Open Access

Alzheimer's disease, a metabolic disorder: Clinical advances and basic model studies (Review)

  • Authors:
    • Shanhu Zhou
    • Limin Tu
    • Wei Chen
    • Gangli Yan
    • Hongmei Guo
    • Xinhua Wang
    • Qian Hu
    • Huiqing Liu
    • Fengguang Li
  • View Affiliations

  • Published online on: December 14, 2023     https://doi.org/10.3892/etm.2023.12351
  • Article Number: 63
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Alzheimer's disease (AD) is a type of neurodegenerative disease characterized by cognitive impairment that is aggravated with age. The pathological manifestations include extracellular amyloid deposition, intracellular neurofibrillary tangles and loss of neurons. As the world population ages, the incidence of AD continues to increase, not only posing a significant threat to the well‑being and health of individuals but also bringing a heavy burden to the social economy. There is epidemiological evidence suggesting a link between AD and metabolic diseases, which share pathological similarities. This potential link would deserve further consideration; however, the pathogenesis and therapeutic efficacy of AD remain to be further explored. The complex pathogenesis and pathological changes of AD pose a great challenge to the choice of experimental animal models. To understand the role of metabolic diseases in the development of AD and the potential use of drugs for metabolic diseases, the present article reviews the research progress of the comorbidity of AD with diabetes, obesity and hypercholesterolemia, and summarizes the different roles of animal models in the study of AD to provide references for researchers.

1. Introduction

Population aging is becoming a difficult problem faced by all countries worldwide, with the gradual improvement of living standards, life expectancy increases, which implies an increase in age-related diseases (1,2). Alzheimer's disease (AD) is one of the biggest obstacles to coping with a healthy aging population. AD is defined by the World Health Organization (WHO) as a neurodegenerative disease of unknown etiology, characterized by progressive deterioration of memory and cognitive function, accounting for 50-75% of all dementia cases (3,4). AD may present with clinical symptoms such as progressive memory loss, impaired executive function, difficulty in daily activities, altered thought and behavior patterns and impaired language function (5). A total of two clinical manifestations of AD are mainly recognized by the academic community: Senile plaques composed of β amyloid (Aβ) and neurofibrillary tangles composed of tau proteins with hyperphosphorylation (6). This series of processes slowly deprives the patient of memory and cognitive ability, and the patient gradually forgets recent events. The patient is unable to analyze, think and judge the events, and finds it difficult to deal with complex problems. Patients are unable to take care of themselves in daily life, making them and their family helpless over time (7,8). Although the exact process by which AD molecular cascades are triggered remains unclear, a series of epidemiological studies suggest that comorbid risk factors for metabolic disease are crucial in the pathogenesis of this disease (9-11). This suggests that physicians also associate metabolic disease with AD.

In the early nineties, some investigators noticed common mechanistic features between metabolic diseases and AD, and proposed the concept of type 3 diabetes (12-14). Researchers focused on close links between diabetes mellitus (DM) and AD, such as insulin, insulin-like growth factor, oxidative stress, glycogen synthase kinase 3β, Aβ and tau hyperphosphorylation (15-17). Since then, several studies have been carried out worldwide to explore possible links between metabolic diseases and AD, and to turn attention to AD as a type 3 diabetes; therefore, new therapeutic options for AD are explored from the perspective of metabolic disease (13,18,19). In light of recent research, it has become increasingly apparent that AD and various metabolic diseases exhibit numerous common characteristics.

However, the mechanism by which metabolic diseases affect the progression of AD remains unclear and the selection of therapeutic drugs and animal models for AD remains to be further discussed. Moreover, the relevant literature has not been fully reviewed at present. In the present study, using ‘Alzheimer's disease’, ‘metabolic disease’, ‘obesity’, ‘hypercholesterolemia’ and ‘animal model’ as keywords, four electronic databases such as Springer (https://link.springer.com/), PUBMED (https://pubmed.ncbi.nlm.nih.gov/), ScienceDirect (https://www.sciencedirect.com/search) and Wiley (https://onlinelibrary.wiley.com/), were searched for relevant literature. The present article systematically discusses the research progress of metabolic diseases and the pathogenesis of AD, and summarizes the different roles of animal models in AD research, to provide a reference for researchers (for the use of acronyms see Table I).

Table I

Abbreviation table.

Table I

Abbreviation table.

Full nameAbbreviation
Fludeoxyglucose18F
Alzheimer's diseaseAD
Apolipoprotein EAPOE
Amyloid precursor proteinAPP
β amyloid
Blood-brain barrierBBB
Body mass indexBMI
Protein kinase A systemcAMP/MPK
Diabetes mellitusDM
Endogenous melatonin reductionEMR
18F-fludeoxyglucoseFDG
HypercholesterolemiaFH
HypercholesterolemiaHC
Insulin-degrading enzymeIDE
Insulin receptorINSR
Low-density lipoproteinLDL
Mild cognitive impairmentMCI
m.p Eparviflora leaf hydroalcoholic extractMpHE
Magnetic resonance spectroscopyMRS
Non-alcoholic steatosisNAFLD
Neurofibrillary tangleNFT
Positron emission tomographyPET
Soluble APP βsAPP β
Subcutaneous adipose tissueSAT
Sodium-glucose CO-TRANSPORTER-2 inhibitorsSGLT2is
Thy1-C/EBP β transgenic miceTG mice
Visceral adipose tissueVAT

2. Metabolic diseases and AD

Diabetes and AD

DM is the most common metabolic disorder and the direct cause of its occurrence is usually due to defective insulin action or insufficient insulin secretion (20,21). Several real-world clinical cases suggest that brain-related mild cognitive impairment complications in diabetes may lead to cognitive deficits, which gradually develops into AD (22,23).

To date, several groups have focused on exploring and explaining the link between DM and AD. Based on anatomy, some parameters showed that the Alzheimer-like pathology of diabetic rats is increased; these parameters include increased levels of Aβ plaques in the hippocampus and frontal cortex, reduced hippocampal volume, reduced protein levels in the cerebral cortex and reduced dendritic spine density in diabetic animals (24,25). Similarly, clinical evidence suggests that amygdala and hippocampal volumes in patients with diabetes are altered compared with normal patients, with a trend toward decline (26).

From a pathological perspective, the cleavage of amyloid precursor protein (APP) and the formation of Aβ plaque requires the involvement of β-secretase, which also regulates the cleavage of insulin receptor, this strengthens the link between AD and diabetes mellitus (27,28). Furthermore, soluble (s)APPβ, a product of β-secretase, is a major determinant of insulin resistance (29). Glycotoxicity can lead to structural and functional damage of brain cells and nerves, cerebral vascular hemorrhage and increased β-amyloid protein accumulation (30). These are potential mechanisms of diabetes-related dementia.

