Next Article in Journal
Genistein Promotes Anti-Heat Stress and Antioxidant Effects via the Coordinated Regulation of IIS, HSP, MAPK, DR, and Mitochondrial Pathways in Caenorhabditis elegans
Next Article in Special Issue
Potential Benefits of Antioxidant Phytochemicals on Endogenous Antioxidants Defences in Chronic Diseases
Previous Article in Journal
Intermediate-Term Storage of Spotted Halibut (Verasper variegatus) Sperm: Effects of Storage Methods, Extenders Supplemented with Antibiotics and Antioxidants on Sperm Quality
Previous Article in Special Issue
Effects of Phenolic-Rich Pinus densiflora Extract on Learning, Memory, and Hippocampal Long-Term Potentiation in Scopolamine-Induced Amnesic Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications

1
School of Basic Sciences, All American Institute of Medical Sciences, Black River, Saint Elizabeth Parish JMDEH03, Jamaica
2
Department of Biochemistry, Michael Okpara University of Agriculture, Umudike 440109, Nigeria
3
Department of Ophthalmology and Visual Sciences, The University of British Columbia, Vancouver, BC V5Z 0A6, Canada
*
Authors to whom correspondence should be addressed.
Antioxidants 2023, 12(1), 123; https://doi.org/10.3390/antiox12010123
Submission received: 30 October 2022 / Revised: 10 December 2022 / Accepted: 21 December 2022 / Published: 4 January 2023

Abstract

:
The global prevalence of diabetes continues to increase partly due to rapid urbanization and an increase in the aging population. Consequently, this is associated with a parallel increase in the prevalence of diabetic vascular complications which significantly worsen the burden of diabetes. For these diabetic vascular complications, there is still an unmet need for safe and effective alternative/adjuvant therapeutic interventions. There is also an increasing urge for therapeutic options to come from natural products such as plants. Hyperglycemia-induced oxidative stress is central to the development of diabetes and diabetic complications. Furthermore, oxidative stress-induced inflammation and insulin resistance are central to endothelial damage and the progression of diabetic complications. Human and animal studies have shown that polyphenols could reduce oxidative stress, hyperglycemia, and prevent diabetic complications including diabetic retinopathy, diabetic nephropathy, and diabetic peripheral neuropathy. Part of the therapeutic effects of polyphenols is attributed to their modulatory effect on endogenous antioxidant systems. This review attempts to summarize the established effects of polyphenols on endogenous antioxidant systems from the literature. Moreover, potential therapeutic strategies for harnessing the potential benefits of polyphenols for diabetic vascular complications are also discussed.

1. Introduction

The change in nutrition habits due to rapid urbanization and an increase in the ageing population have contributed to an increase in the global prevalence of diabetes [1,2]. Consequently, diabetes is now a growing health concern, as the population of adults (aged 20 to 79) living with diabetes was estimated to increase from 463 to 536.6 million people between 2019 and 2021. An increment of a further 46%, to an average of 783.2 million diabetic adults, has been estimated to occur up to 2045 [3]. The latter increased level is projected to be significantly higher in middle-income countries compared to underdeveloped and developed nations [3].
Diabetes is the root cause of several macrovascular and microvascular complications. Macrovascular complications associated with diabetes include atherosclerosis and stroke; and microvascular complications include diabetic retinopathy, nephropathy, and neuropathy. Generally, diabetes vascular complications effectuate the majority of the burden of diabetes. The characteristics of diabetic complications may vary from context to context—in high-income countries, recent studies suggest a decline in hospitalization and death toll among older people with severe diabetic complications in the last 30 years [2,4]. However, in low- and middle-income countries (LMIC) such as Brazil, Ghana, Columbia, and China, and also among young adults, an increase in the prevalence of severe diabetic complications was observed [2]. This trend in LMIC was associated with healthcare deficiencies [2].
In this respect, especially of LMIC, a parallel buildup of diabetes vascular complications is set to have huge economic implications for these countries and the patients living with diabetes in these regions [1,2]. For instance, in North America and Caribbean (NAC) region, the cost of care for diabetes-related health conditions was estimated to be USD 414.5 billion. In contrast, South East Asian countries such as Cambodia, Myanmar which are LMIC, have double the population of diabetic patients compared to NAC regions and were estimated to spend USD 10.1 billion in 2021 [3], and in 2016 the cost of diabetes care in Africa was evaluated to be over USD 4 billion per annum [1]. Moreover, health expenditure increases parallelly by diabetes complications [1]. Furthermore, as a result of the financial burden and physical disabilities associated with diabetic complications, they significantly reduce the quality of life of those living with them [5]. In light of the growth of diabetes and the associated socio-economic impact of diabetes complications, interventions targeting improved therapy and lifestyle of diabetic patients are urgently needed.
Phytochemicals are secondary chemical metabolites that occur naturally in fruits, vegetables, and herbs that are known to possess different structures and can promote health benefits in humans [6]. Phytochemicals are critical in plant growth, development, and protection, including protection from harmful insects and microorganisms as well as extreme temperature and ultraviolet light exposure [7].
Phytochemicals are categorized principally based on their structures as alkaloids, flavonoids, carotenoids, phenolics, terpenoids, steroids, and essential oils [8,9,10]. Several phytochemicals are recognized as the bioactive components of commonly available medicines, examples including salicylates sourced from willow bark which is present in aspirin which is used as pain and fever relief [11,12], quinine found in cinchona bark and used to treat malaria [13]. The largest group of phytochemicals in plants are the polyphenols (e.g., flavanones, flavonoids) known for their free radical scavenging activity. Free radical scavenging activity of flavonoids, for example, is dependent on the number and position of hydroxyl groups on aromatic rings and a C ring with a double bond conjugated with a 4-oxo function, which confers on them their reducing capacity [14,15].
While phytochemicals provide the surest and quickest pathway to the discovery and development of new drugs, they also have some drawbacks which continue to restrict their overall acceptability. Challenges include the lack of a standardized dosage regimen, the preparation, drug–herb interactions, and low bioavailability, for example, clearance of oral contraceptives is increased by co-ingestion with St John’s wort [16]. Therefore, research into ways to maximize phytochemicals for therapy is required.
The purpose of the current review is to discuss the body of literature on diabetic micro-and macrovascular complications with a focus on: (1) the roles of oxidative stress (2) the activation of endogenous antioxidant defenses by exogenous phytochemicals, and (3) the therapeutic potentials of phytochemicals as alternative/adjuvant options.

2. Diabetic Complications

Due to the progressive nature of diabetes, it eventually leads to complications which are mainly microvascular and macrovascular. These complications have intense consequences on the anatomy, structure, and function of several cells, tissues, and organs, and consequently on the patient’s well-being status [17].

2.1. Diabetic Retinopathy (DR)

DR is the most common microvascular complication of diabetes that can affect the retinal metabolism, physiology, and microvasculature of the eyes, it is also determined recently as the prominent cause of blindness in the working population [18]. The impact of DR on normal vision varies broadly depending on the stage of DR and the earliest stage of DR may be asymptomatic [19,20,21,22]. General symptoms of DR which usually develop gradually could include temporarily blurred vision, floaters, and flashes of light or blind spots in the field of vision, and gradual or sudden loss of vision [23,24].
Generally, DR is classically categorized into two major classes: non-proliferative DR (NPDR) and proliferative DR (PDR). NPDR symptoms are defects of the retinal vasculature including hemorrhages such as dot hemorrhages and blot hemorrhages [25,26], hard exudation [26,27], cotton wool spots [25,26,27,28], microaneurysms [25,26,27], and vascular leakage [27,28]. The major PDR hallmarks are related to pathological retinal angiogenesis which involves the growth of new abnormal blood vessels from the pre-existing vascular network, and pathological retinal neovascularization which encompasses the development of new blood vessels by de novo formation (vasculogenesis) and angiogenesis [26,29,30,31,32,33]. The progression from NPDR to PDR is characterized by ischemia, hypoxia, and the increased expression of proangiogenic growth factors including vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2), platelet-derived growth factor (PDGF), and angiopoietin-2 (Ang-2) which activate the growth of aberrant retina blood vessels which can protrude into the preretinal space [30,31,34].
Apart from the clinical features already discussed, other hallmark pathological changes in DR include pericyte loss, microglial activation, modifications to macroglial functions (Müller cells and astrocytes), thickening of the basement membrane, retinal leukocyte adhesion, blood–retinal barrier (BRB) breakdown, leukostasis, capillary nonperfusion, and capillary endothelial cell injury and death [25,26,27]; and degeneration of retinal neurons [35,36].
Prevailing recommended treatments for DR are laser pan-retinal photocoagulation (PRP) [37,38], vitreoretinal surgery [27], and anti-VEGF intravitreal injections [39,40]. Although the aforesaid treatments may slow down the progression of DR towards blindness, they are not effective in tackling every instance of the disease and have significant side effects [38,41,42,43,44,45]; therefore, it becomes imperative to find exigent alternatives or adjuvant therapeutic options to prevent DR or slow down its progression towards blindness.

2.2. Diabetic Peripheral Neuropathy (DPN)

DPN is a microvascular diabetic complication that is characterized by peripheral nerve dysfunction. DPN is a significant cause of morbidity in diabetic patients and about half of all diabetic individuals suffer from this complication [46]. In the clinic, the manifestations of DPN include painful neuropathic sensations and insensitivity, the latter significantly increases the risk of unintentional injuries, burns, and foot ulcers, which potentially leads to non-traumatic amputation and a significant reduction in the health status of patients [47,48]. Once DPN initiates, it would be irreversible, but progression can be slowed down [49].
The Toronto Diabetic Neuropathy Expert group categorized DPN into three major classes: (I) possible DPN, wherein any of the following signs or symptoms may be present: symptoms–decreased sensation, positive neuropathic sensory symptoms (e.g., “asleep numbness”, prickling or stabbing, burning or aching pain) that predominantly occur in the toes, feet, or legs, signs—a symmetric decrease in distal sensation or unequivocally decreased or absent ankle reflexes; (II) probable DPN, wherein two or more of the following signs and symptoms of neuropathy are present: neuropathic symptoms, decreased distal sensation, or unequivocally decreased or absent ankle reflexes; and (III) confirmed DPN, wherein nerve conduction is anomalous and a sign or symptom of neuropathy is present [47].
DPN is currently managed by tight glycemic control focusing on reducing HbA1c, however, for many patients, it is challenging to accomplish glycemic control [48]. The pain symptoms of DPN are managed with pharmacological agents including anticonvulsants (e.g., pregabalin and gabapentin) as the first-line pain therapy, serotonin–norepinephrine reuptake inhibitors (e.g., duloxetine and venlafaxine), secondary amine tricyclic antidepressants (e.g., nortriptyline and desipramine) and opioid agonists (e.g., oxycodone and methadone) [48,49].

2.3. Diabetic Nephropathy (DN)

DN is another microvascular diabetic complication that is the global leading cause of end-stage renal disease (ESRD) and dialysis, constituting about 40% of total patients who need renal replacement therapy [50,51]. The mortality rate is about 30 times higher in DN patients compared with other diabetic patients without DN [52]. Glomerular basement membrane (GBM) thickening, mesangial matrix expansion, proteinuria (classically manifested as albuminuria), the development of characteristic Kimmelstiel–Wilson nodules, and progressive regression in glomerular filtration rate (GFR) are characteristic features of DN [53,54].
DN is classically categorized into five stages based on urinary albumin excretion (UAE), GFR, and blood pressure (BP) [52,54]: (1) glomerular hyperfiltration wherein, GFR is normal “>90 mL/min/1.73 m2” or increased, UAE value is <30 mg/day and BP is normotensive; (2) silent stage, wherein, GFR is normal. UAE rate is <30 mg/day and BP is ±hypertensive along with thickened basement membrane; (3) incipient nephropathy with GFR of <60 mL/min/1.73 m2, microalbuminuria (UAE of 30–300 mg/day; the earliest clinically detectable sign), and BP of ±hypertensive; (4) overt nephropathy, wherein, GFR is <30 mL/min/1.73 m2, macroalbuminuria (UAE of >300 mg/day) and hypertensive BP are present; and ultimately, (5) ESRD, wherein, GFR value is <15 mL/min/1.73 m2, with conditions of hypertensive BP and macroalbuminuria in which usually dialysis or transplantation is required [54].
Currently, tight glycemic and BP control, and the inhibition of the renin–angiotensin-aldosterone system (RAAS) via angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers are the central approaches of DN therapy [52,54]. However, these management approaches have limitations in preventing DN progression towards ESRD, hence, effective therapies are urgently needed [54].

2.4. Diabetes-Induced Cardiovascular and Cerebrovascular Diseases

Diabetics are at a greater risk of both cardiovascular and cerebrovascular diseases including atherosclerosis, cardiomyopathy, and stroke, which significantly increase morbidity and mortality in these patients [55,56,57,58].
Atherosclerosis is a macrovascular disease that is the primary cause of various heart diseases and stroke; diabetes-induced atherosclerosis is a significant cause of morbidity amongst diabetics [56,59]. It manifests as intimal thickening, inflammation, and narrowing of arteries by the build-up of plaques [59]. The Insulin Resistance Atherosclerosis (IRAS) study [56] has shown that compared with non-diabetics, diabetics have an increased rate of progression of carotid atherosclerosis, with diabetics having twice the mean progression of intimal-medial thickness in both the common carotid artery and the internal carotid artery.
Diabetic cardiomyopathy is the diabetes-induced functional and structural change of the myocardium in the absence of other risk factors including hypertension, coronary artery disease, and significant valvular disease [55]. The lack of specific standardized guidelines governing both the diagnosis and treatment of diabetic cardiomyopathy makes a definitive diagnosis, and treatment problematic [60]. Clinical pathological changes include cardiac remodeling, diastolic dysfunction, myocardial fibrosis, cardiac stiffness, impaired calcium handling, increased atrial filling, enlargement, and increased left ventricular end-diastolic pressure [55,61].
Stroke (or cerebrovascular accident) emerges due to the combination of extracranial carotid artery disease and intracranial vessel diseases [62]. The risk of stroke is increased by 36% in diabetics compared with non-diabetics [63] and one-third of all stroke patients are diabetics [64]. There are two main forms of stroke that may feature in diabetes, ischemic and hemorrhagic stroke, with both having the potential of significantly increasing morbidity and mortality amongst diabetics [65]; and in the clinic, stroke is manifested as asymptomatic carotid artery occlusion or cerebral small vessel disease to transitory ischemic attack [62].
Strict glycemic and lipid control as well as management of hypertension are essential for managing both diabetes-induced cardiovascular and cerebrovascular diseases; furthermore, pharmacological interventions for diabetes-induced cardiovascular diseases including glucagon-like peptide 1 (GLP-1) receptor agonists, agonists of peroxisome proliferator-activated receptor gamma-γ (PPAR-γ) (e.g., pioglitazone), and the pharmacological inhibition of sodium-glucose cotransporter type 2 (SGLT2) and RAAS (e.g., by an angiotensin-converting enzyme (ACE) inhibitor or an angiotensin-receptor blocker) have shown clinical benefits [60,66,67,68]. Pharmacological interventions that can prevent stroke in diabetics include pioglitazone and anti-platelet drugs, such as, clopidogrel and aspirin; however, to reduce the combined risk of ischemic stroke, myocardial infarction or vascular death, clopidogrel may be more effective than aspirin [68,69,70]. Although these pharmacological interventions are available, they are inadequate for most diabetics and novel alternative adjuvant treatment options are needed.

3. Implications of Oxidative Stress in Diabetic Complications

Insulin deficiency due to the destruction/deficiency of β-cells is the main trigger of hyperglycemia in type 1 diabetes. A combination of factors such as hormonal and autoimmune dysfunction and environmental factors are associated with the destruction of β-cells [71]. In type 2 diabetes, hyperglycemia occurs due to impaired glucose uptake in skeletal muscles, deranged hormonal regulation of glucose metabolism, and impaired β-cell secretory function [72]. In essence, hyperglycemia due to β-cell dysfunction is common in diabetes. The progression of diabetes and the development of vascular complications stem from hyperglycemia-induced β-cell and vascular damage. This damage is known to be associated with reactive oxygen and nitrogen species (e.g., superoxide ion and nitric oxide).
Although various processes are implicated in the pathophysiology of diabetic retinopathy, five major metabolic pathways are prevalently accepted to significantly contribute to the pathophysiology of diabetic complications: formation and accumulation of AGEs (advanced glycation end-products), increased expression of the receptor for AGEs (RAGE) and its activating ligands, activation of protein kinase C isoforms (PKCs), increased flux via the hexosamine pathway, and increased polyol pathway flux [73,74,75,76,77].
The accumulation of AGEs and their interaction with RAGE can influence the permeability and self-regulation of vascular endothelial cells, inducing inflammation and releasing various proangiogenic cytokines and growth factors [76,78]. In DR, AGE accumulation and RAGE activation mediate the death of retinal pericytes, endothelial dysfunction, BRB breakdown, activation of NF-κB, and the induction of VEGF [78,79]. In diabetic complications, hyperglycemia promotes the production of diacylglycerol (DAG), an important co-factor of PKC-α, β, and δ isoforms [75,76,79]. PKC–DAG interactions are observed in the retina, glomerulus, heart, and aorta where they may promote the remodeling of the basement membrane and consequently promote vascular dysfunction in the aforementioned tissues [76,80,81,82].
The hexosamine pathway functions as an alternative pathway to the normal glycolysis pathway in the hyperglycemic milieu [77]. As a result, excessive uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), the end-product of this pathway, is produced which can form glycolipids, glycoproteins, and proteoglycans which increases the burden of oxidative stress and worsen diabetic complications in various organs including the eyes and the heart [78,83,84,85]. In hyperglycemic conditions, one way that the activation of the polyol pathway contributes to the pathophysiology of diabetic complications is via increased aldose reductase (AR) activity which metabolizes excess intracellular glucose into sorbitol using NADPH as a cofactor and producing NADH [77,78,86]. As sorbitol accumulates within cells, osmotic damage and oxidative stress increase, and eventually, sorbitol is oxidized to fructose by sorbitol dehydrogenase (SDH) [78,86]. In DR, the accumulation of sorbitol and fructose in the retina correlates with increased oxidative stress, neuronal apoptosis, and astrocytes and Müller cell abnormalities [86,87]. The suppression of the polyol pathway has also been shown to impede the progression of experimental DPN, and clinical DPN, DN, and DR [88,89].
These pathways are major reactive oxygen species (ROS) pathways thereby key contributors to increased oxidative stress, a crucial phenomenon in the pathogenesis of diabetic complications [74]. Furthermore, these five pathways lead to elevated inflammation, inflammatory mediators, angiogenesis, and vascular anomalies [73,74,75]. However, of importance is the possibility that these pathways are not independent of themselves but likely stimulate pathological changes in diabetic complications via a complex interplay [75]. In fact, Brownlee [75] has provided multiple lines of evidence that these five pathways are controlled by a unified upstream event—the overproduction of mitochondrial superoxide by the mitochondrial electron transport chain.

