Next Article in Journal
Environmental Microbial Community Proteomics: Status, Challenges and Perspectives
Next Article in Special Issue
Protective Effects of Berberine on Renal Injury in Streptozotocin (STZ)-Induced Diabetic Mice
Previous Article in Journal
The Ribonuclease A Superfamily in Humans: Canonical RNases as the Buttress of Innate Immunity
Previous Article in Special Issue
Association of Serum Uric Acid Concentration with Diabetic Retinopathy and Albuminuria in Taiwanese Patients with Type 2 Diabetes Mellitus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Coadjuvants in the Diabetic Complications: Nutraceuticals and Drugs with Pleiotropic Effects

by
Thiago Melo Costa Pereira
1,2,
Fabio Silva Pimenta
1,3,
Marcella Lima Porto
2,
Marcelo Perim Baldo
4,
Bianca Prandi Campagnaro
1,
Agata Lages Gava
5,6,
Silvana Santos Meyrelles
5 and
Elisardo Corral Vasquez
1,5,*
1
Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Av. Comissario Jose Dantas Melo 21, Boa Vista, 29102-920 Vila Velha, Brazil
2
Federal Institute of Education, Science and Technology (IFES), 29106-010 Vila Velha, Brazil
3
Burn Treatment Center, Children State Hospital, 29056-030 Vitoria, Brazil
4
Department of Pathophysiology, Montes Claros State University, 39401-089, Montes Claros, Brazil
5
Laboratory of Translational Physiology, Federal University of Espirito Santo (Ufes), 29047-100 Vitoria, Brazil
6
Division of Nephrology, McMaster University, Hamilton, ON L8N 4A6, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2016, 17(8), 1273; https://doi.org/10.3390/ijms17081273
Submission received: 5 July 2016 / Revised: 27 July 2016 / Accepted: 29 July 2016 / Published: 5 August 2016
(This article belongs to the Special Issue Diabetic Complications: Pathophysiology, Mechanisms, and Therapies)

Abstract

:
Because diabetes mellitus (DM) is a multifactorial metabolic disease, its prevention and treatment has been a constant challenge for basic and clinical investigators focused on translating their discoveries into clinical treatment of this complex disorder. In this review, we highlight recent experimental and clinical evidences of potential coadjuvants in the management of DM, such as polyphenols (quercetin, resveratrol and silymarin), cultured probiotic microorganisms and drugs acting through direct/indirect or pleiotropic effects on glycemic control in DM. Among several options, we highlight new promising therapeutic coadjuvants, including chemical scavengers, the probiotic kefir and the phosphodiesterase 5 inhibitors, which besides the reduction of hyperglycemia and ameliorate insulin resistance, they reduce oxidative stress and improve endothelial dysfunction in the systemic vascular circulation. In the near future, experimental studies are expected to clear the intracellular pathways involving coadjuvants. The design of clinical trials may also contribute to new strategies with coadjuvants against the harmful effects of diabetic complications.

Graphical Abstract

1. Introduction

Diabetes mellitus (DM) is an important public health issue because it is highly associated with increased morbidity and mortality [1]. In fact, the prevalence of diagnosed diabetes is increasing worldwide, as demonstrated by the rise from 6.5% (1999 to 2002) to 7.8% (2003 to 2006) of the population in just a few years [2]. Type 2 DM is the most common form of the disease and affects 90% to 95% of individuals with diabetes. The main issue of this pandemic is the increase in mortality associated with diabetes due to the risk of cardiovascular diseases (CVD), which are the leading cause of death in this population. This information became clear after an analysis of the First National Health and Nutrition Examination Survey (NHAHES), which covered the period of 1971–1993 and revealed that more than 65% of deaths of people with diabetes were due to CVD [3]. In addition, diabetes is a leading cause of morbidity and leads to microvascular and macrovascular complications [3,4,5].
Even with the already reported increase in the prevalence of diabetes over the years, only approximately 13% of individuals diagnosed as diabetic were in conformity with the control of the established levels of serum glucose, blood arterial pressure and total cholesterol at the same time [6]. Indeed, it is well known that most of the type 2 DM fail to control glycemia to normal levels when subjected only to diet and physical exercise and, consequently, it is necessary to treat them with anti-diabetic pharmacotherapy [1]. For instance, in a period of two years, among those patients who have been diagnosed as diabetics, the percentages of success in the control of glycated hemoglobin, blood pressure and total cholesterol are higher than 7%, 35% and 37%, respectively. As a result, good management of type 2 diabetes with pharmacological as well as non-pharmacological therapy (including reduction of caloric intake and intermittent fasting) is important [7,8]. Lately, with the mission of ameliorating this health problem, eight different classes of drugs for treatment of type 2 DM, with variations in their side effects and costs, have been approved by the US Food and Drug Administration (FDA) [9]. In this regard, investigators have been challenged to test potential therapies for DM based on functional foods, which are of low cost and very accessible (e.g., substances derived from marine algae [10]).
In the present review, we discuss some epidemiological aspects of diabetic complications resulting from hyperglycemia and the therapeutic advances with antioxidant substances based on experimental and clinical studies. Among different alternatives discussed in this review, we highlight the putative coadjuvants in the management of DM, such as functional foods rich in polyphenols and the probiotic kefir. In addition, we discuss drugs with pleiotropic effects, such as phosphodiesterase 5 (PDE5) inhibitors, which lately have been the main focus of investigation in our laboratory.

2. The Impact of Chronic Hyperglycemia on Diabetic Complication

2.1. Epidemiological Aspects of Diabetes

The high mortality and morbidity observed in DM patients characterized by chronic high levels of blood glucose and HbA1c, which compromises the function of the target organs heart and kidneys [11,12]. Therefore, the desired goal of treatment for diabetes is to maintain euglycemic levels as much as possible. Studies from early last century have also highlighted several effects of uncontrolled diabetes, such as dyslipidemia [13], reduced serum protein [14], skeletal muscle changes [15], and other complications. In the last decade, several epidemiological studies have been conducted to identify the risks associated to diabetes.
It is well known that diabetes doubles the risk for acute coronary syndrome with an additional risk once the event has occurred. This risk was evident in the Tehran Lipid and Glucose Study, a population-based cohort study that took place in Iran. The authors found that in type 2 diabetic patients, hypercholesterolemia and central adiposity were independent risk factors for death by cardiovascular causes, and poor glycemic control is an independent risk factor for both cardiovascular and all-cause mortality [16]. In the United Kingdom, a cohort study of myocardial infarction risk in men and women with and without diabetes was carried out using a large, nationwide primary care database. The overall adjusted relative risk of myocardial infarction was higher in individuals with diabetes versus no diabetes and was greater in women compared to men [17].
After a 23-year follow-up to determine the prevalence of diabetes and associated characteristics, the Da Qing IGT and Diabetes Study showed that CVD was the leading cause of death in individuals with diabetes (47.5% in men and 49.7% in women), and almost half of the deaths were due to stroke [18]. This excessive risk of stroke associated with diabetes was significantly higher in women than men, and there were no sex differences for other major cardiovascular risk factors [19]. It is noteworthy that high glucose levels alone did not account for the increased risk associated with diabetes. A meta-analysis of 15 prospective studies in approximately 760,000 patients showed that people with pre-diabetes, which was defined as impaired fasting glucose of 110 to 124 mg/dL (6.1 to 6.9 mmol/L) or both, exhibited a moderate higher risk of stroke events [20].
Another condition associated with diabetes is chronic kidney disease. In fact, diabetes is one of the leading causes of chronic kidney disease in the United States [21], where the prevalence of diabetic nephropathy in the population of patients with type 1 and 2 DM is 20% to 40% [22,23]. The Madrid Diabetes Study, which is a prospective cohort study of 3443 type-2 diabetic outpatients, showed that the unadjusted hazard ratio for all-cause mortality in diabetic patients with eGFR < 60 mL/min/1.73 m2 was approximately 3 after five years of follow-up. Patients with chronic kidney disease at baseline had an increased risk of cardiovascular mortality [24].

2.2. Toxic Effects of Hyperglycemia

Chronic hyperglycemia can promote toxic effects in a myriad of tissues, especially in neurons, because they are more susceptible to glucose uptake [25].
Uncontrolled diabetes cam result in a pathological state characterized by severe hyperglycemia, elevation of plasma osmolarity and diabetic ketoacidosis [26]. Its classic manifestation consists of the biochemical triad of hyperglycemia, increased ketones in bloodstream, and metabolic acidosis, and it might be caused by several factors, including reduced secretion and action of insulin, and raised levels of anti-insulin hormones [27,28].
In general, patients with chronic hyperglycemia exhibit many other characteristics, such as altered expression of matrix degrading enzymes, increased synthesis and deposition of extracellular matrix (ECM), generation of advanced glycation end products (AGE), upregulation of pro-inflammatory cytokines and growth factors, and augmented flux of hexosamines and polyols [29]. Moreover, in chronic hyperglycemia conditions, the augmented glycation of intracellular proteins appears to attack other proteins and worsen the exacerbate formation of AGEs [30,31]. Consequently, it leads to the inhibition of mitochondrial respiration, increased production of reactive oxygen species (ROS) and inflammatory cytokines, culminating with marked alterations in the systemic vascular function. Also, it is well known that the augmented production of ROS causes DNA damage and results in alteration in the expression of ECM glycoproteins, which corroborates the concept that augmented oxidative stress accounts for DM complications [32,33].
In chronic hyperglycemia, aldose reductase is activated and catalyzes the first reaction in the polyol pathway, resulting in exacerbated productions and accumulation of sorbitol [34,35,36] and causing cellular toxicity by osmotic effects. NADPH is consumed and NADH is produced with accumulation of sorbitol and fructose that can also affect cellular osmosis. While there is an oversupply of NADH in individuals with diabetes due to chronic hyperglycemia and enhanced fatty acid oxidation, NAD+ could be depleted due to the activation of poly ADP ribose polymerase (PARP) by oxidative DNA damage during oxidative stress [37,38].
The activation of apoptosis in chronic hyperglycemia has received much attention in recent years. Several mechanisms regulate the complicated signaling pathways that mediate apoptosis by hyperglycemia. This process is initiated by interruption of mitochondrial electron transport, resulting in an incomplete reduction of molecular oxygen, generating superoxide anion (· O 2 ) . This free radical can react with nitric oxide (NO), resulting in the production of peroxynitrite (ONOO), which is a highly toxic molecule [39,40] that causes endothelial cell death. Dysfunction of endothelial cells, which causes loss of multiple endothelium-derived substances, has been hypothesized to play a key role in the progression of vascular disease in diabetes [41,42].

2.3. Role of Oxidative Stress in Diabetic Complications

Oxidative stress is induced by elevations in glucose and free fatty acid levels and has a key role in the pathogenesis of both types of DM and on diabetic complications, as has been reviewed by Wei et al. [43]. Recent evidence suggests oxidative stress is a key participant in the development and progression of diabetes as well as its micro- and macrovascular complications [44,45,46]. Paradoxically, not much attention has been given to other possible therapeutic interventions besides glucose reduction.
ROS are a group of short-lived molecules derived from aerobic respiration and other oxygen reactions that include · O 2 , hydrogen peroxide (H2O2), hydroxyl radical (·OH), ONOO and hydroxyl (OH) [47,48]. The major sources of ROS are the mitochondria, NADPH oxidases, xanthine oxidase, uncoupled NO synthase (NOS), lipoxygenase, cyclooxygenases and CYP450, but they vary in their pathological role and importance depending on the disease and the organ [40,49]. Mitochondria and NADPH oxidases (Nox) are the most important sites for ROS production and are responsible for cardiovascular complications in diabetes [50]. In 1999, Ide et al. [51] observed enhanced cardiomyocyte mitochondrial · O 2 in the failing myocardium. Moreover, Selemidis et al. [52] suggested that NADPH is a primary ROS-producer not only in vascular smooth muscle cells but also in cardiomyocytes, vascular endothelial cells and adventitial fibroblasts. Furthermore, increased expression of Nox isoforms has been associated with myocardial hypertrophy and fibrosis in diabetes [52,53].
Hyperglycemia is characterized not only by a high-level production of ROS but also by an impairment of the intracellular antioxidant defense system, such as the nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a master upregulator of several antioxidant enzymes [54,55]; consequently, the induction of genes encoding antioxidant molecules, including superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase is also affected [56]. Additionally, reduced SOD, catalase and GPx activity have been reported in both experimental and clinical diabetic conditions due to excessive glycation [57,58]. Batinic-Haberle et al. [59] found that diabetic blood vessels exhibited an improved endothelium-dependent relaxant response when treated with SOD. Interestingly, a recent study showed that the antioxidant curcumin may have a protective role against oxidative stress in diabetic mice (db/db) [60]. Therefore, it is important to emphasize that the nutraceutical compounds that require the activation of Nrf2 have been considered as relevant therapeutic strategy for prevention/treatment of diabetic complications [55,56].
Regardless of the imbalance between the generation of ROS and the activity/intracellular levels of the antioxidant defense mechanisms, excessive generation of ROS is a deleterious factor that leads to pathological consequences, including irreversible cellular damage by oxidation of proteins, lipids, carbohydrates and nucleic acids [61]. Recent evidence indicated that increased levels of urinary markers of oxidative DNA and RNA damage occur with diabetic complications [62]. Furthermore, Palem and Abraham [63] observed that diabetic patients taking both oral antidiabetic drugs and insulin still present high levels of oxidative stress, which emphasizes the need for adding antioxidants to reduce the impact of diabetic complications.
In addition to the direct damage to cells, increased ROS levels also cyclically activates pathways associated with diabetes complications, such as the polyol pathway, increased production of AGEs, activation of PKC isoforms and the hexosamine pathway [54,64]. ROS overproduction and increased oxidative stress can also cause vascular endothelial and smooth muscle dysfunction. On the other hand, it has been shown that neutralization of reactive molecules in patients with diabetes was capable of preventing cardiomyopathy, retinopathy, nephropathy and neuropathy [65]. To avoid diabetes disorders, hyperglycemia should be treated promptly through stimulation of insulin secretion (not the best choice) or increasing insulin sensitivity. However, adopting a causal antioxidant therapeutic approach might be a modern adjuvant strategy to prevent the overproduction of ROS and consequently complications from diabetes.

3. Potential of Natural Products with Antioxidant Effects for Treating Diabetes

Polyphenolic compounds are widely found in plants and provide several pharmacological properties, including antidiabetic effects [66,67,68]. Although not focused in the present discussion, it is important to recognize that the Chinese medicine has demonstrated the efficacy of several natural products that have been used in the treatment of DM as reviewed elsewhere [69]. In this subsection, the main polyphenols with potential antidiabetic activity investigated by us as well as others will be addressed.

