plasma vitamin C were enrolled in a randomized, double-blind, placebo-controlled study of vitamin C (800 mg/day for 4 wk). No significant changes in fasting glucose, insulin, SI (determined by glucose clamp), or forearm blood flow in response to ACh, SNP, or insulin were observed after vitamin C treatment. These results indicate that that high-dose oral vitamin C therapy, resulting in incomplete replenishment of vitamin C levels, is ineffective at improving endothelial dysfunction and insulin resistance in T2D.

 

Coenzyme Q10

Another antioxidant reported to have beneficial effects for diabetes is coenzyme Q10.  Coenzyme Q is a vitamin-like molecule that has is frequently used in the treatment of several disorders primarily related to suboptimal cellular energy metabolism and oxidative stress [346]. The effects of orally administered coenzyme Q10 were evaluated in a double-blind, placebo-controlled study of 30 patients with coronary heart disease [347]. Following 8 weeks of treatment with coenzyme Q10 (60 mg twice daily), patients exhibited reduced plasma levels of glucose, insulin, (fasting and 2 hour), and lipid peroxides (a marker of oxidative stress) compared to controls. These results indicate that coenzyme Q10 decreased oxidative stress and improved insulin sensitivity. Additional evaluation of coenzyme Q10 is clearly warranted.

To improve their lipid profiles, many patients with T2D are taking one of the HMG-CoA reductase inhibitors (i.e. statin class of drugs). Statins can reduce serum levels of coenzyme Q10 by up to 40% [348][349]. The logical option is supplementation with coenzyme Q10 as a routine adjunct to any treatment that may reduce the endogenous production of coenzyme Q10, based on a balance of likely benefit against very small risk.

 

Vitamin E

Cardiovascular disease is the leading cause or morbidity and mortality in the Western world, and the major macrovascular complication of diabetes [350]. It is associated with increased oxidative stress [351], and studies both in vitro and in vivo have provided the rationale for numerous prospective clinical studies evaluating the effects of vitamin E (α-tocopherol) on diabetes and cardiovascular events in different populations [352][353](see below). A review of these data by Jialal and colleagues has led to the overall conclusion that four of the five major prospective trials (data reported through 2001) have reported a beneficial effect on cardiovascular end-points, including cardiovascular death, nonfatal myocardial infraction, ischemic stroke, peripheral vascular disease, and others [354]. The one major study (HOPE Study [355]) that was negative for all end-points, had three limitations [356]. It was terminated early due to the overwhelming positive effects of the angiotensin-converting enzyme ramipril, it lacked data on the dietary intake of other antioxidants, and only evaluated synthetic vitamin E (a mixture of tocopherols and tocotrienols) and not α-tocopherol, the most potent and effective tocopherol.

In a study in patients with T2D evaluating the effects of vitamin E on biochemical risk factors for the development of cardiovascular disease, vitamin E treatment significantly reduced low-density lipoprotein oxidizability and soluble cell adhesion molecules [357]. Taken together, the evidence suggests a beneficial effect of vitamin E in patients with pre-existing cardiovascular disease, and in those who are at a greater risk for its development.

 

Oral vitamin E treatment appears to be effective in normalizing abnormalities in retinal hemodynamic, and improving renal function in patients with type 1 diabetes of short (disease) duration [358]. Vitamin E was beneficial in those individuals with poorest glycemic control and the most impaired retinal blood flow [359]. In a well-controlled study, short-term (4 weeks) supplementation of patients with T2D with persistent micro/macroalbuminuria with both vitamins E and C significantly lowered their urinary albumin excretion rate [360]. Four months treatment of patients with T2D with autonomic neuropathy with vitamin E improved the ratio of cardiac sympathetic to parasympathetic tone coincident with lowering of several indices of oxidative stress [361]. Interestingly, the study also reported a lowering of glycated hemoglobin, insulin, norepinephrine, and the homeoststatic model assessment index, indicative of increased insulin sensitivity and glycemic control. These data suggest that vitamin E and perhaps other antioxidant supplementation may provide a benefit in the treatment of microvascular complications of diabetes including diabetic retinopathy or nephropathy.

 

Initial reports of a positive effect of vitamin E on insulin action in insulin resistant patients with T2D were published almost ten years ago [362][363]. Twenty-five patients with T2D were treated with vitamin E (d-α-tocopherol; 900 mg/day) or placebo for three months in a double-blind, crossover design [364]. There was a trend in the reduction of plasma glucose, along with significant reductions in HbA1c levels (7.8 vs. 7.1), triglycerides, free fatty acids, total cholesterol, low-density lipoprotein cholesterol, and apoprotein B [365]. The β response to glucose was unaffected. These intriguing results prompted additional evaluations by Paolisso and colleagues using a more sensitive technique to measure insulin sensitivity, the euglycemic-hyperinsulinemic clamp.

 

Ten healthy subjects and 15 patients with T2D underwent an oral glucose tolerance test and euglycemic-hyperinsulinemic clamp before and after vitamin E supplementation (900 mg/d for 4 mo) [366]. In patients with T2D, vitamin E supplementation significantly increased both whole-body glucose disposal (i.e. insulin sensitivity) by approximately 50%, and non-oxidative glucose disposal by approximately 60%. Vitamin E also improved insulin action in the healthy subjects.

 

Vitamin E also improved insulin action in elderly people [367]. Twenty elderly, non-obese subjects with normal glucose tolerance were submitted to euglycemic-hyperinsulinemic clamp in a double-blind, crossover, and randomized study after 4 months treatment with either vitamin E (900 mg/d) or placebo. Whole-body glucose disposal was significantly potentiated by vitamin E compared to placebo. Furthermore, plasma vitamin E concentrations were correlated with net changes in insulin-stimulated whole-body glucose disposal.

In a 4-week, double-blind, randomized study of vitamin E administration (600 mg/d) versus placebo in 24 hypertensive patients, whole-body glucose disposal was measured by the euglycemic-hyperinsulinemic clamp [368]. In hypertensive subjects, vitamin E administration significantly increased whole-body glucose disposal, along with the ratio of reduced glutathione/oxidized glutathione in plasma. Four months treatment of patients with T2D with cardiac autonomic neuropathy with vitamin E lowered of glycated hemoglobin, insulin, norepinephrine, and the homeoststatic model assessment index, indicative of increased insulin sensitivity and improved glycemic control [369].

 

The results from a large number of major intervention studies have all concluded that treatment with vitamin E is ineffective at improving glycemic control or endothelial function, altering the development of T2D, or at preventing cardiovascular disease [370][371][372][373][374][375][376][377]; some studies have even raised questions of its safety [378][379].  In light of these results, the use of supplemental vitamin E (mixture or an individual isomer) for diabetes or cardiovascular health is no longer recommended.

 

Niacin

Niacin (nicotinic acid) has been used for many years to reduce elevated cholesterol and triglycerides. In addition, niacin has been shown to decrease cardiovascular events and mortality [380]. Some degree of angiographic regression has also being shown with niacin when used with other cholesterol medications. However, the use of niacin for the treatment of dyslipidemia-associated T2D has been limited, due to the adverse effect of high doses on glycemic control. Niacin is a B-vitamin (B-3), but when used in the doses necessary for blood cholesterol control, it should be considered a drug and not a vitamin. Recently, it has been reported that niacin has the potential ability, when given in low doses, to be well tolerated and efficacious. In this study, treatment of individuals with dyslipidemia-associated T2D with extended-release niacin (Niaspan™) led to significantly improved lipid levels and minimal changes in glycemic control [381]. The extended-release form was designed to circumvent the bothersome side effects of regular niacin, such as flushing of the skin.

 

In this 16-week, double-blind, placebo-controlled trial, 148 patients were randomized to placebo (n = 49) or 1000 (n = 45) or 1500 milligrams per day (n = 52) of Niaspan™. About half of the study participants continued taking their prescribed statin drugs for cholesterol lowering during the trial, and 81 percent continued their medications for diabetes. Dose-dependent increases in high-density lipoprotein cholesterol levels (+19% to +24%; P < 0.05) vs. placebo for both niacin dosages) and reductions in triglyceride levels (-13% to -28%; P < 0.05) vs. placebo for the 1500-mg Niaspan™) were observed. Baseline and week 16 values for glycosylated hemoglobin levels were 7.13% and 7.11%, respectively, in the placebo group; 7.28% and 7.35%, respectively, in the 1000-mg Niaspan™ group (P < 0.16 vs. placebo); and 7.2% and 7.5%, respectively, in the 1500-mg Niaspan™ group (P < 0.048 vs placebo). Four patients discontinued participation because of inadequate glucose control. Rates of adverse event rates other than flushing were similar for the niacin and placebo groups. Four patients discontinued participation owing to flushing (including 1 receiving placebo). No hepatotoxic effects or myopathy were observed. The authors concluded that low doses of Niaspan™ (1000 or 1500 mg/d) are a treatment option for dyslipidemia in patients with T2D.

Patients with diabetic dyslipidemia are commonly treated with triglyceride-lowering fibrate drugs, but niacin appears more effective than the fibrates for raising HDL. Since most patients with diabetes will require lipid-lowering therapy, the use of statins to lower LDL cholesterol has become routine therapy for the majority of patients. This study suggests that the addition of extended release low-dose niacin to statin therapy could provide an additional benefit for improvement of blood lipids and lipoproteins in patients with diabetes. However, the impact of niacin on glycemic control will still require regular monitoring.

 

Minerals and Trace Elements

 

Chromium

Second only to calcium, chromium is very popular mineral supplement in the US, with over 10 million individual users; in 2005, retail sales of chromium picolinate-containing products totaled over $130 million, and represent approximately 20% of the total market for chromium-containing products. Several authors, mostly on the basis of small studies of short duration, have suggested dietary trivalent chromium supplementation as an attractive option for the management of T2D and for glycemic control in persons at high risk for T2D [382][383]. Thus, chromium has emerged as the most widely used dietary supplement for the treatment of T2D in the US. The link between chromium and carbohydrate metabolism was proposed over 40 years ago, when it was identified as a component of the biologically active ‘glucose tolerance factor’ [384]. Chromium deficiency has been associated with decreased insulin action in both diabetic animals and humans [385][386][387]. Some but not all human studies have found that chromium supplementation has beneficial effects in individuals with impaired glucose tolerance and diabetes [388][389][390][391][392]. Oral administration of trivalent chromium is associated with favorable safety profile in animals and in humans [393][394].

 

To critically evaluate the clinical studies with chromium-treatment reported to date, a systematic review and meta-analysis of the RCTs were performed [395]. The objective was to determine the effect of chromium on glucose and insulin responses in healthy subjects and in individuals with glucose intolerance or T2D. The authors identified 20 reports of RCTs assessing the effects of chromium on glucose, insulin, or HbA1c. Their analyses summarized data on 618 participants from the 15 trials that reported adequate data: 193 participants had T2D and 425 were in good health or had impaired glucose tolerance. The meta-analysis showed no association between chromium and glucose or insulin concentrations among non-diabetic subjects. A study of 155 diabetic subjects in China reported that chromium reduced glucose and insulin concentrations [396]; the combined data from the 38 diabetic subjects in the other studies did not. Three trials reported data on HbA1c: one study each of persons with T2D [397], persons with impaired glucose tolerance [398], and healthy subjects [399]. The study of diabetic subjects in China was the only one to report that chromium significantly reduced HbA1c [400]. Thus, this meta-analysis of RCTs showed no effect of chromium on glucose or insulin concentrations in non-diabetic subjects, and data for persons with diabetes are inconclusive.

 

More recent reports suggest that the ability of chromium to improve glycemic control and/or increase insulin sensitivity is better observed in subjects with glucose intolerance, insulin resistance, type 1 diabetes, T2D, or gestational diabetes, rather than in healthy normal subjects [401]. Furthermore, these studies suggest that the form of chromium influences the study results, and that the picolinate form provides greater efficacy [402]. Absorption and bioavailability studies of the various forms of commercially available chromium have shown that chromium picolinate is more readily absorbed and is more bioavailable than the other forms of chromium that have been clinically tested in subjects with diabetes [403][404]. This may account for some of the disparity seen in the glycemic response observed in the clinical studies conducted to date.

 

In contrast to the results reported by others [405][406], a recent study has found that chromium picolinate (500 and 1000 μg daily for 6 months) was ineffective at reducing HbA1c in obese, poorly-controlled, insulin-dependent individuals with T2D [407]. Possibilities to explain this contrasting result are the limited statistical power to detect a significant change due to the small number of subjects (n = 17 for placebo group; n = 14 for 500 μg group; n = 15 for 1000 μg group), greater degree of obesity (and insulin resistance) at baseline (BMI = 33-35 kg/m2), and the severity of diabetes control at baseline (HbA1c = 9.4-9.7%). Furthermore, these subjects were unable to achieve adequate glycemic control even with anti-hyperglycemic medication, and required a very high dose of insulin.

Although the exact mechanism of chromium action has not been definitively established, data from a recent in vivo study suggest that chromium might exhibit its insulin sensitizing effect by reducing the content and activity of the tyrosine phosphatase PTP-1B [408]. PTP-1B has long been implicated in the regulation of insulin receptor tyrosine phosphorylation and tyrosine kinase activity [409], and has been validated as a bona fide pharmacological target for increasing insulin sensitivity [410][411][412][413][414]. In animals, small molecules that inhibit PTP-1B increase insulin sensitivity and lower plasma glucose [415][416][417]. Alternatively, chromium might act directly on the insulin receptor, and increase its tyrosine kinase activity [418], as has been observed with other small molecules [419][420].

 

Additional RCTs in well-characterized, at-risk populations are necessary to determine the effects of chromium on glucose, insulin, and HbA1c. To this end, the Office of Dietary Supplements (ODS), the National Center for Complementary and Alternative Medicine (NCCAM), and the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) invited basic and clinical applications to study the role of chromium as adjuvant therapy in T2D and/or impaired glucose tolerance ( http://grants1.nih.gov/grants/guide/pa-files/PA-01-114.html; program announcement expired 10/1/2004).

 

Magnesium

Magnesium, the fourth most abundant cation in humans, is an essential mineral in human nutrition, and is required for wide array of biological functions [421][422]. It is a cofactor in over 300 enzymatic reactions, and is important for the electrical stability of cells, maintenance of membrane integrity, muscle contraction, nerve conduction and vascular tone. Magnesium deficiency is linked to a number of clinical disorders including insulin resistance, T2D, hypertension, and cardiovascular disease [423][424][425][426][427][428][429][430]. Epidemiological studies strongly indicate that high daily magnesium intake is predictive of a lower risk for T2D in both men and women [431][432][433][434]. The plasma magnesium level is inversely related to insulin sensitivity in adults [435][436][437] and obese children [438], and parenteral magnesium supplementation improves insulin sensitivity as well as insulin secretion in patients with T2D [439][440][441]. However, until recently (see below), no beneficial effect of oral magnesium supplementation has been demonstrated on glycemic control either in patients with type 1 or T2D. Nonetheless, RCTs in well-characterized, at-risk populations are warranted to see whether magnesium replacement therapy will prove efficacious in the treatment of T2D.

 

To this end, it has recently been reported that oral magnesium supplementation (as a solution of magnesium chloride, MgCl22) restores serum magnesium levels, and improves insulin sensitivity and metabolic control in patients with T2D [442]. This study was a randomized double-blind placebo-controlled design, in which 63 subjects with decreased serum magnesium (≤ 0.74 mmol/l) treated by glibenclamide received either 50 ml of MgCl2 solution (50 g/l) or placebo daily for 16 weeks. At the end of the study, MgCl2-treated subjects showed a significantly higher serum magnesium concentration (0.74 ± 0.10 vs. 0.65 ± 0.07 mmol/l, P < 0.02) and lower HOMA-IR index (3.8 ± 1.1 vs. 5.0 ± 1.3, P < 0.005), fasting glucose levels (8.0 ± 2.4 vs. 10.3 ± 2.1 mmol/l, P < 0.01), and HbA1c (8.0 ± 2.4 vs. 10.1 ± 3.3%, P < 0.04) compared to placebo-treated subjects.

 

Subsequently, in a smaller pilot study (n = 9), the effects of magnesium (Mg) supplementation on patients with T2D with stable glycemic control were investigated [443]. Water from a salt lake with a high natural Mg content (7.1%) (MAG21) was used for supplementation after dilution with distilled water to 100mg/100mL; 300mL/day was given for 30 days. Fasting serum immunoreactive insulin level decreased significantly, as did HOMA-IR (both P < 0.05). There was also a marked decrease of the mean triglyceride level after supplementation. The patients with hypertension showed significant reduction of systolic (P < 0.01), diastolic (P = 0.0038), and mean (P < 0.01) blood pressure. Taken together, these results support the use of oral magnesium supplementation in patients with T2D who are magnesium-depleted.

 

Zinc

Zinc is another essential mineral in human nutrition with a wide range of biological functions. Zinc fulfills catalytic, structural, or regulatory roles in more than 200 zinc-requiring metalloenzymes [444]. The interaction of zinc with insulin induces conformational changes and enhances binding to the insulin receptor [445][446]. Zinc ions possess insulin mimetic activity, perhaps through their ability to inhibit protein tyrosine phosphatases, including PTP-1B [447]. With regard to glucose metabolism, zinc is a co-factor of several key enzymes. Zinc is an activator of fructose-1-6-bisphosphate aldolase, and an inhibitor of fructose-1-6-biphosphatase [448]. Zinc can also exert antioxidant activity [449], and is a cofactor in copper/zinc superoxide dismutase, a major antioxidant enzyme [450].

 

Some studies have reported zinc deficiency along with alterations in zinc metabolism in patients with diabetes [451][452][453][454]. Zinc supplementation studies in patients with diabetes are few, and have yielded contradictory results with regard to effects on glycemic control [455][456]. Interestingly, a recent study has confirmed previous reports that diabetic patients (both Type 1 and T2D) have significantly lower mean serum zinc levels compared with healthy controls [457], and that zinc supplementation (30 mg/day for 12 weeks) in the patients with T2D elevated their serum zinc level and significantly decreased HbA1c. In a 12-week randomized, double-blind placebo controlled study (n =18 per group) designed to evaluate the effects vitamin/mineral combination therapy on indices of nephropathy in patients with T2D, zinc (30 mg /day), when used in combination with magnesium (200 mg/day), vitamin C (200 mg), and vitamin E (100 IU/day) decreased the level of urinary albumin excretion (P = 0.005, respectively), without affecting urinary N-acetyl-beta-d-glucosaminidase activity [458]. The combination also resulted in a significant decrease in fasting serum glucose (P = 0.035) and malondialdehyde (P = 0.004)[459], along with an increase in HDL cholesterol and apolipoprotein A1 levels (P = 0.019)[460]. These results provide evidence for the beneficial effects of combination of magnesium, zinc, and vitamins C and E supplementation on improving glomerular (but not tubular) renal function in type 2 diabetic patients, and on metabolic control.