Hyperglycemia and Oxidative Stress

Glucose-dependent insulin secretion relies on calcium ion influx into β-cells. This voltage-dependent calcium ion influx is triggered by an increased ATP:ADP ratio due to rising glucose intracellular concentrations. In physiological conditions, the intracellular ATP:ADP ratio increases transiently and returns back. However, hyperglycemic conditions cause an excessive reduction in free ADP concentrations and an increase in the production of reducing equivalents (NADH and FADH2). This results in an increase in the availability of reducing equivalents and accelerated oxidative phosphorylation. Consequently, there is an excess of mitochondrial reductants such as coenzyme Q due to the lack of sufficient ADP for ATP formation. In this condition, the reduction in available oxygen occurs at a faster rate than ATP formation and ROS production ensues [90].
Endogenous antioxidant systems such as thioredoxin, glutathione, peroxidases, and the like (to be discussed in later sections) maintain ROS within optimal concentrations. However, in hyperglycemic conditions, antioxidant systems become dysfunctional and ROS-dependent signaling pathways are activated [91]. Furthermore, ROS generation from mitochondria and nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) is implicated in insulin signaling. NOX1 is proposed to be upregulated in hyperglycemic conditions possibly due to increased diacyl glyceride-induced protein kinase C (PKC) levels. Consequently, excessive NOX activity causes uncoupling of endothelial nitric oxide synthase (eNOS) and increased production of peroxynitrite and superoxide [92,93].
ROS overproduction is associated with the activation of various pro-inflammatory and growth pathways. For example, ROS mitochondrial overproduction has been associated with reduced mammalian target of rapamycin (mTOR) activity. In chronic hyperglycemic conditions, reduced mTOR activity has been associated with 5′ adenosine monophosphate-activated protein kinase (AMPK) -induced increase in ROS production [94] and increased expression of thioredoxin-interacting protein [95]; all of which result in reduced β-cell differentiation and function [96]. Furthermore, increased ROS production has been associated with a reduction in protein kinase B (AKT)/mTOR activation leading to β-cell-induced apoptosis [97].
In the same vein, the AKT/mTOR pathway is associated with insulin signaling and activation of this pathway has been associated with improved survival of cardiomyocytes in diabetes-related myocardial infarction [98]. However, insulin resistance is proposed to be induced by mitochondrial ROS generation [99] and parallel inhibition of the phosphatidylinositol 3-kinase (PI3K)/AKT insulin signaling pathway [100]. Therefore, mitochondrial ROS generation is implicated in insulin resistance [99] and cardiac hypertrophy via mTOR-dependent mechanisms in diabetic conditions [98,101]. Furthermore, inhibition of insulin receptor substrate (IRS) via ROS inactivation of the PI3K/AKT pathway leads to inhibition of both glucose transporter 4 (GLUT4) translocation and glucose reabsorption in the skeletal muscles [99].
In essence, during over-nutrition states, a shift in redox status is implicated in insulin resistance and this results in impaired response to insulin in the skeletal/cardiac muscle and hepatic tissues with the characteristic dysfunctional control of glucose uptake and uncontrolled gluconeogenesis. In line with this, it is proposed that ROS-induced insulin resistance plays a major role in endothelial damage [73,102] and cardiovascular diabetic complications [73,93]. Overall, ROS modulation of insulin signaling and growth pathways such as the AKT/PI3K and mTOR pathways contribute to the progression of diabetes and the development of diabetic vascular complications.
In addition, ROS contributes to the progression of diabetes and the development of diabetic vascular complications by eliciting inflammation. For instance, downregulation of the mTOR signaling pathway via AMPK-dependent mechanisms has been shown to protect cardiomyocytes and skeletal muscles from inflammation [103,104]. Moreover, ROS is implicated in activation of redox-dependent NF-kB activation via PKC-dependent mechanisms [92,105]; redox-dependent serine/threonine phosphorylation and activation of JUN amino-terminal kinase (JNK)/extracellular signal-regulated kinase (ERK) pathways [105]. ROS activation of pro-inflammatory pathways is implicated in β-cell dysfunction and apoptosis; and endothelial damage. This ultimately causes the progression and development of cardiovascular complications [106].

4. Enzymatic Endogenous Antioxidants and Diabetic Complications

The endogenous antioxidant system consists of non-enzymatic and enzymatic antioxidants. Enzymatic antioxidants include superoxide dismutase (SOD) which can include metalloproteins such as manganese, copper/zinc SOD (MnSOD, Cu/ZnSOD), catalase (CAT), heme-oxygenases (HO), paraoxonase-1 (PON1) and the thioredoxins and glutaredoxins system which include: thioredoxin (TRx) and glutaredoxin (GRx), TRx reductase (TrxR), glutathione reductase (GR), glutathione (GSH), and NAD(P)H. Other Trx/Grx-dependent systems include glutathione peroxidase (GPx) and thioredoxin peroxidase (TrxP) [107] (Figure 1). These are the common antioxidant defense systems in the vascular wall to counteract ROS generation [108]. Non-enzymatic endogenous antioxidants include albumin, ceruloplasmin, and metal-binding proteins [109]. The transcription and expression of endogenous antioxidants are regulated by the nuclear factor erythroid 2-related factor 2/antioxidant response element (Nrf-2/ARE) system [110]. Generally, endogenous antioxidants diffuse reactive oxygen species, via redox reactions. In essence, mitochondrial enzymes such as MnSOD convert superoxide ion to hydroperoxide (Figure 1). Hydrogen peroxide in the presence of intracellular iron (Fe2+) forms hydroxyl ions (Figure 1) which oxidize proteins and cell membranes which can lead to deranged cellular signaling and apoptosis. However, TRx/GRx systems as shown in Figure 1, reduce hydroxyl ions and hydrogen peroxide thereby maintaining cellular redox homeostasis and cell viability [111]. The next paragraphs of this review will focus mostly on enzymatic endogenous antioxidant systems and their implications in diabetic complications.

4.1. Enzymatic Endogenous Systems Implicated in Diabetic Retinopathy

As mentioned earlier, oxidative stress due to a compromised antioxidant system (Figure 1) is central to the development of diabetic complications. In the retina, the formation of glycosylated proteins leads to the generation of free radicals and causes oxidative tissue damage due to glutathione (GSH) depletion [112]. Moreover, reduced activity of Nrf-2 and a parallel reduction in MnSOD, GPx, CAT, and mitochondrial function are suggested to occur in the retina in diabetic conditions [113]. Animal studies have shown that increased SOD3 (an isoform of extracellular SOD) prevented retinal muller cell activation and pericyte dysfunction [114]. Furthermore, extracellular SOD, CAT, and GPx are reduced in patients with DR; consequently, they are suggested to be potential biomarkers for DR [115,116]. Moreover, thioredoxin-interacting protein (TXNIP), an endogenous thioredoxin (Trx) inhibitor, works by inhibiting Trx reducing capacity and promoting cellular oxidative stress [117]. TXNIP is upregulated in diabetes and plays an important role in the pathophysiology of DR [118]. Taken together, the regulation of endogenous antioxidant enzymes in the diabetic retina is important in the management of DR.

4.2. Endogenous Antioxidants in Diabetic Nephropathy (DN)

The development of DN is attributed mostly to oxidative stress (OS) [119]. Some intracellular components, including the endoplasmic reticulum, peroxisomes, and mitochondria, as well as particular enzyme clusters, such as NADPH oxidases (NOX), are involved in the formation of ROS in kidney cells [120]. Increased ROS levels are caused by either increased production, a failure of antioxidant mechanisms, or a combination of the two [121]. OS causes inflammation, kidney scarring, and other pathophysiological changes, resulting in further glomerular filtration rate (GFR) decline and DN progression [122]. Podocyte damage, proteinuria, and tubulointerstitial fibrosis are also linked to OS [123]. Trx, CAT, cytochrome c oxidase, SOD, and GPx perform antioxidant defense in the kidney [124].
SOD is the first line of enzymatic antioxidant defense in the kidney [125]. The findings of studies on SOD in diabetes-induced chronic kidney disease (CKD) patients are contradictory and challenging to interpret. Some researchers discovered no significant difference in SOD levels between CKD patients and controls [126]. Others discovered lower SOD activity in hemodialysis and peritoneal dialysis patients when compared to controls [127]. However, in animal studies, SOD1-transgenic db/db mice and STZ-treated SOD1-transgenic mice had lower albuminuria, transforming growth factor (TGF)-β1, and collagen IV expression, as well as mesangial matrix expansion and lower oxidative stress markers than control diabetic mice [128]. SOD2 deficiency has been linked to worsening renal dysfunction, tubulointerstitial fibrosis, inflammation, and apoptosis [129] while SOD3 expression was significantly increased in the glomerulus and tubular area of db/db mice after supplementation with recombinant human SOD3. Recombinant human SOD3 supplements improved diabetic nephropathy by preventing ROS and the phosphorylation of the ERK pathway [130].
Hong et al. [131] also reported on the benefit of targeting extracellular SOD as a treatment for type 2 DN through intrarenal AMPK-PGC-1α-Nrf2 and AMPK-FoxOs signaling. Extracellular SOD increased phosphorylation of AMPK, activation of peroxisome proliferative-activated receptor γ coactivator 1α (PGC-1α), and dephosphorylation of forkhead box O transcription factor (FoxO)1 and FoxO3a. The protective effects were attributed to enhanced nuclear translocation of Nrf2 and subsequently increased expression of NADPH dehydrogenase 1 and HO-1 [131].
Furthermore, in STZ-treated diabetic mice and db/db mice, proximal tubule-specific overexpression of CAT in DN reduced angiotensinogen, p53, and proapoptotic Bcl-2 associated X-protein (BAX) gene expression and decreased renal ROS generation and tubulointerstitial fibrosis [132,133]. This suggests a role of CAT in DN. In addition, glomerular GPx expression was lower in diabetic rats than in the normal control rats, and plasma and urine GPx levels were significantly lower in patients with diabetic glomerulosclerosis than in patients without glomerulosclerosis [134].
Wang et al. [135] reported on the role of kidney GPx4 expression level in DN progression. The GPx4 expression level was significantly lower in DN patients than in healthy controls and was associated with disease severity and progression [135]. Similarly, mice with DN showed significant changes in ferroptosis-associated markers such as increased expression levels of acyl-CoA synthetase long-chain family member 4 (ACSL4) and decreased expression levels of GPx4 [136]. Hence, the endogenous antioxidant systems CAT, SOD, and GPx are implicated in DN, and their upregulation may provide some benefit to DN patients.

4.3. Endogenous Antioxidants in Diabetic Cardiomyopathy

The endogenous antioxidants present in the heart protect against ROS toxicity. They include antioxidant enzymes such as SOD, CAT, GPx, GST, and glucose-6-phosphate dehydrogenase. ROS, such as superoxide anion, hydroxyl radicals, and hydrogen peroxide, play a role in mediating oxidative reactions in the development of diabetic cardiovascular complications [137,138]. Indeed, increased levels of malondialdehyde, thiobarbituric acid reactive substances (TBARS), hydroxynonenal (HNE), 8-hydroxyguanosine, 8-hydroxy-2′-deoxyguanosine, and 8-iso-prostaglandin F2α are biomarkers of chronic oxidative stress in diabetic cardiomyopathy [139].
Cardiomyocyte enlargement, myocardial interstitial and perivascular fibrosis, inflammation, apoptosis, and elevated markers of oxidative stress are the main pathological characteristics of diabetic cardiomyopathy [140]. Apoptosis has been found to be significantly increased in the myocardium of diabetic subjects [141]. Overexpression of the mitochondrial tetrameric mnSOD or catalase in the diabetic heart enhanced respiration, shielded cardiac mitochondria from oxidative damage, and restored bulk in diabetic mitochondria [142].
Overexpression of GPx resulted in an improvement in left ventricular diastolic function, and a reduction in cardiomyocyte hypertrophy, interstitial fibrosis, and myocardial apoptosis in mice with streptozotocin-induced diabetes [143]. Diabetic mice failed to develop cardiomyopathy when GPx was overexpressed under the control of a housekeeping gene promoter [143]. Many natural and synthetic Nrf2 activators have been shown in animal models to be effective in preventing diabetic cardiomyopathy [144]. In view of this, it can be inferred that an altered endogenous antioxidant system is implicated in the pathophysiology of diabetic cardiomyopathy and its upregulation might prove beneficial in managing the disease.

4.4. Endogenous Antioxidants in Cerebrovascular Diabetic Complications

The maintenance of the brain against oxidative stress involves both enzymatic and non-enzymatic antioxidants [145]. In diabetic cerebrovascular disorders including stroke and cognitive impairment, ROS increases the susceptibility to neuronal damage and functional decline via brain oxidation [146].
Endothelial dysfunction characterizes ischemic stroke with an increase in ROS and lower levels of endogenous antioxidants in humans [147,148]. Both stroke patients and animal models of stroke with middle cerebral artery occlusion (MCAO) have been shown to exhibit increased protein and lipid oxidation [149]. Moreover, a considerable rise in malondialdehyde (MDA) levels was observed in patients with acute ischemic stroke 24 h and seven days after stroke, together with a decline in the levels of antioxidant enzymes [150].
Resveratrol improved endothelial dysfunction by reducing the release of cytokines that promote inflammation, such as TNF-α and IL-1 [151], lowering oxidative stress in a sirtuin1-dependent way [152] and promoting endogenous antioxidant enzymes such as SOD, CAT, and GPx [153]. Green tea extract and epigallocatechin-3-gallate (EGCG) reduced lipid peroxidation, which was increased after MCAO [154]. Additional improvements in neurological scores, decreased apoptotic neuronal death, increased GSH levels, decreased infarct size, and enhanced Nrf2 expression were observed after EGCG treatment [155] similar to the findings of Alfieri et al. on the benefit of Nrf2 activation in stroke injury [156].
Diabetes-induced cognitive impairment is characterized by deficits in intelligence, attention, psychomotor efficiency, information processing speed, cognitive flexibility, and visual perception [157]. Resveratrol hindered cognitive impairment induced by type 2 diabetes mellitus (T2DM) [157]. Resveratrol treatment prevented learning and memory decline in T2DM mice when compared to control mice [136]. Resveratrol also prevented T2DM-induced hippocampal neuronal destruction and synaptic ultrastructural damage and increased the expression of Nrf2 and its downstream genes which were decreased in the T2DM group compared to the control [136]. These findings were similar to the report of Han et al. on the prevention of cognitive impairment through activation of the Sirt1/Nrf2 signaling pathway in diabetic rats through antioxidative mechanisms [158].
A weakened endogenous antioxidant system is seen in various diabetes-induced cerebral complications including ischemic stroke and cognitive impairment, while upregulating the endogenous antioxidant system provided some benefits in experimental models.

5. Effects of Antioxidant Phytochemicals on Endogenous Antioxidant Pathways

5.1. Effects of Antioxidant Phytochemicals on Mitochondrial Uncoupling Protein 2 (UCP2) and Mitochondrial Function in Diabetic Complications

Mitochondrial ROS generation is central to oxidative stress induced by hyperglycemia and contributes to the development and progression of diabetic complications. Excessive mitochondrial ROS generation is associated with an increase in mitochondrial membrane potential. In fact, a reduction in membrane potential by mitochondrial uncoupling protein 2 (UCP2) is known to reduce ROS production and prevent cell apoptosis [159].
Caffeic acid phenylethyl ester (CAPE), a polyphenol has been shown to protect retinal cells against tert-butyl hydroperoxide induced oxidative stress. CAPE is known to have a similar structure to the 4-hydroxy-2E-nonenal natural inducer of UCPs. However, in this study [160], CAPE was observed to be less toxic than 4-hydroxy-2E-nonenal. It was also shown to effectively reduce mitochondrial membrane at lesser concentrations than 4-hydroxy-2E-nonenal. This activity of CAPE was inhibited by UCP2 siRNA and inhibitors of UCP2 without affecting mRNA levels of UCP although a minimum but significant increase in ATP production was observed [160]. This implies that, CAPE is similar to the endogenous UCP inducer-4-hydroxy-2E-nonenal: as it also reduces ROS production by reducing mitochondrial membrane potential via induction of UCP (Figure 2). Albeit, it has minimum effect on cellular energy metabolism and cell viability compared to 4-hydroxy-2E-nonenal.
Resveratrol is proposed to prevent arrhythmia in diabetic conditions by alteration of UCP2 levels [161]. The altered expression of UCP2 has been linked to peroxisome proliferator-activated receptor gamma coactivator1-alpha (PCG-1α), a regulator of mitochondrial biogenesis [162] which is known to both upregulate and indirectly downregulate the expression of UCP2 as negative feedback response.
Mitochondrial membrane potential is also regulated by the alteration of mitochondrial permeability via the formation of mitochondrial permeability transition pore (mPTP). The voltage-dependent anion channels (VDAC) and adenine nucleotide translocator (ANT) form the major channels for the exchange of ATP, proteins, water, and ions across the mitochondrial membrane. Opening or Closing of these channels is regulated by binding of anti/pro-apoptotic proteins (i.e., Bcl-2 /Bax), cyclophilin D, outer membrane transporter protein (18 kDa) (TSPO), hexokinase, and creatinine kinase [163]. Polyphenols are proposed to modulate mPTP by altering the interaction of these “regulatory” proteins with VDAC and ANT; or by directly binding to ANT as extensively reviewed by Naoi et al. [163]. For example, resveratrol prevents mitochondria membrane potential collapse by preferentially increasing the interaction of antiapoptotic protein Bcl-2 with VDAC in the outer membrane of the mitochondria. This causes inhibition of mPTP opening, preventing the release of cytochrome c and inhibition of apoptosis [163]. This effect could be beneficial in conditions such as myocardial infarction, which is characterized by the accumulation of mitochondrial ROS and cell death due to the opening of mPTP, leading to the collapse of mitochondrial membrane potential, the disruption of ATP production and induction of necroptosis [164].
Furthermore, anthocyanins (cyanidin-3-glucoside, delphinidin 3-O-glucoside, pelargonidin 3-O-glucoside) are reported to prevent the uncoupling of mitochondria in ischemic cardiac tissue. Skemiene et al. [165] observed that anthocyanins act as a substrate for mitochondria complex 1, reduce cytochrome c, and thereby preserve mitochondria function. A reduction in cytochrome c has been associated with the inhibition of caspase-dependent apoptosis and restoration of cardiac function in diabetic rats [166]. Taken together, polyphenols exert a beneficial effect against ROS-induced vascular damage by maintaining mitochondrial membrane potential via UCP2 and mPTP regulation; improving mitochondrial biogenesis, improving electron transport chain function; thereby preventing necroptosis and cell death [167,168].