3.1. Polyphenolic Compounds

3.1.1. Quercetin

Quercetin (2-(3,4-dihydroxyphenyl)-3,5,7-trihydroxychromen-4-one) is the major flavonoid involved in vegetables and fruits, and it exhibits metabolic, anti-oxidative, anti-apoptotic and renoprotective effects at adequate doses [45,46,68,70]. Although this molecule is widely consumed in the diet, it was surprisingly reported as mutagenic in the 1970s in a study with unusual methods and with no reproducible results [70]. Only in 1999 did the International Agency for Research on Cancer conclude that quercetin should not be classified as carcinogenic to humans [70]. However, in parallel, the investigations with quercetin related to diabetes began in 1975 with an initial interest in preventing cataracts through the inhibition of the aldose reductase that blocks polyol accumulation in intact lenses [71]. Only in the 1990s did quercetin studies extend to other targets in diabetes complications.
Many studies have demonstrated that this bioflavonoid may act through diverse pathways to decrease the tissue-damaging effects of chronic hyperglycemia, such as stimulation of glucose uptake via GLUT4 [72,73,74], inhibiting hepatic glycogenolysis and gluconeogenesis [72,75], and inhibiting α-glucosidase in the small intestine [76] or intestinal glucose transporter GLUT2 [77]. At the same time, another potential advantage is that quercetin exhibits all the characteristics of an adequate antioxidant for diabetes treatment: free radical scavenger ability [78,79], long half-life (~20 h in humans) [80,81], capacity to suppress pro-oxidant enzymes (NADPH oxidase, xanthine oxidase and CYP) [82,83,84] and the ability to stimulate antioxidant enzymes (SOD, catalase, glutathione peroxidase and glutathione reductase) [68,85,86] with high mitochondrial permeability [46,87], which are an important source of ROS in diabetes [88,89]. Given these multiple potential mechanisms, quercetin becomes an important protective molecule against the consequences of long-term diabetes (e.g., microvascular and macrovascular damage, nephropathy, neuropathy associated with the risks autonomic disturbance, amputations and foot ulcers) [67,90,91], as demonstrated in the experimental and clinical investigations as discussed below.
In streptozotocin (STZ)-induced type 1 diabetes models, varying doses of quercetin have shown several benefits. At 50 mg/kg/day (oral dose), quercetin prevented retinal degeneration [92] and vascular complications by inhibiting NF-κB signaling [93]. In rats, quercetin ameliorated erectile dysfunction by inhibiting oxidative stress and upregulating eNOS [94], and it protected against the progression of neuropathy even with a low dose of quercetin (10 mg/kg) as well as attenuating cold allodynia and hyperalgesia [95]. Recently, for the first time, we demonstrated that the same low dose of quercetin attenuates hyperglycemia and nephropathy in STZ-induced diabetes in apolipoprotein E-deficient mice [46] and in C57BL/6J mice [45] (or in rats in a study conducted by others [96,97]), and quercetin treatment exhibited antioxidant benefits. With different doses (25 to 100 mg/kg/day), quercetin was also capable of suppressing the kidney inflammatory response at least partly via anti-hyperuricemic and anti-dyslipidemic effects [98]. In db/db mice (the most popular mouse model for type 2 DM), quercetin also demonstrated satisfactory effects [76]. At doses ranging between 50 and 100 mg/kg/day, quercetin treatment improved postprandial blood glucose (similarly to acarbose) [76] in addition to avoiding hyperglycemia and hyperlipidemia and increasing the antioxidant status [99]. Although experimental studies clearly support the protective effects of quercetin in diabetes, clinical data with this isolated compound are still insufficient and inconclusive. Recently, 500 mg of daily quercetin (for four weeks) was capable of reducing hyperuricemia in healthy men [100], which is a relevant factor associated with insulin resistance and progression of diabetic complications [91]. On the other hand, quercetin administered at the same dosage in women with type 2 DM, has been shown to decrease systolic arterial pressure, without significant effects on other cardiovascular risk factors [101]. Similarly, recent data from Brüll et al. [102] revealed that quercetin (162 mg/day) decreased day- and nighttime systolic blood pressure in overweight-to-obese patients without changing any other metabolic risk factor. More recently, another study reported no effect on flow-mediated dilation or insulin resistance with an analogue of quercetin (quercetin-3-glucoside, at 160 mg/day) in healthy men and women aged 40–80 years [103]. Therefore, more studies about quercetin will be necessary to establish the ideal dosage and to identify the real efficacy in diabetic patients.

3.1.2. Resveratrol

This non-flavonoid polyphenolic compound (3,5,4′-trihydroxystilbene, notably present in peanuts, grapes, grape juice and red wine) might be the main molecule responsible for cardiovascular protective effects in the French population despite a high intake of saturated fats, which is known as “French Paradox” [66,104,105,106]. For that reason, this potent molecule (even with a short half-life) also would be highly beneficial as an adjuvant therapy for diabetes. Additionally, under in vitro [107,108] and in vivo [109,110,111] experimental conditions that mimic human diabetes, resveratrol has been shown to have a potential benefit in several multi-target mechanisms for diabetic complications, as presented below.
Recently, Yan et al. [112] showed that 40 mg/kg/day of oral resveratrol (a high dose—according to Zhou et al. [112]) reduced proteinuria and attenuated the progress of renal fibrosis in db/db mice [112,113]. At the other extreme, it was demonstrated that a low dose of oral resveratrol (0.5 mg/kg) ameliorated classical DM symptoms (e.g., polydipsia, polyphagia, and body weight loss) and delayed the onset of insulin resistance in an STZ model [66], which probably occurred through improved glucose homeostasis. This evidence was supported by Palsamy et al., who in 2009 [114] showed decreased activity of key enzymes for gluconeogenesis by treating rats with mild diabetes (STZ-nicotinamide model) with a low dose (5 mg/kg) of resveratrol. Moreover, several in vitro studies have shown that resveratrol can increase glucose uptake by targeting insulin-affected cells (skeletal muscle, adipocytes and hepatocytes) [115,116,117,118,119], thereby improving the insulin signaling probably through improvement of insulin sensitivity in a SIRT1-dependent manner [120,121,122] or by other distinct mechanisms [66]. This stimulation of SIRT1 (a pivotal mediator of the metabolic effects of resveratrol) also may promote an increase of antioxidant enzymes (SOD, catalase, GPx and glutathione-S-transferase) in pancreatic β-cells and decrease the function of pro-inflammatory mediators (IL-6, NF-kB and COX-2) in many diabetic target tissues [119,123,124,125], which explains the relevant protective effects against apoptosis, neurodegeneration and cardiovascular complications [106,126,127]. Interestingly, resveratrol also seems to contribute to endothelial repair (which is an important tissue affected by chronic diabetes) through free-radical scavenging and/or restoration of eNOS functionality that culminates with increased bioavailability of NO [106,109,127,128] and consequently reduces diabetic complications. Recently, Neves et al. [129] showed another possible pathway of cellular protection through the regulation of cell membrane structure and fluidity (similar to cholesterol). In addition, it was observed that resveratrol might reduce endoplasmic reticulum stress by avoiding misglycosylation, depletion of calcium stores and DNA damage [127]. Therefore, resveratrol not only acts by glycemic control per se but also provides antioxidant and other pleiotropic effects [125,129,130].
Even though the preclinical evidence includes experimental evidence that clearly demonstrated that resveratrol has a significant antidiabetic effect in a wide dose range (0.1 to 1.500 mg/kg body weight), recommending resveratrol as a therapeutic supplement or treatment for diabetes patients is still controversial, and there is a similar controversy for quercetin [119,127,131,132]. This is a problem because it has generated serious doubts about the potential usefulness of these substances, particularly for dietary prevention strategies [133,134,135,136]. For instance, Thazhath et al. [137] have recently demonstrated that 1000 mg/day resveratrol in diet-controlled type-2 DM patients for five weeks did not change body weight, glycemic control or GLP-1 secretion. Similar data were also obtained by Poulsen et al. [132], who gave 1500 mg/day of resveratrol for four weeks to obese patients and found no effects on metabolic biomarkers, blood pressure or resting energy expenditure. These apparent unsuccessful studies also may be explained by variability between volunteers (age, body weight, nutrition, severity of diabetes) and/or duration of treatments [119]. In agreement with this hypothesis, another study of patients with metabolic syndrome treated the patients with 1500 mg/day of resveratrol for 90 days (~13 weeks) and revealed a significant reduction in body weight and insulin secretion [137]. Additionally, Goh et al. [138] showed improvement of insulin sensitivity via SIRT1 through 3000 mg of resveratrol for 12 weeks in type-2 diabetic patients. The advantage of this regulation is the promotion of survival and longevity, associated with telomere length [7]. Even for a shorter period of time, it was shown that resveratrol (1000 mg daily in first week followed by 2000 mg daily in second week) was able to reduce hepatic and intestinal lipoprotein production [139]. It is important to consider that although there are several investigations on the tolerability of resveratrol in humans, we cannot ignore the fact that studies about long-term resveratrol toxicity (or analogues such as pterostilbene) are still needed.

3.1.3. Silymarin

Silymarin is a dry flavonoid mixture extracted after processing the seeds of Silybum marium with ethanol, methanol, and acetone [140], which contains seven major components: taxifolin (the most effective antioxidant), silychristin, silydianin, silybin A, silybin B, isosilybin A and isosilybin B [141,142]. Although silymarin has mainly been used to treat liver diseases [143], its antidiabetic activity was recently reported and is associated with an anti-glycation profile [144,145], inhibition of aldose reductase [143], partial agonist activity in peroxisome proliferator-activated receptor γ (PPARγ) [143], antioxidant capacity and radical scavenging [144,146]. All these characteristics make silymarin an interesting candidate for the prevention and treatment of diabetic complications, which has recently been demonstrated both in experimental models and in humans (the same as for quercetin and resveratrol).
In 2013, Sheela et al. [146] demonstrated more fully that silymarin (60 and 120 mg/kg/day, i.m., for eight weeks) was able of reduce the classical signs of DM and attenuate the progression of the disease in a STZ-nicotinamide-induced nephropathy model (although the possible pathways were not investigated). However, in parallel, an in vitro study revealed that podocytes exposed to high glucose restored the · O 2 production and NADPH oxidase activity to basal levels through 10 μM of isolated compound silybin. In the same paper, Khazim et al. [147] obtained similar results in an in vivo experiment using 100 mg/kg/day of same substance (i.p., six weeks) in an advanced new model of diabetic nephropathy (OVE26 mice) with an additional reduction in albuminuria. These data also corroborated the findings of Vessal et al. [148], who used silymarin (100 mg/kg/day, i.p., for four weeks) in an STZ-rat model to obtain a reduction in kidney lipid peroxidation and increase the activity of catalase and GPx under hyperglycemia conditions.
Further studies are necessary to explore the other cytoprotective effects of silymarin. For example, Tuorkey et al. [140] recently showed that this flavonoid mixture (120 mg/kg, i.p., for 10 days) could protect cardiomyocytes against apoptosis in diabetic (alloxan) rats via restoration of caspase-3 and Bcl-2 to control levels. For neuroprotection, silymarin (100 mg/kg/day for eight weeks) ameliorated hyperalgesia and sciatic motor nerve conduction velocity in STZ-diabetic neuropathic rat by reducing lipoperoxidation and increasing SOD activity [149]. Moreover, silibinin in db/db mice provided DNA protection and reduced oxidative stress in a brain-specific area in rodents [150]. Therefore, these preliminary studies also revealed the potential of silymarin as a valid tool to counteract oxidative stress in the central nervous system under diabetic conditions [151].
Although there are still only a few clinical studies with silymarin, the results have reflected the findings of the laboratory studies. Approximately two decades ago, a study with silymarin supplementation (600 mg/day for 12 months) was conducted in insulin-treated diabetics with alcoholic cirrhosis, and the study had encouraging results. Beyond the antioxidant effects, there was a reduction of insulin resistance, a decrease in endogenous insulin hypersecretion and a reduced need for exogenous insulin administration [152]. Corroborating this observation, 10 years later, 25 diabetic (but non-cirrhotic) patients treated with silymarin (600 mg/day, for 16 weeks) showed reductions in glycemia, glycated hemoglobin, and an improved lipid profile in liver biomarkers [153]. In addition, Hussain et al. [154] showed that silymarin (200 mg/day, for 12 weeks) could be an important adjuvant for improving the glycemic control target by increasing insulin sensitivity in peripheral tissues through sulfonylureas (glibenclamide). This finding was recently corroborated by a study in which type 2 DM patients aged 25–50 years old who were on stable medications were supplemented with silymarin (420 mg/day, for six weeks). These patients also showed improvements in some antioxidant indices (SOD, GPx and total antioxidant capacity) as well as decreased lipid peroxidation besides hs-CRP levels without reporting any adverse effects of silymarin treatment [155]. Based on these results, more studies are still needed for the evaluation of the possible synergistic effects of silymarin with other antidiabetic classes (e.g., biguanides/metformin, dipeptidyl peptidase-4 inhibitors/sitagliptin; glucagon-like peptide-1 analogues/liraglutide; sodium-glucose cotransporter 2 inhibitors/dapagliflozin). Interestingly, silymarin has also been demonstrated to be an alternative treatment for diabetic renal patients who are using the maximum doses of angiotensin-converting enzyme (ACE) inhibitors or AT1 antagonists; even after a short duration treatment with silymarin (420 mg/day, for 12 weeks), these patients showed a reduction of 50% in albuminuria, urinary TNFα levels besides serum and urinary lipid peroxidation [156,157], which reflects a potential nephroprotective activity.
Although clinical trials with these polyphenols are still insufficient to define the optimal doses for treatment, the dosing range of silymarin used for diabetic patients is the closest to the ideal (when compared to diverse doses of resveratrol and quercetin) because it has clinically been investigated in several studies since the 1970s [143,158] compared to quercetin in 1995 [159] and resveratrol in 2007 [160]. The literature still describes that the therapeutic dose for the benefits of silymarin ranges between 210–800 mg/day, and silymarin appears to be safe and well tolerated up to 2100 mg/day [161], which reflects a wide therapeutic index [162]. Another additional advantage offered by silymarin in DM compared to other isolated polyphenols might be related to the relevance of silymarin use in combination with other antioxidants, which prevents individual antioxidant vulnerability and promotes synergistic effects against the chronic oxidative stress induced by diabetes [15].