 

Vanadium

Vanadium is a transition metal that can exist in several oxidation states (-1 to +5), and is widely present in nature in the form of minerals [461]. It is also found in animals and humans, primarily as the tetravalent vanadyl cation (VO2+) and the pentavalent vanadate (VO3-). The tetravalent form is the most common intracellular form whereas the pentavalent form predominates in extracellular body fluids. Animals fed vanadium-deficient diets exhibited an increased rate of spontaneous abortion, depressed milk production, decreased growth, and premature death. The nutritional necessity in humans has not been established. The ‘average’ diet in the US supplies approximately 15-60 micrograms of vanadium daily. Foods relatively rich in vanadium include black pepper, mushrooms, shellfish, parsley, and dill seed. Fresh fruits, vegetables, and oils contain little or no vanadium.

 

In vitro and in vivo, vanadium-containing compounds exhibit insulin-mimetic activity primarily due to their ability to inhibit tyrosine phosphatase activity [462][463][464][465] and activate a cytosolic tyrosine kinase [466]. Many of the metabolic effects of insulin including the stimulation of glucose transport, glycogen synthesis, glucose oxidation, and lipogenesis and anti-lipolysis are mimicked by vanadate and related peroxovanadium compounds [467][468][469]. In vivo, vanadate and peroxovanadium compounds significantly lower blood glucose in insulin-dependent and insulin-resistant diabetic animals in the absence of overt toxicity [470][471][472][473][474]. The glucose-lowering effect of vanadate is achieved without elevating serum insulin, indicating an insulinomimetic effect and, in some cases, an insulin sensitizing effect.

 

In humans with T2D, several small studies of 2-4 weeks duration have indicated small but significant beneficial effects of vanadate treatment on various indicators of glucose metabolism [475][476][477][478][479][480]. The most common side effects were gastrointestinal disturbances: nausea, vomiting, diarrhea, and cramps. In the study from Cusi et al [481], eleven patients with T2D were treated with vanadyl sulfate (VS) at a higher dose (150 mg/day) and for a longer period of time (6 weeks) than in the previous studies. Before and after treatment insulin secretion during an oral glucose tolerance test, and hepatic glucose production (HGP) along with whole body insulin-mediated glucose disposal were measured. Treatment significantly improved glycemic control: fasting plasma glucose (FPG) decreased from 194 ± 16 to 155 ± 15 mg/dl, fructosamine decreased from 348 ± 26 to 293 ± 12 μmol/l, and HbA1C decreased from 8.1 ± 0.4 to 7.6 ± 0.4% (all P < 0.01) without any change in body weight. Subjects had an increased rate of HGP compared with non-diabetic controls (4.1 ± 0.2 vs. 2.7 ± 0.2 mg/kg lean body mass/min; P < 0.001), which was closely correlated with FPG (r = 0.56; P < 0.006). Vanadyl sulfate reduced HGP by about 20% (P < 0.01), and the decline in HGP was correlated with the reduction in FPG (r = 0.60; P < 0.05). VS also caused a modest increase in insulin-mediated glucose disposal (from 4.3 ± 0.4 to 5.1 ± 0.6 mg/kg lean body mass/min; P < 0.03), although the improvement in insulin sensitivity did not correlate with the decline in FPG after treatment (r = -0.16; P = NS). Thus, VS at a dose of 150 mg/day for 6 weeks improves hepatic and muscle insulin sensitivity in patients with T2D. The glucose-lowering effect of VS correlated well with the reduction in HGP, but not with insulin-mediated glucose disposal, suggesting that liver, rather than muscle, is the primary target of VS action at therapeutic doses.

 

Vanadium has a poor therapeutic index, and attempts have been made to reduce the dose of vanadium required for therapeutic effectiveness [482]. Organic forms of vanadium, as opposed to the inorganic VS, are appear to be safer (in animal studies), more absorbable, and able to deliver a therapeutic effect up to 50% greater than the inorganic forms [483][484]. An ongoing goal has been to provide vanadium with increased bioavailability, and in a form that is best able to produce the desired biological effects [485][486]. As a result, numerous organic complexes of vanadium have been developed including bis(maltolato)oxovanadium (BMOV), bis(cysteinamide N-octyl)oxovanadium known as Naglivan, bis(pyrrolidine-N-carbodithioato)oxovanadium, vanadyl-cysteine methyl ester, and bis-glycinato oxovanadium (BGOV) [487][488][489]. Other forms of vanadium, including polyoxovanadium [490] and vanadylpicolinate complexes [491] have also been proposed. The usefulness of these newer formulations as clinical agents for T2D remains to be determined. Despite the encouraging results of the clinical studies published to date, the safety of larger doses and use of vanadium salts (or related compounds) for longer periods is unknown [492]. Thus, the use of supplemental vanadium for the management of diabetes or impaired glucose tolerance is not recommended at this time.

 

Fatty Acids

 

Polyunsaturated Fatty Acids

Dietary ω-3 polyunsaturated fatty acids (ω-3 PUFAs) exhibit a broad array of biological activities in health and disease, including anti-inflammatory, lipid-lowering, and the prevention of coronary heart disease [493][494][495][496][497][498]. The most prominent dietary sources of ω-3 PUFAs include fish oils abundant in eicosapentanoic (EPA) and docosahexanoic (DHA) acids along with plants rich in α-linolenic acid. A primary mechanism of action of ω-3 PUFAs is achieved by altering gene expression mediated by the regulation of the activities or abundance of four families of transcription factors [499][500][501][502]. These include the peroxisome proliferator activated receptor (PPAR α,γ,δ), liver X receptors (α,β), hepatic nuclear factor-4 α, and the sterol regulatory element binding proteins 1 and 2. These transcription factors play major roles in the regulation of hepatic carbohydrate, fatty acid, triglyceride, cholesterol and bile acid metabolism.

 

A large body of epidemiological and clinical trial data suggests that ω-3 PUFAs play a significant role in the prevention of coronary artery disease [503][504][505]. The most convincing evidence is derived from four major intervention trials evaluating either fish meal, fish oil, or an α-linolenic acid-enriched spread on hard clinical end-points including myocardial infarction, death from coronary heart disease, and total mortality [506][507][508][509]. In essence, these studies found that supplementation significantly reduced cardiovascular events (cardiovascular death, non-fatal myocardial infarction and stoke)[510][511][512] and total mortality [513]. The average recommended intake by an expert panel of US nutritional scientists is 2.2 g/d of α-linolenic acid and 0.65 g/d of EPA plus DHA [514], while the British Nutrition Foundation has recommended 2.4 g/d of α-linolenic acid and 1.2 g/d of EPA plus DHA [515].

 

An interim report from The Japan EPA Lipid Intervention Study (JELIS) was presented at the American Heart Association Meeting in November 2005 ( http://www.americanheart.org/presenter.jhtml?identifier=3035468)[516]). Of 18,645 eligible participants, 9,326 were given 1,800 milligrams (mg)/day of highly purified EPA capsules as add-on therapy to a statin. The primary endpoint of the study was experiencing any one of a group of outcomes that included sudden cardiac death, heart attack, unstable angina (sustained chest pain due to the heart’s oxygen starvation), or undergoing procedures to reopen blocked arteries, such as angioplasty/stenting or coronary artery bypass surgery. After more than 4.5 years of follow-up, the primary endpoint was seen in 2.8 percent of patients treated with statins plus EPA compared to 3.5 percent in the statin-only group, represents an approximate 20 percent reduction in risk EPA plus statin treatment compared to statin treatment alone.

 

Conflicting results have been reported regarding the effects of fish oil supplementation on glycemic control in those with glucose intolerance including individuals with T2D. Several early studies reported detrimental effects [517][518], but subsequent studies with improved design have not replicated the earlier findings [519][520][521][522]. Results from a meta-analysis of pooled data from all RCTs in which fish oil supplementation was the only intervention in subjects with T2D was recently published [523]. Eighteen trials including 823 subjects followed for a mean of 12 weeks were included. Doses of fish oil used ranged from 3 to 18 g/day. The outcomes studied were glycemic control and lipid levels. Meta-analysis demonstrated a statistically significant effect of fish oil on lowering triglycerides (-0.56 mmol/l) and raising LDL cholesterol (0.21 mmol/). No statistically significant effect was observed for fasting glucose, HbA1c, total cholesterol, or HDL cholesterol. The triglyceride-lowering effect and the elevation in LDL cholesterol were most evident in those trials that recruited hypertriglyceridemic subjects and used higher doses of fish oil. Thus, this meta-analysis of RCTs showed that fish oil supplementation in T2D lowers triglycerides, raises LDL cholesterol, and has no statistically significant effect on glycemic control. There is no evidence that fish oil supplementation adversely effects glucose tolerance, insulin action, or insulin secretion in non-diabetic individuals [524]. There is some evidence that fish oil may improves defects in insulin action and prevent alterations in glucose homeostasis and the further development of T2D [525].

 

A recent report has shown that, in individuals with T2D but without hypertriglyceridemia, fish oil supplementation for 9 weeks moderately increased blood glucose and decreases insulin sensitivity [526].

 

Conjugated Linoleic Acid (CLA)

Conjugated linoleic acid (CLA) refers to a group of polyunsaturated fatty acids that are positional and geometric conjugated dieonic isomers of linoleic acid [527]. The biological activity of CLA was originally discovered due to its ability to inhibit chemically induced carcinogenesis in rodents [528][529]. Subsequently, numerous health benefits have been attributed to CLA including activity as an anti-obesogenic, anti-diabetogenic, and anti-atherosclerortic agent [530][531]. The major isomers of CLA are the cis-9,trans-11 and the trans-10, cis-12, with their biological activities being isomer-specific [532]. The major dietary sources of CLA are meat and dairy products. CLA concentrations in dairy products typically range from 2.8 to 7.0 mg/g fat (frozen yogurt and condensed milk, respectively), of which the cis-9,trans-11 isomer comprises ~75%-95% of the total CLA [533]. CLA concentrations in meat typically range from 0.6 to 5.8 mg/g fat (pork and lamb, respectively), of which the cis-9,trans-11 isomer comprises ~55%-85% of the total CLA [534]. Similar to ω-3 PUFAs, CLA isomers are ligands and activators of PPARα, but with an approximate 10-fold higher affinity (~140-260 nM) [535]. CLA isomers readily undergo extensive metabolism including elongation and desaturation, yielding additional potential bioactive molecules [536].

 

In several animal models, CLA has been shown to reduce body fat accumulation, improve glucose tolerance, and increase insulin sensitivity [537][538]. In individuals with T2D (n =12), plasma trans-10, cis-12 but not cis-9,trans-11 CLA is inversely correlated with body weight (P < 0.05) and serum leptin (P < 0.02) [539].  Thus, CLA supplementation has been suggested as a potential new nutraceutical approach for obesity, a major risk factor for the development of T2D. However, conflicting results have been reported regarding the beneficial effects of CLA supplementation on adiposity and metabolism in humans [540][541][542][543]. In several studies, administration of CLA (1.8-4.2 g/day) for 12 weeks has been reported to decrease body fat mass (~4%; P < 0.001) in healthy individuals [544][545] and in overweight and obese individuals [546]. There were no changes in body weight, serum lipids, or glucose metabolism in these studies.

 

However, in another study, abdominally obese men (n = 60) were treated with 3.4 g/day CLA (isomer mixture), purified trans-10, cis-12 CLA, or placebo [547]. Euglycemic-hyperinsulinemic clamp, serum hormones, lipids, and anthropometry were assessed before and after 12 weeks of treatment. Baseline metabolic status was similar between groups. Unexpectedly, trans-10, cis-12 CLA increased insulin resistance (19%; P < 0.01) and glycemia (4%; P < 0.001) and reduced HDL cholesterol (-4%; P < 0.01) compared with placebo. Body fat, sagittal abdominal diameter, and weight decreased versus baseline, but the difference was not significantly different from placebo. The CLA mixture did not change glucose metabolism, body composition, or weight compared with placebo but lowered HDL cholesterol (-2%; P < 0.05). Trans-10, cis-12 CLA also increases markers of oxidative stress and inflammation [548], thus revealing important isomer-specific metabolic actions of CLA in abdominally obese men.

 

The detrimental effect of the CLA mixture on insulin sensitivity was confirmed by another group. The effect of CLA supplementation on markers of glucose and insulin metabolism, lipoprotein metabolism, and inflammatory markers of CVD in subjects with T2D [549]. The study was a randomized, double-blind, placebo-controlled trial. Thirty-two subjects with stable, diet-controlled type 2 diabetes received CLA (3.0 g/d; 50:50 blend of cis-9, trans-11 CLA and trans-10, cis-12 CLA) or control for 8 wk. A 3-h 75-g oral-glucose-tolerance test was performed, and fasting plasma lipid concentrations and inflammatory markers were measured before and after the intervention. CLA supplementation significantly increased fasting glucose concentrations (6.3%; P < 0.05) and reduced insulin sensitivity as measured by homeostasis model assessment, oral glucose insulin sensitivity, and the insulin sensitivity index (composite) (P = 0.05). Total HDL-cholesterol concentrations increased by 8% (P < 0.05), which was due to a significant increase in HDL(2)-cholesterol concentrations (P < 0.05). The ratio of LDL to HDL cholesterol was significantly reduced (P < 0.01). CLA supplementation reduced fibrinogen concentrations (P < 0.01), but had no effect on inflammatory markers of CVD. Thus, supplementation with a CLA mixture had an adverse effect on insulin and glucose metabolism.

 

The effects of trans-10, cis-12 CLA supplementation on plasma proinsulin, insulin, C-peptide and adiponectin concentrations, including their associations with change in insulin sensitivity, has also been evaluated [550]. The study was a randomized, double-blind, placebo-controlled trial. Fifty-seven non-diabetic abdominally obese men received either 3.4 g trans-10, cis-12 CLA, CLA-isomer mixture, or control oil for 12 weeks. Insulin sensitivity (hyperinsulinemic-euglycemic clamp), intact proinsulin, insulin, the proinsulin : insulin ratio, C-peptide, glucose, and adiponectin were assessed before and after supplementation. Supplementation with trans-10, cis-12 CLA increased proinsulin (P < 0.01), the proinsulin : insulin ratio (P < 0.05) and C-peptide concentrations (P < 0.001) in comparison with control subjects. Adiponectin did not change significantly. The change in proinsulin, but not the proinsulin : insulin ratio, was related to impaired insulin sensitivity (r = -0.58; P <0.0001), independently of changes in insulin, C-peptide, glucose, adiponectin and BMI. Thus, in obese, non-diabetic men, trans-10, cis-12 CLA induced hyperproinsulinemia that was related to impaired insulin sensitivity, independently of changes in insulin concentrations. These results are of clinical interest, as hyperproinsulinemia predicts diabetes and cardiovascular disease. In light of these results, the use of supplemental CLA (mixture or an individual isomer) for the management of obesity, impaired glucose tolerance, or diabetes is definitely not recommended.

 

 

SUMMARY AND POSSIBILITIES FOR TREATMENT

Clearly, many natural products including botanicals and other nutraceuticals have hypoglycemic, anti-hyperglycemic, insulin sensitizing, anti-hyperlipidemic, anti-hypertensive, and anti-inflammatory activities. There are published studies reporting the anti-diabetic activity of well-over a thousand different botanicals and nutraceuticals. The number of those treatments evaluated in clinical trials is approximately 100 [551]. In the vast major of these trials, the botanicals and nutraceuticals were evaluated as an adjunct to diet and prescription medications. Fifty-eight of the trials were controlled, and conducted in individuals with diabetes or impaired glucose tolerance. Of these, statistically significant treatment effects were reported in 88% of trials (23 of 26) evaluating a single botanical, and 67% of trials (18 of 27) evaluating individual vitamin or mineral supplements (reviewed in [552]). When reported, side effects were few and generally mild (gastrointestinal irritation and nausea).

 

However, many of the studies suffered from design flaws including small (< 10 subjects) sample sizes, heterogeneity of subjects, and short-duration of treatment. Furthermore, there is a lack of multiple studies for many of the individual supplements. Despite the apparent lack of side effects of these treatments, it would be prudent to be aware of the potential for dietary supplements, especially botanicals, to interact with a patient’s prescription medication. One of the most important potential botanical-drug interactions is that of garlic, Trigonella, and Ginkgo biloba with non-steroidal anti-inflammatory drugs (including aspirin) or warfarin, as these botanicals possess limited anti-coagulant activity [553][554]. Another potential interaction of concern is one involving G. biloba, a botanical widely used for the treatment of memory and concentration problems, confusion, depression, anxiety, dizziness, tinnitus, and headache [555][556]. Ingestion of G. biloba extract by patients with T2D may increase the hepatic metabolic clearance rate of not only insulin but also hypoglycemic medications, resulting in reduced insulin-mediated glucose metabolism and elevated blood glucose [557]. Another issue to consider with botanicals is the potential for batch-to-batch variation due to age of the plant, geographic source, time of harvest, and method of drying and preparation, all of which can dramatically impact the purity and potency of active ingredients. None of the agents discussed here is recommended for use in pregnant or lactating women, or in children. Furthermore, patients should be advised on the proper use of any alternative treatment to avoid the risk of hypoglycemia.

 

That being stated, several botanical and nutraceutical agents merit consideration as complimentary approaches for use in patients with T2D. Botanical treatments with the strongest evidence of clinical safety and efficacy include I. batas (caiapo), T. foenum-graecum (fenugreek), and C.cassia (cinnamon). Non-botanical nutraceutical agents with promise for improving insulin sensitivity and glycemic control include α-lipoic acid, chromium picolinate, and magnesium. If the safety profile of the newly proposed organic vanadium complexes could be confirmed with chronic use, this agent would also be regarded as a promising treatment for glycemic control. In addition, there is evidence that α-lipoic acid improve the symptoms of individuals with microvascular complications, especially neuropathy. Clearly, ω-3 PUFAs (EPA, DHA, α-linolenic acid) merit strong consideration for lipid lowering, and overall cardiovascular health.

The increasing movement for the public in general and patients with diabetes (and other diseases) to self-treat using botanicals and nutraceuticals cannot be disputed and should not be ignored. Health care professionals are urged to increase their knowledge base in this area on an ongoing basis. They are also urged to pro-actively query patients on their use of these agents, and record the information obtained in the patient record. Many patients are reluctant to discuss their use of botanicals and nutraceuticals, so it is important for health care professionals to keep an open mind and be non-judgmental. Since patients cannot be expected to distinguish between the marketing hype of manufacturers and evidence derived from credible scientific studies, health care professionals must be positioned to provide an informed opinion and recommendation.

