Open access peer-reviewed chapter

Cognitive Impairment in Diabetes Mellitus and Its Management by Transcription Factor Nrf2-Mediated Antioxidant Defense System

Written By

Xian Zhou, Gerald Münch and Dennis Chang

Submitted: 05 July 2022 Reviewed: 24 October 2022 Published: 28 November 2022

DOI: 10.5772/intechopen.108733

From the Edited Volume

Importance of Oxidative Stress and Antioxidant System in Health and Disease

Edited by Suna Sabuncuoğlu and Ahmet Yalcinkaya

Chapter metrics overview

124 Chapter Downloads

View Full Metrics

Abstract

Diabetes mellitus has been an epidemic in the twenty-first century and an approximately 50% risk of diabetes predisposed to cognitive decline leading to dementia in humans. There is an urgent need to understand the pathophysiology and identify molecular targets of cognitive impairment in diabetes mellitus that might lead to improved therapy. Mounting evidence indicates that nuclear factor erythroid 2-related factor 2 (Nrf2) and its regulated downstream antioxidant genes are emerging therapeutic targets. In this chapter, we introduce cognitive dysfunction in diabetes mellitus and its hallmarks, particularly its pathological mechanisms related to oxidative stress in the brain, then justify the role of the transcription factor Nrf2-mediated antioxidant defense system in attenuating cognitive decline in diabetes mellitus. Studies on Nrf2 inducers sourced from natural products (i.e., sulforaphane, astaxanthin, resveratrol, quercetin) that have shown potent cognitive improvement in diabetic models are discussed. These studies have demonstrated that Nrf2 inducers drive the antioxidant and anti-inflammatory responses in the hippocampus region and effectively improve the spatial and memory function in diabetic rats/mice. However, evidence from large and well-designed clinical trials is warranted to support Nrf2 inducers as promising therapeutic agents in the management of cognitive impairment in diabetes mellitus.

Keywords

  • cognitive impairment in diabetes mellitus
  • Nrf2
  • natural compounds
  • oxidative stress
  • NLRP3 inflammasome
  • neuroinflammation

1. Introduction

Diabetes mellitus (DM) is a chronic disease characterized by excessively high levels of blood glucose either resulting from insulin resistance or inadequate insulin secretion [1]. High blood glucose levels (hyperglycemia) in DM can cause serious damage to the heart, blood vessels, eyes, kidneys and nerves, and thus DM complications can significantly impact the quality of life and reduce life expectancy [2]. DM neuropathy is one of the major DM complications affecting 50% of DM sufferers which describes a type of nerve damage throughout the body induced by hyperglycemia [3, 4]. In the central nervous system, both type 1 (T1DM) and type 2 DM (T2DM) can cause nerve damage and result in various degrees of cognitive decline. Mounting epidemiological studies showed that the risk of cognitive decline in middle-aged diabetic patients was 2–3 times higher compared with healthy peers [5, 6]. Moreover, the prevalence of cognitive impairment is predicted to increase dramatically in the future with a longer survival time for diabetic patients [7]. Cognitive decline among children with T1DM is mainly manifested as subtle changes in cognitive development, whereas adults present subtle decrements in cognitive performance compared to age-matched controls [8]. The severity of cognitive impairment may worsen substantially over time [9]. In T2DM adult patients, the cognitive impairment can be divided into three different stages based on the severity [1011]: diabetes-associated cognitive decrements, mild cognitive impairment (MCI) and dementia. Diabetes-associated cognitive decrements refer to subtle changes in cognitive function. MCI affects one or more cognitive domains with largely preserved activities of daily life [12]. Dementia is the most severe stage that is defined as acquired objective cognitive impairment affecting multiple cognitive domains and daily activities [13, 14].

Cognitive impairment is a shared abnormality between DM and many neurodegenerative and neuropsychiatric disorders, such as Alzheimer’s disease (AD) and schizophrenia [15]. In DM, cognitive impairment can be prevented or slowed down by consistent blood sugar management and a healthy lifestyle. However, there is no effective therapeutic agent to treat the condition at present [16]. The pathological mechanism is not entirely clear. Despite the widespread view that hyperglycemia and insulin resistance are predominant risk factors, convincing evidence demonstrated that oxidative stress plays an important role in contributing to the development of cognitive impairment in DM [17, 18, 19]. High-fat diet (HFD) is shown to directly increase oxidative damage and impair cognitive and memory capacity [20]. The dysfunctional metabolism of blood glucose damages the basilar membranes of capillaries leading to a narrowed cavity in the cerebra, thus decreasing blood supply to the brain and resulting in oxygen-free radical injury [21]. Oxidative stress is observed in cognitive impairment by increased levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and decreased phase II antioxidant enzymes activities of glutathione peroxidase (GPx), chloramphenicol acetyltransferase (CAT) and superoxide dismutases (SOD) [22, 23]. The brain is highly susceptible to such oxidative damage partially due to its high oxygen demand, and the fact that high amounts of polyunsaturated fatty acids are easily targeted by free radicals [24]. Consequently, persistent hypoxia and oxygen-free radical injury can lead to neuronal cell injury and apoptosis [25]. Insulin is shown to penetrate the blood–brain barrier (BBB), and protects neurons against excessive free radicals, and the action is related to the nuclear factor erythroid 2-related factor 2 (Nrf2) regulated pathway [25]. Noticeably, insulin shows no protective effect on Nrf2-knockdown PC12 cells after H2O2-induced damage, while it significantly alleviated damage in Nrf2+/+ cells [25]. Furthermore, insulin was shown to inhibit oxidative stress in neuronal cells as a potential antioxidant agent through the Nrf2-Keap1/antioxidant response element (ARE) signaling pathway [26]. Thus, Nrf2 may play an important role to protect cells against oxidative stress as an oxidative stress-responsive transcription factor. Since the induction of Nrf2 is shown to ameliorate cognitive impairment in neurological disorders [27], Nrf2 has been considered an emerging therapeutic target for the prevention and treatment of cognitive impairment in DM.

In this chapter, we aimed to investigate the mechanisms of Nrf2 as an antioxidant master regulator to attenuate the pathological development of cognitive impairment in DM. We also reviewed current evidence on Nrf2 inducers sourced from natural products as promising therapeutic agents to help manage the pathological conditions and symptoms of cognitive impairment in DM.

Advertisement

2. Nrf2 acts as an important therapeutic target for cognitive impairment in DM

2.1 Ameliorate oxidative stress

Oxidative stress is constantly involved in the development and progression of cognitive impairment in DM. The persistent hyperglycemia, excessive lipid, and high-level advanced glycation end products (AGEs) all contribute to the excessive production of ROS [28]. Thus, ROS stress is considered a link between neurodegenerative diseases and T2DM, which induced oxidized DNA, RNA, protein, and lipid products that are used as disease progression marks in patients with both AD and T2DM [1]. The overproduction of ROS also triggers a prolonged state of inflammation through the high generation of white adipose tissue that secretes proinflammatory mediators [29], and subsequent neuronal death that affects the function of organ systems [3031]. The chronic oxidative stress and the associated neuroinflammation, in turn, can exacerbate abnormal insulin secretion, insulin action, and immune responses [32].

A number of studies suggested that a high-fat diet induced-adiposity and insulin resistance increased cerebral oxidative stress and downregulated Nrf2 signaling [2, 20, 33, 34, 35]. This might be the main reason that triggers the declined cognitive performance in the aged brain [20]. In addition, the diabetes experimental models suggested that enhancing Sirt1/Nrf2 signaling pathway activity prevented oxidative stress-induced neuronal injury, and thus improved cognitive function [2]. Nrf2 is a core transcription factor of antioxidative stress which regulates the cellular redox status. It interacts with ARE and plays a wide range of cytoprotective roles in the prevention of oxidative stress, neurological damage and inflammatory responses [36]. Nrf2 modulates the expression of more than 200 downstream genes encoding Phase II response enzymes during the oxidative challenge, including heme Oxygenase (HO-1), glutathione S-transferase (GST), catalase (CAT), superoxide dismutase (SOD), and NADPH quinone oxidoreductase (NQO1) [37]. The mechanisms of action to suppress oxidative stress by the activation of Nrf2 is mediated through the restored production of many downstream antioxidant genes which directly reduce the intracellular ROS accumulation [38]. The upregulation of Nrf2 target genes in the CNS protects neurons against oxidative insults as shown by a broad array of published studies [24, 39, 40, 41, 42, 43, 44, 45].

2.2 Suppress NLR family pyrin domain containing 3 inflammasome-induced neuroinflammation

The nod-like receptor pyrin containing 3 inflammasomes (NLRP3) is a multimeric protein complex containing cytosolic NLRP3, the adaptor protein ASC and caspase-1. Hyperglycaemia and other repetitive stress stimuli assemble and activate the NLRP3 inflammasome which then triggers the activation of caspase-1 by proteolytic cleavage [46]. The latter converts pro-IL-1β into bioactive IL-1β and consequently initiates inflammatory responses. The sustained inflammatory responses can cause neurotransmitter dysfunction which subsequently leads to a decreased production of brain-derived neurotrophic factor (BDNF), a protein responsible for the learning and memory process [47]. Thereby, it results in neuroinflammation-mediated neuronal damage as well as deterioration of cognitive function [48, 49, 50]. The aggravated neurodegeneration caused by the accumulation of IL-1β can be rescued by blocking IL-1β signaling in APP/PS1 mice [50, 51, 52, 53], suggesting NLRP3 inflammasome-mediated neuroinflammation is an important therapeutic target. Moreover, inflammatory responses can also induce the overproduction of ROS which then exacerbate the oxidative damage to neurons [54].

Recent studies have shown that Nrf2/ARE signaling is directly implicated in the regulation of inflammation rather than relying on regulating oxidative stress to control inflammatory response [48, 49, 55, 56]. Although not fully understood, Nrf2 regulated AGEs/RAGE pathway is shown to mediate the activation of NLRP3 inflammation by providing a priming signal at a transcriptional level [57, 58] which is essential for the assembly of the NLRP3 inflammasome [59]. Excessive activation of this process exhausts cytosolic NADPH, increases the accumulation of ROS, and disrupts Nrf2/ARE pathway, finally leading to oxidative stress [60]. Besides, several studies revealed that suppressed intracellular ROS accumulation by the activation of Nrf2 resulted in reduced activation of the NLRP3 inflammasome, and subsequently inhibited neuroinflammation [38]. Additionally, Nrf2 activation triggers the restored expressions of HO-1 and NQO1 which then further contribute to the suppression of inflammation-associated cytokine productions and NLRP3 inflammasome formation [61, 62, 63].