From a molecular mechanistic perspective, it has been suggested that the protein kinase A system (cAMP/PKA) signaling pathway and insulin-degrading enzyme may contribute to the type 2 diabetes-accelerated AD pathological process by causing Aβ accumulation and neuronal apoptosis (31). In addition, studies focused on protein phosphorylation have demonstrated that overexpression of protein kinase Cα (PKCα) is associated with insulin signaling interfering with insulin receptor substrate (IRS)-1 and Akt phosphorylation in skeletal muscles (32-34). PKCα inhibits insulin signaling through the IRS-Akt pathway, and inhibition and silencing of PKC-α enhances insulin sensitivity by increasing GLUT-4 translocation to the plasma membrane and glucose uptake (32). The aforementioned results demonstrate the role of PKCα in regulating neuronal insulin resistance and diabetes and open new avenues for the treatment of metabolic disorders and neurodegeneration (34). P38γ signal transduction is characterized by its unique reciprocal regulation of the phosphatase protein tyrosine phosphatase H1 antibody, and by its direct binding to promoter DNA, which is also involved in the pathogenesis of diabetes and AD, suggesting its potential as a therapeutic target (35).

Several prospective trials have used sodium-glucose co-transporter-2 (SGLT2) inhibitors (is) as an anti-diabetic drug (36,37). Inhibition of SGLT2, which accounts for ~90% of glucose reabsorption, leads to a significant reduction in blood glucose levels (36). The activation of insulin signaling associated with neuronal survival, in particular the canonical pathway of Nev (pIR, pY-IRS-1, Pakt), has been demonstrated (36). In addition, brain magnetic resonance spectroscopy has been used to detect decreased concentrations of the excitatory neurotransmitter glutamate and its precursor glutamine after administration, given that glutamate excitotoxicity has been consistently associated with AD pathology (37). These findings may inspire the reuse of anti-diabetic drugs (such as SGLT2is) in AD and other related diseases characterized by downregulation of IGF-1/insulin signaling and excitotoxicity in neurons (37). Thus, several studies conducted in this direction have shown a link between diabetes and AD (36,37), and more links between these two diseases remain to be explored.

Obesity and AD

Obesity refers to a state of being overweight or obese, often caused by excessive accumulation of fat in the body, and is closely associated with cognitive impairment and AD (38-41). Body mass index (BMI) is the most common measure of obesity worldwide. BMI is calculated as weight (kg) divided by height (m2) squared. Obesity is defined as BMI ≥30 kg/m2 (42,43). However, BMI does not represent regional fat distribution, which varies by sex, age, ethnicity and residential area (44). Regional fat distribution may have different effects on cognitive decline and AD-related brain changes (45). However, different regions of the fat pool may have different cognitive outcomes and have different effects on the brain.

To date, several groups have focused on exploring and explaining the link between obesity and AD. The authors classify fat as visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT). Hepatic fat in VAT and non-alcoholic steatosis (NAFLD) is the most studied regional fat (46). Researcher has used MRI and functional MRI for structural brain measurements to assess the strong association between obesity and brain changes in different regions (47). A study has demonstrated that increased VAT is associated with decreased grey matter density and cognitive function and that such a relationship is age-dependent (48). For patients with NAFLD, hepatic fat deposits are significantly associated with smaller overall brain volumes as well as smaller cingulate and hippocampal volumes (49). Even after weight loss, NAFLD is still associated with smaller total brain volume (50). Structural measures indicate that higher VAT and SAT are associated with smaller total brain volumes (51). Elevated VAT is associated with cortical thinning, particularly with decreased hippocampal volume (52). The aforementioned study showed that higher VAT is associated with higher brain network damage in cognitive decline, suggesting a strong link between VAT and accelerated brain ageing.

From a pathological point of view, existing experimental results compare individuals with higher VAT metabolism (higher metabolic capacity of visceral adipose tissue) with individuals with lower VAT metabolism (lower metabolic capacity of visceral adipose tissue) (53,54). Individuals with higher VAT metabolism have been found to exhibit higher brain Aβ levels, suggesting a close relationship between VAT dysfunction and AD disease development (53). In addition, another study using brain 18F-fludeoxyglucose positron emission tomography (PET) as a neurodegenerative biomarker of AD yielded the same results (54).

Analyzed from a possible molecular mechanism standpoint, potential factors related to brain changes and cognition may be explained by the release of different secretory factors from different fat deposits (55,56). These different fat deposits release different secreted factors that can cross the blood-brain barrier (BBB) and cause damage, increase cognitive impairment and accelerate AD progression (55). Pro-inflammatory factors secreted by adipocytes, such as leptin, IL-6 and TNF-α, can cross the BBB and lead to neuroinflammation, thus playing a role in cognitive impairment and AD (56). Another study showed that a high-fat diet stimulates diabetes and insulin resistance in Thy1-C/EBPβ transgenic (TG) mice, with significant Aβ accumulation and hyperphosphorylation of Tau protein in the brain, triggering cognitive impairment (57). A study investigated the anti-inflammatory effects of M. parviflora leaf hydroalcoholic extract (MpHE) on obese mice with AD, showing that MpHE effectively reduces astrocyte proliferation, the presence of insoluble Aβ peptides in the hippocampus and spatial learning impairment in lean and obese 5XFAD mice (57). Furthermore, a study investigated the association between AD and obesity from the perspective of the gut microbiota. Endogenous melatonin reduction can cause systemic changes mediated by dysbiosis of the gut microbiota, which may be one of the causative factors of AD and obesity (58). Thus, several studies in this direction demonstrate a link between obesity and AD, and more links between these two diseases remain to be explored.

Hypercholesterolemia and AD

Familial hypercholesterolemia is a particularly severe type of hyperlipidemia. The clinical features were hypercholesterolemia, characteristic xanthoma and family history of early-onset cardiovascular disease (59). Patients have abnormally high levels of low-density lipoprotein (LDL) cholesterol, which is 4-6 times higher in patients with homozygous LDL cholesterol compared with in normal individuals (60). Animal studies showed that diet-induced hypercholesterolemia increases the accumulation of Aβ and accelerates the pathological process of AD (61).

To date, several research groups have focused on exploring and explaining the association between hypercholesterolemia and AD. The researchers used fludeoxyglucose (18F) PET to study different populations, revealing common anatomical structures between individuals at risk for hypercholesterolemia and AD (62-65). The analysis showed that higher serum total cholesterol levels are associated with lower bilateral CMRgl in areas of the anterior cuneiform, parietal-temporal and prefrontal lobes previously found to be preferentially affected by AD, as well as other frontal regions previously found to be preferentially affected by normal aging (65). In certain brain regions affected by AD, the association is greater in apolipoprotein E (APOE)-4 carriers compared with in non-carriers (66). A study showed that higher serum total cholesterol levels in middle age would accelerate brain processes associated with normal aging and act in concert with other risk factors for AD predisposition (67).

The APOE gene is the strongest genetic risk factor for AD, accounting for 60-80% of all dementia cases (68,69). APOE plays an important role in lipid transport and metabolism, accounting for ~7% of phenotypic variation in serum total cholesterol and 14% of polygenic variation (68). APOE also contributes ~1-8.3% of phenotypic variation and 16% of genetic variation in LDL cholesterol (70). Compared with non-carriers, APOE4 carriers tend to have higher total and LDL cholesterol as well as lower HDL cholesterol levels (71). Furthermore, higher levels of total and LDL cholesterol are associated with greater deposition of neuropathological markers of AD in the cerebrum (72). Given that lipids in APOE4 carriers are most sensitive to diet, these findings suggested that lipid management through dietary adjustment can reduce AD risk (73). Menopause itself is associated with a more adverse lipid profile compared with premenopause, especially for APOE3 and APOE4 carriers. Furthermore, APOE4-related AD risk is stronger in females compared with in males (74).