 

 

TABLES

 

Table 1. Major Botanicals Used for Type 2 Diabetes

Botanical 

Putative Bioactives 

Anti- Diabetic Activity 

Mode of Action 

Typical Daily Dose 

Potential Side Effects 

Relative Rating

I. batas (Caiapo)

High-molecular weight acid glycoprotein

Glucose control; reduces HbA1c

Not well characterized; Possibly increases insulin sensitivity

4 g of white-skin sweet potato (capsule)

None reported

++++

T. foenum- graecum

(Fenugreek) 

Fiber, 4- hydroxyisoleucine, saponins, coumarins, alkaloids, glycosides 

Glucose control; Anti- hyperlipidemic 

Delays gastric emptying; Inhibits glucose absorption in gut; Enhances insulin secretion 

2.5-15 g (defatted seeds) 

Hypoglycemia; Additive with other glucose- lowering agents and insulin; GI irritation; Anti- coagulant 

+++

C. cassia (Cinnamon)

Methyl hydroxyl chalcone polymer (MHCP)

Glucose control 

Possibly insulin mimetic 

3-6 g (capsule)

None reported

+++

M. charantia

(Bitter

melon) 

Charantin, vicine, mormordicine (alkaloid), polypeptide P 

Glucose control 

Inhibits glucose absorption in gut; Enhances insulin secretion; increases glucose transport and glycogen synthesis 

300-600 mg (juice extract); 1.8 g (capsule) 

Hypoglycemia; Additive with other glucose- lowering agents and insulin; GI irritation 

++

G. sylvestre

(Gurmar) 

Gymnemic acids, gymnemosides 

Glucose control 

Inhibits glucose absorption in gut; Enhances insulin secretion 

200-600 mg 

Hypoglycemia; Additive with other glucose- lowering agents and insulin;GI irritation 

++

O. streptacantha

(Nopal; prickly pear) 

Fiber, pectin 

Glucose control; Anti- hyperlipidemic 

Delays gastric emptying; Inhibits glucose absorption in gut 

2.4 g 

None reported (limited data) 

++

C. indica

Not yet characterized 

Glucose control 

Not characterized; possibly insulin mimetic 

1.8 g (powdered leaves) 

None reported (limited data) 

++

P. quinquefolius (American ginseng) 

Ginsenosides (saponins), polysaccharides, peptides, fatty acids 

Glucose control 

Delays gastric emptying; Inhibits glucose absorption in gut; Hormonal & CNS activity 

100-200 mg 

Estrogenic effects; Ginseng abuse syndrome; Interacts with many drugs 

++

A. vera

(Aloe) 

Fiber (glucomannan), aloins, anthraquinones, barbaloin, polysaccharides, salicylic acids 

Glucose control 

Not characterized; possibly delays gastric emptying and inhibits glucose absorption in gut 

1.2 g (capsule) 

None reported (limited data) 

++

A. sativum

(Garlic) 

Allicin, allyl methyl thiosulfonate, 1-propenyl allyl thiosulfonate, γ-L-glutamyl- S-alkyl-L- cysteine 

Anti- hyperlipidemic;Anti- hypertensive 

Anti- inflammatory (Antioxidant) 

600-1000 mg 

GI irritation; Anti- coagulant; heartburn; garlic odor 

+

 

 

Table 2. Major Non-Botanical Nutraceuticals Used for Type 2 Diabetes

Botanical 

Anti-Diabetic Activity 

Mode of Action 

Typical Daily Dose 

Potential Side Effects 

Relative Rating

ω-3 PUFAs

Anti- hyperlipidemic 

PPARα,γ,δ agonists 

0.65-1.2 g (EPA + DHA); 2.4 g ALA 

GI irritation; halitosis 

+++++

α-Lipoic acid

Insulin sensitizer; Anti-neuropathy

Anti- inflammatory (Antioxidant) 

900-1800 mg 

GI irritation 

++++

Chromium

Glucose control 

Enhances insulin action 

200-400 μg 

Potential for renal toxicity (rare) 

++++

Magnesium 

Insulin sensitizer; Glucose control; Anti- hypertensive 

Not characterized; Possibly enhances insulin action 

300-400 mg; (2.5 g used for glucose control) 

Diarrhea 

++++

Zinc

Glucose control 

Anti- inflammatory (Antioxidant); insulin binding 

30-80 mg 

GI irritation; metallic taste; headache 

+++

L-Arginine 

Improves endothelial dysfunction 

Nitric oxide donor 

3-6 g 

None reported 

+++

Vitamin C

Insulin sensitizer; Improves endothelial dysfunction 

Anti-inflammatory (Antioxidant) 

500-2000 mg 

None reported 

++

Coenzyme Q10

Insulin sensitizer 

Anti-inflammatory (Antioxidant) 

100-150 mg 

GI irritation 

+

Vitamin E

Insulin sensitizer 

Anti-inflammatory (Antioxidant) 

600-900 mg 

None reported 

Not recommended

Vanadium 

Insulin sensitizer; Glucose control 

Insulin mimetic; Pan-tyrosine phosphatase inhibitor 

100-150 mg 

GI irritation; tissue accumulation; Uncertain long-term safety profile 

Not recommended for long term human use

CLA 

Anti-obesity 

PPARα agonist 

2-4 g 

GI irritation; Increased inflammation and oxidative stress 

Not recommended

Niacin

Anti- hyperlipidemic 

Anti-lipolytic; decreases rate of hepatic synthesis of VLDL and LDL 

1000-1500 mg 

Impaired glucose tolerance; flushing 

Available as Rx

 

 

Table 3. Credible Internet Sources for Information on Botanicals and Other Nutraceuticals

Source 

Website Address 

Thumbnail Sketch

American Botanical Council

http://www.herbalgram.org/

The leading independent, non-profit, international member-based organization providing education using science-based and traditional information to promote the responsible use of herbal medicine

American Nutraceutical Association

http://www.ana-jana.org/

A professional organization founded to develop and provide educational materials and programs on nutraceuticals and nutrition for health care professionals and consumers

American Society of Pharmacognosy 

http://www.phcog.org

Professional organization dedicated to discipline of pharmacognosy (the science and study of drugs from natural sources)

The Cochrane Library

http://www.nelh.nhs.uk/cochrane.asp

An electronic database designed to provide high quality scientific evidence

Consumer Lab.com 

http://www.consumerlab.com

Independent product review site that provides information on the content of nutritional products including dietary supplements (Subscription required for access to some content)

DiaMedBase

http://www.progenebio.in/DMP/DMP.htm

An interactive database of approximately 400 medicinal plants for diabetes

Dr. Duke’s Phytochemical and Ethnobotanical Databases

http://www.ars-grin.gov/duke/

An interactive, electronic database developed by James Duke, PhD, former chief botanist at the USDA

HerbMed 

http://www.herbmed.org/index.asp

An interactive, electronic herbal database on the use of herbs for health

MD Anderson Complementary/ Integrative Medicine

http://www.mdanderson.org/departments/CIMER/

Website of prestigious cancer treatment center

National Center for Complimentary and Alternative Medicine 

http://nccam.nih.gov;

http://nccam.nih.gov/health/diabetes/

National (US) center that supports and disseminates research results on complementary and alternative medicine; NCCAM research report (2005): Treating type 2 diabetes with dietary supplements

Natural Medicines Comprehensive Database 

http://www.naturaldatabase.com

An extremely comprehensive, scientifically-based, and practical database on natural medicines (Subscription required)

NIH Office of Dietary Supplements (ODS) 

http://ods.od.nih.gov/index.aspx

National (US) center that supports and disseminates research results on dietary supplements

NAPRALERT™: Natural Products Alert

http://www.napralert.org/

NAPRALERT™ is an extensive relational database developed at the University of IL at Chicago, and contains ethnomedical, chemical, biochemical, pharmacological, and clinical data derived from the evaluation of plants and other natural products (Search charges apply)

National Institute of Diabetes& Digestive & Kidney Diseases: Alternative Therapies for Diabetes 

http://diabetes.niddk.nih.gov/

http://diabetes.niddk.nih.gov/dm/pubs/alternativetherapies/index.htm 

Clearinghouse (albeit abridged) for some alternative treatments for diabetes

Quackwatch

http://www.quackwatch.org/

A non-profit corporation whose purpose is to combat health-related frauds, myths, fads, fallacies, and misconduct

Rick Mendosa's Diabetes Directory 

http://www.mendosa.com/http://www.mendosa.com/diabetes.htm 

An extensive and unbiased private site published by a freelance journalist and consultant specializing in (and afflicted with) diabetes

Supplement Watch 

http://www.supplementwatch.com

A self-funded, privately held corporation consisting of scientists, physiologists, nutritionists, and other health professionals dedicated to educating the public about the pros and cons of dietary supplementation. (Subscription required for access to some content)

US FDA Office of Nutritional Products, Labeling, and Dietary Supplements 

http://www.cfsan.fda.gov/http://www.cfsan.fda.gov/~dms/supplmnt.html 

FDA office responsible for developing policy and regulations for dietary supplements, medical foods, and related areas, as well as for their scientific evaluation

WebMD Health 

http://www.webmd.com/

WebMD provides comprehensive health information and tools for managing health care. For health care professionals and their patients

N.B. Sources cited above are selected examples, and not meant to be exhaustive

 

 

 

 

 