5.2. Effects of Antioxidant Phytochemicals on Glutathione Peroxidase 4 (GPx4) and Coenzyme Q (CoQ) in Diabetic Complications and Ferroptosis

Ferroptosis is a programmed cell death that occurs due to iron-dependent lipid peroxidation eliciting necro-inflammatory processes [169]. Iron overload is associated with the progression of diabetes [170] and diabetic complications [171], likely due to hyperglycemia-induced expression of iron transporters in the intestines [172]. Iron propagates the formation of lipid peroxide and eventual cell death via the Fenton reaction, enzymatic autoxidation of polyunsaturated fatty acids, and lipoxygenase catalytic formation of arachidonoyl-(AA-)OOH-phosphatidylethanolamine/arachidonic acid [173].
Furthermore, lipid peroxides break down into hydroxy fatty acids or reactive aldehydes MDA [174]; the latter is a known marker of oxidative stress in type 2 diabetic patients [175]. GPx4 and coenzyme Q (CoQ) are two major antioxidants associated with reducing lipid peroxidation of mitochondrial and cellular membranes leading to ferroptosis [169,176].
As mentioned earlier, anthocyanins and quercetin are known to display “CoQ1 like” activity by acting as a complex 1 substrate during oxidative phosphorylation [165]. They may also bind lipid peroxides, thereby stabilizing the mitochondrial membrane during ischemia [165]. In addition, kaempferol and apigenin have been shown to increase CoQ biosynthesis in kidney cells [177] and resveratrol and para-coumarate serve as precursors for CoQ synthesis in human cells [178]. Based on the study by Fernández-del-Río et al. [177], it appears that flavonoid structure with a single hydroxyl group located on the C4′ in the B ring together with a pie bond between the A and C ring is important for the significant synthesis of CoQ. This is a common feature for resveratrol, kaempferol, and apigenin. In addition, the presence of these features and a hydroxyl group situated at C3 of the C ring elicits a much higher synthesis of CoQ10 as observed for kaempferol. Beyond CoQ synthesis, resveratrol was observed to increase mRNA expression of genes for CoQ synthesis and increase CoQ10 levels in the liver of high-fat diet mice; however, the change in CoQ10 levels was not significant [179].
Furthermore, kaempferol is reported to reduce the risk of diabetic complications in diabetic rats by decreasing lipid peroxidation and increasing glutathione/glutathione peroxidases in liver, kidney, and heart tissues [180], but the effect on CoQ levels is not reported. Polyphenols may contribute to CoQ levels and prevent lipid peroxidation and ferroptosis. However, further studies are required.
The role of GPx4 in ferroptosis and the implication for diabetic complications such as retinal damage; kidney damage, cardiomyopathy, and cognitive dysfunction is exemplified with evidence reviewed by Yang and Yang [176]. In brief, GPx4 is dependent on GSH availability for the reduction in lipid peroxides. GSH in turn is influenced by cysteine synthesis and xc transporter system, and NAD(P)H. The latter is affected by NOX activity which is implicated in cardiac injury [181] and diabetic cardiovascular diseases [182]. Therefore, inhibiting NOX activity, upregulating GSH/GPx4 activity, and reducing iron overload could be beneficial for preventing hyperglycemia-induced ferroptosis and related organ damage [176].
In line with the above, glabridin reduced genetic and protein expression of transferrin receptor 1, while increasing genetic and protein expression of GSH/GPx4 and xc transporter system. The study suggests that the reno-protective effect of glabridin was in part related to the reduction in iron load in the kidney and prevention of lipid peroxidation via upregulation of the GSH/GPx4 system [183].
Similarly, calycosin, an isoflavone, reduced markers for cellular labile iron pool and increased GPx4 in kidney cells exposed to hyperglycemic conditions [184]. In the kidneys of diabetic mice model, calycosin was proposed to inhibit hyperglycemia-induced injury by modulating ferroptosis [184]. In relation to diabetic cardiac diseases, naringenin was observed to reduce lipid peroxidation, and NOX2 expression in the cardiac tissue of diabetic mice, indicating potential ferroptosis mechanisms [185]. Moreover, astragaloside-IV is reported to reduce high-glucose-induced oxidative stress in retinal pigment epithelial cells by increasing cysteine synthesis and GPx4 expression via Nrf2-dependent mechanisms [186].
Prevention of vascular complications could also be attributed to the preservation of B-cell function. Cryptochlorogenic acid, a major active component of mulberry leaf, and quercetin are known to prevent lipid peroxidation and consequent ferroptosis by upregulating GPx4 and reducing the accumulation of iron in pancreatic cells in animal diabetic models and in vitro studies [187,188]. Taken together, polyphenols prevent ferroptosis by modulating GSH synthesis, iron intracellular transport, and upregulating the GPx4 antioxidant system (Figure 2), which could be beneficial for preventing the progression of diabetes and vascular complications.
Figure 2. Summary of the effects of polyphenols on endogenous antioxidant systems. In hyperglycemia-induced oxidative stress, polyphenols increase UCP expression (1), reduces mitochondria membrane potential ΔΨm (2) which leads to reduction in O2 (3); preventing mitochondrial damage and apoptosis/necroptosis. Polyphenols enhance Gpx/GSH/Trx function by increase in synthesis of GSH via increased activity of Xc transporter system, reduction in intracellular transport of Fe2+. Moreover, there is an increase in deacetylation activity of SIRT 3 which increases activity of isocitrate dehydrogenase activity (not shown) [189] and NADH leading to regeneration of GSH. Both the latter and former prevent ferroptosis. Polyphenols also enhance SOD activity via SIRT1/3-dependent mechanisms. Finally, increased SIRT3 activity also increases translation of several antioxidant enzymes including NOQ1, HO-1, and CAT via Nrf2-dependent mechanisms. Activation of Nrf2 inhibits NF-kB transcription activity, thereby preventing oxidative stress-induced inflammation which is associated with insulin resistance and endothelial damage.
Figure 2. Summary of the effects of polyphenols on endogenous antioxidant systems. In hyperglycemia-induced oxidative stress, polyphenols increase UCP expression (1), reduces mitochondria membrane potential ΔΨm (2) which leads to reduction in O2 (3); preventing mitochondrial damage and apoptosis/necroptosis. Polyphenols enhance Gpx/GSH/Trx function by increase in synthesis of GSH via increased activity of Xc transporter system, reduction in intracellular transport of Fe2+. Moreover, there is an increase in deacetylation activity of SIRT 3 which increases activity of isocitrate dehydrogenase activity (not shown) [189] and NADH leading to regeneration of GSH. Both the latter and former prevent ferroptosis. Polyphenols also enhance SOD activity via SIRT1/3-dependent mechanisms. Finally, increased SIRT3 activity also increases translation of several antioxidant enzymes including NOQ1, HO-1, and CAT via Nrf2-dependent mechanisms. Activation of Nrf2 inhibits NF-kB transcription activity, thereby preventing oxidative stress-induced inflammation which is associated with insulin resistance and endothelial damage.
Antioxidants 12 00123 g002

5.3. SIRTs and Polyphenols

Silent information regulator proteins (SIRTs) are regulator proteins that couple cellular nutrient sensing to intracellular redox homeostasis via NAD+-dependent mechanisms. SIRTs belong to the class III histone deacetylases and exist as SIRT 1-7 isoforms. SIRTs are localized in the cytoplasm, mitochondria, and nucleus. They are involved with the regulation of redox signaling, transcription of Nrf2 with the expression of antioxidant enzymes, and mitochondrial biogenesis [189,190]. The functions of SIRTs are extensively reviewed in Singh et al. [189].
Resveratrol has been shown to activate SIRT1 activity thereby reducing oxidative stress and the progression of diabetes-induced kidney dysfunction in rats. SIRT1 activity reduced the acetylation of FOXO3a, a key regulator of MnSOD expression [189,191]. Upregulation of SIRT1 antioxidant activity by resveratrol is also associated with PGC-1α activation and the reduction in apoptosis with the improvement of mitochondria biogenesis [192]. Similarly, resveratrol is also shown to improve cardiomyopathy via the SIRT1/PGC-1α-dependent pathway [162]. Resveratrol is also reported to prevent mitochondrial membrane collapse by preventing mPTP pore opening. The suggested mechanism was related to SIRT3-dependent inhibition of cyclophilin-D interaction with ANT [193].
Furthermore, luteolin and fisetin were reported to increase the expression of SIRT1, 3, and 6 in human monocytic cells subjected to hyperglycemic conditions [194]. An increase in SIRT 1, 3, and 6 was associated with increased FOXO3a activity suggesting an effect of antioxidant enzymes [194]. In this respect, it can be said that resveratrol increases the activity of antioxidant systems (GSH, CAT, SOD) via SIRT1-dependent mechanisms. This implies that it could be useful for the prevention of diabetic kidney damage [195]. Interestingly, resveratrol [196], luteolin, and fisetin [194] are also observed to reduce NOX activity, suggesting the potential effect of preserving NADH for GSH antioxidant activity and related systems in the previous sections.
In light of the above, it can be inferred that polyphenols activate SIRTs which in turn upregulate enzyme antioxidants (Figure 2). Polyphenol activation of SIRTs also preserves mitochondrial function and induces mitochondrial biogenesis. This effect may also contribute to the prevention of ferroptosis by preserving GSH/GPx antioxidant activity (Figure 2).

5.4. Effects of Polyphenols on the Nuclear Factor (erythroid-derived 2)-like 2 (Nrf2) and Endogenous Antioxidant System

5.4.1. Nrf2 Regulation

There are seven Nrf2 ECH homology domains (Neh 1-7) that regulate both the activation and inhibition of Nrf2 activity [197]. Under normal conditions, Nrf2 is targeted by Kelch-like ECH-associated protein 1 (KEAP-1) for cytosol degradation. KEAP-1 binding to Nrf2 leads to ubiquitination by Cul3-Rbx1-E3 ligase. This negative regulator of Nrf2 (KEAP-1) attaches to the amino-terminal of the Neh-2 domain containing DLG and ETGE as its binding motif through a double glycine repeat domain. Ubiquitination of Nrf2 leads to its proteasomal degradation. In essence, KEAP-1 inhibits Nrf2 transcriptional activity [198].
Nrf2 stability is negatively controlled by the Neh-6 domain (Serine rich) which acts independently of KEAP-1. DSGIS and DSAPGS, its conserved peptide motifs are recognized by β-transducing repeat-containing protein (β-TrCP). Glycogen synthase kinase -3β (GSK-3β)-mediated phosphorylation of the DSGIS motif causes β-TrCP to bind more effectively to Neh-6 and promotes S-phase kinase 1 (SKP1)-cullin1 (Cul1)-F-box protein 1 (SCF) ubiquitin ligase complex leading to Nrf2 proteasomal degradation [199]. Moreover, the Neh-7 domain through binding with retinoid x receptor (RXRα) mediates repression of Nrf2 transcriptional activity [200].
Excess ROS causes redox disequilibrium and Nrf2 is translocated to the nucleus after Nrf2-KEAP-1 is disassembled. Once inside the nucleus, the Nrf2 Neh-5 domain functions as a transcription factor and has a redox-sensitive nuclear export signal. Furthermore, the CNC-bZIP region on Neh-1 plays a crucial role in the binding of Nrf2 to DNA and the small musculoaponeurotic fibrosarcoma (smaf) proteins [201] allowing Nrf2 to bind to antioxidant response elements (AREs: enhancer sequences in the regulatory region of Nrf2 target genes). This ultimately leads to activation and the expression of antioxidant genes and iron genes involved in the stress response [110]. In addition, ubiquitination and phosphorylation of KEAP-1 by protein kinase C (PKC) phosphorylation also regulate Nrf2 activity [202]. Other KEAP-1-independent regulation of Nrf2 includes the antagonism of GSK-3β action by PI3k-/AKT signaling [197].
Nrf2 activators including phytochemicals can upregulate Nrf2 activity to maximize its antioxidant activity either through structural inhibition of KEAP-1, activation of the Nfe2 (nuclear factor, erythroid 2) gene transcription, micro-RNA induced degradation of KEAP-1, inhibition of proteasomal Nrf2 protein degradation and modulation of upstream regulators of Nrf2. The following paragraphs will discuss examples of polyphenols that activate Nrf2 in select diabetic complications.

5.4.2. Nrf2 in Diabetic Cardiomyopathy

Naringenin, a compound with known antioxidant and anti-inflammatory effects, has been investigated for its potential benefits in STZ-induced diabetic cardiomyopathy. Naringenin decreased blood sugar levels along with a reduction in myocardial fibrosis and cardiomyocyte death [185]. Naringenin also significantly increased the expression of antioxidant enzymes and Nrf2, while inhibiting pro-inflammatory cytokines, reactive oxygen species levels, and nuclear factor kappa-B (NF-kB) expression [185]. Naringenin may control Nrf2 and NF-kB pathways to prevent diabetes-related cardiac damage by lowering oxidative stress and reducing inflammation [185].
Furthermore, garlic oil (diallyl trisulfide) boosted Nrf2 protein stability and nuclear translocation, protecting against hyperglycemia-induced ROS-mediated apoptosis by upregulating PI3K/AKT/Nrf2 pathways [203], in cardiomyocytes exposed to high glucose. This suggests that garlic oil may improve insulin sensitivity in cardiac myocytes via Nrf2-dependent mechanisms.

5.4.3. Nrf2 in Diabetic Nephropathy

Nrf2 is elevated in diabetic kidneys in both humans and animals in the early stages of the disease while its capacity to translocate into the nucleus is decreased, indicating functional impairment of Nrf2 [204]. Consequently, impaired NrF2 activity in DN leads to the accumulation of peroxide radicals which harm kidney cells [205].
EGCG reduced DN by activating the Nrf2/ARE signaling pathway by downregulating KEAP-1 and disrupting the Nrf2-KEAP-1 interaction, thereby increasing nuclear Nrf2 [204]. Sun et al., 2017 confirmed these findings by reporting that EGCG increased Nrf2 expression to prevent diabetic nephropathy [206]. A 24-week EGCG treatment activated Nrf2 expression and function in wild-type mice without affecting KEAP-1 expression. Diabetes-induced renal oxidative damage, inflammation, fibrosis, and albuminuria were all reversed by Nrf2 activation, while deletion of the Nrf2 gene negated the effects of EGCG [206]. Taken together, the data suggest that EGCG could activate Nrf2 via the KEAP-1-dependent and independent pathway.
Altamimi et al. [207] investigated the role of ellagic acid in the prevention of DN. Ellagic acid reduced NF-κβ, upregulated GSH, γGCL, and SOD, and increased nuclear translocation of Nrf2 while preventing renal damage, oxidative stress, inflammation, and apoptosis. Ellagic acid also inhibited KEAP-1 cytoplasmic expression and interaction with Nrf2, as well as increased AKT and GS3KB phosphorylation [207].
Sinapic acid, a polyphenolic metabolite, was found to have antioxidant and anti-inflammatory properties in DN by upregulating the Nrf2/HO-1 signaling pathway [208]. A similar effect has also been observed for astaxanthin in diabetic rats. In addition to upregulating the Nrf2/HO-1 signaling pathway, astaxanthin also increased SOD-1 and reduced fibronectin and collagen IV accumulation in the kidneys of diabetic rats. Astaxanthin also reduced MDA levels suggesting a reduction in oxidative stress in diabetic rats [209].
Paeonol (PA), a phenolic compound derived from the cortex mountain root bark, was tested for antioxidant activity in glomerular mesangial cells and STZ diabetic mice by Zhang et al. [210]. PA provided renoprotection by activating the Nrf2/ARE pathways via SIRT1, suggesting that PA slows the progression of diabetic renal fibrosis [210].
Hesperitin is also reported to provide renoprotection against DN by upregulating the Nrf2/ARE/glyoxylase pathway and augmenting γGCL, a target gene of Nrf2/ARE signaling [211].
These evidences suggest that polyphenols activate the endogenous antioxidant systems in DN via upregulation of Nrf2 activity by inhibiting its proteasomal degradation and improving its translocation to the nucleus and could provide an additional option in the pharmacotherapy of diabetic complications including DN.

5.4.4. Nrf2 in Diabetic Retinopathy

Shi et al. [212] investigated the effects of rhaponticin, a polyphenol stilbene, in DR of diabetic rats. Rhaponticin caused upregulation of Nrf2, NQO-1, and HO-1, while MDA levels were significantly reduced, along with increased activity of SOD, CAT, and GPx. Other effects of rhaponticin include the downregulation of TNF-α, matrix metalloproteinase-2, and the upregulation of IL-10 and tissue inhibitor of metalloproteinase-1 (TIMP-1) in the retina. These findings demonstrate the importance of Nrf2 in maintaining redox homeostasis in the retina, as well as the ability of rhaponticin to prevent diabetic retinal changes via antioxidant, anti-inflammatory, and hypoglycemic effects [212].
Curcumin, a natural polyphenol significantly reduced retinal edema and outperformed insulin in terms of anti-photoreceptor apoptosis [213]. Curcumin alleviated compensatory Nrf2 pathway activation by directly reducing free radicals when combined with insulin, and it better maintained Nrf2 pathway homeostasis compared to insulin alone in the early stages of diabetes [213]. When combined with hypoglycemic agents, curcumin was beneficial in the management of diabetic complications [213]. Bucolo et al. also reported a similar effect of curcumin in high glucose-induced DR [214]. Antioxidant activity was linked to the activation of the Nrf2/HO-1 pathway through the ERK pathway [214]. This suggests antioxidant and anti-inflammatory effects of curcumin in DR.
Blueberry anthocyanin extract (BAE) protected human retinal capillary endothelial cells from high glucose injury [215]. BAE increased the levels of Nrf2 and HO-1 mRNA while decreasing vascular endothelial growth factor (VEGF) and increasing antioxidant enzymes such as GSH and GPx. The antioxidant effect was achieved by activating the Nrf2/HO-1 pathways [215]. A comparison of BAE and the anthocyanin constituents (malvidin, and malvidin glucosides) showed that malvidin-3-glucoside was the most effective for inhibiting high glucose-induced ROS production in human retinal capillary endothelial cells (HRCECs). Moreover, malvidin-3-glucoside did not inhibit NOX4 expression but increased CAT and SOD expression. Furthermore, BAE, malvidin, and glucosides inhibited VEGF, ICAM, and NF-κB expression [216]. Malvidin-3-glucoside and other anthocyanins in grape pomace were suggested to modify KEAP1/Nrf2 interaction and increase the expression of HO-1 and NQO1 antioxidants [217]. This suggests that malvidin-3-glucoside and the likes inhibit inflammation in endothelial cells by improving antioxidant enzymatic expression via Nrf2 signaling-dependent mechanisms.
Pterostilbene, a natural stilbene with documented antioxidant activity has shown protective activities against DR [218]. Pterostilbene conferred protection by reducing protein and lipid oxidative damage and restoring antioxidant enzyme activities, which it accomplished by activating the PI3K/Akt/GSK3/Nrf2 pathways [218]. Matos et al. [219] extensively reviewed the benefits of flavonoids in DR [219].
In summary, polyphenols can confer protection against oxidative stress via upregulating endogenous antioxidants through the Nrf2/KEAP-1 pathway in DR and could be pharmacologically targeted in the future management of DR.

5.4.5. Nrf2 in Diabetic Peripheral Neuropathy

DPN pathogenesis is influenced by oxidative stress and neuroinflammation [220]. Phytochemicals have also been studied as Nrf2 activators in the management of DPN. Fisetin, a phytoflavonoid, has been shown in experimental DPN to protect from oxidative stress and neuroinflammation via NF-κB inhibition and Nrf2 upregulation [221]. Nrf2 increased the levels of antioxidant enzymes such as HO-1, NQO-1, and GCL [221].
Morin, a bioflavonoid with documented antioxidant capacity has been investigated for its benefits in experimental DPN [222]. Morin reduced high glucose-induced mitochondrial superoxide production, membrane depolarization, and ROS generation while increasing Nrf2 activity and inhibiting Nf-KB-mediated neuro-inflammation [222]. Rutin was also beneficial in the STZ model of DPN. When compared to DPN control rats, rutin combined with the cyclooxygenase 2 (COX-2) inhibitor nimesulide reversed oxidative damage as evidenced by increased levels of SOD, CAT, and GSH, as well as Nrf2/HO-1 activation in the sciatic nerve [223].
Overall, Nrf2 regulates the expression of several antioxidant enzymes and can be said to be central to increased endogenous antioxidant activity. Moreso, polyphenol antioxidant activity via modulation of Nrf2 signaling inhibits hyperglycemia-induced inflammation and apoptosis (Figure 2). This emphasizes the benefits of polyphenols for targeting the Nrf2 signaling pathway in the management of diabetic complications. Table 1 below highlights examples of polyphenols and their effect on antioxidants for the benefit of various diabetic complications.