A reasonable mechanism by which diet and lipids may contribute to dementia pathology is by altering levels of oxymethanol, the oxidized product of cholesterol (75). Dyslipidemia and dietary cholesterol intake may result in unbalanced oxidant levels, which appear to result in an unbalanced oxidative type of reduction (75). Studies in mice further support the possibility of reducing AD risk through dietary adjustment. For example, mice fed with a high-cholesterol diet subsequently have higher levels of total cholesterol in plasma and Aβ protein in the brain compared with controls (76). Similarly, a high-fat diet results in greater Aβ deposition and impaired neuroinflammation, sensorimotor function and social interaction, as well as a tendency for APP/PS1 mice to have poorer short-term memory compared with mice fed a control diet, but this trend was not significant (77). Thus, several studies conducted in this direction showed a link between hypercholesterolemia and AD, and more links between these two diseases remain to be explored.

3. Application of animal models in AD

To find effective therapeutic measures, researchers construct different animal models based on pathogenesis, but different animal models of AD have different advantages and disadvantages (76,77).

Human beings and animals have great similarities in physiology and pathology. It is a common research method to simulate disease in animals to explore its biological mechanism. In the research history of AD, common AD model-making animals include Caenorhabditis elegans, Drosophila melanogaster, zebrafish, mice, rats, dogs, rhesus monkeys and chimpanzees, among others (78,79).

These experimental animals differ in species and conditions, each with different strengths and weaknesses. For Caenorhabditis elegans, Drosophila melanogaster and zebrafish, their small size and short life span make them convenient for researchers to reproduce and manipulate, but their brain structures differ considerably from those of humans and lack high-level cognitive behavior (80,81). For rhesus monkeys and chimpanzees, these mammalian primates have the most human-like brain structures and are ideal for receiving sensory tasks that mimic cognitive impairment. At the same time, rhesus macaques and chimpanzees as animal models are expensive and their use requires careful ethical consideration (82,83). Canines are also ideal animal models, which can exhibit age-related cognitive impairment similarly to humans, but canines do not present with neural plaques and tangles (84). Mice and rats are the most economical choice in most laboratories. They have similar mammalian physiology, similar brain structure to humans and lower feeding costs. However, their selection is not perfect, and they must face the disadvantages of a long breeding cycle and high time cost (85,86). Nevertheless, mice and rats are preferred animal models in most brain science laboratories (for a classification of artificial intervention AD animal models see Table II) (87-95).

Table II

Classification table of artificial intervention AD animal models.

Table II

Classification table of artificial intervention AD animal models.

ModelAnimalChemical substances/physical methodsMethod of operationInjury siteAdvantagesDisadvantages(Refs.)
Cholinergic injury modelWistar rat/SD ratPhysical damageCholinergic injuryHippocampal fimbriaSimulate cholinergic system damage, spatial orientation and memory impairmentNo Aβ and tau pathology(87)
Common carotid artery ligation modelSD rat/C57BL/6JmicePhysical ligationCommon carotid artery ligationCarotid arteryChronic cerebral ischemia, cognitive, impairmentNo Aβ and tau pathology(88)
Aβ injection modelBALB/c miceAβ1-42410 pmol, 3 µl brain localization injection, injection time 1 min, needle retention 3 minLateral ventricle (0.5 mm behind bregma point, 1.0 mm lateral to midline, 2.5 mm in depth)--(89)
 SD ratsAB1-401 g/l,1 µl, brain localization injection, injection time 5 min, needle retention 5 minThe dorsal side of the hippocampal dentate gyrus (3.3 mm posterior to the bregma, 2.0 mm lateral to the right, 3.0 mm below the dura mater, and the incisor hook plane is 2.4 mm below the interaural line)AB deposition, inflammation, learning and memory impairmentDoes not meet the characteristics of the progressive onset of AD, AB accumulates as the injection site 
 SD ratsAB25-3510 µg/µl, brain localization injection, 1 µl each on the left and right, after 5 min injection, keep the needle for 5 minCA1 area of the hippocampus on both sides (the bregma is the zero point, the puncture point is 3.5 mm behind the bregma, 2 mm on the right side of the midline, and the needle is vertically inserted 3 mm from the brain surface with a micro syringe)-- 
IBO infusion modelSD ratsIBO5 µg/µl, 1 µlMeynert basal nucleus (1.0 mm behind bregma, 3.0 mm next to midline, 7.3 mm deep)Aβ deposition and tau protein increase, and memory impairmentNo neurofibrillary tangles(90)
Streptozotocin infusion modelLong Evans ratsSTZ40 mg/kg, injection time 3 minIn both sides of the brain (1.0 mm behind the bregma, 1.0 mm lateral to the right side of the midline, 2.5 mm below the skull)Aβ deposition, tau protein hyperphosphorylation, cholinergic loss, oxidative stressNo neurofibrillary tangles and senile plaques(91)
D-gal infusion modelSwiss albino miceD-gal150 mg/kg, once a day, continue injection for 42 daysSubcutaneous injection/intraperitoneal injectionTissue oxidative stress and inflammation, cognitive and cholinergic system disorders, tau protein hyperphosphorylationNo Aβ, neurofibrillary tangles and senile plaques(92)
Aluminum trichloride infusion modelWistar ratsAluminum trichloride100 mg/kg, continuous injection for 60 dayIntraperitoneal injectionAβ aggregation, neuronal degeneration, learning and memory impairmentModeling time is long, central cholinergic is not reduced, NFTs are different from patients with AD(93)
OKA infusion modelSD ratsOKA40 ng/µl, 5 µl, injection time 5 min, needle retention 5 minLateral ventricle (0.8 mm posterior to the bregma, 1.5 mm lateral to the midline, 3.6 mm vertical needle insertion)Shows Tau protein hyperphosphorylation and Aβ pathological manifestationsNo neurofibrillary tangles(94)
SCOP infusion modelWistar ratsSCOP0.2 ml/150 g, continuous injection for 14 dayAbdominal cavitySpace and memory impairmentNo typical pathological features of Aβ, neurofibrillary(95)

[i] AD, Alzheimer's disease; IBO, ibotenic acid; STZ, streptozotocin; D-gal, D-galactose; NFTs, neurofibrillary tangles; SCOP, scopolamine; OKA, okadaic acid.

Injection of streptozotocin (STZ) into the lateral ventricles of animals disrupts brain energy metabolism, and it is a common method to model AD in animals with corresponding Aβ deposition, hyperphosphorylation of tau protein, abnormal cholinergic function and oxidative stress (96-98). It is worth mentioning that STZ is also the primary modeling drug for diabetes. This underscores the potential co-morbid mechanisms of metabolic disease with AD in another way.