Footnotes

  1. Zimmet P, Alberti KG, Shaw J. Global and societal implications of the diabetes epidemic. Nature 2001; 414:782-787.
  2. Reaven GM. Insulin resistance and its consequences: type 2 diabetes mellitus and coronary heart disease. In: LeRoith D, Taylor SI, Olefsky JM, editors. Diabetes Mellitus: A Fundamental and Clinical Text. Philadelphia: Lippincott Williams & Wilkins, 2000: 604-615.
  3. Reaven GM. Role of insulin resistance in human disease. Diabetes 1988; 37:1595-1607.
  4. DeFronzo RA. Pathogenesis of type 2 diabetes: metabolic and molecular implications for identifying diabetes genes. Diabet Rev 1997; 5:177-269.
  5. Hill JO, Wyatt HR, Reed GW, Peters JC. Obesity and the environment: where do we go from here? Science 2003; 299:853-855.
  6. Turner RC, Cull CA, Frighi V, Holman RR. Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus - Progressive requirement for multiple therapies (UKPDS 49). J Am Med Assoc 1999; 281:2005-2012.
  7. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med 1999; 131:281-303.
  8. Evans JL, Rushakoff RJ. Oral Pharmacological Agents for Type 2 Diabetes: Sulfonylureas, Meglitinides, Metformin, Thiazolidinediones, a-Glucosidase Inhibitors, and Emerging Approaches. In: Degroot L, Goldfine ID, Rushakoff RJ, editors. Endotext.com: Diabetes and Carbohydrate Metabolism. http://www.medtext.com:MDtext.com, 2002.
  9. Fuhr JP, He H, Goldfarb N, Nash DB. Use of chromium picolinate and biotin in the management of type 2 diabetes: an economic analysis. Dis Manag 2005; 8:265-275.
  10. Altman SH, Parks-Thomas C. Controlling spending for prescription drugs. N Engl J Med 2002; 346:855-856.
  11. Dham S, Shah V, Hirsch S, Banerji MA. The role of complementary and alternative medicine in diabetes. Curr Diab Rep 2006; 6:251-258.
  12. Cicero AF, Derosa G, Gaddi A. What do herbalists suggest to diabetic patients in order to improve glycemic control? Evaluation of scientific evidence and potential risks. Acta Diabetol 2004; 41:91-98.
  13. Saxena A, Vikram NK. Role of selected Indian plants in management of type 2 diabetes: a review. J Altern Complement Med 2004; 10:369-378.
  14. Shekelle PG, Hardy M, Morton SC et al. Are Ayurvedic herbs for diabetes effective? J Fam Pract 2005; 54:876-886.
  15. Andrade-Cetto A, Heinrich M. Mexican plants with hypoglycemic effect used in the treatment of diabetes. J Ethnopharmacol 2005; 99:325-348.
  16. Johnson L, Strich H, Taylor A et al. Use of herbal remedies by diabetic Hispanic women in the southwestern United States. Phytother Res 2006; 20:250-255.
  17. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  18. Berman BM, Swyers JP, Kaczmarczyk J. Complementary and alternative medicine: herbal therapies for diabetes. J Assoc Acad Minor Phys 1999; 10:10-14.
  19. Grover JK, Yadav S, Vats V. Medicinal plants of India with anti-diabetic potential. J Ethnopharmacol 2002; 81:81-100.
  20. Murray MT, Pizzorno JE. Botanical medicine ¿ a modern perspective. Textbook of Natural Medicine. Edinburgh: Churchill Livingstone, 1999.
  21. AACE Nutrition Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for the clinical use of dietary supplements and nutraceuticals. Endocr Pract 2003; 9:417-470.
  22. Fontanarosa PB, Rennie D, DeAngelis CD. The need for regulation of dietary supplements-lessons from ephedra. J Am Med Assoc 2003; 289:1568-1570.
  23. Kinsel JF, Straus SE. Complementary and alternative therpaeutics: rigorous research is needed to support claims. Annu Rev Pharmacol Toxicol 2003; 43:463-484.
  24. Wolfe SM. Ephedra--scientific evidence versus money/politics. Science 2003; 300(5618):437.
  25. Fontanarosa PB, Rennie D, DeAngelis CD. The need for regulation of dietary supplements-lessons from ephedra. J Am Med Assoc 2003; 289:1568-1570.
  26. Wolfe SM. Ephedra--scientific evidence versus money/politics. Science 2003; 300(5618):437.
  27. Bent S, Tiedt TN, Odden MC, Shlipak MG. The relative safety of ephedra compared with other herbal products. Ann Intern Med 2003; 138:468-471.
  28. Shekelle PG, Hardy ML, Morton SC et al. Efficacy and safety of ephedra and ephedrine for weight loss and athletic performance: a meta-analysis. J Am Med Assoc 2003; 289:1537-1545.
  29. Kinsel JF, Straus SE. Complementary and alternative therpaeutics: rigorous research is needed to support claims. Annu Rev Pharmacol Toxicol 2003; 43:463-484.
  30. AACE Nutrition Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for the clinical use of dietary supplements and nutraceuticals. Endocr Pract 2003; 9:417-470.
  31. Yeh GY, Eisenberg DM, Davis RB, Phillips RS. Use of complementary and alternative medicine among persons with diabetes mellitus: results of a national survey. Am J Public Health 2002; 92:1648-1652.
  32. Berman BM, Swyers JP, Kaczmarczyk J. Complementary and alternative medicine: herbal therapies for diabetes. J Assoc Acad Minor Phys 1999; 10:10-14.
  33. Kim C, Kwok YS. Navajo use of native healers. Arch Intern Med 1998; 158:2245-2249.
  34. Mull DS, Nguyen N, Mull JD. Vietnamese diabetic patients and their physicians: what ethnography can teach us. West J Med 2001; 175:307-311.
  35. Noel PH, Pugh JA, Larme AC, Marsh G. The use of traditional plant medicines for non-insulin dependent diabetes mellitus in South Texas. Phytother Res 2003; 11:512-517.
  36. Eisenberg DM, Davis RB, Ettner SL et al. Trends in alternative medicine use in the United States, 1990-1997: results of a follow-up national survey. J Am Med Assoc 1998; 280:1569-1575.
  37. Yeh GY, Eisenberg DM, Davis RB, Phillips RS. Use of complementary and alternative medicine among persons with diabetes mellitus: results of a national survey. Am J Public Health 2002; 92:1648-1652.
  38. American Diabetes Association. Unproven therapies. Diabetes Care 2003; 26 (Suppl 1):S142.
  39. AACE Nutrition Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for the clinical use of dietary supplements and nutraceuticals. Endocr Pract 2003; 9:417-470.
  40. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  41. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  42. Dey L, Attele AS, Yuan CS. Alternative therapies for type 2 diabetes. Altern Med Rev 2002; 7:45-58.
  43. Evans JL, Maddux BA, Goldfine ID. Antioxidants in diabetic complications and insulin resistance. In: Raz I, Skyler JS, Shafrir E, editors. Diabetes: From Research to Diagnosis and Treatment. London: Martin Dunitz, 2003: 479-496.
  44. Antioxidants in Diabetes Management. 1st ed. New York: Marcel Dekker, 2000.
  45. Ha T, Lean MEJ. Diet and lifestyle modification in the management of non-insulin-dependent diabetes mellitus. In: Pickup JC, Williams G, editors. Textbook of Diabetes. Oxford: Blackwell Science, 1997: 1-18.
  46. Jenkins DJA, Jenkins AL, Kendall CWC. Dietary therapy in type 2 diabetes mellitus: spreading the nutrient load. In: LeRoith D, Taylor SI, Olefsky JM, editors. Diabetes Mellitus: A Fundamental and Clinical Text. Philadelphia: Lippincott Williams & Wilkins, 2002: 757-765.
  47. Costacou T, Mayer-Davis EJ. Nutrition and prevention of type 2 diabetes. Annu Rev Nutr 2003; 23:147-170.
  48. Youngren JF. Exercise and the regulation of blood glucose. In: Degroot L, Goldfine ID, Rushakoff RJ, editors. Endotext.org: Diabetes and Carbohydrate Metabolism. Internet-based: http://www.mdtext.com/,3 A.D.: http://www.mdtext.com/diabetes/diabetes14/diabetesframe14.htm.
  49. Watts NB, Spanheimer RG, DiGirolamo M et al. Prediction of glucose response to weight loss in patients with non-insulin-dependent diabetes mellitus. Arch Intern Med 1990; 150:803-806.
  50. Wing RR, Koeske R, Epstein LH, Nowalk MP, Gooding W, Becker D. Long-term effects of modest weight loss in type II diabetic patients. Arch Intern Med 1987; 147:1749-1753.
  51. Goldstein DJ. Beneficial health effects of modest weight loss. Int J Obes Relat Metab Disord 1992; 16:397-415.
  52. Bantle JP, Wylie-Rosett J, Albright AL et al. Nutrition recommendations and interventions for diabetes--2006: a position statement of the American Diabetes Association. Diabetes Care 2006; 29:2140-2157.
  53. Diabetes Medical Guidelines Task Force. The American Association of Clinical Endocrinologists medical guidelines for the management of diabetes mellitus: the AACE system of intensive diabetes self-management-2002 update. Endocr Pract 2002; 8 (Suppl.1):40-82.
  54. Chandalia M, Garg A, Lutjohann D, von Bergmann K, Grundy SM, Brinkley LJ. Beneficial effects of high dietary fiber intake in patients with type 2 diabetes mellitus. N Engl J Med 2000; 342:1392-1398.
  55. Vuksan V, Sievenpiper JL, Owen R et al. Beneficial effects of viscous dietary fiber from Konjac-mannan in subjects with the insulin resistance syndrome: results of a controlled metabolic trial. Diabetes Care 2000; 23:9-14.
  56. Frape DL, Jones AM. Chronic and postprandial responses of plasma insulin, glucose and lipids in volunteers given dietary fibre supplements. Br J Nutr 1995; 73:733-751.
  57. Lakhdar A, Farish E, McLaren EH. Fibre and patients with diabetes. Br Med J 1988; 296:1471.
  58. He FJ, Nowson CA, MacGregor GA. Fruit and vegetable consumption and stroke: meta-analysis of cohort studies. Lancet 2006; 367:320-326.
  59. Lichtenstein AH, Appel LJ, Brands M et al. Diet and lifestyle recommendations revision 2006: a scientific statement from the American Heart Association Nutrition Committee. Circulation 2006; 114:82-96.
  60. Stanner SA, Hughes J, Kelly CN, Buttriss J. A review of the epidemiological evidence for the 'antioxidant hypothesis'. Public Health Nutr 2004; 7:407-422.
  61. van Dam RM, Hu FB. Coffee consumption and risk of type 2 diabetes: a systematic review. J Am Med Assoc 2005; 294:97-104.
  62. Bantle JP, Wylie-Rosett J, Albright AL et al. Nutrition recommendations and interventions for diabetes--2006: a position statement of the American Diabetes Association. Diabetes Care 2006; 29:2140-2157.
  63. Stanner SA, Hughes J, Kelly CN, Buttriss J. A review of the epidemiological evidence for the 'antioxidant hypothesis'. Public Health Nutr 2004; 7:407-422.
  64. Lee DH, Folsom AR, Harnack L, Halliwell B, Jacobs DR, Jr. Does supplemental vitamin C increase cardiovascular disease risk in women with diabetes? Am J Clin Nutr 2004; 80:1194-1200.
  65. Liu S, Lee IM, Song Y et al. Vitamin E and risk of type 2 diabetes in the women's health study randomized controlled trial. Diabetes 2006; 55:2856-2862.
  66. Lonn E, Bosch J, Yusuf S et al. Effects of long-term vitamin E supplementation on cardiovascular events and cancer: a randomized controlled trial. J Am Med Assoc 2005; 293:1338-1347.
  67. Boshtam M, Rafiei M, Golshadi ID, Ani M, Shirani Z, Rostamshirazi M. Long term effects of oral vitamin E supplement in type II diabetic patients. Int J Vitam Nutr Res 2005; 75:341-346.
  68. Bantle JP, Wylie-Rosett J, Albright AL et al. Nutrition recommendations and interventions for diabetes--2006: a position statement of the American Diabetes Association. Diabetes Care 2006; 29:2140-2157.
  69. Keys A. Mediterranean diet and public health: personal reflections. Am J Clin Nutr 1995; 619 (6 Suppl):1321S-1323S.
  70. Curtis BM, O'Keefe JH, Jr. Understanding the Mediterranean diet. Could this be the new "gold standard" for heart disease prevention? Postgrad Med 2002; 112:35.
  71. de LM, Salen P. The Mediterranean-style diet for the prevention of cardiovascular diseases. Public Health Nutr 2006; 9:118-123.
  72. Willett WC. The Mediterranean diet: science and practice. Public Health Nutr 2006; 9:105-110.
  73. Serra-Majem L, Roman B, Estruch R. Scientific evidence of interventions using the Mediterranean diet: a systematic review. Nutr Rev 2006; 64:S27-S47.
  74. Trichopoulou A. Mediterranean diet: the past and the present. Nutr Metab Cardiovasc Dis 2001; 11 (4 Suppl):1-4.
  75. Schroder H. Protective mechanisms of the Mediterranean diet in obesity and type 2 diabetes. J Nutr Biochem 2006; Epub ahead of print.
  76. Estruch R, Martinez-Gonzalez MA, Corella D et al. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145:1-11.
  77. Biesalski HK. Diabetes preventive components in the Mediterranean diet. Eur J Nutr 2004; 43 Suppl 1:I/26-I/30.
  78. Panagiotakos DB, Pitsavos C, Chrysohoou C, Stefanadis C. The epidemiology of type 2 diabetes mellitus in Greek adults: the ATTICA study. Diabet Med 2005; 22:1581-1588.
  79. Hung HC, Joshipura KJ, Jiang R et al. Fruit and vegetable intake and risk of major chronic disease. J Natl Cancer Inst 2004; 96:1577-1584.
  80. Fraga CG. Cocoa, diabetes, and hypertension: should we eat more chocolate? Am J Clin Nutr 2005; 81:541-542.
  81. Fraga CG. Cocoa, diabetes, and hypertension: should we eat more chocolate? Am J Clin Nutr 2005; 81:541-542.
  82. Fisher ND, Hughes M, Gerhard-Herman M, Hollenberg NK. Flavanol-rich cocoa induces nitric-oxide-dependent vasodilation in healthy humans. J Hypertens 2003; 21:2281-2286.
  83. Fisher ND, Hollenberg NK. Flavanols for cardiovascular health: the science behind the sweetness. J Hypertens 2005; 23:1453-1459.
  84. Rein D, Lotito S, Holt RR, Keen CL, Schmitz HH, Fraga CG. Epicatechin in human plasma: in vivo determination and effect of chocolate consumption on plasma oxidation status. J Nutr 2000; 130:2109S-2114S.
  85. Rein D, Lotito S, Holt RR, Keen CL, Schmitz HH, Fraga CG. Epicatechin in human plasma: in vivo determination and effect of chocolate consumption on plasma oxidation status. J Nutr 2000; 130:2109S-2114S.
  86. Vlachopoulos C, Aznaouridis K, Alexopoulos N, Economou E, Andreadou I, Stefanadis C. Effect of dark chocolate on arterial function in healthy individuals. Am J Hypertens 2005; 18:785-791.
  87. Grassi D, Lippi C, Necozione S, Desideri G, Ferri C. Short-term administration of dark chocolate is followed by a significant increase in insulin sensitivity and a decrease in blood pressure in healthy persons. Am J Clin Nutr 2005; 81:611-614.
  88. Grassi D, Necozione S, Lippi C et al. Cocoa reduces blood pressure and insulin resistance and improves endothelium-dependent vasodilation in hypertensives. Hypertension 2005; 46:398-405.
  89. Griggs B. Green Pharmacy. A history of herbal medicine. 1st ed. London: Robert Hale, 1981.
  90. Murray MT, Pizzorno JE. Botanical medicine ¿ a modern perspective. Textbook of Natural Medicine. Edinburgh: Churchill Livingstone, 1999.
  91. Bailey CJ, Turner RC. Metformin. N Engl J Med 1996; 334:574-579.
  92. Witters LA. The blooming of the French lilac. J Clin Invest 2001; 108:1105-1107.
  93. Cusi K, DeFronzo RA. Metformin: a review of its metabolic effects. Diabetes Rev 1998; 6:89-131.
  94. Oubre AY, Carlson TJ, King SR, Reaven GM. From plant to patient: An ethnomedical approach to the identification of new drugs for the treatment of NIDDM. Diabetologia 1997; 40:614-617.
  95. Ackerknecht EH. A Short History of Medicine (revised edition). Baltimore: Johns Hopkins Unversity Press, 1982.
  96. Oubre AY, Carlson TJ, King SR, Reaven GM. From plant to patient: An ethnomedical approach to the identification of new drugs for the treatment of NIDDM. Diabetologia 1997; 40:614-617.
  97. Bailey CJ, Day C. Traditional plant medicine as treatments for diabetes. Diabetes Care 1989; 12:553-564.
  98. Marles RJ, Farnsworth NR. Plants as sources of antidiabetic agents. Economic and Medical Plant Research 1993; 6:149-187.
  99. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  100. Haddad PS, Depot M, Settaf A, Cherrah Y. Use of antidiabetic plants in Morocco and Quebec. Diabetes Care 2001; 24:608-609.
  101. Jung M, Park M, Lee HC, Kang YH, Kang ES, Kim SK. Antidiabetic agents from medicinal plants. Curr Med Chem 2006; 13:1203-1218.
  102. Kusano S, Abe H. Antidiabetic activity of white skinned sweet potato (Ipomoea batatas L.) in obese Zucker fatty rats. Biol Pharm Bull 2000; 23:23-26.
  103. Kusano S, Abe H, Tamura H. Isolation of antidiabetic components from white-skinned sweet potato (Ipomoea batatas L.). Biosci Biotechnol Biochem 2001; 65:109-114.
  104. Ludvik BH, Mahdjoobian K, Waldhaeusl W et al. The effect of Ipomoea batatas (Caiapo) on glucose metabolism and serum cholesterol in patients with type 2 diabetes: a randomized study. Diabetes Care 2002; 25:239-240.
  105. Ludvik B, Waldhausl W, Prager R, Kautzky-Willer A, Pacini G. Mode of action of ipomoea batatas (Caiapo) in type 2 diabetic patients. Metabolism 2003; 52:875-880.
  106. Ludvik B, Neuffer B, Pacini G. Efficacy of Ipomoea batatas (Caiapo) on diabetes control in type 2 diabetic subjects treated with diet. Diabetes Care 2004; 27:436-440.
  107. Evans JL, Rushakoff RJ. Oral Pharmacological Agents for Type 2 Diabetes: Sulfonylureas, Meglitinides, Metformin, Thiazolidinediones, a-Glucosidase Inhibitors, and Emerging Approaches. In: Degroot L, Goldfine ID, Rushakoff RJ, editors. Endotext.com: Diabetes and Carbohydrate Metabolism. http://www.medtext.com:MDtext.com, 2002.
  108. Bloomgarden ZT, Dodis R, Viscoli CM, Holmboe ES, Inzucchi SE. Lower baseline glycemia reduces apparent oral agent glucose-lowering efficacy: a meta-regression analysis. Diabetes Care 2006; 29:2137-2139.
  109. Anonymous. PDR for Herbal Medicines. 1st ed ed. Montvale, NJ: Medical Economics Co, 1998.
  110. Brasch E, Ulbricht C, Kuo G, Szapary P, Smith M. Therapeutic applications of fenugreek. Altern Med Rev 2003; 8:20-27.
  111. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  112. Marles RJ, Farnsworth NR. Plants as sources of antidiabetic agents. Economic and Medical Plant Research 1993; 6:149-187.
  113. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  114. Sauvaire Y, Petit P, Broca C et al. 4-Hydroxyisoleucine: a novel amino acid potentiator of insulin secretion. Diabetes 1998; 47:206-210.
  115. Broca C, Manteghetti M, Gross R et al. 4-Hydroxyisoleucine: effects of synthetic and natural analogues on insulin secretion. Eur J Pharmacol 2000; 390:339-345.
  116. Broca C, Gross R, Petit P et al. 4-Hydroxyisoleucine: experimental evidence of its insulinotropic and antidiabetic properties. Am J Physiol 1999; 277:E617-E623.
  117. Broca C, Gross R, Petit P et al. 4-Hydroxyisoleucine: experimental evidence of its insulinotropic and antidiabetic properties. Am J Physiol 1999; 277:E617-E623.