4. Beneficial Effects of Probiotics: Highlights of Treatments with Kefir

In recent years, besides traditional drug treatments for DM, many efforts have been made in complementary or adjuvant therapy for the treatment of this complex disease [163]. Inadequate human dietary changes have been thought to be of major importance for the increased prevalence of DM. Overall, DM is estimated to afflict 350 million people globally and cost hundreds of billions of dollars annually [1,2]. Millions of cases could be prevented by including dietary modification to functional nutrition, which is a primary option for preventing metabolic disturbances and for reducing undesirable outcomes in DM [164,165]. Intestinal microbiota is a relevant therapeutic source for treatment of different diseases. Although there have been proposed different strategies including pre/probiotics and fecal microbiota transplantation interventions [166], in this section we review the main experimental and clinical studies that have focused on the beneficial effects of dairy cultured probiotics (live microorganisms) as coadjuvants in the prevention/treatment of this metabolic disorder.
Although many studies have been focused on the identification and use of innately occurring dairy components for the prevention and correction of metabolic dysfunctions accompanying DM [167], there is a growing and remarkable body of research showing the beneficial effects of non-innately cultured probiotics or bioactive end products [168]. These health benefits are achieved by stimulating beneficial gastrointestinal indigenous microflora proliferation [169]. Fermented milk kefir, which originated in the Northern Caucasus Mountains, is now commercially available in some countries, and in others it has been domestically produced and is spreading hand-to-hand [170]. The probiotic kefir has been associated with a range of health benefits, which have been reviewed by others [171], and its continuous intake has been shown to modulate complex cardiovascular and metabolic dysfunctions, including arterial hypertension [172] and DM [164].
In contrast with non-cultured dairy products, kefir grains are small clusters of microorganisms held together by an exopolysaccharide matrix named kefiran, which is the main functional component of the beverage [169,173,174,175]. Kefir grains are produced during the fermentation of milk by a complex symbiotic mixture of yeasts as well as lactic and acetic acid bacteria [170,173,174,176]. The dominating populations of bacterial genera in cultured kefir are Lactobacillus, Lactococcus and Streptococcus [169].
Rats administered with STZ (type 1 DM) or fed a hypercaloric diet (type 2 DM) are experimental models of DM [177]. In STZ-induced DM, it has been shown that daily administration of kefir caused an improvement in the increased levels of glycemia and glucose tolerance compared to conventional fermented milk [168,178,179]. Interestingly, kefiran, which is an exopolysaccharide isolated from kefir grains, has been shown to decrease blood pressure and blood glucose in animal models of hypertension [180] and an animal model of intolerance to glucose overload [181]. Kefiran-kefir also enhanced glucose uptake into insulin-responsive muscle cells, probably through activation of PI 3-kinases or another related signaling pathway [182]. Kefir also decreased polyuria, polydipsia and polyphagia [178]. In this model of DM, it has also been shown that administration of kefir results in a decrease in total cholesterol, triglycerides, LDL-cholesterol and an increase of HDL-cholesterol levels [179]. Moreover, kefir treatment of type 1 DM rats led to a decrease in the pro-inflammatory cytokines IL-1 and IL-6 as well as an increase of anti-inflammatory IL-10 compared to control groups [167]. These studies support the concept that kefir can be useful as a complementary or adjuvant therapy for better control of glycemia. However, the mechanisms by which probiotic kefir modulate hyperglycemia are not fully understood.
The beneficial effects of cultured probiotics have also been demonstrated in experimental type 2 DM. Administration of a strain of the probiotic microorganism Lactococcus lactis in rats with type 2 DM induced by a high-fructose diet resulted in significantly lower fasting blood glucose, HbA1c, insulin, free fatty acids and triglyceride levels than untreated DM rats [183]. By reducing the hyperglycemia, insulin resistance and hyperlipidemia, kefir actions were reflected in amelioration of the intracellular metabolic imbalance. In untreated DM animals, the excessive production of ROS overwhelmed the endogenous antioxidant defenses and resulted in oxidative stress, but this sequence of events can be attenuated through kefir treatment [183].
The use of antioxidant agents for therapeutic approaches in DM has been an attractive focus [184,185]. Accordingly, Friques et al. [172] observed through flow cytometry assays that kefir attenuated the endothelial dysfunction of spontaneously hypertensive rats by reducing the production of · O 2 , ONOO and H2O2. Augmented oxidative stress has also been shown to play a role in DM [185,186,187,188] and arterial hypertension [172]. First, it was shown that kefir reduced the intracellular levels of ROS in insulin-responsive muscle cells [182]. Second, the antioxidative effects of kefir in STZ-induced DM led to an improvement in the ROS levels [178,179]. The antioxidative effect seems to be the main mechanism by which kefir reduced proteinuria and azotemia, which consequently improved the progression of renal injury in type 1 DM rats [178]. These results indicated that kefir treatment may exert beneficial effects on the oxidative stress that accompanies DM and suggests it could be used as a non-pharmacological adjuvant to delay the progression of this disease [178].
Recently, our laboratory has assessed the actions of kefir on cardiac dysautonomia and impaired baroreflex control of cardiovascular function in SHR [189]. The main action of kefir on cardiac autonomic imbalance and impaired baroreflex appears to be through attenuation of the cardiac and vascular sympathetic hyperactivity as well as augmenting cardiac parasympathetic hypoactivity [189]. Some of these effects are also expected to occur in animal models of diabetes because they present similar disturbances in the cardiovascular system [190]. For example, in the model of type 1 DM, an important imbalance of the cardiac autonomic nerves, located at both tissues and molecular pathways, has been observed. Recently, in the first 10 weeks of experimental DM, a marked cardiac dysfunction and an incomplete recovery of the cardioinhibitory vagal nerves, accompanied by a remodeling process in the stimulatory noradrenergic nerves [191], have been shown.
Most clinical studies, including trials, have been conducted with patients who have type 2 DM, and most were treated with probiotic fermented milk kefir containing one, two or multi-strains of bacteria, such as Lactobacillus casei, L. acidophilus, L. bulgaricus, Streptococcus thermophiles and Bifidobacterium lactis [169]. The kefir effects observed on primary outcomes included decreased fasting blood glucose and HbA1c levels as well as improved insulin resistance [166,192,193,194]. The latter effect could be a consequence of a kefir-induced reduction in the inflammatory response [192]. In agreement with these results, it has been shown that kefir reduced pro-inflammatory cytokines, including TNFα, in DM [168,195]. The secondary outcomes included an improved lipid profile, blood pressure and hs-CRP, but kefir administration did not significantly change these parameters [191]. In contrast with experimental studies, it is still not clear whether kefir has beneficial effects on the lipid profile. An important finding after comparing the quantity of Lactobacillus and Bifidobacterium before and after the intervention was that there was successful passage of the probiotic supplement through the gastrointestinal tract [166,196]. The above studies support the concept that kefir can be useful as a complementary or adjuvant therapy for a better control of glycemia, insulin resistance and kidney function in diabetic individuals.
An important characteristic of DM is endothelial dysfunction, which has been shown in experimental and clinical studies [197]. Our laboratory has demonstrated that kefir administered for at least 60 days in spontaneously hypertensive rats resulted in a significant attenuation of endothelial dysfunction [172]. Therefore, there is a need for more studies to test the hypothesis that kefir administration also exhibits benefits against this abnormality.
A limitation in the therapeutic use of the probiotic kefir in DM is that this is a heterogeneous and a multiple systems-derived disease that results in multiple complications. Therefore, it makes it hard to prevent or to treat DM with traditional medicine and functional nutrition, especially when treating different people with different needs. However, there is evidence that kefir has great potential to become an adjuvant alternative for control of glycemia and other diabetes-related outcomes. Further studies are needed not only to clarify the mechanisms behind the effects of kefir but also to determine which microorganisms in kefir are responsible for its benefits.

5. Beneficial Effects of Phosphodiesterase Inhibitors in Diabetes Mellitus: New Insights

Several investigations have demonstrated that increased cyclic GMP (cGMP) signaling might be an important strategy for reducing the progression of diabetes through multiple pathways [198]. Concerning glycemic control, even if an increase in intracellular calcium is the principal signal that activates insulin exocytosis, cGMP may also participate through distinct signals [199,200] and potentiating the stimulation of glucose [201,202,203]. In parallel, some in vitro studies have shown that cGMP may enhance insulin sensitivity in target organs (muscle and adipocytes) by stimulating GLUT4 recruitment into the plasma membrane [204,205,206]. In addition, because NO/cGMP signaling is fundamental to vascular protection [207,208,209], the increment of this pathway may be an attractive strategy to attenuate endothelial dysfunction development in diabetic complications, which is as major cause of disability and death in patients with DM [4,44].
Pharmacological strategies to increase cGMP signaling may be achieved through two main routes: (1) direct activation of guanylate cyclase directly by augmentation of NO; and/or (2) decreasing cGMP hydrolysis through PDE5 inhibitors (sildenafil/Viagra™, vardenafil/Levitra™, tadalafil/Cialis™, avanafil/Stendra™), which is currently considered an important tool to treat endothelial dysfunction in DM [209,210,211]. Because PDE5 is expressed in some tissues of the body (e.g., corpus cavernosum, platelets, systemic arteries and veins) [209,212], the diminished NO bioavailability in diabetic vasculature can be partially compensated through PDE5 inhibitors. Interestingly, although some recent studies demonstrated that association with antioxidants (e.g., polyphenols or vitamin E) potentiates vascular protection [213,214], sildenafil may also provide intrinsic antioxidant effects through NADPH oxidase activity inhibition [215]. This evidence was complemented by our research in various models of hypertension, nephropathy or atherosclerosis and demonstrated protective effects for endothelial, cardiac and kidney functions in physiological parameters, such as morphological analyses (Figure 1) [209,212,216,217,218,219]. Therefore, our studies and other experimental evidence support the clinical findings of improvement in endothelial function, reduction of markers of vascular inflammation, and beneficial effects for conditions beyond erectile dysfunction [220,221,222,223,224].
In relation to glycemic control, PDE5 inhibitors may have improved insulin sensitivity both in isolated human endothelial cells [225] and in high fat-fed mice [226], which corroborates the results related to cGMP that were previously discussed. Moreover, recent findings showed that chronic treatment with tadalafil reduced inflammatory cytokines, infarct size and oxidative stress in the hearts of diabetic mice by reducing NADPH oxidase activity, oxidized glutathione and lipid peroxidation [227,228]. These results show a potential role for PDE5 inhibitors in treating diabetes-related cardiac and inflammatory complications. Interestingly, clinical investigations confirmed that sildenafil could improve insulin sensitivity in addition to fibrinolytic balance and albuminuria in hyperglycemic patients [198,229]. These studies suggest that PDE inhibitors can be effective (and safely used) in patients with multiple comorbidities and therapies, except for patients treated with continuous nitrates [209,223,230].

6. Conclusions

In the present review, besides showing the importance of lifestyle modification, diet and weight control to prevent DM and its aggravation, we highlight recent experimental and clinical evidences of potential coadjuvants in the management of DM without compromising the function of β-cells via hyperinsulinism. Here, we have discussed the substances that exhibit direct/indirect or pleiotropic effects on glycemic control in DM and on oxidative stress that is one of the most contributors to the complications of this disease by affecting the different target organs. Among several options, we have highlighted new promising therapeutic coadjuvants, including the cultured probiotic microorganisms (such as kefir grains) and the PDE5 inhibitors (such as sildenafil), which besides the reduction of hyperglycemia and ameliorate insulin resistance, they have been shown to reduce the oxidative stress and improved the endothelial dysfunction in the systemic vascular circulation. In the near future, there are expected experimental studies designed to clear the intracellular pathways involving those coadjuvants discussed in this review as well as promoting clinical trials that may contribute to new strategies against the harmful effects of diabetic complications.

Acknowledgments

Elisardo Corral Vasquez is supported by National Council for the Development of Science and Technology (CNPq) (Grant 303001/2015-1) and State Foundation for Science and Technology (Fapes) (Grant Universal 2014 Proc 67597483). Silvana S. Meyrelles is supported by CNPq (307584/2015-1). Bianca P. Campagnaro is supported by CNPq (445736/2014-3). Thiago Melo C. Pereira is supported by CNPq (445080/2014-0).

Author Contributions

Elisardo Corral Vasquez supervised the analysis of publication data, and edited and approved the final version of this review. The contribution of the collaborators for each section is as follows. Section 1: Marcelo Perim Baldo and Silvana Santos Meyrelles; Section 2: Marcella Lima Porto and Agata Lages Gava; Section 3: Thiago Melo Costa Pereira and Bianca Prandi Campagnaro; Section 4: Fabio Silva Pimenta and Silvana Santos Meyrelles; Section 5: Thiago Melo Costa Pereira and Elisardo Corral Vasquez and creation of Figure 1: Thiago Melo Costa Pereira and Elisardo Corral Vasquez.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACEangiotensin-converting enzyme
AGEsglycation end products
cGMPcyclic guanosine monophosphate
COXcyclooxygenase
CRPC-reactive protein
CVDcardiovascular diseases
CYP450cytochrome P450
DMdiabetes mellitus
ECMextracellular matrix
eGFRestimated glomerular filtration rate
eNOSendothelial nitric oxide synthase
GLP-1glucagon-like peptide-1
GLUTglucose transporter
GPxglutathione peroxidase
H2O2hydrogen peroxide
HbA1cglycated hemoglobin level
HDLhigh density lipoprotein
hs-CRPhigh-sensitivity C-reactive protein
IGTimpaired glucose tolerance
ILinterleukin
iPDEPDE inhibitors
LDLlow density lipoprotein
NADHnicotinamide adenine dinucleotide
NADPHnicotinamide adenine dinucleotide phosphate
NF-κBnuclear factor-κB
NHAHESnational health and nutrition examination survey
NOnitric oxide
NOSnitric oxide synthase
NoxNADPH oxidases
· O 2 superoxide anion
ONOOperoxynitrite
OHhydroxyl
·OHhydroxyl radical
PARPpoly ADP ribose polymerase
PDE5phosphodiesterase 5
PKCprotein kinase C
PPARγproliferator-activated receptor γ
ROSreactive oxygen species
SHRspontaneously hypertensive rats
SIRTsirtuin 1
SODsuperoxide dismutase
STZstreptozotocin
TNFαtumor necrosis factor