4. Discussion

The WHO estimates that the proportion of the population of the world aged >60 years old will rise to 22% by 2050 (97,98). Emerging evidence now indicates an increasing trend in patients with AD and related age-related diseases (97). For example, in the United States, the number of patients aged ≥65 years with AD and related dementia is increasing and is expected to reach 13.9 million by 2060(98). These epidemiological studies suggest that the decline in quality of life in older adults and the increased risk of AD and related aging-related diseases pose a serious threat to global health (Fig. 1).

As the relationship between metabolic diseases and AD deepens, it is necessary to understand the reasonable relationship between the two. The present review discusses the new role of diabetes, obesity and hypercholesterolemia in AD. Changes in the hippocampus and frontal cortex have been found in both patients with metabolic disease and those with AD, using a variety of diagnostic instruments or anatomical studies. Most of these changes occur in the volume of different brain regions and the level of cortical proteins. This series of changes points to commonalities between metabolic diseases and cognitive changes in brain injury (13,19,20,41,55-57). Another study found that long-term high-sugar and high-fat diets can induce metabolic syndrome in experimental animals, and their brain tissue can exhibit typical characteristic changes of AD (99). Excessive lipid deposition in brain tissue can induce chronic inflammation, which plays an important role in the onset of AD. Exploring safe and effective intervention measures is currently one of the urgent issues that need to be addressed in the interdisciplinary treatment of metabolic syndrome. Excessive nutrition can cause changes in the hypothalamic immune system, leading to a hypothalamic inflammatory response, The activation of pro-inflammatory factors and other pro-inflammatory molecules persists in the pathological processes associated with metabolic syndrome, indicating the importance of improving obesity and other metabolic syndromes in the treatment of AD (100,101).

From a pathological perspective, the amyloid hypothesis has long been the dominant theory asserting that AD is caused by the accumulation of Aβ protein in the brain, leading to neuronal toxicity in the central nervous system (58). Metabolic diseases also happen to influence the pathogenesis of AD from different perspectives. These metabolic diseases are involved either through a process of Aβ plaque formation or increased Aβ accumulation (21-24,67).

In addition, the aforementioned metabolic diseases also affect the course of AD through some potential mechanisms. The cAMP/PKA signaling pathway, the IRS-Akt pathway, neuronal apoptosis, neuroinflammation and oxidative stress are all factors that have been verified by several research groups; they become common ground between metabolic diseases and cognitive impairment (25-28,46-49,65-67). These commonalities lead the present authors to focus on the potential of drugs commonly used to treat metabolic diseases for the treatment of AD. There are rich pathophysiological links between AD and diabetes. It is not difficult to imagine that some anti-diabetic drugs may be used to treat AD. Among them, insulin is the most prominent example. Aβ senile plaque formation and tau protein hyperphosphorylation are the main histopathological manifestations of AD, and insulin signaling and insulin resistance play important regulatory roles (102). Research shows that insulin can protect the brains of rats from Aβ formation, thereby having beneficial effects on them (103). Another example is metformin. The effect of metformin on insulin is achieved through AMP-activated protein kinase (104). In previous experiments, metformin has been shown to reduce tau phosphorylation and prevent pathological changes in AD neurons (105).

Animal models play an important role in the study of the pathogenesis and potential treatment of AD. The present review summarizes the common animal models and their advantages and disadvantages to provide a reference for researchers. Although several groups studied the subtle link between metabolic disease and AD, several questions remain to be answered. A common question is about the causal relationship that exists between metabolic disease and cognitive impairment. The present study has a clear understanding that these two diseases usually occur together, but the sequence and causality between them are not yet supported by strong experimental results. In addition, the criteria for characterizing AD and metabolic disease in different animal models are not consistent, which makes it difficult to map to clinical patients, which requires more data.

Acknowledgements

Not applicable.

Funding

Funding: This work was supported by the General Project of the Natural Science Foundation of Hubei Province (grant no. 2021CFB585), the General Project of Health Commission of Hubei Province (grant no. WJ2021M030).

Availability of data and materials

Not applicable.

Author contributions

FL contributed to the design of the review. HL and SZ prepared the manuscript. LT, WC, GY, HG, XW and QH made substantial contributions to conception and design. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kovács Z, Brunner B and Ari C: Beneficial effects of exogenous ketogenic supplements on aging processes and age-related neurodegenerative diseases. Nutrients. 13(2197)2021.PubMed/NCBI View Article : Google Scholar

2 

Jensen L, Monnat SM, Green JJ, Hunter LM and Sliwinski MJ: Rural population health and aging: Toward a multilevel and multidimensional research agenda for the 2020s. Am J Public Health. 110:1328–1331. 2020.PubMed/NCBI View Article : Google Scholar

3 

Li Y, Xu H, Wang H, Yang K, Luan J and Wang S: TREM2: Potential therapeutic targeting of microglia for Alzheimer's disease. Biomed Pharmacother. 165(115218)2023.PubMed/NCBI View Article : Google Scholar

4 

Maria C and Rauter AP: Nucleoside analogues: N-glycosylation methodologies, synthesis of antiviral and antitumor drugs and potential against drug-resistant bacteria and Alzheimer's disease. Carbohydr Res. 532(108889)2023.PubMed/NCBI View Article : Google Scholar

5 

Stocker H, Trares K, Beyer L, Perna L, Rujescu D, Holleczek B, Beyreuther K, Gerwert K, Schöttker B and Brenner H: Alzheimer's polygenic risk scores, APOE, Alzheimer's disease risk, and dementia-related blood biomarker levels in a population-based cohort study followed over 17 years. Alzheimers Res Ther. 15(129)2023.PubMed/NCBI View Article : Google Scholar

6 

Weller J and Budson A: Current understanding of Alzheimer's disease diagnosis and treatment. F1000Res 7: F1000 Faculty Rev-1161, 2018.

7 

Ismail Z, Leon R, Creese B, Ballard C, Robert P and Smith EE: Optimizing detection of Alzheimer's disease in mild cognitive impairment: A 4-year biomarker study of mild behavioral impairment in ADNI and MEMENTO. Mol Neurodegener. 18(50)2023.PubMed/NCBI View Article : Google Scholar

8 

Antoniou A, Stavrou M, Evripidou N, Georgiou E, Kousiappa I, Koupparis A, Papacostas SS, Kleopa KA and Damianou C: FUS-mediated blood-brain barrier disruption for delivering anti-Aβ antibodies in 5XFAD Alzheimer's disease mice. J Ultrasound: Jul 29, 2023 (Epub ahead of print). doi: 10.1007/s40477-023-00805-4.

9 

Śliwińska S and Jeziorek M: The role of nutrition in Alzheimer's disease. Rocz Panstw Zakl Hig. 72:29–39. 2021.PubMed/NCBI View Article : Google Scholar

10 

Fan R, Peng X, Xie L, Dong K, Ma D, Xu W, Shi X, Zhang S, Chen J, Yu X, et al: Importance of Bmal1 in Alzheimer's disease and associated aging-related diseases. Mechanisms and interventions. Aging Cell. 21(e13704)2022.PubMed/NCBI View Article : Google Scholar

11 

Carvalho C and Moreira PI: Metabolic defects shared by Alzheimer's disease and diabetes: A focus on mitochondria. Curr Opin Neurobiol. 79(102694)2023.PubMed/NCBI View Article : Google Scholar

12 

Nguyen TT, Ta QTH, Nguyen TKO, Nguyen TTD and Giau VV: Type 3 diabetes and its role implications in Alzheimer's disease. Int J Mol Sci. 21(3165)2020.PubMed/NCBI View Article : Google Scholar

13 

Janoutová J, Machaczka O, Zatloukalová A and Janout V: Is Alzheimer's disease a type 3 diabetes? A review. Cent Eur J Public Health. 30:139–143. 2022.PubMed/NCBI View Article : Google Scholar

14 

Kandimalla R, Thirumala V and Reddy PH: Is Alzheimer's disease a type 3 diabetes? A critical appraisal. Biochim Biophys Acta Mol Basis Dis. 1863:1078–1089. 2017.PubMed/NCBI View Article : Google Scholar

15 

Michailidis M, Moraitou D, Tata DA, Kalinderi K, Papamitsou T and Papaliagkas V: Alzheimer's disease as type 3 diabetes: Common pathophysiological mechanisms between Alzheimer's disease and type 2 diabetes. Int J Mol Sci. 23(2687)2022.PubMed/NCBI View Article : Google Scholar

16 

Diniz Pereira J, Gomes Fraga V, Morais Santos AL, Carvalho MDG, Caramelli P and Braga Gomes K: Alzheimer's disease and type 2 diabetes mellitus: A systematic review of proteomic studies. J Neurochem. 156:753–776. 2021.PubMed/NCBI View Article : Google Scholar

17 

Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang HY, Ahima RS, Craft S, Gandy S, Buettner C, Stoeckel LE, et al: Brain insulin resistance in type 2 diabetes and Alzheimer disease: Concepts and conundrums. Nat Rev Neurol. 14:168–181. 2018.PubMed/NCBI View Article : Google Scholar

18 

Hamzé R, Delangre E, Tolu S, Moreau M, Janel N, Bailbé D and Movassat J: Type 2 diabetes mellitus and Alzheimer's disease: Shared molecular mechanisms and potential common therapeutic targets. Int J Mol Sci. 23(15287)2022.PubMed/NCBI View Article : Google Scholar

19 

Chen Z and Zhong C: Decoding Alzheimer's disease from perturbed cerebral glucose metabolism: Implications for diagnostic and therapeutic strategies. Prog Neurobiol. 108:21–43. 2013.PubMed/NCBI View Article : Google Scholar

20 

Candasamy M, Mohamed Elhassan SA, Kumar Bhattamisra S, Hua WY, Sern LM, Binti Busthamin NA, Mohamad Ilni NB, Shun NS, Baohong L, Ya NS and Ying NW: Type 3 diabetes (Alzheimer's disease): New insight for promising therapeutic avenues. Panminerva Med. 62:155–163. 2020.PubMed/NCBI View Article : Google Scholar

21 

Zhang Y, Huang NQ, Yan F, Jin H, Zhou SY, Shi JS and Jin F: Diabetes mellitus and Alzheimer's disease: GSK-3β as a potential link. Behav Brain Res. 339:57–65. 2018.PubMed/NCBI View Article : Google Scholar

22 

Moayedi K, Orandi S, Ebrahimi R, Tanhapour M, Moradi M, Abbastabar M and Golestani A: A novel approach to type 3 diabetes mechanism: The interplay between noncoding RNAs and insulin signaling pathway in Alzheimer's disease. J Cell Physiol. 237:2838–2861. 2022.PubMed/NCBI View Article : Google Scholar

23 

de la Monte SM: The full spectrum of Alzheimer's disease is rooted in metabolic derangements that drive type 3 diabetes. Adv Exp Med Biol. 1128:45–83. 2019.PubMed/NCBI View Article : Google Scholar

24 

Ma LY, Fei YL, Wang XY, Wu SD, Du JH, Zhu M, Jin L, Li M, Li HL, Zhai JJ, et al: The research on the relationship of RAGE, LRP-1, and Aβ accumulation in the hippocampus, prefrontal lobe, and amygdala of STZ-Induced diabetic rats. J Mol Neurosci. 62:1–10. 2017.PubMed/NCBI View Article : Google Scholar

25 

XiaoMing Z, Xi Z, Fang S and Jilin Z: Specific changes of somatostatin mRNA expression in the frontal cortex and hippocampus of diabetic rats. J Anat. 204:221–225. 2004.PubMed/NCBI View Article : Google Scholar

26 

Lachmann G, Spies C, Windmann V, Wollersheim T, Engelhardt LJ, Winterer G and Kuehn S: BIOCOG Study Group. Impact of intraoperative hyperglycemia on brain structures and volumes. J Neuroimaging. 29:260–267. 2019.PubMed/NCBI View Article : Google Scholar

27 

Bao H, Liu Y, Zhang M, Chen Z, Zhang W, Ge Y, Kang D, Gao F and Shen Y: Increased β-site APP cleaving enzyme 1-mediated insulin receptor cleavage in type 2 diabetes mellitus with cognitive impairment. Alzheimers Dement. 17:1097–1108. 2021.PubMed/NCBI View Article : Google Scholar

28 

Braun M, Ramracheya R and Rorsman P: Autocrine regulation of insulin secretion. Diabetes Obes Metab. 14 (Suppl 3):S143–S151. 2012.PubMed/NCBI View Article : Google Scholar

29 

Botteri G, Salvadó L, Gumà A, Lee Hamilton D, Meakin PJ, Montagut G, Ashford MLJ, Ceperuelo-Mallafré V, Fernández-Veledo S, Vendrell J, et al: The BACE1 product sAPPβ induces ER stress and inflammation and impairs insulin signaling. Metabolism. 85:59–75. 2018.PubMed/NCBI View Article : Google Scholar

30 

Wang Q, Duan L, Li X, Wang Y, Guo W, Guan F and Ma S: Glucose metabolism, neural cell senescence and Alzheimer's disease. Int J Mol Sci. 23(4351)2022.PubMed/NCBI View Article : Google Scholar

31 

Li H, Yang S, Wu J, Ji L, Zhu L, Cao L, Huang J, Jiang Q, Wei J, Liu M, et al: cAMP/PKA signaling pathway contributes to neuronal apoptosis via regulating IDE expression in a mixed model of type 2 diabetes and Alzheimer's disease. J Cell Biochem. 119:1616–1626. 2018.PubMed/NCBI View Article : Google Scholar

32 

Farr SA, Roesler E, Niehoff ML, Roby DA, McKee A and Morley JE: Metformin improves learning and memory in the SAMP8 mouse model of Alzheimer's disease. J Alzheimers Dis. 68:1699–1710. 2019.PubMed/NCBI View Article : Google Scholar

33 

Mishra D, Reddy I and Dey CS: PKCα Isoform inhibits insulin signaling and aggravates neuronal insulin resistance. Mol Neurobiol. 60:6642–6659. 2023.PubMed/NCBI View Article : Google Scholar

34 

Oriente F, Andreozzi F, Romano C, Perruolo G, Perfetti A, Fiory F, Miele C, Beguinot F and Formisano P: Protein kinase C-alpha regulates insulin action and degradation by interacting with insulin receptor substrate-1 and 14-3-3 epsilon. J Biol Chem. 280:40642–40649. 2005.PubMed/NCBI View Article : Google Scholar

35 

Qi XM and Chen G: p38γ MAPK inflammatory and metabolic signaling in physiology and disease. Cells. 12(1674)2023.PubMed/NCBI View Article : Google Scholar

36 

Esterline R, Oscarsson J and Burns J: A role for sodium glucose cotransporter 2 inhibitors (SGLT2is) in the treatment of Alzheimer's disease? Int Rev Neurobiol. 155:113–140. 2020.PubMed/NCBI View Article : Google Scholar

37 

Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, Fuino RL, Kawaguchi KR, Samoyedny AJ, Wilson RS, et al: Demonstrated brain insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest. 122:1316–1338. 2012.PubMed/NCBI View Article : Google Scholar

38 

Frith E and Loprinzi PD: Fitness Fatness Index and Alzheimer-specific mortality. Eur J Intern Med. 42:51–53. 2017.PubMed/NCBI View Article : Google Scholar

39 

Pugazhenthi S, Qin L and Reddy PH: Common neurodegenerative pathways in obesity, diabetes, and Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis. 1863:1037–1045. 2016.PubMed/NCBI View Article : Google Scholar

40 

Silva MVF, Loures CMG, Alves LCV, de Souza LC, Borges KBG and Carvalho MDG: Alzheimer's disease: Risk factors and potentially protective measures. J Biomed Sci. 26(33)2019.PubMed/NCBI View Article : Google Scholar

41 

Flores-Cordero JA, Pérez-Pérez A, Jiménez-Cortegana C, Alba G, Flores-Barragán A and Sánchez-Margalet V: Obesity as a risk factor for dementia and Alzheimer's disease: The Role of Leptin. Int J Mol Sci. 23(5202)2022.PubMed/NCBI View Article : Google Scholar

42 

Khanna D, Peltzer C, Kahar P and Parmar MS: Body mass index (BMI): A screening tool analysis. Cureus. 14(e22119)2022.PubMed/NCBI View Article : Google Scholar

43 

Flegal KM, Kit BK, Orpana H and Graubard BI: Association of all-cause mortality with overweight and obesity using standard body mass index categories: A systematic review and meta-analysis. JAMA. 309:71–82. 2013.PubMed/NCBI View Article : Google Scholar

44 

Ashwell M, Gunn P and Gibson S: Waist-to-height ratio is a better screening tool than waist circumference and BMI for adult cardiometabolic risk factors: Systematic review and meta-analysis. Obes Rev. 13:275–286. 2012.PubMed/NCBI View Article : Google Scholar

45 

Diehl-Wiesenecker E, von Armin CA, Dupuis L, Müller HP, Ludolph AC and Kassubek J: Adipose tissue distribution in patients with Alzheimer's disease: A whole body MRI Case-Control study. J Alzheimers Dis. 48:825–832. 2015.PubMed/NCBI View Article : Google Scholar

46 

Boccara E, Golan S and Beeri MS: The association between regional adiposity, cognitive function, and dementia-related brain changes: A systematic review. Front Med (Lausanne). 10(1160426)2023.PubMed/NCBI View Article : Google Scholar

47 

Gómez-Apo E, Mondragón-Maya A, Ferrari-Díaz M and Silva-Pereyra J: Structural Brain Changes associated with overweight and obesity. J Obes. 2021(6613385)2021.PubMed/NCBI View Article : Google Scholar

48 

Li X, Chen H, Lv Y, Chao HH, Gong L, Li CR and Cheng H: Diminished gray matter density mediates chemotherapy dosage-related cognitive impairment in breast cancer patients. Sci Rep. 8(13801)2018.PubMed/NCBI View Article : Google Scholar

49 

Miao Y, Zhang B, Sun X, Ma X, Fang D, Zhang W, Wu T, Xu X, Yu C, Hou Y, et al: The presence and severity of NAFLD are associated with cognitive impairment and hippocampal damage. J Clin Endocrinol Metab. 108:3239–3249. 2023.PubMed/NCBI View Article : Google Scholar

50 

Weinstein G, Zelber-Sagi S, Preis SR, Beiser AS, DeCarli C, Speliotes EK, Satizabal CL, Vasan RS and Seshadri S: Association of nonalcoholic fatty liver disease with lower brain volume in healthy Middle-aged adults in the framingham study. JAMA Neurol. 75:97–104. 2018.PubMed/NCBI View Article : Google Scholar

51 

Ruchinskas R, Nguyen T, Womack K, Khera A, Yu FF and Kelley BJ: Diagnostic utility of hippocampal volumetric data in a memory disorder clinic setting. Cogn Behav Neurol. 35:66–75. 2022.PubMed/NCBI View Article : Google Scholar

52 

van Oostveen WM and de Lange ECM: Imaging techniques in Alzheimer's disease: A review of applications in early diagnosis and longitudinal monitoring. Int J Mol Sci. 22(2110)2021.PubMed/NCBI View Article : Google Scholar

53 

Prem Kumar A, Singh N, Nair D and Justin A: Neuronal PET tracers for Alzheimer's disease. Biochem Biophys Res Commun. 587:58–62. 2022.PubMed/NCBI View Article : Google Scholar

54 

So SW, Fleming KM, Nixon JP and Butterick TA: Early life obesity increases neuroinflammation, amyloid beta deposition, and cognitive decline in a mouse model of Alzheimer's disease. Nutrients. 15(2494)2023.PubMed/NCBI View Article : Google Scholar

55 

Jayaraman A, Lent-Schochet D and Pike CJ: Diet-induced obesity and low testosterone increase neuroinflammation and impair neural function. J Neuroinflammation. 11(162)2014.PubMed/NCBI View Article : Google Scholar

56 

Liu P, Wang ZH, Kang SS, Liu X, Xia Y, Chan CB and Ye K: High-fat diet-induced diabetes couples to Alzheimer's disease through inflammation-activated C/EBPβ/AEP pathway. Mol Psychiatry. 27:3396–3409. 2022.PubMed/NCBI View Article : Google Scholar

57 

Medrano-Jiménez E, Jiménez-Ferrer Carrillo I, Pedraza-Escalona M, Ramírez-Serrano CE, Álvarez-Arellano L, Cortés-Mendoza J, Herrera-Ruiz M, Jiménez-Ferrer E, Zamilpa A, Tortoriello J, et al: Malva parviflora extract ameliorates the deleterious effects of a high fat diet on the cognitive deficit in a mouse model of Alzheimer's disease by restoring microglial function via a PPAR-γ-dependent mechanism. J Neuroinflammation. 16(143)2019.PubMed/NCBI View Article : Google Scholar

58 

Zhang B, Chen T, Cao M, Yuan C, Reiter RJ, Zhao Z, Zhao Y, Chen L, Fan W, Wang X, et al: Gut microbiota dysbiosis induced by decreasing endogenous melatonin mediates the pathogenesis of Alzheimer's disease and obesity. Front Immunol. 13(900132)2022.PubMed/NCBI View Article : Google Scholar

59 

Obradovic M, Zaric B, Sudar-Milovanovic E, Ilincic B, Stokic E, Perovic M and Isenovic ER: PCSK9 and hypercholesterolemia: Therapeutic Approach. Curr Drug Targets. 19:1058–1067. 2018.PubMed/NCBI View Article : Google Scholar

60 

Mannarino MR, Ministrini S and Pirro M: Nutraceuticals for the treatment of hypercholesterolemia. Eur J Intern Med. 25:592–599. 2014.PubMed/NCBI View Article : Google Scholar

61 

Loera-Valencia R, Goikolea J, Parrado-Fernandez C, Merino-Serrais P and Maioli S: Alterations in cholesterol metabolism as a risk factor for developing Alzheimer's disease: Potential novel targets for treatment. J Steroid Biochem Mol Biol. 190:104–114. 2019.PubMed/NCBI View Article : Google Scholar

62 

Appleton JP, Scutt P, Sprigg N and Bath PM: Hypercholesterolaemia and vascular dementia. Clin Sci (Lond). 131:1561–1578. 2017.PubMed/NCBI View Article : Google Scholar

63 

Sandebring-Matton A, Goikolea J, Björkhem I, Paternain L, Kemppainen N, Laatikainen T, Ngandu T, Rinne J, Soininen H, Cedazo-Minguez A, et al: 27-Hydroxycholesterol, cognition, and brain imaging markers in the FINGER randomized controlled trial. Alzheimers Res Ther. 13(56)2021.PubMed/NCBI View Article : Google Scholar

64 

Reiman EM, Chen K, Langbaum JB, Lee W, Reschke C, Bandy D, Alexander GE and Caselli RJ: Higher serum total cholesterol levels in late middle age are associated with glucose hypometabolism in brain regions affected by Alzheimer's disease and normal aging. Neuroimage. 49:169–176. 2010.PubMed/NCBI View Article : Google Scholar

65 

Gottesman RF, Schneider AL, Zhou Y, Coresh J, Green E, Gupta N, Knopman DS, Mintz A, Rahmim A, Sharrett AR, et al: Association between midlife vascular risk factors and estimated brain amyloid deposition. JAMA. 317:1443–1450. 2017.PubMed/NCBI View Article : Google Scholar

66 

Serrano-Pozo A, Das S and Hyman BT: APOE and Alzheimer's disease: Advances in genetics, pathophysiology, and therapeutic approaches. Lancet Neurol. 20:68–80. 2021.PubMed/NCBI View Article : Google Scholar

67 

Martens YA, Zhao N, Liu CC, Kanekiyo T, Yang AJ, Goate AM, Holtzman DM and Bu G: ApoE Cascade Hypothesis in the pathogenesis of Alzheimer's disease and related dementias. Neuron. 110:1304–1317. 2022.PubMed/NCBI View Article : Google Scholar

68 

Raulin AC, Doss SV, Trottier ZA, Ikezu TC, Bu G and Liu CC: ApoE in Alzheimer's disease: Pathophysiology and therapeutic strategies. Mol Neurodegener. 17(72)2022.PubMed/NCBI View Article : Google Scholar

69 

Koutsodendris N, Nelson MR, Rao A and Huang Y: Apolipoprotein E and Alzheimer's Disease: Findings, hypotheses, and potential mechanisms. Annu Rev Pathol. 17:73–99. 2022.PubMed/NCBI View Article : Google Scholar

70 

Lanfranco MF, Ng CA and Rebeck GW: ApoE lipidation as a therapeutic target in Alzheimer's disease. Int J Mol Sci. 21(6336)2020.PubMed/NCBI View Article : Google Scholar

71 

Zhao N, Liu CC, Qiao W and Bu G: Apolipoprotein E, receptors, and modulation of Alzheimer's disease. Biol Psychiatry. 83:347–357. 2018.PubMed/NCBI View Article : Google Scholar

72 

Fernández-Calle R, Konings SC, Frontiñán-Rubio J, García-Revilla J, Camprubí-Ferrer L, Svensson M, Martinson I, Boza-Serrano A, Venero JL, Nielsen HM, et al: APOE in the bullseye of neurodegenerative diseases: Impact of the APOE genotype in Alzheimer's disease pathology and brain diseases. Mol Neurodegener. 17(62)2022.PubMed/NCBI View Article : Google Scholar

73 

McNamara DJ: Dietary cholesterol, heart disease risk and cognitive dissonance. Proc Nutr Soc. 73:161–166. 2014.PubMed/NCBI View Article : Google Scholar

74 

Lee SB, Kim HG, Lee JS, Kim WY, Lee MM, Kim YH, Lee JO, Kim HS and Son CG: Intermittent restraint-induced sympathetic activation attenuates hepatic steatosis and inflammation in a high-fat diet-fed mouse model. Am J Physiol Gastrointest Liver Physiol. 317:G811–G823. 2019.PubMed/NCBI View Article : Google Scholar

75 

Ettcheto M, Petrov D, Pedrós I, Alva N, Carbonell T, Beas-Zarate C, Pallas M, Auladell C, Folch J and Camins A: Evaluation of neuropathological effects of a High-Fat diet in a presymptomatic Alzheimer's disease stage in APP/PS1 Mice. J Alzheimers Dis. 54:233–251. 2016.PubMed/NCBI View Article : Google Scholar

76 

Bromley-Brits K, Deng Y and Song W: Morris water maze test for learning and memory deficits in Alzheimer's disease model mice. J Vis Exp. 20(2920)2011.PubMed/NCBI View Article : Google Scholar

77 

Song J: Animal model of aluminum-induced Alzheimer's disease. Adv Exp Med Biol. 1091:113–127. 2018.PubMed/NCBI View Article : Google Scholar

78 

Drummond E and Wisniewski T: Alzheimer's disease: Experimental models and reality. Acta Neuropathol. 133:155–175. 2017.PubMed/NCBI View Article : Google Scholar

79 

Zhang L, Chen C, Mak MS, Lu J, Wu Z, Chen Q, Han Y, Li Y and Pi R: Advance of sporadic Alzheimer's disease animal models. Med Res Rev. 40:431–458. 2020.PubMed/NCBI View Article : Google Scholar

80 

Thawkar BS and Kaur G: Zebrafish as a promising tool for modeling Neurotoxin-Induced Alzheimer's disease. Neurotox Res. 39:949–965. 2021.PubMed/NCBI View Article : Google Scholar

81 

Tsuda L and Lim YM: Alzheimer's disease model system using drosophila. Adv Exp Med Biol. 1076:25–40. 2018.PubMed/NCBI View Article : Google Scholar

82 

Souder DC, Dreischmeier IA, Smith AB, Wright S, Martin SA, Sagar MAK, Eliceiri KW, Salamat SM, Bendlin BB, Colman RJ, et al: Rhesus monkeys as a translational model for late-onset Alzheimer's disease. Aging Cell. 20(e13374)2021.PubMed/NCBI View Article : Google Scholar

83 

Edler MK, Munger EL, Meindl RS, Hopkins WD, Ely JJ, Erwin JM, Mufson EJ, Hof PR, Sherwood CC and Raghanti MA: Neuron loss associated with age but not Alzheimer's disease pathology in the chimpanzee brain. Philos Trans R Soc Lond B Biol Sci. 375(20190619)2020.PubMed/NCBI View Article : Google Scholar

84 

Dewey CW, Davies ES, Xie H and Wakshlag JJ: Canine cognitive dysfunction: Pathophysiology, diagnosis, and treatment. Vet Clin North Am Small Anim Pract. 49:477–499. 2019.PubMed/NCBI View Article : Google Scholar

85 

Esquerda-Canals G, Montoliu-Gaya L, Güell-Bosch J and Villegas S: Mouse models of Alzheimer's Disease. J Alzheimers Dis. 57:1171–1183. 2017.PubMed/NCBI View Article : Google Scholar

86 

Puzzo D, Gulisano W, Palmeri A and Arancio O: Rodent models for Alzheimer's disease drug discovery. Expert Opin Drug Discov. 10:703–711. 2015.PubMed/NCBI View Article : Google Scholar

87 

Hefti F, Dravid A and Hartikka J: Chronic intraventricular injections of nerve growth factor elevate hippocampal choline acetyltransferase activity in adult rats with partial septo-hippocampal lesions. Brain Res. 293:305–311. 1984.PubMed/NCBI View Article : Google Scholar

88 

Washida K, Hattori Y and Ihara M: Animal models of chronic cerebral hypoperfusion: From mouse to primate. Int J Mol Sci. 20(6176)2019.PubMed/NCBI View Article : Google Scholar

89 

Yi J, Liu BY and Cai GX: Study on effect of Ultra-powder Liuwei Dihuang decoction on recognize and bFGF in Alzheime's disease rats bFGF. Chin J Exp Tradit Med Formulae. 17:139–142. 2011.

90 

Chen HF, Mo YS and Yang C: Protective effect and mechanism of quercetin on hippocampal neurons in IBO-injured rats. Tradit Chin Drug Res Clin Pharmacol. 29:552–556. 2018.

91 

Lester-Coll N, Rivera EJ, Soscia SJ, Doiron K, Wands JR and de la Monte SM: Intracerebral Streptozotocin model of type 3 diabetes: Relevance to sporadic Alzheimer's disease. J Alzheimers Dis. 9:13–33. 2006.PubMed/NCBI View Article : Google Scholar

92 

Wang C, Cai Z, Wang W, Wei M, Si X, Shang Y, Yang Z, Li T, Guo H and Li S: Piperine regulates glycogen synthase kinase-3β-related signaling and attenuates cognitive decline in D-galactose-induced aging mouse model. J Nutr Biochem. 75(108261)2020.PubMed/NCBI View Article : Google Scholar

93 

Mustafa HN: Neuro-amelioration of cinnamaldehyde in aluminum-induced Alzheimer's disease rat model. J Histotechnol. 43:11–20. 2020.PubMed/NCBI View Article : Google Scholar

94 

Song XY, Hu JF, Chu SF, Zhang Z, Xu S, Yuan YH, Han N, Liu Y, Niu F, He X and Chen NH: Ginsenoside Rg1 attenuates okadaic acid induced spatial memory impairment by the GSK3β/tau signaling pathway and the Aβ formation prevention in rats. Eur J Pharmacol. 710:29–38. 2013.PubMed/NCBI View Article : Google Scholar

95 

Haider S, Tabassum S and Perveen T: Scopolamine-induced greater alterations in neurochemical profile and increased oxidative stress demonstrated a better model of dementia: A comparative study. Brain Res Bull. 127:234–247. 2016.PubMed/NCBI View Article : Google Scholar

96 

Zangerolamo L, Vettorazzi JF, Solon C, Bronczek GA, Engel DF, Kurauti MA, Soares GM, Rodrigues KS, Velloso LA, Boschero AC, et al: The bile acid TUDCA improves glucose metabolism in streptozotocin-induced Alzheimer's disease mice model. Mol Cell Endocrinol. 521(111116)2021.PubMed/NCBI View Article : Google Scholar

97 

Liu RM: Aging, cellular senescence, and Alzheimer's Disease. Int J Mol Sci. 23(1989)2022.PubMed/NCBI View Article : Google Scholar

98 

Stern Y: Cognitive reserve in ageing and Alzheimer's disease. Lancet Neurol. 11:1006–1012. 2012.PubMed/NCBI View Article : Google Scholar

99 

Leuti A, Fazio D, Fava M, Piccoli A, Oddi S and Maccarrone M: Bioactive lipids, inflammation and chronic diseases. Adv Drug Deliv Rev. 159:133–169. 2020.PubMed/NCBI View Article : Google Scholar

100 

Kälin S, Heppner FL, Bechmann I, Prinz M, Tschöp MH and Yi CX: Hypothalamic innate immune reaction in obesity. Nat Rev Endocrinol. 11:339–351. 2015.PubMed/NCBI View Article : Google Scholar

101 

de Git KC and Adan RA: Leptin resistance in diet-induced obesity: The role of hypothalamic inflammation. Obes Rev. 16:207–224. 2015.PubMed/NCBI View Article : Google Scholar

102 

Boccardi V, Murasecco I and Mecocci P: Diabetes drugs in the fight against Alzheimer's disease. Ageing Res Rev. 54(100936)2019.PubMed/NCBI View Article : Google Scholar

103 

Kellar D and Craft S: Brain insulin resistance in Alzheimer's disease and related disorders: Mechanisms and therapeutic approaches. Lancet Neurol. 19:758–766. 2020.PubMed/NCBI View Article : Google Scholar

104 

Campbell JM, Stephenson MD, de Courten B, Chapman I, Bellman SM and Aromataris E: Metformin use associated with reduced risk of dementia in patients with diabetes: A systematic review and Meta-Analysis. J Alzheimers Dis. 65:1225–1236. 2018.PubMed/NCBI View Article : Google Scholar

105 

Briggs R, Kennelly SP and O'Neill D: Drug treatments in Alzheimer's disease. Clin Med (Lond). 16:247–253. 2016.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

February-2024
Volume 27 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou S, Tu L, Chen W, Yan G, Guo H, Wang X, Hu Q, Liu H and Li F: Alzheimer's disease, a metabolic disorder: Clinical advances and basic model studies (Review). Exp Ther Med 27: 63, 2024
APA
Zhou, S., Tu, L., Chen, W., Yan, G., Guo, H., Wang, X. ... Li, F. (2024). Alzheimer's disease, a metabolic disorder: Clinical advances and basic model studies (Review). Experimental and Therapeutic Medicine, 27, 63. https://doi.org/10.3892/etm.2023.12351
MLA
Zhou, S., Tu, L., Chen, W., Yan, G., Guo, H., Wang, X., Hu, Q., Liu, H., Li, F."Alzheimer's disease, a metabolic disorder: Clinical advances and basic model studies (Review)". Experimental and Therapeutic Medicine 27.2 (2024): 63.
Chicago
Zhou, S., Tu, L., Chen, W., Yan, G., Guo, H., Wang, X., Hu, Q., Liu, H., Li, F."Alzheimer's disease, a metabolic disorder: Clinical advances and basic model studies (Review)". Experimental and Therapeutic Medicine 27, no. 2 (2024): 63. https://doi.org/10.3892/etm.2023.12351