  118. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  119. Madar Z, Abel R, Samish S, Arad J. Glucose-lowering effect of fenugreek in non-insulin dependent diabetics. Eur J Clin Nutr 1988; 42:51-54.
  120. Sharma RD, Sarkar A, Hazra DK. Use of fenugreek seed powder in the management of non-inslin dependent diabetes mellitus. Nutr Res 1996; 16:1331-1339.
  121. Gupta A, Gupta R, Lal B. Effect of Trigonella foenum-graecum (fenugreek) seeds on glycaemic control and insulin resistance in type 2 diabetes mellitus: a double blind placebo controlled study. J Assoc Physicians India 2001; 49:1057-1061.
  122. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  123. Kochhar A, Nagi M. Effect of supplementation of traditional medicinal plants on blood glucose in non-insulin-dependent diabetics: a pilot study. J Med Food 2005; 8:545-549.
  124. Flammang AM, Cifone MA, Erexson GL, Stankowski LF, Jr. Genotoxicity testing of a fenugreek extract. Food Chem Toxicol 2004; 42:1769-1775.
  125. Khan A, Bryden NA, Polansky MM, Anderson RA. Insulin potentiating factor and chromium content of selected foods and spices. Biol Trace Element Res 1990; 24:183-188.
  126. Berrio LF, Polansky MM, Anderson RA. Insulin activity: stimulatory effects of cinnamon and brewer's yeast as influenced by albumin. Horm Res 1992; 37:225-229.
  127. Berrio LF, Polansky MM, Anderson RA. Insulin activity: stimulatory effects of cinnamon and brewer's yeast as influenced by albumin. Horm Res 1992; 37:225-229.
  128. Flammang AM, Cifone MA, Erexson GL, Stankowski LF, Jr. Genotoxicity testing of a fenugreek extract. Food Chem Toxicol 2004; 42:1769-1775.
  129. Evans JL, Jallal B. Protein tyrosine phosphatases: their role in insulin action and potential as drug targets. Exp Opin Invest Drugs 1999; 8:139-160.
  130. Imparl-Radosevich J, Deas S, Polansky MM et al. Regulation of PTP-1 and insulin receptor kinase by fractions from cinnamon: implications for cinnamon regulation of insulin signalling. Horm Res 1998; 50:177-182.
  131. Khan A, Safdar M, li Khan MM, Khattak KN, Anderson RA. Cinnamon improves glucose and lipids of people with type 2 diabetes. Diabetes Care 2003; 26:3215-3218.
  132. Mang B, Wolters M, Schmitt B et al. Effects of a cinnamon extract on plasma glucose, HbA, and serum lipids in diabetes mellitus type 2. Eur J Clin Invest 2006; 36:340-344.
  133. Bloomgarden ZT, Dodis R, Viscoli CM, Holmboe ES, Inzucchi SE. Lower baseline glycemia reduces apparent oral agent glucose-lowering efficacy: a meta-regression analysis. Diabetes Care 2006; 29:2137-2139.
  134. Vanschoonbeek K, Thomassen BJ, Senden JM, Wodzig WK, van Loon LJ. Cinnamon supplementation does not improve glycemic control in postmenopausal type 2 diabetes patients. J Nutr 2006; 136:977-980.
  135. Marles RJ, Farnsworth NR. Plants as sources of antidiabetic agents. Economic and Medical Plant Research 1993; 6:149-187.
  136. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  137. Grover JK, Yadav S, Vats V. Medicinal plants of India with anti-diabetic potential. J Ethnopharmacol 2002; 81:81-100.
  138. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  139. Grover JK, Yadav S, Vats V. Medicinal plants of India with anti-diabetic potential. J Ethnopharmacol 2002; 81:81-100.
  140. Nag B, Medicherla S, Sharma SD. Orally active fraction of momordica charantia, active peptides thereof, and their use in the treatment of diabetes. US Patent 2002; 6,391,854:1-14.
  141. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  142. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  143. Krawinkel MB, Keding GB. Bitter gourd (Momordica Charantia): A dietary approach to hyperglycemia. Nutr Rev 2006; 64:331-337.
  144. Nag B, Medicherla S, Sharma SD. Orally active fraction of momordica charantia, active peptides thereof, and their use in the treatment of diabetes. US Patent 2002; 6,391,854:1-14.
  145. Krawinkel MB, Keding GB. Bitter gourd (Momordica Charantia): A dietary approach to hyperglycemia. Nutr Rev 2006; 64:331-337.
  146. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  147. Nag B, Medicherla S, Sharma SD. Orally active fraction of momordica charantia, active peptides thereof, and their use in the treatment of diabetes. US Patent 2002; 6,391,854:1-14.
  148. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  149. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  150. Basch E, Gabardi S, Ulbricht C. Bitter melon (Momordica charantia): a review of efficacy and safety. Am J Health Syst Pharm 2003; 60:356-359.
  151. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  152. Basch E, Gabardi S, Ulbricht C. Bitter melon (Momordica charantia): a review of efficacy and safety. Am J Health Syst Pharm 2003; 60:356-359.
  153. Grover JK, Yadav S, Vats V. Medicinal plants of India with anti-diabetic potential. J Ethnopharmacol 2002; 81:81-100.
  154. Marles RJ, Farnsworth NR. Plants as sources of antidiabetic agents. Economic and Medical Plant Research 1993; 6:149-187.
  155. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  156. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  157. Shapiro K, Gong WC. Natural products used for diabetes. J Am Pharm Assoc (Wash ) 2002; 42:217-226.
  158. Roman-Ramos R, Flores-Saenz JL, Alarcon-Aguilar FJ. Anti-hyperglycemic effect of some edible plants. J Ethnopharmacol 1995; 48:25-32.
  159. Marles RJ, Farnsworth NR. Plants as sources of antidiabetic agents. Economic and Medical Plant Research 1993; 6:149-187.
  160. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  161. El Kossori RL, Villaume C, El Boustani E, Sauvaire Y, Mejean L. Composition of pulp, skin and seeds of prickly pears fruit (Opuntia ficus indica sp.). Plant Foods Hum Nutr 1998; 52:263-270.
  162. Roman-Ramos R, Flores-Saenz JL, Alarcon-Aguilar FJ. Anti-hyperglycemic effect of some edible plants. J Ethnopharmacol 1995; 48:25-32.
  163. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  164. Roman-Ramos R, Flores-Saenz JL, Alarcon-Aguilar FJ. Anti-hyperglycemic effect of some edible plants. J Ethnopharmacol 1995; 48:25-32.
  165. Frati AC, Gordillo BE, Altamirano P, Ariza CR, Cortes-Franco R, Chavez-Negrete A. Acute hypoglycemic effect of Opuntia streptacantha Lemaire in NIDDM. Diabetes Care 1990; 13:455-456.
  166. Frati-Munari AC, Gordillo BE, Altamirano P, Ariza CR. Hypoglycemic effect of Opuntia streptacantha Lemaire in NIDDM. Diabetes Care 1988; 11:63-66.
  167. Frati AC, Gordillo BE, Altamirano P, Ariza CR, Cortes-Franco R, Chavez-Negrete A. Acute hypoglycemic effect of Opuntia streptacantha Lemaire in NIDDM. Diabetes Care 1990; 13:455-456.
  168. Grover JK, Yadav S, Vats V. Medicinal plants of India with anti-diabetic potential. J Ethnopharmacol 2002; 81:81-100.
  169. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  170. Azad Khan AK, Akhtar S, Mahtab H. Coccinia indica in the treatment of patients with diabetes mellitus. Bangladesh Med Res Counc Bull 1979; 5:60-66.
  171. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  172. Griggs B. Green Pharmacy. A history of herbal medicine. 1st ed. London: Robert Hale, 1981.
  173. Ackerknecht EH. A Short History of Medicine (revised edition). Baltimore: Johns Hopkins Unversity Press, 1982.
  174. Xie JT, Mchendale S, Yuan CS. Ginseng and diabetes. Am J Chin Med 2005; 33:397-404.
  175. Vogler BK, Pittler MH, Ernst E. The efficacy of ginseng. A systematic review of randomised clinical trials. Eur J Clin Pharmacol 1999; 55:567-575.
  176. Marles RJ, Farnsworth NR. Plants as sources of antidiabetic agents. Economic and Medical Plant Research 1993; 6:149-187.
  177. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  178. Marles RJ, Farnsworth NR. Plants as sources of antidiabetic agents. Economic and Medical Plant Research 1993; 6:149-187.
  179. Marles RJ, Farnsworth NR. Antidiabetic plants and their active constituents. Phytomed 1995; 2:137-189.
  180. Vogler BK, Pittler MH, Ernst E. The efficacy of ginseng. A systematic review of randomised clinical trials. Eur J Clin Pharmacol 1999; 55:567-575.
  181. Sievenpiper JL, Arnason JT, Vidgen E, Leiter LA, Vuksan V. A systematic quantitative analysis of the literature of the high variability in ginseng (Panax spp.): should ginseng be trusted in diabetes? Diabetes Care 2004; 27:839-840.
  182. Vuksan V, Sievenpiper JL. Herbal remedies in the management of diabetes: lessons learned from the study of ginseng. Nutr Metab Cardiovasc Dis 2005; 15:149-160.
  183. Sotaniemi EA, Haapakoski E, Rautio A. Ginseng therapy in non-insulin-dependent diabetic patients. Diabetes Care 1995; 18:1373-1375.
  184. Vuksan V, Sievenpiper JL, Koo VY et al. American ginseng (Panax quinquefolius L) reduces postprandial glycemia in nondiabetic subjects and subjects with type 2 diabetes mellitus. Arch Intern Med 2000; 160:1009-1013.
  185. Vogler BK, Ernst E. Aloe vera: a systematic review of its clinical effectiveness. Br J Gen Pract 1999; 49:823-828.
  186. Grover JK, Yadav S, Vats V. Medicinal plants of India with anti-diabetic potential. J Ethnopharmacol 2002; 81:81-100.
  187. Al Rowais NA. Herbal medicine in the treatment of diabetes mellitus. Saudi Med J 2002; 23:1327-1331.
  188. Ghannam N, Kingston M, Al Meshaal IA, Tariq M, Parman NS, Woodhouse N. The antidiabetic activity of aloes: preliminary clinical and experimental observations. Horm Res 1986; 24:288-294.
  189. Yongchaiyudha S, Rungpitarangsi V, Bunyapraphatsara N, Chokechaijaroenporn O. Antidiabetic activity of Aloe vera L. juice: I. Clinical trial in new cases of diabetes mellitus. Phytomed 1996; 3:241-243.
  190. Bunyapraphatsara N, Yongchaiyudha S, Rungpitarangsi V, Chokechaijaroenporn O. Antidiabetic activity of Aloe vera L. juice: II. Clinical trial in diabetes mellitus patients in combination with glibenclamide. Phytomed 1996; 3:245-248.
  191. Vuksan V, Sievenpiper JL, Owen R et al. Beneficial effects of viscous dietary fiber from Konjac-mannan in subjects with the insulin resistance syndrome: results of a controlled metabolic trial. Diabetes Care 2000; 23:9-14.
  192. Vuksan V, Jenkins DJA, Spadafora P et al. Konjac-Mannan (Glucomannan) improves glycemia and other associated risk factors for coronary heart disease in type 2 diabetes - A randomized controlled metabolic trial. Diabetes Care 1999; 22:913-919.
  193. Banerjee SK, Maulik SK. Effect of garlic on cardiovascular disorders: a review. Nutr J 2002; 1:4.
  194. Banerjee SK, Maulik SK. Effect of garlic on cardiovascular disorders: a review. Nutr J 2002; 1:4.
  195. Borek C. Garlic reduces dementia and heart-disease risk. J Nutr 2006; 136:810S-812S.
  196. Budoff M. Aged garlic extract retards progression of coronary artery calcification. J Nutr 2006; 136:741S-744S.
  197. Ahmad MS, Ahmed N. Antiglycation properties of aged garlic extract: possible role in prevention of diabetic complications. J Nutr 2006; 136:796S-799S.
  198. Kendler BS. Garlic (Allium sativum) and onion (Allium cepa): a review of their relationship to cardiovascular disease. Prev Med 1987; 16:670-685.
  199. Keys A. Wine, garlic, and CHD in seven countries. Lancet 1980; 1:145-146.
  200. Kris-Etherton PM, Etherton TD, Carlson J, Gardner C. Recent discoveries in inclusive food-based approaches and dietary patterns for reduction in risk for cardiovascular disease. Curr Opin Lipidol 2002; 13:397-407.
  201. Banerjee SK, Maulik SK. Effect of garlic on cardiovascular disorders: a review. Nutr J 2002; 1:4.
  202. Ashraf R, Aamir K, Shaikh AR, Ahmed T. Effects of garlic on dyslipidemia in patients with type 2 diabetes mellitus. J Ayub Med Coll Abbottabad 2005; 17:60-64.
  203. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  204. Banerjee SK, Maulik SK. Effect of garlic on cardiovascular disorders: a review. Nutr J 2002; 1:4.
  205. Belleville,J: The French paradox: possible involvement of ethanol in the protective effect against cardiovascular diseases. Nutrition 18:173-177, 2002
  206. Opie,LH, Lecour,S: The red wine hypothesis: from concepts to protective signalling molecules. Eur Heart J 28:1683-1693, 2007
  207. Providencia,R: Cardiovascular protection from alcoholic drinks: scientific basis of the French Paradox. Rev Port Cardiol 25:1043-1058, 2006
  208. Baur,JA, Sinclair,DA: Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov 5:493-506, 2006
  209. Baur,JA, Sinclair,DA: Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov 5:493-506, 2006
  210. Jang,M, Cai,L, Udeani,GO, Slowing,KV, Thomas,CF, Beecher,CW, Fong,HH, Farnsworth,NR, Kinghorn,AD, Mehta,RG, Moon,RC, Pezzuto,JM: Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 275:218-220, 1997
  211. Harikumar,KB, Aggarwal,BB: Resveratrol: A multitargeted agent for age-associated chronic diseases. Cell Cycle 7:1-18, 2008
  212. Orallo,F: Trans-resveratrol: a magical elixir of eternal youth? Curr Med Chem 15:1887-1898, 2008
  213. Bass,TM, Weinkove,D, Houthoofd,K, Gems,D, Partridge,L: Effects of resveratrol on lifespan in Drosophila melanogaster and Caenorhabditis elegans. Mech Ageing Dev 128:546-552, 2007
  214. Barger,JL, Kayo,T, Vann,JM, Arias,EB, Wang,J, Hacker,TA, Wang,Y, Raederstorff,D, Morrow,JD, Leeuwenburgh,C, Allison,DB, Saupe,KW, Cartee,GD, Weindruch,R, Prolla,TA: A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS ONE 3:e2264 (1)-e2264 (10), 2008
  215. Pearson,KJ, Baur,JA, Lewis,KN, Peshkin,L, Price,NL, Labinskyy,N, Swindell,WR, Kamara,D, Minor,RK, Perez,E, Jamieson,HA, Zhang,Y, Dunn,SR, Sharma,K, Pleshko,N, Woollett,LA, Csiszar,A, Ikeno,Y, Le,CD, Elliott,PJ, Becker,KG, Navas,P, Ingram,DK, Wolf,NS, Ungvari,Z, Sinclair,DA, de,CR: Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab 8:157-168, 2008
  216. Lagouge,M, Argmann,C, Gerhart-Hines,Z, Meziane,H, Lerin,C, Daussin,F, Messadeq,N, Milne,J, Lambert,P, Elliott,P, Geny,B, Laakso,M, Puigserver,P, Auwerx,J: Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 127:1109-1122, 2006
  217. Milne,JC, Lambert,PD, Schenk,S, Carney,DP, Smith,JJ, Gagne,DJ, Jin,L, Boss,O, Perni,RB, Vu,CB, Bemis,JE, Xie,R, Disch,JS, Ng,PY, Nunes,JJ, Lynch,AV, Yang,H, Galonek,H, Israelian,K, Choy,W, Iffland,A, Lavu,S, Medvedik,O, Sinclair,DA, Olefsky,JM, Jirousek,MR, Elliott,PJ, Westphal,CH: Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450:712-716, 2007
  218. Sakamoto,K: Silencing metabolic disorders by novel SIRT1 activators. Cell Metab 7:3-4, 2008
  219. Pirola,L, Frojdo,S: Resveratrol: one molecule, many targets. IUBMB Life 60:323-332, 2008
  220. Lagouge,M, Argmann,C, Gerhart-Hines,Z, Meziane,H, Lerin,C, Daussin,F, Messadeq,N, Milne,J, Lambert,P, Elliott,P, Geny,B, Laakso,M, Puigserver,P, Auwerx,J: Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 127:1109-1122, 2006
  221. Milne,JC, Lambert,PD, Schenk,S, Carney,DP, Smith,JJ, Gagne,DJ, Jin,L, Boss,O, Perni,RB, Vu,CB, Bemis,JE, Xie,R, Disch,JS, Ng,PY, Nunes,JJ, Lynch,AV, Yang,H, Galonek,H, Israelian,K, Choy,W, Iffland,A, Lavu,S, Medvedik,O, Sinclair,DA, Olefsky,JM, Jirousek,MR, Elliott,PJ, Westphal,CH: Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450:712-716, 2007
  222. Zabolotny,JM, Kim,YB: Silencing insulin resistance through SIRT1. Cell Metab 6:247-249, 2007
  223. Sun,C, Zhang,F, Ge,X, Yan,T, Chen,X, Shi,X, Zhai,Q: SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab 6:307-319, 2007
  224. Elliott,PJ, Jirousek,M: Sirtuins: Novel targets for metabolic disease. Curr Opin Investig Drugs 9:371-378, 2008
  225. Covington MB. Traditional Chinese medicine in the treament of diabetes. Diabetes Spectrum 2001; 14:154-159.
  226. Jia W, Gao W, Tang L. Antidiabetic herbal drugs officially approved in China. Phytother Res 2003; 17:1127-1134.
  227. Liu JP, Zhang M, Wang WY, Grimsgaard S. Chinese herbal medicines for type 2 diabetes mellitus. Cochrane Database Syst Rev 2004; Issue 3:CD003642.
  228. Li WL, Zheng HC, Bukuru J, De KN. Natural medicines used in the traditional Chinese medical system for therapy of diabetes mellitus. J Ethnopharmacol 2004; 92:1-21.
  229. Jia W, Gao WY, Xiao PG. Antidiabetic drugs of plant origin used in China: compositions, pharmacology, and hypoglycemic mechanisms. Zhongguo Zhong Yao Za Zhi 2003; 28:108-113.
  230. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001; 414:813-820.
  231. Evans JL, Maddux BA, Goldfine ID. The molecular basis for oxidative stress-induced insulin resistance. Antioxid Redox Signal 2004; 7:1040-1052.
  232. Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative stress-activated signaling pathways mediators of insulin resistance and dysfunction? Diabetes 2003; 52:1-8.
  233. Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Oxidative stress and stress-activated signaling pathways: a unifying hypothesis of type 2 diabetes. Endocr Rev 2002; 23:599-622.
  234. Evans JL, Maddux BA, Goldfine ID. Antioxidants in diabetic complications and insulin resistance. In: Raz I, Skyler JS, Shafrir E, editors. Diabetes: From Research to Diagnosis and Treatment. London: Martin Dunitz, 2003: 479-496.
  235. Antioxidants in Diabetes Management. 1st ed. New York: Marcel Dekker, 2000.
  236. Evans JL, Youngren JF, Goldfine ID. Effective treatments for insulin resistance: trim the fat and douse the fire. Trends Endocrinol Metab 2004; 15:425-431.
  237. Henriksen EJ. Exercise training and the antioxidant alpha-lipoic acid in the treatment of insulin resistance and type 2 diabetes. Free Radic Biol Med 2006; 40:3-12.
  238. Packer L, Witt EH, Tritschler HJ. alpha-Lipoic acid as a biological antioxidant. Free Radic Biol Med 1995; 19:227-250.
  239. Packer L, Witt EH, Tritschler HJ. Antioxidant properties and clinical applications of alpha-lipoic acid and dihydrolipoic acid. In: Cadenas E, Packer L, editors. Handbook of Antioxidants. New York: Dekker, 1998: 545-591.
  240. Fuchs J, Packer L, Zimmer G. Lipoic Acid in Health and Disease. 1st ed. New York: Marcel Dekker, 1997.
  241. Reed LJ, DeBusk BG, Gunsalus IC, Hornberger JrCS. Crystalline a-lipoic acid: a catalytic agent associated with pyruvate dehydrogenase. Science 1951; 114:93-94.
  242. Biewenga GP, Haenen GR, Bast A. The pharmacology of the antioxidant lipoic acid. Gen Pharmacol 1997; 29:315-331.
  243. Biewenga GP, Haenen GR, Bast A. The role of lipoic acid in the treatment of diabetic polyneuropathy. Drug Metab Rev 1997; 29:1025-1054.
