- 1Emergency Department, Jining No.1 People's Hospital, Jining, Shandong, China
- 2Institute of Emergency and Critical Care of Jining Medical Research Academy, Jining, Shandong, China
- 3Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- 4Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
1 Introduction
A pooled analysis of 1,108 population-representative studies published in 2024 noted that type 2 diabetes mellitus (T2DM) affects 828 million adults worldwide (1). Researchers recognize diabetes-associated cognitive dysfunction (DACD) as a critical comorbidity of T2DM, reflecting the intersection of metabolic dysfunction and neurodegeneration. Demographic trends for DACD very closely resemble those seen in diabetes mellitus (2). A systematic review and meta-analysis encompassing >25 original studies with millions of participants, estimates that the relative risk (RR) for all types of cognitive dysfunction is 1.73 (95% CI 1.65–1.82) for people with diabetes compared with people without diabetes (3). DACD shares pathological features overlapping with Alzheimer's disease (AD), including insulin resistance, chronic neuroinflammation, and amyloid-β accumulation (4). While existing therapies focus primarily on glycemic control, few interventions target the brain-specific consequences of T2DM, such as cognitive impairment. The Memory in Diabetes (MIND) sub-study of the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial is the largest intervention study of cognitive impairment in diabetes mellitus to date, found no benefit of intensive glycemic control on cognitive function (5). Therefore, “novel” interventions to address DACD are urgently needed.
Intermittent fasting (IF), a dietary regimen alternating periods of fasting and feeding, has emerged as a promising intervention to mitigate both metabolic and cognitive deficits in T2DM (6). IF—encompassing regimens like time-restricted feeding and 5:2 fasting (7)—induces metabolic switching from glucose to ketone metabolism, activating pathways that may counteract neurodegeneration (8, 9). The objective of this opinion article is to examine whether IF can improve T2DM-associated cognitive dysfunction by enhancing insulin sensitivity, reducing neuroinflammation, mitigating oxidative stress, and restoring gut microbiota homeostasis, and thoroughly analyzed the potential challenges associated with the clinical translation of IF.
2 Pathological links between T2DM and cognitive dysfunction
Evidence exists of a link between type 2 diabetes mellitus (T2DM), cognitive decline, and dementia (10). Given the complexity of the phenotype of T2DM and cognitive dysfunction, we will address the potential pathomechanistic links between the two in the following key areas.
2.1 Insulin resistance
All brain cell types express insulin receptors, with the highest densities localized to the olfactory bulb, hypothalamus, hippocampus, cerebral cortex, striatum, and cerebellum (11, 12). Emerging evidence indicates that insulin influences cerebral bioenergetics, enhances synaptic viability and dendritic spine formation, increases the turnover of neurotransmitters, and facilitates clearance of amyloid β peptide while modulating tau phosphorylation (13, 14). T2DM, the predominant form of diabetes mellitus, is generally characterized by chronic hyperglycemia, hyperinsulinemia, dyslipidemia, as well as lipotoxicity, which result in progressive deterioration of insulin secretion and insulin action (15–18). Insulin resistance (IR) is defined as the lack or decreased response of the target tissues to insulin (19, 20). Notably, evidence has shown that peripheral IR results in loss of brain function, which in turn is strongly associated with brain degeneration, cognitive dysfunction, depression, and AD (21–24). Similarly, brain insulin resistance (bIR) can be defined as the failure of brain cells to respond to insulin as they normally would, resulting in impairments in synaptic, metabolic, and immune response functions (25). Individuals with relatively diminished brain insulin sensitivity have a particularly high risk for an AD-like brain pattern (26). Indeed, preclinical and clinical findings support the hypothesis that bIR underlies the basic neuropathological mechanism of cognitive impairment in the aging-related, T2DM-associated, and neurodegenerative context (26). Through these multiple pathways, we hypothesize that insulin resistance could contribute to neurodegeneration, which in turn mediates and promotes the development of AD, vascular cognitive impairment, and other dementias. Notably, metabolites of the intestinal flora, such as bile acids (BAs), short-chain fatty acids (SCFAs) and amino acids (AAs) may influence to some extent the decreased insulin sensitivity associated with T2DM dysfunction and regulate metabolic as well as immune homeostasis (27).
