Next Article in Journal
RHO-Associated Coiled-Coil-Containing Protein Kinase Inhibitors Significantly Modulate the Epithelial–Mesenchymal Transition Induced by TGF-β2 in the 2-D and 3-D Cultures of Human Corneal Stroma Fibroblasts
Previous Article in Journal
Adalimumab Autoantibodies in Uveitis Patients: Do We Need Routine Drug Monitoring?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Promising Potency of Sodium–Glucose Cotransporter 2 Inhibitors in the Prevention of and as Treatment for Cognitive Impairment Among Type 2 Diabetes Patients

1
Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
2
Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
3
Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(12), 2783; https://doi.org/10.3390/biomedicines12122783
Submission received: 3 November 2024 / Revised: 28 November 2024 / Accepted: 4 December 2024 / Published: 6 December 2024
(This article belongs to the Section Endocrinology and Metabolism Research)

Abstract

:
Type 2 diabetes mellitus (T2DM), accounting for the majority of diabetes mellitus prevalence, is associated with an increased risk of cognition decline and deterioration of cognition function in diabetic patients. The sodium–glucose cotransporter 2 (SGLT2), located in the renal proximal tubule, plays a role in urine glucose reabsorption. SGLT2 inhibitors (SGLT2i), have shown potential benefits beyond cardiac and renal improvement in preventing and treating cognitive impairment (CI), including mild cognitive impairment, Alzheimer’s disease and vascular dementia in T2DM patients. Studies suggest that SGLT2i may ameliorate diabetic CI through metabolism pathways, inflammation, oxidative stress, neurotrophic factors and AChE inhibition. Clinical trials and meta-analyses have reported significant and insignificant results. Given their vascular effects, SGLT2i may offer unique protection against vascular CI. This review compiles mechanisms and clinical evidence, emphasizing the need for future analysis, evaluation, trials and meta-analyses to verify and recommend optimal SGLT2i selection and dosage for specific patients.

1. Introduction

DM is a metabolic syndrome characterized by chronic hyperglycemia. T2DM, as a subtype of DM, originates from insulin resistance, leading to progressive pancreatic β cell failure and insulin insufficiency [1,2,3]. It is estimated that 529 million people worldwide live with diabetes, with a global age-standardized total diabetes prevalence of 6.1%, and this number is projected to exceed 1.31 billion by 2050 [4]. T2DM accounts for nearly 96.0% of all diabetes cases [4]. Additionally, the number of people with IGT and IFG is expected to rise from 464 million and 298 million in 2021 to 638 million and 414 million, respectively, by 2050, posing a huge burden on global health [5].
CI, which is classified into MCI and dementia, is defined as a significant and progressive decline in one or more cognitive domains compared to a person’s previous level, including learning and memory, social cognition, complex attention, executive function, perceptual-motor function and language [6]. MCI, also known as predementia, is distinguished from dementia by its impact on functional abilities conducting one’s daily life, with no insignificant impairment in social or occupational functioning [6]. Dementia is a group of heterogeneous diseases with various etiologies, including AD, VaD and other subtypes [7]. AD, the most common subtype of dementia, typically presents as an amnestic variant [8]. Confirming a diagnosis of AD requires neuropathologic evidence of both amyloid plaques from Aβ peptides and NFTs composed of hyperphosphorylated and abnormally folded tau proteins [9]. VaD, the second most common cause of dementia, accounts for 15% of cases [10] and is characterized by vascular changes, such as infarcts, atherosclerosis, arteriolosclerosis and CAA, which have independent and additive effects on the risk of CI [11]
Diabetic patients are at significantly higher risk of various complications, including CI. A population-based prospective follow-up study conducted by A. Ott et al. in 1996 suggested that diabetes may increase the risk of dementia onset, with VaD contributing modestly to the incidence of dementia in T2DM [12]. Recent analysis of cohort studies has identified diabetes as a risk factor for dementia, with a weighted population attributable fraction of 1.9% [13]. Another meta-analysis of DM patients in Africa showed that poorly controlled diabetes and a diabetes duration of over 10 years are risk factors associated with CI, with pooled OR (a statistical measure used to summarize the OR from multiple studies in a meta-analysis) of 5.85 and 1.13, respectively [14]. In 2024, a systematic umbrella review of diabetic CI evidence revealed a significant association between diabetes and an increased risk of AD and VaD [15]. A longitudinal study in 2024 reported that patients experienced a significantly faster rate of cognitive decline after the onset of diabetes (difference in slope after diabetes onset −0.23, 95%CI −0.043 to −0.004, p = 0.019), concluding that diabetes might accelerate the deterioration of cognitive function [16]. The ADA in 2024 stated that elderly individuals with diabetes are at higher risk of CI and recommended they be screened and monitored for CI using simple assessment methods such as the MMSE or MoCA [17].
SGLT2 is primarily expressed in the renal proximal tubules and is responsible for glucose reabsorption. SGLT2i, including canagliflozin, dapagliflozin, empagliflozin, ertugliflozin and sotagliflozin, act as the antagonists of SGLT2, leading to reduced glucose reabsorption and glycosuria, and have been shown to benefit metabolism and cardiorenal outcomes in patients with or without diabetes [18]. Meta-analyses have reported more compelling results. In 2024, a Bayesian network meta-analysis favored metformin and SGLT2i to reduce the risk of dementia and AD compared to DPP−4i, SUs and TZDs [19]. A network meta-analysis in 2023 concluded the treatment effects of SGLT2i were comparable to GLP-1 Ras and superior to TZDs and DPP-4i for the cognition outcomes related to T2DM, as measured by SUCRA [20]. Antidiabetic agents are increasingly recognized for their potential in treating T2DM-associated CI.
To elucidate the mechanism of SGLT2i in treating CI related to diabetes and T2DM, this review aims to summarize the pathogenesis and treatments and compile experiments and reviews to provide a comprehensive understanding after secondary integration. In addition to AD, VaD is also discussed. Clinical trials and evidence-based meta-analyses are sorted to support and verify the use of SGLT2i in this field. Since evidence on VaD is relatively scarce, the association between SGLT2i and ACVEs, such as stroke, ischemic and hemorrhage cerebral diseases, has been included based on the pathophysiology of the VaD.

2. Potential Pathophysiological Pathways About Diabetic CI

(A) Impaired metabolism through the insulin pathway and others, (B) inflammation and oxidative stress and (C) cerebral vessel pathology in diabetes might all contribute to CI, its subtypes, or mixed types. Many experiments focus on AD, but VaD in diabetes should not be overlooked. Future studies on vascular factors contributing to CI are warranted.

2.1. GSK3β: Impaired Insulin Signal Pathways for Insulin Resistance

Intracellularly, insulin binds to IR and regulates glycogen synthesis via the IRS/PI3K/AKT/GSK3β pathway, where GSK3β negatively affects glucose tolerance and homeostasis is inhibited by AKT in this signaling pathway [21,22]. Similarly, insulin signaling in the brain consists of IR/IRS/PI3K/AKT regulating food intake and endogenous glucose production, etc., based on animal studies and intranasal insulin studies [23]. When insulin resistance occurs in the brain, a reduced amount of insulin transported by the BBB causes a relative insulin deficiency and the inability of the insulin signaling pathway, which ultimately impairs central and peripheral metabolic function, possibly affecting mood and cognition [23]. The leptin receptor in the CNS regulates appetite, body weight, neuroendocrine functions and glycemia, initiating the cascade of PI3K/AKT [24]. T2DM patients with obesity commonly experience leptin resistance, and there is evidence of the therapeutic potential of leptin due to its glucoregulatory and antidiabetic actions based on clinical trial results [24]. When the Wnt/β–catenin pathway is disheveled, leptin is activated by the Wnt-coreceptor low-density lipoprotein receptor-related protein complex and also acts as an inhibitor of GSK3β [25]. Experiments have shown evidence that attributes tau hyperphosphorylation, an AD biomarker, to PI3K/AKT/GSK3β signaling pathway damage due to diabetes mellitus. As a kinase, the derepressed GSK3β increases phosphorylation of the tau protein in the damaged signaling pathway, ultimately contributing to CI in AD or poststroke dementia, etc. [26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48]. Leptin produced by high-intensity interval training ameliorates tau phosphorylation via PI3K/AKT/GSK3β downstream to bypass leptin signaling suffering caused by leptin receptor insufficiency in the hippocampus due to T2DM [49]. Canonical Wnt/β-catenin repairs the insulin/PI3K/AKT/GSK3β function, showing the neuroprotective potential of manually amplifying the Wnt/β-catenin/Ins2 pathway [50,51,52,53,54]. During T2DM, elevated glucocorticoids and hyperglycemia might directly affect GSK3β, accelerating the progression of tau pathology or AD [55,56]. All experiments have demonstrated that GSK3β and relevant pathways might be therapeutic targets in the future.

2.2. IDE: Insulin Resistance and Aβ Protein

In addition to the tau protein, another biomarker of AD is the Aβ protein, whose deposition is involved in core pathophysiological mechanisms [9,57]. The generation of APP requires cleavage by the β-site Aβ precursor protein-cleaving enzyme 1 [58]. When experiencing impaired insulin-related signaling pathways, insulin resistance and hyperinsulinemia, IDE, a degrading enzyme of insulin and Aβ, is considered as a bridge linking diabetes and CI and AD via the potential signaling pathways of PI3K/AKT/GSK3β, PPARγ and NFκB [59]. With diverse cellular localization, the IDE becomes disabled due to the aberrant S-nitrosylation and competitive blocking by insulin at the Aβ binding site, which might provide a rough explanation of how the IDE connects AD with T2DM with a background of hyperglycemia and hyperinsulinemia caused by insulin resistance [60,61,62,63,64,65]. Experiments have demonstrated that the expression, transcription, translation and activity of the cerebral IDE affected by age, insulin or glycemia participate in the onset and progression of CI and AD [66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86]. As mentioned above, CAA is one of the key vascular wall modifications contributing to VaD [11]. It should be recognized that not only does the IDE promote the progress of the AD stage but also VaD in T2DM, meaning that the IDE might contribute to VaD or mixed dementia [87,88,89]. On the contrary, the role of insulin is disputed, with some studies concluding a protective role, whereas cerebral hyperinsulinemia is considered a risk factor for CI due to T2DM, which probably contributes to the diabetic model or various other standpoints [90,91,92,93]. In another trial, the hypothesis that hyperinsulinemia would significantly elevate the Aβ load and thus exacerbate the extent of AD is challenged by the results showing no influence of the Aβ load on the brains of 134 diabetic and 567 non-diabetic subjects [94]. There are also studies reporting Aβ deposition where the IDE is not affected [90,95].
IDEs inactivated by an oxidant might be the dominant reason for the massive Aβ accumulation. Another explanation is that the state called hyperinsulinemia in the brain is relative and regional due to insulin resistance, where the insulin receptor becomes insensitive. Not only relative hyperinsulinemia but also oxidants, such as NO, attack the IDE, which ultimately loses its proteolysis function towards insulin and Aβ. With insulin accumulating regionally, this positive feedback deteriorates cerebral Aβ homeostasis, and then CI is classified as a branch of the pathophysiological mechanism of T2DM.

2.3. Neuroinflammation and Oxidative Stress

Microglial, the main defenders in the brain, can be activated by a range of insults, including hyperglycemia, metabolic stress, ischemia and hypoxia in diabetes, as well as Aβ proteins. It is then polarized into the proinflammatory M1 classification with upregulated inflammatory mediators such as IL-1β, IL-, and TNF-α [96]. In addition to inactivating the IDE, as mentioned above, oxidants might have a direct effect on the CNS in diabetes. Coupled with an inflammatory reaction, cerebral glycemia overload damages the balance between glycogenesis and glycolysis and mitochondria function, therefore producing excessive ROS, such as superoxide (O2), hydroxyl (HO) and hydrogen peroxide (H2O2), via NFκB and NOX2, all of which ultimately results in DNA oxidation, lipid peroxidation, advanced oxidation of proteins, and the formation of AGEs [96]. In a combined analysis of the brain transcriptome from db/db mice with CI, 222 upregulated gene sets and 85 downregulated gene sets have been defined, mainly related to mitochondrial respiratory and oxidative stress, and glycolytic and inflammation [97]. Recently, basic studies involving complex pathway crosstalk have deeply explored the role of M1 microglial polarization, inflammatory signaling pathways and ROS, which injure neurons and cause apoptosis or necrosis, in the decline in cognition function related to diabetes [98,99,100,101,102,103,104,105,106]. In a 12-year followed-up cohort of older adults with healthy cognition function (n = 1840) or MCI (n = 682), diabetes patients (HbA1c ≥ 7.5%) had a significantly higher (triple) risk of the progression of MCI to dementia (HR 3.30, 95%CI 1.02–9.93) accompanied by elevated CRP compared to normoglycemia, while those with diabetes with normal CRP showed an insignificant risk (HR 0.9, 0.48–1.67) [107].

2.4. Cerebral Vascular Modification

In 2024, a systematic review and meta-analysis focusing on stroke sorted the evidence on risk factors for poststroke CI and dementia, reporting that diabetes is a significant treatable risk factor for VaD, independent of age and stroke severity [108]. According to the vascular impairment of cognition classification consensus study, the subtypes of VaD include poststroke dementia and mixed dementias, such as subcortical ischemic VaD and multiple-infarct dementia [109]. Another classification defined subtypes of VaD from another perspective: multi-infract (cortical VaD), small vessel (subcortical VaD), strategic infarct, hypoperfusion and hemorrhagic dementias; hereditary VaD and AD with cardiovascular disease [10]. Obviously, VaD is a potential secondary outcome of cerebrovascular disease.
When vascular risk factors strike cerebral vessels, vascular wall modification occurs, including atherosclerosis, arteriolosclerosis and CAA, followed by tissue injuries such as macroscopic infarcts, microinfarcts, hemorrhages, microbleeds and white matter hyperintensities, along with neurodegeneration, including neuronal and/or volume loss, oligodendrocyte loss, astrocytosis and microgliosis, elucidating the mechanism of VaD [11]. Diabetes itself confers an independent risk of atherosclerotic cardiovascular disease, such as coronary atherosclerotic heart disease and cerebral atherosclerosis [110]. The main risk factors associated with arteriolosclerosis are hypertension and diabetes, and T2DM is associated with an increased risk of brain arteriolosclerosis, small and large brain infarcts and white matter hyperintensity [111].
Therefore, when referring to the preventive and treatment effects of SGLT2i, the vascular effects have been summarized in response to the rare outcomes of VaD reported in clinical trials.