References

  1. Garcia-Perez, L.E.; Alvarez, M.; Dilla, T.; Gil-Guillen, V.; Orozco-Beltran, D. Adherence to therapies in patients with type 2 diabetes. Diabetes Ther. 2013, 4, 175–194. [Google Scholar] [CrossRef] [PubMed]
  2. Cheung, B.M.; Ong, K.L.; Cherny, S.S.; Sham, P.C.; Tso, A.W.; Lam, K.S. Diabetes prevalence and therapeutic target achievement in the United States, 1999 to 2006. Am. J. Med. 2009, 122, 443–453. [Google Scholar] [CrossRef] [PubMed]
  3. Gu, K.; Cowie, C.C.; Harris, M.I. Mortality in adults with and without diabetes in a national cohort of the U.S. population, 1971–1993. Diabetes Care 1998, 21, 1138–1145. [Google Scholar] [CrossRef] [PubMed]
  4. Potenza, M.A.; Gagliardi, S.; Nacci, C.; Carratu, M.R.; Montagnani, M. Endothelial dysfunction in diabetes: From mechanisms to therapeutic targets. Curr. Med. Chem. 2009, 16, 94–112. [Google Scholar] [CrossRef] [PubMed]
  5. Santi, D.; Giannetta, E.; Isidori, A.M.; Vitale, C.; Aversa, A.; Simoni, M. Therapy of endocrine disease. Effects of chronic use of phosphodiesterase inhibitors on endothelial markers in type 2 diabetes mellitus: A meta-analysis. Eur. J. Endocrinol. 2015, 172, R103–R114. [Google Scholar] [CrossRef] [PubMed]
  6. Ong, K.L.; Cheung, B.M.; Wong, L.Y.; Wat, N.M.; Tan, K.C.; Lam, K.S. Prevalence, treatment, and control of diagnosed diabetes in the U.S. National Health and Nutrition Examination Survey 1999–2004. Ann. Epidemiol. 2008, 18, 222–229. [Google Scholar] [CrossRef] [PubMed]
  7. Wegman, M.P.; Guo, M.H.; Bennion, D.M.; Shankar, M.N.; Chrzanowski, S.M.; Goldberg, L.A.; Xu, J.; Williams, T.A.; Lu, X.; Hsu, S.I.; et al. Practicality of intermittent fasting in humans and its effect on oxidative stress and genes related to aging and metabolism. Rejuvenation Res. 2015, 18, 162–172. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, Y. Molecular Links between Caloric Restriction and Sir2/SIRT1 Activation. Diabetes Metab. J. 2014, 38, 321–329. [Google Scholar] [CrossRef] [PubMed]
  9. Kuecker, C.M.; Vivian, E.M. Patient considerations in type 2 diabetes—Role of combination dapagliflozin-metformin XR. Diabetes Metab. Syndr. Obes. 2016, 9, 25–35. [Google Scholar] [PubMed]
  10. Sharma, B.R.; Kim, H.J.; Rhyu, D.Y. Caulerpa lentillifera extract ameliorates insulin resistance and regulates glucose metabolism in C57BL/KsJ-db/db mice via PI3K/AKT signaling pathway in myocytes. J. Transl. Med. 2015, 13, 62. [Google Scholar] [CrossRef] [PubMed]
  11. Shera, A.S.; Jawad, F.; Maqsood, A.; Jamal, S.; Azfar, M.; Ahmed, U. Prevalence of chronic complications and associated factors in type 2 diabetes. J. Pak. Med. Assoc. 2004, 54, 54–59. [Google Scholar] [PubMed]
  12. Siddiqui, F.J.; Avan, B.I.; Mahmud, S.; Nanan, D.J.; Jabbar, A.; Assam, P.N. Uncontrolled diabetes mellitus: Prevalence and risk factors among people with type 2 diabetes mellitus in an Urban District of Karachi, Pakistan. Diabetes Res. Clin. Pract. 2015, 107, 148–156. [Google Scholar] [CrossRef] [PubMed]
  13. Hamwi, G.J.; Garcia, O.; Kruger, F.A.; Gwinup, G.; Cornwell, D.G. Hyperlipidemia in uncontrolled diabetes. Metabolism 1962, 11, 850–862. [Google Scholar] [PubMed]
  14. Moinat, P.; Nichols, N.; Tuller, E.F. Changes in serum proteins and polysaccharides in rats with uncontrolled diabetes. Diabetes 1956, 5, 468–474. [Google Scholar] [PubMed]
  15. Litchfield, J.A. Biochemical changes in skeletal muscle in patients with uncontrolled diabetes mellitus. Diabetes 1959, 8, 257–260. [Google Scholar] [CrossRef] [PubMed]
  16. Afsharian, S.; Akbarpour, S.; Abdi, H.; Sheikholeslami, F.; Moeini, A.S.; Khalili, D.; Momenan, A.A.; Azizi, F.; Hadaegh, F. Risk factors for cardiovascular disease and mortality events in adults with type 2 diabetes: A 10 year follow-up: Tehran lipid and glucose study. Diabetes Metab. Res. Rev. 2016. [Google Scholar] [CrossRef] [PubMed]
  17. Mulnier, H.E.; Seaman, H.E.; Raleigh, V.S.; Soedamah-Muthu, S.S.; Colhoun, H.M.; Lawrenson, R.A.; de Vries, C.S. Risk of myocardial infarction in men and women with type 2 diabetes in the UK: A cohort study using the General Practice Research Database. Diabetologia 2008, 51, 1639–1645. [Google Scholar] [CrossRef] [PubMed]
  18. An, Y.; Zhang, P.; Wang, J.; Gong, Q.; Gregg, E.W.; Yang, W.; Li, H.; Zhang, B.; Shuai, Y.; Chen, Y.; et al. Cardiovascular and all-cause mortality over a 23-year period among chinese with newly diagnosed diabetes in the Da Qing IGT and Diabetes Study. Diabetes Care 2015, 38, 1365–1371. [Google Scholar] [CrossRef] [PubMed]
  19. Peters, S.A.; Huxley, R.R.; Woodward, M. Diabetes as a risk factor for stroke in women compared with men: A systematic review and meta-analysis of 64 cohorts, including 775,385 individuals and 12,539 strokes. Lancet 2014, 383, 1973–1980. [Google Scholar] [CrossRef]
  20. Lee, M.; Saver, J.L.; Hong, K.S.; Song, S.; Chang, K.H.; Ovbiagele, B. Effect of pre-diabetes on future risk of stroke: Meta-analysis. BMJ 2012, 344. [Google Scholar] [CrossRef] [PubMed]
  21. Collins, A.J.; Foley, R.N.; Herzog, C.; Chavers, B.; Gilbertson, D.; Ishani, A.; Kasiske, B.; Liu, J.; Mau, L.W.; McBean, M.; et al. US renal data system 2010 annual data report. Am. J. Kidney Dis. 2011, 57, e1–e526. [Google Scholar] [CrossRef] [PubMed]
  22. Radbill, B.; Murphy, B.; LeRoith, D. Rationale and strategies for early detection and management of diabetic kidney disease. Mayo Clin. Proc. 2008, 83, 1373–1381. [Google Scholar] [CrossRef] [PubMed]
  23. de Boer, I.H.; Rue, T.C.; Hall, Y.N.; Heagerty, P.J.; Weiss, N.S.; Himmelfarb, J. Temporal trends in the prevalence of diabetic kidney disease in the United States. JAMA 2011, 305, 2532–2539. [Google Scholar] [CrossRef] [PubMed]
  24. Salinero-Fort, M.Á.; San Andrés-Rebollo, F.J.; de Burgos-Lunar, C.; Abánades-Herranz, J.C.; Carrillo-de-Santa-Pau, E.; Chico-Moraleja, R.M.; Jiménez-García, R.; López-de-Andrés, A.; Gómez-Campelo, P. Cardiovascular and all-cause mortality in patients with type 2 diabetes mellitus in the MADIABETES Cohort Study: Association with chronic kidney disease. J. Diabetes Complic. 2016, 30, 227–236. [Google Scholar] [CrossRef] [PubMed]
  25. Brownlee, M. The pathobiology of diabetic complications: A unifying mechanism. Diabetes 2005, 54, 1615–1625. [Google Scholar] [CrossRef] [PubMed]
  26. Kitabchi, A.E.; Umpierrez, G.E.; Miles, J.M.; Fisher, J.N. Hyperglycemic crises in adult patients with diabetes. Diabetes Care 2009, 32, 1335–1343. [Google Scholar] [CrossRef] [PubMed]
  27. Kitabchi, A.E.; Nyenwe, E.A. Hyperglycemic crises in diabetes mellitus: Diabetic ketoacidosis and hyperglycemic hyperosmolar state. Endocrinol. Metab. Clin. N. Am. 2006, 35, 725–751. [Google Scholar] [CrossRef] [PubMed]
  28. Davis, S.N.; Umpierrez, G.E. Diabetic ketoacidosis in type 2 diabetes mellitus-pathophysiology and clinical presentation. Nat. Clin. Pract. Endocrinol. Metab. 2007, 3, 730–731. [Google Scholar] [CrossRef] [PubMed]
  29. Lewko, B.; Stepinski, J. Hyperglycemia and mechanical stress: Targeting the renal podocyte. J. Cell. Physiol. 2009, 221, 288–295. [Google Scholar] [CrossRef] [PubMed]
  30. Di Mario, U.; Pugliese, G. 15th Golgi lecture: From hyperglycaemia to the dysregulation of vascular remodelling in diabetes. Diabetologia 2001, 44, 674–692. [Google Scholar] [CrossRef] [PubMed]
  31. Beisswenger, P.J.; Howell, S.K.; Nelson, R.G.; Mauer, M.; Szwergold, B.S. α-Oxoaldehyde metabolism and diabetic complications. Biochem. Soc. Trans. 2003, 31, 1358–1363. [Google Scholar] [CrossRef] [PubMed]
  32. Nishikawa, T.; Edelstein, D.; Du, X.L.; Yamagishi, S.; Matsumura, T.; Kaneda, Y.; Yorek, M.A.; Beebe, D.; Oates, P.J.; Hammes, H.P.; et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000, 404, 787–790. [Google Scholar] [PubMed]
  33. Lee, H.B.; Yu, M.R.; Yang, Y.; Jiang, Z.; Ha, H. Reactive oxygen species-regulated signaling pathways in diabetic nephropathy. J. Am. Soc. Nephrol. 2003, 14, S241–S245. [Google Scholar] [CrossRef] [PubMed]
  34. Dunlop, M. Aldose reductase and the role of the polyol pathway in diabetic nephropathy. Kidney Int. Suppl. 2000, 77, S3–S12. [Google Scholar] [CrossRef] [PubMed]
  35. Tomlinson, D.R.; Gardiner, N.J. Glucose neurotoxicity. Nat. Rev. Neurosci. 2008, 9, 36–45. [Google Scholar] [CrossRef] [PubMed]
  36. Luo, X.; Wu, J.; Jing, S.; Yan, L.J. Hyperglycemic stress and carbon stress in diabetic glucotoxicity. Aging Dis. 2016, 7, 90–110. [Google Scholar] [CrossRef] [PubMed]
  37. Ying, W. NAD+/NADH and NADP+/NADPH in cellular functions and cell death: Regulation and biological consequences. Antioxid. Redox Signal. 2008, 10, 179–206. [Google Scholar] [CrossRef] [PubMed]
  38. Dolle, C.; Rack, J.G.; Ziegler, M. NAD and ADP-ribose metabolism in mitochondria. FEBS J. 2013, 280, 3530–3541. [Google Scholar] [CrossRef] [PubMed]
  39. Du, X.L.; Edelstein, D.; Rossetti, L.; Fantus, I.G.; Goldberg, H.; Ziyadeh, F.; Wu, J.; Brownlee, M. Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc. Natl. Acad. Sci. USA 2000, 97, 12222–12226. [Google Scholar] [CrossRef] [PubMed]
  40. Porto, M.L.; Rodrigues, B.P.; Menezes, T.N.; Ceschim, S.L.; Casarini, D.E.; Gava, A.L.; Pereira, T.M.; Vasquez, E.C.; Campagnaro, B.P.; Meyrelles, S.S. Reactive oxygen species contribute to dysfunction of bone marrow hematopoietic stem cells in aged C57BL/6 J mice. J. Biomed. Sci. 2015, 22, 97. [Google Scholar] [CrossRef] [PubMed]
  41. Nakagami, H.; Morishita, R.; Yamamoto, K.; Yoshimura, S.I.; Taniyama, Y.; Aoki, M.; Matsubara, H.; Kim, S.; Kaneda, Y.; Ogihara, T. Phosphorylation of p38 mitogen-activated protein kinase downstream of bax-caspase-3 pathway leads to cell death induced by high d-glucose in human endothelial cells. Diabetes 2001, 50, 1472–1481. [Google Scholar] [CrossRef] [PubMed]
  42. Nakagami, H.; Kaneda, Y.; Ogihara, T.; Morishita, R. Endothelial dysfunction in hyperglycemia as a trigger of atherosclerosis. Curr. Diabetes Rev. 2005, 1, 59–63. [Google Scholar] [CrossRef] [PubMed]
  43. Wei, W.; Liu, Q.; Tan, Y.; Liu, L.; Li, X.; Cai, L. Oxidative stress, diabetes, and diabetic complications. Hemoglobin 2009, 33, 370–377. [Google Scholar] [CrossRef] [PubMed]
  44. Domingueti, C.P.; Dusse, L.M.; Carvalho, M.D.; de Sousa, L.P.; Gomes, K.B.; Fernandes, A.P. Diabetes mellitus: The linkage between oxidative stress, inflammation, hypercoagulability and vascular complications. J. Diabetes Complic. 2016, 30, 738–745. [Google Scholar] [CrossRef] [PubMed]
  45. Gomes, I.B.; Porto, M.L.; Santos, M.C.; Campagnaro, B.P.; Pereira, T.M.; Meyrelles, S.S.; Vasquez, E.C. Renoprotective, anti-oxidative and anti-apoptotic effects of oral low-dose quercetin in the C57BL/6J model of diabetic nephropathy. Lipids Health Dis. 2014, 6, 184. [Google Scholar] [CrossRef] [PubMed]
  46. Gomes, I.B.; Porto, M.L.; Santos, M.C.; Campagnaro, B.P.; Gava, A.L.; Meyrelles, S.S.; Pereira, T.M.; Vasquez, E.C. The protective effects of oral low-dose quercetin on diabetic nephropathy in hypercholesterolemic mice. Front. Physiol. 2015, 6, 247. [Google Scholar] [CrossRef] [PubMed]
  47. Holmström, K.M.; Finkel, T. Cellular mechanisms and physiological consequences of redox-dependent signaling. Nat. Rev. Mol. Cell Biol. 2014, 15, 411–421. [Google Scholar] [CrossRef] [PubMed]
  48. Kayama, Y.; Raaz, U.; Jagger, A.; Adam, M.; Schellinger, I.N.; Sakamoto, M.; Suzuki, H.; Toyama, K.; Spin, J.M.; Tsao, P.S. Diabetic cardiovascular disease induced by oxidative stress. Int. J. Mol. Sci. 2015, 16, 25234–25263. [Google Scholar] [CrossRef] [PubMed]
  49. Münzel, T.; Gori, T.; Bruno, R.M.; Taddei, S. Is oxidative stress a therapeutic target in cardiovascular disease? Eur. Heart J. 2010, 31, 2741–2748. [Google Scholar] [CrossRef] [PubMed]
  50. Ceriello, A.; Testa, R.; Genovese, S. Clinical implications of oxidative stress and potential role of natural antioxidants in diabetic vascular complications. Nutr. Metab. Cardiovasc. Dis. 2016, 26, 285–292. [Google Scholar] [CrossRef] [PubMed]
  51. Ide, T.; Tsutsui, H.; Kinugawa, S.; Utsumi, H.; Kang, D.; Hattori, N.; Uchida, K.; Arimura, K.; Egashira, K.; Takeshita, A. Mitochondrial electron transport complex I is a potential source of oxygen free radicals in the failing myocardium. Circ. Res 1999, 85, 357–363. [Google Scholar] [CrossRef] [PubMed]
  52. Selemidis, S.; Sobey, C.G.; Wingler, K.; Schmidt, H.H.; Drummond, G.R. NADPH oxidases in the vasculature: Molecular features, roles in disease and pharmacological inhibition. Pharmacol. Ther. 2008, 120, 254–291. [Google Scholar] [CrossRef] [PubMed]
  53. Zhou, G.; Li, X.; Hein, D.W.; Xiang, X.; Marshall, J.P.; Prabhu, S.D.; Cai, L. Metallothionein suppresses angiotensin II-induced nicotinamide adenine dinucleotide phosphate oxidase activation, nitrosative stress, apoptosis, and pathological remodeling in the diabetic heart. J. Am. Coll. Cardiol. 2008, 52, 655–666. [Google Scholar] [CrossRef] [PubMed]
  54. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef] [PubMed]
  55. Tan, S.M.; de Haan, J.B. Combating oxidative stress in diabetic complications with Nrf2 activators: How much is too much? Redox Rep. 2014, 19, 107–117. [Google Scholar] [CrossRef] [PubMed]
  56. Xu, X.; Luo, P.; Wang, Y.; Cui, Y.; Miao, L. Nuclear factor (erythroid-derived 2)-like 2 (NFE2L2) is a novel therapeutic target for diabetic complications. J. Int. Med. Res. 2013, 41, 13–19. [Google Scholar] [CrossRef] [PubMed]
  57. Murali, R.; Karthikeyan, A.; Saravanan, R. Protective effects of d-limonene on lipid peroxidation and antioxidant enzymes in streptozotocin-induced diabetic rats. Basic Clin. Pharmacol. Toxicol. 2013, 112, 175–181. [Google Scholar] [CrossRef] [PubMed]
  58. Ghattas, M.H.; Abo-Elmatty, D.M. Association of polymorphic markers of the catalase and superoxide dismutase genes with type 2 diabetes mellitus. DNA Cell Biol. 2012, 31, 1598–1603. [Google Scholar] [CrossRef] [PubMed]
  59. Batinic-Haberle, I.; Reboucas, J.S.; Spasojevic, I. Superoxide dismutase mimics: Chemistry, pharmacology, and therapeutic potential. Antioxid. Redox Signal. 2010, 13, 877–918. [Google Scholar] [CrossRef] [PubMed]
  60. Soto-Urquieta, M.G.; López-Briones, S.; Pérez-Vázquez, V.; Saavedra-Molina, A.; González-Hernández, G.A.; Ramírez-Emiliano, J. Curcumin restores mitochondrial functions and decreases lipid peroxidation in liver and kidneys of diabetic db/db mice. Biol. Res. 2014, 47, 74. [Google Scholar] [CrossRef] [PubMed]