5.4.6. Polyphenol and Endogenous Antioxidants Effect

From the previous sections, the polyphenol effect on SIRT1/3 activity can be considered a signaling connector between ROS production, GPx function, and Nrf2 signaling. This is because they activate MnSOD, increase NADH which is crucial for GPx function and increase Nrf2 signaling. The latter then leads to the upregulation of other antioxidants and anti-inflammatory effects (Figure 2). Moreover, ROS overproduction is associated with insulin resistance [99,103], and polyphenol upregulation of endogenous antioxidants via Nrf2-dependent mechanisms can be said to be beneficial for insulin resistance by the upregulation of the PI3K/AKT pathway [218]. Polyphenol modulation of mTOR via the PI3K/AKT pathway could be beneficial for the prevention of cardiac hypertrophy and preserving β-cell function [97,98,101]. Therefore, targeting the Nrf2 pathway and the SIRTs could be beneficial for the prevention of insulin resistance and associated diabetic cardiovascular complications. Regulation of the Nrf2 pathway by microRNAs has been studied. microRNAs could also be modulated by polyphenols and can indirectly modulate antioxidant activity [224].
Other antioxidants such as paraoxonase are modulated by polyphenols. Glabridin, at levels lower than antioxidant concentrations, was shown to increase expression and transcription of PON2, in addition to MnSOD and CAT in hyperglycemic mice. The mechanism of action was suggested to be via protein binding with PON2 [225]. A similar study with curcumin-enriched yoghurts and metformin in diabetic rats showed that curcumin sustained the function of PON1 in diabetic conditions probably via preventing glycation of PON1 [226]. Taken together, glabridin and curcumin could prevent atherosclerosis by upregulating PON activity and reducing lipid peroxidation [225,226].
Glabridin was also shown to inhibit lipopolysaccharide-induced inflammation in THP-1 cells by downregulating the expression of inducible nitric oxide synthase (iNOS). Downregulation of iNOS and nitrotyrosine production was also observed in the liver of high-fat-fed mice. This suggests that glabridin could prevent inflammation associated with nutritional-induced oxidative stress (i.e., hyperglycemia) [227].
In relation to inflammation, polyphenols such as punicalagin, prevented nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) induced pyroptosis (inflammation-induced cell death) by inhibiting NOX4 activity. This in turn reduces Trx-TXNIP dissociation and subsequent activation of NLRP3. The data suggest the role of polyphenol modulation of the Trx antioxidant system for pyroptosis in the development of DN [228].
Polyphenols could also indirectly exert antioxidant activity through highly lipophilic gut microbial-derived metabolites [229]. Therefore, polyphenol gut microbiota interactions contribute to systemic polyphenol antioxidant activity. Iatcu et al. [230] extensively reviewed the role of gut microbiota in various type-2 diabetes complications. In general, the proportion of Bifidobacterium spp. and Lactobacillus spp. to Enterococcus spp. and Clostridium spp. is reduced in patients with type 2 diabetes and complications.
Gallic acid is known to modulate gut microbiota in different conditions including stress and inflammatory bowel disease [231]. An evaluation of healthy dog fecal content revealed that long-term supplementation of gallic acid increased anti-inflammatory short-chain fatty acid concentrations. This increase was positively correlated with an increase in Erysipelotrichaceae_unclassified spp. and Faecalibacterium spp., and a decrease in the proliferation of pro-inflammatory Parasutterella spp.; suggesting a beneficial prebiotic and anti-inflammatory effect of gallic acid. In this study, gallic acid supplementation was also associated with increased levels of CAT and SOD [231].
In a similar study, olive oil enriched with polyphenols and thyme elicited an increase in Bifidobacterium spp. in humans. Furthermore, a reduction in Clostridium spp. was associated with a decrease in isolithocholic acid; the latter is associated with a reduced risk of cardiometabolic disease in chronic insomnia [232]. Furthermore, the prebiotic effect of olive oil enriched with polyphenols was associated with reduced oxidized low-density lipoproteins and increased microbial metabolites of procatechuric and hydroxytyrosol, suggesting a relationship of gut-derived metabolites and antioxidant activity [233].
In fact, specific gut microbiota-derived polyphenolic metabolites also retain the ability to modulate antioxidant systems. For example, in vitro studies on urolithins A and B (gut microbial metabolites derived from polyphenols) are known to preserve podocyte function by upregulation of Bcl-2 expression and nephrin expression. Urolithin A, specifically, upregulated Bcl-2 in high glucose conditions [229]. In vivo studies with urolithin A have been shown to prevent renal kidney injury in vivo via the Nrf2 signaling pathway [234]. These results suggest that polyphenols–gut microbiota interactions contribute to the modulatory effect of polyphenols on endogenous antioxidants.
Overall, polyphenols influence endogenous antioxidant systems via multiple mechanisms. A summary of the effects of polyphenols on some endogenous antioxidant systems is represented in Figure 2 below.

6. Potential Therapeutic Strategies to Alleviate Diabetic Complications via the Activation of Endogenous Antioxidants by Exogenous Antioxidant Phytochemicals

One therapeutic strategy for harnessing the beneficial effects of polyphenols could be drug synthesis. For example, redesigning polyphenols to include imidazole derivatives [235,236] might prove a potential way to harness polyphenols for insulin resistance via Nrf2-dependent mechanisms. Structure-related activity of studies could assist with the identification of phenolic components with high affinity for inducing antioxidant systems. For instance, cyanidin is a potential drug candidate for antioxidant therapy as it was identified as a potent activator of SIRT6 [237]. Further drug discovery studies are required to harness this potential of cyanidin. For example, the chemical synthesis of cyanidin analogs coupled with in silico studies could facilitate the discovery of cyanidin analogs with optimized pharmacokinetic properties and less toxicity [238]; making it useful for therapy of diabetic complications. Taken together, further studies of chemically modified polyphenols and their effect on antioxidant systems are required.
Beyond synthetic drug designs, harnessing the health benefits of polyphenols in their natural form depends on their bioavailability. However, bioavailability is influenced by several factors such as food source, food processing, gut microbiome/hepatic biotransformation, and individual variability. Rodriguez-Mateos et al. [239] reported the bioavailability of polyphenols after the consumption of berry drinks compared to berry bun in healthy volunteers. Bioavailability and physiological effects on the endothelium were associated with heat processing and gut microbiome biotransformation—only the latter had an effect six hours post-consumption and was associated with unprocessed berry drink [239]. With respect to studies by Rodriguez-Mateos et al. [239], cooking may also improve the availability of polyphenols as reported in the study by Abukhabt et al. [240]. Heat processing causes a release of polyphenols from the food matrix and can increase absorption and colonic concentrations both of which are crucial for polyphenol bioactivity.
As discussed earlier, it is accepted knowledge that the gut microbiome is implicated in the therapeutic benefits of polyphenols. Gut microbiome dysbiosis is implicated in the disruption of intestinal tight junctions, a common pathological feature of aging and diabetes. Hidalgo-Liberona et al. [241] suggest that decreased intestinal integrity may reduce the bioavailability of gut microbiome-derived polyphenolic metabolites but increase the bioavailability of phase II metabolites. The implication of this could mean that when patients with compromised intestinal integrity (e.g., diabetic patients) consume flavonoid-rich metabolites, for instance, there may be increased anti-inflammatory activity of the flavonoids in these patients when compared to consuming the intact polyphenol due to increased bioavailability of phase II metabolites as reviewed in Williams et al. [242]. Furthermore, genetic polymorphisms of phase II enzymes may influence the bioavailability of polyphenols as reviewed in Manach et al. [243]. Therefore, in the future, understanding genetic polymorphisms and their effect on phase II polyphenolic metabolites would be useful for reducing variability to polyphenol therapy and improving the efficacy of polyphenols for the management of diabetic complications.
Moreover, given that gut microbiome-transformed metabolites were associated with a beneficial effect on the endothelium long after consumption [239]; it can be recommended that polyphenol benefits in diabetic patients may need to be enhanced with probiotics [244] for the prevention of endothelial damage, which is central to diabetic complications.
In addition, recent evidence suggests that the production of gut microbiome-derived metabolites from polyphenols could differ within individuals. Iglesias-Aguirre et al. [245] observed that resveratrol is metabolized to lunularin by mostly male compared to female individuals. According to Cortés-Martín et al. [246], such variations should be regarded as markers for human polyphenol-related metabotypes. However, given the evidence on the impact of equol metabotype/equol gut microbiota metabolizing ability on cardiometabolic risk factors as reviewed in Frankenfield [247], consideration of human polyphenol-related metabotypes might be important to identify specific metabotypes to maximize the benefits of polyphenols. This is promising for precision medicine. Besides, pharmacological therapy for metabolic syndrome could also have a beneficial influence on prebiotic effect of polyphenols, depending on cardiovascular risk factors and the associated medications [248]. Taken together, further research into individual variations due to sex, race, polyphenol-related metabotypes, and pharmacotherapy presents the opportunity for understanding how favorable drug–polyphenol–gut microbial interactions can be harnessed for the therapy of diabetes and diabetic complications.
Finally, the use of various nanoparticle platforms and encapsulation in food has been shown to improve the efficacy of polyphenols by increasing systemic and local delivery of polyphenols as extensively reviewed in [249,250]. It will be important to consider stability, especially in food particles, and the risk that metal-based and lipid-based nanoparticles may cause oxidative stress. Therefore, the design of polyphenol-based nanoparticles should take into consideration the route and could include a combination of polyphenols to mitigate any potential toxicity due to oxidative stress [251,252]. Further clinical including toxicity studies are required to validate the use of nano-based polyphenols in the management of diabetic complications.

7. Conclusions

The development and progression of diabetic complications are associated with various oxidative stress and inflammatory pathways. Various phytochemicals possess antioxidant, anti-inflammatory, and anti-hyperglycemic properties which can potentially prevent or impede the progression of diabetic complications. However, the low bioavailability and absorption of antioxidant phytochemicals currently limit their utility in the clinic. Further studies are needed to further explore the antidiabetic potentials of phytochemicals, improve their absorption and bioavailability, and explore their potential side effects in clinical trials. A future in which more diabetic patients will rely on phytochemicals as an alternative/adjuvant therapy for preventing or treating diabetic complications is one in which bioavailability is improved, and dosage standardization has been achieved either for the crude extracts or the purified bioactive compounds. The hope is that in combination with currently available treatment approaches, antioxidant phytochemicals would significantly reduce the burden of diabetic complications.