  244. Ziegler D, Gries FA. Alpha-lipoic acid in the treatment of diabetic peripheral and cardiac autonomic neuropathy. Diabetes 1997; 46 Suppl 2:S62-S66.
  245. Ziegler D, Reljanovic M, Mehnert H, Gries FA. Alpha-lipoic acid in the treatment of diabetic polyneuropathy in Germany: current evidence from clinical trials. Exp Clin Endocrinol Diabetes 1999; 107:421-430.
  246. Ziegler D, Reljanovic M, Mehnert H, Gries FA. Alpha-lipoic acid in the treatment of diabetic polyneuropathy in Germany: current evidence from clinical trials. Exp Clin Endocrinol Diabetes 1999; 107:421-430.
  247. Ziegler D, Nowak H, Kempler P, Vargha P, Low PA. Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a meta-analysis. Diabet Med 2004; 21:114-121.
  248. Ziegler D. Thioctic acid for patients with symptomatic diabetic polyneuropathy: a critical review. Treat Endocrinol 2004; 3:173-189.
  249. Gidal BE. New and emerging treatment options for neuropathic pain. Am J Manag Care 2006; 12 (9 Suppl):S269-S278.
  250. Ziegler D, Ametov A, Barinov A et al. Oral treatment with alpha-lipoic acid improves symptomatic diabetic polyneuropathy: the SYDNEY 2 trial. Diabetes Care 2006; 29:2365-2370.
  251. Doggrell SA. Alpha-lipoic acid, an anti-obesity agent? Expert Opin Investig Drugs 2004; 13:1641-1643.
  252. Jacob S, Henriksen EJ, Schiemann AL et al. Enhancement of glucose disposal in patients with type 2 diabetes by alpha-lipoic acid. Arzneimittel-Forschung 1995; 45:872-874.
  253. Jacob S, Henriksen EJ, Tritschler HJ, Augustin HJ, Dietze GJ. Improvement of insulin-stimulated glucose-disposal in type 2 diabetes after repeated parenteral administration of thioctic acid. Exp Clin Endocrinol Diabetes 1996; 104:284-288.
  254. Evans JL, Goldfine ID. a-Lipoic acid: a multi-functional antioxidant that improves insulin sensitivity in patients with type 2 diabetes. Diabetes Technol Therap 2000; 2:401-413.
  255. Jacob S, Rett K, Henriksen EJ, Haring HU. Thioctic acid: effects on insulin sensitivity and glucose metabolism. BioFactors 1999; 10:169-174.
  256. Jacob S, Henriksen EJ, Schiemann AL et al. Enhancement of glucose disposal in patients with type 2 diabetes by alpha-lipoic acid. Arzneimittel-Forschung 1995; 45:872-874.
  257. Jacob S, Henriksen EJ, Tritschler HJ, Augustin HJ, Dietze GJ. Improvement of insulin-stimulated glucose-disposal in type 2 diabetes after repeated parenteral administration of thioctic acid. Exp Clin Endocrinol Diabetes 1996; 104:284-288.
  258. Jacob S, Ruus P, Hermann R et al. Oral administration of RAC-alpha-lipoic acid modulates insulin sensitivity in patients with type 2 diabetes mellitus: a placebo-controlled pilot trial. Free Radic Biol Med 1999; 27:309-314.
  259. Konrad T, Vicini P, Kusterer K et al. Alpha-lipoic acid treatment decreases serum lactate and pyruvate concentrations and improves glucose effectiveness in lean and obese patients with type 2 diabetes. Diabetes Care 1999; 22:280-287.
  260. Evans JL, Heymann CJ, Goldfine ID, Gavin LA. Pharmacokinetics, tolerability, and fructosamine-lowering effect of a novel, controlled release formulation of a-lipoic acid. Endocr Pract 2002; 8:29-35.
  261. Evans JL, Heymann CJ, Goldfine ID, Gavin LA. Pharmacokinetics, tolerability, and fructosamine-lowering effect of a novel, controlled release formulation of a-lipoic acid. Endocr Pract 2002; 8:29-35.
  262. Pjodkowaki RA, Grotz VL, Mudalar S, Henry RR. The effects of alpha-lipoic acid on glucose homeostasis in type 2 diabetes. J Invest Med 50, 71A-71A (Abstract 382). 2002. Ref Type: Abstract
  263. Rudich A, Tirosh A, Potashnik R, Khamaisi M, Bashan N. Lipoic acid protects against oxidative stress induced impairment in insulin stimulation of protein kinase B and glucose transport in 3T3-L1 adipocytes. Diabetologia 1999; 42:949-957.
  264. Maddux BA, See W, Lawrence JC, Jr., Goldfine AL, Goldfine ID, Evans JL. Protection against oxidative stress-induced insulin resistance in rat L6 muscle cells by micromolar concentrations of a-lipoic acid. Diabetes 2001; 50:404-410.
  265. Zhang WJ, Frei B. Alpha-lipoic acid inhibits TNF-alpha-induced NF-kappaB activation and adhesion molecule expression in human aortic endothelial cells. FASEB J 2001; 15:2423-2432.
  266. Birnbaum MJ. Turning down insulin signaling. J Clin Invest 2001; 108:655-659.
  267. Evans JL, Maddux BA, Goldfine ID. The molecular basis for oxidative stress-induced insulin resistance. Antioxid Redox Signal 2004; 7:1040-1052.
  268. Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 1997; 336:1066-1071.
  269. Mohamed AK, Bierhaus A, Schiekofer S, Tritschler H, Ziegler R, Nawroth PP. The role of oxidative stress and NF-kB activation in late diabetic complications. BioFactors 1999; 10:157-167.
  270. Bierhaus A, Schiekofer S, Schwaninger M et al. Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB. Diabetes 2001; 50:2792-2808.
  271. Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 1997; 336:1066-1071.
  272. Bierhaus A, Schiekofer S, Schwaninger M et al. Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB. Diabetes 2001; 50:2792-2808.
  273. Zhang WJ, Frei B. Alpha-lipoic acid inhibits TNF-alpha-induced NF-kappaB activation and adhesion molecule expression in human aortic endothelial cells. FASEB J 2001; 15:2423-2432.
  274. Bierhaus A, Chevion S, Chevion M et al. Advanced glycation end product-induced activation of NF-kappaB is suppressed by alpha-lipoic acid in cultured endothelial cells. Diabetes 1997; 46:1481-1490.
  275. Packer L. alpha-Lipoic acid: a metabolic antioxidant which regulates NF-kappa B signal transduction and protects against oxidative injury. Drug Metab Rev 1998; 30:245-275.
  276. Hofmann MA, Schiekofer S, Kanitz M et al. Insufficient glycemic control increases nuclear factor-kappa B binding activity in peripheral blood mononuclear cells isolated from patients with type 1 diabetes. Diabetes Care 1998; 21:1310-1316.
  277. Hofmann MA, Schiekofer S, Isermann B et al. Peripheral blood mononuclear cells isolated from patients with diabetic nephropathy show increased activation of the oxidative-stress sensitive transcription factor NF-kappaB. Diabetologia 1999; 42:222-232.
  278. Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative stress-activated signaling pathways mediators of insulin resistance and ¿ dysfunction? Diabetes 2003; 52:1-8.
  279. Ruan H, Hacohen N, Golub TR, Van Parijs L, Lodish HF. Tumor necrosis factor-alpha suppresses adipocyte-specific genes and activates expression of preadipocyte genes in 3T3-L1 adipocytes: nuclear factor-kappaB activation by TNF-alpha is obligatory. Diabetes 2002; 51:1319-1336.
  280. Rossi A, Kapahi P, Natoli G et al. Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors of IkappaB kinase. Nature 2000; 403:103-108.
  281. Yuan M, Konstantopoulos N, Lee J et al. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of IKKb. Science 2001; 293:1673-1677.
  282. Kim JK, Kim YJ, Fillmore JJ et al. Prevention of fat-induced insulin resistance by salicylate. J Clin Invest 2001; 108:437-446.
  283. Hundal RS, Petersen KF, Mayerson AB et al. Mechanism by which high-dose aspirin improves glucose metabolism in type 2 diabetes. J Clin Invest 2002; 109(10):1321-1326.
  284. Ziegler D, Reljanovic M, Mehnert H, Gries FA. Alpha-lipoic acid in the treatment of diabetic polyneuropathy in Germany: current evidence from clinical trials. Exp Clin Endocrinol Diabetes 1999; 107:421-430.
  285. Mohamed AK, Bierhaus A, Schiekofer S, Tritschler H, Ziegler R, Nawroth PP. The role of oxidative stress and NF-kB activation in late diabetic complications. BioFactors 1999; 10:157-167.
  286. Barbul A. Arginine: biochemistry, physiology, and therapeutic implications. J Parenter Enteral Nutr 1986; 10:227-238.
  287. Moncada S, Higgs A. The L-arginine-nitric oxide pathway. N Engl J Med 1993; 329:2002-2012.
  288. Fried R, Merrell WC. The Arginine Solution. 1 ed. New York: Warner Books, 1999.
  289. Palmer RM, Ashton DS, Moncada S. Vascular endothelial cells synthesize nitric oxide from L-arginine. Nature 1988; 333:664-666.
  290. Moncada S, Higgs A. The L-arginine-nitric oxide pathway. N Engl J Med 1993; 329:2002-2012.
  291. Kobzik L, Reid MB, Bredt DS, Stamler JS. Nitric oxide in skeletal muscle. Nature 1994; 372:546-548.
  292. Reid MB. Role of nitric oxide in skeletal muscle: synthesis, distribution and functional importance. Acta Physiol Scand 1998; 162:401-409
  293. Luscher TF, Noll G. The pathogenesis of cardiovascular disease: role of the endothelium as a target and mediator. Atherosclerosis 1995; 118 (Suppl):S81-S90.
  294. Zeiher AM, Drexler H, Saurbier B, Just H. Endothelium-mediated coronary blood flow modulation in humans. Effects of age, atherosclerosis, hypercholesterolemia, and hypertension. J Clin Invest 1993; 92:652-662.
  295. Tschudi MR, Barton M, Bersinger NA et al. Effect of age on kinetics of nitric oxide release in rat aorta and pulmonary artery. J Clin Invest 1996; 98:899-905.
  296. Pieper GM. Review of alterations in endothelial nitric oxide production in diabetes: protective role of arginine on endothelial dysfunction. Hypertension 1998; 31:1047-1060.
  297. Honing ML, Morrison PJ, Banga JD, Stroes ES, Rabelink TJ. Nitric oxide availability in diabetes mellitus. Diabetes Metab Rev 1998; 14:241-249.
  298. Honing ML, Morrison PJ, Banga JD, Stroes ES, Rabelink TJ. Nitric oxide availability in diabetes mellitus. Diabetes Metab Rev 1998; 14:241-249.
  299. Gewaltig MT, Kojda G. Vasoprotection by nitric oxide: mechanisms and therapeutic potential. Cardiovasc Res 2002; 55:250-260.
  300. Gewaltig MT, Kojda G. Vasoprotection by nitric oxide: mechanisms and therapeutic potential. Cardiovasc Res 2002; 55:250-260.
  301. Boger RH, Bode-Boger SM. The clinical pharmacology of L-arginine. Annu Rev Pharmacol Toxicol 2001; 41:79-99.
  302. Fried R, Merrell WC. The Arginine Solution. 1 ed. New York: Warner Books, 1999.
  303. Cooke JP, Zimmer J. The Cardiovascular Cure. How to Strengthen Your Self-Defense Against Heart Attack and Stroke. 1st ed. New York: Broadway, 2002.
  304. Preli RB, Klein KP, Herrington DM. Vascular effects of dietary L-arginine supplementation. Atherosclerosis 2002; 162:1-15.
  305. Preli RB, Klein KP, Herrington DM. Vascular effects of dietary L-arginine supplementation. Atherosclerosis 2002; 162:1-15.
  306. Preli RB, Klein KP, Herrington DM. Vascular effects of dietary L-arginine supplementation. Atherosclerosis 2002; 162:1-15.
  307. Lekakis JP, Papathanassiou S, Papaioannou TG et al. Oral L-arginine improves endothelial dysfunction in patients with essential hypertension. Int J Cardiol 2002; 86:317-323.
  308. Maxwell AJ, Anderson B, Zapien MP, Cooke JP. Endothelial dysfunction in hypercholesterolemia is reversed by a nutritional product designed to enhance nitric oxide activity. Cardiovasc Drugs Ther 2000; 14:309-316.
  309. Clarkson P, Adams MR, Powe AJ et al. Oral L-arginine improves endothelium-dependent dilation in hypercholesterolemic young adults. J Clin Invest 1996; 97:1989-1994.
  310. Adams MR, McCredie R, Jessup W, Robinson J, Sullivan D, Celermajer DS. Oral L-arginine improves endothelium-dependent dilatation and reduces monocyte adhesion to endothelial cells in young men with coronary artery disease. Atherosclerosis 1997; 129:261-269.
  311. Lerman A, Burnett JC, Jr., Higano ST, McKinley LJ, Holmes DR, Jr. Long-term L-arginine supplementation improves small-vessel coronary endothelial function in humans. Circulation 1998; 97:2123-2128.
  312. Huynh NT, Tayek JA. Oral arginine reduces systemic blood pressure in type 2 diabetes: its potential role in nitric oxide generation. J Am Coll Nutr 2002; 21:422-427.
  313. Mehta S, Stewart DJ, Levy RD. The hypotensive effect of L-arginine is associated with increased expired nitric oxide in humans. Chest 1996; 109:1550-1555.
  314. Facchinetti F, Longo M, Piccinini F, Neri I, Volpe A. L-arginine infusion reduces blood pressure in preeclamptic women through nitric oxide release. J Soc Gynecol Investig 1999; 6:202-207.
  315. Kelly BS, Alexander JW, Dreyer D et al. Oral arginine improves blood pressure in renal transplant and hemodialysis patients. J Parenter Enteral Nutr 2001; 25:194-202.
  316. Setoguchi S, Hirooka Y, Eshima K, Shimokawa H, Takeshita A. Tetrahydrobiopterin improves impaired endothelium-dependent forearm vasodilation in patients with heart failure. J Cardiovasc Pharmacol 2002; 39:363-368.
  317. Blum A, Hathaway L, Mincemoyer R et al. Effects of oral L-arginine on endothelium-dependent vasodilation and markers of inflammation in healthy postmenopausal women. J Am Coll Cardiol 2000; 35:271-276.
  318. Blum A, Hathaway L, Mincemoyer R et al. Oral L-arginine in patients with coronary artery disease on medical management. Circulation 2000; 101:2160-2164.
  319. Preli RB, Klein KP, Herrington DM. Vascular effects of dietary L-arginine supplementation. Atherosclerosis 2002; 162:1-15.
  320. Preli RB, Klein KP, Herrington DM. Vascular effects of dietary L-arginine supplementation. Atherosclerosis 2002; 162:1-15.
  321. Knekt P, Ritz J, Pereira MA et al. Antioxidant vitamins and coronary heart disease risk: a pooled analysis of 9 cohorts. Am J Clin Nutr 2004; 80:1508-1520.
  322. Osganian SK, Stampfer MJ, Rimm E et al. Vitamin C and risk of coronary heart disease in women. J Am Coll Cardiol 2003; 42:246-252.
  323. Wannamethee SG, Lowe GD, Rumley A, Bruckdorfer KR, Whincup PH. Associations of vitamin C status, fruit and vegetable intakes, and markers of inflammation and hemostasis. Am J Clin Nutr 2006; 83:567-574.
  324. Boekholdt SM, Meuwese MC, Day NE et al. Plasma concentrations of ascorbic acid and C-reactive protein, and risk of future coronary artery disease, in apparently healthy men and women: the EPIC-Norfolk prospective population study. Br J Nutr 2006; 96:516-522.
  325. Lee DH, Folsom AR, Harnack L, Halliwell B, Jacobs DR, Jr. Does supplemental vitamin C increase cardiovascular disease risk in women with diabetes? Am J Clin Nutr 2004; 80:1194-1200.
  326. Levine GN, Keaney JF, Jr., Vita JA. Cholesterol reduction in cardiovascular disease. Clinical benefits and possible mechanisms. N Engl J Med 1995; 332(8):512-521.
  327. Honing ML, Morrison PJ, Banga JD, Stroes ES, Rabelink TJ. Nitric oxide availability in diabetes mellitus. Diabetes Metab Rev 1998; 14:241-249.
  328. Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res 2000; 87:840-844.
  329. Laight DW, Carrier MJ, Anggard EE. Antioxidants, diabetes and endothelial dysfunction. Cardiovasc Res 2000; 47:457-464.
  330. Laight DW, Carrier MJ, Anggard EE. Antioxidants, diabetes and endothelial dysfunction. Cardiovasc Res 2000; 47:457-464.
  331. Baron AD, Quon MJ. Insulin action and endothelial function. In: Reaven GM, Laws A, editors. Insulin Resistance: The Metabolic Syndrome X. Totowa: Humana Press, 1999: 247-263.
  332. Calles-Escandon J, Cipolla M. Diabetes and endothelial dysfunction: a clinical perspective. Endocr Rev 2001; 22:36-52.
  333. Perticone F, Ceravolo R, Candigliota M et al. Obesity and body fat distribution induce endothelial dysfunction by oxidative stress: protective effect of vitamin C. Diabetes 2001; 50:159-165.
  334. Timimi FK, Ting HH, Haley EA, Roddy MA, Ganz P, Creager MA. Vitamin C improves endothelium-dependent vasodilation in patients with insulin-dependent diabetes mellitus. J Am Coll Cardiol 1998; 31:552-557.
  335. Ting HH, Timimi FK, Boles KS, Creager SJ, Ganz P, Creager MA. Vitamin C improves endothelium-dependent vasodilation in patients with non-insulin-dependent diabetes mellitus. J Clin Invest 1996; 97:22-28.
  336. Lekakis JP, Anastasiou EA, Papamichael CM et al. Short-term oral ascorbic acid improves endothelium-dependent vasodilatation in women with a history of gestational diabetes mellitus. Diabetes Care 2000; 23:1432-1434.
  337. Levine GN, Frei B, Koulouris SN, Gerhard MD, Keaney JF, Jr., Vita JA. Ascorbic acid reverses endothelial vasomotor dysfunction in patients with coronary artery disease. Circulation 1996; 93:1107-1113.
  338. Gokce N, Keaney JF, Jr., Frei B et al. Long-term ascorbic acid administration reverses endothelial vasomotor dysfunction in patients with coronary artery disease. Circulation 1999; 99(25):3234-3240.
  339. Gokce N, Keaney JF, Jr., Frei B et al. Long-term ascorbic acid administration reverses endothelial vasomotor dysfunction in patients with coronary artery disease. Circulation 1999; 99(25):3234-3240.
  340. Baron AD, Quon MJ. Insulin action and endothelial function. In: Reaven GM, Laws A, editors. Insulin Resistance: The Metabolic Syndrome X. Totowa: Humana Press, 1999: 247-263.
  341. Hirai N, Kawano H, Hirashima O et al. Insulin resistance and endothelial dysfunction in smokers: effects of vitamin C. Am J Physiol 2000; 279:H1172-H1178.
  342. Hirashima O, Kawano H, Motoyama T et al. Improvement of endothelial function and insulin sensitivity with vitamin C in patients with coronary spastic angina: possible role of reactive oxygen species. J Am Coll Cardiol 2000; 35:1860-1866.
  343. Antoniades C, Tousoulis D, Tountas C et al. Vascular endothelium and inflammatory process, in patients with combined Type 2 diabetes mellitus and coronary atherosclerosis: the effects of vitamin C. Diabet Med 2004; 21:552-558.
  344. Anderson RA, Evans LM, Ellis GR et al. Prolonged deterioration of endothelial dysfunction in response to postprandial lipaemia is attenuated by vitamin C in Type 2 diabetes. Diabet Med 2006; 23:258-264.
  345. Chen H, Karne RJ, Hall G et al. High-dose oral vitamin C partially replenishes vitamin C levels in patients with Type 2 diabetes and low vitamin C levels but does not improve endothelial dysfunction or insulin resistance. Am J Physiol Heart Circ Physiol 2006; 290:H137-H145.
  346. Bonakdar RA, Guarneri E. Coenzyme Q10. Am Fam Physician 2005; 72:1065-1070.
  347. Singh RB, Niaz MA, Rastogi SS, Shukla PK, Thakur AS. Effect of hydrosoluble coenzyme Q10 on blood pressure and insullin resistance in hypertensive patients with coronary heart disease. J Hum Hypertens 1999; 13:203-208.