2.3 Alleviate endoplasmic reticulum stress

The endoplasmic reticulum (ER) is involved in glucose-modulated secretory protein folding, intracellular Ca2+ storage, synthesis of unsaturated fatty acids, sterols, and phospholipids [64, 65]. High glucose levels in the body disrupt the internal balance of the ER, resulting in the accumulation of unfolded or misfolded proteins, which leads to ER stress (ERS) [66]. Oxidative stress also can induce ER stress responses [67] which are triggered by the accumulation of ROS [68].

Excessive ERS impairs neuronal function through various pathways. In hippocampal cells, high glucose and palmitic acid-induced ERS significantly decrease proBDNF, but were restored when ER stress was reduced [69]. In the neuroinflammatory pathway, ERS can activate the NLRP3 inflammasome and lead to the subsequent secretion of proinflammatory cytokines such as IL-1β [676870] which is detrimental to the neuronal function as described above. In addition, ERS can further impair neuronal survival. The double-stranded RNA-dependent protein kinase (PKR)-like ER-resident kinase (PERK) pathway is one of the three ERS-related protective signaling pathways identified so far. PERK pathway can activate caspase-12, which is a specific mediator of ER stress-induced apoptosis, to impair protein synthesis and synaptic functions [70]. Excessive or prolonged ER stress could up-regulate the transcription factor C/EBP Homologous Protein (CHOP), which is a critical mediator of the Bax/Bcl-2 -dependent pathway, thus converting neuronal cells from the pro-survival to pro-apoptosis phase [71, 72]. Thus, ERS is considered a valuable therapeutic target in neuroprotection such as the cognitive decline in DM [73].

ERS can cause and exacerbate oxidative stress, whereas oxidative stress can worsen ERS. In response to ERS, the onset of the PERK pathway quickly activates Nrf2 as an antioxidant defensive system [66], although the clear link remains to be explored. As an endogenous defense system against oxidative stress, Nrf2 regulates the expression of a subset of detoxifying enzymes including NQO1 and γ-GCS. This suggests an important cytoprotective role of Nrf2 against ERS, and that the Nrf2 pathway represents a useful therapeutic strategy in cognitive decline by attenuating ERS. However, the moderate self-defense response of PERK and Nrf2 maybe not be enough to prevent damage [73]. The other pathway that Nrf2 may exert a beneficial effect in attenuating ERS is that the Nrf2 activation contributes to the maintenance of glutathione levels, which, in turn, functions as a buffer for the accumulation of ROS during the unfolded protein response in ER and thus may attenuate ERS. In addition, the reduced level of ROS may reduce apoptotic induction as a consequent event after ERS [74].

2.4 Repair a leaking BBB

Impairment of BBB induced by hyperglycemia, oxidative damage, and inflammation is a critical neurovascular complication of DM that adversely affects the microvascular environment, health, and function of the central nervous system. Many studies have demonstrated that hyperglycemia-driven neuroinflammation is a risk factor for BBB disruption leading to a high permeability of BBB [75, 76, 77]. The BBB leakage leads to many adverse impacts on the central nervous system including decreased waste transport, increased infiltration of immune cells and subsequent glial and neuron dysfunction, over-active immune sensitivity, hormone dysregulation, and cognitive impairment [78]. The excessive production of ROS also induces endothelial oxidative stress and mitochondrial damage which has been recognized as a central pathological mechanism for BBB dysfunction in DM.

A study from Sajja et al. suggested a close link of Nrf2 protein expression to that of the BBB permeability, as evidenced by a significantly down-regulated brain Nrf2 protein expression in the db/db diabetic mice with a strong increase in BBB permeability (to 70 kDa dextran) [79]. The role of Nrf2 induction in the repairment of BBB appeared to be associated with the mitochondrial transporter ABCB10 which is considered as an essential player in mitochondrial function and redox balance at BBB endothelium. The ABCB10 knockdown resulted in a strong induction of Nrf2-driven antioxidant responses manifested as increased expression of Nrf2 and its downstream antioxidant genes [79]. In addition, several studies suggested that Nrf2-driven free radical detoxification pathways are essential in protecting against oxidative stress-induced endothelial injuries [80], and thus may in turn contribute to the protection of BBB [81].

2.5 Protect mitochondria function

Mitochondrial dysfunction is implicated in the pathogenesis of most neurodegenerative disorders including cognitive impairment in DM [82]. Mitochondria is the main source of energy in the cell, by providing ATP through oxidative phosphorylation and harboring several metabolic pathways such as fatty acid oxidation (FAO) and the tricarboxylic acid cycle (TCA) cycle [82]. Accumulated oxidative damage causes an altered oxidative phosphorylation and redox imbalance in mitochondria [83]. The long-lasting effect can lead to the des-regulation of mitochondrial protein homeostasis, and consequent proteotoxic stress in the neurons [84]. Neurons in the central nervous system have a high requirement for energy as attributed to their constant activity and signaling, and thus are especially vulnerable to mitochondrial dysfunction. Decreased ATP production is a common hallmark of neurodegenerative diseases and can be caused by various mechanisms, such as the impaired activity of any of the complexes of the respiratory chain, alterations in glucose uptake, glycolysis, TCA cycle, or uncoupling [85].

Nrf2 can lead to mitochondrial bioenergetic improvement, although the mechanisms of action are not fully explored [82]. There are several mechanisms of action that underpin the role of Nrf2 in protecting mitochondrial function. Nrf2 prevents the oxidative thiol modifications that can modulate the function of proteins implicated in metabolic pathways [86, 87, 88]. The accumulative mitochondrial ROS can reversibly modify thiol groups presented in several enzymes implicated in carbohydrate and lipid metabolism. The activation of Nrf2 that scavenge or inhibit the overproduction of mitochondrial ROS may prevent the redox modifications of the sensitive thiol groups [89, 90, 91]. Consequently, such antioxidant modification enables the restoration of normal glycolytic metabolism and reduces neuronal apoptosis [92].

2.6 Modulation of glucose metabolism

Diabetic patients (both type 1 and type 2) have a higher risk of AD and vascular dementia, mainly caused by abnormal glucose metabolism induced by hyperglycemia and consequent cerebrovascular lesions in the frontal lobe [93]. The elevated glucose concentration leads to enhanced oxidative phosphorylation and increased levels of glutamate, an excitatory neurotransmitter, whose enhanced levels provoke cognitive dysfunction by causing neuronal damage.

Although the majority of evidence stands for the antioxidant properties of Nrf2 activation, Nrf2 is also involved in glucose metabolism [94, 95]. Nrf2 can directly activate the transcription of enzymes containing ARE-sequences in the gene promoters, which have been described in the promoters of the genes encoding the peroxisome proliferator-activated receptor ɣ (PPAR- ɣ) [96]. PPAR-ɣ is known to regulate adipocyte differentiation and lipid metabolism, as well as the network regulation of glucose homeostasis [97]. It also exhibited a versatile role in early brain development and post-injury brain repair [98]. The activation of PPAR- ɣ is shown to repair damaged tissue through angiogenesis, alleviate neurological deficits, and exhibit neuroprotective activity [96]. Indeed, studies have revealed the connection of Nrf2 and PPAR-ɣ via a positive feedback loop that simultaneously strengthens each other’s expression [94, 95]. For example, PPAR-ɣ expression was found to be significantly lower in Nrf2 knockout mice [99]. It is also identified that one of the many Nrf2 downstream targets, TALDO1, contains an ARE sequence which is a key enzyme in the nonoxidative pentose phosphate pathway, providing ribose-5-phosphatase and NADPH for nucleic acid and lipid biosynthesis [92].

Taken together, the recently recognized abilities of Nrf2 to ameliorate oxidative and ERS, repair BBB, protect mitochondrial function and regulate glucose metabolism make the Nrf2 activation an attractive and comprehensive therapeutic target for the management of cognitive impairment in DM. A summarized diagram of Nrf2-associated pathways in the pathological mechanisms is shown in Figure 1.

Figure 1.

A summarized diagram of Nrf2-associated pathways in the pathological mechanisms of DCI.

Advertisement

3. Nrf2 activators from natural products that prevent or treat cognitive impairment in DM

Natural products have served as a rich source of novel drug discovery and development. This section focuses on the promising Nrf2 inducers sourced from natural products that have shown beneficial effects in the prevention or treatment of cognitive impairment in DM. A literature search was conducted in scientific databases (PubMed, MEDLINE and EMBASE) and Google Scholar for English-published studies between January 2000 and September 2022. Clinical and pre-clinical studies of natural products (i.e., herbal extracts, vegetables, plants, chemicals, nutraceuticals, and supplements sourced from plant or plant extract) that showed effects in attenuating cognitive impairment in DM and the mechanism of action was associated with the Nrf2 activation were included. Studies investigating pharmaceutical drugs, analog drugs, and synthesized compounds were excluded.

Nineteen preclinical studies demonstrated that various natural compounds attenuated cognitive impairment in DM at least partially through the induction of Nrf2 (see Table 1). The effects on cognitive impairment by the natural products were mostly demonstrated in the diabetic animal models, either on high-fat diet (HDF) or streptozotocin (STZ) injection-induced diabetic rats [111, 113, 114], db/db mice (model phase 1 to 3 of diabetes type II and obesity) [100, 115], or diabetic Goto-Kakizaki rats (non-obese type 2 diabetic model) [110]. One cellular study was included which utilized chronic high glucose-exposed SHSY5Y neurons [116]. The studies on natural Nrf2 activators with strong preclinical evidence and associated molecular mechanisms are discussed in detail below.

Natural compoundsSourceAdministrationKey resultsMechanism related to Nrf2 activation
Sulforaphane [100, 101]Vegetables1 mg/kg, i.p. for 28 daysMitigated cognitive impairment of db/db mice; decreased Aβ oligomers and plaques, phosphor-tau and Thr231 in hippocampus↑Nrf2 → ↑HO-1 and NQO1 → ↓ROS level
25 mg/kg, orally once daily for consecutive 14 daysPrevented the memory impairment, decreased the apoptosis of hippocampal
neurons in STZ-injected SD rats
↓ Caspase-3 and myeloid cell leukemia 1 (MCL-1);
↑ p-Akt,
p-GSK3β, NGF and BDNF
Troxerutin [102103]Sophora japonica60 mg/kg, 1 mL/kg, i.p., 12 weeksImproved learning and memory levels in STZ-induced diabetic rats↑Nrf2 → ↑SOD and ↓MDA
150 mg/kg/day intraperitoneally for 6 weeksImproved cognitive impairment in STZ-induced diabetic rats↑ Nrf2 translocation → ↑ HO-1 and NQO1 → NOX subunits → ↓MDA and ↑SOD
Strawberry leaf extract [104]Strawberry tree200 mg/kg for 4 weeksAlleviated cognitive impairment in STZ-induced diabetic rats↑Nrf2 -HO-1 signaling → ↓ROS, ↓MDA, ↑SOD and CAT; ↓IL-6 and TNF-α; ↓caspase-3 and caspase-9 in hippocampus
Betulin [105]Birch tree bark20 or 40 mg/kg for 4 weeksImproved glucose intolerance and modify basal learning performance in STZ-induced diabetic rats↑Nrf2 -HO-1 signaling and ↓NF-κB → ↑SOD and ↓MDA; ↓inflammatory cytokines in serum and hippocampus
S-allyl cysteine [106]Aged garlic extract150 mg/kg/day for 7 weeks (p.o.)Lowered serum glucose, improved spatial recognition memory, discrimination ratio in novel object recognition task, and restored step-through latency (STL) in
passive avoidance paradigm in STZ-induced diabetic rats
↑Nrf2-HO-1 signaling and ↓TLR4-NF-κB signaling → ↓ acetylcholinesterase activity, MDA; ↑SOD and GSH
Caffeic acid phenethyl ester [107]Propolis200 and 400 mg/kg p.o. for 14 daysRescued the diabetic brain atrophy and diminish CA1 and CA3 cells of hippocampus and cerebral cortex in STZ-induced diabetic mice; decrease Aβ and p-tau (S396)↑Nrf2-related anti-oxidation mechanisms → antioxidation, anti-inflammation and autophagy induction
AB-38b [108]Fructus Schisandrae chinensis0, 10, 20 and 40 mg/kg by gavage for 8 weeksIncreased the preference index to novel object and the number of neurons in hippocampal CA1 area of diabetic mice↑Nrf2 expression and phosphorylation → ɣ-GCS
Fish oil supplementation [109]Fish oil1.5 g/kg/d (34% EPA, 24%
DHA) for 10 weeks
Improved spatial learning and
memory in STZ-induced diabetic rats
Nrf2 and HO-1 in cortex and hippocampus → ↓oxidative stress → ↓ IL-1β, IL-6, and TNF-α, ↑ IL-4 and IL-10
Soy isoflavones [110]Soy20 mg/kg once a day for 4 weeksAlleviated the cognitive
dysfunction of the diabetic Goto-Kakizaki rats
↑ Nrf2, HO-1 and NQO1 → oxidative reactions
Resveratrol [111, 112]Grapes, berries, etc.30 mg/kg every other day for 4 months by intragastric administrationPrevented the learning and memory
decline and hippocampal neuron
destruction and synaptic ultrastructural damage in HFD and STZ- induced T2DM mice
↑ Nrf2 → ↑ HO-1 and NQO1 proteins; ↑ SOD and CAT → ↓ TNF-α and IL-1β
20 mg/kg, intraperitoneally once daily for 4 weeksSignificantly elevated total oxidant species (1.22-fold) and
Malonedialdehyde (MDA) (1.38-fold) contents in diabetic rat brain cortex tissues in STZ (55 mg/
kg)-injected Wistar rats
↓oxidative conditions
Thymol (2-isopropyl-5-methylphenol) [113]Thyme20, 40 mg/kg for 12 weeksReversed the gain of body weight and peripheral insulin resistance induced by
HFD; improved the cognitive impairments; decreased
HFD-induced Aβ deposition and tau hyperphosphorylation
↑ Nrf2-HO-1 signaling → ↓ oxidative stress and inflammation
Astaxanthin [114]Algae, yeast, salmon, trout, krill, shrimp, and crayfish25 mg/kg 3 times/week for 6 weeks by intraperitoneallyAmeliorated the impairment in the
neurons of HFD and STZ-induced diabetic rats
↑Nrf2-HO-1 and ↓NF-κB signalings → ↑ SOD and ↓ MDA; ↓IL-1β and IL-6
Notoginsenoside R1 [115]Panax notoginseng10, 30 mg/kg once daily administrated intragastrically for 10 weeksAmeliorated cognitive dysfunction,
depression-like behaviors, insulin resistance, hyperinsulinemia, dyslipidemia, and
inflammation in db/db mice
↑Akt → ↑ Nrf2 and ↓ NLRP3
Albiflorin [7]Paeonia lactiflora100, 200 mg/kg by gastric gavage for 10 weeksImproved spatial and learning ability; decrease in Aβ plaque density in the hippocampus of rats;Regulate Nrf2/HO-1/HMGB1/NF-κB → ↓ oxidative stress and inflammation
Quercetin [116, 117]Fruits and vegetables5, 10, 20 μmol/L for 72 hIncreased cell viability, and enhanced Glo-1 functions in SHSY5Y neurons↑Nrf2 and p-Nrf2 levels → ↑ ɣ-GCS protein and mRNA
50 mg/kg i.p. for 18 daysSignificantly mitigated the STZ-induced increase in cholinergic dysfunction in STZ (3 mg/kg)-induced brain mitochondrial toxicity in Alzheimer’s disease -like rats↑α7nAChR/Nrf2/HO-1-mediated
neuroprotection

Table 1.

Summarized studies for natural products that attenuate cognitive impairment in DM and associated symptoms via the mechanism of Nrf2 activation.

3.1 Sulforaphane

Sulforaphane is a hydrolysis product of glucoraphanin, a group of sulfur-containing glycosides, found in many raw vegetables such as broccoli, cauliflower, bok choy, and watercress [118, 119, 120, 121]. Upon consumption, glucoraphanin is hydrolyzed to sulforaphane which has been demonstrated to penetrate BBB [122] and exhibit a neuroprotective effect via a number of mechanisms [123]. In particular, strong evidence has indicated that sulforaphane is a potent Nrf2 activator. The molecular mechanisms are linked with increased Nrf2 transcription by reducing methylation of the first 15 CpGs of Nrf2 promoters [124], modulation of Kelch-like ECH-associated protein 1 (Keap1) [125, 126, 127], prevention of ubiquitination of Nrf2 [128], and enhanced Nrf2 translocation [128]. The activation of Nrf2 by sulforaphane was reported to mediate a long-lasting effect of the upregulation of cytoprotective enzymes (4-hour exposure of sulforaphane led to 24 hrs NQO1 and HO-1 mRNA elevation and over 48 hrs increase of corresponding proteins) [129].

Two in vivo studies have investigated the effect of IP/oral injection of sulforaphane on mitigating cognitive impairment in diabetic models [100, 101]. In particular, the study by Pu et al. suggested that the IP injection of sulforaphane (1 mg/kg for 28 days) improved the spatial learning and memory function of db/db mice examined by the Morris water maze tests. Sulforaphane was shown to decrease the levels of Aβ oligomers, Aβ 1–42 plaques, phosphor-tau at Ser396 and Thr231 in the hippocampus. The molecular mechanisms were related to Nrf2-mediated upregulation of HO-1 and NQO1 protein expression, and reduced ROS/RNS levels [100]. In line with the research finding, a study by Wang et al. revealed that the oral administration of sulforaphane (25 mg/kg) for consecutive 14 days prevented memory impairment in STZ-induced diabetic rats [101]. It was revealed that sulforaphane markedly reduced apoptotic neurons levels and it was associated with decreased phospho-protein kinase B (Akt), phosphor-glycogen synthase kinase 3β (GSK3β), nerve growth factor (NGF) and BDNF expressions. However, the link between the reduced neuronal apoptotic level and the Nrf2 activation remains to be explored. Broadly speaking, many studies have agreed the strong neuroprotective effect of sulforaphane by activating Nrf2 antioxidant signaling cascade, and such activity can be used in many neurodegenerative diseases including AD, traumatic brain injury, and ischemic stroke [130]. Future studies are suggested to explore the safety profile and pharmacokinetic characteristics of sulforaphane to further demonstrate its therapeutic value in improving cognitive function in humans.

3.2 Astaxanthin

Astaxanthin is a red pigment belonging to a group of carotenoids that causes the pink-red color in salmon. Astaxanthin has various biological and pharmacological activities such as antioxidant, anti-inflammatory, and antidiabetes [131]. Many studies demonstrated that astaxanthin exhibited potential neuroprotective effects, of which the mechanism was associated with Nrf2 activation [131, 132, 133, 134]. Astaxanthin administration was shown to accelerate nuclear translocation of Nrf2 via inducing PI3K/Akt signaling, resulting in a decrease in ROS levels, and inhibiting apoptosis in neuronal cells [135].

Orally or parenterally administered astaxanthin has been shown to improve insulin resistance and insulin secretion, attenuate hyperglycemia, and exert protective effects against many DM complications including retinopathy, nephropathy, and neuropathy [136]. Specifically, for cognitive impairment in DM, astaxanthin administration improves cognitive function [137] and diminish oxidative stress, nitric oxide synthase, and inflammation in DM rat [137]. Furthermore, Feng et al. suggested that IP injection of astaxanthin (25 mg/kg) ameliorated the impairment in the neurons of HFD and STZ-induced diabetic rats, and the action was associated with upregulated the Nrf2-HO-1 signaling pathway which drove the increased expressions of SOD and declined MDA. The intermediate metabolites from the Nrf2-HO-1 axis contribute to the suppressed inflammatory response by inhibiting the activation of NF-κB leading to decreased levels of IL-1β and IL-6 [114].

3.3 Resveratrol

Resveratrol (3,4′,5-trihydroxy-stilbene, RES) is a very popular nutraceutical that is a naturally occurring compound found in grapes, cereals, vegetables, dry legumes and plant-derived beverages, including tea, coffee, and wine. A large number of studies supported its beneficial use in anti-diabetes, anti-inflammatory, and antioxidative conditions [138, 139, 140, 141, 142]. In diabetes, resveratrol is shown to exhibit an insulin-sensitizing effect, enhance glucose uptake and metabolism, preserve islet β-cells and release insulin from β-cells [138]. Among all the associated molecular mechanisms of resveratrol in treating diabetic complications, one of the key pathways was the upregulation of Nrf2 nuclear translocation which then induced the increased expressions of GSH, SOD, NQD1, and HO-1, reduced ROS production, and declined oxidative stress [138]. Two studies specifically investigated the protective effect of resveratrol on cognitive function in diabetic rats via regulating oxidative biomarkers and antioxidant enzymes in the brain [111, 112]. A study from Sadi et al. suggested that resveratrol significantly elevated total oxidant species in diabetic rat brain cortex tissues in STZ-injected Wistar rats [112]. Moreover, Wang et al. showed that resveratrol prevented the learning and memory decline in STZ-induced T2DM mice [111], and such action was linked with upregulated Nrf2 and its mediated HO-1 and NQO1 proteins, SOD and CAT enzymes, as well as reduced IL-1β and TNF-α. A randomized, double-blind, placebo-controlled clinical trial demonstrated that a total of 48 patients with T2D received 800 mg/day of resveratrol for 2 months markedly increased Nrf2 and SOD expressions in peripheral blood mononuclear cells. Resveratrol was well tolerated without causing any serious adverse events [143]. Thus, it is encouraging that resveratrol may exhibit beneficial clinical outcomes in T2D patients with cognitive decline as a stronger Nrf2 activator.

3.4 Quercetin

Quercetin is one of the most abundant polyphenolic flavonoids, which is present in fruits and vegetables and displays a strong health-promoting effect [144]. In addition, quercetin plays an indirect role in neutralizing oxidative stress by activating the Nrf2-ARE antioxidant pathway and inducing the expression of antioxidant enzymes like CAT and SOD. In lipopolysaccharides (LPS)-induced murine BV-2 microglial cells, quercetin produced a greater stimulating effect on Nrf2-induced increase expression of heme-oxygenase-1 (HO-1) protein than cyanidin against endotoxic stress via the participation of mitogen-activated protein kinase (MAPKs) [145, 146]. In particular, quercetin was found to increase cell viability and enhance glyoxalase-I (Glo-1) functions in SHSY5Y cells which were related to activated Nrf2 and phosphor-Nrf2 levels [116]. Furthermore, IP injection of quercetin (50 mg/kg) for 18 days significantly mitigated the STZ-induced increase in cholinergic dysfunction in STZ (3 mg/kg)-induced brain mitochondrial toxicity in AD-like rats, which the mechanism was associated with α7nAChR/Nrf2/HO-1-mediated neuroprotection [117]. However, a clinical trial is warranted to further investigate the effect of quercetin in humans.

Advertisement

4. Conclusion and future perspectives

A number of natural compounds, such as sulforaphane, astaxanthin, resveratrol, and quercetin [115] were found to significantly improve the spatial and memory function in diabetic animals models (rats/mice) with the effective daily dosage ranging from 1 mg/kg to 30 mg/kg. Most of the treatments were given after the induction of DM. In addition, most treatments from natural products were long-term treatment duration (14 days to 4 months) with a daily administration to see an obvious effect. The summarized information is listed in Table 1. However, clinical trials are largely lacking to further demonstrate their safety and efficacy in humans.

Nrf2 inducers sourced from natural products upregulated Nrf2 activity, Nrf2 translocation (nuclear Nrf2 protein expression) and/or phosphorated Nrf2 expression. This leads to the activation of Nrf2-regulated downstream antioxidant genes including HO-1, NQO1, SOD and CAT [102, 103, 104, 106, 111114]. The reduced levels of ROS and MDA in the hippocampus were often reported as the consequent events. Interestingly, many studies also mentioned the inhibited NF-κB signaling in addition to the Nrf2-mediated antioxidant response [7, 106, 114]. The upregulated expression of HO-1 protein in response to the Nrf2 upregulation suppresses the NF-κB activation which governs the transcriptions and productions of proinflammatory cytokines. Thus, Nrf2/HO-1 pathway not only drives the antioxidant response but also plays a critical role in regulating the proinflammatory reactions. The suppressed proinflammatory response in the animals is evidenced by the reduced production of IL-6 and TNF-α and increased levels of IL-4 and IL-10 [109114]. However, the inner relationship among Nrf2-mediated antioxidants and anti-inflammatory by the Nrf2 activators is not fully explored.

It is worth mentioning that the Nrf2 activation in cognitive impairment was shown to be mediated by the induction of protein kinase B (Akt) as an upstream mediator. A study from Zhai et al. suggested that notoginsenoside R1 (NR1) activated the Nrf2 pathway as well as inhibited NLRP3 inflammasome which both contributed to ameliorated cognitive function. However, the neuroprotective effect after the Nrf2 activation and NLRP3 inflammasome inhibition was abolished by the co-treatment of Akt inhibitor and NR1 [115]. It has been well-studied that the activation of Akt regulates cell survival and apoptosis as well as glucose transport [147, 148]. NR1 was capable to improve glucose tolerance and insulin sensitivity in db/db mice and reducing neuron apoptosis in the hippocampal CA1 region, and the mechanisms may be associated with the activation of the Akt and Nrf2 signaling cascade [115]. These two studies highlight the possible molecular action of Nrf2 inducers mediated by the Akt pathway in protecting neuronal survival in the hippocampal region. Further studies are warranted to elucidate the link between Akt and Nrf2 pathways.

Taken together, this book chapter introduces cognitive impairment in DM as the major complication in both type 1 and type 2 DM, and explained the associated pathological mechanisms, particularly the detrimental role of oxidative stress that impairs cognitive and memory capacity. Nrf2, the core intracellular transcription factor of antioxidative stress, has been considered an emerging therapeutic target against cognitive impairment in DM. The induction of Nrf2 signaling drives the antioxidant defense system to ameliorate oxidative and ER stress, repair the BBB, protect mitochondrial function, and regulate glucose metabolism, which eventually protects neurons against oxidative stresses and hyperglycemia and helps to restore cognitive function. A number of studies, predominantly based on diabetic animal models, have demonstrated that Nrf2 inducers sourced from natural products such as vegetables, soy, garlic, and strawberry leaf improved cognitive and memory function. The associated mechanisms of action were shown as Nrf2-mediated antioxidant and anti-inflammatory responses. Although the results are supportive, conclusive evidence for each proposed therapeutic candidate is weak due to the low quantity of studies. The safety human trials to further demonstrate their safety and efficacy are warranted. However, the findings of this chapter may shed light on the development of natural Nrf2 activators as promising pharmacological agents to prevent or slow the progress of cognitive impairment in DM.

Advertisement

Acknowledgments

I am grateful to Professor Gerald Münch who inspires me greatly in the research field of the application of Nrf2-mediated antioxidant defense system in the prevention and treatment of many diseases including neurodegenerative diseases. Thanks to Mr. John Truong for his kind support.

Advertisement

Conflict of interest

The authors declare no conflict of interest. As a medical research institute, NICM Health Research Institute receives research grants and donations from foundations, universities, government agencies, individuals and industry. Sponsors and donors also provide untied funding for work to advance the vision and mission of the Institute.

References

  1. 1. He L, Sun Y. The potential role of Keap1-Nrf2 pathway in the pathogenesis of Alzheimer’s disease, type 2 diabetes, and type 2 diabetes-related Alzheimer’s disease. Metabolic Brain Disease. 2021;36:1469-1479. DOI: 10.1007/s11011-021-00762-z
  2. 2. 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. Molecular Medicine Reports. 2018;17:8414-8422. DOI: 10.3892/mmr.2018.8876
  3. 3. Vinik AI, Nevoret M-L, Casellini C, Parson H. Diabetic neuropathy. Endocrinology and Metabolism Clinics of North America. 2013;42:747-787. DOI: 10.1016/j.ecl.2013.06.001
  4. 4. Figueroa-Romero C, Sadidi M, Feldman EL. Mechanisms of disease: The oxidative stress theory of diabetic neuropathy. Reviews in Endocrine & Metabolic Disorders. 2008;9:301-314. DOI: 10.1007/s11154-008-9104-2
  5. 5. Degen C, Toro P, Schönknecht P, Sattler C, Schröder J. Diabetes mellitus type II and cognitive capacity in healthy aging, mild cognitive impairment and Alzheimer's disease. Psychiatry Research. 2016;240:42-46. DOI: 10.1016/j.psychres.2016.04.009
  6. 6. Luchsinger JA, Reitz C, Patel B, Tang M-X, Manly JJ, Mayeux R. Relation of diabetes to mild cognitive impairment. Archives of Neurology. 2007;64:570-575. DOI: 10.1001/archneur.64.4.570
  7. 7. Ma X, Song M, Yan Y, Ren G, Hou J, Qin G, et al. Albiflorin alleviates cognitive dysfunction in STZ-induced rats. Aging (Albany NY). 2021;13:18287. DOI: 10.18632/aging.203274
  8. 8. Ryan CM, van Duinkerken E, Rosano C. Neurocognitive consequences of diabetes. The American Psychologist. 2016;71:563. DOI: 10.1037/a0040455
  9. 9. Nunley KA, Rosano C, Ryan CM, Jennings JR, Aizenstein HJ, Zgibor JC, et al. Clinically relevant cognitive impairment in middle-aged adults with childhood-onset type 1 diabetes. Diabetes Care. 2015;38:1768-1776. DOI: 10.2337/dc15-0041
  10. 10. Koekkoek PS, Kappelle LJ, van den Berg E, Rutten GE, Biessels GJ. Cognitive function in patients with diabetes mellitus: Guidance for daily care. Lancet Neurology. 2015;14:329-340. DOI: 10.1016/S1474-4422(14)70249-2
  11. 11. Dolores EB. Diagnostic and statistical manual of mental disorders. Codas. 2013;21:591-643. DOI: 10.1590/s2317-17822013000200017
  12. 12. Petersen RC. Mild cognitive impairment as a diagnostic entity. Journal of Internal Medicine. 2004;256:183-194. DOI: 10.1111/j.1365-2796.2004.01388.x
  13. 13. Petersen RC, Lopez O, Armstrong MJ, Getchius TS, Ganguli M, Gloss D, et al. Practice guideline update summary: Mild cognitive impairment: Report of the guideline development, dissemination, and implementation Subcommittee of the American Academy of neurology. Neurology. 2018;90:126-135. DOI: 10.1212/WNL.0000000000004826
  14. 14. Dubois B, Feldman HH, Jacova C, Cummings JL, DeKosky ST, Barberger-Gateau P, et al. Revising the definition of Alzheimer's disease: A new lexicon. Lancet Neurology. 2010;9:1118-1127. DOI: 10.1016/S1474-4422(10)70223-4
  15. 15. Chen Q , Cao T, Li N, Zeng C, Zhang S, Wu X, et al. Repurposing of anti-diabetic agents as a new opportunity to alleviate cognitive impairment in neurodegenerative and neuropsychiatric disorders. Frontiers in Pharmacology. 2021;12:667874. DOI: 10.3389/fphar.2021.667874
  16. 16. Sharma G, Parihar A, Talaiya T, Dubey K, Porwal B, Parihar MS. Cognitive impairments in type 2 diabetes, risk factors and preventive strategies. Journal of Basic and Clinical Physiology and Pharmacology. 2020;31:20190105. DOI: 10.1515/jbcpp-2019-0105
  17. 17. Muriach M, Flores-Bellver M, Romero FJ, Barcia JM. Diabetes and the brain: Oxidative stress, inflammation, and autophagy. Oxidative Medicine and Cellular Longevity. 2014;2014:102158. DOI: 10.1155/2014/102158
  18. 18. Domínguez R, Pagano M, Marschoff E, González S, Repetto M, Serra J. Alzheimer disease and cognitive impairment associated with diabetes mellitus type 2: Associations and a hypothesis. Neurología. 2014;29:567-572. DOI: 10.1016/j.nrl.2013.05.006
  19. 19. Hoyos CM, Colagiuri S, Turner A, Ireland C, Naismith SL, Duffy SL. Brain oxidative stress and cognitive function in older adults with diabetes and pre-diabetes who are at risk for dementia. Diabetes Research and Clinical Practice. 2022;184:109178. DOI: 10.1016/j.diabres.2021.109178
  20. 20. Morrison CD, Pistell PJ, Ingram DK, Johnson WD, Liu Y, Fernandez-Kim SO, et al. High fat diet increases hippocampal oxidative stress and cognitive impairment in aged mice: Implications for decreased Nrf2 signaling. Journal of Neurochemistry. 2010;114:1581-1589. DOI: 10.1111/j.1471-4159.2010.06865.x
  21. 21. Aruoma OI, Neergheen VS, Bahorun T, Jen L-S. Free radicals, antioxidants and diabetes: Embryopathy, retinopathy, neuropathy, nephropathy and cardiovascular complications. Neuroembryology Aging. 2006;4:117-137. DOI: 10.1159/000109344
  22. 22. Padurariu M, Ciobica A, Hritcu L, Stoica B, Bild W, Stefanescu C. Changes of some oxidative stress markers in the serum of patients with mild cognitive impairment and Alzheimer's disease. Neuroscience Letters. 2010;469:6-10. DOI: 10.1016/j.neulet.2009.11.033
  23. 23. Tayanloo-Beik A, Kiasalari Z, Roghani M. Paeonol ameliorates cognitive deficits in streptozotocin murine model of sporadic Alzheimer’s disease via attenuation of oxidative stress, inflammation, and mitochondrial dysfunction. Journal of Molecular Neuroscience. 2022;72:336-348. DOI: 10.1007/s12031-021-01936-1
  24. 24. Vasconcelos AR, Dos Santos NB, Scavone C, Munhoz CD. Nrf2/ARE pathway modulation by dietary energy regulation in neurological disorders. Frontiers in Pharmacology. 2019;10:33. DOI: 10.3389/fphar.2019.00033
  25. 25. Song Y, Ding W, Bei Y, Xiao Y, Tong H-D, Wang L-B, et al. Insulin is a potential antioxidant for diabetes-associated cognitive decline via regulating Nrf2 dependent antioxidant enzymes. Biomedicine & Pharmacotherapy. 2018;104:474-484. DOI: 10.1016/j.biopha.2018.04.097
  26. 26. Bhakkiyalakshmi E, Sireesh D, Rajaguru P, Paulmurugan R, Ramkumar KM. The emerging role of redox-sensitive Nrf2–Keap1 pathway in diabetes. Pharmacological Research. 2015;91:104-114. DOI: 10.1016/j.phrs.2014.10.004
  27. 27. Uruno A, Matsumaru D, Ryoke R, Saito R, Kadoguchi S, Saigusa D, et al. Nrf2 suppresses oxidative stress and inflammation in App knock-in Alzheimer’s disease model mice. Molecular and Cellular Biology. 2020;40:e00467-e00419. DOI: 10.1128/MCB.00467-19
  28. 28. Ahmad W, Ijaz B, Shabbiri K, Ahmed F, Rehman S. Oxidative toxicity in diabetes and Alzheimer’s disease: Mechanisms behind ROS/RNS generation. Journal of Biomedical Science. 2017;24:1-10. DOI: 10.1186/s12929-017-0379-z
  29. 29. Knight JA. Free radicals, antioxidants, and the immune system. Annals of Clinical and Laboratory Science. 2000;30:145-158
  30. 30. Zhao Y, Zhao B. Oxidative stress and the pathogenesis of Alzheimer's disease. Longevity OMAC. 2013;2013:316523. DOI: 10.1155/2013/316523
  31. 31. Selfridge JE, Lezi E, Lu J, Swerdlow RH. Role of mitochondrial homeostasis and dynamics in Alzheimer's disease. Neurobiology of Disease. 2013;51:3-12. DOI: 10.1016/j.nbd.2011.12.057
  32. 32. Newsholme P, Cruzat VF, Keane KN, Carlessi R, de Bittencourt JPIH. Molecular mechanisms of ROS production and oxidative stress in diabetes. The Biochemical Journal. 2016;473:4527-4550. DOI: 10.1042/BCJ20160503C
  33. 33. Zhang Z, Yang H, Yang J, Xie J, Xu J, Liu C, et al. Pseudoginsenoside-F11 attenuates cognitive impairment by ameliorating oxidative stress and neuroinflammation in d-galactose-treated mice. International Immunopharmacology. 2019;67:78-86. DOI: 10.1016/j.intimp.2018.11.026
  34. 34. Xiang T, Luo X, Ye L, Huang H, Wu Y. Klotho alleviates NLRP3 inflammasome-mediated neuroinflammation in a temporal lobe epilepsy rat model by activating the Nrf2 signaling pathway. Epilepsy & Behavior. 2022;128:108509. DOI: 10.1016/j.yebeh.2021.108509
  35. 35. Madhu LN, Kodali M, Attaluri S, Shuai B, Melissari L, Rao X, et al. Melatonin improves brain function in a model of chronic gulf war illness with modulation of oxidative stress, NLRP3 inflammasomes, and BDNF-ERK-CREB pathway in the hippocampus. Redox Biology. 2021;43:101973. DOI: 10.1016/j.redox.2021.101973
  36. 36. Singh S, Vrishni S, Singh BK, Rahman I, Kakkar P. Nrf2-ARE stress response mechanism: A control point in oxidative stress-mediated dysfunctions and chronic inflammatory diseases. Free Radical Research. 2010;44:1267-1288. DOI: 10.3109/10715762.2010.507670
  37. 37. Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. The Journal of Biological Chemistry. 2009;284:13291-13295. DOI: 10.1074/jbc.R900010200
  38. 38. Du L, Wang J, Chen Y, Li X, Wang L, Li Y, et al. Novel biphenyl diester derivative AB-38b inhibits NLRP3 inflammasome through Nrf2 activation in diabetic nephropathy. Cell Biology and Toxicology. 2020;36:243-260. DOI: 10.1007/s10565-019-09501-8
  39. 39. Sun Y, Yang T, Leak K. Preventive and protective roles of dietary Nrf2 activators against central nervous system diseases. CNS & Neurological Disorders Drug Targets. 2017;16:326-338. DOI: 10.2174/1871527316666170102120211
  40. 40. Scannevin RH, Chollate S, et al. Fumarates promote cytoprotection of central nervous system cells against oxidative stress via the nuclear factor (erythroid-derived 2)-like 2 pathway. Journal of Pharmacology. 2012;341:274-284
  41. 41. Lee JM, Li J, Johnson DA, Stein TD, Kraft AD, Calkins MJ, et al. Nrf2, a multi-organ protector? The FASEB Journal. 2005;19:1061-1066. DOI: 10.1096/fj.04-2591hyp
  42. 42. Heurtaux T, Bouvier DS, Benani A, Helgueta Romero S, Frauenknecht KB, Mittelbronn M, et al. Normal and pathological NRF2 signalling in the central nervous system. Antioxidants. 2022;11:1426. DOI: 10.3390/antiox11081426
  43. 43. Yang Y, Jiang S, Yan J, Li Y, Xin Z, Lin Y, et al. An overview of the molecular mechanisms and novel roles of Nrf2 in neurodegenerative disorders. Cytokine & Growth Factor Reviews. 2015;26:47-57. DOI: 10.1016/j.cytogfr.2014.09.002
  44. 44. Tavakkoli A, Iranshahi M, Hasheminezhad SH, Hayes AW, Karimi G. The neuroprotective activities of natural products through the Nrf2 upregulation. Phytotherapy Research. 2019;33:2256-2273. DOI: 10.1002/ptr.6427
  45. 45. Zhang M, An C, Gao Y, Leak RK, Chen J, Zhang F. Emerging roles of Nrf2 and phase II antioxidant enzymes in neuroprotection. Progress in Neurobiology. 2013;100:30-47. DOI: 10.1016/j.pneurobio.2012.09.003
  46. 46. Wang J, Shen X, Liu J, Chen W, Wu F, Wu W, et al. High glucose mediates NLRP3 inflammasome activation via upregulation of ELF3 expression. Cell Death & Disease. 2020;11:1-14. DOI: 10.1038/s41419-020-2598-6
  47. 47. Hung WL, Ho CT, Pan MH. Targeting the NLRP3 inflammasome in neuroinflammation: Health promoting effects of dietary phytochemicals in neurological disorders. Molecular Nutrition & Food Research. 2020;64:e1900550. DOI: 10.1002/mnfr.201900550
  48. 48. Hu H, Zhu T, Gong L, Zhao Y, Shao Y, Li S, et al. Transient receptor potential melastatin 2 contributes to neuroinflammation and negatively regulates cognitive outcomes in a pilocarpine-induced mouse model of epilepsy. International Immunopharmacology. 2020;87:106824. DOI: 10.1016/j.intimp.2020.106824
  49. 49. Rong S, Wan D, Fan Y, Liu S, Sun K, Huo J, et al. Amentoflavone affects epileptogenesis and exerts neuroprotective effects by inhibiting NLRP3 inflammasome. Frontiers in Pharmacology. 2019;10:856. DOI: 10.3389/fphar.2019.00856
  50. 50. Kanneganti T-D. Central roles of NLRs and inflammasomes in viral infection. Nature Reviews. Immunology. 2010;10:688-698. DOI: 10.1038/nri2851
  51. 51. Kitazawa M, Cheng D, Tsukamoto MR, Koike MA, Wes PD, Vasilevko V, et al. Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal β-catenin pathway function in an Alzheimer’s disease model. Journal of Immunology. 2011;187:6539-6549. DOI: 10.4049/jimmunol.1100620
  52. 52. Lu J, Wu D-m, Zheng Y-l, Hu B, Zhang Z-f, Ye Q , et al. Ursolic acid attenuates D-galactose-induced inflammatory response in mouse prefrontal cortex through inhibiting AGEs/RAGE/NF-κB pathway activation. Cerebral Cortex. 2010;20:2540-2548. DOI: 10.1093/cercor/bhq002
  53. 53. Cui X, Zuo P, Zhang Q , Li X, Hu Y, Long J, et al. Chronic systemic D-galactose exposure induces memory loss, neurodegeneration, and oxidative damage in mice: Protective effects of R-α-lipoic acid. Journal of Neuroscience Research. 2006;83:1584-1590. DOI: 10.1002/jnr.20899
  54. 54. Wang H, Lv J, Jiang N, Huang H, Wang Q , Liu X. Ginsenoside Re protects against chronic restraint stress-induced cognitive deficits through regulation of NLRP3 and Nrf2 pathways in mice. Phytotherapy Research. 2021;35:2523-2535. DOI: 10.1002/ptr.6947
  55. 55. Kobayashi EH, Suzuki T, Funayama R, Nagashima T, Hayashi M, Sekine H, et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nature Communications. 2016;7:1-14. DOI: 10.1038/ncomms11624
  56. 56. Mills EL, Ryan DG, Prag HA, Dikovskaya D, Menon D, Zaslona Z, et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature. 2018;556:113-117. DOI: 10.1038/nature25986
  57. 57. Yeh W-J, Yang H-Y, Pai M-H, Wu C-H, Chen J-R. Long-term administration of advanced glycation end-product stimulates the activation of NLRP3 inflammasome and sparking the development of renal injury. The Journal of Nutritional Biochemistry. 2017;39:68-76. DOI: 10.1016/j.jnutbio.2016.09.014
  58. 58. Song Y, Wang Y, Zhang Y, Geng W, Liu W, Gao Y, et al. Advanced glycation end products regulate anabolic and catabolic activities via NLRP3-inflammasome activation in human nucleus pulposus cells. Journal of Cellular and Molecular Medicine. 2017;21:1373-1387. DOI: 10.1111/jcmm.13067
  59. 59. Juliana C, Fernandes-Alnemri T, Kang S, Farias A, Qin F, Alnemri ES. Non-transcriptional priming and deubiquitination regulate NLRP3 inflammasome activation. The Journal of Biological Chemistry. 2012;287:36617-36622. DOI: 10.1074/jbc.M112.407130
  60. 60. Li Y-N, Guo Y, Xi M-M, Yang P, Zhou X-Y, Yin S, et al. Saponins from Aralia taibaiensis attenuate D-galactose-induced aging in rats by activating FOXO3a and Nrf2 pathways. Oxidative Medicine and Cellular Longevity. 2014;2014:320513. DOI: 10.1155/2014/320513
  61. 61. Lv J, Su W, Yu Q , Zhang M, Di C, Lin X, et al. Heme oxygenase-1 protects airway epithelium against apoptosis by targeting the proinflammatory NLRP3–RXR axis in asthma. The Journal of Biological Chemistry. 2018;293:18454-18465. DOI: 10.1074/jbc.RA118.004950
  62. 62. Kim S-J, Lee S-M. NLRP3 inflammasome activation in D-galactosamine and lipopolysaccharide-induced acute liver failure: Role of heme oxygenase-1. Free Radical Biology & Medicine. 2013;65:997-1004. DOI: 10.1016/j.freeradbiomed.2013.08.178
  63. 63. Wang K, Lv Q , Miao Y-m, Qiao S-m, Dai Y, Wei Z-f. Cardamonin, a natural flavone, alleviates inflammatory bowel disease by the inhibition of NLRP3 inflammasome activation via an AhR/Nrf2/NQO1 pathway. Biochemical Pharmacology. 2018;155:494-509. DOI: 10.1016/j.bcp.2018.07.039
  64. 64. Schröder M. Endoplasmic reticulum stress responses. Cellular and Molecular Life Sciences. 2008;65:862-894. DOI: 10.1007/s00018-007-7383-5
  65. 65. Tengholm A, Hellman B, Gylfe E. The endoplasmic reticulum is a glucose-modulated high-affinity sink for Ca2+ in mouse pancreatic β-cells. The Journal of Physiology. 2001;530:533. DOI: 10.1111/j.1469-7793.2001.0533k.x
  66. 66. Guo Y, Zhang C, Wang C, Huang Y, Liu J, Chu H, et al. Thioredoxin-1 is a target to attenuate alzheimer-like pathology in diabetic encephalopathy by alleviating endoplasmic reticulum stress and oxidative stress. Frontiers in Physiology. 2021;12:651105. DOI: 10.3389/fphys.2021.651105
  67. 67. Gregersen N, Bross P. Protein misfolding and cellular stress: An overview. Methods in Molecular Biology. 2010;648:3-23. DOI: 10.1007/978-1-60761-756-3_1
  68. 68. Ye J, Han Y, Chen X, Xie J, Liu X, Qiao S, et al. L-carnitine attenuates H2O2-induced neuron apoptosis via inhibition of endoplasmic reticulum stress. Neurochemistry International. 2014;78:86-95. DOI: 10.1016/j.neuint.2014.08.009
  69. 69. Cai M et al. The molecular mechanisms of excessive hippocampal endoplasmic reticulum stress depressing cognition-related proteins expression and the regulatory effects of Nrf2. Neuroscience. 2020;431:152-165. DOI: 10.1016/j.neuroscience.2020.02.001
  70. 70. Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, et al. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-β. Nature. 2000;403:98-103. DOI: 10.1038/47513
  71. 71. Oyadomari S, Mori M. Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death and Differentiation. 2004;11:381-389. DOI: 10.1038/sj.cdd.4401373
  72. 72. Ye T, Meng X, Zhai Y, Xie W, Wang R, Sun G, et al. Gastrodin ameliorates cognitive dysfunction in diabetes rat model via the suppression of endoplasmic reticulum stress and NLRP3 inflammasome activation. Frontiers in Pharmacology. 2018;9:1346. DOI: 10.3389/fphar.2018.01346
  73. 73. Zhu Y-F, Li X-H, Yuan Z-P, Li C-Y, Tian R-B, Jia W, et al. Allicin improves endoplasmic reticulum stress-related cognitive deficits via PERK/Nrf2 antioxidative signaling pathway. European Journal of Pharmacology. 2015;762:239-246. DOI: 10.1016/j.ejphar.2015.06.002
  74. 74. Cullinan SB, Diehl JA. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. The Journal of Biological Chemistry. 2004;279:20108-20117. DOI: 10.1074/jbc.M314219200
  75. 75. Rom S, Zuluaga-Ramirez V, Gajghate S, Seliga A, Winfield M, Heldt NA, et al. Hyperglycemia-driven neuroinflammation compromises BBB leading to memory loss in both diabetes mellitus (DM) type 1 and type 2 mouse models. Molecular Neurobiology. 2019;56:1883-1896. DOI: 10.1007/s12035-018-1195-5
  76. 76. Bogush M, Heldt NA, Persidsky Y. Blood brain barrier injury in diabetes: Unrecognized effects on brain and cognition. Journal of Neuroimmune Pharmacology. 2017;12:593-601. DOI: 10.1007/s11481-017-9752-7
  77. 77. Prasad S, Sajja RK, Naik P, Cucullo L. Diabetes mellitus and blood-brain barrier dysfunction: An overview. Journal of Pharmacovigil. 2014;2:125. DOI: 10.4172/2329-6887.1000125
  78. 78. Van Dyken P, Lacoste B. Impact of metabolic syndrome on neuroinflammation and the blood–brain barrier. Frontiers in Neuroscience. 2018;12:930. DOI: 10.3389/fnins.2018.00930
  79. 79. Sajja RK, Prasad S, Tang S, Kaisar MA, Cucullo L. Blood-brain barrier disruption in diabetic mice is linked to Nrf2 signaling deficits: Role of ABCB10? Neuroscience Letters. 2017;653:152-158. DOI: 10.1016/j.neulet.2017.05.059
  80. 80. Chen B, Lu Y, Chen Y, Cheng J. The role of Nrf2 in oxidative stress-induced endothelial injuries. The Journal of Endocrinology. 2015;225:R83-R99. DOI: 10.1530/JOE-14-0662
  81. 81. Yang M-y, Fan Z, Zhang Z, Fan J. MitoQ protects against high glucose-induced brain microvascular endothelial cells injury via the Nrf2/HO-1 pathway. Journal of Pharmacological Sciences. 2021;145:105-114. DOI: 10.1016/j.jphs.2020.10.007
  82. 82. Esteras N, Dinkova-Kostova AT, Abramov AY. Nrf2 activation in the treatment of neurodegenerative diseases: A focus on its role in mitochondrial bioenergetics and function. Biological Chemistry. 2016;397:383-400. DOI: 10.1515/hsz-2015-0295
  83. 83. Fernandes V, Choudhary M, Kumar A, Singh SB. Proteotoxicity and mitochondrial dynamics in aging diabetic brain. Pharmacological Research. 2020;159:104948. DOI: 10.1016/j.phrs.2020.104948
  84. 84. Luo J-S, Ning J-Q , Chen Z-Y, Li W-J, Zhou R-L, Yan R-Y, et al. The role of mitochondrial quality control in cognitive dysfunction in diabetes. Neurochemical Research. 2022;8:2158-2172. DOI: 10.1007/s11064-022-03631-y
  85. 85. Burchell VS, Gandhi S, Deas E, Wood NW, Abramov AY, Plun-Favreau H. Targeting mitochondrial dysfunction in neurodegenerative disease: Part I. Expert Opinion on Therapeutic Targets. 2010;14:369-385. DOI: 10.1517/14728221003652489
  86. 86. Biswas S, Chida AS, Rahman I. Redox modifications of protein–thiols: Emerging roles in cell signaling. Biochemical Pharmacology. 2006;71:551-564. DOI: 10.1016/j.bcp.2005.10.044
  87. 87. Dodson M, Redmann M, Rajasekaran NS, Darley-Usmar V, Zhang J. KEAP1–NRF2 signalling and autophagy in protection against oxidative and reductive proteotoxicity. The Biochemical Journal. 2015;469:347-355. DOI: 10.1042/BJ20150568
  88. 88. Tebay LE, Robertson H, Durant ST, Vitale SR, Penning TM, Dinkova-Kostova AT, et al. Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it attenuates degenerative disease. Free Radical Biology & Medicine. 2015;88:108-146. DOI: 10.1016/j.freeradbiomed.2015.06.021
  89. 89. Chen H, Tang X, Zhou B, Zhou Z, Xu N, Wang Y. A ROS-mediated mitochondrial pathway and Nrf2 pathway activation are involved in BDE-47 induced apoptosis in Neuro-2a cells. Chemosphere. 2017;184:679-686. DOI: 10.1016/j.chemosphere.2017.06.006
  90. 90. Gao J, Liu S, Xu F, Liu Y, Lv C, Deng Y, et al. Trilobatin protects against oxidative injury in neuronal PC12 cells through regulating mitochondrial ROS homeostasis mediated by AMPK/Nrf2/Sirt3 signaling pathway. Frontiers in Molecular Neuroscience. 2018;11:267. DOI: 10.3389/fnmol.2018.00267
  91. 91. Ebrahimi R, Sepand MR, Seyednejad SA, Omidi A, Akbariani M, Gholami M, et al. Ellagic acid reduces methotrexate-induced apoptosis and mitochondrial dysfunction via up-regulating Nrf2 expression and inhibiting the IĸBα/NFĸB in rats. Daru. 2019;27:721-733. DOI: 10.1007/s40199-019-00309-9
  92. 92. Chorley BN, Campbell MR, Wang X, Karaca M, Sambandan D, Bangura F, et al. Identification of novel NRF2-regulated genes by ChIP-Seq: Influence on retinoid X receptor alpha. Nucleic Acids Research. 2012;40:7416-7429. DOI: 10.1093/nar/gks409
  93. 93. Kawamura T, Umemura T, Hotta N. Cognitive impairment in diabetic patients: Can diabetic control prevent cognitive decline? Journal of Diabetes Investigation. 2012;3:413-423. DOI: 10.1111/j.2040-1124.2012.00234.x
  94. 94. Polvani S, Tarocchi M, Galli A. PPARγ and oxidative stress: Con (β) catenating NRF2 and FOXO. PPAR Research. 2012;2012:641087. DOI: 10.1155/2012/641087
  95. 95. Sharma V, Kaur A, Singh TG. Counteracting role of nuclear factor erythroid 2-related factor 2 pathway in Alzheimer's disease. Biomedicine & Pharmacotherapy. 2020;129:110373. DOI: 10.1016/j.biopha.2020.110373
  96. 96. Mannan A, Garg N, Singh TG, Kang HK. Peroxisome proliferator-activated receptor-gamma (PPAR-ɣ): Molecular effects and its importance as a novel therapeutic target for cerebral ischemic injury. Neurochemical Research. 2021;46:2800-2831. DOI: 10.1007/s11064-021-03402-1
  97. 97. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, et al. PPARγ signaling and metabolism: The good, the bad and the future. Nature Medicine. 2013;19:557-566. DOI: 10.1038/nm.3159
  98. 98. Cai W, Yang T, Liu H, Han L, Zhang K, Hu X, et al. Peroxisome proliferator-activated receptor γ (PPARγ): A master gatekeeper in CNS injury and repair. Progress in Neurobiology. 2018;163:27-58. DOI: 10.1016/j.pneurobio.2017.10.002
  99. 99. Thapa K, Khan H, Sharma U, Grewal AK, Singh TG. Poly (ADP-ribose) polymerase-1 as a promising drug target for neurodegenerative diseases. Life Sciences. 2021;267:118975. DOI: 10.1016/j.lfs.2020.118975
  100. 100. Pu D, Zhao Y, Chen J, Lv A, Zhu S, Luo C, et al. Protective effects of sulforaphane on cognitive impairments and AD-like lesions in diabetic mice are associated with the upregulation of Nrf2 transcription activity. Neuroscience. 2018;381:35-45. DOI: 10.1016/j.neuroscience.2018.04.017
  101. 101. Wang G, Fang H, Zhen Y, Xu G, Tian J, Zhang Y, et al. Sulforaphane prevents neuronal apoptosis and memory impairment in diabetic rats. Cellular Physiology and Biochemistry. 2016;39:901-907. DOI: 10.1159/000447799
  102. 102. Zhang S, Yuan L, Zhang L, Li C, Li J. Prophylactic use of troxerutin can delay the development of diabetic cognitive dysfunction and improve the expression of Nrf2 in the hippocampus on STZ diabetic rats. Behavioural Neurology. 2018;2018:8678539. DOI: 10.1155/2018/8678539
  103. 103. Gao M, Kang Y, Zhang L, Li H, Qu C, Luan X, et al. Troxerutin attenuates cognitive decline in the hippocampus of male diabetic rats by inhibiting NADPH oxidase and activating the Nrf2/ARE signaling pathway. International Journal of Molecular Medicine. 2020;46:1239-1248. DOI: 10.3892/ijmm.2020.4653
  104. 104. Zhang L, Ma Q , Zhou Y. Strawberry leaf extract treatment alleviates cognitive impairment by activating Nrf2/HO-1 signaling in rats with streptozotocin-induced diabetes. Frontiers in Aging Neuroscience. 2020;12:201. DOI: 10.3389/fnagi.2020.00201
  105. 105. Ma C, Long H. Protective effect of betulin on cognitive decline in streptozotocin (STZ)-induced diabetic rats. Neurotoxicology. 2016;57:104-111
  106. 106. Baluchnejadmojarad T, Kiasalari Z, Afshin-Majd S, Ghasemi Z, Roghani M. S-allyl cysteine ameliorates cognitive deficits in streptozotocin-diabetic rats via suppression of oxidative stress, inflammation, and acetylcholinesterase. European Journal of Pharmacology. 2017;794:69-76. DOI: 10.1016/j.ejphar.2016.11.033
  107. 107. Luo Z, Wan Q , Han Y, Li Z, Li B. CAPE-pNO2 ameliorates diabetic brain injury through modulating Alzheimer's disease key proteins, oxidation, inflammation and autophagy via a Nrf2-dependent pathway. Life Sciences. 2021;287:119929
  108. 108. Chen Y-J, Tang Z-Z, Du L, Liu Y, Lu Q , Ma T-F, et al. A novel compound AB-38b improves diabetes-associated cognitive decline in mice via activation of Nrf2/ARE pathway. Brain Research Bulletin. 2019;150:160-167
  109. 109. Wang G, Zhang X, Lu X, Liu J, Zhang Z, Wei Z, et al. Fish oil supplementation attenuates cognitive impairment by inhibiting neuroinflammation in STZ-induced diabetic rats. Aging (Albany NY). 2020;12:15281-15289. DOI: 10.18632/aging.103426
  110. 110. Ke B, Zhang T, An T, Lu R. Soy isoflavones ameliorate the cognitive dysfunction of Goto-Kakizaki rats by activating the Nrf2-HO-1 signalling pathway. Aging (Albany NY). 2020;12:21344-21354. DOI: 10.18632/aging.103877
  111. 111. Wang X, Fang H, Xu G, Yang Y, Xu R, Liu Q , et al. Resveratrol prevents cognitive impairment in type 2 diabetic mice by upregulating Nrf2 expression and transcriptional level. Diabetes Metabolosm Syndrome in Obesity. 2020;13:1061-1075. DOI: 10.2147/DMSO.S243560
  112. 112. Sadi G, Konat D. Resveratrol regulates oxidative biomarkers and antioxidant enzymes in the brain of streptozotocin-induced diabetic rats. Pharmaceutical Biology. 2016;54:1156-1163. DOI: 10.3109/13880209.2015
  113. 113. Li H, Qin T, Li M, Ma S. Thymol improves high-fat diet-induced cognitive deficits in mice via ameliorating brain insulin resistance and upregulating NRF2/HO-1 pathway. Metabolic Brain Disease. 2017;32:385-393. DOI: 10.1007/s11011-016-9921-z
  114. 114. Feng Y, Chu A, Luo Q , Wu M, Shi X, Chen Y. The protective effect of astaxanthin on cognitive function via inhibition of oxidative stress and inflammation in the brains of chronic T2DM rats. Frontiers in Pharmacology. 2018;9:748. DOI: 10.3389/fphar.2018.00748
  115. 115. Zhai Y, Meng X, Luo Y, Wu Y, Ye T, Zhou P, et al. Notoginsenoside R1 ameliorates diabetic encephalopathy by activating the Nrf2 pathway and inhibiting NLRP3 inflammasome activation. Oncotarget. 2018;9:9344-9363. DOI: 10.18632/oncotarget.24295
  116. 116. Liu Y-W, Liu X-L, Kong L, Zhang M-Y, Chen Y-J, Zhu X, et al. Neuroprotection of quercetin on central neurons against chronic high glucose through enhancement of Nrf2/ARE/glyoxalase-1 pathway mediated by phosphorylation regulation. Biomedicine & Pharmacotherapy. 2019;109:2145-2154. DOI: 10.1016/j.biopha.2018.11.066
  117. 117. Singh NK, Garabadu D. Quercetin exhibits α7nAChR/Nrf2/HO-1-mediated neuroprotection against STZ-induced mitochondrial toxicity and cognitive impairments in experimental rodents. Neurotoxicity Research. 2021;39:1859-1879. DOI: 10.1007/s12640-021-00410-5
  118. 118. Ghawi SK, Methven L, Niranjan K. The potential to intensify sulforaphane formation in cooked broccoli (Brassica oleracea var. italica) using mustard seeds (Sinapis alba). Food Chemistry. 2013;138:1734-1741. DOI: 10.1016/j.foodchem.2012.10.119
  119. 119. Hwang E-S, Jeffery EH. Induction of quinone reductase by sulforaphane and sulforaphane N-acetylcysteine conjugate in murine hepatoma cells. Journal of Medicinal Food. 2005;8:198-203. DOI: 10.1089/jmf.2005.8.198
  120. 120. McNaughton S, Marks G. Development of a food composition database for the estimation of dietary intakes of glucosinolates, the biologically active constituents of cruciferous vegetables. The British Journal of Nutrition. 2003;90:687-697. DOI: 10.1079/bjn2003917
  121. 121. Sones K, Heaney RK, Fenwick GR. An estimate of the mean daily intake of glucosinolates from cruciferous vegetables in the UK. Journal of the Science of Food and Agriculture. 1984;35:712-720
  122. 122. Dash PK, Zhao J, Orsi SA, Zhang M, Moore AN. Sulforaphane improves cognitive function administered following traumatic brain injury. Neuroscience Letters. 2009;460:103-107. DOI: 10.1016/j.neulet.2009.04.028
  123. 123. Sun Y, Yang T, Mao L, Zhang F. Sulforaphane protects against brain diseases: Roles of cytoprotective enzymes. Austin Journal of Cerebrovascular Diseases. 2017:4
  124. 124. Su Z-Y, Zhang C, Lee JH, Shu L, Wu T-Y, Khor TO, et al. Requirement and epigenetics reprogramming of Nrf2 in suppression of tumor promoter TPA-induced mouse skin cell transformation by SulforaphaneActivating Nrf2 blocks TPA-induced JB6 transformation by SFN. Cancer Prevention Research (Philadelphia, Pa.). 2014;7:319-329. DOI: 10.1158/1940-6207.CAPR-13-0313-T
  125. 125. Hu R, Xu C, Shen G, Jain MR, Khor TO, Gopalkrishnan A, et al. Gene expression profiles induced by cancer chemopreventive isothiocyanate sulforaphane in the liver of C57BL/6J mice and C57BL/6J/Nrf2 (−/−) mice. Cancer Letters. 2006;243:170-192. DOI: 10.1016/j.canlet.2005.11.050
  126. 126. Hu C, Eggler AL, Mesecar AD, Van Breemen RB. Modification of keap1 cysteine residues by sulforaphane. Chemical Research in Toxicology. 2011;24:515-521. DOI: 10.1021/tx100389r
  127. 127. Hong F, Freeman ML, Liebler DC. Identification of sensor cysteines in human Keap1 modified by the cancer chemopreventive agent sulforaphane. Chemical Research in Toxicology. 2005;18:1917-1926. DOI: 10.1021/tx0502138
  128. 128. Shang G, Tang X, Gao P, Guo F, Liu H, Zhao Z, et al. Sulforaphane attenuation of experimental diabetic nephropathy involves GSK-3 beta/Fyn/Nrf2 signaling pathway. The Journal of Nutritional Biochemistry. 2015;26:596-606. DOI: 10.1016/j.jnutbio.2014.12.008
  129. 129. Bergström P, Andersson HC, Gao Y, Karlsson J-O, Nodin C, Anderson MF, et al. Repeated transient sulforaphane stimulation in astrocytes leads to prolonged Nrf2-mediated gene expression and protection from superoxide-induced damage. Neuropharmacology. 2011;60:343-353. DOI: 10.1016/j.neuropharm.2010.09.023
  130. 130. Uddin MS, Al Mamun A, Jakaria M, Thangapandiyan S, Ahmad J, Rahman MA, et al. Emerging promise of sulforaphane-mediated Nrf2 signaling cascade against neurological disorders. Science Total Environment. 2020;707:135624. DOI: 10.1016/j.scitotenv.2019.135624
  131. 131. Ashrafizadeh M, Ahmadi Z, Yaribeygi H, Sathyapalan T, Sahebkar A. Astaxanthin and Nrf2 signaling pathway: A novel target for new therapeutic approaches. Mini Reviews in Medicinal Chemistry. 2022;22:312-321. DOI: 10.2174/1389557521666210505112834
  132. 132. Chen Y, Tang J, Zhang Y, Du J, Wang Y, Yu H, et al. Astaxanthin alleviates gestational diabetes mellitus in mice through suppression of oxidative stress. Naunyn-Schmiedeberg's Archives of Pharmacology. 2020;393:2517-2527. DOI: 10.1007/s00210-020-01861-x
  133. 133. Jeong SM, Kim Y-J. Astaxanthin treatment induces maturation and functional change of myeloid-derived suppressor cells in tumor-bearing mice. Antioxidants. 2020;9:350. DOI: 10.3390/antiox9040350
  134. 134. Mohammadi S, Barzegari A, Dehnad A, Barar J, Omidi Y. Astaxanthin protects mesenchymal stem cells from oxidative stress by direct scavenging of free radicals and modulation of cell signaling. Chemico-Biological Interactions. 2021;333:109324. DOI: 10.1016/j.cbi.2020.109324
  135. 135. Yuan L, Qu Y, Li Q , An T, Chen Z, Chen Y, et al. Protective effect of astaxanthin against La2O3 nanoparticles induced neurotoxicity by activating PI3K/AKT/Nrf-2 signaling in mice. Food and Chemical Toxicology. 2020;144:111582. DOI: 10.1016/j.fct.2020.111582
  136. 136. Landon R, Gueguen V, Petite H, Letourneur D, Pavon-Djavid G, Anagnostou F. Impact of astaxanthin on diabetes pathogenesis and chronic complications. Marine Drugs. 2020;18:357. DOI: 10.3390/md18070357
  137. 137. Xu L, Zhu J, Yin W, Ding X. Astaxanthin improves cognitive deficits from oxidative stress, nitric oxide synthase and inflammation through upregulation of PI3K/Akt in diabetes rat. International Journal of Clinical Pathology. 2015;8:6083
  138. 138. Huang D-D, Shi G, Jiang Y, Yao C, Zhu C. A review on the potential of resveratrol in prevention and therapy of diabetes and diabetic complications. Biomedicine & Pharmacotherapy. 2020;125:109767. DOI: 10.1016/j.biopha.2019.109767
  139. 139. Pan MH, Wu JC, Ho CT, Lai CS. Antiobesity molecular mechanisms of action: Resveratrol and pterostilbene. BioFactors. 2018;44:50-60. DOI: 10.1002/biof.1409
  140. 140. Yang DK, Kang H-S. Anti-diabetic effect of cotreatment with quercetin and resveratrol in streptozotocin-induced diabetic rats. Biomolecular Therapy (Seoul). 2018;26:130. DOI: 10.4062/biomolther.2017.254
  141. 141. Elshaer M, Chen Y, Wang XJ, Tang X. Resveratrol: An overview of its anti-cancer mechanisms. Life Sciences. 2018;207:340-349. DOI: 10.1016/j.lfs.2018.06.028
  142. 142. De La Lastra CA, Villegas I. Resveratrol as an anti-inflammatory and anti-aging agent: Mechanisms and clinical implications. Molecular Nutrition & Food Research. 2005;49:405-430. DOI: 10.1002/mnfr.200500022
  143. 143. Seyyedebrahimi S, Khodabandehloo H, Nasli Esfahani E, Meshkani R. The effects of resveratrol on markers of oxidative stress in patients with type 2 diabetes: A randomized, double-blind, placebo-controlled clinical trial. Acta Diabetologica. 2018;55:341-353. DOI: 10.1007/s00592-017-1098-3
  144. 144. Ebrahimpour S, Zakeri M, Esmaeili A. Crosstalk between obesity, diabetes, and alzheimer’s disease: Introducing quercetin as an effective triple herbal medicine. Ageing Research Reviews. 2020;62:101095. DOI: 10.1016/j.arr.2020.101095
  145. 145. Sun GY, Chen Z, Jasmer KJ, Chuang DY, Gu Z, Hannink M, et al. Quercetin attenuates inflammatory responses in BV-2 microglial cells: Role of MAPKs on the Nrf2 pathway and induction of heme oxygenase-1. PLoS One. 2015;10:e0141509. DOI: 10.1371/journal.pone.0141509
  146. 146. Wang D, Zhao J, Li S, Shen G, Hu S. Quercetin attenuates domoic acid-induced cognitive deficits in mice. Nutritional Neuroscience. 2018;21:123-131. DOI: 10.1080/1028415X.2016.1231438
  147. 147. Yano S, Tokumitsu H, Soderling TR. Calcium promotes cell survival through CaM-K kinase activation of the protein-kinase-B pathway. Nature. 1998;396:584-587. DOI: 10.1038/25147
  148. 148. Hajduch E, Litherland GJ, Hundal HS. Protein kinase B (PKB/Akt)–a key regulator of glucose transport? FEBS Letters. 2001;492:199-203. DOI: 10.1016/s0014-5793(01)02242-6

Written By

Xian Zhou, Gerald Münch and Dennis Chang

Submitted: 05 July 2022 Reviewed: 24 October 2022 Published: 28 November 2022