Author Contributions

Conceptualization, G.O. and O.-O.P.A.; investigation, O.-O.P.A., E.E.U. and G.O.; writing—original draft preparation, O.-O.P.A., E.E.U. and G.O.; writing—review and editing, O.-O.P.A. and G.O.; visualization, O.-O.P.A. and G.O.; supervision, G.O.; project administration, G.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Misra, A.; Gopalan, H.; Jayawardena, R.; Hills, A.P.; Soares, M.; Reza-Albarrán, A.A.; Ramaiya, K.L. Diabetes in Developing Countries. J. Diabetes 2019, 11, 522–539. [Google Scholar] [CrossRef] [PubMed]
  2. Ali, M.K.; Pearson-Stuttard, J.; Selvin, E.; Gregg, E.W. Interpreting Global Trends in Type 2 Diabetes Complications and Mortality. Diabetologia 2022, 65, 3–13. [Google Scholar] [CrossRef]
  3. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Stein, C.; Basit, A.; Chan, J.C.N.; Mbanya, J.C. IDF Diabetes Atlas: Global, Regional and Country-Level Diabetes Prevalence Estimates for 2021 and Projections for 2045. Diabetes Res. Clin. Pract. 2021, 183, 109119. [Google Scholar] [CrossRef] [PubMed]
  4. Harding, J.L.; Pavkov, M.E.; Magliano, D.J.; Shaw, J.E.; Gregg, E.W. Global Trends in Diabetes Complications: A Review of Current Evidence. Diabetologia 2019, 62, 3–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jing, X.; Chen, J.; Dong, Y.; Han, D.; Zhao, H.; Wang, X.; Gao, F.; Li, C.; Cui, Z.; Liu, Y. Related Factors of Quality of Life of Type 2 Diabetes Patients: A Systematic Review and Meta-Analysis. Health Qual. Life Outcomes 2018, 16, 1–14. [Google Scholar] [CrossRef] [PubMed]
  6. Hasler, C.M.; Blumberg, J.B. Phytochemicals: Biochemistry and Physiology. Introduction. J. Nutr. 1999, 129, 756S–757S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Martinez, K.B.; Mackert, J.D.; McIntosh, M.K. Polyphenols and Intestinal Health. In Nutrition and Functional Foods for Healthy Aging; Elsevier: Amsterdam, The Netherlands, 2017; pp. 191–210. [Google Scholar]
  8. Koche, D.; Shirsat, R.; Kawale, M. An Overview of Major Classes of Phytochemicals: Their Types and Role in Disease Prevention. Hislopia J. 2016, 9, 976–2124. [Google Scholar]
  9. Ramawat, K.G.; Dass, S.; Mathur, M. The Chemical Diversity of Bioactive Molecules and Therapeutic Potential of Medicinal Plants. In Herbal Drugs: Ethnomedicine to Modern Medicine; Springer: Berlin, Germany, 2009; pp. 7–32. [Google Scholar]
  10. Oran, S.A.; Althaher, A.R.; Mubarak, M.S. Cinchona Officinalis (Cinchona Tree) and Corylus Avellana (Common Hazel), 1st ed.; Herbs, Shrubs, and Trees of Potential Medicinal Benefits; CRC Press: Boca Raton, FL, USA, 2022; ISBN 978-1-00-320506-7. [Google Scholar]
  11. Desborough, M.J.R.; Keeling, D.M. The Aspirin Story—From Willow to Wonder Drug. Br. J. Haematol. 2017, 177, 674–683. [Google Scholar] [CrossRef] [Green Version]
  12. Drummond, E.M.; Harbourne, N.; Marete, E.; Martyn, D.; Jacquier, J.; O’Riordan, D.; Gibney, E.R. Inhibition of Proinflammatory Biomarkers in THP1 Macrophages by Polyphenols Derived from Chamomile, Meadowsweet and Willow Bark. Phytother. Res. 2013, 27, 588–594. [Google Scholar] [CrossRef]
  13. Bharadwaj, K.C.; Gupta, T.; Singh, R.M. Chapter 9—Alkaloid Group of Cinchona Officinalis: Structural, Synthetic, and Medicinal Aspects. In Synthesis of Medicinal Agents from Plants; Tewari, A., Tiwari, S., Eds.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 205–227. ISBN 978-0-08-102071-5. [Google Scholar]
  14. Pietta, P.G. Flavonoids as Antioxidants. J. Nat. Prod. 2000, 63, 1035–1042. [Google Scholar] [CrossRef]
  15. Gu, R.; Wang, Y.; Long, B.; Kennelly, E.; Wu, S.; Liu, B.; Li, P.; Long, C. Prospecting for Bioactive Constituents from Traditional Medicinal Plants through Ethnobotanical Approaches. Biol. Pharm. Bull. 2014, 37, 903–915. [Google Scholar] [CrossRef] [PubMed]
  16. Hall, S.D.; Wang, Z.; Huang, S.-M.; Hamman, M.A.; Vasavada, N.; Adigun, A.Q.; Hilligoss, J.K.; Miller, M.; Gorski, J.C. The Interaction between St John’s Wort and an Oral Contraceptive. Clin. Pharmacol. Ther. 2003, 74, 525–535. [Google Scholar] [CrossRef] [PubMed]
  17. Cade, W.T. Diabetes-Related Microvascular and Macrovascular Diseases in the Physical Therapy Setting. Phys. Ther. 2008, 88, 1322–1335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Cheung, N.; Mitchell, P.; Wong, T.Y. Diabetic Retinopathy. Lancet 2010, 376, 124–136. [Google Scholar] [CrossRef] [PubMed]
  19. Biallosterski, C.; van Velthoven, M.E.J.; Michels, R.P.J.; Schlingemann, R.O.; DeVries, J.H.; Verbraak, F.D. Decreased Optical Coherence Tomography-Measured Pericentral Retinal Thickness in Patients with Diabetes Mellitus Type 1 with Minimal Diabetic Retinopathy. Br. J. Ophthalmol. 2007, 91, 1135–1138. [Google Scholar] [CrossRef] [Green Version]
  20. Lamoureux, E.L.; Tai, E.S.; Thumboo, J.; Kawasaki, R.; Saw, S.-M.; Mitchell, P.; Wong, T.Y. Impact of Diabetic Retinopathy on Vision-Specific Function. Ophthalmology 2010, 117, 757–765. [Google Scholar] [CrossRef]
  21. Fenwick, E.K.; Pesudovs, K.; Rees, G.; Dirani, M.; Kawasaki, R.; Wong, T.Y.; Lamoureux, E.L. The Impact of Diabetic Retinopathy: Understanding the Patient’s Perspective. Br. J. Ophthalmol. 2011, 95, 774–782. [Google Scholar] [CrossRef]
  22. Shi, R.; Chen, L.; Wang, W.; Deng, Y.; Liu, Y.; Zhou, H.; Lin, R. Plasma MiR-26a-5p Is a Biomarker for Retinal Neurodegeneration of Early Diabetic Retinopathy. Eye 2021, 35, 1587–1599. [Google Scholar] [CrossRef]
  23. Coyne, K.S.; Margolis, M.K.; Kennedy-Martin, T.; Baker, T.M.; Klein, R.; Paul, M.D.; Revicki, D.A. The Impact of Diabetic Retinopathy: Perspectives from Patient Focus Groups. Fam. Pract. 2004, 21, 447–453. [Google Scholar] [CrossRef] [Green Version]
  24. Watkinson, S.; Seewoodhary, R. Ocular Complications Associated with Diabetes Mellitus. Nurs. Stand. 2008, 22, 51–57, quiz 58, 60. [Google Scholar] [CrossRef]
  25. Stitt, A.W.; Lois, N.; Medina, R.J.; Adamson, P.; Curtis, T.M. Advances in Our Understanding of Diabetic Retinopathy. Clin. Sci. 2013, 125, 1–17. [Google Scholar] [CrossRef] [PubMed]
  26. Duh, E.J.; Sun, J.K.; Stitt, A.W. Diabetic Retinopathy: Current Understanding, Mechanisms, and Treatment Strategies. JCI Insight 2017, 2, e93751. [Google Scholar] [CrossRef] [PubMed]
  27. Stitt, A.W.; Curtis, T.M.; Chen, M.; Medina, R.J.; McKay, G.J.; Jenkins, A.; Gardiner, T.A.; Lyons, T.J.; Hammes, H.-P.; Simó, R. The Progress in Understanding and Treatment of Diabetic Retinopathy. Prog. Retin. Eye Res. 2016, 51, 156–186. [Google Scholar] [CrossRef] [PubMed]
  28. Meyerle, C.B.; Chew, E.Y.; Ferris, F.L. Nonproliferative Diabetic Retinopathy. In Diabetic Retinopathy; Duh, E.J., Ed.; Humana Press: Totowa, NJ, USA, 2008; pp. 3–27. ISBN 978-1-934115-83-1. [Google Scholar]
  29. Isner, J.M.; Asahara, T. Angiogenesis and Vasculogenesis as Therapeutic Strategies for Postnatal Neovascularization. J. Clin. Investig. 1999, 103, 1231–1236. [Google Scholar] [CrossRef] [Green Version]
  30. Crawford, T.N.; Alfaro, D.V., 3rd; Kerrison, J.B.; Jablon, E.P. Diabetic Retinopathy and Angiogenesis. Curr. Diabetes Rev. 2009, 5, 8–13. [Google Scholar] [CrossRef]
  31. Capitão, M.; Soares, R. Angiogenesis and Inflammation Crosstalk in Diabetic Retinopathy. J. Cell. Biochem. 2016, 117, 2443–2453. [Google Scholar] [CrossRef]
  32. Gupta, N.; Mansoor, S.; Sharma, A.; Sapkal, A.; Sheth, J.; Falatoonzadeh, P.; Kuppermann, B.; Kenney, M. Diabetic Retinopathy and VEGF. Open Ophthalmol. J. 2013, 7, 4–10. [Google Scholar] [CrossRef] [Green Version]
  33. Campbell, M.; Doyle, S.L. Current Perspectives on Established and Novel Therapies for Pathological Neovascularization in Retinal Disease. Biochem. Pharmacol. 2019, 164, 321–325. [Google Scholar] [CrossRef]
  34. Praidou, A.; Androudi, S.; Brazitikos, P.; Karakiulakis, G.; Papakonstantinou, E.; Dimitrakos, S. Angiogenic Growth Factors and Their Inhibitors in Diabetic Retinopathy. Curr. Diabetes Rev. 2010, 6, 304–312. [Google Scholar] [CrossRef]
  35. Barber, A.J.; Lieth, E.; Khin, S.A.; Antonetti, D.A.; Buchanan, A.G.; Gardner, T.W. Neural Apoptosis in the Retina during Experimental and Human Diabetes. J. Clin. Investig. 1998, 102, 783–791. [Google Scholar] [CrossRef] [Green Version]
  36. Kern, T.S.; Barber, A.J. Retinal Ganglion Cells in Diabetes. J. Physiol. 2008, 586, 4401–4408. [Google Scholar] [CrossRef] [PubMed]
  37. Mohamed, Q.; Gillies, M.C.; Wong, T.Y. Management of Diabetic Retinopathy: A Systematic Review. JAMA 2007, 298, 902–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Fong, D.S.; Girach, A.; Boney, A. Visual Side Effects of Successful Scatter Laser Photocoagulation Surgery for Proliferative Diabetic Retinopathy: A Literature Review. Retina 2007, 27, 816–824. [Google Scholar] [CrossRef] [PubMed]
  39. Gross, J.G.; Glassman, A.R.; Jampol, L.M.; Inusah, S.; Aiello, L.P.; Antoszyk, A.N.; Baker, C.W.; Berger, B.B.; Bressler, N.M.; Browning, D. Panretinal Photocoagulation vs Intravitreous Ranibizumab for Proliferative Diabetic Retinopathy: A Randomized Clinical Trial. JAMA 2015, 314, 2137–2146. [Google Scholar] [PubMed] [Green Version]
  40. Sivaprasad, S.; Prevost, A.T.; Vasconcelos, J.C.; Riddell, A.; Murphy, C.; Kelly, J.; Bainbridge, J.; Tudor-Edwards, R.; Hopkins, D.; Hykin, P. Clinical Efficacy of Intravitreal Aflibercept versus Panretinal Photocoagulation for Best Corrected Visual Acuity in Patients with Proliferative Diabetic Retinopathy at 52 Weeks (CLARITY): A Multicentre, Single-Blinded, Randomised, Controlled, Phase 2b, Non-Inferiority Trial. Lancet 2017, 389, 2193–2203. [Google Scholar]
  41. Preti, R.C.; Ramirez, L.M.; Monteiro, M.L.; Carra, M.K.; Pelayes, D.E.; Takahashi, W.Y. Contrast Sensitivity Evaluation in High Risk Proliferative Diabetic Retinopathy Treated with Panretinal Photocoagulation Associated or Not with Intravitreal Bevacizumab Injections: A Randomised Clinical Trial. Br. J. Ophthalmol. 2013, 97, 885–889. [Google Scholar] [CrossRef]
  42. McCannel, C.A. Meta-Analysis of Endophthalmitis after Intravitreal Injection of Anti-Vascular Endothelial Growth Factor Agents: Causative Organisms and Possible Prevention Strategies. Retina 2011, 31, 654–661. [Google Scholar] [CrossRef]
  43. Wubben, T.J.; Johnson, M.W. Anti-Vascular Endothelial Growth Factor Therapy for Diabetic Retinopathy: Consequences of Inadvertent Treatment Interruptions. Am. J. Ophthalmol. 2019, 204, 13–18. [Google Scholar] [CrossRef]
  44. Zehden, J.A.; Mortensen, X.M.; Reddy, A.; Zhang, A.Y. Systemic and Ocular Adverse Events with Intravitreal Anti-VEGF Therapy Used in the Treatment of Diabetic Retinopathy: A Review. Curr. Diabetes Rep. 2022, 22, 525–536. [Google Scholar] [CrossRef]
  45. Gonzalez, V.H.; Campbell, J.; Holekamp, N.M.; Kiss, S.; Loewenstein, A.; Augustin, A.J.; Ma, J.; Ho, A.C.; Patel, V.; Whitcup, S.M. Early and Long-Term Responses to Anti-Vascular Endothelial Growth Factor Therapy in Diabetic Macular Edema: Analysis of Protocol I Data. Am. J. Ophthalmol. 2016, 172, 72–79. [Google Scholar] [CrossRef] [Green Version]
  46. Faselis, C.; Katsimardou, A.; Imprialos, K.; Deligkaris, P.; Kallistratos, M.; Dimitriadis, K. Microvascular Complications of Type 2 Diabetes Mellitus. Curr. Vasc. Pharmacol. 2020, 18, 117–124. [Google Scholar] [CrossRef] [PubMed]
  47. Tesfaye, S.; Selvarajah, D. Advances in the Epidemiology, Pathogenesis and Management of Diabetic Peripheral Neuropathy: Advances in Epidemiology, Pathogenesis and Management of DPN. Diabetes Metab. Res. Rev. 2012, 28, 8–14. [Google Scholar] [CrossRef] [PubMed]
  48. Iqbal, Z.; Azmi, S.; Yadav, R.; Ferdousi, M.; Kumar, M.; Cuthbertson, D.J.; Lim, J.; Malik, R.A.; Alam, U. Diabetic Peripheral Neuropathy: Epidemiology, Diagnosis, and Pharmacotherapy. Clin. Ther. 2018, 40, 828–849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Gandhi, M.; Fargo, E.; Prasad-Reddy, L.; Mahoney, K.M.; Isaacs, D. Diabetes: How to Manage Diabetic Peripheral Neuropathy. Drugs Context 2022, 11, 1–13. [Google Scholar] [CrossRef] [PubMed]
  50. Nitta, K.; Masakane, I.; Hanafusa, N.; Taniguchi, M.; Hasegawa, T.; Nakai, S.; Goto, S.; Wada, A.; Hamano, T.; Hoshino, J.; et al. Annual Dialysis Data Report 2017, JSDT Renal Data Registry. Ren. Replace. Ther. 2019, 5, 53. [Google Scholar] [CrossRef] [Green Version]
  51. Rossing, K.; Christensen, P.K.; Hovind, P.; Tarnow, L.; Rossing, P.; Parving, H.-H. Progression of Nephropathy in Type 2 Diabetic Patients. Kidney Int. 2004, 66, 1596–1605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Sagoo, M.K.; Gnudi, L. Diabetic Nephropathy: An Overview. In Diabetic Nephropathy; Gnudi, L., Long, D.A., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2020; Volume 2067, pp. 3–7. ISBN 978-1-4939-9840-1. [Google Scholar]
  53. Nakhoul, F.; Nakhoul, N.; Asleh, R.; Miller-Lotan, R.; Levy, A.P. Is the Hp 2-2 Diabetic Mouse Model a Good Model to Study Diabetic Nephropathy? Diabetes Res. Clin. Pract. 2013, 100, 289–297. [Google Scholar] [CrossRef]
  54. Gnudi, L.; Gentile, G.; Ruggenenti, P. The Patient with Diabetes Mellitus. In Oxford Textbook of Clinical Nephrology; Turner, N., Lameire, N., Goldsmith, D.J., Winearls, C.G., Himmelfarb, J., Remuzzi, G., Eds.; Oxford University Press: Oxford, UK, 2015; pp. 1199–1247. ISBN 978-0-19-959254-8. [Google Scholar]
  55. Jia, G.; Hill, M.A.; Sowers, J.R. Diabetic Cardiomyopathy: An Update of Mechanisms Contributing to This Clinical Entity. Circ. Res. 2018, 122, 624–638. [Google Scholar] [CrossRef]
  56. Wagenknecht, L.E.; Zaccaro, D.; Espeland, M.A.; Karter, A.J.; O’Leary, D.H.; Haffner, S.M. Diabetes and Progression of Carotid Atherosclerosis: The Insulin Resistance Atherosclerosis Study. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1035–1041. [Google Scholar] [CrossRef] [Green Version]
  57. Folsom, A.R.; Rasmussen, M.L.; Chambless, L.E.; Howard, G.; Cooper, L.S.; Schmidt, M.I.; Heiss, G. Prospective Associations of Fasting Insulin, Body Fat Distribution, and Diabetes with Risk of Ischemic Stroke. The Atherosclerosis Risk in Communities (ARIC) Study Investigators. Diabetes Care 1999, 22, 1077–1083. [Google Scholar] [CrossRef]
  58. Ergul, A.; Kelly-Cobbs, A.; Abdalla, M.; Fagan, S.C. Cerebrovascular Complications of Diabetes: Focus on Stroke. Endocrine Metab. Immune Disord.-Drug Targets 2012, 12, 148–158. [Google Scholar] [CrossRef] [PubMed]
  59. Lusis, A.J. Atherosclerosis. Nature 2000, 407, 233–241. [Google Scholar] [CrossRef]
  60. Borghetti, G.; von Lewinski, D.; Eaton, D.M.; Sourij, H.; Houser, S.R.; Wallner, M. Diabetic Cardiomyopathy: Current and Future Therapies. Beyond Glycemic Control. Front. Physiol. 2018, 9, 1514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Westermeier, F.; Riquelme, J.A.; Pavez, M.; Garrido, V.; Díaz, A.; Verdejo, H.E.; Castro, P.F.; García, L.; Lavandero, S. New Molecular Insights of Insulin in Diabetic Cardiomyopathy. Front. Physiol. 2016, 7, 125. [Google Scholar] [CrossRef] [Green Version]
  62. Dal Canto, E.; Ceriello, A.; Rydén, L.; Ferrini, M.; Hansen, T.B.; Schnell, O.; Standl, E.; Beulens, J.W. Diabetes as a Cardiovascular Risk Factor: An Overview of Global Trends of Macro and Micro Vascular Complications. Eur. J. Prev. Cardiol. 2019, 26, 25–32. [Google Scholar] [CrossRef] [Green Version]
  63. O’Donnell, M.J.; Xavier, D.; Liu, L.; Zhang, H.; Chin, S.L.; Rao-Melacini, P.; Rangarajan, S.; Islam, S.; Pais, P.; McQueen, M.J.; et al. Risk Factors for Ischaemic and Intracerebral Haemorrhagic Stroke in 22 Countries (the INTERSTROKE Study): A Case-Control Study. Lancet 2010, 376, 112–123. [Google Scholar] [CrossRef] [PubMed]
  64. Lau, L.; Lew, J.; Borschmann, K.; Thijs, V.; Ekinci, E.I. Prevalence of Diabetes and Its Effects on Stroke Outcomes: A Meta-analysis and Literature Review. J. Diabetes Investig. 2019, 10, 780–792. [Google Scholar] [CrossRef] [Green Version]
  65. Mankovsky, B.N.; Ziegler, D. Stroke in Patients with Diabetes Mellitus. Diabetes Metab. Res. Rev. 2004, 20, 268–287. [Google Scholar] [CrossRef]
  66. Tun, N.N.; Arunagirinathan, G.; Munshi, S.K.; Pappachan, J.M. Diabetes Mellitus and Stroke: A Clinical Update. World J. Diabetes 2017, 8, 235. [Google Scholar] [CrossRef]
  67. Zinman, B.; Wanner, C.; Lachin, J.M.; Fitchett, D.; Bluhmki, E.; Hantel, S.; Mattheus, M.; Devins, T.; Johansen, O.E.; Woerle, H.J.; et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N. Engl. J. Med. 2015, 373, 2117–2128. [Google Scholar] [CrossRef] [Green Version]
  68. Zhou, Y.; Huang, Y.; Ji, X.; Wang, X.; Shen, L.; Wang, Y. Pioglitazone for the Primary and Secondary Prevention of Cardiovascular and Renal Outcomes in Patients with or at High Risk of Type 2 Diabetes Mellitus: A Meta-Analysis. J. Clin. Endocrinol. Metab. 2020, 105, 1670–1681. [Google Scholar] [CrossRef] [PubMed]