  348. Ghirlanda G, Oradei A, Manto A et al. Evidence of plasma CoQ10-lowering effect by HMG-CoA reductase inhibitors: a double-blind, placebo-controlled study. J Clin Pharmacol 1993; 33:226-229.
  349. Colquhoun DM, Jackson R, Walters M et al. Effects of simvastatin on blood lipids, vitamin E, coenzyme Q10 levels and left ventricular function in humans. Eur J Clin Invest 2005; 35:251-258.
  350. Pyorala K, Laakso M, Uusitupa M. Diabetes and atherosclerosis: an epidemiologic view. Diabetes Metab Rev 1987; 3:463-524.
  351. Paolisso G, Esposito R, D'Alessio MA, Barbieri M. Primary and secondary prevention of atherosclerosis: is there a role for antioxidants? Diabetes Metab 1999; 25(4):298-306.
  352. Devaraj S, Jialal I. Antioxidants and vitamins to reduce cardiovascular disease. Curr Atheroscler Rep 2000; 2:342-351.
  353. Jialal I, Traber M, Devaraj S. Is there a vitamin E paradox? Curr Opin Lipidol 2001; 12:49-53.
  354. Jialal I, Traber M, Devaraj S. Is there a vitamin E paradox? Curr Opin Lipidol 2001; 12:49-53.
  355. Yusuf S, Dagenais G, Pogue J, Bosch J, Sleight P. Vitamin E supplementation and cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med 2000; 342:154-160.
  356. Jialal I, Traber M, Devaraj S. Is there a vitamin E paradox? Curr Opin Lipidol 2001; 12:49-53.
  357. Devaraj S, Jialal I. Low-density lipoprotein postsecretory modification, monocyte function, and circulating adhesion molecules in type 2 diabetic patients with and without macrovascular complications: the effect of alpha-tocopherol supplementation. Circulation 2000; 102:191-196.
  358. Bursell SE, Clermont AC, Aiello LP et al. High-dose vitamin E supplementation normalizes retinal blood flow and creatinine clearance in patients with type 1 diabetes. Diabetes Care 1999; 22:1245-1251.
  359. Bursell SE, Clermont AC, Aiello LP et al. High-dose vitamin E supplementation normalizes retinal blood flow and creatinine clearance in patients with type 1 diabetes. Diabetes Care 1999; 22:1245-1251.
  360. Gaede P, Poulsen HE, Parving HH, Pedersen O. Double-blind, randomised study of the effect of combined treatment with vitamin C and E on albuminuria in Type 2 diabetic patients. Diabet Med 2001; 18:756-760.
  361. Manzella D, Barbieri M, Ragno E, Paolisso G. Chronic administration of pharmacologic doses of vitamin E improves the cardiac autonomic nervous system in patients with type 2 diabetes. Am J Clin Nutr 2001; 73:1052-1057.
  362. Caballero B. Vitamin E improves the action of insulin. Nutrit Rev 1993; 51:339-340.
  363. Paolisso G, D'Amore A, Galzerano D et al. Daily vitamin E supplements improve metabolic control but not insulin secretion in elderly type II diabetic patients. Diabetes Care 1993; 16:1433-1437.
  364. Ferrannini E, Mari A. How to measure insulin sensitivity. J Hypertension 1998; 6:895-906.
  365. Paolisso G, D'Amore A, Galzerano D et al. Daily vitamin E supplements improve metabolic control but not insulin secretion in elderly type II diabetic patients. Diabetes Care 1993; 16:1433-1437.
  366. Paolisso G, D'Amore A, Giugliano D, Ceriello A, Varricchio M, D'Onofrio F. Pharmacological doses of vitamin E improve insulin action in healthy subjects and non-insulin-dependent diabetic patients. Am J Clin Nutrit 1993; 57:650-656.
  367. Paolisso G, Di Maro G, Galzerano D et al. Pharmacological doses of vitamin E and insulin action in elderly subjects. Am J Clin Nutr 1994; 59:1291-1296.
  368. Barbagallo M, Dominguez LJ, Tagliamonte MR, Resnick LM, Paolisso G. Effects of vitamin E and glutathione on glucose metabolism: role of magnesium. Hypertension 1999; 34:1002-1006.
  369. Manzella D, Barbieri M, Ragno E, Paolisso G. Chronic administration of pharmacologic doses of vitamin E improves the cardiac autonomic nervous system in patients with type 2 diabetes. Am J Clin Nutr 2001; 73:1052-1057.
  370. Stanner SA, Hughes J, Kelly CN, Buttriss J. A review of the epidemiological evidence for the 'antioxidant hypothesis'. Public Health Nutr 2004; 7:407-422.
  371. Liu S, Lee IM, Song Y et al. Vitamin E and risk of type 2 diabetes in the women's health study randomized controlled trial. Diabetes 2006; 55:2856-2862.
  372. Lonn E, Bosch J, Yusuf S et al. Effects of long-term vitamin E supplementation on cardiovascular events and cancer: a randomized controlled trial. J Am Med Assoc 2005; 293:1338-1347.
  373. Boshtam M, Rafiei M, Golshadi ID, Ani M, Shirani Z, Rostamshirazi M. Long term effects of oral vitamin E supplement in type II diabetic patients. Int J Vitam Nutr Res 2005; 75:341-346.
  374. Sacco M, Pellegrini F, Roncaglioni MC, Avanzini F, Tognoni G, Nicolucci A. Primary prevention of cardiovascular events with low-dose aspirin and vitamin E in type 2 diabetic patients: results of the Primary Prevention Project (PPP) trial. Diabetes Care 2003; 26:3264-3272.
  375. Czernichow S, Couthouis A, Bertrais S et al. Antioxidant supplementation does not affect fasting plasma glucose in the Supplementation with Antioxidant Vitamins and Minerals (SU.VI.MAX) study in France: association with dietary intake and plasma concentrations. Am J Clin Nutr 2006; 84:395-399.
  376. Economides PA, Khaodhiar L, Caselli A et al. The effect of vitamin E on endothelial function of micro- and macrocirculation and left ventricular function in type 1 and type 2 diabetic patients. Diabetes 2005; 54:204-211.
  377. McSorley PT, Bell PM, Young IS et al. Endothelial function, insulin action and cardiovascular risk factors in young healthy adult offspring of parents with Type 2 diabetes: effect of vitamin E in a randomized double-blind, controlled clinical trial. Diabet Med 2005; 22:703-710.
  378. Lonn E, Bosch J, Yusuf S et al. Effects of long-term vitamin E supplementation on cardiovascular events and cancer: a randomized controlled trial. J Am Med Assoc 2005; 293:1338-1347.
  379. Economides PA, Khaodhiar L, Caselli A et al. The effect of vitamin E on endothelial function of micro- and macrocirculation and left ventricular function in type 1 and type 2 diabetic patients. Diabetes 2005; 54:204-211.
  380. Guyton JR. Effect of niacin on atherosclerotic cardiovascular disease. Am J Cardiol 1998; 82:18U-23U.
  381. Grundy SM, Vega GL, McGovern ME et al. Efficacy, safety, and tolerability of once-daily niacin for the treatment of dyslipidemia associated with type 2 diabetes: results of the assessment of diabetes control and evaluation of the efficacy of niaspan trial. Arch Intern Med 2002; 162:1568-1576.
  382. Singh RB, Niaz MA, Rastogi SS, Shukla PK, Thakur AS. Effect of hydrosoluble coenzyme Q10 on blood pressure and insullin resistance in hypertensive patients with coronary heart disease. J Hum Hypertens 1999; 13:203-208.
  383. Mccarty MF. Exploiting complementary therapeutic strategies for the treatment of type II diabetes and prevention of its complications. Med Hypotheses 1997; 49:143-152.
  384. Mertz W. Chromium research from a distance: from 1959 to 1980. J Am Coll Nutr 1998; 17:544-547.
  385. Morris BW, Kouta S, Robinson R, MacNeil S, Heller S. Chromium supplementation improves insulin resistance in patients with Type 2 diabetes mellitus. Diabet Med 2000; 17:684-685.
  386. Jeejeebhoy KN, Chu RC, Marliss EB, Greenberg GR, Bruce-Robertson A. Chromium deficiency, glucose intolerance, and neuropathy reversed by chromium supplementation, in a patient receiving long-term total parenteral nutrition. Am J Clin Nutr 1977; 30:531-538.
  387. Mooradian AD, Failla M, Hoogwerf B, Maryniuk M, Wylie-Rosett J. Selected vitamins and minerals in diabetes. Diabetes Care 1994; 17:464-479.
  388. Trumbo PR, Ellwood KC. Chromium picolinate intake and risk of type 2 diabetes: an evidence-based review by the United States Food and Drug Administration. Nutr Rev 2006; 64:357-363.
  389. Kleefstra N, Houweling ST, Jansman FG et al. Chromium treatment has no effect in patients with poorly controlled, insulin-treated type 2 diabetes in an obese Western population: a randomized, double-blind, placebo-controlled trial. Diabetes Care 2006; 29:521-525.
  390. Martin J, Wang ZQ, Zhang XH et al. Chromium picolinate supplementation attenuates body weight gain and increases insulin sensitivity in subjects with type 2 diabetes. Diabetes Care 2006; 29:1826-1832.
  391. Racek J, Trefil L, Rajdl D, Mudrova V, Hunter D, Senft V. Influence of chromium-enriched yeast on blood glucose and insulin variables, blood lipids, and markers of oxidative stress in subjects with type 2 diabetes mellitus. Biol Trace Elem Res 2006; 109:215-230.
  392. Anderson RA. Chromium in the prevention and control of diabetes. Diabetes Metab 2000; 26:22-27.
  393. Lukaski HC. Chromium as a supplement. Annu Rev Nutr 1999; 19:279-302.
  394. Lamson DS, Plaza SM. The safety and efficacy of high-dose chromium. Altern Med Rev 2002; 7:218-235.
  395. Althuis MD, Jordan NE, Ludington EA, Wittes JT. Glucose and insulin responses to dietary chromium supplements: a meta-analysis. Am J Clin Nutr 2002; 76:148-155.
  396. Anderson RA, Cheng N, Bryden NA, Polansky MM, Chi J, Feng J. Elevated intakes of supplemental chromium improve glucose and insulin variables in individuals with type 2 diabetes. Diabetes 1997; 46:1786-1791.
  397. Anderson RA, Cheng N, Bryden NA, Polansky MM, Chi J, Feng J. Elevated intakes of supplemental chromium improve glucose and insulin variables in individuals with type 2 diabetes. Diabetes 1997; 46:1786-1791.
  398. Uusitupa MI, Mykkanen L, Siitonen O et al. Chromium supplementation in impaired glucose tolerance of elderly: effects on blood glucose, plasma insulin, C-peptide and lipid levels. Br J Nutr 1992; 68:209-216.
  399. Grant KE, Chandler RM, Castle AL, Ivy JL. Chromium and exercise training: effect on obese women. Med Sci Sports Exerc 1997; 29:992-998.
  400. Anderson RA, Cheng N, Bryden NA, Polansky MM, Chi J, Feng J. Elevated intakes of supplemental chromium improve glucose and insulin variables in individuals with type 2 diabetes. Diabetes 1997; 46:1786-1791.
  401. Cefalu WT, Hu FB. Role of chromium in human health and in diabetes. Diabetes Care 2004; 27:2741-2751.
  402. Juturu V, Komorowski JR. Chromium supplements, glucose, and insulin responses. Am J Clin Nutr 2003; 78:190-193.
  403. Anderson RA, Polansky MM, Bryden NA. Stability and absorption of chromium and absorption of chromium histidinate complexes by humans. Biol Trace Elem Res 2004; 101:211-218.
  404. DiSilvestro RA, Dy E. Acute absorption comparison of commercially available chromium supplements and one new supplement. Biol Trace Elem Res. In press.
  405. Anderson RA. Chromium in the prevention and control of diabetes. Diabetes Metab 2000; 26:22-27.
  406. Cefalu WT, Hu FB. Role of chromium in human health and in diabetes. Diabetes Care 2004; 27:2741-2751.
  407. Kleefstra N, Houweling ST, Jansman FG et al. Chromium treatment has no effect in patients with poorly controlled, insulin-treated type 2 diabetes in an obese Western population: a randomized, double-blind, placebo-controlled trial. Diabetes Care 2006; 29:521-525.
  408. Wang ZQ, Zhang XH, Russell JC, Hulver M, Cefalu WT. Chromium picolinate enhances skeletal muscle cellular insulin signaling in vivo in obese, insulin-resistant JCR:LA-cp rats. J Nutr 2006; 136:415-420.
  409. Byon JCH, Kusari AB, Kusari J. Protein-tyrosine phosphatase-1B acts as a negative regulator of insulin signal transduction. Mol Cell Biochem 1998; 182:101-108.
  410. Evans JL, Jallal B. Protein tyrosine phosphatases: their role in insulin action and potential as drug targets. Exp Opin Invest Drugs 1999; 8:139-160.
  411. Elchebly M, Payette P, Michaliszyn E et al. Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene. Science 1999; 283:1544-1548.
  412. Klaman LD, Boss O, Peroni OD et al. Increased energy expenditure, decreased adiposity, and tissue-specific insulin sensitivity in protein-tyrosine phosphatase 1B-deficient mice. Mol Cell Biol 2000; 20:5479-5489.
  413. Goldstein BJ. Protein-tyrosine phosphatase 1B (PTP1B): a novel therapeutic target for type 2 diabetes mellitus, obesity, and related states of insulin resistance. Curr Drug Targets Immune Endocr Metabol Disord 2001; 1:265-275.
  414. Ramachandran C, Kennedy BP. Protein tyrosine phosphatase 1B: a novel target for type 2 diabetes and obesity. Curr Top Med Chem 2003; 3:749-757.
  415. Malamas MS, Sredy J, Gunawan I et al. New azolidinediones as inhibitors of protein tyrosine phosphatase 1B with antihyperglycemic properties. J Med Chem 2000; 43:995-1010.
  416. Cheon HG, Kim SM, Yang SD, Ha JD, Choi JK. Discovery of a novel protein tyrosine phosphatase-1B inhibitor, KR61639: potential development as an antihyperglycemic agent. Eur J Pharmacol 2004; 485:333-339.
  417. Liu G, Szczepankiewicz BG, Pei Z et al. Discovery and structure-activity relationship of oxalylarylaminobenzoic acids as inhibitors of protein tyrosine phosphatase 1B. J Med Chem 2003; 46:2093-2103.
  418. Wang H, Kruszewski A, Brautigan DL. Cellular chromium enhances activation of insulin receptor kinase. Biochemistry 2005; 44:8167-8175.
  419. Pender C, Youngren JF, Manchem VP et al. Regulation of insulin receptor function by a small molecule insulin receptor activator. J Biol Chem 2002
  420. Li M, Youngren JF, Dunaif A et al. Decreased insulin receptor (IR) autophosphorylation in fibroblasts from patients with PCOS: effects of sering kinase inhibitors and IR activators. J Clin Endocrinol Metab 2002; 87:4088-4093.
  421. Laires MJ, Monteiro CP, Bicho M. Role of cellular magnesium in health and human disease. Front Biosci 2004; 9:262-276.
  422. Anonymous. PDR for nutritional supplements. 1st ed ed. Montvale, NJ: Medical Economics Co, 2001.
  423. Laires MJ, Monteiro CP, Bicho M. Role of cellular magnesium in health and human disease. Front Biosci 2004; 9:262-276.
  424. Yokota K. Diabetes mellitus and magnesium. Clin Calcium 2005; 15:203-212.
  425. Takaya J, Higashino H, Kobayashi Y. Intracellular magnesium and insulin resistance. Magnes Res 2004; 17:126-136.
  426. Barbagallo M, Dominguez LJ, Galioto A et al. Role of magnesium in insulin action, diabetes and cardio-metabolic syndrome X. Mol Aspects Med 2003; 24:39-52.
  427. Tosiello L. Hypomagnesemia and diabetes mellitus: A review of clinical implications. Arch Intern Med 1996; 156:1143-1148.
  428. Chakraborti S, Chakraborti T, Mandal M, Mandal A, Das S, Ghosh S. Protective role of magnesium in cardiovascular diseases: a review. Mol Cell Biochem 2002; 238:163-179.
  429. Paolisso G, Barbagallo M. Hypertension, diabetes mellitus, and insulin resistance: the role of intracellular magnesium. Am J Hypertens 1997; 10:346-355.
  430. Fox C, Ramsoomair D, Carter C. Magnesium: its proven and potential clinical significance. South Med J 2001; 94:1195-1201.
  431. Paolisso G, Barbagallo M. Hypertension, diabetes mellitus, and insulin resistance: the role of intracellular magnesium. Am J Hypertens 1997; 10:346-355.
  432. Lopez-Ridaura R, Willett WC, Rimm EB et al. Magnesium intake and risk of type 2 diabetes in men and women. Diabetes Care 2004; 27:134-140.
  433. Murakami K, Okubo H, Sasaki S. Effect of dietary factors on incidence of type 2 diabetes: a systematic review of cohort studies. J Nutr Sci Vitaminol (Tokyo) 2005; 51:292-310.
  434. Song Y, Manson JE, Buring JE, Liu S. Dietary magnesium intake in relation to plasma insulin levels and risk of type 2 diabetes in women. Diabetes Care 2004; 27:59-65.
  435. Ma B, Lawson AB, Liese AD, Bell RA, Mayer-Davis EJ. Dairy, magnesium, and calcium intake in relation to insulin sensitivity: approaches to modeling a dose-dependent association. Am J Epidemiol 2006; 164:449-458.
  436. Nadler JL, Buchanan T, Natarajan R, Antonipillai I, Bergman RN, Rude R. Magnesium deficiency produces insulin resistance and increased thromboxane synthesis. Hypertension 1993; 21:1024-1029.
  437. de Valk HW. Magnesium in diabetes mellitus. Neth J Med 1999; 54:139-146.
  438. Huerta MG, Roemmich JN, Kington ML et al. Magnesium deficiency is associated with insulin resistance in obese children. Diabetes Care 2005; 28(5):1175-1181.
  439. Sjogren A, Floren CH, Nilsson A. Oral administration of magnesium hydroxide to subjects with insulin-dependent diabetes mellitus: effects on magnesium and potassium levels and on insulin requirements. Magnesium 1988; 7(3):117-122.
  440. Paolisso G, Sgambato S, Pizza G, Passariello N, Varricchio M, D'Onofrio F. Improved insulin response and action by chronic magnesium administration in aged NIDDM subjects. Diabetes Care 1989; 12:265-269.
  441. Paolisso G, Passariello N, Pizza G et al. Dietary magnesium supplements improve ¿ response to glucose and arginine in elderly non-insulin dependent diabetic subjects. Acta Endocrinol 1989; 121:16-20.
  442. Rodriguez-Moran M, Guerrero-Romero F. Oral magnesium supplementation improves insulin sensitivity and metabolic control in type 2 diabetic subjects: a randomized double-blind controlled trial. Diabetes Care 2003; 26:1147-1152.
  443. Yokota K, Kato M, Lister F et al. Clinical efficacy of magnesium supplementation in patients with type 2 diabetes. J Am Coll Nutr 2004; 23:506S-509S.
  444. Anonymous. PDR for nutritional supplements. 1st ed ed. Montvale, NJ: Medical Economics Co, 2001.
  445. Faure P, Roussel A, Coudray C, Richard MJ, Halimi S, Favier A. Zinc and insulin sensitivity. Biol Trace Elem Res 1992; 32:305-310.
  446. Arquilla ER, Packer S, Tarmas W, Miyamoto S. The effect of zinc on insulin metabolism. Endocrinology 1978; 103:1440-1449.
  447. Haase H, Maret W. Protein tyrosine phosphatases as targets of the combined insulinomimetic effects of zinc and oxidants. Biometals 2005; 18:333-338.
  448. Salgueiro MJ, Krebs N, Zubillaga MB et al. Zinc and diabetes mellitus: is there a need of zinc supplementation in diabetes mellitus patients? Biol Trace Elem Res 2001; 81:215-228.
  449. DiSilvestro RA. Zinc in relation to diabetes and oxidative disease. J Nutr 2000; 130 (5S Suppl):1509S-1511S.
  450. Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med 2002; 33:337-349.
  451. Salgueiro MJ, Krebs N, Zubillaga MB et al. Zinc and diabetes mellitus: is there a need of zinc supplementation in diabetes mellitus patients? Biol Trace Elem Res 2001; 81:215-228.
  452. DiSilvestro RA. Zinc in relation to diabetes and oxidative disease. J Nutr 2000; 130 (5S Suppl):1509S-1511S.