2.2 Gut-brain axis dysregulation
The gut microbiome is known for playing a major role in human health as well as being increasingly recognized as being involved in the pathogenesis of metabolic diseases. Accumulating preclinical and clinical data over the past years has shown that alterations in the gut microbiota affect many organs involved in T2DM and the clinical onset of hyperglycemia (28, 29). Multi-omics (OMICs) studies have shown that single-dose streptozotocin (STZ) -induced hyperglycemia (HG) is sufficient to induce and exacerbate intestinal dysbiosis through modulation of the cecum metabolite pool by analyzing the taxonomic composition, transcriptional activity, and small molecule libraries of the cecum (30). A two-stage case-control metagenome-wide association study (MGWAS) based on deep next-generation shotgun sequencing showed that patients with T2DM were characterized by gut microbial dysbiosis, a decrease in the abundance of some universal butyrate-producing bacteria and an increase in various opportunistic pathogens, as well as an enrichment of other microbial functions conferring sulfate reduction and oxidative stress resistance (31). Intestinal dysbiosis promotes insulin resistance and inflammation, exacerbating diabetes; diabetes further worsens the intestinal dysbiosis, creating a mutually reinforcing mechanism. GM dysbiosis synergistically results in (i) a general increase in pro-inflammatory bacteria and a decrease in anti-inflammatory bacteria; (ii) impair intestinal tight junction integrity by increasing production of inflammatory metabolites and intestinal inflammation; and (iii) induced neuroinflammation, accelerated Aβ fibrillogenesis and parenchymal plaque burden (32). Several studies report that a variety of intestinal bacteria responsible for the production of lipopolysaccharides (LPS), a neurotoxin that disrupts paracellular barriers by cleaving intercellular proteins, such as E-cadherin in epithelial cells, leading to the “leaky gut” phenomenon (33, 34). LPS activates microglia and astrocytes, triggering neuroinflammation that promotes amyloid precursor protein accumulation, Aβ42 fibrillogenesis, plaque formation, and ultimately neuronal loss—a key pathway in neurodegeneration (35–37). Dysbiosis of gut microbiota promotes the harmful intestinal substances enter the systemic circulation through a compromised intestinal barrier, triggering systemic inflammation, which in turn destroys the blood–brain barrier and activates the TLR4/NF-κB signaling pathway in the brain, causing neuroinflammation (38, 39).
2.3 Oxidative stress and neuroinflammation
An increasing number of studies have indicated that increased oxidative stress is associated with neuronal damage and is a key factor contributing to the onset and progression of DCAD (40–42). ROS are normally produced as by-products of oxygen metabolism, but various factors can elevate its production. Among them, diabetes is a major cause of increased ROS generation by auto-oxidation of glucose, protein glycation, and through the polyol pathway (43). Oxidative stress has been shown to be a major causal factor compromising neuronal loss and synaptic disruption by impairing brain mitochondrial homeostasis, as seen in diabetic mice models, ultimately having deleterious effects on cognitive performance (44). In addition, a wide range of clinical studies have noted that oxidative stress is contribute significantly to the pathogenesis and progression of cognitive dysfunction (23, 45–47). On the other hand, oxidative stress has also been shown to be a facilitator of neuroinflammation, which is another primary contributor to the progression of cognitive decline in T2DM (48).
During T2DM, influenced by HG, microglial activation can exacerbate cytotoxicity and neuronal damage. Previous studies have revealed that in T2DM rat models, microglia activation in the brain is evident, resulting in overexpression of proinflammatory cytokines in the brains of these model rats, along with a marked decline in their learning and memory abilities (49). In contrast, treatment of T2DM model mice with drugs significantly suppressed the over-activation of microglia in the CNS, accompanied by a notable downregulation of pro-inflammatory cytokines and a significant amelioration of cognitive impairment symptom in the mice (50, 51). In the CNS, the functions of microglia are highly dynamic and can adopt different phenotypes based on the microenvironmental signals they receive. This ability to change phenotype, known as polarization, is a key characteristic of microglia, enabling them to adapt to various physiological and pathological conditions (52). However, they are generally classified into two main phenotypes: M1 and M2 (53). M1 phenotype microglia are typically considered pro-inflammatory, playing a key role in immune responses and inflammatory reactions. On the other hand, M2 phenotype microglia are mainly involved in neuroprotection and anti-inflammatory responses. In db/db diabetic mouse model, microglia are polarized into a pro-inflammatory M1 phenotype, along with low levels of a neuroprotective M2 phenotype, and significant cognitive impairment was observed through the Morris water maze test (54). The link between regulatory T (Treg) function and microglia polarization is well established in the brain (55), dipeptidyl peptidase-4 (DPP4)-mediated impairment of Tregs function polarize microglia toward a pro-inflammatory phenotype and subsequently lead to neuroinflammation and cognitive dysfunction in T2DM patients (54). In the T2DM mouse model, the pharmacological intervention inhibited the overactivated microglia and reversed the polarization of microglial phenotypes under T2DM conditions, shifting them from the proinflammatory M1 type to the anti-inflammatory M2 type, with concomitant improvement of cognitive impairment in T2DM mice (56, 57). IF, gut flora dysbiosis, neuroinflammation, and oxidative stress form a self-reinforcing network that underlies DACD.