  69. Committee, C.S. A Randomised, Blinded, Trial of Clopidogrel versus Aspirin in Patients at Risk of Ischaemic Events (CAPRIE). Lancet 1996, 348, 1329–1339. [Google Scholar] [CrossRef]
  70. Dormandy, J.A.; Charbonnel, B.; Eckland, D.J.; Erdmann, E.; Massi-Benedetti, M.; Moules, I.K.; Skene, A.M.; Tan, M.H.; Lefèbvre, P.J.; Murray, G.D.; et al. Secondary Prevention of Macrovascular Events in Patients with Type 2 Diabetes in the PROactive Study (PROspective PioglitAzone Clinical Trial in MacroVascular Events): A Randomised Controlled Trial. Lancet 2005, 366, 1279–1289. [Google Scholar] [CrossRef] [PubMed]
  71. Asmat, U.; Abad, K.; Ismail, K. Diabetes Mellitus and Oxidative Stress—A Concise Review. Saudi Pharm. J. 2016, 24, 547–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Bhatti, J.S.; Sehrawat, A.; Mishra, J.; Sidhu, I.S.; Navik, U.; Khullar, N.; Kumar, S.; Bhatti, G.K.; Reddy, P.H. Oxidative Stress in the Pathophysiology of Type 2 Diabetes and Related Complications: Current Therapeutics Strategies and Future Perspectives. Free Radic. Biol. Med. 2022, 184, 114–134. [Google Scholar] [CrossRef] [PubMed]
  73. Brownlee, M. Biochemistry and Molecular Cell Biology of Diabetic Complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef]
  74. Giacco, F.; Brownlee, M. Oxidative Stress and Diabetic Complications. Circ. Res. 2010, 107, 1058–1070. [Google Scholar] [CrossRef] [Green Version]
  75. Brownlee, M. The Pathobiology of Diabetic Complications: A Unifying Mechanism. Diabetes 2005, 54, 1615–1625. [Google Scholar] [CrossRef] [Green Version]
  76. Paul, S.; Ali, A.; Katare, R. Molecular Complexities Underlying the Vascular Complications of Diabetes Mellitus—A Comprehensive Review. J. Diabetes Its Complicat. 2020, 34, 107613. [Google Scholar] [CrossRef]
  77. Díaz-Flores, M.; Baiza-Gutman, L.A. Biochemical Mechanisms of Vascular Complications in Diabetes. In The Diabetes Textbook; Rodriguez-Saldana, J., Ed.; Springer International Publishing: Cham, Switzerland, 2019; pp. 695–707. ISBN 978-3-030-11814-3. [Google Scholar]
  78. Wu, M.Y.; Yiang, G.T.; Lai, T.T.; Li, C.J. The Oxidative Stress and Mitochondrial Dysfunction during the Pathogenesis of Diabetic Retinopathy. Oxidative Med. Cell. Longev. 2018, 2018, 3420187. [Google Scholar] [CrossRef]
  79. Xu, J.; Chen, L.J.; Yu, J.; Wang, H.J.; Zhang, F.; Liu, Q.; Wu, J. Involvement of Advanced Glycation End Products in the Pathogenesis of Diabetic Retinopathy. Cell. Physiol. Biochem. 2018, 48, 705–717. [Google Scholar] [CrossRef] [PubMed]
  80. Hayden, M.R.; Sowers, J.R.; Tyagi, S.C. The Central Role of Vascular Extracellular Matrix and Basement Membrane Remodeling in Metabolic Syndrome and Type 2 Diabetes: The Matrix Preloaded. Cardiovasc. Diabetol. 2005, 4, 9. [Google Scholar] [CrossRef] [PubMed]
  81. Koya, D.; King, G.L. Protein Kinase C Activation and the Development of Diabetic Complications. Diabetes 1998, 47, 859–866. [Google Scholar] [CrossRef]
  82. Mima, A.; Kitada, M.; Geraldes, P.; Li, Q.; Matsumoto, M.; Mizutani, K.; Qi, W.; Li, C.; Leitges, M.; Rask-Madsen, C. Glomerular VEGF Resistance Induced by PKCδ/SHP-1 Activation and Contribution to Diabetic Nephropathy. FASEB J. 2012, 26, 2963–2974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Buse, M.G. Hexosamines, Insulin Resistance, and the Complications of Diabetes: Current Status. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E1–E8. [Google Scholar] [CrossRef]
  84. Chatham, J.C.; Young, M.E.; Zhang, J. Role of O-Linked N-Acetylglucosamine (O-GlcNAc) Modification of Proteins in Diabetic Cardiovascular Complications. Curr. Opin. Pharmacol. 2020, 57, 1–12. [Google Scholar] [CrossRef]
  85. Liu, C.; Dong, W.; Li, J.; Kong, Y.; Ren, X. O-GlcNAc Modification and Its Role in Diabetic Retinopathy. Metabolites 2022, 12, 725. [Google Scholar] [CrossRef]
  86. Frey, T.; Antonetti, D.A. Alterations to the Blood-Retinal Barrier in Diabetes: Cytokines and Reactive Oxygen Species. Antioxid. Redox Signal. 2011, 15, 1271–1284. [Google Scholar] [CrossRef] [Green Version]
  87. Asnaghi, V.; Gerhardinger, C.; Hoehn, T.; Adeboje, A.; Lorenzi, M. A Role for the Polyol Pathway in the Early Neuroretinal Apoptosis and Glial Changes Induced by Diabetes in the Rat. Diabetes 2003, 52, 506–511. [Google Scholar] [CrossRef] [Green Version]
  88. Li, Q.R.; Wang, Z.; Zhou, W.; Fan, S.R.; Ma, R.; Xue, L.; Yang, L.; Li, Y.S.; Tan, H.L.; Shao, Q.H. Epalrestat Protects against Diabetic Peripheral Neuropathy by Alleviating Oxidative Stress and Inhibiting Polyol Pathway. Neural Regen. Res. 2016, 11, 345–351. [Google Scholar]
  89. Hotta, N.; Kawamori, R.; Fukuda, M.; Shigeta, Y. Long-Term Clinical Effects of Epalrestat, an Aldose Reductase Inhibitor, on Progression of Diabetic Neuropathy and Other Microvascular Complications: Multivariate Epidemiological Analysis Based on Patient Background Factors and Severity of Diabetic Neuropathy. Diabet. Med. A J. Br. Diabet. Assoc. 2012, 29, 1529–1533. [Google Scholar]
  90. Fridlyand, L.E.; Philipson, L.H. Does the Glucose-Dependent Insulin Secretion Mechanism Itself Cause Oxidative Stress in Pancreatic β-Cells? Diabetes 2004, 53, 1942–1948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Munteanu, C.; Rotariu, M.; Turnea, M.-A.; Anghelescu, A.; Albadi, I.; Dogaru, G.; Silișteanu, S.C.; Ionescu, E.V.; Firan, F.C.; Ionescu, A.M. Topical Reappraisal of Molecular Pharmacological Approaches to Endothelial Dysfunction in Diabetes Mellitus Angiopathy. Curr. Issues Mol. Biol. 2022, 44, 3378–3397. [Google Scholar] [CrossRef] [PubMed]
  92. Elumalai, S.; Karunakaran, U.; Moon, J.-S.; Won, K.-C. NADPH Oxidase (NOX) Targeting in Diabetes: A Special Emphasis on Pancreatic β-Cell Dysfunction. Cells 2021, 10, 1573. [Google Scholar] [CrossRef]
  93. Förstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef]
  94. Kim, W.-H.; Lee, J.W.; Suh, Y.H.; Lee, H.J.; Lee, S.H.; Oh, Y.K.; Gao, B.; Jung, M.H. AICAR Potentiates ROS Production Induced by Chronic High Glucose: Roles of AMPK in Pancreatic β-Cell Apoptosis. Cell. Signal. 2007, 19, 791–805. [Google Scholar] [CrossRef]
  95. Chau, G.C.; Im, D.U.; Kang, T.M.; Bae, J.M.; Kim, W.; Pyo, S.; Moon, E.-Y.; Um, S.H. MTOR Controls ChREBP Transcriptional Activity and Pancreatic β Cell Survival under Diabetic Stress. J. Cell Biol. 2017, 216, 2091–2105. [Google Scholar] [CrossRef] [Green Version]
  96. Wang, W.; Zhang, C. Targeting β-Cell Dedifferentiation and Transdifferentiation: Opportunities and Challenges. Endocr. Connect. 2021, 10, R213–R228. [Google Scholar] [CrossRef]
  97. Wang, F.; Wang, L.; Qu, C.; Chen, L.; Geng, Y.; Cheng, C.; Yu, S.; Wang, D.; Yang, L.; Meng, Z. Kaempferol Induces ROS-Dependent Apoptosis in Pancreatic Cancer Cells via TGM2-Mediated Akt/MTOR Signaling. BMC Cancer 2021, 21, 396. [Google Scholar] [CrossRef]
  98. Zhao, D.; Yang, J.; Yang, L. Insights for Oxidative Stress and MTOR Signaling in Myocardial Ischemia/Reperfusion Injury under Diabetes. Oxidative Med. Cell. Longev. 2017, 2017, 6437467. [Google Scholar] [CrossRef] [Green Version]
  99. James, D.E.; Stöckli, J.; Birnbaum, M.J. The Aetiology and Molecular Landscape of Insulin Resistance. Nat. Rev. Mol. Cell Biol. 2021, 22, 751–771. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, X.; Pan, J.; Liu, D.; Zhang, M.; Li, X.; Tian, J.; Liu, M.; Jin, T.; An, F. Nicorandil Alleviates Apoptosis in Diabetic Cardiomyopathy through PI3K/Akt Pathway. J. Cell. Mol. Med. 2019, 23, 5349–5359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Zhao, L.-G.; Li, P.-L.; Dai, Y.; Deng, J.-L.; Shan, M.-Y.; Chen, B.; Zhang, K.-B.; Guo, S.-D.; Xu, Z.-H. Mibefradil Alleviates High-Glucose–Induced Cardiac Hypertrophy by Inhibiting PI3K/Akt/MTOR-Mediated Autophagy. J. Cardiovasc. Pharmacol. 2020, 76, 246–254. [Google Scholar] [CrossRef] [PubMed]
  102. Paneni, F.; Costantino, S.; Cosentino, F. Role of Oxidative Stress in Endothelial Insulin Resistance. World J. Diabetes 2015, 6, 326. [Google Scholar] [CrossRef] [PubMed]
  103. Shamshoum, H.; Vlavcheski, F.; MacPherson, R.E.; Tsiani, E. Rosemary Extract Activates AMPK, Inhibits MTOR and Attenuates the High Glucose and High Insulin-Induced Muscle Cell Insulin Resistance. Appl. Physiol. Nutr. Metab. 2021, 46, 819–827. [Google Scholar] [CrossRef]
  104. Tang, Z.; Wang, P.; Dong, C.; Zhang, J.; Wang, X.; Pei, H. Oxidative Stress Signaling Mediated Pathogenesis of Diabetic Cardiomyopathy. Oxidative Med. Cell. Longev. 2022, 2022, 5913374. [Google Scholar] [CrossRef]
  105. Sies, H.; Jones, D.P. Reactive Oxygen Species (ROS) as Pleiotropic Physiological Signalling Agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  106. Volpe, C.M.O.; Villar-Delfino, P.H.; Dos Anjos, P.M.F.; Nogueira-Machado, J.A. Cellular Death, Reactive Oxygen Species (ROS) and Diabetic Complications. Cell Death Dis. 2018, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
  107. Ahsan, M.K.; Lekli, I.; Ray, D.; Yodoi, J.; Das, D.K. Redox Regulation of Cell Survival by the Thioredoxin Superfamily: An Implication of Redox Gene Therapy in the Heart. Available online: https://www.liebertpub.com/doi/10.1089/ars.2009.2683 (accessed on 12 November 2022).
  108. Santilli, F.; D’Ardes, D.; Davì, G. Oxidative Stress in Chronic Vascular Disease: From Prediction to Prevention. Vasc. Pharmacol. 2015, 74, 23–37. [Google Scholar] [CrossRef]
  109. Mirończuk-Chodakowska, I.; Witkowska, A.M.; Zujko, M.E. Endogenous Non-Enzymatic Antioxidants in the Human Body. Adv. Med. Sci. 2018, 63, 68–78. [Google Scholar] [CrossRef]
  110. Jung, K.-A.; Kwak, M.-K. The Nrf2 System as a Potential Target for the Development of Indirect Antioxidants. Molecules 2010, 15, 7266–7291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Seco-Cervera, M.; González-Cabo, P.; Pallardó, F.V.; Romá-Mateo, C.; García-Giménez, J.L. Thioredoxin and Glutaredoxin Systems as Potential Targets for the Development of New Treatments in Friedreich’s Ataxia. Antioxidants 2020, 9, 1257. [Google Scholar] [CrossRef] [PubMed]
  112. Kowluru, R.A.; Kern, T.S.; Engerman, R.L. Abnormalities of Retinal Metabolism in Diabetes or Experimental Galactosemia. IV. Antioxidant Defense System. Free Radic. Biol. Med. 1997, 22, 587–592. [Google Scholar] [CrossRef] [PubMed]
  113. Kowluru, R.A.; Mishra, M. Oxidative Stress, Mitochondrial Damage and Diabetic Retinopathy. Biochim. Et Biophys. Acta (BBA) Mol. Basis Dis. 2015, 1852, 2474–2483. [Google Scholar] [CrossRef]
  114. Lee, J.-Y.; Kim, M.; Oh, S.B.; Kim, H.-Y.; Kim, C.; Kim, T.-Y.; Park, Y.-H. Superoxide Dismutase 3 Prevents Early Stage Diabetic Retinopathy in Streptozotocin-Induced Diabetic Rat Model. PLoS ONE 2022, 17, e0262396. [Google Scholar] [CrossRef]
  115. Kwong-Han, K.; Zunaina, E.; Hanizasurana, H.; Che-Badariah, A.A.; Che-Maraina, C.H. Comparison of Catalase, Glutathione Peroxidase and Malondialdehyde Levels in Tears among Diabetic Patients with and without Diabetic Retinopathy. J. Diabetes Metab. Disord. 2022, 21, 681–688. [Google Scholar] [CrossRef]
  116. Zhao, J.-S.; Jin, H.-X.; Gao, J.-L.; Pu, C.; Zhang, P.; Huang, J.-J.; Cheng, L.; Feng, G. Serum Extracellular Superoxide Dismutase Is Associated with Diabetic Retinopathy Stage in Chinese Patients with Type 2 Diabetes Mellitus. Dis. Mrk. 2018, 2018, 8721379. [Google Scholar] [CrossRef]
  117. Lu, J.; Holmgren, A. The Thioredoxin Antioxidant System. Free Radic. Biol. Med. 2014, 66, 75–87. [Google Scholar] [CrossRef]
  118. Singh, L.P. Thioredoxin Interacting Protein (TXNIP) and Pathogenesis of Diabetic Retinopathy. J. Clin. Exp. Ophthalmol. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
  119. Cheng, Y.; Zhang, X.; Ma, F.; Sun, W.; Wang, W.; Yu, J.; Shi, Y.; Cai, L.; Xu, Z. The Role of Akt2 in the Protective Effect of Fenofibrate against Diabetic Nephropathy. Int. J. Biol. Sci. 2020, 16, 553–567. [Google Scholar] [CrossRef] [Green Version]
  120. Wei, P.Z.; Szeto, C.C. Mitochondrial Dysfunction in Diabetic Kidney Disease. Clin. Chim. Acta 2019, 496, 108–116. [Google Scholar] [CrossRef] [PubMed]
  121. Sakashita, M.; Tanaka, T.; Inagi, R. Metabolic Changes and Oxidative Stress in Diabetic Kidney Disease. Antioxidants 2021, 10, 1143. [Google Scholar] [CrossRef] [PubMed]
  122. Nezu, M.; Suzuki, N. Roles of Nrf2 in Protecting the Kidney from Oxidative Damage. Int. J. Mol. Sci. 2020, 21, 2951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Duni, A.; Liakopoulos, V.; Roumeliotis, S.; Peschos, D.; Dounousi, E. Oxidative Stress in the Pathogenesis and Evolution of Chronic Kidney Disease: Untangling Ariadne’s Thread. Int. J. Mol. Sci. 2019, 20, 3711. [Google Scholar] [CrossRef]
  124. Hong, Y.A.; Park, C.W. Catalytic Antioxidants in the Kidney. Antioxidants 2021, 10, 130. [Google Scholar] [CrossRef]
  125. Tucker, P.S.; Dalbo, V.J.; Han, T.; Kingsley, M.I. Clinical and Research Markers of Oxidative Stress in Chronic Kidney Disease. Biomarkers 2013, 18, 103–115. [Google Scholar] [CrossRef]
  126. Modlinger, P.S.; Wilcox, C.S.; Aslam, S. Nitric Oxide, Oxidative Stress, and Progression of Chronic Renal Failure. Semin. Nephrol. 2004, 24, 354–365. [Google Scholar] [CrossRef]
  127. Shurtz-Swirski, R.; Mashiach, E.; Kristal, B.; Shkolnik, T.; Shasha, S.M. Antioxidant Enzymes Activity in Polymorphonuclear Leukocytes in Chronic Renal Failure. Nephron 1995, 71, 176–179. [Google Scholar] [CrossRef]
  128. DeRubertis, F.R.; Craven, P.A.; Melhem, M.F.; Salah, E.M. Attenuation of Renal Injury in Db / Db Mice Overexpressing Superoxide Dismutase. Diabetes 2004, 53, 762–768. [Google Scholar] [CrossRef] [Green Version]
  129. Kitada, M.; Xu, J.; Ogura, Y.; Monno, I.; Koya, D. Manganese Superoxide Dismutase Dysfunction and the Pathogenesis of Kidney Disease. Front. Physiol. 2020, 11, 755. [Google Scholar] [CrossRef]
  130. Kuo, C.-W.; Shen, C.-J.; Tung, Y.-T.; Chen, H.-L.; Chen, Y.-H.; Chang, W.-H.; Cheng, K.-C.; Yang, S.-H.; Chen, C.-M. Extracellular Superoxide Dismutase Ameliorates Streptozotocin-Induced Rat Diabetic Nephropathy via Inhibiting the ROS/ERK1/2 Signaling. Life Sci. 2015, 135, 77–86. [Google Scholar] [CrossRef]
  131. Hong, Y.A.; Lim, J.H.; Kim, M.Y.; Kim, Y.; Park, H.S.; Kim, H.W.; Choi, B.S.; Chang, Y.S.; Kim, H.W.; Kim, T.-Y.; et al. Extracellular Superoxide Dismutase Attenuates Renal Oxidative Stress Through the Activation of Adenosine Monophosphate-Activated Protein Kinase in Diabetic Nephropathy. Antioxid. Redox Signal. 2018, 28, 1543–1561. [Google Scholar] [CrossRef] [PubMed]
  132. Liu, F.; Tran, S.; Sachetelli, S.; Filep, J.G.; Ingelfinger, J.R.; Chan, J.S.D. Catalase Overexpression Attenuates Angiotensinogen Expression and Apoptosis in Diabetic Mice. Kidney Int. 2007, 71, 912–923. [Google Scholar] [CrossRef] [Green Version]
  133. Brezniceanu, M.-L.; Liu, F.; Wei, C.-C.; Chénier, I.; Godin, N.; Zhang, S.-L.; Filep, J.G.; Ingelfinger, J.R.; Chan, J.S.D. Attenuation of Interstitial Fibrosis and Tubular Apoptosis in Db/Db Transgenic Mice Overexpressing Catalase in Renal Proximal Tubular Cells. Diabetes 2008, 57, 451–459. [Google Scholar] [CrossRef] [PubMed]
  134. Chiu, Y.I.-W.; Kuo, M.-C.; Kuo, H.-T.; Chang, J.-M.; Guh, J.-Y.; Lai, Y.-H.; Chen, H.-C. Alterations of Glomerular and Extracellular Levels of Glutathione Peroxidase in Patients and Experimental Rats with Diabetic Nephropathy. J. Lab. Clin. Med. 2005, 145, 181–186. [Google Scholar] [CrossRef]
  135. Wang, Y.; Chang, D.; Zhao, M.; Chen, M. Glutathione Peroxidase 4 Is a Predictor of Diabetic Kidney Disease Progression in Type 2 Diabetes Mellitus. Oxidative Med. Cell. Longev. 2022, 2022, 2948248. [Google Scholar] [CrossRef]
  136. Wang, X.; Fang, H.; Xu, G.; Yang, Y.; Xu, R.; Liu, Q.; Xue, X.; Liu, J.; Wang, H. Resveratrol Prevents Cognitive Impairment in Type 2 Diabetic Mice by Upregulating Nrf2 Expression and Transcriptional Level. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 1061–1075. [Google Scholar] [CrossRef] [Green Version]
  137. Sharma, A.; Tate, M.; Mathew, G.; Vince, J.E.; Ritchie, R.H.; de Haan, J.B. Oxidative Stress and NLRP3-Inflammasome Activity as Significant Drivers of Diabetic Cardiovascular Complications: Therapeutic Implications. Front. Physiol. 2018, 9, 114. [Google Scholar] [CrossRef] [Green Version]
  138. Tan, Y.; Zhang, Z.; Zheng, C.; Wintergerst, K.A.; Keller, B.B.; Cai, L. Mechanisms of Diabetic Cardiomyopathy and Potential Therapeutic Strategies: Preclinical and Clinical Evidence. Nat. Rev. Cardiol. 2020, 17, 585–607. [Google Scholar] [CrossRef]
  139. Bigagli, E.; Lodovici, M. Circulating Oxidative Stress Biomarkers in Clinical Studies on Type 2 Diabetes and Its Complications. Oxidative Med. Cell. Longev. 2019, 2019, 5953685. [Google Scholar] [CrossRef] [Green Version]