  61. Pitocco, D.; Tesauro, M.; Alessandro, R.; Ghirlanda, G.; Cardillo, C. Oxidative stress in diabetes: Implications for vascular and other complications. Int. J. Mol. Sci. 2013. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, X.; Gan, W.; Zou, Y.; Yang, B.; Su, Z.; Deng, J.; Wang, L.; Cai, J. Elevated levels of urinary markers of oxidative DNA and RNA damage in type 2 diabetes with complications. Oxid. Med. Cell. Longev. 2016, 2016, 4323198. [Google Scholar] [CrossRef] [PubMed]
  63. Palem, S.P.; Abraham, P. A study on the level of oxidative stress and inflammatory markers in type 2 diabetes mellitus patients with different treatment modalities. J. Clin. Diagn. Res. 2015. [Google Scholar] [CrossRef] [PubMed]
  64. Araki, E.; Nishikawa, T. Oxidative stress: A cause and therapeutic target of diabetic complications. J. Diabetes Investig. 2010, 1, 90–96. [Google Scholar] [CrossRef] [PubMed]
  65. Zatalia, S.R.; Sanusi, H. The role of antioxidants in the pathophysiology, complications, and management of diabetes mellitus. Acta Med. Indones 2013, 45, 141–147. [Google Scholar] [PubMed]
  66. Su, H.C.; Hung, L.M.; Chen, J.K. Resveratrol, a red wine antioxidant, possesses an insulin-like effect in streptozotocin-induced diabetic rats. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E1339–E1346. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, Z.; Li, Y.; Li, Y. Grape seed proanthocyanidin extracts prevent hyperglycemia-induced monocyte adhesion to aortic endothelial cells and ameliorates vascular inflammation in high-carbohydrate/high-fat diet and streptozotocin-induced diabetic rats. Int. J. Food Sci. Nutr. 2015, 67, 524–534. [Google Scholar] [CrossRef] [PubMed]
  68. D’Andrea, G. Quercetin: A flavonol with multifaceted therapeutic applications? Fitoterapia 2015, 106, 256–271. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, J.Y.; Chen, X.X.; Tang, S.C.; Sze, S.C.; Feng, Y.; Lee, K.F.; Zhang, K.Y. Chinese medicines in the treatment of experimental diabetic nephropathy. Chin. Med. 2016, 11, 6. [Google Scholar] [CrossRef] [PubMed]
  70. Okamoto, T. Safety of quercetin for clinical application (Review). Int. J. Mol. Med. 2005, 16, 275–278. [Google Scholar] [CrossRef] [PubMed]
  71. Varma, S.D.; Mikuni, I.; Kinoshita, J.H. Flavonoids as inhibitors of lens aldose reductase. Science 1975, 188, 1215–1216. [Google Scholar] [CrossRef] [PubMed]
  72. Alam, M.M.; Meerza, D.; Naseem, I. Protective effect of quercetin on hyperglycemia, oxidative stress and DNA damage in alloxan induced type 2 diabetic mice. Life Sci. 2014, 109, 8–14. [Google Scholar] [CrossRef] [PubMed]
  73. Xu, M.; Hu, J.; Zhao, W.; Gao, X.; Jiang, C.; Liu, K.; Liu, B.; Huang, F. Quercetin differently regulates insulin-mediated glucose transporter 4 translocation under basal and inflammatory conditions in adipocytes. Mol. Nutr. Food Res. 2014, 58, 931–941. [Google Scholar] [CrossRef] [PubMed]
  74. Henagan, T.M.; Lenard, N.R.; Gettys, T.W.; Stewart, L.K. Dietary quercetin supplementation in mice increases skeletal muscle PGC1α expression, improves mitochondrial function and attenuates insulin resistance in a time-specific manner. PLoS ONE 2014, 9, e89365. [Google Scholar] [CrossRef] [PubMed]
  75. Eid, H.M.; Nachar, A.; Thong, F.; Sweeney, G.; Haddad, P.S. The molecular basis of the antidiabetic action of quercetin in cultured skeletal muscle cells and hepatocytes. Pharmacogn. Mag. 2015, 11, 74–81. [Google Scholar] [PubMed]
  76. Kim, J.H.; Kang, M.J.; Choi, H.N.; Jeong, S.M.; Lee, Y.M.; Kim, J.I. Quercetin attenuates fasting and postprandial hyperglycemia in animal models of diabetes mellitus. Nutr. Res. Pract. 2011, 5, 107–111. [Google Scholar] [CrossRef] [PubMed]
  77. Kwon, O.; Eck, P.; Chen, S.; Corpe, C.P.; Lee, J.H.; Kruhlak, M.; Levine, M. Inhibition of the intestinal glucose transporter GLUT2 by flavonoids. FASEB J. 2007, 21, 366–377. [Google Scholar] [CrossRef] [PubMed]
  78. Mahesh, T.; Menon, V.P. Quercetin allievates oxidative stress in streptozotocin-induced diabetic rats. Phytother. Res. 2004, 18, 123–127. [Google Scholar] [CrossRef] [PubMed]
  79. Oršolić, N.; Gajski, G.; Garaj-Vrhovac, V.; Dikić, D.; Prskalo, Z.Š.; Sirovina, D. DNA-protective effects of quercetin or naringenin in alloxan-induced diabetic mice. Eur. J. Pharmacol. 2011, 656, 110–118. [Google Scholar] [CrossRef] [PubMed]
  80. Hollman, P.C.; van Trijp, J.M.; Buysman, M.N.; van der Gaag, M.S.; Mengelers, M.J.; de Vries, J.H.; Katan, M.B. Relative bioavailability of the antioxidant flavonoid quercetin from various foods in man. FEBS Lett. 1997, 418, 152–156. [Google Scholar] [CrossRef]
  81. De Boer, V.C.; Dihal, A.A.; van der Woude, H.; Arts, I.C.; Wolffram, S.; Alink, G.M.; Rietjens, I.M.; Keijer, J.; Hollman, P.C. Tissue distribution of quercetin in rats and pigs. J. Nutr. 2005, 135, 1718–1725. [Google Scholar] [PubMed]
  82. Bakhshaeshi, M.; Khaki, A.; Fathiazad, F.; Khaki, A.A.; Ghadamkheir, E. Anti-oxidative role of quercetin derived from Allium cepa on aldehyde oxidase (OX-LDL) and hepatocytes apoptosis in streptozotocin-induced diabetic rat. Asian Pac. J. Trop. Biomed. 2012, 2, 528–531. [Google Scholar] [CrossRef]
  83. Dodda, D.; Ciddi, V. Plants used in the management of diabetic complications. Indian J. Pharm. Sci. 2014, 76, 97–106. [Google Scholar] [PubMed]
  84. Umathe, S.N.; Dixit, P.V.; Kumar, V.; Bansod, K.U.; Wanjari, M.M. Quercetin pretreatment increases the bioavailability of pioglitazone in rats: Involvement of CYP3A inhibition. Biochem. Pharmacol. 2008, 75, 1670–1676. [Google Scholar] [CrossRef] [PubMed]
  85. Sanders, R.A.; Rauscher, F.M.; Watkins, J.B. Effects of quercetin on antioxidant defense in streptozotocin-induced diabetic rats. J. Biochem. Mol. Toxicol. 2001, 15, 143–149. [Google Scholar] [CrossRef] [PubMed]
  86. Elbe, H.; Esrefoglu, M.; Vardi, N.; Taslidere, E.; Ozerol, E.; Tanbek, K. Melatonin, quercetin and resveratrol attenuates oxidative hepatocellular injury in streptozotocin-induced diabetic rats. Hum. Exp. Toxicol. 2015, 34, 859–868. [Google Scholar] [CrossRef] [PubMed]
  87. Ortega, R.; García, N. The flavonoid quercetin induces changes in mitochondrial permeability by inhibiting adenine nucleotide translocase. J. Bioenerg. Biomembr. 2009, 41, 41–47. [Google Scholar] [CrossRef] [PubMed]
  88. Sivitz, W.I.; Yorek, M.A. Mitochondrial dysfunction in diabetes: From molecular mechanisms to functional significance and therapeutic opportunities. Antioxid. Redox Signal. 2010, 12, 537–577. [Google Scholar] [CrossRef] [PubMed]
  89. Kowluru, R.A.; Mishra, M. Oxidative stress, mitochondrial damage and diabetic retinopathy. Biochim. Biophys. Acta 2015, 1852, 2474–2483. [Google Scholar] [CrossRef] [PubMed]
  90. Gonzalez Suarez, M.L.; Thomas, D.B.; Barisoni, L.; Fornoni, A. Diabetic nephropathy: Is it time yet for routine kidney biopsy? World J. Diabetes 2013, 4, 245–255. [Google Scholar] [PubMed]
  91. Kushiyama, A.; Tanaka, K.; Hara, S.; Kawazu, S. Linking uric acid metabolism to diabetic complications. World J. Diabetes 2014, 5, 787–795. [Google Scholar] [CrossRef] [PubMed]
  92. Kumar, B.; Gupta, S.K.; Nag, T.C.; Srivastava, S.; Saxena, R.; Jha, K.A.; Srinivasan, B.P. Retinal neuroprotective effects of quercetin in streptozotocin-induced diabetic rats. Exp. Eye Res. 2014, 125, 193–202. [Google Scholar] [CrossRef] [PubMed]
  93. Mahmoud, M.F.; Hassan, N.A.; El Bassossy, H.M.; Fahmy, A. Quercetin protects against diabetes-induced exaggerated vasoconstriction in rats: Effect on low grade inflammation. PLoS ONE 2013, 8, e63784. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, W.; Wang, Y.; Yang, Z.; Qiu, J.; Ma, J.; Zhao, Z.; Bao, T. Antioxidant treatment with quercetin ameliorates erectile dysfunction in streptozotocin-induced diabetic rats. J. Biosci. Bioeng. 2011, 112, 215–218. [Google Scholar] [CrossRef] [PubMed]
  95. Anjaneyulu, M.; Chopra, K. Quercetin attenuates thermal hyperalgesia and cold allodynia in STZ-induced diabetic rats. Indian J. Exp. Biol. 2004, 42, 766–769. [Google Scholar] [PubMed]
  96. Anjaneyulu, M.; Chopra, K. Quercetin, an anti-oxidant bioflavonoid, attenuates diabetic nephropathy in rats. Clin. Exp. Pharmacol. Physiol. 2004, 31, 244–248. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, P.; Chen, J.; Zheng, Q.; Chen, W.; Wang, Y.; Xu, X. Pioglitazone, extract of compound Danshen dripping pill, and quercetin ameliorate diabetic nephropathy in diabetic rats. J. Endocrinol. Investig. 2013, 36, 422–427. [Google Scholar]
  98. Wang, C.; Pan, Y.; Zhang, Q.Y.; Wang, F.M.; Kong, L.D. Quercetin and allopurinol ameliorate kidney injury in STZ-treated rats with regulation of renal NLRP3 inflammasome activation and lipid accumulation. PLoS ONE 2012, 7, e38285. [Google Scholar] [CrossRef] [PubMed]
  99. Jeong, S.M.; Kang, M.J.; Choi, H.N.; Kim, J.H.; Kim, J.I. Quercetin ameliorates hyperglycemia and dyslipidemia and improves antioxidant status in type 2 diabetic db/db mice. Nutr. Res Pract. 2012, 6, 201–207. [Google Scholar] [CrossRef] [PubMed]
  100. Shi, Y.; Williamson, G. Quercetin lowers plasma uric acid in pre-hyperuricaemic males: A randomised, double-blinded, placebo-controlled, cross-over trial. Br. J. Nutr. 2016, 115, 800–806. [Google Scholar] [CrossRef] [PubMed]
  101. Zahedi, M.; Ghiasvand, R.; Feizi, A.; Asgari, G.; Darvish, L. Does quercetin improve cardiovascular risk factors and inflammatory biomarkers in women with type 2 diabetes: A double-blind randomized controlled clinical trial. Int. J. Prev. Med. 2013, 4, 777–785. [Google Scholar] [PubMed]
  102. Brüll, V.; Burak, C.; Stoffel-Wagner, B.; Wolffram, S.; Nickenig, G.; Müller, C.; Langguth, P.; Alteheld, B.; Fimmers, R.; Naaf, S.; et al. Effects of a quercetin-rich onion skin extract on 24 h ambulatory blood pressure and endothelial function in overweight-to-obese patients with (pre-)hypertension: A randomised double-blinded placebo-controlled cross-over trial. Br. J. Nutr. 2015, 114, 1263–1277. [Google Scholar] [CrossRef] [PubMed]
  103. Dower, J.I.; Geleijnse, J.M.; Gijsbers, L.; Zock, P.L.; Kromhout, D.; Hollman, P.C. Effects of the pure flavonoids epicatechin and quercetin on vascular function and cardiometabolic health: A randomized, double-blind, placebo-controlled, crossover trial. Am. J. Clin. Nutr. 2015, 101, 914–921. [Google Scholar] [CrossRef] [PubMed]
  104. Renaud, S.; de Lorgeril, M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet 1992, 339, 1523–1526. [Google Scholar] [CrossRef]
  105. Nishizuka, T.; Fujita, Y.; Sato, Y.; Nakano, A.; Kakino, A.; Ohshima, S.; Kanda, T.; Yoshimoto, R.; Sawamura, T. Procyanidins are potent inhibitors of LOX-1: A new player in the French Paradox. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2011, 87, 104–113. [Google Scholar] [CrossRef] [PubMed]
  106. Bonnefont-Rousselot, D. Resveratrol and cardiovascular diseases. Nutrients 2016, 8, E250. [Google Scholar] [CrossRef] [PubMed]
  107. Lin, Y.L.; Chang, H.C.; Chen, T.L.; Chang, J.H.; Chiu, W.T.; Lin, J.W.; Chen, R.M. Resveratrol protects against oxidized LDL-induced breakage of the blood-brain barrier by lessening disruption of tight junctions and apoptotic insults to mouse cerebrovascular endothelial cells. J. Nutr. 2010, 140, 2187–2192. [Google Scholar] [CrossRef] [PubMed]
  108. Posadino, A.M.; Cossu, A.; Giordo, R.; Zinellu, A.; Sotgia, S.; Vardeu, A.; Hoa, P.T.; Nguyen le, H.V.; Carru, C.; Pintus, G. Resveratrol alters human endothelial cells redox state and causes mitochondrial-dependent cell death. Food Chem. Toxicol. 2015, 78, 10–16. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, H.; Zhang, J.; Ungvari, Z.; Zhang, C. Resveratrol improves endothelial function: Role of TNFα and vascular oxidative stress. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 1164–1171. [Google Scholar] [CrossRef] [PubMed]
  110. Petit, M.; Guihot, A.L.; Grimaud, L.; Vessieres, E.; Toutain, B.; Menet, M.C.; Nivet-Antoine, V.; Arnal, J.F.; Loufrani, L.; Procaccio, V.; et al. Resveratrol improved flow-mediated outward arterial remodeling in ovariectomized rats with hypertrophic effect at high dose. PLoS ONE 2016, 11, e0146148. [Google Scholar] [CrossRef] [PubMed]
  111. Hu, M.; Liu, B. Resveratrol via activation of LKB1-AMPK signaling suppresses oxidative stress to prevent endothelial dysfunction in diabetic mice. Clin. Exp. Hypertens. 2016, 5, 1–7. [Google Scholar] [CrossRef] [PubMed]
  112. Yan, C.; Xu, W.; Huang, Y.; Li, M.; Shen, Y.; You, H.; Liang, X. HRD1-mediated IGF-1R ubiquitination contributes to renal protection of resveratrol in db/db mice. Mol. Endocrinol. 2016. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, Y.; Chen, K.; He, L.; Xia, Y.; Dai, W.; Wang, F.; Li, J.; Li, S.; Liu, T.; Zheng, Y.; et al. The Protective effect of resveratrol on concanavalin-A-induced acute hepatic injury in mice. Gastroenterol. Res. Pract. 2015, 2015, 506390. [Google Scholar] [CrossRef] [PubMed]
  114. Palsamy, P.; Subramanian, S. Ameliorative potential of resveratrol on proinflammatory cytokines, hyperglycemia mediated oxidative stress, and pancreatic β-cell dysfunction in streptozotocin-nicotinamide-induced diabetic rats. J. Cell. Physiol. 2010, 224, 423–432. [Google Scholar] [CrossRef] [PubMed]
  115. Breen, D.M.; Sanli, T.; Giacca, A.; Tsiani, E. Stimulation of muscle cell glucose uptake by resveratrol through sirtuins and AMPK. Biochem. Biophys. Res. Commun. 2008, 374, 117–122. [Google Scholar] [CrossRef] [PubMed]
  116. Borriello, A.; Cucciolla, V.; della Ragione, F.; Galletti, P. Dietary polyphenols: Focus on resveratrol, a promising agent in the prevention of cardiovascular diseases and control of glucose homeostasis. Nutr. Metab. Cardiovasc. Dis. 2010, 20, 618–625. [Google Scholar] [CrossRef] [PubMed]
  117. Patel, M.I.; Gupta, A.; Dey, C.S. Potentiation of neuronal insulin signaling and glucose uptake by resveratrol: The involvement of AMPK. Pharmacol. Rep. 2011, 63, 1162–1168. [Google Scholar] [CrossRef]
  118. Gomez-Zorita, S.; Tréguer, K.; Mercader, J.; Carpéné, C. Resveratrol directly affects in vitro lipolysis and glucose transport in human fat cells. J. Physiol. Biochem. 2013, 69, 585–593. [Google Scholar] [CrossRef] [PubMed]
  119. Szkudelski, T.; Szkudelska, K. Resveratrol and diabetes: From animal to human studies. Biochim. Biophys. Acta 2015, 1852, 1145–1154. [Google Scholar] [CrossRef] [PubMed]
  120. 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. 2007, 6, 307–319. [Google Scholar] [CrossRef] [PubMed]