  453. DiSilvestro RA. Zinc in relation to diabetes and oxidative disease. J Nutr 2000; 130 (5S Suppl):1509S-1511S.
  454. Al-Maroof RA, Al-Sharbatti SS. Serum zinc levels in diabetic patients and effect of zinc supplementation on glycemic control of type 2 diabetics. Saudi Med J 2006; 27:344-350.
  455. Salgueiro MJ, Krebs N, Zubillaga MB et al. Zinc and diabetes mellitus: is there a need of zinc supplementation in diabetes mellitus patients? Biol Trace Elem Res 2001; 81:215-228.
  456. Al-Maroof RA, Al-Sharbatti SS. Serum zinc levels in diabetic patients and effect of zinc supplementation on glycemic control of type 2 diabetics. Saudi Med J 2006; 27:344-350.
  457. Al-Maroof RA, Al-Sharbatti SS. Serum zinc levels in diabetic patients and effect of zinc supplementation on glycemic control of type 2 diabetics. Saudi Med J 2006; 27:344-350.
  458. Farvid MS, Jalali M, Siassi F, Hosseini M. Comparison of the effects of vitamins and/or mineral supplementation on glomerular and tubular dysfunction in type 2 diabetes. Diabetes Care 2005; 28:2458-2464.
  459. Farvid MS, Jalali M, Siassi F, Hosseini M. Comparison of the effects of vitamins and/or mineral supplementation on glomerular and tubular dysfunction in type 2 diabetes. Diabetes Care 2005; 28:2458-2464.
  460. Farvid MS, Siassi F, Jalali M, Hosseini M, Saadat N. The impact of vitamin and/or mineral supplementation on lipid profiles in type 2 diabetes. Diabetes Res Clin Pract 2004; 65:21-28.
  461. Anonymous. Vanadium. In: Hendler SS, Rorvik D, Fleming T, Deutsch M, Wyble C, editors. PDR for nutritional supplements. Montvale, NJ: Medical Economics Co, 2001: 459-460.
  462. Evans JL, Jallal B. Protein tyrosine phosphatases: their role in insulin action and potential as drug targets. Exp Opin Invest Drugs 1999; 8:139-160.
  463. Mehdi MZ, Pandey SK, Theberge JF, Srivastava AK. Insulin signal mimicry as a mechanism for the insulin-like effects of vanadium. Cell Biochem Biophys 2006; 44:73-81.
  464. Srivastava AK, Mehdi MZ. Insulino-mimetic and anti-diabetic effects of vanadium compounds. Diabet Med 2005; 22:2-13.
  465. Posner BI, Faure R, Burgess JW et al. Peroxovanadium compounds. A new class of potent phosphotyrosine phosphatase inhibitors which are insulin mimetics. J Biol Chem 1994; 269:4596-4604.
  466. Elberg G, He ZB, Li JP, Sekar N, Shechter Y. Vanadate activates membranous nonreceptor protein tyrosine kinase in rat adipocytes. Diabetes 1997; 46:1684-1690.
  467. Posner BI, Faure R, Burgess JW et al. Peroxovanadium compounds. A new class of potent phosphotyrosine phosphatase inhibitors which are insulin mimetics. J Biol Chem 1994; 269:4596-4604.
  468. Tsiani E, Fantus IG. Vanadium compounds. Biological actions and potential as pharmacological agents. Trends Endocrin Metabol 1997; 8:51-58.
  469. Sekar N, Li JP, Shechter Y. Vanadium salts as insulin substitutes: Mechanisms of action, a scientific and therapeutic tool in diabetes mellitus research. Crit Rev Biochem Molec Biol 1996; 31:339-359.
  470. Heyliger CE, Tahiliani AG, McNeill JH. Effect of vanadate on elevated blood glucose and depressed cardiac performance of diabetic rats. Science 1985; 227:1474-1477.
  471. Yale JF, Lachance D, Bevan AP, Vigeant C, Shaver A, Posner BI. Hypoglycemic effects of peroxovanadium compoundsin Sprague-Dawley and diabetic BB rats. Diabetes 1995; 44:1274-1279.
  472. Yao J, Battell ML, McNeill JH. Acute and chronic response to vanadium following two methods of streptozotocin-diabetes induction. Can J Physiol Pharmacol 1997; 75:83-90.
  473. Meyerovitch J, Rothenberg P, Shechter Y, Bonner-Weir S, Kahn CR. Vanadate normalizes hyperglycemia in two mouse models of non-insulin-dependent diabetes mellitus. J Clin Invest 1991; 87:1286-1294.
  474. Verma S, Cam MC, McNeill JH. Nutritional factors that that favorably influence the glucose/insulin system: Vanadium. J Am Coll Nutr 1998; 17:11-18.
  475. Goldfine AB, Simonson DC, Folli F, Patti E, Kahn CR. In vivo and in vitro studies of vanadate in human and rodent diabetes mellitus. Mol Cell Biochem 1995; 153:217-231.
  476. Goldfine AB, Simonson DC, Folli F, Patti ME, Kahn CR. Metabolic effects of sodium metavanadate in humans with insulin-dependent and noninsulin-dependent diabetes mellitus in vivo and in vitro studies. J Clin Endocrinol Metab 1995; 80:3311-3320.
  477. Halberstam M, Cohen N, Shlimovich P, Rossetti L, Shamoon H. Oral vanadyl sulfate improves insulin sensitivity in NIDDM but not in obese nondiabetic subjects. Diabetes 1996; 45:659-666.
  478. Cohen N, Halberstam M, Shlimovich P, Chang CJ, Shamoon H, Rossetti L. Oral vanadyl sulfate improves hepatic and peripheral insulin sensitivity in patients with non-insulin-dependent diabetes mellitus. J Clin Invest 1995; 95:2501-2509.
  479. Boden G, Chen XH, Ruiz J, Vanrossum GDV, Turco S. Effects of vanadyl sulfate on carbohydrate and lipid metabolism in patients with non-insulin-dependent diabetes mellitus. Metabolism 1996; 45:1130-1135.
  480. Cusi K, Cukier S, DeFronzo RA, Torres M, Puchulu FM, Redondo JC. Vanadyl sulfate improves hepatic and muscle insulin sensitivity in type 2 diabetes. J Clin Endocrinol Metab 2001; 86:1410-1417.
  481. Cusi K, Cukier S, DeFronzo RA, Torres M, Puchulu FM, Redondo JC. Vanadyl sulfate improves hepatic and muscle insulin sensitivity in type 2 diabetes. J Clin Endocrinol Metab 2001; 86:1410-1417.
  482. Anonymous. Vanadium. In: Hendler SS, Rorvik D, Fleming T, Deutsch M, Wyble C, editors. PDR for nutritional supplements. Montvale, NJ: Medical Economics Co, 2001: 459-460.
  483. Verma S, Cam MC, McNeill JH. Nutritional factors that that favorably influence the glucose/insulin system: Vanadium. J Am Coll Nutr 1998; 17:11-18.
  484. Thompson KH, Orvig C. Vanadium compounds in the treatment of diabetes. Met Ions Biol Syst 2004; 41:221-252.
  485. Thompson KH, Orvig C. Vanadium compounds in the treatment of diabetes. Met Ions Biol Syst 2004; 41:221-252.
  486. Kofuji K, Qian CJ, Murata Y, Kawashima S. The controlled release of insulin-mimetic metal ions by the multifunction of chitosan. J Inorg Biochem 2005; 99:1329-1334.
  487. Verma S, Cam MC, McNeill JH. Nutritional factors that that favorably influence the glucose/insulin system: Vanadium. J Am Coll Nutr 1998; 17:11-18.
  488. Thompson KH, Orvig C. Vanadium compounds in the treatment of diabetes. Met Ions Biol Syst 2004; 41:221-252
  489. Badmaev V, Prakash S, Majeed M. Vanadium: a review of its potential role in the fight against diabetes. J Altern Complement Med 1999; 5:273-291.
  490. Hasenknopf B. Polyoxometalates: introduction to a class of inorganic compounds and their biomedical applications. Front Biosci 2005; 10:275-287.
  491. Sakurai H. Therapeutic potential of vanadium in treating diabetes mellitus. Clin Calcium 2005; 15:49-57.
  492. Domingo JL. Vanadium and tungsten derivatives as antidiabetic agents: a review of their toxic effects. Biol Trace Elem Res 2002; 88:97-112.
  493. Sethi S, Ziouzenkova O, Ni H, Wagner DD, Plutzky J, Mayadas TN. Oxidized omega-3 fatty acids in fish oil inhibit leukocyte-endothelial interactions through activation of PPAR alpha. Blood 2002; 100:1340-1346.
  494. Jump DB. The biochemistry of n-3 polyunsaturated fatty acids. J Biol Chem 2002; 277:8755-8758.
  495. Hu FB, Bronner L, Willett WC et al. Fish and omega-3 fatty acid intake and risk of coronary heart disease in women. J Am Med Assoc 2002; 287:1815-1821.
  496. Harper CR, Jacobson TA. The fats of life: the role of omega-3 fatty acids in the prevention of coronary heart disease. Arch Intern Med 2001; 161:2185-2192.
  497. Weber P, Raederstorff D. Triglyceride-lowering effect of omega-3 LC-polyunsaturated fatty acids--a review. Nutr Metab Cardiovasc Dis 2000; 10:28-37.
  498. Clarke SD. Polyunsaturated fatty acid regulation of gene transcription: a molecular mechanism to improve the metabolic syndrome. J Nutr 2001; 131:1129-1132.
  499. Evans JL, Lin JJ, Goldfine ID. Novel approach to treat insulin resistance, type 2 diabetes, and the metabolic syndrome: simultaneous activation of PPARa, PPARg, and PPARd. Current Diabetes Reviews 2005; 1:299-307.
  500. Jump DB. Dietary polyunsaturated fatty acids and regulation of gene transcription. Curr Opin Lipidol 2002; 13:155-164.
  501. Clarke SD. Polyunsaturated fatty acid regulation of gene transcription: a mechanism to improve energy balance and insulin resistance. Br J Nutr 2000; 83 Suppl 1:S59-S66.
  502. Xu HE, Lambert MH, Montana VG et al. Molecular recognition of fatty acids by peroxisome proliferator-activated receptors. Mol Cell 1999; 3:397-403.
  503. Hu FB, Bronner L, Willett WC et al. Fish and omega-3 fatty acid intake and risk of coronary heart disease in women. J Am Med Assoc 2002; 287:1815-1821.
  504. Harper CR, Jacobson TA. The fats of life: the role of omega-3 fatty acids in the prevention of coronary heart disease. Arch Intern Med 2001; 161:2185-2192.
  505. Weber P, Raederstorff D. Triglyceride-lowering effect of omega-3 LC-polyunsaturated fatty acids--a review. Nutr Metab Cardiovasc Dis 2000; 10:28-37.
  506. Burr ML, Fehily AM, Gilbert JF et al. Effects of changes in fat, fish, and fibre intakes on death and myocardial reinfarction: diet and reinfarction trial (DART). Lancet 1989; 2:757-761.
  507. de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation 1999; 99:779-785.
  508. Singh RB, Niaz MA, Sharma JP, Kumar R, Rastogi V, Moshiri M. Randomized, double-blind, placebo-controlled trial of fish oil and mustard oil in patients with suspected acute myocardial infarction: the Indian experiment of infarct survival--4. Cardiovasc Drugs Ther 1997; 11:485-491.
  509. GISSI-Prevenzione Investigators. Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: results of the GISSI-Prevenzione trial. Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto miocardico. Lancet 1999; 354:447-455.
  510. de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation 1999; 99:779-785.
  511. Singh RB, Niaz MA, Sharma JP, Kumar R, Rastogi V, Moshiri M. Randomized, double-blind, placebo-controlled trial of fish oil and mustard oil in patients with suspected acute myocardial infarction: the Indian experiment of infarct survival--4. Cardiovasc Drugs Ther 1997; 11:485-491.
  512. GISSI-Prevenzione Investigators. Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: results of the GISSI-Prevenzione trial. Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto miocardico. Lancet 1999; 354:447-455.
  513. Burr ML, Fehily AM, Gilbert JF et al. Effects of changes in fat, fish, and fibre intakes on death and myocardial reinfarction: diet and reinfarction trial (DART). Lancet 1989; 2:757-761.
  514. Kris-Etherton PM, Taylor DS, Yu-Poth S et al. Polyunsaturated fatty acids in the food chain in the United States. Am J Clin Nutr 2000; 71 (1 Suppl):179S-188S.
  515. Roche HM. Unsaturated fatty acids. Proc Nutr Soc 1999; 58:397-401.
  516. Cleland JG, Freemantle N, Coletta AP, Clark AL. Clinical trials update from the American Heart Association: REPAIR-AMI, ASTAMI, JELIS, MEGA, REVIVE-II, SURVIVE, and PROACTIVE. Eur J Heart Fail 2006; 8:105-110.
  517. Friday KE, Childs MT, Tsunehara CH, Fujimoto WY, Bierman EL, Ensinck JW. Elevated plasma glucose and lowered triglyceride levels from omega-3 fatty acid supplementation in type II diabetes. Diabetes Care 1989; 12:276-281.
  518. Vessby B, Boberg M. Dietary supplementation with n-3 fatty acids may impair glucose homeostasis in patients with non-insulin-dependent diabetes mellitus. J Intern Med 1990; 228:165-171.
  519. Mcmanus RM, Jumpson J, Finegood DT, Clandinin MT, Ryan EA. A comparison of the effects of n-3 fatty acids from linseed oil and fish oil in well-controlled type II diabetes. Diabetes Care 1996; 19:463-467.
  520. Luo J, Rizkalla SW, Vidal H et al. Moderate intake of n-3 fatty acids for 2 months has no detrimental effect on glucose metabolism and could ameliorate the lipid profile in type 2 diabetic men: Results of a controlled study. Diabetes Care 1998; 21:717-724.
  521. Prince MJ, Deeg MA. Do n-3 fatty acids improve glucose tolerance and lipemia in diabetics? Curr Opin Lipidol 1997; 8:7-11.
  522. Montori VM, Farmer A, Wollan PC, Dinneen SF. Fish oil supplementation in type 2 diabetes: a quantitative systematic review. Diabetes Care 2000; 23:1407-1415.
  523. Montori VM, Farmer A, Wollan PC, Dinneen SF. Fish oil supplementation in type 2 diabetes: a quantitative systematic review. Diabetes Care 2000; 23:1407-1415.
  524. Anonymous. PDR for nutritional supplements. 1st ed ed. Montvale, NJ: Medical Economics Co, 2001.
  525. Carpentier YA, Portois L, Malaisse WJ. n-3 fatty acids and the metabolic syndrome. Am J Clin Nutr 2006; 83 (6 Suppl):1499S-1504S.
  526. Mostad IL, Bjerve KS, Bjorgaas MR, Lydersen S, Grill V. Effects of n-3 fatty acids in subjects with type 2 diabetes: reduction of insulin sensitivity and time-dependent alteration from carbohydrate to fat oxidation. Am J Clin Nutr 2006; 84:540-550.
  527. Pariza MW, Park Y, Cook ME. The biologically active isomers of conjugated linoleic acid. Prog Lipid Res 2001; 40:283-298.
  528. Ha YL, Grimm NK, Pariza MW. Anticarcinogens from fried ground beef: heat-altered derivatives of linoleic acid. Carcinogenesis 1987; 8:1881-1887.
  529. Ip C, Chin SF, Scimeca JA, Pariza MW. Mammary cancer prevention by conjugated dienoic derivative of linoleic acid. Cancer Res 1991; 51:6118-6124.
  530. Belury MA. Dietary conjugated linoleic acid in health: physiological effects and mechanisms of action. Annu Rev Nutr 2002; 22:505-531.
  531. MacDonald HB. Conjugated linoleic acid and disease prevention: a review of current knowledge. J Am Coll Nutr 2000; 19 (2 Suppl):111S-118S.
  532. Pariza MW, Park Y, Cook ME. The biologically active isomers of conjugated linoleic acid. Prog Lipid Res 2001; 40:283-298.
  533. MacDonald HB. Conjugated linoleic acid and disease prevention: a review of current knowledge. J Am Coll Nutr 2000; 19 (2 Suppl):111S-118S.
  534. MacDonald HB. Conjugated linoleic acid and disease prevention: a review of current knowledge. J Am Coll Nutr 2000; 19 (2 Suppl):111S-118S.
  535. Moya-Camarena SY, Vanden Heuvel JP, Blanchard SG, Leesnitzer LA, Belury MA. Conjugated linoleic acid is a potent naturally occurring ligand and activator of PPARa. J Lipid Res 1999; 40:1426-1433.
  536. Belury MA. Dietary conjugated linoleic acid in health: physiological effects and mechanisms of action. Annu Rev Nutr 2002; 22:505-531.
  537. DeLany JP, West DB. Changes in body composition with conjugated linoleic acid. J Am Coll Nutr 2000; 19:487S-493S.
  538. Ryder JW, Portocarrero CP, Song XM et al. Isomer-specific antidiabetic properties of conjugated linoleic acid. Improved glucose tolerance, skeletal muscle insulin action, and UCP-2 gene expression. Diabetes 2001; 50:1149-1157.
  539. Belury MA, Mahon A, Banni S. The conjugated linoleic acid (CLA) isomer, t10c12-CLA, is inversely associated with changes in body weight and serum leptin in subjects with type 2 diabetes mellitus. J Nutr 2003; 133(1):257S-260S.
  540. Salas-Salvado J, Marquez-Sandoval F, Bullo M. Conjugated linoleic acid intake in humans: a systematic review focusing on its effect on body composition, glucose, and lipid metabolism. Crit Rev Food Sci Nutr 2006; 46:479-488.
  541. Brown JM, McIntosh MK. Conjugated linoleic acid in humans: regulation of adiposity and insulin sensitivity. J Nutr 2003; 133:3041-3046
  542. Bhattacharya A, Banu J, Rahman M, Causey J, Fernandes G. Biological effects of conjugated linoleic acids in health and disease. J Nutr Biochem 2006.
  543. Wang YW, Jones PJ. Conjugated linoleic acid and obesity control: efficacy and mechanisms. Int J Obes Relat Metab Disord 2004; 28:941-955.
  544. Smedman A, Vessby B. Conjugated linoleic acid supplementation in humans--metabolic effects. Lipids 2001; 36:773-781.
  545. Thom E, Wadstein J, Gudmundsen O. Conjugated linoleic acid reduces body fat in healthy exercising humans. J Int Med Res 2001; 29:392-396.
  546. Blankson H, Stakkestad JA, Fagertun H, Thom E, Wadstein J, Gudmundsen O. Conjugated linoleic acid reduces body fat mass in overweight and obese humans. J Nutr 2000; 130:2943-2948.
  547. Riserus U, Arner P, Brismar K, Vessby B. Treatment with dietary trans10cis12 conjugated linoleic acid causes isomer-specific insulin resistance in obese men with the metabolic syndrome. Diabetes Care 2002; 25:1516-1521.
  548. Riserus U, Basu S, Jovinge S, Fredrikson GN, Arnlov J, Vessby B. Supplementation with conjugated linoleic acid causes isomer-dependent oxidative stress and elevated C-reactive protein: a potential link to fatty acid-induced insulin resistance. Circulation 2002; 106:1925-1929.
  549. Moloney F, Yeow TP, Mullen A, Nolan JJ, Roche HM. Conjugated linoleic acid supplementation, insulin sensitivity, and lipoprotein metabolism in patients with type 2 diabetes mellitus. Am J Clin Nutr 2004; 80:887-895.
  550. Riserus U, Vessby B, Arner P, Zethelius B. Supplementation with trans10cis12-conjugated linoleic acid induces hyperproinsulinaemia in obese men: close association with impaired insulin sensitivity. Diabetologia 2004; 47:1016-1019.
  551. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  552. Yeh GY, Eisenberg DM, Kaptchuk TJ, Phillips RS. Systematic review of herbs and dietary supplements for glycemic control in diabetes. Diabetes Care 2003; 26:1277-1294.
  553. Anonymous. PDR for Herbal Medicines. 1st ed ed. Montvale, NJ: Medical Economics Co, 1998.
  554. Abebe W. Herbal medication: potential for adverse interactions with analgesic drugs. J Clin Pharm Ther 2002; 27:391-401.
  555. Canter PH, Ernst E. Ginkgo biloba: a smart drug? A systematic review of controlled trials of the cognitive effects of ginkgo biloba extracts in healthy people. Psychopharmacol Bull 2002; 36:108-123.
  556. Birks J, Grimley EV, Van Dongen M. Ginkgo biloba for cognitive impairment and dementia. Cochrane Database Syst Rev 2002;CD003120.
  557. Kudolo GB. The effect of 3-month ingestion of Ginkgo biloba extract (EGb 761) on pancreatic beta-cell function in response to glucose loading in individuals with non-insulin-dependent diabetes mellitus. J Clin Pharmacol 2001; 41:600-611.