  140. Saisho, Y. Glycemic Variability and Oxidative Stress: A Link between Diabetes and Cardiovascular Disease? Int. J. Mol. Sci. 2014, 15, 18381–18406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Frustaci, A.; Kajstura, J.; Chimenti, C.; Jakoniuk, I.; Leri, A.; Maseri, A.; Nadal-Ginard, B.; Anversa, P. Myocardial Cell Death in Human Diabetes. Circ. Res. 2000, 87, 1123–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Shen, X.; Zheng, S.; Metreveli, N.S.; Epstein, P.N. Protection of Cardiac Mitochondria by Overexpression of MnSOD Reduces Diabetic Cardiomyopathy. Diabetes 2006, 55, 798–805. [Google Scholar] [CrossRef] [Green Version]
  143. Matsushima, S.; Kinugawa, S.; Ide, T.; Matsusaka, H.; Inoue, N.; Ohta, Y.; Yokota, T.; Sunagawa, K.; Tsutsui, H. Overexpression of Glutathione Peroxidase Attenuates Myocardial Remodeling and Preserves Diastolic Function in Diabetic Heart. Am. J. Physiol.-Heart Circ. Physiol. 2006, 291, H2237–H2245. [Google Scholar] [CrossRef] [PubMed]
  144. Ge, Z.-D.; Lian, Q.; Mao, X.; Xia, Z. Current Status and Challenges of NRF2 as a Potential Therapeutic Target for Diabetic Cardiomyopathy. Int. Heart J. 2019, 60, 512–520. [Google Scholar] [CrossRef] [Green Version]
  145. Jiang, T.; Sun, Q.; Chen, S. Oxidative Stress: A Major Pathogenesis and Potential Therapeutic Target of Antioxidative Agents in Parkinson’s Disease and Alzheimer’s Disease. Prog. Neurobiol. 2016, 147, 1–19. [Google Scholar] [CrossRef] [PubMed]
  146. Niedzielska, E.; Smaga, I.; Gawlik, M.; Moniczewski, A.; Stankowicz, P.; Pera, J.; Filip, M. Oxidative Stress in Neurodegenerative Diseases. Mol. Neurobiol. 2016, 53, 4094–4125. [Google Scholar] [CrossRef] [Green Version]
  147. Allen, C.L.; Bayraktutan, U. Oxidative Stress and Its Role in the Pathogenesis of Ischaemic Stroke. Int. J. Stroke 2009, 4, 461–470. [Google Scholar] [CrossRef]
  148. Cherubini, A.; Polidori, M.C.; Bregnocchi, M.; Pezzuto, S.; Cecchetti, R.; Ingegni, T.; di Iorio, A.; Senin, U.; Mecocci, P. Antioxidant Profile and Early Outcome in Stroke Patients. Stroke 2000, 31, 2295–2300. [Google Scholar] [CrossRef] [Green Version]
  149. Domínguez, C.; Delgado, P.; Vilches, A.; Martín-Gallán, P.; Ribó, M.; Santamarina, E.; Molina, C.; Corbeto, N.; Rodríguez-Sureda, V.; Rosell, A.; et al. Oxidative Stress after Thrombolysis-Induced Reperfusion in Human Stroke. Stroke 2010, 41, 653–660. [Google Scholar] [CrossRef] [Green Version]
  150. Cojocaru, I.M.; Cojocaru, M.; Sapira, V.; Ionescu, A. Evaluation of Oxidative Stress in Patients with Acute Ischemic Stroke. Rom. J. Intern. Med. 2013, 51, 97–106. [Google Scholar] [PubMed]
  151. Chekalina, N.I.; Kazakov, Y.M.; Mamontova, T.V.; Vesnina, L.E.; Kaidashev, I.P. Resveratrol More Effectively than Quercetin Reduces Endothelium Degeneration and Level of Necrosis Factor α in Patients with Coronary Artery Disease. Wiad. Lek. 2016, 69, 475–479. [Google Scholar] [PubMed]
  152. Csiszar, A.; Labinskyy, N.; Pinto, J.T.; Ballabh, P.; Zhang, H.; Losonczy, G.; Pearson, K.; de Cabo, R.; Pacher, P.; Zhang, C.; et al. Resveratrol Induces Mitochondrial Biogenesis in Endothelial Cells. Am. J. Physiol.-Heart Circ. Physiol. 2009, 297, H13–H20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Li, H.; Xia, N.; Förstermann, U. Cardiovascular Effects and Molecular Targets of Resveratrol. Nitric Oxide 2012, 26, 102–110. [Google Scholar] [CrossRef]
  154. Della-Morte, D.; Dave, K.R.; DeFazio, R.A.; Bao, Y.C.; Raval, A.P.; Perez-Pinzon, M.A. Resveratrol Pretreatment Protects Rat Brain from Cerebral Ischemic Damage via a Sirtuin 1–Uncoupling Protein 2 Pathway. Neuroscience 2009, 159, 993–1002. [Google Scholar] [CrossRef] [Green Version]
  155. Han, J.; Wang, M.; Jing, X.; Shi, H.; Ren, M.; Lou, H. (−)-Epigallocatechin Gallate Protects Against Cerebral Ischemia-Induced Oxidative Stress via Nrf2/ARE Signaling. Neurochem. Res. 2014, 39, 1292–1299. [Google Scholar] [CrossRef]
  156. Alfieri, A.; Srivastava, S.; Siow, R.C.M.; Modo, M.; Fraser, P.A.; Mann, G.E. Targeting the Nrf2-Keap1 Antioxidant Defence Pathway for Neurovascular Protection in Stroke: Nrf2-Mediated Neurovascular Protection. J. Physiol. 2011, 589, 4125–4136. [Google Scholar] [CrossRef]
  157. Biessels, G.J.; Koffeman, A.; Scheltens, P. Diabetes and Cognitive Impairment: Clinical Diagnosis and Brain Imaging in Patients Attending a Memory Clinic. J. Neurol. 2006, 253, 477–482. [Google Scholar] [CrossRef]
  158. Han, J.; Liu, X.; Li, Y.; Zhang, J.; Yu, H. Sirt1/Nrf2 Signalling Pathway Prevents Cognitive Impairment in Diabetic Rats through Anti-oxidative Stress Induced by MiRNA-23b-3p Expression. Mol. Med. Rep. 2018, 17, 8414–8422. [Google Scholar] [CrossRef] [Green Version]
  159. Donadelli, M.; Dando, I.; Fiorini, C.; Palmieri, M. UCP2, a Mitochondrial Protein Regulated at Multiple Levels. Cell. Mol. Life Sci. 2014, 71, 1171–1190. [Google Scholar] [CrossRef]
  160. Zhang, M.; Wang, L.; Wen, D.; Ren, C.; Chen, S.; Zhang, Z.; Hu, L.; Yu, Z.; Tombran-Tink, J.; Zhang, X.; et al. Neuroprotection of Retinal Cells by Caffeic Acid Phenylethyl Ester (CAPE) Is Mediated by Mitochondrial Uncoupling Protein UCP2. Neurochem. Int. 2021, 151, 105214. [Google Scholar] [CrossRef]
  161. Bhagani, H.; Nasser, S.A.; Dakroub, A.; El-Yazbi, A.F.; Eid, A.A.; Kobeissy, F.; Pintus, G.; Eid, A.H. The Mitochondria: A Target of Polyphenols in the Treatment of Diabetic Cardiomyopathy. Int. J. Mol. Sci. 2020, 21, 4962. [Google Scholar] [CrossRef]
  162. Fang, W.; Wang, C.; He, Y.; Zhou, Y.; Peng, X.; Liu, S. Resveratrol Alleviates Diabetic Cardiomyopathy in Rats by Improving Mitochondrial Function through PGC-1α Deacetylation. Acta Pharm. Sin. 2018, 39, 59–73. [Google Scholar] [CrossRef] [Green Version]
  163. Naoi, M.; Wu, Y.; Shamoto-Nagai, M.; Maruyama, W. Mitochondria in Neuroprotection by Phytochemicals: Bioactive Polyphenols Modulate Mitochondrial Apoptosis System, Function and Structure. Int. J. Mol. Sci. 2019, 20, 2451. [Google Scholar] [CrossRef]
  164. Zhu, H.; Toan, S.; Mui, D.; Zhou, H. Mitochondrial Quality Surveillance as a Therapeutic Target in Myocardial Infarction. Acta Physiol. 2021, 231, e13590. [Google Scholar] [CrossRef]
  165. Skemiene, K.; Liobikas, J.; Borutaite, V. Anthocyanins as Substrates for Mitochondrial Complex I—Protective Effect against Heart Ischemic Injury. FEBS J. 2015, 282, 963–971. [Google Scholar] [CrossRef]
  166. Huang, P.-C.; Wang, G.-J.; Fan, M.-J.; Asokan Shibu, M.; Liu, Y.-T.; Padma Viswanadha, V.; Lin, Y.-L.; Lai, C.-H.; Chen, Y.-F.; Liao, H.-E.; et al. Cellular Apoptosis and Cardiac Dysfunction in STZ-Induced Diabetic Rats Attenuated by Anthocyanins via Activation of IGFI-R/PI3K/Akt Survival Signaling. Environ. Toxicol. 2017, 32, 2471–2480. [Google Scholar] [CrossRef]
  167. Diao, J.; Zhao, H.; You, P.; You, H.; Wu, H.; Shou, X.; Cheng, G. Rosmarinic Acid Ameliorated Cardiac Dysfunction and Mitochondrial Injury in Diabetic Cardiomyopathy Mice via Activation of the SIRT1/PGC-1α Pathway. Biochem. Biophys. Res. Commun. 2021, 546, 29–34. [Google Scholar] [CrossRef]
  168. Diao, J.; Wei, J.; Yan, R.; Fan, G.; Lin, L.; Chen, M. Effects of Resveratrol on Regulation on UCP2 and Cardiac Function in Diabetic Rats. J. Physiol. Biochem. 2019, 75, 39–51. [Google Scholar] [CrossRef]
  169. Zheng, J.; Conrad, M. The Metabolic Underpinnings of Ferroptosis. Cell Metab. 2020, 32, 920–937. [Google Scholar] [CrossRef]
  170. Liu, J.; Li, Q.; Yang, Y.; Ma, L. Iron Metabolism and Type 2 Diabetes Mellitus: A Meta-Analysis and Systematic Review. J. Diabetes Investig. 2020, 11, 946–955. [Google Scholar] [CrossRef]
  171. Chaudhary, K.; Promsote, W.; Ananth, S.; Veeranan-Karmegam, R.; Tawfik, A.; Arjunan, P.; Martin, P.; Smith, S.B.; Thangaraju, M.; Kisselev, O.; et al. Iron Overload Accelerates the Progression of Diabetic Retinopathy in Association with Increased Retinal Renin Expression. Sci. Rep. 2018, 8, 3025. [Google Scholar] [CrossRef] [Green Version]
  172. Zhao, L.; Bartnikas, T.; Chu, X.; Klein, J.; Yun, C.; Srinivasan, S.; He, P. Hyperglycemia Promotes Microvillus Membrane Expression of DMT1 in Intestinal Epithelial Cells in a PKCα-dependent Manner. FASEB J. 2019, 33, 3549–3561. [Google Scholar] [CrossRef]
  173. Sha, W.; Hu, F.; Xi, Y.; Chu, Y.; Bu, S. Mechanism of Ferroptosis and Its Role in Type 2 Diabetes Mellitus. J. Diabetes Res. 2021, 2021, 9999612. [Google Scholar] [CrossRef]
  174. Jakaria, M.; Belaidi, A.A.; Bush, A.I.; Ayton, S. Ferroptosis as a Mechanism of Neurodegeneration in Alzheimer’s Disease. J. Neurochem. 2021, 159, 804–825. [Google Scholar] [CrossRef]
  175. Chandra, K.; Jain, V.; Jabin, A.; Dwivedi, S.; Joshi, S.; Ahmad, S.; Jain, S.K. Effect of Cichorium Intybus Seeds Supplementation on the Markers of Glycemic Control, Oxidative Stress, Inflammation, and Lipid Profile in Type 2 Diabetes Mellitus: A Randomized, Double-Blind Placebo Study. Phytother. Res. 2020, 34, 1609–1618. [Google Scholar] [CrossRef]
  176. Yang, X.-D.; Yang, Y.-Y. Ferroptosis as a Novel Therapeutic Target for Diabetes and Its Complications. Front. Endocrinol. 2022, 13, 853822. [Google Scholar] [CrossRef]
  177. Fernández-del-Río, L.; Nag, A.; Gutiérrez Casado, E.; Ariza, J.; Awad, A.M.; Joseph, A.I.; Kwon, O.; Verdin, E.; de Cabo, R.; Schneider, C.; et al. Kaempferol Increases Levels of Coenzyme Q in Kidney Cells and Serves as a Biosynthetic Ring Precursor. Free Radic. Biol. Med. 2017, 110, 176–187. [Google Scholar] [CrossRef]
  178. Xie, L.X.; Williams, K.J.; He, C.H.; Weng, E.; Khong, S.; Rose, T.E.; Kwon, O.; Bensinger, S.J.; Marbois, B.N.; Clarke, C.F. Resveratrol and Para-Coumarate Serve as Ring Precursors for Coenzyme Q Biosynthesis [S]. J. Lipid Res. 2015, 56, 909–919. [Google Scholar] [CrossRef] [Green Version]
  179. Meza-Torres, C.; Hernández-Camacho, J.D.; Cortés-Rodríguez, A.B.; Fang, L.; Bui Thanh, T.; Rodríguez-Bies, E.; Navas, P.; López-Lluch, G. Resveratrol Regulates the Expression of Genes Involved in CoQ Synthesis in Liver in Mice Fed with High Fat Diet. Antioxidants 2020, 9, 431. [Google Scholar] [CrossRef]
  180. Al-Numair, K.S.; Chandramohan, G.; Veeramani, C.; Alsaif, M.A. Ameliorative Effect of Kaempferol, a Flavonoid, on Oxidative Stress in Streptozotocin-Induced Diabetic Rats. Redox Rep. 2015, 20, 198–209. [Google Scholar] [CrossRef]
  181. Yu, Q.; Lee, C.F.; Wang, W.; Karamanlidis, G.; Kuroda, J.; Matsushima, S.; Sadoshima, J.; Tian, R. Elimination of NADPH Oxidase Activity Promotes Reductive Stress and Sensitizes the Heart to Ischemic Injury. J. Am. Heart Assoc. 2014, 3, e000555. [Google Scholar] [CrossRef] [Green Version]
  182. Zhang, Y.; Murugesan, P.; Huang, K.; Cai, H. NADPH Oxidases and Oxidase Crosstalk in Cardiovascular Diseases: Novel Therapeutic Targets. Nat. Rev. Cardiol. 2020, 17, 170–194. [Google Scholar] [CrossRef]
  183. Tan, H.; Chen, J.; Li, Y.; Li, Y.; Zhong, Y.; Li, G.; Liu, L.; Li, Y. Glabridin, a Bioactive Component of Licorice, Ameliorates Diabetic Nephropathy by Regulating Ferroptosis and the VEGF/Akt/ERK Pathways. Mol. Med. 2022, 28, 58. [Google Scholar] [CrossRef]
  184. Huang, D.; Shen, P.; Wang, C.; Gao, J.; Ye, C.; Wu, F. Calycosin Plays a Protective Role in Diabetic Kidney Disease through the Regulation of Ferroptosis. Pharm. Biol. 2022, 60, 990–996. [Google Scholar] [CrossRef]
  185. He, Y.; Wang, S.; Sun, H.; Li, Y.; Feng, J. Naringenin Ameliorates Myocardial Injury in STZ-Induced Diabetic Mice by Reducing Oxidative Stress, Inflammation and Apoptosis via Regulating the Nrf2 and NF-ΚB Signaling Pathways. Front. Cardiovasc. Med. 2022, 9, 946766. [Google Scholar] [CrossRef]
  186. Tang, X.; Li, X.; Zhang, D.; Han, W. Astragaloside-IV Alleviates High Glucose-Induced Ferroptosis in Retinal Pigment Epithelial Cells by Disrupting the Expression of MiR-138-5p/Sirt1/Nrf2. Bioengineered 2022, 13, 8238–8253. [Google Scholar] [CrossRef]
  187. Zhou, Y. The Protective Effects of Cryptochlorogenic Acid on β-Cells Function in Diabetes in Vivo and Vitro via Inhibition of Ferroptosis. Diabetes Metab. Syndr. Obes. 2020, 13, 1921–1931. [Google Scholar] [CrossRef]
  188. Li, D.; Jiang, C.; Mei, G.; Zhao, Y.; Chen, L.; Liu, J.; Tang, Y.; Gao, C.; Yao, P. Quercetin Alleviates Ferroptosis of Pancreatic β Cells in Type 2 Diabetes. Nutrients 2020, 12, 2954. [Google Scholar] [CrossRef]
  189. Singh, C.K.; Chhabra, G.; Ndiaye, M.A.; Garcia-Peterson, L.M.; Mack, N.J.; Ahmad, N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid. Redox Signal. 2018, 28, 643–661. [Google Scholar] [CrossRef]
  190. Liu, J.; Chen, S.; Biswas, S.; Nagrani, N.; Chu, Y.; Chakrabarti, S.; Feng, B. Glucose-Induced Oxidative Stress and Accelerated Aging in Endothelial Cells Are Mediated by the Depletion of Mitochondrial SIRTs. Physiol. Rep. 2020, 8, e14331. [Google Scholar] [CrossRef]
  191. Wang, X.; Meng, L.; Zhao, L.; Wang, Z.; Liu, H.; Liu, G.; Guan, G. Resveratrol Ameliorates Hyperglycemia-Induced Renal Tubular Oxidative Stress Damage via Modulating the SIRT1/FOXO3a Pathway. Diabetes Res. Clin. Pract. 2017, 126, 172–181. [Google Scholar] [CrossRef] [PubMed]
  192. Zhang, T.; Chi, Y.; Kang, Y.; Lu, H.; Niu, H.; Liu, W.; Li, Y. Resveratrol Ameliorates Podocyte Damage in Diabetic Mice via SIRT1/PGC-1α Mediated Attenuation of Mitochondrial Oxidative Stress. J. Cell. Physiol. 2019, 234, 5033–5043. [Google Scholar] [CrossRef] [PubMed]
  193. Bernal-Ramirez, J.; Silva-Platas, C.; Jerjes-Sánchez, C.; Chapoy-Villanueva, H.; Ramos, M.; Garcia, N.; García-Rivas, G. Resveratrol Preserves Mitochondrial Function By Sirt3 Activation in a Right Ventricle Dysfunction Model. J. Card. Fail. 2022, 28, S82. [Google Scholar] [CrossRef]
  194. Kim, A.; Lee, W.; Yun, J.-M. Luteolin and Fisetin Suppress Oxidative Stress by Modulating Sirtuins and Forkhead Box O3a Expression under in vitro Diabetic Conditions. Nutr. Res. Pract. 2017, 11, 430–434. [Google Scholar] [CrossRef] [Green Version]
  195. Hu, H.-C.; Lei, Y.-H.; Zhang, W.-H.; Luo, X.-Q. Antioxidant and Anti-Inflammatory Properties of Resveratrol in Diabetic Nephropathy: A Systematic Review and Meta-Analysis of Animal Studies. Front. Pharm. 2022, 13, 841818. [Google Scholar] [CrossRef] [PubMed]
  196. Xian, Y.; Gao, Y.; Lv, W.; Ma, X.; Hu, J.; Chi, J.; Wang, W.; Wang, Y. Resveratrol Prevents Diabetic Nephropathy by Reducing Chronic Inflammation and Improving the Blood Glucose Memory Effect in Non-Obese Diabetic Mice. Naunyn-Schmiedeberg’s Arch. Pharm. 2020, 393, 2009–2017. [Google Scholar] [CrossRef] [PubMed]
  197. Hayes, J.D.; Dinkova-Kostova, A.T. The Nrf2 Regulatory Network Provides an Interface between Redox and Intermediary Metabolism. Trends Biochem. Sci. 2014, 39, 199–218. [Google Scholar] [CrossRef]
  198. Nguyen, P.M.; Park, M.S.; Chow, M.; Chang, J.H.; Wrischnik, L.; Chan, W.K. Benzo[a]Pyrene Increases the Nrf2 Content by Downregulating the Keap1 Message. Toxicol. Sci. 2010, 116, 549–561. [Google Scholar] [CrossRef] [Green Version]
  199. Rada, P.; Rojo, A.I.; Evrard-Todeschi, N.; Innamorato, N.G.; Cotte, A.; Jaworski, T.; Tobón-Velasco, J.C.; Devijver, H.; García-Mayoral, M.F.; Van Leuven, F.; et al. Structural and Functional Characterization of Nrf2 Degradation by the Glycogen Synthase Kinase 3/β-TrCP Axis. Mol. Cell. Biol. 2012, 32, 3486–3499. [Google Scholar] [CrossRef] [Green Version]
  200. Wang, H.; Liu, K.; Geng, M.; Gao, P.; Wu, X.; Hai, Y.; Li, Y.; Li, Y.; Luo, L.; Hayes, J.D.; et al. RXRα Inhibits the NRF2-ARE Signaling Pathway through a Direct Interaction with the Neh7 Domain of NRF2. Cancer Res. 2013, 73, 3097–3108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Motohashi, H.; Katsuoka, F.; Engel, J.D.; Yamamoto, M. Small Maf Proteins Serve as Transcriptional Cofactors for Keratinocyte Differentiation in the Keap1–Nrf2 Regulatory Pathway. Proc. Natl. Acad. Sci. USA 2004, 101, 6379–6384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Kopacz, A.; Kloska, D.; Forman, H.J.; Jozkowicz, A.; Grochot-Przeczek, A. Beyond Repression of Nrf2: An Update on Keap1. Free Radic. Biol. Med. 2020, 157, 63–74. [Google Scholar] [CrossRef] [PubMed]
  203. Tsai, C.Y.; Wang, C.C.; Lai, T.-I.; Tsu, H.N.; Wang, C.H.; Liang, H.Y.; Kuo, W.W. Antioxidant Effects of Diallyl Trisulfide on High Glucose-Induced Apoptosis Are Mediated by the PI3K/Akt-Dependent Activation of Nrf2 in Cardiomyocytes. Int. J. Cardiol. 2013, 168, 1286–1297. [Google Scholar] [CrossRef] [PubMed]