  121. Deng, J.Y.; Hsieh, P.S.; Huang, J.P.; Lu, L.S.; Hung, L.M. Activation of estrogen receptor is crucial for resveratrol-stimulating muscular glucose uptake via both insulin-dependent and -independent pathways. Diabetes 2008, 57, 1814–1823. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, Q.; Sun, X.; Li, X.; Dong, X.; Li, P.; Zhao, L. Resveratrol attenuates intermittent hypoxia-induced insulin resistance in rats: Involvement of Sirtuin 1 and the phosphatidylinositol-4,5-bisphosphate 3-kinase/AKT pathway. Mol. Med. Rep. 2015, 11, 151–158. [Google Scholar] [CrossRef] [PubMed]
  123. Bagul, P.K.; Deepthi, N.; Sultana, R.; Banerjee, S.K. Resveratrol ameliorates cardiac oxidative stress in diabetes through deacetylation of NFκB-p65 and histone 3. J. Nutr. Biochem. 2015, 26, 1298–1307. [Google Scholar] [CrossRef] [PubMed]
  124. Diaz-Gerevini, G.T.; Repossi, G.; Dain, A.; Tarres, M.C.; Das, U.N.; Eynard, A.R. Beneficial action of resveratrol: How and why? Nutrition 2016, 32, 174–178. [Google Scholar] [CrossRef] [PubMed]
  125. Granados-Soto, V. Pleiotropic effects of resveratrol. Drug News Perspect. 2003, 16, 299–307. [Google Scholar] [CrossRef] [PubMed]
  126. Rodrigo, R.; Miranda, A.; Vergara, L. Modulation of endogenous antioxidant system by wine polyphenols in human disease. Clin. Chim. Acta 2011, 412, 410–424. [Google Scholar] [CrossRef] [PubMed]
  127. Kakoti, B.B.; Hernandez-Ontiveros, D.G.; Kataki, M.S.; Shah, K.; Pathak, Y.; Panguluri, S.K. Resveratrol and Omega-3 Fatty Acid: Its Implications in Cardiovascular Diseases. Front. Cardiovasc. Med. 2015, 11, 38. [Google Scholar] [CrossRef] [PubMed]
  128. Xia, N.; Förstermann, U.; Li, H. Resveratrol and endothelial nitric oxide. Molecules 2014, 19, 16102–16121. [Google Scholar] [CrossRef] [PubMed]
  129. Neves, A.R.; Nunes, C.; Amenitsch, H.; Reis, S. Effects of resveratrol on the structure and fluidity of lipid bilayers: A membrane biophysical study. Soft Matter 2016, 12, 2118–2126. [Google Scholar] [CrossRef] [PubMed]
  130. Marques, F.Z.; Markus, M.A.; Morris, B.J. Resveratrol: Cellular actions of a potent natural chemical that confers a diversity of health benefits. Int. J. Biochem. Cell Biol. 2009, 41, 2125–2128. [Google Scholar] [CrossRef] [PubMed]
  131. Cho, S.J.; Jung, U.J.; Choi, M.S. Differential effects of low-dose resveratrol on adiposity and hepatic steatosis in diet-induced obese mice. Br. J. Nutr. 2012, 108, 2166–2175. [Google Scholar] [CrossRef] [PubMed]
  132. Poulsen, M.M.; Vestergaard, P.F.; Clasen, B.F.; Radko, Y.; Christensen, L.P.; Stødkilde-Jørgensen, H.; Møller, N.; Jessen, N.; Pedersen, S.B.; Jørgensen, J.O. High-dose resveratrol supplementation in obese men: An investigator-initiated, randomized, placebo-controlled clinical trial of substrate metabolism, insulin sensitivity, and body composition. Diabetes 2013, 62, 1186–1195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
  134. la Porte, C.; Voduc, N.; Zhang, G.; Seguin, I.; Tardiff, D.; Singhal, N.; Cameron, D.W. Steady-State pharmacokinetics and tolerability of trans-resveratrol 2000 mg twice daily with food, quercetin and alcohol (ethanol) in healthy human subjects. Clin. Pharmacokinet. 2010, 49, 449–454. [Google Scholar] [CrossRef] [PubMed]
  135. Kapetanovic, I.M.; Muzzio, M.; Huang, Z.; Thompson, T.N.; McCormick, D.L. Pharmacokinetics, oral bioavailability, and metabolic profile of resveratrol and its dimethylether analog, pterostilbene, in rats. Cancer Chemother. Pharmacol. 2011, 68, 593–601. [Google Scholar] [CrossRef] [PubMed]
  136. Malhotra, A.; Bath, S.; Elbarbry, F. An organ system approach to explore the antioxidative, anti-inflammatory, and cytoprotective actions of resveratrol. Oxid. Med. Cell. Longev. 2015, 2015, 803971. [Google Scholar] [CrossRef] [PubMed]
  137. Thazhath, S.S.; Wu, T.; Bound, M.J.; Checklin, H.L.; Standfield, S.; Jones, K.L.; Horowitz, M.; Rayner, C.K. Administration of resveratrol for 5 wk has no effect on glucagon-like peptide 1 secretion, gastric emptying, or glycemic control in type 2 diabetes: A randomized controlled trial. Am. J. Clin. Nutr. 2016, 103, 66–70. [Google Scholar] [CrossRef] [PubMed]
  138. Goh, K.P.; Lee, H.Y.; Lau, D.P.; Supaat, W.; Chan, Y.H.; Koh, A.F. Effects of resveratrol in patients with type 2 diabetes mellitus on skeletal muscle SIRT1 expression and energy expenditure. Int. J. Sport Nutr. Exerc. Metab. 2014, 24, 2–13. [Google Scholar] [CrossRef] [PubMed]
  139. Dash, S.; Xiao, C.; Morgantini, C.; Szeto, L.; Lewis, G.F. High-dose resveratrol treatment for 2 weeks inhibits intestinal and hepatic lipoprotein production in overweight/obese men. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2895–2901. [Google Scholar] [CrossRef] [PubMed]
  140. Tuorkey, M.J.; El-Desouki, N.I.; Kamel, R.A. Cytoprotective effect of silymarin against diabetes-induced cardiomyocyte apoptosis in diabetic rats. Biomed. Environ. Sci. 2015, 28, 36–43. [Google Scholar] [PubMed]
  141. Lee, D.Y.; Liu, Y. Molecular structure and stereochemistry of silybin A, silybin B, isosilybin A, and isosilybin B, Isolated from Silybum marianum (milk thistle). J. Nat. Prod. 2003, 66, 1171–1174. [Google Scholar] [CrossRef] [PubMed]
  142. Anthony, K.P.; Saleh, M.A. Free radical scavenging and antioxidant activities of silymarin components. Antioxidants 2013, 2, 398–407. [Google Scholar] [CrossRef] [PubMed]
  143. Kazazis, C.E.; Evangelopoulos, A.A.; Kollas, A.; Vallianou, N.G. The therapeutic potential of milk thistle in diabetes. Rev. Diabet. Stud. 2014, 11, 167–174. [Google Scholar] [CrossRef] [PubMed]
  144. Wu, C.H.; Huang, S.M.; Yen, G.C. Silymarin: A novel antioxidant with antiglycation and antiinflammatory properties in vitro and in vivo. Antioxid. Redox Signal. 2011, 14, 353–366. [Google Scholar] [CrossRef] [PubMed]
  145. Shin, S.; Lee, J.A.; Kim, M.; Kum, H.; Jung, E.; Park, D. Anti-glycation activities of phenolic constituents from Silybum marianum (Milk Thistle) flower in vitro and on human explants. Molecules 2015, 20, 3549–3564. [Google Scholar] [CrossRef] [PubMed]
  146. Sheela, N.; Jose, M.A.; Sathyamurthy, D.; Kumar, B.N. Effect of silymarin on streptozotocin-nicotinamide-induced type 2 diabetic nephropathy in rats. Iran J. Kidney Dis. 2013, 7, 117–123. [Google Scholar] [PubMed]
  147. Khazim, K.; Gorin, Y.; Cavaglieri, R.C.; Abboud, H.E.; Fanti, P. The antioxidant silybin prevents high glucose-induced oxidative stress and podocyte injury in vitro and in vivo. Am. J. Physiol. Ren. Physiol. 2013, 305, F691–F700. [Google Scholar] [CrossRef] [PubMed]
  148. Vessal, G.; Akmali, M.; Najafi, P.; Moein, M.R.; Sagheb, M.M. Silymarin and milk thistle extract may prevent the progression of diabetic nephropathy in streptozotocin-induced diabetic rats. Ren. Fail. 2010, 32, 733–739. [Google Scholar] [CrossRef] [PubMed]
  149. Baluchnejadmojarad, T.; Roghani, M.; Khastehkhodaie, Z. Chronic treatment of silymarin improves hyperalgesia and motor nerve conduction velocity in diabetic neuropathic rat. Phytother. Res. 2010, 24, 1120–1125. [Google Scholar] [CrossRef] [PubMed]
  150. Marrazzo, G.; Bosco, P.; la Delia, F.; Scapagnini, G.; di Giacomo, C.; Malaguarnera, M.; Galvano, F.; Nicolosi, A.; Li Volti, G. Neuroprotective effect of silibinin in diabetic mice. Neurosci. Lett. 2011, 504, 252–256. [Google Scholar] [CrossRef] [PubMed]
  151. Borah, A.; Paul, R.; Choudhury, S.; Choudhury, A.; Bhuyan, B.; Das Talukdar, A.; Dutta Choudhury, M.; Mohanakumar, K.P. Neuroprotective potential of silymarin against CNS disorders: Insight into the pathways and molecular mechanisms of action. CNS Neurosci. Ther. 2013, 19, 847–853. [Google Scholar] [CrossRef] [PubMed]
  152. Velussi, M.; Cernigoi, A.M.; De Monte, A.; Dapas, F.; Caffau, C.; Zilli, M. Long-term (12 months) treatment with an anti-oxidant drug (silymarin) is effective on hyperinsulinemia, exogenous insulin need and malondialdehyde levels in cirrhotic diabetic patients. J. Hepatol. 1997, 26, 871–879. [Google Scholar] [CrossRef]
  153. Huseini, H.F.; Larijani, B.; Heshmat, R.; Fakhrzadeh, H.; Radjabipour, B.; Toliat, T.; Raza, M. The efficacy of Silybum marianum (L.) Gaertn. (silymarin) in the treatment of type II diabetes: A randomized, double-blind, placebo-controlled, clinical trial. Phytother. Res. 2006, 20, 1036–1039. [Google Scholar] [CrossRef] [PubMed]
  154. Hussain, S.A. Silymarin as an adjunct to glibenclamide therapy improves long-term and postprandial glycemic control and body mass index in type 2 diabetes. J. Med. Food 2007, 10, 543–547. [Google Scholar] [CrossRef] [PubMed]
  155. Ebrahimpour Koujan, S.; Gargari, B.P.; Mobasseri, M.; Valizadeh, H.; Asghari-Jafarabadi, M. Effects of Silybum marianum (L.) Gaertn. (silymarin) extract supplementation on antioxidant status and hs-CRP in patients with type 2 diabetes mellitus: A randomized, triple-blind, placebo-controlled clinical trial. Phytomedicine 2015, 22, 290–296. [Google Scholar] [CrossRef] [PubMed]
  156. Fallahzadeh, M.K.; Dormanesh, B.; Sagheb, M.M.; Roozbeh, J.; Vessal, G.; Pakfetrat, M.; Daneshbod, Y.; Kamali-Sarvestani, E.; Lankarani, K.B. Effect of addition of silymarin to renin-angiotensin system inhibitors on proteinuria in type 2 diabetic patients with overt nephropathy: A randomized, double-blind, placebo-controlled trial. Am. J. Kidney Dis. 2012, 60, 896–903. [Google Scholar] [CrossRef] [PubMed]
  157. Gueutin, V.; Gauthier, M.; Cazenave, M.; Izzedine, H. Diabetic nephropathy: Emerging treatments. Nephrol. Ther. 2014, 10, 210–215. [Google Scholar] [CrossRef] [PubMed]
  158. Magliulo, E.; Gagliardi, B.; Fiori, G.P. Results of a double blind study on the effect of silymarin in the treatment of acute viral hepatitis, carried out at two medical centres. Med. Klin. 1978, 73, 1060–1065. [Google Scholar] [PubMed]
  159. Hollman, P.C.; de Vries, J.H.; van Leeuwen, S.D.; Mengelers, M.J.; Katan, M.B. Absorption of dietary quercetin glycosides and quercetin in healthy ileostomy volunteers. Am. J. Clin. Nutr. 1995, 62, 1276–1282. [Google Scholar] [PubMed]
  160. Boocock, D.J.; Faust, G.E.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J.; et al. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1246–1252. [Google Scholar] [CrossRef] [PubMed]
  161. Hawke, R.L.; Schrieber, S.J.; Soule, T.A.; Wen, Z.; Smith, P.C.; Reddy, K.R.; Wahed, A.S.; Belle, S.H.; Afdhal, N.H.; Navarro, V.J.; et al. Silymarin ascending multiple oral dosing phase I study in noncirrhotic patients with chronic hepatitis C. J. Clin. Pharmacol. 2010, 50, 434–449. [Google Scholar] [CrossRef] [PubMed]
  162. Lecomte, J. Pharmacologic properties of silybin and silymarin. Rev. Med. Liege 1975, 30, 110–114. [Google Scholar] [PubMed]
  163. O'Connor, L.M.; Lentjes, M.A.; Luben, R.N.; Khaw, K.T.; Wareham, N.J.; Forouhi, N.G. Dietary dairy product intake and incident type 2 diabetes: A prospective study using dietary data from a 7-day food diary. Diabetologia 2014, 57, 909–917. [Google Scholar] [CrossRef] [PubMed]
  164. Pasin, G.; Comerford, K.B. Dairy foods and dairy proteins in the management of type 2 diabetes: A systematic review of the clinical evidence. Adv. Nutr. 2015, 6, 245–259. [Google Scholar] [CrossRef] [PubMed]
  165. Astrup, A. Yogurt and dairy product consumption to prevent cardiometabolic diseases: Epidemiologic and experimental studies. Am. J. Clin. Nutr. 2014, 99, 1235S–1242S. [Google Scholar] [CrossRef] [PubMed]
  166. Ejtahed, H.S.; Soroush, A.R.; Angoorani, P.; Larijani, B.; Hasani-Ranjbar, S. Gut microbiota as a target in the pathogenesis of metabolic disorders: A new approach to novel therapeutic agents. Horm. Metab. Res. 2016, 48, 349–358. [Google Scholar] [CrossRef] [PubMed]
  167. Aune, D.; Norat, T.; Romundstad, P.; Vatten, L.J. Dairy products and the risk of type 2 diabetes: A systematic review and dose-response meta-analysis of cohort studies. Am. J. Clin. Nutr. 2013, 98, 1066–1083. [Google Scholar] [CrossRef] [PubMed]
  168. Hadisaputro, S.; Djokomoeljanto, R.R.; Judiono Soesatyo, M.H. The effects of oral plain kefir supplementation on proinflammatory cytokine properties of the hyperglycemia Wistar rats induced by streptozotocin. Acta Med. Indones. 2012, 44, 100–104. [Google Scholar] [PubMed]
  169. Bourrie, B.C.; Willing, B.P.; Cotter, P.D. The microbiota and health promoting characteristics of the fermented beverage kefir. Front. Microbiol. 2016, 7, 647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Magalhães, K.T.; de Melo Pereira, G.V.; Campos, C.R.; Dragone, G.; Schwan, R.F. Brazilian kefir: Structure, microbial communities and chemical composition. Braz. J. Microbiol. 2011, 42, 693–702. [Google Scholar] [CrossRef] [PubMed]
  171. De Oliveira Leite, A.M.; Miguel, M.A.; Peixoto, R.S.; Rosado, A.S.; Silva, J.T.; Paschoalin, V.M. Microbiological, technological and therapeutic properties of kefir: A natural probiotic beverage. Braz. J. Microbiol. 2013, 44, 341–349. [Google Scholar] [CrossRef] [PubMed]
  172. Friques, A.G.; Arpini, C.M.; Kalil, I.C.; Gava, A.L.; Leal, M.A.; Porto, M.L.; Nogueira, B.V.; Dias, A.T.; Andrade, T.U.; Pereira, T.M.; et al. Chronic administration of the probiotic kefir improves the endothelial function in spontaneously hypertensive rats. J. Transl. Med. 2015, 13, 390. [Google Scholar] [CrossRef] [PubMed]
  173. Zhou, J.; Liu, X.; Jiang, H.; Dong, M. Analysis of the microflora in Tibetan kefir grains using denaturing gradient gel electrophoresis. Food Microbiol. 2009, 26, 770–775. [Google Scholar] [CrossRef] [PubMed]
  174. Leite, A.M.; Mayo, B.; Rachid, C.T.; Peixoto, R.S.; Silva, J.T.; Paschoalin, V.M.; Delgado, S. Assessment of the microbial diversity of Brazilian kefir grains by PCR-DGGE and pyrosequencing analysis. Food Microbiol. 2012, 31, 215–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Hamet, M.F.; Londero, A.; Medrano, M.; Vercammen, E.; van Hoorde, K.; Garrote, G.L.; Huys, G.; Vandamme, P.; Abraham, A.G. Application of culture-dependent and culture-independent methods for the identification of Lactobacillus kefiranofaciens in microbial consortia present in kefir grains. Food Microbiol. 2013, 36, 327–334. [Google Scholar] [CrossRef] [PubMed]
  176. Marsh, A.J.; O’Sullivan, O.; Hill, C.; Ross, R.P.; Cotter, P.D. Sequencing-based analysis of the bacterial and fungal composition of kefir grains and milks from multiple sources. PLoS ONE 2013, 8, e69371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Gonçalves, N.; Gomes-Ferreira, C.; Moura, C.; Roncon-Albuquerque, R., Jr.; Leite-Moreira, A.F.; Falcão-Pires, I. Worse cardiac remodeling in response to pressure overload in type 2 diabetes mellitus. Int. J. Cardiol. 2016, 217, 195–204. [Google Scholar] [CrossRef] [PubMed]