3 Mechanisms of intermittent fasting in neuroprotection
IF is defined as a dietary pattern that restricts the time of eating, rather than the amount or composition of food, in the absence of malnutrition. Popular intermittent fasting diets involve daily time-restricted feeding or intermittent full-day fasting for 2 to 4 days per week. After an 8- to 12-h period of fasting, the liver starts to break down fatty acids to produce ketone bodies, which play a neuroprotective role by improving brain neuronal function, decreasing inflammatory expression and reactive oxygen species (ROS) production, activating brain-derived neurotrophic factor (BDNF) expression in neurons, and restore neuronal metabolism (58, 59). A clinical study in overweight adults suggests that IF increases BDNF levels and may have anti-aging effects (60). Moreover, studies suggest that IF-induced alterations in brain energy metabolism favor the modulation of microglial polarization from the M1 to the M2 phenotype and play an important role in degenerative diseases (61). Studies in diabetic mice have demonstrated that a 28-day IF treatment alleviated diabetes-induced cognitive dysfunction via a microbiota-metabolites-brain axis, benefiting from comprehensive investigations on diabetic mice behavior/synaptic structure, mitochondrial/energy metabolism-related signaling, and an integrated analysis of multi-OMICs (6). Clinical studies on the 5:2 intermittent fasting (IF) and the USDA healthy living (HL) diet have shown that both regimens are effective in improving peripheral IR, lipid metabolism, and cognition (62, 63). Emerging evidence suggests positive relationship between energy limitation, human health and cognition (7). A recent human intervention study showed that IF may influence memory function possibly through modulating adult hippocampal neurogenesis with the potential to be used as an intervention to prevent or boost cognitive decline (64). Ooi and colleagues found that a 3-year IF diet enhanced cognitive functioning in older adults with mild cognitive impairment compared to age-matched adults who irregularly practice IF and age-matched adults who do not practice IF (65). Moreover, in a randomized clinical trial conducted by Kapogiannis et al., both the 5:2 IF regimen and HL diet approaches were effective in reducing brain insulin resistance and improving memory and executive function in in patients with AD, and the improvements were more pronounced in the IF group (66). Interestingly, IF can also reduce the level of circulating insulin in the blood, thereby improving the sensitivity of insulin receptors and upregulating the insulin/IGF-1 signaling pathway, which ultimately enhances the absorption and utilization of glucose by neurons and ameliorates hypometabolism in neurodegenerative disorders (59, 67). In addition, other studies have shown that IF changes the structure of the gut microbiota, increases the abundance of anti-inflammatory bacterial strains, and decreases the level of proinflammatory endotoxins in the gut and serum (68); and leads to increased diversity of gut bacteria, and leads to an increase in the diversity of intestinal bacteria, as well as an enhancement of several antioxidant microbial metabolic pathways (69). Overall, it is strongly hypothesized that IF regimens may be effective in exerting neuroprotection through a variety of pathways, including reduction of insulin resistance, oxidative stress, immune-inflammatory responses, and modulation of intestinal flora dysbiosis, which ultimately ameliorates cerebral energy metabolism and the symptoms of neurocognitive dysfunction.