  204. Mohan, T.; Narasimhan, K.K.S.; Ravi, D.B.; Velusamy, P.; Chandrasekar, N.; Chakrapani, L.N.; Srinivasan, A.; Karthikeyan, P.; Kannan, P.; Tamilarasan, B.; et al. Role of Nrf2 Dysfunction in the Pathogenesis of Diabetic Nephropathy: Therapeutic Prospect of Epigallocatechin-3-Gallate. Free Radic. Biol. Med. 2020, 160, 227–238. [Google Scholar] [CrossRef]
  205. Nezu, M.; Souma, T.; Yu, L.; Suzuki, T.; Saigusa, D.; Ito, S.; Suzuki, N.; Yamamoto, M. Transcription Factor Nrf2 Hyperactivation in Early-Phase Renal Ischemia-Reperfusion Injury Prevents Tubular Damage Progression. Kidney Int. 2017, 91, 387–401. [Google Scholar] [CrossRef]
  206. Sun, W.; Liu, X.; Zhang, H.; Song, Y.; Li, T.; Liu, X.; Liu, Y.; Guo, L.; Wang, F.; Yang, T.; et al. Epigallocatechin Gallate Upregulates NRF2 to Prevent Diabetic Nephropathy via Disabling KEAP1. Free Radic. Biol. Med. 2017, 108, 840–857. [Google Scholar] [CrossRef]
  207. ALTamimi, J.Z.; AlFaris, N.A.; Alshammari, G.M.; Alagal, R.I.; Aljabryn, D.H.; Aldera, H.; Alrfaei, B.M.; Alkhateeb, M.A.; Yahya, M.A. Ellagic Acid Protects against Diabetic Nephropathy in Rats by Regulating the Transcription and Activity of Nrf2. J. Funct. Foods 2021, 79, 104397. [Google Scholar] [CrossRef]
  208. Alaofi, A.L. Sinapic Acid Ameliorates the Progression of Streptozotocin (STZ)-Induced Diabetic Nephropathy in Rats via NRF2/HO-1 Mediated Pathways. Front. Pharmacol. 2020, 11, 1119. [Google Scholar] [CrossRef]
  209. Zhu, X.; Chen, Y.; Chen, Q.; Yang, H.; Xie, X. Astaxanthin Promotes Nrf2/ARE Signaling to Alleviate Renal Fibronectin and Collagen IV Accumulation in Diabetic Rats. J. Diabetes Res. 2018, 2018, e6730315. [Google Scholar] [CrossRef]
  210. Zhang, L.; Chen, Z.; Gong, W.; Zou, Y.; Xu, F.; Chen, L.; Huang, H. Paeonol Ameliorates Diabetic Renal Fibrosis Through Promoting the Activation of the Nrf2/ARE Pathway via Up-Regulating Sirt1. Front. Pharmacol. 2018, 9, 512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Chen, Y.-J.; Kong, L.; Tang, Z.-Z.; Zhang, Y.-M.; Liu, Y.; Wang, T.-Y.; Liu, Y.-W. Hesperetin Ameliorates Diabetic Nephropathy in Rats by Activating Nrf2/ARE/Glyoxalase 1 Pathway. Biomed. Pharmacother. 2019, 111, 1166–1175. [Google Scholar] [CrossRef] [PubMed]
  212. Shi, Q.; Cheng, Y.; Dong, X.; Zhang, M.; Pei, C.; Zhang, M. Effects of Rhaponticin on Retinal Oxidative Stress and Inflammation in Diabetes through NRF2/HO-1/NF-κB Signalling. J. Biochem. Mol. Toxicol. 2020, 34, e22568. [Google Scholar] [CrossRef] [PubMed]
  213. Xie, T.; Chen, X.; Chen, W.; Huang, S.; Peng, X.; Tian, L.; Wu, X.; Huang, Y. Curcumin Is a Potential Adjuvant to Alleviates Diabetic Retinal Injury via Reducing Oxidative Stress and Maintaining Nrf2 Pathway Homeostasis. Front. Pharmacol. 2021, 12, 796565. [Google Scholar] [CrossRef]
  214. Bucolo, C.; Drago, F.; Maisto, R.; Romano, G.L.; D’Agata, V.; Maugeri, G.; Giunta, S. Curcumin Prevents High Glucose Damage in Retinal Pigment Epithelial Cells through ERK1/2-mediated Activation of the Nrf2/HO-1 Pathway. J. Cell. Physiol. 2019, 234, 17295–17304. [Google Scholar] [CrossRef]
  215. Song, Y.; Huang, L.; Yu, J. Effects of Blueberry Anthocyanins on Retinal Oxidative Stress and Inflammation in Diabetes through Nrf2/HO-1 Signaling. J. Neuroimmunol. 2016, 301, 1–6. [Google Scholar] [CrossRef]
  216. Huang, W.; Yan, Z.; Li, D.; Ma, Y.; Zhou, J.; Sui, Z. Antioxidant and Anti-Inflammatory Effects of Blueberry Anthocyanins on High Glucose-Induced Human Retinal Capillary Endothelial Cells. Oxidative Med. Cell. Longev. 2018, 2018, e1862462. [Google Scholar] [CrossRef]
  217. Herrera-Bravo, J.; Beltrán, J.F.; Huard, N.; Saavedra, K.; Saavedra, N.; Alvear, M.; Lanas, F.; Salazar, L.A. Anthocyanins Found in Pinot Noir Waste Induce Target Genes Related to the Nrf2 Signalling in Endothelial Cells. Antioxidants 2022, 11, 1239. [Google Scholar] [CrossRef]
  218. Millán, I.; Desco, M.d.C.; Torres-Cuevas, I.; Pérez, S.; Pulido, I.; Mena-Mollá, S.; Mataix, J.; Asensi, M.; Ortega, Á.L. Pterostilbene Prevents Early Diabetic Retinopathy Alterations in a Rabbit Experimental Model. Nutrients 2019, 12, 82. [Google Scholar] [CrossRef] [Green Version]
  219. Matos, A.L.; Bruno, D.F.; Ambrósio, A.F.; Santos, P.F. The Benefits of Flavonoids in Diabetic Retinopathy. Nutrients 2020, 12, 3169. [Google Scholar] [CrossRef]
  220. Liang, F.; Wang, J.; Zhu, X.; Wang, Z.; Zheng, J.; Sun, Z.; Xu, S.; Zhang, J.; Zhou, J.; Shi, L. Melatonin Alleviates Neuronal Damage After Intracerebral Hemorrhage in Hyperglycemic Rats. Drug Des. Dev. Ther. 2020, 14, 2573–2584. [Google Scholar] [CrossRef] [PubMed]
  221. Sandireddy, R.; Yerra, V.G.; Komirishetti, P.; Areti, A.; Kumar, A. Fisetin Imparts Neuroprotection in Experimental Diabetic Neuropathy by Modulating Nrf2 and NF-ΚB Pathways. Cell. Mol. Neurobiol. 2016, 36, 883–892. [Google Scholar] [CrossRef] [PubMed]
  222. Bachewal, P.; Gundu, C.; Yerra, V.G.; Kalvala, A.K.; Areti, A.; Kumar, A. Morin Exerts Neuroprotection via Attenuation of ROS Induced Oxidative Damage and Neuroinflammation in Experimental Diabetic Neuropathy: Pharmacological Effects of Morin in Diabetic Neuropathy. BioFactors 2018, 44, 109–122. [Google Scholar] [CrossRef] [PubMed]
  223. Mittal, R.; Kumar, A.; Singh, D.P.; Bishnoi, M.; Nag, T.C. Ameliorative Potential of Rutin in Combination with Nimesulide in STZ Model of Diabetic Neuropathy: Targeting Nrf2/HO-1/NF-KB and COX Signalling Pathway. Inflammopharmacol 2018, 26, 755–768. [Google Scholar] [CrossRef]
  224. Malakoti, F.; Mohammadi, E.; Akbari Oryani, M.; Shanebandi, D.; Yousefi, B.; Salehi, A.; Asemi, Z. Polyphenols Target MiRNAs as a Therapeutic Strategy for Diabetic Complications. Crit. Rev. Food Sci. Nutr. 2022, 1–17. [Google Scholar] [CrossRef]
  225. Yehuda, I.; Madar, Z.; Leikin-Frenkel, A.; Szuchman-Sapir, A.; Magzal, F.; Markman, G.; Tamir, S. Glabridin, an Isoflavan from Licorice Root, Upregulates Paraoxonase 2 Expression under Hyperglycemia and Protects It from Oxidation. Mol. Nutr. Food Res. 2016, 60, 287–299. [Google Scholar] [CrossRef]
  226. Roxo, D.F.; Arcaro, C.A.; Gutierres, V.O.; Costa, M.C.; Oliveira, J.O.; Lima, T.F.O.; Assis, R.P.; Brunetti, I.L.; Baviera, A.M. Curcumin Combined with Metformin Decreases Glycemia and Dyslipidemia, and Increases Paraoxonase Activity in Diabetic Rats. Diabetol. Metab. Syndr. 2019, 11, 33. [Google Scholar] [CrossRef] [Green Version]
  227. Yehuda, I.; Madar, Z.; Leikin-Frenkel, A.; Tamir, S. Glabridin, an Isoflavan from Licorice Root, Downregulates INOS Expression and Activity under High-Glucose Stress and Inflammation. Mol. Nutr. Food Res. 2015, 59, 1041–1052. [Google Scholar] [CrossRef]
  228. An, X.; Zhang, Y.; Cao, Y.; Chen, J.; Qin, H.; Yang, L. Punicalagin Protects Diabetic Nephropathy by Inhibiting Pyroptosis Based on TXNIP/NLRP3 Pathway. Nutrients 2020, 12, 1516. [Google Scholar] [CrossRef]
  229. Kotewicz, M.; Krauze-Baranowska, M.; Daca, A.; Płoska, A.; Godlewska, S.; Kalinowski, L.; Lewko, B. Urolithins Modulate the Viability, Autophagy, Apoptosis, and Nephrin Turnover in Podocytes Exposed to High Glucose. Cells 2022, 11, 2471. [Google Scholar] [CrossRef]
  230. Iatcu, C.O.; Steen, A.; Covasa, M. Gut Microbiota and Complications of Type-2 Diabetes. Nutrients 2022, 14, 166. [Google Scholar] [CrossRef] [PubMed]
  231. Yang, K.; Jian, S.; Guo, D.; Wen, C.; Xin, Z.; Zhang, L.; Kuang, T.; Wen, J.; Yin, Y.; Deng, B. Fecal Microbiota and Metabolomics Revealed the Effect of Long-Term Consumption of Gallic Acid on Canine Lipid Metabolism and Gut Health. Food Chem. X 2022, 15, 100377. [Google Scholar] [CrossRef] [PubMed]
  232. Jiang, Z.; Zhuo, L.; He, Y.; Fu, Y.; Shen, L.; Xu, F.; Gou, W.; Miao, Z.; Shuai, M.; Liang, Y.; et al. The Gut Microbiota-Bile Acid Axis Links the Positive Association between Chronic Insomnia and Cardiometabolic Diseases. Nat. Commun. 2022, 13, 3002. [Google Scholar] [CrossRef]
  233. Martín-Peláez, S.; Mosele, J.I.; Pizarro, N.; Farràs, M.; de la Torre, R.; Subirana, I.; Pérez-Cano, F.J.; Castañer, O.; Solà, R.; Fernandez-Castillejo, S.; et al. Effect of Virgin Olive Oil and Thyme Phenolic Compounds on Blood Lipid Profile: Implications of Human Gut Microbiota. Eur. J. Nutr. 2017, 56, 119–131. [Google Scholar] [CrossRef] [PubMed]
  234. Zhang, Y.; Liu, M.; Zhang, Y.; Tian, M.; Chen, P.; Lan, Y.; Zhou, B. Urolithin A Alleviates Acute Kidney Injury Induced by Renal Ischemia Reperfusion through the P62-Keap1-Nrf2 Signaling Pathway. Phytother. Res. 2022, 36, 984–995. [Google Scholar] [CrossRef] [PubMed]
  235. Gerasimova, E.L.; Gazizullina, E.R.; Borisova, M.V.; Igdisanova, D.I.; Nikiforov, E.A.; Moseev, T.D.; Varaksin, M.V.; Chupakhin, O.N.; Charushin, V.N.; Ivanova, A.V. Design and Antioxidant Properties of Bifunctional 2H-Imidazole-Derived Phenolic Compounds—A New Family of Effective Inhibitors for Oxidative Stress-Associated Destructive Processes. Molecules 2021, 26, 6534. [Google Scholar] [CrossRef] [PubMed]
  236. Yates, M.S.; Tauchi, M.; Katsuoka, F.; Flanders, K.C.; Liby, K.T.; Honda, T.; Gribble, G.W.; Johnson, D.A.; Johnson, J.A.; Burton, N.C.; et al. Pharmacodynamic Characterization of Chemopreventive Triterpenoids as Exceptionally Potent Inducers of Nrf2-Regulated Genes. Mol. Cancer Ther. 2007, 6, 154–162. [Google Scholar] [CrossRef] [Green Version]
  237. Rahnasto-Rilla, M.; Tyni, J.; Huovinen, M.; Jarho, E.; Kulikowicz, T.; Ravichandran, S.; Bohr, V.A.; Ferrucci, L.; Lahtela-Kakkonen, M.; Moaddel, R. Natural Polyphenols as Sirtuin 6 Modulators. Sci. Rep. 2018, 8, 4163. [Google Scholar] [CrossRef] [Green Version]
  238. Kancheva, V.D.; Dettori, M.A.; Fabbri, D.; Alov, P.; Angelova, S.E.; Slavova-Kazakova, A.K.; Carta, P.; Menshov, V.A.; Yablonskaya, O.I.; Trofimov, A.V.; et al. Natural Chain-Breaking Antioxidants and Their Synthetic Analogs as Modulators of Oxidative Stress. Antioxidants 2021, 10, 624. [Google Scholar] [CrossRef]
  239. Rodriguez-Mateos, A.; Pino-García, R.D.; George, T.W.; Vidal-Diez, A.; Heiss, C.; Spencer, J.P.E. Impact of Processing on the Bioavailability and Vascular Effects of Blueberry (Poly)Phenols. Mol. Nutr. Food Res. 2014, 58, 1952–1961. [Google Scholar] [CrossRef]
  240. Abukhabta, S.; Khalil Ghawi, S.; Karatzas, K.A.; Charalampopoulos, D.; McDougall, G.; Allwood, J.W.; Verrall, S.; Lavery, S.; Latimer, C.; Pourshahidi, L.K.; et al. Sulforaphane-Enriched Extracts from Glucoraphanin-Rich Broccoli Exert Antimicrobial Activity against Gut Pathogens in Vitro and Innovative Cooking Methods Increase in Vivo Intestinal Delivery of Sulforaphane. Eur. J. Nutr. 2021, 60, 1263–1276. [Google Scholar] [CrossRef] [PubMed]
  241. Hidalgo-Liberona, N.; González-Domínguez, R.; Vegas, E.; Riso, P.; Del Bo’, C.; Bernardi, S.; Peron, G.; Guglielmetti, S.; Gargari, G.; Kroon, P.A.; et al. Increased Intestinal Permeability in Older Subjects Impacts the Beneficial Effects of Dietary Polyphenols by Modulating Their Bioavailability. J. Agric. Food Chem. 2020, 68, 12476–12484. [Google Scholar] [CrossRef] [PubMed]
  242. Williamson, G.; Kay, C.D.; Crozier, A. The Bioavailability, Transport, and Bioactivity of Dietary Flavonoids: A Review from a Historical Perspective. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1054–1112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Manach, C.; Milenkovic, D.; Van de Wiele, T.; Rodriguez-Mateos, A.; de Roos, B.; Garcia-Conesa, M.T.; Landberg, R.; Gibney, E.R.; Heinonen, M.; Tomás-Barberán, F.; et al. Addressing the Inter-Individual Variation in Response to Consumption of Plant Food Bioactives: Towards a Better Understanding of Their Role in Healthy Aging and Cardiometabolic Risk Reduction. Mol. Nutr. Food Res. 2017, 61, 1600557. [Google Scholar] [CrossRef] [PubMed]
  244. Polia, F.; Pastor-Belda, M.; Martínez-Blázquez, A.; Horcajada, M.-N.; Tomás-Barberán, F.A.; García-Villalba, R. Technological and Biotechnological Processes To Enhance the Bioavailability of Dietary (Poly)Phenols in Humans. J. Agric. Food Chem. 2022, 70, 2092–2107. [Google Scholar] [CrossRef]
  245. Iglesias-Aguirre, C.E.; Vallejo, F.; Beltrán, D.; Aguilar-Aguilar, E.; Puigcerver, J.; Alajarín, M.; Berná, J.; Selma, M.V.; Espín, J.C. Lunularin Producers versus Non-Producers: Novel Human Metabotypes Associated with the Metabolism of Resveratrol by the Gut Microbiota. J. Agric. Food Chem. 2022, 70, 10521–10531. [Google Scholar] [CrossRef]
  246. Cortés-Martín, A.; Selma, M.V.; Tomás-Barberán, F.A.; González-Sarrías, A.; Espín, J.C. Where to Look into the Puzzle of Polyphenols and Health? The Postbiotics and Gut Microbiota Associated with Human Metabotypes. Mol. Nutr. Food Res. 2020, 64, 1900952. [Google Scholar] [CrossRef]
  247. Frankenfeld, C.L. Cardiometabolic Risk and Gut Microbial Phytoestrogen Metabolite Phenotypes. Mol. Nutr. Food Res. 2017, 61, 1500900. [Google Scholar] [CrossRef]
  248. Cortés-Martín, A.; Iglesias-Aguirre, C.E.; Meoro, A.; Selma, M.V.; Espín, J.C. Pharmacological Therapy Determines the Gut Microbiota Modulation by a Pomegranate Extract Nutraceutical in Metabolic Syndrome: A Randomized Clinical Trial. Mol. Nutr. Food Res. 2021, 65, 2001048. [Google Scholar] [CrossRef]
  249. Islam, F.; Khadija, J.F.; Islam, M.R.; Shohag, S.; Mitra, S.; Alghamdi, S.; Babalghith, A.O.; Theyab, A.; Rahman, M.T.; Akter, A.; et al. Investigating Polyphenol Nanoformulations for Therapeutic Targets against Diabetes Mellitus. Evid.-Based Complement. Altern. Med. 2022, 2022, e5649156. [Google Scholar] [CrossRef]
  250. de Oliveira, W.Q.; Neri-Numa, I.A.; Arruda, H.S.; McClements, D.J.; Pastore, G.M. Encapsulation of Flavonoids in Foods for Diabetics: The Emerging Paradigm for an Effective Therapy. Trends Food Sci. Technol. 2022, 127, 198–206. [Google Scholar] [CrossRef]
  251. Haider, M.; Abdin, S.M.; Kamal, L.; Orive, G. Nanostructured Lipid Carriers for Delivery of Chemotherapeutics: A Review. Pharmaceutics 2020, 12, 288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Medici, S.; Peana, M.; Pelucelli, A.; Zoroddu, M.A. An Updated Overview on Metal Nanoparticles Toxicity. Semin. Cancer Biol. 2021, 76, 17–26. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Endogenous antioxidants reactions with reactive oxygen species. MnSOD reacts with superoxide ion (O2) converting it to hydrogen peroxide (H2O2). H2O2 diffuses to the cytoplasm and is converted to hydroxyl ion (OH) via Fenton reaction with Fe2+. OH cellular damage is prevented by glutaredoxin/thioredoxin, peroxidases (GPx, GRx, TRx), glutathione (GSH), and catalase (not shown). GSH is regenerated by nicotinamide adenine dinucleotide (NADH).
Figure 1. Endogenous antioxidants reactions with reactive oxygen species. MnSOD reacts with superoxide ion (O2) converting it to hydrogen peroxide (H2O2). H2O2 diffuses to the cytoplasm and is converted to hydroxyl ion (OH) via Fenton reaction with Fe2+. OH cellular damage is prevented by glutaredoxin/thioredoxin, peroxidases (GPx, GRx, TRx), glutathione (GSH), and catalase (not shown). GSH is regenerated by nicotinamide adenine dinucleotide (NADH).
Antioxidants 12 00123 g001
Table 1. Summary of examples of polyphenols and their effects on endogenous antioxidant systems in diabetic conditions.
Table 1. Summary of examples of polyphenols and their effects on endogenous antioxidant systems in diabetic conditions.
Antioxidant SystemExamples of PolyphenolEffects
UCP2/mitochondrial Complex 1Caffeic acid
Anthocyanins
In vitro protection of retinal cells, improving mitochondrial function [160].
Improve cardiac function by preserving mitochondrial function in diabetic rat model [166].
SIRTsResveratrolImprove GSH, CAT SOD activity and mitochondria function via activation of SIRT1/PGC-1α/FOXO3a.
GSH/GPx4/Xc system
(Inhibition of ferroptosis)
Glabridin and calycosinReno-protective effect in DN rat model [183,184].
Naringenin Cardio protective effect [185].
Astragaloside-IVIn vitro protection of retinal cells [186].
Quercetin and cryptochlorogenic acidPreserving β-cell function [87,188].
Nrf-2Epigallocatechin-3-gallate
Curcumin
Reno-protective effect in DN [206].
Protective effective against retinal injury in diabetes [213].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Akpoveso, O.-O.P.; Ubah, E.E.; Obasanmi, G. Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications. Antioxidants 2023, 12, 123. https://doi.org/10.3390/antiox12010123

AMA Style

Akpoveso O-OP, Ubah EE, Obasanmi G. Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications. Antioxidants. 2023; 12(1):123. https://doi.org/10.3390/antiox12010123

Chicago/Turabian Style

Akpoveso, Oke-Oghene Philomena, Emeka Emmanuel Ubah, and Gideon Obasanmi. 2023. "Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications" Antioxidants 12, no. 1: 123. https://doi.org/10.3390/antiox12010123

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