Vildagliptin prevents cognitive deficits and neuronal apoptosis in a rat model of Alzheimer's disease

  • Authors:
    • Qing‑Hua Ma
    • Liu‑Fang Jiang
    • Jian‑Liang Mao
    • Wen‑Xin Xu
    • Min Huang
  • View Affiliations

  • Published online on: December 18, 2017     https://doi.org/10.3892/mmr.2017.8289
  • Pages: 4113-4119
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Diabetes has been identified to be a risk factor for Alzheimer's disease (AD). Vildagliptin, a novel oral hypoglycemic agent, has been demonstrated to exert protective effects on the pancreas and cardiovascular system. The present study examined the potential protective effects of vildagliptin on neurons in an AD rat model. Treatment with vildagliptin improved memory deficits and decreased neuronal apoptosis in the hippocampus. The expression levels of B cell lymphoma 2 (Bcl‑2) were increased, and the expression levels of caspase‑3, Bcl‑2 associated X protein and AD‑associated proteins were decreased in the hippocampus following treatment with vildagliptin. Additionally, the AD model‑induced decrease in phosphorylated (p) protein kinase B (p‑Akt), p‑glycogen synthase kinase 3β (p‑GSK3β), post‑synaptic density 95 and synaptophysin expression was reversed. These results indicate that vildagliptin administration exerts a protective effect against cognitive deficits by reducing tau phosphorylation and increasing the expression of proteins associated with synaptic plasticity in the hippocampus. Targeting of the Akt/GSK3β signaling pathway may be a key mechanism in preventing the disease progression of AD.

Introduction

Alzheimer's disease (AD) is one of the most severe types of neurodegenerative disease, characterized by a decline in memory capacity and other cognitive abilities (1). The incidence of AD has grown rapidly in the past few decades. It was estimated in 2010 that ~40 million people worldwide were living with AD, and this figure is expected to double every 20 years (2). At present, there are no treatments that are able to completely cure AD and only symptomatic therapeutics are available (3,4). The brains of patients with AD have a number of hallmark features. For instance, twisted strands of hyperphosphorylated tau would cause neurodegeneration as a consequence of structural incompleteness of microtubules, which is indeed observed in the widespread intraneuronal fibrillary tangles and intracellular formation of neurofibrillary tangles (NFTs) (5). Senile plaques consist of modified β amyloid (Aβ) peptides which are able to induce neuronal apoptosis. Aβ is rapidly degraded by a number of proteases which maintain its concentration in normal neuronal cells (6). Impaired balance between Aβ production and clearance contribute to the amyloidogenic pathway (6). NFTs primarily consist of phosphorylated tau protein (7). Therefore, the degree of plaque deposition and synaptic plasticity in the hippocampus are critical indices for evaluating the anti-AD efficacy of potential therapeutics.

Type 2 diabetes is a risk factor for AD development and may exacerbate the progression of the disease (8). Overlapping mechanisms between type 2 diabetes and brain disorders suggest that antidiabetic drugs may have beneficial effects on brain-cell metabolism, which may be of clinical importance for the treatment of brain complications in diabetes and AD (9). Dipeptidyl peptidase-IV (DPP4) inhibitors are a class of hypoglycemic drugs used in clinical practice that have been demonstrated to improve memory in various animal models of neurodegeneration (1012). The results of the GDMD Study in China confirmed a positive association between plasma DPP4 activity and mild cognitive impairment in elderly patients with type 2 diabetes. DPP4 inhibitors have been demonstrated to ameliorate cognitive impairment through the suppression of inflammation and oxidative stress in mouse models (13). Vildagliptin is a DPP4 inhibitor that effectively inhibits the degradation of glucagon-like peptide-1 (GLP-1). Vildagliptin was demonstrated to increase the expression and activity of GLP-1 in the peripheral blood and to reduce Aβ, phosphorylated tau and inflammation in an AD mouse model (14). Thus, the use of vildagliptin to reduce plaque formation and improve synaptic plasticity warrants further investigation. In the present study, a rat model of AD was used to investigate the effects of vildagliptin on cognitive function and to examine its underlying mechanisms. In combination with other evidence (14), the present study indicates that vildagliptin may inhibit plaque formation and regulate synaptic plasticity in AD, providing novel evidence to suggest the development of vildagliptin as a potential therapy for AD.

Materials and methods

Animals

Male Sprague-Dawley rats (n=40; 210–230 g) were obtained from the Animal Center of Suzhou University (Suzhou, China) and maintained in plastic cages (5 rats per cage) at 20–24°C and 50±10% humidity, in a 12-h light-dark cycle with free access to food and water. All animal experiments were approved by The Institutional Review Board of Soochow University (Suzhou, China) and experimental procedures were performed in accordance with guidelines for The Care and Use of Laboratory Animals, and The National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals (NIH Publication no. 80–23, revised 1996; NIH, Bethesda, MD, USA).

Preparation of animal models

Rats were randomly divided into the following four groups: Sham, AD, AD + low-dose vildagliptin (AD + Vilda-L) and AD + high-dose vildagliptin (AD + Vilda-H). Rats were anesthetized with 1% pentobarbital sodium (40 mg/kg) via intraperitoneal injection and were subsequently administered 10 µg/µl Aβ1-40 via intracerebral ventricular injection. The sham group was injected with the equivalent volume of saline. All rats were subjected to the Morris water maze test to evaluate whether the AD model was successfully established from day 11–15 following the induction of AD and their behavioral indexes were recorded once daily. There was one mortality incidence in the AD group during the model establishment. Rats in the vildagliptin groups were treated with 5 (AD + Vilda-L) or 10 (AD + Vilda-H) mg/kg vildagliptin (Gulvus; Novartis International AG, Basel, Switzerland) once a day by oral gavage for 4 consecutive weeks. Following vildagliptin treatment, behavioral tests and biochemical analysis was performed.

Morris water maze (MWM) test

The spatial learning and memory of the rats was tested by MWM assessment following treatment with vildagliptin. Rats were placed in a black circular water tank (diameter, 150 cm; depth, 60 cm; 25°C). Reference objects were placed around the pool as visual hints and were left unaltered throughout the MWM test. During the 4-day training period, rats were randomly placed into the water at any point in the four quadrants, with one test performed in every quadrant each morning for 1 min at every turn. The rats that did not locate the platform were placed on the platform and allowed to stand for 15 sec. Rats that successfully located the platform were additionally permitted to stand on it for 15 sec. On the 5th day, the platform was removed and the rats were allowed to swim for 1 min. Maze performance was recorded with a video camera located above the pool, interfaced with a video tracking system (HVS Image, Buckingham, UK). The average escape latency and time spent in each quadrant in a total of five trials was recorded.

Histology

Following completion of the MWM test, rats were anesthetized with sodium pentobarbital (60 mg/kg) and euthanized by transcardial perfusion with cold PBS. The hippocampus of brain was rapidly isolated, part of the tissues was subsequently fixed with cold 4% paraformaldehyde containing 0.2% saturated picric acid for 24 h at 4°C. The remaining samples were stored at −80°C. Paraffin-embedded sections were cut in a coronal plane at a thickness of 5 µm with a microtome. Paraffin-embedded brain sections were de-paraffinized with xylene (3 times × 5 min) and subsequently rehydrated prior to Nissl staining with 0.1% (w/v) cresyl violet to investigate the degree of neuronal damage in the hippocampus. The mean number of morphologically-intact neurons per 100 µm length in the CA1 hippocampal area was calculated to accurately estimate the extent of neuronal damage in comparison to controls.

Western blot analysis

Frozen samples were obtained and lysed in Tissue Protein Lysis Solution (Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 5% Proteinase Inhibitor Cocktail (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). A total of 25 µg extracted protein/lane was separated on a 12% SDS-PAGE gel (Beyotime Institute of Biotechnology, Haimen, China) and transferred onto a nitrocellulose membrane (Beyotime Institute of Biotechnology) using wet transfer. The membranes were blocked in 5% non-fat milk for 2 h at room temperature and washed three times in PBS with Tween 20 (PBST). The membrane was subsequently incubated with the following primary antibodies overnight at 4°C: Rabbit polyclonal caspase-3 (1:1,000; cat. no. AB13847; Abcam, Cambridge, UK), rabbit polyclonal B cell lymphoma 2 (Bcl-2; 1:1,000; cat. no. AB59348; Abcam), rabbit monoclonal Bcl-2 associated X protein (Bax; 1:1,000; cat. no. AB32503; Abcam), rabbit polyclonal phosphorylated (p) protein kinase B (p-Akt; 1:1,000; cat. no. AB38449; Abcam), rabbit polyclonal Akt (1:1,000; cat. no. AB8805; Abcam), rabbit monoclonal glycogen synthase kinase 3β (GSK3β; 1:1,000; cat. no. AB32391; Abcam), rabbit polyclonal p-GSK3β (1:1,000; cat. no. AB131356; Abcam), rabbit polyclonal postsynaptic density protein 95 (PSD-95; 1:1,000; cat. no. AB18258; Abcam), rabbit monoclonal synaptophysin (1:1,000; cat. no. AB32127; Abcam), rabbit polyclonal p-tau (1:1,000; cat. no. AB109390; Abcam), rabbit monoclonal tau (1:1,000; cat. no. AB32057; Abcam), amyloid precursor protein (APP; 1:1,000; cat. no. AB32136; Abcam) and rabbit polyclonal β-actin (1:1,000; cat. no. AB8227; Abcam). Following incubation with primary antibodies, the membranes were washed with PBST three times for 10 min each and subsequently incubated with polyclonal goat anti-rabbit secondary antibody (1:1,000; Abcam; cat. no. AB205718) for 2 h at room temperature. Protein levels were analyzed using Image J software version 1.41 (NIH) following exposure to an ECL kit (BOSTER Biological Technology, Pleasanton, CA, USA).

Statistical analysis

The data are presented as the mean ± standard deviation. Each experiment was repeated a minimum of 3 times. The statistical analysis was performed using one-way analysis of variance, followed by the Bonferroni post hoc test, with SPSS 21.0 (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Vildagliptin improves spatial learning and memory impairment in the AD group

The MWM test was used to assess spatial learning function. Compared with the Sham group, the escape latency in rats were administered 10 µg/µl Aβ1-40 via intracerebral ventricular injection was significantly longer (P<0.01, Fig. 1A) from day 11–15 following the induction of AD. It indicated that the AD model is modeled successfully. After four weeks the AD group still exhibited a significant spatial learning deficit compared with the sham group, and vildagliptin administration significantly reduced the escape latency compared with the AD group, particularly in the Vilda-H group (Fig. 1B). Following the 4-day training period, the platform was removed. The time spent in the target quadrant was significantly lower in the AD group compared with the sham group (Fig. 1C). The time spent in the target quadrant by the AD + Vilda-L group was significantly increased compared with the AD group. The Vilda-H group demonstrated the most significant improvement in the spatial probe test compared to the AD group (P<0.05).

Treatment with vildagliptin attenuates the expression of apoptosis-associated proteins and prevents neuronal cell loss

The Nissl staining method was performed to investigate neuronal alterations in the hippocampal CA1 region of rats of different groups. Hippocampal neurons in the sham group were large and conical with well-demarcated amphophilic cytoplasm and round vesicular nuclei with prominent nucleoli (Fig. 2A). Neurons in the AD group were characterized by pyknotic pyramidal cells and pronounced neuronal body shrinkage with nuclear loss (Fig. 2B). Treatment with vildagliptin reduced the AD model-induced cell loss and pyknosis, although degenerating cells with altered morphology were still observed (Fig. 2C and D). Treatment with vildagliptin exerted significant protection against AD-induced neurotoxicity, as determined by neuronal density (Fig. 2E).

Hippocampal caspase-3, Bax and Bcl-2 expression

The expression of caspase-3 and Bax increased in the AD model compared with the sham group (Fig. 3A and B; P<0.01) and treatment with vildagliptin downregulated this expression. The expression level of Bcl-2 appeared to decrease in the AD model compared with the sham group (Fig. 3B). However, vildagliptin-treated rats exhibited a dose-dependent increase in hippocampal Bcl-2 expression levels by comparison with the AD group (Vilda-L and Vilda-H, P<0.05).

Vildagliptin treatment reduces the expression of AD-associated proteins

The expression levels of APP in the hippocampus were determined by western blot analysis. The expression of APP increased in the AD model group compared with the sham group (Fig. 4A; P<0.05) and APP expression was downregulated by treatment with vildagliptin (Vilda-L and Vilda-H, P<0.05). Additionally, p-tau expression increased in the AD group compared with the sham group (Fig. 4B; P<0.05). The expression levels of p-tau were reduced following treatment with vildagliptin (Vilda-L and Vilda-H, P<0.05).

Treatment with vildagliptin enhances the expression of proteins associated with synaptic plasticity

The hippocampal expression of the synapse-associated proteins synaptophysin and PSD-95 was analyzed. Levels of PSD-95 and synaptophysin in the hippocampus were significantly decreased (P<0.01) in the AD group compared with the sham group (Fig. 5). Treatment with vildagliptin (Vilda-H) significantly enhanced PSD-95 and synaptophysin expression levels in the hippocampus compared with the AD group (P<0.01). However, the low dose of vildagliptin did not significantly increase synaptophysin expression (Fig. 5B).

Vildagliptin increases Akt/GSK3β pathway activity

The total protein levels of Akt and GSK3β did not vary notably between groups. However, levels of p-Akt were significantly decreased in the hippocampus of the compared with the sham group (P<0.05). Rats in the AD group had increased p-GSK3β levels compared with the rats in sham group (P<0.05). The administration of vildagliptin significantly increased p-Akt (Vilda-L, P<0.05; Vilda-H, P<0.01) and reduced p-GSK3β levels (Vilda-L, P<0.05; Vilda-H, P<0.01), in a dose-dependent manner (Fig. 6).

Discussion

AD is a progressive neurodegenerative disease that leads to memory impairment, aphasia, disability, visual impairment, administrative dysfunction, and personality and behavioral changes. In 1907, Alois Alzheimer's experimental observations first identified the pathological modifications of Aβ and tau, which are the hallmark features of AD (15). The deposition of prefibrillar and fibrillar oligomeric Aβ, tau protein phosphorylation, synaptic loss, inflammation and glial cell activation contribute to AD pathogenesis (16). Various hypotheses have been proposed to explain the different causes of AD, but the exact mechanism remains unknown. Currently, FDA approved drugs for AD only offer symptomatic relief through the control of neurotransmitter levels and the activity of neurotransmitters, including donepezil and memantine, which affect the cholinergic and glutamatergic systems, respectively (17,18). Thus, the discovery and identification of novel and effective treatments is required.

DPP4 inhibitors are a class of oral hypoglycemic agents used in monotherapy or in combination with other antidiabetic compounds. DPP4 inhibitors bind reversibly and competitively to DPP4, which indirectly enhances the levels of incretin hormones, particularly GLP-1 and gastric inhibitory polypeptide. Numerous studies have investigated the effect of DPP4 inhibitors on cognitive function. Treatment with sitagliptin was demonstrated to significantly improve the working and reference memories of diabetic rats (19); it may additionally improve the memory and hippocampal neurogenesis of high fat-fed mice (20). Furthermore, Aβ deposition was delayed in an early stage AD transgenic mouse model following treatment with sitagliptin (21).

Vildagliptin has been demonstrated to significantly reduce oxidative stress in the brain, restore brain insulin sensitivity and mitochondrial function, and improve hippocampal synaptic plasticity and cognitive function in an obese rat model (22). In a streptozotocin (STZ)-induced diabetic rat model, treatment with vildagliptin significantly improved memory and learning impairments (23). Furthermore, treatment with vildagliptin ameliorated cognitive deficits in a STZ-induced rat model of AD (14). This evidence demonstrates that vildagliptin may effectively improve cognitive function.

The results of the present study demonstrated that the established AD rat model exhibited a decline in memory performance. Treatment for 1 month with vildagliptin was demonstrated to ameliorate cognitive deficits in the AD model in a dose-dependent manner. Bcl-2 expression was increased, and Bax and caspase-3 expression was decreased by treatment with vildagliptin. The mechanism underlying the anti-apoptotic effect of vildagliptin was not determined, but may have been mediated by the activation of the Akt/GSK3β signaling pathway (24,25), an effect previously reported for vildagliptin in vitro (26). Consistent with this finding, vildagliptin administration following AD model-induced neuronal damage inhibited the activation of caspase-3, decreased Bax and increased Bcl-2 expression, and therefore may reduce apoptosis in the brain.

The AD-afflicted brain is characterized by the presence of senile plaques and NFTs composed of aggregated Aβ peptides and p-tau, respectively (2729). It has been reported that Aβ plaques are present in the brain of approximately one-third to one-half of individuals aged ≥65, and tau inclusions are present almost universally (30). In AD, APP is cleaved by β-secretase and γ-secretase to produce toxic Aβ protein, which is involved in plaque formation (31). In the present study, vildagliptin was demonstrated to significantly reduce APP and p-tau protein expression, with this effect being more pronounced at the higher 10 mg/kg vildagliptin dose. This suggested that vildagliptin was able to reduce Aβ aggregation and tau hyperphosphorylation in AD. Expression of PSD-95 in the AD group was significantly decreased compared with the sham group, and this expression was recovered by treatment with vildagliptin, suggesting that vildagliptin may improve synaptic plasticity, which is important for improving cognitive function. Synaptophysin expression levels were not significantly improved in the AD+Vilda-L group compared with the AD group. However, a significant improvement was observed in the AD+Vilda-H group. In terms of adverse effects, a previous study indicated that the most common adverse events in vildagliptin-treated subjects were mild or moderate, and suspected to be unrelated to the study medication. Nausea occurred in certain vildagliptin-treated subjects, although this was suggested to not be a dose-limiting side effect of DPP-4 inhibition (32).

In conclusion, the present study demonstrated that vildagliptin improved learning and memory deficits induced in an AD rat model, through an increase in the expression of proteins associated with synaptic plasticity, and a decrease in the expression of apoptosis and AD-associated proteins. Additionally, activation of the Akt and GSK-3β inhibition may have contributed to the improvement in cognitive function mediated by vildagliptin. The present study provides evidence for the development of vildagliptin as a potential therapeutic for AD.

Acknowledgements

The present study was supported by a Project of Opening Project of Provincial Key Laboratory of Soochow University (grant no. KJS1513) and the Suzhou Science and Technology Project (grant no. XJ201460).

References

1 

Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S and Van der Flier WM: Alzheimer's disease. Lancet. 388:505–517. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W and Ferri CP: The global prevalence of dementia: A systematic review and metaanalysis. Alzheimers Dement. 9:63–75. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Li X, Bao X and Wang R: Neurogenesis-based epigenetic therapeutics for Alzheimer's disease. Mol Med Rep. 14:1043–1053. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Chen PY, Tsai CT, Ou CY, Hsu WT, Jhuo MD, Wu CH, Shih TC, Cheng TH and Chung JG: Computational analysis of novel drugs designed for use as acetylcholinesterase inhibitors and histamine H3 receptor antagonists for Alzheimer's disease by docking, scoring and de novo evolution. Mol Med Rep. 5:1043–1048. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Henry W, Querfurth HW and LaFerla FM: Mechanisms of disease Alzheimer's disease. New Engl J Med. 362:329–344. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Zuroff L, Daley D, Black KL and Koronyo-Hamaoui M: Clearance of cerebral Aβ in Alzheimer's disease: Reassessing the role of microglia and monocytes. Cell Mol Life Sci. 74:2167–2201. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Kepp KP: Bioinorganic chemistry of Alzheimer's disease. Chem Rev. 112:5193–5239. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Patrone C, Eriksson O and Lindholm D: Diabetes drugs and neurological disorders: New views and therapeutic possibilities. Lancet Diabetes Endocrinol. 2:256–262. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Palleria C, Leporini C, Maida F, Succurro E, De Sarro G, Arturi F and Russo E: Potential effects of current drug therapies on cognitive impairment in patients with type 2 diabetes. Front Neuroendocrinol. 42:76–92. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Darsalia V, Olverling A, Larsson M, Mansouri S, Nathanson D, Nyström T, Klein T, Sjöholm Å and Patrone C: Linagliptin enhances neural stem cell proliferation after stroke in type 2 diabetic mice. Regul Pept 190–191. 1–31. 2014.

11 

Nassar NN, Al-Shorbagy MY, Arab HH and Abdallah DM: Saxagliptin: A novel antiparkinsonian approach. Neuropharmacology. 89:308–317. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Matteucci E and Giampietro O: Mechanisms of Neurodegeration in Type 2 diabetes and the neuroprotective potential of dipeptidyl peptidase 4 inhibitors. Curr Med Chem. 22:1573–1581. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Zheng T, Qin L, Chen B, Hu X, Zhang X, Liu Y, Liu H, Qin S, Li G and Li Q: Association of plasma DPP4 activity with mild cognitive impairment in elderly patients with type 2 diabetes: results from the GDMD study in China. Diabetes Care. 39:1594–1601. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Kosaraju J, Murthy V, Khatwal RB, Dubala A, Chinni S, Muthureddy Nataraj SK and Basavan D: Vildagliptin: An anti-diabetes agent ameliorates cognitive deficits and pathology observed in streptozotocin-induced Alzheimer's disease. J Pharm Pharmacol. 65:1773–1784. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Brion JP, Fraser H, Flament-Durand J and Dickinson AG: Amyloid scrapie plaques in mice and Alzheimer senile plaques, share common antigens with tau, a microtubule-associated protein. Neurosci Lett. 78:113–118. 1987. View Article : Google Scholar : PubMed/NCBI

16 

Gonzalez B, Abud EM, Abud AM, Poon WW and Gylys KH: Tau spread, apolipoprotein E, inflammation and more: Rapidly evolving basic science in alzheimer disease. Neurol Clin. 35:175–190. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Storr T: Ligand design in medicinal inorganic chemistry. John Wiley & Sons; New York, NY: 2014, View Article : Google Scholar : PubMed/NCBI

18 

Jakobroetne R and Jacobsen H: Alzheimer's disease: From pathology to therapeutic approaches. Angew Chem Int Ed Enge. 48:3030–3059. 2009. View Article : Google Scholar

19 

Pintana H, Apaijai N, Chattipakorn N and Chattipakorn SC: DPP-4 inhibitors improve cognition and brain mitochondrial function of insulin-resistant rats. J Endocrinol. 218:1–11. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Gault VA, Lennox R and Flatt PR: Sitagliptin, a dipeptidyl peptidase-4 inhibitor, improves recognition memory, oxidative stress and hippocampal neurogenesis and upregulates key genes involved in cognitive decline. Diabetes Obes Metab. 17:403–413. 2015. View Article : Google Scholar : PubMed/NCBI

21 

D'Amico M, Di Filippo C, Marfella R, Abbatecola AM, Ferraraccio F, Rossi F and Paolisso G: Long-term inhibition of dipeptidyl peptidase-4 in Alzheimer's prone mice. Exp Gerontol. 45:202–207. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Pintana H, Tanajak P, Pratchayasakul W, Sa-Nguanmoo P, Chunchai T, Satjaritanun P, Leelarphat L, Chattipakorn N and Chattipakorn SC: Energy restriction combined with dipeptidyl peptidase-4 inhibitor exerts neuroprotection in obese male rats. Br J Nutr. 1–9. 2016.PubMed/NCBI

23 

El Batsh MM, El Batch MM, Shafik NM and Younos IH: Favorable effects of vildagliptin on metabolic and cognitive dysfunctions in streptozotocin-induced diabetic rats. Eur J Pharmacol. 769:297–305. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Gao C, Hölscher C, Liu Y and Li L: GSK3: A key target for the development of novel treatments for type 2 diabetes mellitus and Alzheimer disease. Rev Neurosci. 23:1–11. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Yu T and Lin W: Small-molecule GSK-3 inhibitor rescued apoptosis and neurodegeneration in anesthetics-injured dorsal root ganglion neurons. Biomed Pharmacother. 84:395–402. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Fan R, Li X, Gu X, Chan JC and Xu G: Exendin-4 protects pancreatic beta cells from human islet amyloid polypeptide-induced cell damage: Potential involvement of AKT and mitochondria biogenesis. Diabetes Obes Metab. 12:815–824. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Savelieff MG, Lee S, Liu Y and Lim MH: Untangling amyloid-β, tau, and metals in Alzheimer's disease. Acs Chem Biol. 8:856–865. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Nadezhdin KD, Bocharova OV, Bocharov EV and Arseniev AS: Structural and dynamic study of the transmembrane domain of the amyloid precursor protein. Acta Naturae. 3:69–76. 2011.PubMed/NCBI

29 

Köpke E, Tung YC, Shaikh S, Alonso AC, Iqbal K and Grundke-Iqbal I: Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease. J Biol Chem. 268:24374–24384. 1993.PubMed/NCBI

30 

Braak H, Thal DR, Ghebremedhin E and Del Tredici K: Stages of the pathologic process in Alzheimer disease: Age categories from 1 to 100 years. J Neuropathol Exp Neurol. 70:960–969. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Do Carmo S, Crynen G, Paradis T, Reed J, Iulita MF, Ducatenzeiler A, Crawford F and Cuello AC: Hippocampal proteomic analysis reveals distinct pathway deregulation profiles at early and late stages in a rat model of alzheimer's-like amyloid pathology. Mol Neurobiol. May 13–2017.(Epub ahead of print). View Article : Google Scholar

32 

Pratley RE, Jauffret-Kamel S, Galbreath E and Holmes D: Twelve-week monotherapy with the DPP-4 inhibitor vildagliptin improves glycemic control in subjects with type 2 diabetes. Horm Metab Res. 38:423–428. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 17 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma QH, Jiang LF, Mao JL, Xu WX and Huang M: Vildagliptin prevents cognitive deficits and neuronal apoptosis in a rat model of Alzheimer's disease. Mol Med Rep 17: 4113-4119, 2018
APA
Ma, Q., Jiang, L., Mao, J., Xu, W., & Huang, M. (2018). Vildagliptin prevents cognitive deficits and neuronal apoptosis in a rat model of Alzheimer's disease. Molecular Medicine Reports, 17, 4113-4119. https://doi.org/10.3892/mmr.2017.8289
MLA
Ma, Q., Jiang, L., Mao, J., Xu, W., Huang, M."Vildagliptin prevents cognitive deficits and neuronal apoptosis in a rat model of Alzheimer's disease". Molecular Medicine Reports 17.3 (2018): 4113-4119.
Chicago
Ma, Q., Jiang, L., Mao, J., Xu, W., Huang, M."Vildagliptin prevents cognitive deficits and neuronal apoptosis in a rat model of Alzheimer's disease". Molecular Medicine Reports 17, no. 3 (2018): 4113-4119. https://doi.org/10.3892/mmr.2017.8289