  178. Punaro, G.R.; Maciel, F.R.; Rodrigues, A.M.; Rogero, M.M.; Bogsan, C.S.; Oliveira, M.N.; Ihara, S.S.; Araujo, S.R.; Sanches, T.R.; Andrade, L.C.; et al. Kefir administration reduced progression of renal injury in STZ-diabetic rats by lowering oxidative stress. Nitric Oxide 2014, 37, 53–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Yadav, H.; Jain, S.; Sinha, P.R. Oral administration of dahi containing probiotic Lactobacillus acidophilus and Lactobacillus casei delayed the progression of streptozotocin-induced diabetes in rats. J. Dairy Res. 2008, 75, 189–195. [Google Scholar] [CrossRef] [PubMed]
  180. Maeda, H.; Zhu, X.; Omura, K.; Suzuki, S.; Kitamura, S. Effects of an exopolysaccharide (kefiran) on lipids, blood pressure, blood glucose, and constipation. Biofactors 2004, 22, 197–200. [Google Scholar] [CrossRef] [PubMed]
  181. Maeda, H.; Zhu, X.; Suzuki, S.; Suzuki, K.; Kitamura, S. Structural characterization and biological activities of an exopolysaccharide kefiran produced by Lactobacillus kefiranofaciens WT-2B(T). J. Agric. Food Chem. 2004, 52, 5533–5338. [Google Scholar] [CrossRef] [PubMed]
  182. Teruya, K.; Yamashita, M.; Tominaga, R.; Nagira, T.; Shim, S.Y.; Katakura, Y.; Tokumaru, S.; Tokumaru, K.; Barnes, D.; Shirahata, S. Fermented milk, Kefram-Kefir enhances glucose uptake into insulin-responsive muscle cells. Cytotechnology 2002, 40, 107–116. [Google Scholar] [CrossRef] [PubMed]
  183. Yadav, H.; Jain, S.; Sinha, P.R. Effect of Dahi containing Lactococcus lactis on the progression of diabetes induced by a high-fructose diet in rats. Biosci. Biotechnol. Biochem. 2006, 70, 1255–1258. [Google Scholar] [CrossRef] [PubMed]
  184. Asemi, Z.; Zare, Z.; Shakeri, H.; Sabihi, S.S.; Esmaillzadeh, A. Effect of multispecies probiotic supplements on metabolic profiles, hs-CRP, and oxidative stress in patients with type 2 diabetes. Ann. Nutr. Metab. 2013. [Google Scholar] [CrossRef] [PubMed]
  185. Rudich, A.; Kozlovsky, N.; Potashnik, R.; Bashan, N. Oxidant stress reduces insulin responsiveness in 3T3-L1 adipocytes. Am. J. Physiol. 1997, 272, E935–E940. [Google Scholar] [PubMed]
  186. Rudich, A.; Tirosh, A.; Potashnik, R.; Hemi, R.; Kanety, H.; Bashan, N. Prolonged oxidative stress impairs insulin-induced GLUT4 translocation in 3T3-L1 adipocytes. Diabetes 1998, 47, 1562–1569. [Google Scholar] [CrossRef] [PubMed]
  187. Baynes, J.W. Role of oxidative stress in development of complications in diabetes. Diabetes 1991, 40, 405–412. [Google Scholar] [CrossRef] [PubMed]
  188. Wolff, S.P.; Jiang, Z.Y.; Hunt, J.V. Protein glycation and oxidative stress in diabetes mellitus and ageing. Free Radic. Biol. Med. 1991, 10, 339–352. [Google Scholar] [CrossRef]
  189. Klippel, B.F.; Duemke, L.B.; Leal, M.A.; Friques, A.G.F.; Dantas, E.M.; Dalvi, R.F.; Gava, A.L.; Pereira, T.M.C.; Andrade, T.U.; Meyrelles, S.S.; et al. Effects of kefir on the cardiac autonomic tones and baroreflex sensitivity in spontaneously hypertensive rats. Front. Physiol. 2016, 7, 211. [Google Scholar] [CrossRef] [PubMed]
  190. Liu, Y.; Liu, S.X.; Zheng, F.; Cai, Y.; Xie, K.L.; Zhang, W.L. Cardiovascular autonomic neuropathy in patients with type 2 diabetes. J. Diabetes Investig. 2015. [Google Scholar] [CrossRef] [PubMed]
  191. Li, X.; Jiang, Y.H.; Jiang, P.; Lin, H.Q.; Yang, J.L.; Ma, D.F.; Wang, X.; Yang, C.H. Analysis of heart rate variability and cardiac autonomic nerve remodeling in streptozotocin-induced diabetic rats. Exp. Clin. Endocrinol. Diabetes 2015, 123, 272–281. [Google Scholar] [CrossRef] [PubMed]
  192. Firouzi, S.; Majid, H.A.; Ismail, A.; Kamaruddin, N.A.; Barakatun-Nisak, M.Y. Effect of multi-strain probiotics (multi-strain microbial cell preparation) on glycemic control and other diabetes-related outcomes in people with type 2 diabetes: A randomized controlled trial. Eur. J. Nutr. 2016. [Google Scholar] [CrossRef] [PubMed]
  193. Ostadrahimi, A.; Taghizadeh, A.; Mobasseri, M.; Farrin, N.; Payahoo, L.; Beyramalipoor Gheshlaghi, Z.; Vahedjabbari, M. Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: A randomized double-blind placebo-controlled clinical trial. Iran J. Public Health 2015, 44, 228–237. [Google Scholar] [PubMed]
  194. Hulston, C.J.; Churnside, A.A.; Venables, M.C. Probiotic supplementation prevents high-fat, overfeeding-induced insulin resistance in human subjects. Br. J. Nutr. 2015, 113, 596–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Tonucci, L.B.; Olbrich Dos Santos, K.M.; Licursi de Oliveira, L.; Rocha Ribeiro, S.M.; Duarte Martino, H.S. Clinical application of probiotics in type 2 diabetes mellitus: A randomized, double-blind, placebo-controlled study. Clin. Nutr. 2015. [Google Scholar] [CrossRef] [PubMed]
  196. Manaer, T.; Yu, L.; Zhang, Y.; Xiao, X.J.; Nabi, X.H. Anti-diabetic effects of shubat in type 2 diabetic rats induced by combination of high-glucose-fat diet and low-dose streptozotocin. J. Ethnopharmacol. 2015, 169, 269–274. [Google Scholar] [CrossRef] [PubMed]
  197. Kuschnerus, K.; Landmesser, U.; Kränkel, N. Vascular repair strategies in type 2 diabetes: Novel insights. Cardiovasc. Diagn. Ther. 2015, 5, 374–386. [Google Scholar] [PubMed]
  198. Ramirez, C.E.; Nian, H.; Yu, C.; Gamboa, J.L.; Luther, J.M.; Brown, N.J.; Shibao, C.A. Treatment with sildenafil improves insulin sensitivity in prediabetes: A randomized, controlled trial. J. Clin. Endocrinol. Metab. 2015, 100, 4533–4540. [Google Scholar] [CrossRef] [PubMed]
  199. Gromada, J.; Høy, M.; Renström, E.; Bokvist, K.; Eliasson, L.; Göpel, S.; Rorsman, P. CaM kinase II-dependent mobilization of secretory granules underlies acetylcholine-induced stimulation of exocytosis in mouse pancreatic B-cells. J. Physiol. 1999, 518, 745–759. [Google Scholar] [CrossRef] [PubMed]
  200. Fu, Z.; Gilbert, E.R.; Liu, D. Regulation of insulin synthesis and secretion and pancreatic β-cell dysfunction in diabetes. Curr. Diabetes Rev. 2013, 9, 25–53. [Google Scholar] [CrossRef] [PubMed]
  201. Nyström, T.; Ortsäter, H.; Huang, Z.; Zhang, F.; Larsen, F.J.; Weitzberg, E.; Lundberg, J.O.; Sjöholm, Å. Inorganic nitrite stimulates pancreatic islet blood flow and insulin secretion. Free Radic. Biol. Med. 2012, 53, 1017–1023. [Google Scholar] [CrossRef] [PubMed]
  202. Méndez-del Villar, M.; González-Ortiz, M.; Martínez-Abundis, E.; Pérez-Rubio, K.G.; Lizárraga-Valdez, R. Effect of resveratrol administration on metabolic syndrome, insulin sensitivity, and insulin secretion. Metab. Syndr. Relat. Disord. 2014, 12, 497–501. [Google Scholar] [CrossRef] [PubMed]
  203. Otunola, G.A.; Oloyede, O.B.; Oladiji, A.T.; Afolayan, A.J. Selected spices and their combination modulate hypercholesterolemia-induced oxidative stress in experimental rats. Biol. Res. 2014, 47, 5. [Google Scholar] [PubMed]
  204. Bergandi, L.; Silvagno, F.; Russo, I.; Riganti, C.; Anfossi, G.; Aldieri, E.; Ghigo, D.; Trovati, M.; Bosia, A. Insulin stimulates glucose transport via nitric oxide/cyclic GMP pathway in human vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 2215–2221. [Google Scholar] [CrossRef] [PubMed]
  205. Tanaka, T.; Nakatani, K.; Morioka, K.; Urakawa, H.; Maruyama, N.; Kitagawa, N.; Katsuki, A.; Araki-Sasaki, R.; Hori, Y.; Gabazza, E.C.; et al. Nitric oxide stimulates glucose transport through insulin-independent GLUT4 translocation in 3T3-L1 adipocytes. Eur. J. Endocrinol. 2003, 149, 61–67. [Google Scholar] [CrossRef] [PubMed]
  206. Lira, V.A.; Soltow, Q.A.; Long, J.H.; Betters, J.L.; Sellman, J.E.; Criswell, D.S. Nitric oxide increases GLUT4 expression and regulates AMPK signaling in skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 2007, 293, E1062–E1068. [Google Scholar] [CrossRef] [PubMed]
  207. Furchgott, R.F.; Zawadzki, J.V. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 1980, 288, 373–376. [Google Scholar] [CrossRef] [PubMed]
  208. Ignarro, L.J. Nitric oxide: A unique endogenous signaling molecule in vascular biology. Biosci. Rep. 1999, 19, 51–71. [Google Scholar] [CrossRef] [PubMed]
  209. Vasquez, E.C.; Gava, A.L.; Graceli, J.B.; Balarini, C.M.; Campagnaro, B.P.; Pereira, T.M.; Meyrelles, S.S. Novel therapeutic targets for phosphodiesterase 5 inhibitors: Current state-of-the-art on systemic arterial hypertension and atherosclerosis. Curr. Pharm. Biotechnol. 2016, 17, 347–364. [Google Scholar] [CrossRef] [PubMed]
  210. Behr-Roussel, D.; Oudot, A.; Caisey, S.; Coz, O.L.; Gorny, D.; Bernabé, J.; Wayman, C.; Alexandre, L.; Giuliano, F.A. Daily treatment with sildenafil reverses endothelial dysfunction and oxidative stress in an animal model of insulin resistance. Eur. Urol. 2008, 53, 1272–1280. [Google Scholar] [CrossRef] [PubMed]
  211. Maiorino, M.I.; Bellastella, G.; Esposito, K. Diabetes and sexual dysfunction: Current perspectives. Diabetes Metab. Syndr. Obes. 2014, 7, 95–105. [Google Scholar] [PubMed]
  212. Leal, M.A.; Balarini, C.M.; Dias, A.T.; Porto, M.L.; Gava, A.L.; Pereira, T.M.; Meyrelles, S.S.; Vasquez, E.C. Mechanisms of enhanced vasoconstriction in the mouse model of atherosclerosis: The beneficial effects of sildenafil. Curr. Pharm. Biotechnol. 2015, 16, 517–530. [Google Scholar] [CrossRef] [PubMed]
  213. Bai, Y.; An, R. Resveratrol and sildenafil synergistically improve diabetes-associated erectile dysfunction in streptozotocin-induced diabetic rats. Life Sci. 2015, 135, 43–48. [Google Scholar] [CrossRef] [PubMed]
  214. Dalaklioglu, S.; Bayram, Z.; Tasatargil, A.; Ozdem, S. Resveratrol reverses diabetes-related decrement in sildenafil-induced relaxation of corpus cavernosum in aged rats. Aging Clin. Exp. Res. 2016. [Google Scholar] [CrossRef] [PubMed]
  215. Bivalacqua, T.J.; Sussan, T.E.; Gebska, M.A.; Strong, T.D.; Berkowitz, D.E.; Biswal, S.; Burnett, A.L.; Champion, H.C. Sildenafil inhibits superoxide formation and prevents endothelial dysfunction in a mouse model of secondhand smoke induced erectile dysfunction. J. Urol. 2009, 181, 899–906. [Google Scholar] [CrossRef] [PubMed]
  216. Balarini, C.M.; Leal, M.A.; Gomes, I.B.; Pereira, T.M.; Gava, A.L.; Meyrelles, S.S.; Vasquez, E.C. Sildenafil restores endothelial function in the apolipoprotein E knockout mouse. J. Transl. Med. 2013, 11, 3. [Google Scholar] [CrossRef] [PubMed]
  217. Dias, A.T.; Cintra, A.S.; Frossard, J.C.; Palomino, Z.; Casarini, D.E.; Gomes, I.B.; Balarini, C.M.; Gava, A.L.; Campagnaro, B.P.; Pereira, T.M.; et al. Inhibition of phosphodiesterase 5 restores endothelial function in renovascular hypertension. J. Transl. Med. 2014, 12, 250. [Google Scholar] [CrossRef] [PubMed]
  218. Fahning, B.M.; Dias, A.T.; Oliveira, J.P.; Gava, A.L.; Porto, M.L.; Gomes, I.B.; Nogueira, B.V.; Campagnaro, B.P.; Pereira, T.M.; Vasquez, E.C.; et al. Sildenafil improves vascular endothelial structure and function in renovascular hypertension. Curr. Pharm. Biotechnol. 2015, 16, 823–831. [Google Scholar] [CrossRef] [PubMed]
  219. de Almeida, L.S.; Barboza, J.R.; Freitas, F.P.; Porto, M.L.; Vasquez, E.C.; Meyrelles, S.S.; Gava, A.L.; Pereira, T.M. Sildenafil prevents renal dysfunction in contrast media-induced nephropathy in Wistar rats. Hum. Exp. Toxicol. 2016. [Google Scholar] [CrossRef] [PubMed]
  220. Aversa, A.; Vitale, C.; Volterrani, M.; Fabbri, A.; Spera, G.; Fini, M.; Rosano, G.M. Chronic administration of Sildenafil improves markers of endothelial function in men with Type 2 diabetes. Diabet Med. 2008, 25, 37–44. [Google Scholar] [CrossRef] [PubMed]
  221. Kondoh, N.; Higuchi, Y.; Maruyama, T.; Nojima, M.; Yamamoto, S.; Shima, H. Salvage therapy trial for erectile dysfunction using phosphodiesterase type 5 inhibitors and vitamin E: Preliminary report. Aging Male 2008, 11, 167–170. [Google Scholar] [CrossRef] [PubMed]
  222. Mandosi, E.; Giannetta, E.; Filardi, T.; Lococo, M.; Bertolini, C.; Fallarino, M.; Gianfrilli, D.; Venneri, M.A.; Lenti, L.; Lenzi, A.; et al. Endothelial dysfunction markers as a therapeutic target for Sildenafil treatment and effects on metabolic control in type 2 diabetes. Expert Opin. Ther. Targets 2015, 19, 1617–1622. [Google Scholar] [CrossRef] [PubMed]
  223. Balhara, Y.P.; Sarkar, S.; Gupta, R. Phosphodiesterase-5 inhibitors for erectile dysfunction in patients with diabetes mellitus: A systematic review and meta-analysis of randomized controlled trials. Indian J. Endocrinol. Metab. 2015, 19, 451–461. [Google Scholar] [CrossRef] [PubMed]
  224. Das, A.; Durrant, D.; Salloum, F.N.; Xi, L.; Kukreja, R.C. PDE5 inhibitors as therapeutics for heart disease, diabetes and cancer. Pharmacol. Ther. 2015, 147, 12–21. [Google Scholar] [CrossRef] [PubMed]
  225. Mammi, C.; Pastore, D.; Lombardo, M.F.; Ferrelli, F.; Caprio, M.; Consoli, C.; Tesauro, M.; Gatta, L.; Fini, M.; Federici, M.; et al. Sildenafil reduces insulin-resistance in human endothelial cells. PLoS ONE 2011, 6, e14542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Ayala, J.E.; Bracy, D.P.; Julien, B.M.; Rottman, J.N.; Fueger, P.T.; Wasserman, D.H. Chronic treatment with sildenafil improves energy balance and insulin action in high fat-fed conscious mice. Diabetes 2007, 56, 1025–1033. [Google Scholar] [CrossRef] [PubMed]
  227. Varma, A.; Das, A.; Hoke, N.N.; Durrant, D.E.; Salloum, F.N.; Kukreja, R.C. Anti-inflammatory and cardioprotective effects of tadalafil in diabetic mice. PLoS ONE 2012, 7, e45243. [Google Scholar] [CrossRef] [PubMed]
  228. Koka, S.; Das, A.; Salloum, F.N.; Kukreja, R.C. Phosphodiesterase-5 inhibitor tadalafil attenuates oxidative stress and protects against myocardial ischemia/reperfusion injury in type 2 diabetic mice. Free Radic. Biol. Med. 2013, 60, 80–88. [Google Scholar] [CrossRef] [PubMed]
  229. Grover-Páez, F.; Villegas Rivera, G.; Guillén Ortíz, R. Sildenafil citrate diminishes microalbuminuria and the percentage of A1c in male patients with type 2 diabetes. Diabetes Res. Clin. Pract. 2007, 78, 136–140. [Google Scholar] [CrossRef] [PubMed]
  230. Chrysant, S.G.; Chrysant, G.S. The pleiotropic effects of phosphodiesterase 5 inhibitors on function and safety in patients with cardiovascular disease and hypertension. J. Clin. Hypertens. 2012, 14, 644–649. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of metabolic complications of diabetes mellitus in two important target organs, and the main effects exhibited by three important coadjuvants currently under investigation with the aim of preventing and treating this complex disease. Arrows up (↑): increase; arrows down (↓): decrease.
Figure 1. Schematic representation of metabolic complications of diabetes mellitus in two important target organs, and the main effects exhibited by three important coadjuvants currently under investigation with the aim of preventing and treating this complex disease. Arrows up (↑): increase; arrows down (↓): decrease.
Ijms 17 01273 g001