3. SGLT2i Improving CI Among Patients with T2DM

SGLT2, encoded by the SLC5A2 gene, is responsible for glucose reabsorption primarily in the kidney. It is a new target for treating diabetes as it actively transports Na+ and glucose with a ratio of 1:1 across cell membranes as they are energized by the sodium gradient [112]. Almost all SGLT2 is located in the renal proximal tubule, performing 90% of glucose reabsorption, with the remaining part is performed by SGLT1 [113]. When it binds to an inhibitor, the activity of SGLT2 effectively decreases, resulting in reducing glucose reabsorption, promoting urinary glucose excretion, and exerting glucose-lowering effects, all of which ultimately relieve the sodium and glucose toxicity in the proximal tubule [113].
SGLT2i, including canagliflozin, dapagliflozin, empagliflozin, ertugliflozin, approved by the European Medicines Agency and the Federal Drug Administration, and sotagliflozin approved only by the European Medicines Agency for use in type 1 diabetes, as well as others still in trials, have been clinically observed to provide T2DM patients with extra cardiorenal benefits in the context of heart failure or CKD [18]. In summary, mechanisms underlying the cardiovascular and renal protective effects of SGLT2i include inhibiting myocardial Na+/H+ exchange, reducing cardiomyocyte apoptosis and improving myocardial fibrosis, reducing the synthesis of adipokines, cytokines and epicardial adipose tissue, improving glomerular hyperfiltration, reducing renal oxygen consumption and inflammatory reactions, etc. [113]. In addition to cardiorenal outcomes, it has been summarized recently that SGLT2i could improve HRQoL by decreasing the multimorbidity risk, lowering body weight, preventing depression incidence and improving physical function [114]. In the standards of diabetes care-2024 by the ADA, it is recommended that the treatment of adult T2DM patients with cardiovascular or/and renal risk should include SGLT2i and/or GLP-1 Ras [115]. However, even with such extra benefits when treating T2DM, there are possible adverse effects, such as polyuria and urinary tract infections, which might lead to treatment discontinuation [18].
Clinical trials on SGLT2i as preventive and treating agents have been collected to fully understand their impact. Meta-analyses synthesizing the results are also in this article. Here, an attempt is made to provide a comprehensive conclusion, with experiments and reviews summarized.

3.1. Evidence from Primary and Secondary Analysis of Clinical Trials

3.1.1. Observational Clinical Trials and RCTs

Table 1 collects the clinical evidence of SGLT2i in treating CI in T2DM, while Figure 1, Figure 2, Figure 3 and Figure 4 (by R.4.4.1 of free, same for the Figure 5, Figure 6 and Figure 7) illustrate the results. In a 5-year cohort study of 106,903 individuals aged 66 years or older, SGLT2i was associated with a significantly lower risk of dementia (adjusted HR 0.80, 95%CI 0.71–0.89) compared to DPP-4i, with dapagliflozin showing the lowest risk (adjusted HR 0.67, 95%CI 0.53–0.0.84), followed by empagliflozin (adjusted HR 0.78, 95%CI 0.69–0.89) and canagliflozin with an insignificant risk (adjusted HR 0.96 95%CI 0.80–1.16) [116]. Another prospective cohort among frail elders over 65 years with T2DM and HFpEF found empagliflozin significantly associated with improved MoCA scores (OR 3.609, 95%CI 1.566–8.321, p = 0.003) [117]. In a Swedish longitudinal prospective cohort of 132,402 subjects, SGLT2i users who showed dementia at baseline had a significantly lower dementia mortality rate (adjusted HR 0.43, 95%CI 0.23–0.80) compared to non-users, while insulin (adjusted HR 1.34, 95%CI 1.23–1.45, p < 0.001) or SUs (adjusted HR 1.19, 95%CI 1.01–1.42, p < 0.05) increased the mortality risk [118]. A cross-sectional study in 2023 observed that SGLT2i had a protective effect on patients at risk of MCI (OR 0.337, 95%CI 0.135–0.843, p = 0.020) under multi-factor analysis by logistic regression [119]. Using multivariable regression analysis where the improvement in MoCA scores was the dependent variable, empagliflozin treatment was calculated to have a coefficient of 0.794 (p = 0.031) [120]. A cohort in Hong Kong, China, associated SGLT2i with a lower risk of PD in addition to dementia and cerebrovascular onset, compared to DPP-4i as shown by Cox regression [121]. A nested case-control in Denmark reported a significantly lower risk of dementia (multi-adjusted OR 0.58, 95%CI 0.42–0.81) with SGLT2i use [122]. Measuring cognition function by the RBANS, including immediate memory, delayed memory, viso-spatial/construction, language and attention, a secondary analysis of the SMART2D cohort reported that SGLT2i was associated with significantly increased language scores (adjusted coefficient 0.74, 95%CI 0.12–1.36, p = 0.019) among 138 patients with a mean age of 60.6 years in Singapore [123]. A later observational trial studying a new SGLT2i reported that henagliflozin might significantly improve the MoCA scores and plasma phosphorylated Tau protein among T2DM patients with MoCA lower than 26 [124].
A cohort study investigating cerebrovascular and cognitive benefits of SGLT2i as a treatment among patients with atrial fibrillation and T2DM reported that non-SGLT2i users had a significantly increased risk of ischemic stroke/TIA, ICH and incident dementia compared to SGLT2i treatment after 3-years of follow-up [125]. When reporting MASE as an outcome including stroke, myocardial infarction and all-cause death, a 1.1-year followed-up cohort showed a decreased risk (HR 0.81, 95%CI 0.70–0.94) when comparing empagliflozin initiation to DPP-4i [128]. In 2023, a population-based cohort study taking gender into account showed that men might obtain more benefits than women when using SGLT2i rather than GLP-1 RAs for the outcomes of MASEs (men/women, sub-distribution HR 0.78/0.99, 95%CI 0.66–0.93/0.77–1.28) and stroke (0.70/1.23, 0.42–1.16/0.59–2.56), which remains to be fully studied in the future [129]. Concentrating only on cardiovascular outcomes, adverse cardiovascular events, including fatal or nonfatal stroke and TIA in EMPA-REG OUTCOME, were determined to have insignificant risks: 1.18 and 0.85, respectively, when using empagliflozin compared to a placebo [130]. Study DECLARE TIMI 58 reported that dapagliflozin was insignificantly associated with ischemic stroke compared to the placebo, with a result of HR = 1.01 [131]. Also, studying the association between SGLT2i and cardiovascular outcomes, VERTIS revealed an insignificant link between ertugliflozin and fatal or nonfatal stroke [132].
Reporting both incident dementia and other subtypes as outcomes, a cohort concluded SGLT2i had a protective effect in reducing risk incidence for dementia (HR 0.705, 95%CI 0.506–0.984), AD (0.939, 0.721–1.222), VaD (0.44, 0.235–0.824) and other/mixed dementia (0.601, 0.296–1.220) [126]. Compared to sulfonylureas, SGLT2i, when newly used, were associated with a significantly lower risk of all-cause dementia, AD, VaD, expressed by a minus risk difference of −2.5% (95%CI −3.0% to −2.1%), −0.47% (−0.61% to −0.33%), −0.39% (−0.55% to −0.23%), respectively, with or without CKD and combined with other glucose-lowering drugs or not [141].
As for RCTs, one study including 55 patients in Nanjing, China, favored liraglutide over dapagliflozin for the enhancement of impaired olfactory neural activation and cognition capacity in T2DM [142]. Another RCT conducted in Italy reported that SGLT2i (canagliflozin, empagliflozin, dapagliflozin) did not improve the cognitive status significantly as tested by verbal fluency, the Babcock Story Recall Test and attentive matrices test, compared to either baseline or incretins [134]. In China, in 2022, one RCT, including 96 patients, reported dapagliflozin combined with CBT could significantly increase cognition function scores (all p < 0.001) of ADL (MD ± SD 6.18 ± 1.96), MMSE (2.59 ± 0.72), QOL-AD (10.69 ± 1.45) [135].
Here, six RCTs are summarized (one is not included in Table 1), comprising 14 cohorts, one cross-sectional and one case-control study. As VaD is rarely reported, the cerebral vascular outcomes in EMPA REG OUTCOME, DECLARE TIMI 58, VERTIS and MACE have been summarized as risk factors or accelerators of VaD to understand the effect of SGLT2i from another perspective. They all reported insignificant results.
RCTs of high quality exploring the association between SGLT2i and CI among T2DM patients are relatively rare, so the evidence is not firmly convincing. Given both the significant and insignificant results, some confounding factors should be taken into consideration. (1) First, the population recruited and region of conduct. The basic incidence of CI varies from ethnicity to ethnicity. Thus, the impact of whether one has been prescribed SGLT2i against T2DM progression or not would be different in different regions as a result of genes, environment, lifestyles or health service, etc. It is similar when it comes to gender, age and other personal characteristics for the same ethnicity. (2) Second, the number of participants in certain clinical trials. It is confusing as to whether significant or insignificant results from a trial with a limited number of subjects are representative or just random errors. The RCT with significant results by Ying Zhao et al. only recruited 96 actual subjects (Table 1) [135]. (3) Third, the intervention (exposure) and control (non-exposure). The specific SGLT2i used were different, and the control was either another antidiabetic agent or a placebo. Given the possible toxic effects of some antidiabetics, which can accelerate the onset of CI, the protective role of SGLT2i becomes doubtful when compared to a placebo or no drug use. The insignificant results from RCTs using a placebo as control here verify this concern. (4) Last but not least, the outcomes of interest. When using the incidence of CI related to T2DM as the outcome, the years of follow-up should be sufficiently long, or there might be bias. When using the cognition function scores to quantify the deterioration by T2DM, not only the amount of change but rate should be taken into consideration in the calculation for comprehensive exploration.
Table 1. Clinical trials studying the association between SGLT2i and CI and cerebrovascular risks.
Table 1. Clinical trials studying the association between SGLT2i and CI and cerebrovascular risks.
AuthorDesignPopulation (Region)InterventionVersusOutcomeEffect SizeResults (95%CI)p for Results
Ding et al. [119].CS222 middle-aged and elderly patients with T2DM (China)SGLT2iNon-useMCIOR0.337 (0.135–0.843)0.020
Wium-Andersen et al. [122].N-CC11,619 dementia cases and 46,476 control all with T2DM (Denmark)SGLT2iNon-useDementiaOR0.58 (0.42–0.81)
S. Low et al. [123].C138 patients with T2DM (Singapore)SGLT2iNon-useRBANSβ0.20 (−0.03–0.44)0.091
IM0.38 (−0.04–0.79)0.074
DM0.09 (−0.34–0.52)0.677
VS−0.11 (−0.67–0.46)0.703
Language0.60 (0.10–1.11)0.019
Attention0.05 (−0.35–0.44)0.806
Mone et al. [117].C162 Frail older adults > 65 years with T2DM and HFpEF (Italy)EmpagliflozinNon-useMoCA scores improvementOR3.609 (1.566–8.321)0.003
P. Mone et al. [120].C166 frail elders with diabetes and CKD (USA)EmpagliflozinNon-useMoCA scoresMD1.9 (1.04) †
β0.794 (0.073–1.516)0.031
Zhang et al. [124].C290 T2DM patients ≥ 45 years with MoCA ≤ 26 (henagliflozin, n = 135; non-use, n = 155) (China)HenagliflozinNon-useMoCA scores improvementOR3.670 (2.224–6.056)<0.0001
Plasma p-Tau level improvement2.595 (1.598–4.213)<0.0001
Chen et al. [126].C2430 patients (age: 55–85 years) with AF and diabetes (Taiwan, China)SGLT2iNon-useDementiaHR0.705 (0.506–0.984)
AD0.939 (0.721–1.222)
VaD0.440 (0.235–0.824)
Other/mixed0.601 (0.296–1.220)
Stroke0.750 (0.601–0.937)
Riester et al. [133].C7710 elder aged ≥ 66 years with T2DM in nursing home (31.08% SGLT2i) (USA)SGLT2iGLP-1 RAsMACEHR1.03 (0.74–1.44)
A. Sharma et al. [129].C8026 patients (55.7% male) with T2DM at baseline (4231 SGLT2i users) (Australia)SGLT2iGLP-1 RAsMACEMHR0.78 (0.66–0.93)
F0.99 (0.77–1.28)
StrokeM0.70 (0.42–1.16)
F1.23 (0.59–2.56)
Tang et al. [141].C35,458 new users aged ≥ 50 years of SGLT2i or SU with T2DM (USA)SGLT2iSUDementiaRD−2.5% (−3.0%, −2.1%)
AD−0.47% (−0.61%, −0.33%)
VaD−0.39% (−0.55%, −0.23%)
Mui et al. [121].C51,460 T2DM users with a median age of 66.3 years (Hong Kong, China)SGLT2iDPP-4iDementiaHR0.41 (0.27–0.61)<0.0001
AD0.25 (0.06–1.04)0.0546
PD0.28 (0.09–0.91)0.0349
CVM0.36 (0.30–0.43)<0.0001
D. Edmonston et al. [128].C62,197 adult patients with T2DM (USA)EmpagliflozinDPP-4iMACEHR0.81 (0.70–0.94)0.007
Riccardo Proietti et al. [125].C89,356 patients aged ≥ 18 years with AF and T2DM (global)Non-useSGLT2iIS or TIAHR1.12 (1.01–1.24)0.029
ICH1.57 (1.25–1.99)0.001
Dementia1.66 (1.30–2.12)0.001
Wu et al. [116].C106,903 Residents of Ontario, Canada ≥ 66 years old free of dementia (Canada)SGLT2iDPP-4iDementiaHR0.80 (0.71–0.89)
Dapagliflozin0.67 (0.53–0.84)
Empagliflozin0.78 (0.69–0.89)
Canagliflozin0.96 (0.80–1.16)
J.Secnik et al. [118].C132,402 subjects with diabetes at baseline (Sweden)SGLT2iNon-useDementia deathHR0.43 (0.23–0.80)< 0.05
0.52 (0.22–1.23) *
Siao et al. [127].C976,972 patients with newly diagnosed T2DM (Taiwan, China)SGLT2iNon-useDementiaOR0.89 (0.82–0.96)0.0021
Perna et al. [134].RCT39 elder aged > 65 years subjects (women:16) with T2DM (Italy)SGLT2iBaselineVFTMC0 (−4.25–4.25)1
BSRT−1.32 (−3.36–0.71)0.178
AMT−0.09 (−0.56–0.38)0.676
Ying Zhao et al. [135].RCT96 patients with T2DM and MCI (China)Dapagliflozin with CBTConvention interventionADLMD6.18 (1.96) †<0.001
MMSE2.59 (0.72) †<0.001
QoL-AD10.69 (1.45) †<0.001
EMPA REG OUTCOMERCT7028 adult patients with T2DMEmpagliflozinPlaceboStrokeHR1.18 (0.89–1.56)0.26
TIA0.85 (0.51–1.42)0.54
VERTISRCT8246 patients aged ≥ 40 years with T2DMErtugliflozinPlaceboStrokeHR1.06 (0.82–1.37)
DECLARE TIMI 58RCT17,160 patients aged ≥ 40 years with T2DMDapagliflozinPlaceboISHR1.01 (0.84–1.21)
Non-use participants who did not use SGLT2i as a treatment. M, male; F, female. β, coefficient in multivariable regression analysis. Blank means not reported. Results in boldface mean significant effect size whose p-value < 0.05. * where the population were free of dementia at baseline and the OR stands for the mortality risk of dementia death. † the data here are mean and standard deviation: M(SD).
In the future, the effects of different SGLT2i choice and dosage could be tested, and the outcomes of interest in trials could include incidence of CI (MCI, dementia, AD, VaD), difference in cognition function scores (MMSE, MoCA, etc.) and deterioration rate of cognition among T2DM patients. More trials should concentrate on VaD in T2DM.

3.1.2. Evidence-Based Meta-Analyses

Meta-analysis is considered as evidence of high quality. Therefore, meta-analyses regarding the use of SGLT2i to treat CI in T2DM were collected and are shown in Table 2. The forest plot (Figure 5) shows the results of different outcomes. Recently, in 2024, an umbrella systemic review and meta-analysis concluded that SGLT2i are associated with a reduced risk of dementia [15]. Compared with other antidiabetics, SGLT2i in a Bayesian network meta-analysis demonstrated a lower risk of dementia (vs. α-glucosidase inhibitors, RR 0.169, 95%CI 0.071–0.384; vs. DPP-4i, 0.455, 0.294–0.667; vs. SUs, 0.417, 0.244–0.667; vs. TZDs, 0.370, 0.169–0.769) and AD (vs. DPP-4i, 0.130, 0.014–0.625; vs. SUs, 0.083, 0.008–0.556; vs. TZDs, 0.067, 0.005–0.556) [19]. Meanwhile, another meta-analysis and network meta-analysis reported that SGLT2i might have the best potency for mitigating CI in T2DM. The meta-analysis used the methods of SUCRA (94%) [20]. Figure 6 and Figure 7 illustrate the risk-lowering effect of SGLT2i compared to control or other interventions. When cognition function was measured using the MMSE or MoCA, a meta-analysis that included one RCT and two cohorts showed that SGLT2i might contribute to a significant elevation (SMD 0.88, 95%CI 0.32–1.44) in cognition function scores [136]. In a meta-analysis in 2020 which included two case-control studies, SGLT2i showed an insignificantly decreased risk of dementia (OR 0.74, 95%CI 0.47–1.15) [137]. On the contrary, a new meta-analysis, including 12 RCTs, summarized that there is no significant effect on either diabetic dementia (OR 1.37, 95%CI 0.70–2.69) or subtypes of dementia (AD: 1.99, 0.59–6.71; PD: 0.63, 0.25–1.61; VaD: 0.40, 0.09–1.85) [138].
The results of Jaiswal et al. were calculated by a fourfold table, but others used logistic regression. That may be the reason why they raise a contrary conclusion. The reported heterogeneity was relatively large (e.g., 96.1%, 87%, 79%, 82.5%, 78.9%). According to funnel plots, the publication bias is acceptable.
Existing analyses have concluded that SGLT2i might be a promising choice in this situation. However, since there are fewer RCTs on this topic, the quality of meta-analysis remains low. An umbrella systemic review of high quality is needed in the future. Furthermore, more meta-analyses might provide evidence of optimal SGLT2i choice and dosage based on more clinical trials conducted in the future, especially different phases of RCTs.
Table 2. Results of meta-analysis reporting an association between SGLT2i treatment and CI in T2DM compared to non-SGLT2i.
Table 2. Results of meta-analysis reporting an association between SGLT2i treatment and CI in T2DM compared to non-SGLT2i.
AuthorsOutcomeEffective SizeResults (95%CI)I2 (%)p for HeterogeneityStudies Included
(RCT/C/CS/CC)
Kuate Defo et al. [15].DementiaRR0.39 (0.200.76)96.10.0005 (0/2/1/2)
Tian et al. [20].CIOR0.41 (0.220.76)
Dementia0.41 (0.220.76)
AD0.16 (0.070.39)
CISUCRA94.0% 12 (3RCTs/9OTs) *
M.Banerjee et al. [139].Non-cardiovascular mortalityRR0.90 (0.820.99)00.758 (8/0/0/0)
Y.J. Youn et al. [136].DementiaHR0.68 (0.500.92)87<0.0014 (0/4/0/0)
OR0.74 (0.47–1.15)790.032 (0/0/0/2)
Dementia ≥ 60 yearsHR0.84 (0.750.95) 2 (0/2/0/0)
Cognitive function scoresSMD0.88 (0.32–1.44) 3 (1/2/0/0)
Tang et al. [140].DementiaRR0.62 (0.390.97)82.50.033 (0/1/0/2)
J.B. Zhou et al. [137].DementiaOR0.74 (0.47–1.15)78.90.032 (0/0/0/2)
Jaiswal et al. [138].DementiaOR1.37 (0.70–2.69)0 6 (6/0/0/0)
AD1.99 (0.59–6.71)0 6 (6/0/0/0)
PD0.63 (0.25–1.61)0 9 (9/0/0/0)
VaD0.40 (0.09–1.85)0 4 (4/0/0/0)
Non-cardiovascular mortality includes infectious diseases, cancers, lung diseases, dementia, renal diseases and other external causes. Blank means not reported. Results in boldface mean significant effect size whose p-value < 0.05. * Three RCTs and nine observational trials were included.

3.2. Potential Mechanism SGLT2i as a Treatment

After reviewing the experiments and reviews, the neuroprotective effects against CI in T2DM could be simply divided into (1) a metabolism effect, (2) anti-inflammation and oxidative stress effects,(3) effects with neurotrophic factors, (4) an AChE inhibition effect. The metabolism effect is driven by the original function of anti-reabsorption to lower glycemia by inhibiting SGLT2 located in the kidney.
As a metabolic result, the body improves its metabolism homeostasis by modulating insulin and AMPK and mTOR signaling pathways, with blood pressure and body weight lowering, which have cardiovascular benefits. Not only does SGLT2i show anti-inflammatory and oxidative stress effects on nervous tissue but also on the vascular wall in the brain via regulating microglial polarization, cytokines, ROS and relative enzyme levels involved in NFκB, NLRP3 Inflammasome, HIF-1α and NADPH oxidase pathways. SGLT2i also crosstalks with neurotrophic factors, including BDNF, NT4 and GDNF, which have neuroprotective effects on neurons and neuroglia cells. Promisingly, experiments have shown that SGLT2i could act as dual inhibitors of SGLT2 and AChE, amplifying its treating potential in CI. Figure 8 and Figure 9, created by PowerPoint (office 2019), illustrate the hypothetical mechanism of the preventative and treating role of SGLT2i.

3.2.1. Studied Mechanism in Animal Experiments

In an SGLT2 spontaneous mutant mouse line (SAMP10-∆Sglt2), SGLT2 mRNA expression reduced to 34% compared to the SAMR (the senescence-resistant mouse model) 1 mice, and APP expression in the hippocampus showed significantly increased expression, while amyloid precursor-like protein (Aplp) was markedly downregulated, where the age-related decline in cognition was detected via membrane translocation of GLUT1 in the brain interfered with by APP and an unclear mechanism that increased APP expression associated with SGLT2 mutations [143]. In an experiment using a mice model of T2D-AD conducted in 2023, empagliflozin was reported to reduce p-Tau levels by activating ACE2/MasR that might crosstalk with IRS1/AKT/GSK3β pathways [86]. Table 3 summarizes some experiments investigating the potential mechanism using SGLT2i as a treatment of CI in T2DM. At the same time, another experiment reported empagliflozin and dapagliflozin as showing protective effects on diabetes-induced CI of diabetic mice, through modulating the expression of cytokines and proteins, including neurotrophic factors (BDNF, NT4), HIF-1α, APP and α-synuclein [144]. In 2021, empagliflozin was proven to attenuate CI in T2DM via suppressing inflammatory mediators, including IL-1β, IL-6 and TNF-α and oxidase stress in T2DM mice caused by a high-fructose diet [145]. In addition, empagliflozin also had a protective effect on NVU via ultrastructural remodeling discovered in type 2 diabetic db/db female mice by transmission electron microscopy [146]. Canagliflozin studied in male and female mice models also reported a neuroprotective effect against CI in aging by the reduction in the inflammatory response and mTOR signaling pathway in a sex-specific manner [147]. In addition to the mechanism mentioned above, a molecular structure experiment concluded that dapagliflozin had an inhibiting effect on AChE, which is the primary target of AD therapy [148]. Another experiment with a similar purpose reported that sotagliflozin and ertugliflozin might be dual inhibitors of SGLT2 and AChE [149].
Table 3. Animal experiments about SGLT2i in treating CI in T2DM. HFD, high-fat diet; STZ, streptozocin; HFruD, high-fructose diet; RVS, rivastigmine; US, ultrastructure; MasR, mitochondrial assembly receptor; NN, nitrite–nitrate; TBARS, thiobarbituric acid reactive substance; SOD, Superoxide dismutase; BM, base membrane.
Table 3. Animal experiments about SGLT2i in treating CI in T2DM. HFD, high-fat diet; STZ, streptozocin; HFruD, high-fructose diet; RVS, rivastigmine; US, ultrastructure; MasR, mitochondrial assembly receptor; NN, nitrite–nitrate; TBARS, thiobarbituric acid reactive substance; SOD, Superoxide dismutase; BM, base membrane.
AuthorModelDrugsControlResultsMechanism
A.Y. Sim et al. [86].Male C57BL/6 mice fed with HFD and injected STZEmpagliflozin, 25 mg/kg/day, orallyVehicle or DPP-4iHippocampal-dependent cognitive functions↑;
Hepatic lipid accumulation↓ and inflammation↓;
Promoting glucose homeostasis
Restore brain insulin signaling; pTau and Aβ↓: IRS1/AKT/GSK3β pathways
pTau↓: ACE2/MasR↑
Iwona Piątkowska-Chmiel et al. [144].CD-1 male mice with administration of 20% aqueous fructose solution and injection of freshly prepared STZ solutionEmpagliflozin, 10 mg/kg/dayVehicleProtective effects of cognitionInflammatory cytokines↓
BDNF↑
NT4↑
α-synuclein in prefrontal cortex↑
Dapagliflozin, 10 mg/kg/dayVehicleProtective effects of cognitionInflammatory cytokines↓
BDNF↑
HIF-1α↑
APP in hippocampus↑
T.Khan et al. [145].CLB57/6 mice fed with HFruDEmpagliflozin, 4.4 mg/kg/dayNormal saline (10 mL/kg) or RVS (1.5 mg/kg)Attenuating memory deficitNN↓, TBARS↓, SOD↑, catalase↑
IL-1β, IL-6, TNF-α↓
TLR4-NFκB pathway↓
GSK3β/pTau↓, Aβ (1–40)↓, Aβ (1–42)↓
M.R Hayden et al. [146].Female db/db mice and wild controlEmpagliflozin, 10 mg/kg/day Ameliorating US remodeling of NVUBM thickening↓, damage on BBB↓
Activated microglia invasion↓
Aberrant mitochondria remodeling↓
myelin remodeling↓
Axonal collapse↓
Myelin disarray↓
Jayarathne et al. [147].UM-HET3 miceCanagliflozin, 14.4 mg/kgPurina 5LG6Insulin sensitivity↑; locomotor activity↑; overall behavior function↑mTOR signaling↓
Inflammatory response↓
microgliosis↓, astrogliosis↓
Hierro-Bujalance et al. [150].APP/PS1, db/db, APP/PS1 & db/db miceEmpagliflozin, 10 mg/kg Learning and memory in AD, T2D and AD-T2D mice↑; Brain atrophy↓; Spontaneous bleeding↓amyloid pathology↓
Iba1+ burden↓
tau phosphorylation↓

3.2.2. Reviewed Mechanism

A review in 2023 summarized that SGLT2i might ameliorate CI via decreasing the leakage of the blood–brain barrier, reducing reactive oxygen species, Aβ levels and TNF-⍺, improving endothelial and microglia functions against neurodegenerative diseases such as AD and PD, although it was given limited space among other outcomes of interest [151]. Concentrating on LOAD, SGLT2i were concluded to be a novel pleiotropic antidiabetic drug class for LOAD and vascular brain damage for both direct (oxidative stress modulation; increase in brain-derived neurotrophic factor, BDNF; protective effect on neuronal loss and reduction in Aβ plaques; acetylcholinesterase, AChE, inhibition) and indirect (reducing blood pressure; improving cardiac function; modulating inflammation; promoting weight loss; reducing atherosclerotic plaque progression) neuroprotective effects compared to hypoglycemic agents and other anti-hyperglycemic agents [152]. Table 4 summarizes the reviewed mechanism of SGLT2i used to treat CI related to T2DM or diabetes. Serving as glucose sensors in response to extracellular glucose changes, SGLT2i showed their neuroprotective and neuro-cognitive effect in T2DM. However, the particular distribution and co-presence of SGLT1/SGLT2 in the CNS needs to be deeply studied as it might play a possible role in brain and cognitive activity [153]. The neuroprotective effects of SGLT2i have been reported to be related to atherosclerosis, inflammation, oxidative stress and mitochondrial dysfunction, mTOR signaling and cerebrovascular dysfunction, and differ in each classification of drug [154]. In a review mainly discussing the mTOR and AMPK/ULK1 pathways and metabolism homeostasis, it was reported that SGLT2i act as a metabolism regulator of glucose and amino acids, the sequence of which activated mTOR signaling calming and AMPK/ULK1 pathway acceleration to promote autophagy and mitophagy, which might be a beneficial target of AD treatment [155].
An article discussing the potential role of SGLT2i in treating cerebrovascular complications or accidents of diabetes suggested they could lower the risk of cerebrovascular dysfunction, including endothelial dysfunction, atherosclerosis, hypercoagulability, oxidative stress, renal reperfusion injury, which probably contribute to VaD [156]. When it comes to ischemia-related cerebral damage, which might contribute to VaD, empagliflozin, one of the SGLT2i, showed a neuroprotective effect at another stage against CI, related vascular disease in T2DM via increasing the expression of HIF-1α and VEGF-A, decreasing the expression of caspase-3, thus limiting the infarct volume [157].
A study to repurpose empagliflozin as a treatment for CI conducted among 21 participants aged ≥ 55 years without diabetes detected that the levels of phosphorylated IGF-1 receptor, phosphorylated insulin receptor, phosphorylated-in-tyrosine insulin receptor substrate-1 and phosphorylated AKT increased in neuronal derived extracellular vesicles after the administration of empagliflozin, suggesting that SGLT2i might be put into use in related disease via activating the IGF-1 and insulin/IR/IRS-1/PI3K/AKT pathways [158].
Table 4. Mechanism of SGLT2i treating CI in T2DM. Blank means not reported.
Table 4. Mechanism of SGLT2i treating CI in T2DM. Blank means not reported.
AuthorsOutcomesPotential Mechanism
F. Mancinetti et al. [152].LOAD and dementiaDirect effectsBP↓
Cardiac function↑
Weight↓
Atherosclerotic plaque progression↓
Indirect effectsOxidative stress↓
BDNF↑
Aβ plaques↓
AChE inhibition↑
M.R. Rizzo et al. [153].Cognitive dysfucntionOxidative stress↓, NADPH oxidase↓
BDNF↑
Insulin sensitivity↑
Mitochondrial brain function↑
Synaptic plasticity↑
DJ-1/Nrf2 pathway
GDNF↑, PI3K/AKT/GSK3β (Ser9) pathway
NF-κB pathway↓, TNF-α levels↓
Brain extra- and intracellular accumulation of Aβ and NFTs↓
mTOR hyperactivation↓
AChE inhibition
A. Pawlos et al. [154].NeuroprotectionCanagliflozinAnti-inflammatory
Promoting M2 Macrophages Polarization
Oxidative Stress↓
mTOR Signaling↓
DapagliflozinAnti-epileptic Potential
CIMT Regression
Anti-inflammatory
NLRP3 Inflammasome↓
Promoting M2 macrophage polarization
Oxidative Stress↓
mTOR Signaling↓
EmpagliflozinBDNF↑
CIMT Regression
Anti-inflammatory
Blood–Brain Barrier protection
NLRP3 Inflammasome↓
Promoting M2 macrophage polarization
Oxidative Stress↓
Neurovascular unit remodeling
Cerebral ischemia/Reperfusion damage↓
mTOR Signaling↓
ErtugliflozinOxidative Stress↓
mTOR Signaling↓
SotagliflozinOxidative Stress↓
S.V. Birajdar et al. [159].NeurodegenerationInsulin sensitivity↑
Oxidative Stress↓
Neuronal damage↓
G. Goodarzi et al. [160].ADEmpagliflozinOxidative Stress↓
BDNF↑
Infarct size
HIF-1α/VEGF↑
CanagliflozinAChE inhibition
DapagliflozinAChE inhibition
Hippocampal synaptic plasticity↑
V. Chavda et al. [156].CVAsEndothelia dysfunction↓
glycemia↓
AGE superoxide↓
C-peptide and NFκB↓
Inflammation↓
J Ariana Noel et al. [161].CIAChE inhibition
Oxidative Stress↓
Inflammation (NFkB)↓
mTOR pathway↓
Mei et al. [162].CINLRP3 inflammasome↓
ROS dependent neuronal apoptosis
Cerebral Glu metabolism↑
BDNF↑, NGF↑
AMPK pathway↑
CRP↓
M.E Youssef et al. [163].CIDapagliflozinDJ-1/Nrf2 pathway
GDNF↑
PI3K/AKT/GSK3β pathway↑
NFκB pathway↓, TNF↓
AChE inhibition
Empagliflozininflammatory mediators↓
oxidative stress↓
p-tau↓
Aβ↓
Sim et al. [164].ADPeripheral insulin sensitivity↑
Body weight↓
Brain mitochondrial function↑
Insulin signaling↑
Cell death↓
Synaptic plasticity↑
mTOR inhibition
NLRP3 inflammasome-IL-1β modulation
Cerebral oxidative stress↓
Riemma et al. [165].CIM2 microglial polarization↑
JAK2/STAT1 pathway↓
Free radicals↓, antioxidant enzymes↑
ADmTOR activity↓
AChE inhibition, acetylcholine M1 receptor↑
Michał Wiciński et al. [157].Ischemia-related cerebral damageEmpagliflozinHIF-1α↑
VEGF-A↑
caspase-3↓
Limiting infarct volume
Preventing US remodeling of NVU’s cell and myelin
Russell Esterline et al. [155].ADmTOR pathway↓
AMPK/ULK1 pathway↑

4. Conclusions, Discussion and Perspectives

SGLT2i, not only in experiments but also in clinical trials, have shown their potential neuroprotective effects against damage from hyperglycemia and insulin resistance. To some extent, the results of the experiments and clinical trials have shown consistency, so the rationality of causal inference is probably convincing.
The outcomes of interest in clinical trials included the onset of CI and its subtypes and the decline in cognition function domains evaluated by rating scales, where both categorical variables (OR, RR, HR) and continuous variables (mean, MD, SMD) were used for description and meta-analysis. Insight from these clinical trials might be constructive starting points for investigations in the future. SGLT2i might have unique protective effects against VaD for verified cardiovascular benefits compared to other anti-hyperglycemia drugs. However, contrary results have been reported that cannot be overlooked. To conclude the convincing treatment effects of SGLT2i, comprehensive analysis and evaluation of those contradictory results are essential.
After summarizing the experiments and reviews, SGLT2i might prevent or alleviate CI among T2DM patients via ameliorating metabolism homeostasis, suppressing inflammation and oxidative stress, cooperating with neurotrophic factors and inhibiting AChE. Nevertheless, the deeper mechanism of SGLT2i and how they regulate the expression or activity of relevant molecules working in specific pathways could be explored in the future.
The promising role of SGLT2i as a treatment in CI in T2DM still needs to be deeply and fully studied and understood. Existing evidence is not enough to give a recommendation of specific SGLT2i as an optimal treatment. Clinical trials in this field, especially RCTs, should be conducted regarding SGLT2i in treating diabetic CI, to provide more evidence of its value in clinical practice. A meta-analysis and umbrella systemic review should also be used to provide evidence-based, convincing results on this topic in the future.

Author Contributions

Y.Z. drafted the manuscript. J.X., X.L., J.Y., S.L., M.L., X.S., S.Z., C.L., W.X., X.Y. and Y.Y. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work is funded by grants from the National Nature Science Foundation of China (Grant number: 81974114).

Acknowledgments

Special thanks to Xuemin Peng for her suggestions for this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

TermsFull Name
DMdiabetes mellitus
T2DMtype 2 diabetes mellitus
IGTimpaired glucose tolerance
IFGimpaired fasting glucose
CIcognitive impairment
MCImild cognitive impairment
ADAlzheimer’s disease
VaDvascular dementia
amyloid β
NFTsneurofibrillary tangles
CAAcerebral amyloid angiopathy
ORodds ratio
ADAAmerican Diabetes Association
MMSEMini-Mental State Examination
MoCAMini-Cog and Montreal Cognitive Assessment
SGLT2sodium–glucose cotransporter 2
SGLT2isodium–glucose cotransporter 2 inhibitors
DPP-4idipeptidyl peptidase-4 inhibitors
SUssulfonylureas
TZDsthiazolidinediones
GLP-1 RAsglucagon-like peptide 1 receptor agonists
SUCRAsurface under the cumulative ranking curves
ACVEsadverse cerebral vascular events
GSK3βglucogen synthase kinase 3β
IRinsulin receptor
IRSinsulin receptor substrate
PI3Kphosphoinositide 3-kinase
AKTprotein kinase B (PKB)
BBBblood–brain barrier
CNScentral neural system
IDEinsulin-degrading enzyme
APPamyloid-β precursor protein
PPARperoxisome proliferators-activated receptors
NFκBnuclear factor κ B
NOnitric oxide
ILinterleukin
TNFtumor necrosis factor
ROSreactive oxygen species
NADPHnicotinamide adenine dinucleotide phosphate
NOX2NADPH oxidase 2
AGEsadvanced glycation end-products
HRhazard ratio
HbA1cglycosylated hemoglobin A1c
CRPC-reactive protein
CKDchronic kidney disease
HRQoLhealth-related quality of life
RCTsrandomized control trials
HFpEFheart failure with a preserved ejection fraction
95%CI95% confidence interval
RBANSRepeatable Battery for the Assessment of Neuropsychological Status
ISischemic stroke
TIAtransient ischemic attack
ICHintracranial hemorrhage
MACEsmajor adverse cardiovascular events
CBTcognitive behavioral training
ADLactivity of daily living
MDmean difference
SDstandard deviation
QOL-ADthe quality of life AD scores
Ccohort
CScross-sectional
N-CCnested case-control
PDParkinson’s disease
CVMcerebrovascular mortality
VFTverbal fluency test
BRSTBabcock story recall test
AMTattentive matrices test
IMimmediate memory
DMdelayed memory
VSvisuospatial/construction
MCmean changes
RDrisk difference
SMDstandard mean difference
RRrisk ratio
AChEacetylcholinesterase
AMPKAMP-activated protein kinase
mTORmammalian target of rapamycin
HIFhypoxia-inducible factor
NLRP3nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3
ACVEsadverse cerebral vascular events
NVUneurovascular unit
NGFnerve growth factor
BDNFbrain-derived neurotrophic factor
NT-4neurotrophins 4
GDNFglial cell-derived neurotrophic factor
PPREperoxisome proliferators reactive element
Nrf2nuclear factor-erythroid 2-related factor 2
TLRtoll-like receptor
IκBinhibitor of NFκB
JAK-STATJanus Kinase-signal transducer and activator of transcription
ACE2angiotensin-converting enzyme 2
LOADLate-onset AD
VEGF-Avascular endothelial growth factor A
IGF-1insulin-like growth factor-1

References

  1. Ahmad, E.; Lim, S.; Lamptey, R.; Webb, D.R.; Davies, M.J. Type 2 diabetes. Lancet 2022, 400, 1803–1820. [Google Scholar] [CrossRef] [PubMed]
  2. Alejandro, E.U.; Gregg, B.; Blandino-Rosano, M.; Cras-Méneur, C.; Bernal-Mizrachi, E. Natural history of β-cell adaptation and failure in type 2 diabetes. Mol. Asp. Med. 2015, 42, 19–41. [Google Scholar] [CrossRef]
  3. Narayan, K.M.V.; Kondal, D.; Chang, H.H.; Mohan, D.; Gujral, U.P.; Anjana, R.M.; Staimez, L.R.; Patel, S.A.; Ali, M.K.; Prabhakaran, D.; et al. Natural History of Type 2 Diabetes in Indians: Time to Progression. Diabetes Care 2024, 47, 858–863. [Google Scholar] [CrossRef]
  4. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: A systematic analysis for the Global Burden of Disease Study 2021. Lancet 2023, 402, 203–234. [CrossRef] [PubMed]
  5. Rooney, M.R.; Fang, M.; Ogurtsova, K.; Ozkan, B.; Echouffo-Tcheugui, J.B.; Boyko, E.J.; Magliano, D.J.; Selvin, E. Global Prevalence of Prediabetes. Diabetes Care 2023, 46, 1388–1394. [Google Scholar] [CrossRef]
  6. Sachdev, P.S.; Blacker, D.; Blazer, D.G.; Ganguli, M.; Jeste, D.V.; Paulsen, J.S.; Petersen, R.C. Classifying neurocognitive disorders: The DSM-5 approach. Nat. Rev. Neurol. 2014, 10, 634–642. [Google Scholar] [CrossRef]
  7. Langa, K.M.; Levine, D.A. The diagnosis and management of mild cognitive impairment: A clinical review. JAMA 2014, 312, 2551–2561. [Google Scholar] [CrossRef] [PubMed]
  8. Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chételat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
  9. Deture, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef]
  10. O’Brien, J.T.; Thomas, A. Vascular dementia. Lancet 2015, 386, 1698–1706. [Google Scholar] [CrossRef]
  11. van der Flier, W.M.; Skoog, I.; Schneider, J.A.; Pantoni, L.; Mok, V.; Chen, C.L.H.; Scheltens, P. Vascular cognitive impairment. Nat. Rev. Dis. Primers 2018, 4, 18003. [Google Scholar] [CrossRef] [PubMed]
  12. Ott, A.; Stolk, R.P.; Hofman, A.; van Harskamp, F.; Grobbee, D.E.; Breteler, M.M. Association of diabetes mellitus and dementia: The Rotterdam Study. Diabetologia 1996, 39, 1392–1397. [Google Scholar] [CrossRef]
  13. Mukadam, N.; Wolters, F.J.; Walsh, S.; Wallace, L.; Brayne, C.; Matthews, F.E.; Sacuiu, S.; Skoog, I.; Seshadri, S.; Beiser, A.; et al. Changes in prevalence and incidence of dementia and risk factors for dementia: An analysis from cohort studies. Lancet Public Health 2024, 9, e443–e460. [Google Scholar] [CrossRef] [PubMed]
  14. Chekol Tassew, W.; Ferede, Y.A.; Zeleke, A.M. Cognitive impairment and associated factors among patients with diabetes mellitus in Africa: A systematic review and meta-analysis. Front. Endocrinol. 2024, 15, 1386600. [Google Scholar] [CrossRef] [PubMed]
  15. Kuate Defo, A.; Bakula, V.; Pisaturo, A.; Labos, C.; Wing, S.S.; Daskalopoulou, S.S. Diabetes, antidiabetic medications and risk of dementia: A systematic umbrella review and meta-analysis. Diabetes Obes. Metab. 2024, 26, 441–462. [Google Scholar] [CrossRef]
  16. Chen, Q.; Zhu, S.; Shang, J.; Fang, Q.; Xue, Q.; Hua, J. Trends in Cognitive Function Before and After Diabetes Onset: The China Health and Retirement Longitudinal Study. Neurology 2024, 102, e209165. [Google Scholar] [CrossRef] [PubMed]
  17. American Diabetes Association Professional Practice Committee. 13. Older Adults: Standards of Care in Diabetes-2024. Diabetes Care 2024, 47, S244–S257. [Google Scholar] [CrossRef]
  18. Brown, E.; Heerspink, H.J.L.; Cuthbertson, D.J.; Wilding, J.P.H. SGLT2 inhibitors and GLP-1 receptor agonists: Established and emerging indications. Lancet 2021, 398, 262–276. [Google Scholar] [CrossRef]
  19. Sunwoo, Y.; Park, J.; Choi, C.-Y.; Shin, S.; Choi, Y.J. Risk of Dementia and Alzheimer’s Disease Associated With Antidiabetics: A Bayesian Network Meta-Analysis. Am. J. Prev. Med. 2024, 67, 434–443. [Google Scholar] [CrossRef]
  20. Tian, S.; Jiang, J.; Wang, J.; Zhang, Z.; Miao, Y.; Ji, X.; Bi, Y. Comparison on cognitive outcomes of antidiabetic agents for type 2 diabetes: A systematic review and network meta-analysis. Diabetes Metab. Res. Rev. 2023, 39, e3673. [Google Scholar] [CrossRef]
  21. Haeusler, R.A.; McGraw, T.E.; Accili, D. Biochemical and cellular properties of insulin receptor signalling. Nat. Rev. Mol. Cell Biol. 2018, 19, 31–44. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, L.; Li, J.; Di, L.-J. Glycogen synthesis and beyond, a comprehensive review of GSK3 as a key regulator of metabolic pathways and a therapeutic target for treating metabolic diseases. Med. Res. Rev. 2022, 42, 946–982. [Google Scholar] [CrossRef] [PubMed]
  23. Scherer, T.; Sakamoto, K.; Buettner, C. Brain insulin signalling in metabolic homeostasis and disease. Nat. Rev. Endocrinol. 2021, 17, 468–483. [Google Scholar] [CrossRef]
  24. Coppari, R.; Bjørbæk, C. Leptin revisited: Its mechanism of action and potential for treating diabetes. Nat. Rev. Drug Discov. 2012, 11, 692–708. [Google Scholar] [CrossRef]
  25. Welters, H.J.; Kulkarni, R.N. Wnt signaling: Relevance to beta-cell biology and diabetes. Trends Endocrinol. Metab. 2008, 19, 349–355. [Google Scholar] [CrossRef]
  26. Meske, V.; Albert, F.; Ohm, T.G. Coupling of mammalian target of rapamycin with phosphoinositide 3-kinase signaling pathway regulates protein phosphatase 2A- and glycogen synthase kinase-3 -dependent phosphorylation of Tau. J. Biol. Chem. 2008, 283, 100–109. [Google Scholar] [CrossRef]
  27. Jolivalt, C.G.; Lee, C.A.; Beiswenger, K.K.; Smith, J.L.; Orlov, M.; Torrance, M.A.; Masliah, E. Defective insulin signaling pathway and increased glycogen synthase kinase-3 activity in the brain of diabetic mice: Parallels with Alzheimer’s disease and correction by insulin. J. Neurosci. Res. 2008, 86, 3265–3274. [Google Scholar] [CrossRef]
  28. Zhang, T.; Pan, B.-S.; Sun, G.-C.; Sun, X.; Sun, F.-Y. Diabetes synergistically exacerbates poststroke dementia and tau abnormality in brain. Neurochem. Int. 2010, 56, 955–961. [Google Scholar] [CrossRef] [PubMed]
  29. Qu, Z.; Jiao, Z.; Sun, X.; Zhao, Y.; Ren, J.; Xu, G. Effects of streptozotocin-induced diabetes on tau phosphorylation in the rat brain. Brain Res. 2011, 1383, 300–306. [Google Scholar] [CrossRef]
  30. Hu, S.H.; Jiang, T.; Yang, S.S.; Yang, Y. Pioglitazone ameliorates intracerebral insulin resistance and tau-protein hyperphosphorylation in rats with type 2 diabetes. Exp. Clin. Endocrinol. Diabetes 2013, 121, 220–224. [Google Scholar] [CrossRef]
  31. Yang, Y.; Ma, D.; Wang, Y.; Jiang, T.; Hu, S.; Zhang, M.; Yu, X.; Gong, C.-X. Intranasal insulin ameliorates tau hyperphosphorylation in a rat model of type 2 diabetes. J. Alzheimers Dis. 2013, 33, 329–338. [Google Scholar] [CrossRef] [PubMed]
  32. Gao, C.; Liu, Y.; Jiang, Y.; Ding, J.; Li, L. Geniposide ameliorates learning memory deficits, reduces tau phosphorylation and decreases apoptosis via GSK3β pathway in streptozotocin-induced alzheimer rat model. Brain Pathol. 2014, 24, 261–269. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, W.; Yang, Y.; Yuan, G.; Zhu, W.; Ma, D.; Hu, S. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces Alzheimer disease-associated tau hyperphosphorylation in the hippocampus of rats with type 2 diabetes. J. Investig. Med. 2015, 63, 267–272. [Google Scholar] [CrossRef]
  34. Ma, D.-L.; Chen, F.-Q.; Xu, W.-J.; Yue, W.-Z.; Yuan, G.; Yang, Y. Early intervention with glucagon-like peptide 1 analog liraglutide prevents tau hyperphosphorylation in diabetic db/db mice. J. Neurochem. 2015, 135, 301–308. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, Y.; Yin, F.; Liu, J.; Liu, Z. Geniposide Attenuates the Phosphorylation of Tau Protein in Cellular and Insulin-deficient APP/PS1 Transgenic Mouse Model of Alzheimer’s Disease. Chem. Biol. Drug Des. 2016, 87, 409–418. [Google Scholar] [CrossRef]
  36. Wang, X.; Zhao, L. Calycosin ameliorates diabetes-induced cognitive impairments in rats by reducing oxidative stress via the PI3K/Akt/GSK-3β signaling pathway. Biochem. Biophys. Res. Commun. 2016, 473, 428–434. [Google Scholar] [CrossRef]
  37. Xu, W.; Liu, J.; Ma, D.; Yuan, G.; Lu, Y.; Yang, Y. Capsaicin reduces Alzheimer-associated tau changes in the hippocampus of type 2 diabetes rats. PLoS ONE 2017, 12, e0172477. [Google Scholar] [CrossRef]
  38. Ma, Q.-H.; Jiang, L.-F.; Mao, J.-L.; Xu, W.-X.; Huang, M. Vildagliptin prevents cognitive deficits and neuronal apoptosis in a rat model of Alzheimer’s disease. Mol. Med. Rep. 2018, 17, 4113–4119. [Google Scholar] [CrossRef]
  39. Zhou, M.; Chen, S.; Peng, P.; Gu, Z.; Yu, J.; Zhao, G.; Deng, Y. Dulaglutide ameliorates STZ induced AD-like impairment of learning and memory ability by modulating hyperphosphorylation of tau and NFs through GSK3β. Biochem. Biophys. Res. Commun. 2019, 511, 154–160. [Google Scholar] [CrossRef]
  40. Peng, X.; Shi, X.; Huang, J.; Zhang, S.; Yan, Y.; Ma, D.; Xu, W.; Xu, W.; Dong, K.; Tao, J.; et al. Exendin-4 Improves Cognitive Function of Diabetic Mice via Increasing Brain Insulin Synthesis. Curr. Alzheimer Res. 2021, 18, 546–557. [Google Scholar] [CrossRef]
  41. Latina, V.; Giacovazzo, G.; Calissano, P.; Atlante, A.; La Regina, F.; Malerba, F.; Dell’Aquila, M.; Stigliano, E.; Balzamino, B.O.; Micera, A.; et al. Tau Cleavage Contributes to Cognitive Dysfunction in Strepto-Zotocin-Induced Sporadic Alzheimer’s Disease (sAD) Mouse Model. Int. J. Mol. Sci. 2021, 22, 12158. [Google Scholar] [CrossRef] [PubMed]
  42. He, S.; Peng, W.-B.; Fu, X.-J.; Zhou, H.-L.; Wang, Z.-G. Deep Sea Water Alleviates Tau Phosphorylation and Cognitive Impairment via PI3K/Akt/GSK-3β Pathway. Mar. Biotechnol. 2022, 24, 68–81. [Google Scholar] [CrossRef]
  43. Zaki, M.O.; El-Desouky, S.; Elsherbiny, D.A.; Salama, M.; Azab, S.S. Glimepiride mitigates tauopathy and neuroinflammation in P301S transgenic mice: Role of AKT/GSK3β signaling. Inflammopharmacology 2022, 30, 1871–1890. [Google Scholar] [CrossRef]
  44. Jingxuan, L.; Litian, M.; Yanyang, T.; Jianfang, F. Knockdown of CLC-3 may improve cognitive impairment caused by diabetic encephalopathy. Diabetes Res. Clin. Pract. 2022, 190, 109970. [Google Scholar] [CrossRef]
  45. Chen, J.; Liu, Y.; Zhou, K.; Zhang, W.; Wen, B.; Xu, K.; Liu, Y.; Chen, L.; Huang, Y.; He, B.; et al. DISC1 inhibits GSK3β activity to prevent tau hyperphosphorylation under diabetic encephalopathy. Biofactors 2023, 49, 173–184. [Google Scholar] [CrossRef] [PubMed]
  46. Khoramipour, K.; Bejeshk, M.A.; Rajizadeh, M.A.; Najafipour, H.; Dehghan, P.; Farahmand, F. High-Intensity Interval Training Ameliorates Molecular Changes in the Hippocampus of Male Rats with the Diabetic Brain: The Role of Adiponectin. Mol. Neurobiol. 2023, 60, 3486–3495. [Google Scholar] [CrossRef] [PubMed]
  47. Moghazy, H.M.; Abdelhaliem, N.G.; Mohammed, S.A.; Hassan, A.; Abdelrahman, A. Liraglutide versus pramlintide in protecting against cognitive function impairment through affecting PI3K/AKT/GSK-3β/TTBK1 pathway and decreasing Tau hyperphosphorylation in high-fat diet- streptozocin rat model. Pflug. Arch. 2024, 476, 779–795. [Google Scholar] [CrossRef] [PubMed]
  48. Lu, Y.-W.; Xie, L.-Y.; Qi, M.-H.; Ren, S.; Wang, Y.-Q.; Hu, J.-N.; Wang, Z.; Tang, S.; Zhang, J.-T.; Li, W. Platycodin D Ameliorates Cognitive Impairment in Type 2 Diabetes Mellitus Mice via Regulating PI3K/Akt/GSK3β Signaling Pathway. J. Agric. Food Chem. 2024, 72, 12516–12528. [Google Scholar] [CrossRef]
  49. Rezaei, M.H.; Madadizadeh, E.; Aminaei, M.; Abbaspoor, M.; Schierbauer, J.; Moser, O.; Khoramipour, K.; Chamari, K. Leptin Signaling Could Mediate Hippocampal Decumulation of Beta-Amyloid and Tau Induced by High-Intensity Interval Training in Rats with Type 2 Diabetes. Cell Mol. Neurobiol. 2023, 43, 3465–3478. [Google Scholar] [CrossRef]
  50. Zhao, Y.; Wang, Z.; Mao, Y.; Li, B.; Zhu, Y.; Zhang, S.; Wang, S.; Jiang, Y.; Xu, N.; Xie, Y.; et al. NEAT1 regulates microtubule stabilization via FZD3/GSK3β/P-tau pathway in SH-SY5Y cells and APP/PS1 mice. Aging 2020, 12, 23233–23250. [Google Scholar] [CrossRef]
  51. Muneeb, M.; Mansou, S.M.; Saleh, S.; Mohammed, R.A. Vitamin D and rosuvastatin alleviate type-II diabetes-induced cognitive dysfunction by modulating neuroinflammation and canonical/noncanonical Wnt/β-catenin signaling. PLoS ONE 2022, 17, e0277457. [Google Scholar] [CrossRef] [PubMed]
  52. El-Safty, H.; Ismail, A.; Abdelsalam, R.M.; El-Sahar, A.E.; Saad, M.A. Dapagliflozin diminishes memory and cognition impairment in Streptozotocin induced diabetes through its effect on Wnt/β-Catenin and CREB pathway. Brain Res. Bull. 2022, 181, 109–120. [Google Scholar] [CrossRef] [PubMed]
  53. Kang, X.; Wang, D.; Zhang, L.; Huang, T.; Liu, S.; Feng, X.; Guo, Y.; Zhang, Z.; Wang, Z.; Ren, H.; et al. Exendin-4 ameliorates tau hyperphosphorylation and cognitive impairment in type 2 diabetes through acting on Wnt/β-catenin/NeuroD1 pathway. Mol. Med. 2023, 29, 118. [Google Scholar] [CrossRef] [PubMed]
  54. Zhao, F.; Guo, L.; Huang, T.; Liu, C.; Wu, D.; Fang, L.; Min, W. Interaction between the Neuroprotective and Hyperglycemia Mitigation Effects of Walnut-Derived Peptide LVRL via the Wnt3a/β-Catenin/GSK-3β Pathway in a Type 2 Diabetes Mellitus Model. J. Agric. Food Chem. 2024, 72, 16204–16220. [Google Scholar] [CrossRef] [PubMed]
  55. Dey, A.; Hao, S.; Wosiski-Kuhn, M.; Stranahan, A.M. Glucocorticoid-mediated activation of GSK3β promotes tau phosphorylation and impairs memory in type 2 diabetes. Neurobiol. Aging 2017, 57, 75–83. [Google Scholar] [CrossRef]
  56. Elahi, M.; Motoi, Y.; Shimonaka, S.; Ishida, Y.; Hioki, H.; Takanashi, M.; Ishiguro, K.; Imai, Y.; Hattori, N. High-fat diet-induced activation of SGK1 promotes Alzheimer’s disease-associated tau pathology. Hum. Mol. Genet. 2021, 30, 1693–1710. [Google Scholar] [CrossRef]
  57. Jack, C.R.; Andrews, J.S.; Beach, T.G.; Buracchio, T.; Dunn, B.; Graf, A.; Hansson, O.; Ho, C.; Jagust, W.; McDade, E.; et al. Revised criteria for diagnosis and staging of Alzheimer’s disease: Alzheimer’s Association Workgroup. Alzheimers Dement. 2024, 20, 5143–5169. [Google Scholar] [CrossRef]
  58. Deng, Y.; Wang, Z.; Wang, R.; Zhang, X.; Zhang, S.; Wu, Y.; Staufenbiel, M.; Cai, F.; Song, W. Amyloid-β protein (Aβ) Glu11 is the major β-secretase site of β-site amyloid-β precursor protein-cleaving enzyme 1(BACE1), and shifting the cleavage site to Aβ Asp1 contributes to Alzheimer pathogenesis. Eur. J. Neurosci. 2013, 37, 1962–1969. [Google Scholar] [CrossRef]
  59. Tian, Y.; Jing, G.; Zhang, M. Insulin-degrading enzyme: Roles and pathways in ameliorating cognitive impairment associated with Alzheimer’s disease and diabetes. Ageing Res. Rev. 2023, 90, 101999. [Google Scholar] [CrossRef]
  60. Kurochkin, I.V.; Guarnera, E.; Berezovsky, I.N. Insulin-Degrading Enzyme in the Fight against Alzheimer’s Disease. Trends Pharmacol. Sci. 2018, 39, 49–58. [Google Scholar] [CrossRef]
  61. Cordes, C.M.; Bennett, R.G.; Siford, G.L.; Hamel, F.G. Nitric oxide inhibits insulin-degrading enzyme activity and function through S-nitrosylation. Biochem. Pharmacol. 2009, 77, 1064–1073. [Google Scholar] [CrossRef] [PubMed]
  62. Cordes, C.M.; Bennett, R.G.; Siford, G.L.; Hamel, F.G. Redox regulation of insulin degradation by insulin-degrading enzyme. PLoS ONE 2011, 6, e18138. [Google Scholar] [CrossRef]
  63. Akhtar, M.W.; Sanz-Blasco, S.; Dolatabadi, N.; Parker, J.; Chon, K.; Lee, M.S.; Soussou, W.; McKercher, S.R.; Ambasudhan, R.; Nakamura, T.; et al. Elevated glucose and oligomeric β-amyloid disrupt synapses via a common pathway of aberrant protein S-nitrosylation. Nat. Commun. 2016, 7, 10242. [Google Scholar] [CrossRef] [PubMed]
  64. Kubis-Kubiak, A.; Wiatrak, B.; Piwowar, A. Hyper-glycemia and insulinemia induce morphological changes and modulate secretion of S100B, S100A8, amyloid β 1-40 and amyloid β 1-42, in a model of human dopaminergic neurons. Biomed. Pharmacother. 2022, 156, 113869. [Google Scholar] [CrossRef]
  65. Taubes, G. Neuroscience. Insulin insults may spur Alzheimer’s disease. Science 2003, 301, 40–41. [Google Scholar] [CrossRef]
  66. Farris, W.; Mansourian, S.; Chang, Y.; Lindsley, L.; Eckman, E.A.; Frosch, M.P.; Eckman, C.B.; Tanzi, R.E.; Selkoe, D.J.; Guenette, S. Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo. Proc. Natl. Acad. Sci. USA 2003, 100, 4162–4167. [Google Scholar] [CrossRef] [PubMed]
  67. Farris, W.; Mansourian, S.; Leissring, M.A.; Eckman, E.A.; Bertram, L.; Eckman, C.B.; Tanzi, R.E.; Selkoe, D.J. Partial loss-of-function mutations in insulin-degrading enzyme that induce diabetes also impair degradation of amyloid beta-protein. Am. J. Pathol. 2004, 164, 1425–1434. [Google Scholar] [CrossRef]
  68. Zhao, L.; Teter, B.; Morihara, T.; Lim, G.P.; Ambegaokar, S.S.; Ubeda, O.J.; Frautschy, S.A.; Cole, G.M. Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: Implications for Alzheimer’s disease intervention. J. Neurosci. 2004, 24, 11120–11126. [Google Scholar] [CrossRef] [PubMed]
  69. Farris, W.; Leissring, M.A.; Hemming, M.L.; Chang, A.Y.; Selkoe, D.J. Alternative splicing of human insulin-degrading enzyme yields a novel isoform with a decreased ability to degrade insulin and amyloid beta-protein. Biochemistry 2005, 44, 6513–6525. [Google Scholar] [CrossRef]
  70. Im, H.; Manolopoulou, M.; Malito, E.; Shen, Y.; Zhao, J.; Neant-Fery, M.; Sun, C.-Y.; Meredith, S.C.; Sisodia, S.S.; Leissring, M.A.; et al. Structure of substrate-free human insulin-degrading enzyme (IDE) and biophysical analysis of ATP-induced conformational switch of IDE. J. Biol. Chem. 2007, 282, 25453–25463. [Google Scholar] [CrossRef]
  71. Zhao, J.; Li, L.; Leissring, M.A. Insulin-degrading enzyme is exported via an unconventional protein secretion pathway. Mol. Neurodegener. 2009, 4, 4. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, Y.; Liu, L.; Lu, S.; Wang, D.; Liu, X.; Xie, L.; Wang, G. Impaired amyloid β-degrading enzymes in brain of streptozotocin-induced diabetic rats. J. Endocrinol. Investig. 2011, 34, 26–31. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, J.; Zhang, Y.; Deng, X.; Yin, F. Geniposide decreases the level of Aβ1-42 in the hippocampus of streptozotocin-induced diabetic rats. Acta Biochim. Biophys. Sin. 2013, 45, 787–791. [Google Scholar] [CrossRef]
  74. Mittal, K.; Mani, R.J.; Katare, D.P. Type 3 Diabetes: Cross Talk between Differentially Regulated Proteins of Type 2 Diabetes Mellitus and Alzheimer’s Disease. Sci. Rep. 2016, 6, 25589. [Google Scholar] [CrossRef]
  75. Song, M.K.; Bischoff, D.S.; Song, A.M.; Uyemura, K.; Yamaguchi, D.T. Metabolic relationship between diabetes and Alzheimer’s Disease affected by Cyclo(His-Pro) plus zinc treatment. BBA Clin. 2017, 7, 41–54. [Google Scholar] [CrossRef]
  76. Yang, S.; Chen, Z.; Cao, M.; Li, R.; Wang, Z.; Zhang, M. Pioglitazone ameliorates Aβ42 deposition in rats with diet-induced insulin resistance associated with AKT/GSK3β activation. Mol. Med. Rep. 2017, 15, 2588–2594. [Google Scholar] [CrossRef]
  77. Li, H.; Wu, J.; Zhu, L.; Sha, L.; Yang, S.; Wei, J.; Ji, L.; Tang, X.; Mao, K.; Cao, L.; et al. Insulin degrading enzyme contributes to the pathology in a mixed model of Type 2 diabetes and Alzheimer’s disease: Possible mechanisms of IDE in T2D and AD. Biosci. Rep. 2018, 38, BSR20170862. [Google Scholar] [CrossRef] [PubMed]
  78. Kazkayasi, I.; Burul-Bozkurt, N.; Ismail, M.-A.-M.; Merino-Serrais, P.; Pekiner, C.; Cedazo-Minguez, A.; Uma, S. Insulin deprivation decreases insulin degrading enzyme levels in primary cultured cortical neurons and in the cerebral cortex of rats with streptozotocin-induced diabetes. Pharmacol. Rep. 2018, 70, 677–683. [Google Scholar] [CrossRef]
  79. Zhang, Y.; Wang, P. Age-Related Increase of Insulin-Degrading Enzyme Is Inversely Correlated with Cognitive Function in APPswe/PS1dE9 Mice. Med. Sci. Monit. 2018, 24, 2446–2455. [Google Scholar] [CrossRef]
  80. Kang, J.Y.; Park, S.K.; Kim, J.M.; Park, S.B.; Yoo, S.K.; Han, H.J.; Kim, D.O.; Heo, H.J. 4,5-dicaffeyolquinic acid improves high-fat diet-induced cognitive dysfunction through the regulation of insulin degrading enzyme. J. Food Biochem. 2019, 43, e12855. [Google Scholar] [CrossRef]
  81. Hayrabedyan, S.; Todorova, K.; Spinelli, M.; Barnea, E.R.; Mueller, M. The core sequence of PIF competes for insulin/amyloid β in insulin degrading enzyme: Potential treatment for Alzheimer’s disease. Oncotarget 2018, 9, 33884–33895. [Google Scholar] [CrossRef] [PubMed]
  82. Li, H.; Cao, L.; Ren, Y.; Jiang, Y.; Xie, W.; Li, D. GLP-1 receptor regulates cell growth through regulating IDE expression level in Aβ1-42-treated PC12 cells. Biosci. Rep. 2018, 38, BSR20171284. [Google Scholar] [CrossRef]
  83. Fernández-de Frutos, M.; Galán-Chilet, I.; Goedeke, L.; Kim, B.; Pardo-Marqués, V.; Pérez-García, A.; Herrero, J.I.; Fernández-Hernando, C.; Kim, J.; Ramírez, C.M. MicroRNA 7 Impairs Insulin Signaling and Regulates Aβ Levels through Posttranscriptional Regulation of the Insulin Receptor Substrate 2, Insulin Receptor, Insulin-Degrading Enzyme, and Liver X Receptor Pathway. Mol. Cell Biol. 2019, 39, e00170-19. [Google Scholar] [CrossRef] [PubMed]
  84. Bosoi, C.R.; Vandal, M.; Tournissac, M.; Leclerc, M.; Fanet, H.; Mitchell, P.L.; Verreault, M.; Trottier, J.; Virgili, J.; Tremblay, C.; et al. High-Fat Diet Modulates Hepatic Amyloid β and Cerebrosterol Metabolism in the Triple Transgenic Mouse Model of Alzheimer’s Disease. Hepatol. Commun. 2021, 5, 446–460. [Google Scholar] [CrossRef]
  85. Martín-Martín, Y.; Pérez-García, A.; Torrecilla-Parra, M.; Fernández-de Frutos, M.; Pardo-Marqués, V.; Casarejos, M.J.; Busto, R.; Ramírez, C.M. New Insights on the Regulation of the Insulin-Degrading Enzyme: Role of microRNAs and RBPs. Cells 2022, 11, 2538. [Google Scholar] [CrossRef]
  86. Sim, A.Y.; Choi, D.H.; Kim, J.Y.; Kim, E.R.; Goh, A.R.; Lee, Y.-H.; Lee, J.E. SGLT2 and DPP4 inhibitors improve Alzheimer’s disease-like pathology and cognitive function through distinct mechanisms in a T2D-AD mouse model. Biomed. Pharmacother. 2023, 168, 115755. [Google Scholar] [CrossRef] [PubMed]
  87. Morelli, L.; Llovera, R.E.; Mathov, I.; Lue, L.-F.; Frangione, B.; Ghiso, J.; Castaño, E.M. Insulin-degrading enzyme in brain microvessels: Proteolysis of amyloid {beta} vasculotropic variants and reduced activity in cerebral amyloid angiopathy. J. Biol. Chem. 2004, 279, 56004–56013. [Google Scholar] [CrossRef]
  88. Inoue, Y.; Ueda, M.; Masuda, T.; Misumi, Y.; Yamashita, T.; Ando, Y. Memantine, a Noncompetitive N-Methyl-D-Aspartate Receptor Antagonist, Attenuates Cerebral Amyloid Angiopathy by Increasing Insulin-Degrading Enzyme Expression. Mol. Neurobiol. 2019, 56, 8573–8588. [Google Scholar] [CrossRef]
  89. Inoue, Y.; Masuda, T.; Misumi, Y.; Ando, Y.; Ueda, M. Metformin attenuates vascular pathology by increasing expression of insulin-degrading enzyme in a mixed model of cerebral amyloid angiopathy and type 2 diabetes mellitus. Neurosci. Lett. 2021, 762, 136136. [Google Scholar] [CrossRef]
  90. Devi, L.; Alldred, M.J.; Ginsberg, S.D.; Ohno, M. Mechanisms underlying insulin deficiency-induced acceleration of β-amyloidosis in a mouse model of Alzheimer’s disease. PLoS ONE 2012, 7, e32792. [Google Scholar] [CrossRef]
  91. Pandini, G.; Pace, V.; Copani, A.; Squatrito, S.; Milardi, D.; Vigneri, R. Insulin has multiple antiamyloidogenic effects on human neuronal cells. Endocrinology 2013, 154, 375–387. [Google Scholar] [CrossRef] [PubMed]
  92. Shingo, A.S.; Kanabayashi, T.; Kito, S.; Murase, T. Intracerebroventricular administration of an insulin analogue recovers STZ-induced cognitive decline in rats. Behav. Brain Res. 2013, 241, 105–111. [Google Scholar] [CrossRef]
  93. He, Z.; Han, S.; Zhu, H.; Hu, X.; Li, X.; Hou, C.; Wu, C.; Xie, Q.; Li, N.; Du, X.; et al. The Protective Effect of Vanadium on Cognitive Impairment and the Neuropathology of Alzheimer’s Disease in APPSwe/PS1dE9 Mice. Front. Mol. Neurosci. 2020, 13, 21. [Google Scholar] [CrossRef] [PubMed]
  94. Alafuzoff, I.; Aho, L.; Helisalmi, S.; Mannermaa, A.; Soininen, H. Beta-amyloid deposition in brains of subjects with diabetes. Neuropathol. Appl. Neurobiol. 2009, 35, 60–68. [Google Scholar] [CrossRef]
  95. Ramos-Rodriguez, J.J.; Ortiz-Barajas, O.; Gamero-Carrasco, C.; de la Rosa, P.R.; Infante-Garcia, C.; Zopeque-Garcia, N.; Lechuga-Sancho, A.M.; Garcia-Alloza, M. Prediabetes-induced vascular alterations exacerbate central pathology in APPswe/PS1dE9 mice. Psychoneuroendocrinology 2014, 48, 123–135. [Google Scholar] [CrossRef] [PubMed]
  96. Little, K.; Llorián-Salvador, M.; Scullion, S.; Hernández, C.; Simó-Servat, O.; Del Marco, A.; Bosma, E.; Vargas-Soria, M.; Carranza-Naval, M.J.; Van Bergen, T.; et al. Common pathways in dementia and diabetic retinopathy: Understanding the mechanisms of diabetes-related cognitive decline. Trends Endocrinol. Metab. 2022, 33, 50–71. [Google Scholar] [CrossRef]
  97. Song, X.; Zhu, Z.; Qian, X.; Liu, X.; Chen, S.; Tang, H. Multi-Omics Characterization of Type 2 Diabetes Mellitus-Induced Cognitive Impairment in the db/db Mouse Model. Molecules 2022, 27, 1904. [Google Scholar] [CrossRef]
  98. Cassano, V.; Leo, A.; Tallarico, M.; Nesci, V.; Cimellaro, A.; Fiorentino, T.V.; Citraro, R.; Hribal, M.L.; De Sarro, G.; Perticone, F.; et al. Metabolic and Cognitive Effects of Ranolazine in Type 2 Diabetes Mellitus: Data from an in vivo Model. Nutrients 2020, 12, 382. [Google Scholar] [CrossRef]
  99. Hui, Y.; Xu, Z.; Li, J.; Kuang, L.; Zhong, Y.; Tang, Y.; Wei, J.; Zhou, H.; Zheng, T. Nonenzymatic function of DPP4 promotes diabetes-associated cognitive dysfunction through IGF-2R/PKA/SP1/ERp29/IP3R2 pathway-mediated impairment of Treg function and M1 microglia polarization. Metabolism 2023, 138, 155340. [Google Scholar] [CrossRef]
  100. Fan, X.; Zhang, Y.; Song, Y.; Zhao, Y.; Xu, Y.; Guo, F.; Shao, M.; Ma, X.; Zhang, W.; Wei, F.; et al. Compound Danshen Dripping Pills moderate intestinal flora and the TLR4/MyD88/NF-κB signaling pathway in alleviating cognitive dysfunction in type 2 diabetic KK-Ay mice. Phytomedicine 2023, 111, 154656. [Google Scholar] [CrossRef]
  101. Li, Q.; Zhao, Y.; Guo, H.; Li, Q.; Yan, C.; Li, Y.; He, S.; Wang, N.; Wang, Q. Impaired lipophagy induced-microglial lipid droplets accumulation contributes to the buildup of TREM1 in diabetes-associated cognitive impairment. Autophagy 2023, 19, 2639–2656. [Google Scholar] [CrossRef]
  102. Li, Y.; Jiang, T.; Du, M.; He, S.; Huang, N.; Cheng, B.; Yan, C.; Tang, W.; Gao, W.; Guo, H.; et al. Ketohexokinase-dependent metabolism of cerebral endogenous fructose in microglia drives diabetes-associated cognitive dysfunction. Exp. Mol. Med. 2023, 55, 2417–2432. [Google Scholar] [CrossRef]
  103. Yao, Y.; Shi, J.; Zhang, C.; Gao, W.; Huang, N.; Liu, Y.; Yan, W.; Han, Y.; Zhou, W.; Kong, L. Pyruvate dehydrogenase kinase 1 protects against neuronal injury and memory loss in mouse models of diabetes. Cell Death Dis. 2023, 14, 722. [Google Scholar] [CrossRef]
  104. Wang, Z.; Cui, X.; Yan, W.; Liu, N.; Shang, J.; Yi, X.; Guo, T.; Wei, X.; Sun, Y.; Hu, H.; et al. Mollugin activates GLP-1R to improve cognitive dysfunction in type 2 diabetic mice. Life Sci. 2023, 331, 122026. [Google Scholar] [CrossRef]
  105. Poorgholam, P.; Yaghmaei, P.; Noureddini, M.; Hajebrahimi, Z. Artemisin and human endometrial-derived stem cells improve cognitive function and synaptic plasticity in a rat model of Alzheimer disease and diabetes. Metab. Brain Dis. 2023, 38, 1925–1936. [Google Scholar] [CrossRef]
  106. Cho, J.H.; Chae, C.W.; Lim, J.R.; Jung, Y.H.; Han, S.J.; Yoon, J.H.; Park, J.Y.; Han, H.J. Sodium butyrate ameliorates high glucose-suppressed neuronal mitophagy by restoring PRKN expression via inhibiting the RELA-HDAC8 complex. Autophagy 2024, 20, 1505–1522. [Google Scholar] [CrossRef]
  107. Dove, A.; Shang, Y.; Xu, W.; Grande, G.; Laukka, E.J.; Fratiglioni, L.; Marseglia, A. The impact of diabetes on cognitive impairment and its progression to dementia. Alzheimer’s Dement. 2021, 17, 1769–1778. [Google Scholar] [CrossRef]
  108. Filler, J.; Georgakis, M.K.; Dichgans, M. Risk factors for cognitive impairment and dementia after stroke: A systematic review and meta-analysis. Lancet Healthy Longev. 2024, 5, e31–e44. [Google Scholar] [CrossRef]
  109. Skrobot, O.A.; O’Brien, J.; Black, S.; Chen, C.; DeCarli, C.; Erkinjuntti, T.; Ford, G.A.; Kalaria, R.N.; Pantoni, L.; Pasquier, F.; et al. The Vascular Impairment of Cognition Classification Consensus Study. Alzheimers Dement. 2017, 13, 624–633. [Google Scholar] [CrossRef]
  110. American Diabetes Association Professional Practice Committee. 10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes-2024. Diabetes Care 2024, 47, S179–S218. [Google Scholar] [CrossRef]
  111. Blevins, B.L.; Vinters, H.V.; Love, S.; Wilcock, D.M.; Grinberg, L.T.; Schneider, J.A.; Kalaria, R.N.; Katsumata, Y.; Gold, B.T.; Wang, D.J.J.; et al. Brain arteriolosclerosis. Acta Neuropathol. 2021, 141, 1–24. [Google Scholar] [CrossRef]
  112. Wright, E.M. Glucose transport families SLC5 and SLC50. Mol. Aspects Med. 2013, 34, 183–196. [Google Scholar] [CrossRef]
  113. Ni, L.; Yuan, C.; Chen, G.; Zhang, C.; Wu, X. SGLT2i: Beyond the glucose-lowering effect. Cardiovasc. Diabetol. 2020, 19, 98. [Google Scholar] [CrossRef]
  114. Kintzoglanakis, K.; Diamantis, C.; Mariolis, A.; Paschou, S.A. Patient-important outcomes in type 2 diabetes: The paradigm of the sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists. Diabetes Vasc. Dis. Res. 2024, 21, 14791641241269743. [Google Scholar] [CrossRef]
  115. American Diabetes Association Professional Practice Committee. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2024. Diabetes Care 2024, 47, S158–S178. [Google Scholar] [CrossRef]
  116. Wu, C.-Y.; Iskander, C.; Wang, C.; Xiong, L.Y.; Shah, B.R.; Edwards, J.D.; Kapral, M.K.; Herrmann, N.; Lanctôt, K.L.; Masellis, M.; et al. Association of Sodium-Glucose Cotransporter 2 Inhibitors With Time to Dementia: A Population-Based Cohort Study. Diabetes Care 2023, 46, 297–304. [Google Scholar] [CrossRef]
  117. Mone, P.; Lombardi, A.; Gambardella, J.; Pansini, A.; Macina, G.; Morgante, M.; Frullone, S.; Santulli, G. Empagliflozin Improves Cognitive Impairment in Frail Older Adults With Type 2 Diabetes and Heart Failure With Preserved Ejection Fraction. Diabetes Care 2022, 45, 1247–1251. [Google Scholar] [CrossRef]
  118. Secnik, J.; Xu, H.; Schwertner, E.; Hammar, N.; Alvarsson, M.; Winblad, B.; Eriksdotter, M.; Garcia-Ptacek, S.; Religa, D. Glucose-Lowering Medications and Post-Dementia Survival in Patients with Diabetes and Dementia. J. Alzheimers Dis. 2022, 86, 245–257. [Google Scholar] [CrossRef]
  119. Ding, J.; Shi, Q.; Tao, Q.; Su, H.; Du, Y.; Pan, T.; Zhong, X. Correlation between long-term glycemic variability and cognitive function in middle-aged and elderly patients with type 2 diabetes mellitus: A retrospective study. PeerJ 2023, 11, e16698. [Google Scholar] [CrossRef]
  120. Mone, P.; Guerra, G.; Lombardi, A.; Illario, M.; Pansini, A.; Marro, A.; Frullone, S.; Taurino, A.; Sorriento, D.; Verri, V.; et al. Effects of SGLT2 inhibition via empagliflozin on cognitive and physical impairment in frail diabetic elders with chronic kidney disease. Pharmacol. Res. 2024, 200, 107055. [Google Scholar] [CrossRef]
  121. Mui, J.V.; Zhou, J.; Lee, S.; Leung, K.S.K.; Lee, T.T.L.; Chou, O.H.I.; Tsang, S.L.; Wai, A.K.C.; Liu, T.; Wong, W.T.; et al. Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitors vs. Dipeptidyl Peptidase-4 (DPP4) Inhibitors for New-Onset Dementia: A Propensity Score-Matched Population-Based Study With Competing Risk Analysis. Front. Cardiovasc. Med. 2021, 8, 747620. [Google Scholar] [CrossRef]
  122. Wium-Andersen, I.K.; Osler, M.; Jørgensen, M.B.; Rungby, J.; Wium-Andersen, M.K. Antidiabetic medication and risk of dementia in patients with type 2 diabetes: A nested case-control study. Eur. J. Endocrinol. 2019, 181, 499–507. [Google Scholar] [CrossRef]
  123. Low, S.; Goh, K.S.; Ng, T.P.; Moh, A.; Ang, S.F.; Wang, J.; Ang, K.; Tang, W.E.; Lim, Z.; Subramaniam, T.; et al. Association Between Use of Sodium-Glucose Co-Transporter-2 (SGLT2) Inhibitors and Cognitive Function in a Longitudinal Study of Patients with Type 2 Diabetes. J. Alzheimers Dis. 2022, 87, 635–642. [Google Scholar] [CrossRef]
  124. Zhang, S.; Wang, F.; Xie, L.; Xu, J.; Song, X.; Tao, J.; Chen, J.; Ma, D.; Yu, X.; Shi, X.; et al. Sodium-glucose cotransporter 2 inhibition through henagliflozin ameliorates cognitive impairment in patients with type 2 diabetes. J. Diabetes Investig. 2024, 15, 1596–1603. [Google Scholar] [CrossRef]
  125. Proietti, R.; Rivera-Caravaca, J.M.; López-Gálvez, R.; Harrison, S.L.; Marín, F.; Underhill, P.; Shantsila, E.; McDowell, G.; Vinciguerra, M.; Davies, R.; et al. Cerebrovascular, Cognitive and Cardiac Benefits of SGLT2 Inhibitors Therapy in Patients with Atrial Fibrillation and Type 2 Diabetes Mellitus: Results from a Global Federated Health Network Analysis. J. Clin. Med. 2023, 12, 2814. [Google Scholar] [CrossRef]
  126. Chen, Y.-Y.; Chang, H.-C.; Lin, Y.-J.; Chien, K.-L.; Hsieh, Y.-C.; Chung, F.-P.; Lin, C.-H.; Lip, G.Y.H.; Chen, S.-A. The impact of sodium-glucose co-transporter-2 inhibitors on dementia and cardiovascular events in diabetic patients with atrial fibrillation. Diabetes Metab. Res. Rev. 2024, 40, e3775. [Google Scholar] [CrossRef]
  127. Siao, W.-Z.; Lin, T.-K.; Huang, J.-Y.; Tsai, C.-F.; Jong, G.-P. The association between sodium-glucose cotransporter 2 inhibitors and incident dementia: A nationwide population-based longitudinal cohort study. Diabetes Vasc. Dis. Res. 2022, 19, 14791641221098168. [Google Scholar] [CrossRef]
  128. Edmonston, D.; Mulder, H.; Lydon, E.; Chiswell, K.; Lampron, Z.; Shay, C.; Marsolo, K.; Jones, W.S.; Butler, J.; Shah, R.C.; et al. Kidney and Cardiovascular Effectiveness of Empagliflozin Compared With Dipeptidyl Peptidase-4 Inhibitors in Patients With Type 2 Diabetes. Am. J. Cardiol. 2024, 221, 52–63. [Google Scholar] [CrossRef]
  129. Sharma, A.; Wood, S.; Bell, J.S.; De Blasio, M.J.; Ilomäki, J.; Ritchie, R.H. Sex differences in risk of cardiovascular events and mortality with sodium glucose co-transporter-2 inhibitors versus glucagon-like peptide 1 receptor agonists in Australians with type 2 diabetes: A population-based cohort study. Lancet Reg. Health West. Pac. 2023, 33, 100692. [Google Scholar] [CrossRef]
  130. 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]
  131. Wiviott, S.D.; Raz, I.; Bonaca, M.P.; Mosenzon, O.; Kato, E.T.; Cahn, A.; Silverman, M.G.; Zelniker, T.A.; Kuder, J.F.; Murphy, S.A.; et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2019, 380, 347–357. [Google Scholar] [CrossRef]
  132. Cannon, C.P.; Pratley, R.; Dagogo-Jack, S.; Mancuso, J.; Huyck, S.; Masiukiewicz, U.; Charbonnel, B.; Frederich, R.; Gallo, S.; Cosentino, F.; et al. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N. Engl. J. Med. 2020, 383, 1425–1435. [Google Scholar] [CrossRef]
  133. Riester, M.R.; Zullo, A.R.; Joshi, R.; Daiello, L.A.; Hayes, K.N.; Ko, D.; Kim, D.H.; Munshi, M.; Berry, S.D. Comparative safety and cardiovascular effectiveness of sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists in nursing homes. Diabetes Obes. Metab. 2024, 26, 3403–3417. [Google Scholar] [CrossRef]
  134. Perna, S.; Mainardi, M.; Astrone, P.; Gozzer, C.; Biava, A.; Bacchio, R.; Spadaccini, D.; Solerte, S.B.; Rondanelli, M. 12-month effects of incretins versus SGLT2-Inhibitors on cognitive performance and metabolic profile. A randomized clinical trial in the elderly with Type-2 diabetes mellitus. Clin. Pharmacol. 2018, 10, 141–151. [Google Scholar] [CrossRef]
  135. Zhao, Y.; Zhang, R.; Wang, S. Effect of Dapagliflozin Combined with Cognitive Behavior Training on Quality of Life and Cognitive Function in Elderly Patients with Type 2 Diabetes Mellitus Complicated with Mild Cognitive Impairment. Iran. J. Public Health 2022, 51, 1251–1258. [Google Scholar] [CrossRef]
  136. Youn, Y.J.; Kim, S.; Jeong, H.-J.; Ah, Y.-M.; Yu, Y.M. Sodium-glucose cotransporter-2 inhibitors and their potential role in dementia onset and cognitive function in patients with diabetes mellitus: A systematic review and meta-analysis. Front. Neuroendocrinol. 2024, 73, 101131. [Google Scholar] [CrossRef]
  137. Zhou, J.-B.; Tang, X.; Han, M.; Yang, J.; Simó, R. Impact of antidiabetic agents on dementia risk: A Bayesian network meta-analysis. Metabolism 2020, 109, 154265. [Google Scholar] [CrossRef]
  138. Jaiswal, V.; Mashkoor, Y.; Raj, N.; Rajak, K.; Jaiswal, A.; Fonarow, G.C. Association between SGLT2 Inhibitors and Risk of Dementia and Parkinson’s Disease: A Meta-Analysis of 12 Randomized Controlled Trials. Am. J. Med. 2024, 137, 1136–1141. [Google Scholar] [CrossRef]
  139. Banerjee, M.; Pal, R.; Maisnam, I.; Mukhopadhyay, S. GLP-1 receptor agonists, SGLT2 inhibitors and noncardiovascular mortality in type 2 diabetes: Insights from a meta-analysis. Diabetes Metab. Syndr. 2024, 18, 102943. [Google Scholar] [CrossRef]
  140. Tang, H.; Shao, H.; Shaaban, C.E.; Yang, K.; Brown, J.; Anton, S.; Wu, Y.; Bress, A.; Donahoo, W.T.; DeKosky, S.T.; et al. Newer glucose-lowering drugs and risk of dementia: A systematic review and meta-analysis of observational studies. J. Am. Geriatr. Soc. 2023, 71, 2096–2106. [Google Scholar] [CrossRef]
  141. Tang, H.; Donahoo, W.T.; Svensson, M.; Shaaban, C.E.; Smith, G.; Jaffee, M.S.; Huang, Y.; Hu, X.; Lu, Y.; Salloum, R.G.; et al. Heterogeneous treatment effects of sodium-glucose cotransporter 2 inhibitors on risk of dementia in people with type 2 diabetes: A population-based cohort study. Alzheimers Dement. 2024, 20, 5528–5539. [Google Scholar] [CrossRef]
  142. Cheng, H.; Zhang, Z.; Zhang, B.; Zhang, W.; Wang, J.; Ni, W.; Miao, Y.; Liu, J.; Bi, Y. Enhancement of Impaired Olfactory Neural Activation and Cognitive Capacity by Liraglutide, but Not Dapagliflozin or Acarbose, in Patients With Type 2 Diabetes: A 16-Week Randomized Parallel Comparative Study. Diabetes Care 2022, 45, 1201–1210. [Google Scholar] [CrossRef]
  143. Unno, K.; Taguchi, K.; Takagi, Y.; Hase, T.; Meguro, S.; Nakamura, Y. Mouse Models with SGLT2 Mutations: Toward Understanding the Role of SGLT2 beyond Glucose Reabsorption. Int. J. Mol. Sci. 2023, 24, 6278. [Google Scholar] [CrossRef]
  144. Piątkowska-Chmiel, I.; Herbet, M.; Gawrońska-Grzywacz, M.; Pawłowski, K.; Ostrowska-Leśko, M.; Dudka, J. Molecular and neural roles of sodium-glucose cotransporter 2 inhibitors in alleviating neurocognitive impairment in diabetic mice. Psychopharmacology 2023, 240, 983–1000. [Google Scholar] [CrossRef]
  145. Khan, T.; Khan, S.; Akhtar, M.; Ali, J.; Najmi, A.K. Empagliflozin nanoparticles attenuates type2 diabetes induced cognitive impairment via oxidative stress and inflammatory pathway in high fructose diet induced hyperglycemic mice. Neurochem. Int. 2021, 150, 105158. [Google Scholar] [CrossRef]
  146. Hayden, M.R.; Grant, D.G.; Aroor, A.R.; DeMarco, V.G. Empagliflozin Ameliorates Type 2 Diabetes-Induced Ultrastructural Remodeling of the Neurovascular Unit and Neuroglia in the Female db/db Mouse. Brain Sci. 2019, 9, 57. [Google Scholar] [CrossRef]
  147. Jayarathne, H.S.M.; Debarba, L.K.; Jaboro, J.J.; Ginsburg, B.C.; Miller, R.A.; Sadagurski, M. Neuroprotective effects of Canagliflozin: Lessons from aged genetically diverse UM-HET3 mice. Aging Cell 2022, 21, e13653. [Google Scholar] [CrossRef]
  148. Shaikh, S.; Rizvi, S.M.D.; Shakil, S.; Riyaz, S.; Biswas, D.; Jahan, R. Forxiga (dapagliflozin): Plausible role in the treatment of diabetes-associated neurological disorders. Biotechnol. Appl. Biochem. 2016, 63, 145–150. [Google Scholar] [CrossRef]
  149. Shakil, S. Molecular Interaction of Anti-Diabetic Drugs With Acetylcholinesterase and Sodium Glucose Co-Transporter 2. J. Cell Biochem. 2017, 118, 3855–3865. [Google Scholar] [CrossRef]
  150. Hierro-Bujalance, C.; Infante-Garcia, C.; Del Marco, A.; Herrera, M.; Carranza-Naval, M.J.; Suarez, J.; Alves-Martinez, P.; Lubian-Lopez, S.; Garcia-Alloza, M. Empagliflozin reduces vascular damage and cognitive impairment in a mixed murine model of Alzheimer’s disease and type 2 diabetes. Alzheimers Res. Ther. 2020, 12, 40. [Google Scholar] [CrossRef]
  151. O’Keefe, J.H.; Weidling, R.; O’Keefe, E.L.; Franco, W.G. SGLT inhibitors for improving Healthspan and lifespan. Prog. Cardiovasc. Dis. 2023, 81, 2–9. [Google Scholar] [CrossRef]
  152. Mancinetti, F.; Xenos, D.; De Fano, M.; Mazzieri, A.; Porcellati, F.; Boccardi, V.; Mecocci, P. Diabetes-Alzheimer’s connection in older age: SGLT2 inhibitors as promising modulators of disease pathways. Ageing Res. Rev. 2023, 90, 102018. [Google Scholar] [CrossRef]
  153. Rizzo, M.R.; Di Meo, I.; Polito, R.; Auriemma, M.C.; Gambardella, A.; di Mauro, G.; Capuano, A.; Paolisso, G. Cognitive impairment and type 2 diabetes mellitus: Focus of SGLT2 inhibitors treatment. Pharmacol. Res. 2022, 176, 106062. [Google Scholar] [CrossRef]
  154. Pawlos, A.; Broncel, M.; Woźniak, E.; Gorzelak-Pabiś, P. Neuroprotective Effect of SGLT2 Inhibitors. Molecules 2021, 26, 7213. [Google Scholar] [CrossRef]
  155. Esterline, R.; Oscarsson, J.; Burns, J. A role for sodium glucose cotransporter 2 inhibitors (SGLT2is) in the treatment of Alzheimer’s disease? Int. Rev. Neurobiol. 2020, 155, 113–140. [Google Scholar] [CrossRef]
  156. Chavda, V.; Vashi, R.; Patel, S. Cerebrovascular Complications of Diabetes: SGLT-2 Inhibitors as a Promising Future Therapeutics. Curr. Drug Targets 2021, 22, 1629–1636. [Google Scholar] [CrossRef]
  157. Wiciński, M.; Wódkiewicz, E.; Górski, K.; Walczak, M.; Malinowski, B. Perspective of SGLT2 Inhibition in Treatment of Conditions Connected to Neuronal Loss: Focus on Alzheimer’s Disease and Ischemia-Related Brain Injury. Pharmaceuticals 2020, 13, 379. [Google Scholar] [CrossRef]
  158. Avgerinos, K.I.; Mullins, R.J.; Vreones, M.; Mustapic, M.; Chen, Q.; Melvin, D.; Kapogiannis, D.; Egan, J.M. Empagliflozin Induced Ketosis, Upregulated IGF-1/Insulin Receptors and the Canonical Insulin Signaling Pathway in Neurons, and Decreased the Excitatory Neurotransmitter Glutamate in the Brain of Non-Diabetics. Cells 2022, 11, 3372. [Google Scholar] [CrossRef]
  159. Birajdar, S.V.; Mazahir, F.; Alam, M.I.; Kumar, A.; Yadav, A.K. Repurposing and clinical attributes of antidiabetic drugs for the treatment of neurodegenerative disorders. Eur. J. Pharmacol. 2023, 961, 176117. [Google Scholar] [CrossRef]
  160. Goodarzi, G.; Tehrani, S.S.; Fana, S.E.; Moradi-Sardareh, H.; Panahi, G.; Maniati, M.; Meshkani, R. Crosstalk between Alzheimer’s disease and diabetes: A focus on anti-diabetic drugs. Metab. Brain Dis. 2023, 38, 1769–1800. [Google Scholar] [CrossRef]
  161. Noel, J.A.; Hougen, I.; Sood, M.M. The Intersection of SGLT2 Inhibitors, Cognitive Impairment, and CKD. Front. Neurol. 2022, 13, 823569. [Google Scholar] [CrossRef] [PubMed]
  162. Mei, J.; Li, Y.; Niu, L.; Liang, R.; Tang, M.; Cai, Q.; Xu, J.; Zhang, D.; Yin, X.; Liu, X.; et al. SGLT2 inhibitors: A novel therapy for cognitive impairment via multifaceted effects on the nervous system. Transl. Neurodegener. 2024, 13, 41. [Google Scholar] [CrossRef] [PubMed]
  163. Youssef, M.E.; Yahya, G.; Popoviciu, M.S.; Cavalu, S.; Abd-Eldayem, M.A.; Saber, S. Unlocking the Full Potential of SGLT2 Inhibitors: Expanding Applications beyond Glycemic Control. Int. J. Mol. Sci. 2023, 24, 6039. [Google Scholar] [CrossRef] [PubMed]
  164. Sim, A.Y.; Barua, S.; Kim, J.Y.; Lee, Y.-H.; Lee, J.E. Role of DPP-4 and SGLT2 Inhibitors Connected to Alzheimer Disease in Type 2 Diabetes Mellitus. Front. Neurosci. 2021, 15, 708547. [Google Scholar] [CrossRef]
  165. Riemma, M.A.; Mele, E.; Donniacuo, M.; Telesca, M.; Bellocchio, G.; Castaldo, G.; Rossi, F.; De Angelis, A.; Cappetta, D.; Urbanek, K.; et al. Glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors, anti-diabetic drugs in heart failure and cognitive impairment: Potential mechanisms of the protective effects. Front. Pharmacol. 2024, 15, 1422740. [Google Scholar] [CrossRef]
Figure 1. Results of clinical trials using binary variables to describe the effects of SGLT2i against CI compared to non-use. S, SGLT2i; Dapa, dapagliflozin; Empa, empagliflozin; Cana, canagliflozin; (1) where the population was diagnosed as having dementia at baseline; (2) where the population was free of dementia. The blue line represents the 95% confidence interval, which is insignificant when 7intersecting the vertical dotted line [116,118,119,121,122,125,126,127]. The red box indicates the trial result and standard error size of the result. Same for the following Figure 2, Figure 3, Figure 4, Figure 5, Figure 6 and Figure 7.
Figure 1. Results of clinical trials using binary variables to describe the effects of SGLT2i against CI compared to non-use. S, SGLT2i; Dapa, dapagliflozin; Empa, empagliflozin; Cana, canagliflozin; (1) where the population was diagnosed as having dementia at baseline; (2) where the population was free of dementia. The blue line represents the 95% confidence interval, which is insignificant when 7intersecting the vertical dotted line [116,118,119,121,122,125,126,127]. The red box indicates the trial result and standard error size of the result. Same for the following Figure 2, Figure 3, Figure 4, Figure 5, Figure 6 and Figure 7.
Biomedicines 12 02783 g001
Figure 2. The improving effects of SGLT2i compared to non-use. Hena, henagliflozin [117,124].
Figure 2. The improving effects of SGLT2i compared to non-use. Hena, henagliflozin [117,124].
Biomedicines 12 02783 g002
Figure 3. The association between SGLT2i and adverse cerebral vascular events compared to non-use. Empa, empagliflozin; Dapa, dapagliflozin; Ertu, ertugliflozin. (M) where the HR describes the risk among male patients; (F) where the HR describe the risk among female patients [121,125,126,128,129,130,131,132,133].
Figure 3. The association between SGLT2i and adverse cerebral vascular events compared to non-use. Empa, empagliflozin; Dapa, dapagliflozin; Ertu, ertugliflozin. (M) where the HR describes the risk among male patients; (F) where the HR describe the risk among female patients [121,125,126,128,129,130,131,132,133].
Biomedicines 12 02783 g003
Figure 4. The results of continuous variables describing the protective effects of SGLT2i compared to non-use. Dapa, dapagliflozin; β, coefficient in multivariable regression analysis [123,134,135].
Figure 4. The results of continuous variables describing the protective effects of SGLT2i compared to non-use. Dapa, dapagliflozin; β, coefficient in multivariable regression analysis [123,134,135].
Biomedicines 12 02783 g004
Figure 5. Forest plot of meta-analyses which pool the results of SGLT2i against CI in T2DM compared to non-use. Non-cardiovascular mortality includes infectious diseases, cancers, lung diseases, dementia, renal diseases and other external causes [15,20,136,137,138,139,140].
Figure 5. Forest plot of meta-analyses which pool the results of SGLT2i against CI in T2DM compared to non-use. Non-cardiovascular mortality includes infectious diseases, cancers, lung diseases, dementia, renal diseases and other external causes [15,20,136,137,138,139,140].
Biomedicines 12 02783 g005
Figure 6. Network meta-analysis results of SGLT2i for lowering dementia risk compared to control or other interventions from Tian et al. [20].
Figure 6. Network meta-analysis results of SGLT2i for lowering dementia risk compared to control or other interventions from Tian et al. [20].
Biomedicines 12 02783 g006
Figure 7. Bayesian network meta-analysis results of SGLT2i for lowering dementia and AD risk compared to control or other interventions from Sunwoo et al. [19].
Figure 7. Bayesian network meta-analysis results of SGLT2i for lowering dementia and AD risk compared to control or other interventions from Sunwoo et al. [19].
Biomedicines 12 02783 g007
Figure 8. The hypothetical mechanism of the protective and treating effects when using SGLT2i. (A) Renal Glucose Reabsorption. SGLT2i inhibit renal glucose reabsorption to ameliorate glycemic overload, insulin resistance, metabolic disturbance, overweight and even hypertension, which is supposed to be the original power of neuroprotection. (B) Neurovascular protection. The metabolism benefits might manifest as anti-atherosclerosis, arteriosclerosis and CAA in the brain via anti-neuroinflammation, oxidative stress, microglial polarization, NVU remodeling and BBB leakage. (C) AChE inhibition and (D) NGF crosstalk. The possible extra effects of SGLT2i against CI by AChE inhibition, which is the therapeutic strategy of AD, and NGF crosstalk leading to neuronal survival and synaptic plasticity improvement in hippocampal. An upward arrow means increasing, improvement, or being up-regulated, which can be interpreted as any positive effect. A downward arrow indicates lightening, ameliorating or being down-regulated, which can be thought of as the antagonism of any disadvantage. Same for the Table 3 and Table 4.
Figure 8. The hypothetical mechanism of the protective and treating effects when using SGLT2i. (A) Renal Glucose Reabsorption. SGLT2i inhibit renal glucose reabsorption to ameliorate glycemic overload, insulin resistance, metabolic disturbance, overweight and even hypertension, which is supposed to be the original power of neuroprotection. (B) Neurovascular protection. The metabolism benefits might manifest as anti-atherosclerosis, arteriosclerosis and CAA in the brain via anti-neuroinflammation, oxidative stress, microglial polarization, NVU remodeling and BBB leakage. (C) AChE inhibition and (D) NGF crosstalk. The possible extra effects of SGLT2i against CI by AChE inhibition, which is the therapeutic strategy of AD, and NGF crosstalk leading to neuronal survival and synaptic plasticity improvement in hippocampal. An upward arrow means increasing, improvement, or being up-regulated, which can be interpreted as any positive effect. A downward arrow indicates lightening, ameliorating or being down-regulated, which can be thought of as the antagonism of any disadvantage. Same for the Table 3 and Table 4.
Biomedicines 12 02783 g008
Figure 9. The hypothetical molecular mechanism and involved signaling pathways of SGLT2i against CI. The arrow indicates promotion, and the horizontal arrow indicates inhibition.
Figure 9. The hypothetical molecular mechanism and involved signaling pathways of SGLT2i against CI. The arrow indicates promotion, and the horizontal arrow indicates inhibition.
Biomedicines 12 02783 g009
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

Zhang, Y.; Liao, X.; Xu, J.; Yin, J.; Li, S.; Li, M.; Shi, X.; Zhang, S.; Li, C.; Xu, W.; et al. The Promising Potency of Sodium–Glucose Cotransporter 2 Inhibitors in the Prevention of and as Treatment for Cognitive Impairment Among Type 2 Diabetes Patients. Biomedicines 2024, 12, 2783. https://doi.org/10.3390/biomedicines12122783

AMA Style

Zhang Y, Liao X, Xu J, Yin J, Li S, Li M, Shi X, Zhang S, Li C, Xu W, et al. The Promising Potency of Sodium–Glucose Cotransporter 2 Inhibitors in the Prevention of and as Treatment for Cognitive Impairment Among Type 2 Diabetes Patients. Biomedicines. 2024; 12(12):2783. https://doi.org/10.3390/biomedicines12122783

Chicago/Turabian Style

Zhang, Yibin, Xiaobin Liao, Jialu Xu, Jiaxin Yin, Shan Li, Mengni Li, Xiaoli Shi, Shujun Zhang, Chunyu Li, Weijie Xu, and et al. 2024. "The Promising Potency of Sodium–Glucose Cotransporter 2 Inhibitors in the Prevention of and as Treatment for Cognitive Impairment Among Type 2 Diabetes Patients" Biomedicines 12, no. 12: 2783. https://doi.org/10.3390/biomedicines12122783

APA Style

Zhang, Y., Liao, X., Xu, J., Yin, J., Li, S., Li, M., Shi, X., Zhang, S., Li, C., Xu, W., Yu, X., & Yang, Y. (2024). The Promising Potency of Sodium–Glucose Cotransporter 2 Inhibitors in the Prevention of and as Treatment for Cognitive Impairment Among Type 2 Diabetes Patients. Biomedicines, 12(12), 2783. https://doi.org/10.3390/biomedicines12122783

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