4 Conclusions and future directions
The prevalence of T2DM-associated cognitive dysfunction is increasing due to the extension of the human lifespan, and there is currently no cure. It is important to identify preventive interventions and treatment strategies to ameliorate the progression of neurodegenerative disorders. IF regimens may be effective in exerting neuroprotection through a variety of pathways, including reduction of insulin resistance, oxidative stress, immune-inflammatory responses, and modulation of intestinal flora dysbiosis, which in turn improves symptoms of neurocognitive disorders such as dementia. Pilot study shows neuroprotective effects of IF, but limited research on T2DM-related cognitive dysfunction. In addition, there are no reliable studies indicating the optimal duration of IF programs in T2DM-related research, nor have determined whether there is heterogeneity in IF strategies across individuals and their long-term safety. Considering that diabetic patients are subject to strict glycemic control, the administration of hypoglycemic drugs during IF may cause hypoglycemia and its more serious complications. In addition, specific biomarkers to monitor the efficacy of IF have also not been identified.
Given the existing research gaps in non-pharmacological interventions for DACD, we propose a phased investigation. Systematically translate basic science into generalizable interventions through repeated tests of efficacy and effectiveness. Stage I (Intervention Optimization) will focus on identifying the dose-response relationship of intermittent fasting (IF), including optimal intervention duration (e.g., 12-h vs. 16-h daily fasting) and frequency (e.g., alternate-day vs. 5:2 regimens). Mechanistic outcomes such as insulin sensitivity, inflammatory biomarkers, and feasibility metrics such as adherence rates and adverse events will be prioritized. Stage II (Efficacy Evaluation) will involve a multicenter, randomized, controlled, stratified pilot trial to assess the preliminary efficacy of the optimized IF protocol.
Author contributions
CC: Conceptualization, Writing – original draft, Writing – review & editing. DS: Writing – review & editing. YY: Writing – review & editing. XW: Writing – review & editing. RL: Writing – review & editing. SN: Writing – review & editing. WX: Conceptualization, Writing – review & editing.
Funding
The author(s) declare that no financial support was received for the research and/or publication of this article.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.
Generative AI statement
The author(s) declare that no Gen AI was used in the creation of this manuscript.
Publisher's note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Zhou B, Rayner AW, Gregg EW, Sheffer KE, Carrillo-Larco RM, Bennett JE, et al. Worldwide trends in diabetes prevalence and treatment from 1990 to 2022: a pooled analysis of 1108 population-representative studies with 141 million participants. Lancet. (2024) 404:2077–93. doi: 10.1016/S0140-6736(24)02317-1
2. Biessels GJ, Despa F. Cognitive decline and dementia in diabetes mellitus: mechanisms and clinical implications. Nat Rev Endocrinol. (2018) 14:591–604. doi: 10.1038/s41574-018-0048-7
3. Gudala K, Bansal D, Schifano F, Bhansali A. Diabetes mellitus and risk of dementia: a meta-analysis of prospective observational studies. J Diabetes Invest. (2013) 4:640–50. doi: 10.1111/jdi.12087
4. Minami Y, Sonoda N, Hayashida E, Makimura H, Ide M, Ikeda N, et al. P66shc signaling mediates diabetes-related cognitive decline. Sci Rep-UK. (2018) 8:3213. doi: 10.1038/s41598-018-21426-6
5. Launer LJ, Miller ME, Williamson JD, Lazar RM, Gerstein HC, Murray AM, et al. Effects of intensive glucose lowering on brain structure and function in people with type 2 diabetes (accord mind): a randomised open-label substudy. Lancet Neurol. (2011) 10:969–77. doi: 10.1016/S1474-4422(11)70188-0
6. Liu ZG, Dai XS, Zhang HB, Shi RJ, Hui Y, Jin X, et al. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat Commun. (2020) 11:855. doi: 10.1038/s41467-020-14676-4
7. Mattson MP, Longo VD, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Res Rev. (2017) 39:46–58. doi: 10.1016/j.arr.2016.10.005
8. de Cabo R, Mattson MP. Effects of intermittent fasting on health, aging, and disease. New Engl J Med. (2019) 381:2541–51. doi: 10.1056/NEJMra1905136
9. Zhao YH, Jia MZ, Chen WX, Liu ZG. The Neuroprotective effects of intermittent fasting on brain aging and neurodegenerative diseases via regulating mitochondrial function. Free Radical Bio Med. (2022) 182:206–18. doi: 10.1016/j.freeradbiomed.2022.02.021
10. Strachan MWJ, Reynolds RM, Marioni RE, Price JF. Cognitive function, dementia and type 2 diabetes mellitus in the elderly. Nat Rev Endocrinol. (2011) 7:108–14. doi: 10.1038/nrendo.2010.228
11. Selenius JS, Silveira PP, Haapanen MJ, von Bonsdorff M, Lahti J, Eriksson JG, et al. The brain insulin receptor gene network and associations with frailty index. Age Ageing (2024) 53:afae091. doi: 10.1093/ageing/afae091
12. Schulingkamp RJ, Pagano TC, Hung D, Raffa RB. Insulin receptors and insulin action in the brain: review and clinical implications. Neurosci Biobehav R. (2000) 24:855–72. doi: 10.1016/S0149-7634(00)00040-3
13. Tu X, Jain A, Bueno PP, Decker H, Liu XD, Yasuda R. Local autocrine plasticity signaling in single dendritic spines by insulin-like growth factors. Sci Adv. (2023) 9:adg0666. doi: 10.1126/sciadv.adg0666
14. Dewanjee S, Chakraborty P, Bhattacharya H, Chacko L, Singh B, Chaudhary A, et al. Altered glucose metabolism in Alzheimer's disease: role of mitochondrial dysfunction and oxidative stress. Free Radical Bio Med. (2022) 193:134–57. doi: 10.1016/j.freeradbiomed.2022.09.032
15. Matczuk J, Zalewska A, Lukaszuk B, Knas M, Maciejczyk M, Garbowska M, et al. Insulin resistance and obesity affect lipid profile in the salivary glands. J Diabetes Res. (2016) 2016:8163474. doi: 10.1155/2016/8163474
16. Bastien M, Poirier P, Lemieux I, Després JP. Overview of epidemiology and contribution of obesity to cardiovascular disease. Prog Cardiovasc Dis. (2014) 56:369–81. doi: 10.1016/j.pcad.2013.10.016
17. Taylor R. Insulin resistance and type 2 diabetes. Diabetes. (2012) 61:778–9. doi: 10.2337/db12-0073
18. Yazici D, Sezer H. Insulin resistance, obesity and lipotoxicity. Obesity Lipotoxicity. (2017) 960:277–304. doi: 10.1007/978-3-319-48382-5_12
19. Holloway GP, Han XX, Jain SS, Bonen A, Chabowski A. Chronic muscle stimulation improves insulin sensitivity while increasing subcellular lipid droplets and reducing selected diacylglycerol and ceramide species in obese zucker rats. Diabetologia. (2014) 57:832–40. doi: 10.1007/s00125-014-3169-0
20. Maciejczyk M, Matczuk J, Zendzian-Piotrowska M, Niklinska W, Fejfer K, Szarmach I, et al. Eight-week consumption of high-sucrose diet has a pro-oxidant effect and alters the function of the salivary glands of rats. Nutrients. (2018) 10:1530. doi: 10.3390/nu10101530
21. Pugazhenthi S, Qin LM, Reddy PH. Common neurodegenerative pathways in obesity, diabetes, and Alzheimer's disease. Bba-Mol Basis Dis. (2017) 1863:1037–45. doi: 10.1016/j.bbadis.2016.04.017
22. Frisardi V, Solfrizzi V, Seripa D, Capurso C, Santamato A, Sancarlo D, et al. Metabolic-cognitive syndrome: a cross-talk between metabolic syndrome and Alzheimer's disease. Ageing Res Rev. (2010) 9:399–417. doi: 10.1016/j.arr.2010.04.007
23. Butterfield DA, Di Domenico F, Barone E. Elevated risk of type 2 diabetes for development of Alzheimer disease: a key role for oxidative stress in brain. Bba-Mol Basis Dis. (2014) 1842:1693–706. doi: 10.1016/j.bbadis.2014.06.010
24. Tong M, de la Monte SM. Mechanisms of ceramide-mediated neurodegeneration. J Alzheimers Dis. (2009) 16:705–14. doi: 10.3233/JAD-2009-0983
25. Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang HY, Ahima RS, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol. (2018) 14:168–81. doi: 10.1038/nrneurol.2017.185
26. Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Häring HU. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. (2016) 96:1169–209. doi: 10.1152/physrev.00032.2015
27. Liu LL, Zhang JH, Cheng Y, Zhu M, Xiao ZF, Ruan GC, et al. Gut microbiota: a new target for t2dm prevention and treatment. Front Endocrinol. (2022) 13:958218. doi: 10.3389/fendo.2022.958218
28. Tilg H, Moschen AR. Microbiota and diabetes: an evolving relationship. Gut. (2014) 63:1513–21. doi: 10.1136/gutjnl-2014-306928
29. Byndloss M, Devkota S, Duca F, Niess JH, Nieuwdorp M, Orho-Melander M, et al. The gut microbiota and diabetes: research, translation, and clinical applications-2023 diabetes, diabetes care, and diabetologia expert forum. Diabetes Care. (2024) 47:1491–508. doi: 10.2337/dci24-0052
30. Wurster JI, Peterson RL, Brown CE, Penumutchu S, Guzior DV, Neugebauer K, et al. Streptozotocin-induced hyperglycemia alters the cecal metabolome and exacerbates antibiotic-induced dysbiosis. Cell Rep. (2021) 37:110113. doi: 10.1016/j.celrep.2021.110113
31. Qin JJ, Li YR, Cai ZM, Li SH, Zhu JF, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. (2012) 490:55–60. doi: 10.1038/nature11450
32. Qian XH, Liu XL, Chen G, Chen SD, Tang HD. Injection of amyloid-β to lateral ventricle induces gut microbiota dysbiosis in association with inhibition of cholinergic anti-inflammatory pathways in Alzheimer's disease. J Neuroinflamm. (2022) 19:236. doi: 10.1186/s12974-022-02599-4
33. Lukiw WJ. Gastrointestinal (Gi) tract microbiome-derived neurotoxins-potent neuro-inflammatory signals from the Gi tract via the systemic circulation into the brain. Front Cell Infect Microbiol. (2020) 10:22. doi: 10.3389/fcimb.2020.00022
34. Xu HM, Huang HL, Zhou YL, Zhao HL, Xu J, Shou DW, et al. Fecal microbiota transplantation: a new therapeutic attempt from the gut to the brain. Gastroent Res Pract. (2021) 2021:6699268. doi: 10.1155/2021/6699268
35. Batista CRA, Gomes GF, Candelario-Jalil E, Fiebich BL, de Oliveira ACP. Lipopolysaccharide-induced neuroinflammation as a bridge to understand neurodegeneration. Int J Mol Sci. (2019) 20:2293. doi: 10.3390/ijms20092293
36. Khan MS, Ikram M, Park JS, Park TJ, Kim MO. Gut microbiota, its role in induction of Alzheimer's disease pathology, and possible therapeutic interventions: special focus on anthocyanins. Cells-Basel. (2020) 9:853. doi: 10.3390/cells9040853
37. Shabbir U, Arshad MS, Sameen A, Oh DH. Crosstalk between gut and brain in Alzheimer's disease: the role of gut microbiota modulation strategies. Nutrients. (2021) 13:690. doi: 10.3390/nu13020690
38. Wang Q, Luo YQ, Chaudhuri KR, Reynolds R, Tan EK, Pettersson S. The role of gut dysbiosis in Parkinson's disease: mechanistic insights and therapeutic options. Brain. (2021) 144:2571–93. doi: 10.1093/brain/awab156
39. Liu JS, Huang HX, Yang Q, Zhao JX, Zhang H, Chen W, et al. Dietary Supplementation of N-3 lcpufas prevents salmonellosis in a murine model. J Agr Food Chem. (2020) 68:128–37. doi: 10.1021/acs.jafc.9b05899
40. Hoyos CM, Colagiuri S, Turner A, Ireland C, Naismith SL, Duffy SL. Brain oxidative stress and cognitive function in older adults with diabetes and pre-diabetes who are at risk for dementia. Diabetes Res Clin Pract. (2022) 184:109178. doi: 10.1016/j.diabres.2021.109178
41. Wang BN, Wu CB, Chen ZM, Zheng PP, Liu YQ, Xiong J, et al. Dl-3-N-butylphthalide ameliorates diabetes-associated cognitive decline by enhancing Pi3k/Akt signaling and suppressing oxidative stress. Acta Pharmacol Sin. (2021) 42:347–60. doi: 10.1038/s41401-020-00583-3
42. Zhang L, Ma QH, Zhou YL. Strawberry leaf extract treatment alleviates cognitive impairment by activating Nrf2/Ho-1 signaling in rats with streptozotocin-induced diabetes. Front Aging Neurosci. (2020) 12:201. doi: 10.3389/fnagi.2020.00201
43. Tomlinson DR, Gardiner NJ. Glucose neurotoxicity. Nat Rev Neurosci. (2008) 9:36–45. doi: 10.1038/nrn2294
44. Solanki I, Parihar P, Shetty R, Parihar MS. Synaptosomal and mitochondrial oxidative damage followed by behavioral impairments in streptozotocin induced diabetes mellitus: restoration by Malvastrum tricuspidatum. Cell Mol Biol. (2017) 63:94–101. doi: 10.14715/cmb/2017.63.7.16
45. Al-Rawaf HA, Alghadir AH, Gabr SA. Molecular changes in circulating micrornas' expression and oxidative stress in adults with mild cognitive impairment: a biochemical and molecular study. Clin Interv Aging. (2021) 16:57–70. doi: 10.2147/CIA.S285689
46. Suresh S, Begum RF, Singh SA, Chitra V. Anthocyanin as a therapeutic in Alzheimer's disease: a systematic review of preclinical evidences. Ageing Res Rev. (2022) 76:101595. doi: 10.1016/j.arr.2022.101595
47. Martins RN, Villemagnen V, Sohrabi HR, Chatterjee P, Shah TM, Verdile G, et al. Alzheimer's disease: a journey rom amyloid peptides and oxidative stress, to biomarker technologies and disease prevention strategies-gains from aibl and dian cohort studies. J Alzheimers Dis. (2018) 62:965–92. doi: 10.3233/JAD-171145
48. Pang XJ, Makinde EA, Eze FN, Olatunji OJ. Polyphenol rich extract counteracts cognitive deficits, neuropathy, neuroinflammation and oxidative stress in diabetic encephalopathic rats via P38 Mapk/Nrf2/Ho-1 pathways. Front Pharmacol. (2021) 12:737764. doi: 10.3389/fphar.2021.737764
49. Li DL, Liu LL, Li L, Li XG, Huang B, Zhou CQ, et al. Sevoflurane induces exaggerated and persistent cognitive decline in a type II diabetic rat model by aggregating hippocampal inflammation. Front Pharmacol. (2017) 8:886. doi: 10.3389/fphar.2017.00886
50. Cui YX, Yang MM, Wang YL, Ren JN, Lin P, Cui C, et al. Melatonin prevents diabetes-associated cognitive dysfunction from microglia-mediated neuroinflammation by activating autophagy via Tlr4/Akt/Mtor pathway. Faseb J. (2021) 35:e21485. doi: 10.1096/fj.202002247RR
51. Zhang YX, Yuan Y, Zhang JW, Zhao Y, Zhang YQ, Fu JL. Astragaloside IV supplementation attenuates cognitive impairment by inhibiting neuroinflammation and oxidative stress in type 2 diabetic mice. Front Aging Neurosci. (2022) 14:1004557. doi: 10.3389/fnagi.2022.1004557
52. Dräger NM, Sattler SM, Huang CTL, Teter OM, Leng K, Hashemi SH, et al. A crispri/a platform in human IPSC-derived microglia uncovers regulators of disease states. Nat Neurosci. (2022) 25:1149–62. doi: 10.1038/s41593-022-01131-4
53. Ritzel RM, Li Y, Jiao Y, Lei ZF, Doran SJ, He JY, et al. Brain injury accelerates the onset of a reversible age-related microglial phenotype associated with inflammatory neurodegeneration. Sci Adv. (2023) 9:eadd1101. doi: 10.1126/sciadv.add1101
54. Hui Y, Xu ZQ, Li JX, Kuang LY, Zhong YM, Tang YY, et al. 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. doi: 10.1016/j.metabol.2022.155340
55. Machhi J, Kevadiya BD, Muhammad IK, Herskovitz J, Olson KE, Mosley RL, et al. Harnessing regulatory T cell neuroprotective activities for treatment of neurodegenerative disorders. Mol Neurodegener. (2020) 15:32. doi: 10.1186/s13024-020-00375-7
56. Liu WY, Li K, Zheng ML, He L, Chen T. Genipin attenuates diabetic cognitive impairment by reducing lipid accumulation and promoting mitochondrial fusion via Fabp4/Mfn1 signaling in microglia. Antioxidants-Basel. (2023) 12:74. doi: 10.3390/antiox12010074
57. Sood A, Fernandes V, Preeti K, Khot M, Khatri DK, Singh SB. Fingolimod alleviates cognitive deficit in type 2 diabetes by promoting microglial M2 polarization via the Pstat3-Jmjd3 axis. Mol Neurobiol. (2023) 60:901–22. doi: 10.1007/s12035-022-03120-x
58. Bai L, Zhou Y, Zhang J, Ma JP. The role of a ketogenic diet in the treatment of dementia in type 2 diabetes mellitus. Nutrients. (2023) 15:1971. doi: 10.3390/nu15081971
59. Marosi K, Kim SW, Moehl K, Scheibye-Knudsen M, Cheng AW, Cutler R, et al. 3-hydroxybutyrate regulates energy metabolism and induces bdnf expression in cerebral cortical neurons. J Neurochem. (2016) 139:769–81. doi: 10.1111/jnc.13868
60. Jamshed H, Beyl RA, Della Manna DL, Yang ES, Ravussin E, Peterson CM. Early time-restricted feeding improves 24-hour glucose levels and affects markers of the circadian clock, aging, and autophagy in humans. Nutrients. (2019) 11:1234. doi: 10.3390/nu11061234
61. Lv RJ, Liu B, Jiang ZY, Zhou RF, Liu XX, Lu TS, et al. Intermittent fasting and neurodegenerative diseases: molecular mechanisms and therapeutic potential. Metabolism. (2025) 164:156104. doi: 10.1016/j.metabol.2024.156104
62. Mozaffari H, Jalilpiran Y, Suitor K, Bellissimo N, Azadbakht L. Associations between empirically derived dietary patterns and cardiovascular risk factors among older adult men a cross-sectional study. Int J Vitam Nutr Res. (2023) 93:308–18. doi: 10.1024/0300-9831/a000725
63. Yuan XJ, Wang JP, Yang S, Gao M, Cao LX, Li XM, et al. Effect of intermittent fasting diet on glucose and lipid metabolism and insulin resistance in patients with impaired glucose and lipid metabolism: a systematic review and meta-analysis. Int J Endocrinol. (2022) 2022:6999907. doi: 10.1155/2022/6999907
64. Kim C, Pinto AM, Bordoli C, Buckner LP, Kaplan PC, del Arenal IM, et al. Energy restriction enhances adult hippocampal neurogenesis-associated memory after four weeks in an adult human population with central obesity: a randomized controlled trial. Nutrients. (2020) 12:638. doi: 10.3390/nu12030638
65. Ooi TC, Meramat A, Rajab NF, Shahar S, Ismail IS, Azam AA, et al. Intermittent fasting enhanced the cognitive function in older adults with mild cognitive impairment by inducing biochemical and metabolic changes: a 3-year progressive study. Nutrients. (2020) 12:2644. doi: 10.3390/nu12092644
66. Kapogiannis D, Manolopoulos A, Mullins R, Avgerinos K, Delgado-Peraza F, Mustapic M, et al. Brain responses to intermittent fasting and the healthy living diet in older adults. Cell Metab. (2024) 36:1900–4. doi: 10.1016/j.cmet.2024.07.012
67. Rahmani J, Varkaneh HK, Clark C, Zand H, Bawadi H, Ryan PM, et al. The influence of fasting and energy restricting diets on IGF-1. Levels in humans: a systematic review and meta-analysis. Ageing Res Rev. (2019) 53:100910. doi: 10.1016/j.arr.2019.100910
68. Zhang CH, Li SF, Yang L, Huang P, Li WJ, Wang SY, et al. Structural modulation of gut microbiota in life-long calorie-restricted mice. Nat Commun. (2013) 4:2163. doi: 10.1038/ncomms3163
Keywords: intermittent fasting, type 2 diabetes mellitus, diabetes-related cognitive dysfunction, insulin resistance, neuroinflammation, gut-brain axis dysregulation, oxidative stress
Citation: Cui C, Song D, Yang Y, Wang X, Lv R, Nie S and Xu W (2025) The role of intermittent fasting in the treatment of cognitive dysfunction in type 2 diabetes mellitus. Front. Nutr. 12:1603165. doi: 10.3389/fnut.2025.1603165
Received: 31 March 2025; Accepted: 18 April 2025;
Published: 09 May 2025.
Edited by:
Haoqiang Zhang, University of Science and Technology of China, ChinaReviewed by:
Xiaofei Hu, Army Medical University, ChinaQianhui Wang, Xinjiang Medical University, China
Copyright © 2025 Cui, Song, Yang, Wang, Lv, Nie and Xu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Wenwen Xu, eHV3ZW53ZW4zMjQ3QDE2My5jb20=