Share and Cite

MDPI and ACS Style

Pereira, T.M.C.; Pimenta, F.S.; Porto, M.L.; Baldo, M.P.; Campagnaro, B.P.; Gava, A.L.; Meyrelles, S.S.; Vasquez, E.C. Coadjuvants in the Diabetic Complications: Nutraceuticals and Drugs with Pleiotropic Effects. Int. J. Mol. Sci. 2016, 17, 1273. https://doi.org/10.3390/ijms17081273

AMA Style

Pereira TMC, Pimenta FS, Porto ML, Baldo MP, Campagnaro BP, Gava AL, Meyrelles SS, Vasquez EC. Coadjuvants in the Diabetic Complications: Nutraceuticals and Drugs with Pleiotropic Effects. International Journal of Molecular Sciences. 2016; 17(8):1273. https://doi.org/10.3390/ijms17081273

Chicago/Turabian Style

Pereira, Thiago Melo Costa, Fabio Silva Pimenta, Marcella Lima Porto, Marcelo Perim Baldo, Bianca Prandi Campagnaro, Agata Lages Gava, Silvana Santos Meyrelles, and Elisardo Corral Vasquez. 2016. "Coadjuvants in the Diabetic Complications: Nutraceuticals and Drugs with Pleiotropic Effects" International Journal of Molecular Sciences 17, no. 8: 1273. https://doi.org/10.3390/ijms17081273

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop