Open Access

Exendin‑4 antagonizes Aβ1‑42‑induced attenuation of spatial learning and memory ability

  • Authors:
    • Xiaohui Wang
    • Li Wang
    • Ruirui Jiang
    • Yunyun Xu
    • Xueling Zhao
    • Yang Li
  • View Affiliations

  • Published online on: September 21, 2016     https://doi.org/10.3892/etm.2016.3742
  • Pages: 2885-2892
  • Copyright: © Wang 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

β-amyloid protein (Aβ) accumulation in cerebral centers involved in cognition and memory is a pivotal pathological feature of Alzheimer's disease (AD). The onset process of type 2 diabetes mellitus (T2DM) has a number of similarities compared with AD. Thus, it is hypothesized that the pharmacological therapy employed for the treatment of T2DM may help to prevent and ameliorate the symptoms of AD. This study demonstrated that Exendin‑4, which is a glucagon‑like peptide‑1 analogue which is used as a therapeutic drug for T2DM, markedly antagonized Aβ fragment‑induced attenuation of spatial learning and memory ability, as indicated by a Morris water maze experiment. In addition, we investigated the potential underlying electrophysiological and molecular mechanisms. The results indicate that Exendin‑4 rescued long‑term potentiation from Aβ1‑42‑induced damage in the rat hippocampal CA1 region in vivo, and antagonized Aβ1‑42‑induced reduction of cyclic adenosine monophosphate and phosphorylated‑cAMP response element‑binding protein in rat hippocampal tissue using ELISA and western blot analysis, respectively. Thus, the results of the present study provide theoretical support for the application of Exendin‑4 for improving AD.

Introduction

With the increase in the aging world population, the prevalence of major neurodegenerative diseases has become a serious public health problem worldwide, particularly Alzheimer's disease (AD). AD is a degenerative, progressive and incurable neurological syndrome accompanied by a gradual decline in learning, memory and intelligence (1). A series of hypotheses have been proposed for the mechanism of AD, and it is now widely accepted that the deposit of amyloid β protein (Aβ) in cerebral centers involved in cognition and memory is the most important pathological feature of AD (2). There are two types of AD; familial and sporadic AD (3). Familial AD is associated with a genetic predisposition, which is associated by the production and deposition of Aβ (4).

Notably, the pathogenesis of another type of degenerative disease, type 2 diabetes mellitus (T2DM), has certain similarities with AD, such as Aβ deposits in the islets of T2DM patients. Further studies are necessary to assess whether therapeutic methods for T2DM could antagonize the neurotoxicity of Aβ peptides and ameliorate symptoms of AD patients. Glucagon-like peptide-1 (GLP-1) is a T2DM novel therapeutic drug has been suggested as a therapeutic target for AD (5). Due to the relatively short half-life of native GLP-1, which is easily degraded by dipeptidyl peptidase-4 (DPP-4), it has been limited in its clinical application (6). Exendin-4 is a GLP-1 analogue which is more slowly degraded, with a half-life of 9.57 h, and has been approved by the Federal Drug Administration as a therapeutic for T2DM (7).

Recently, studies have been published addressing the neuroprotective effect of GLP-1 analogues on neurodegenerative disorders (8,9). Our previous study indicated that Val8-GLP-1, another GLP-1 analog, was able to effectively antagonize synaptic dysfunction and intracellular Ca2+ overload induced by Aβ1–40 (10). This neuroprotection may be associated with the exhaustive regulation of synaptic transmission and intracellular calcium homeostasis by Val8-GLP-1 (11). Val8-GLP-1 prevented synaptic degeneration and reversed hippocampal synaptic plasticity in mice (12). In addition, Exendin-4 appears to increase the expression level of soluble polypeptide by enhancing the activity level of α-secretase of amyloid precursor protein (APP), thus antagonizing Aβ toxicity (13). The above findings suggest that GLP-1 analogs such as Exendin-4 may exert neuroprotective effects by improving synaptic function and antagonizing Aβ toxicity. However, whether Exendin-4 is able to mitigate spatial learning and memory damage resulting from Aβ fragment remains unclear. The present study investigated the effects of Exendin-4 on Aβ1-42-induced spatial learning and memory impairment, and on the overall behavior of rats in order to evaluate any neuroprotective effects induced by Exendin-4, and further investigated the underlying electrophysiological and molecular mechanisms.

Materials and methods

Experimental animal and reagents

A total of 80 adult male Sprague-Dawley rats (220–260 g) were used for the present study. All animals were provided by the Research Animal Center of Shanxi Medical University (Taiyuan, China). The study protocol received the approval of the Shanxi Animal Research Ethics Committee.

Aβ1–42 [Bachem (UK) Ltd, Saint Helens, UK] was dissolved in dimethyl sulfoxide (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) then diluted with normal saline to 6.25×10−4 mol/l and incubated at 37°C for 36 h to form oligomers (14). Exendin-4 (Sigma-Aldrich) was diluted with normal saline to 2×10−4 mol/l. The administration was performed as follows: Intra-hippocampal injection 0.625 nmol (1 µl) Aβ1–42 (Aβ1–42 group); 0.2 nmol (1 µl) Exendin-4 (Exendin-4 group); Exendin-4 + Aβ1–42 group received Exendin-4 injection at 15 min following Aβ1–42 oligomers; and the control group received 1 µl normal saline injection. The intra-hippocampal injection was performed via a microinjection pump (KD Scientific, Inc., Holliston, MA, USA), with an injection rate of 0.1 µl/min. The experiments were then performed 5 days after.

Morris water maze test

The spatial learning and memory test was performed using a Morris water maze (MWM), as described previously (10). The MWM consists of a circular pool with a diameter of 150 cm (Zhenghua Biological Instrument Equipment Co., Ltd., Anhui, China), containing tap water at ~25°C. A underwater platform (14 cm in diameter) was placed under ~1.0 cm below the horizontal plane. At two weeks after drug injection, the hidden platform test was conducted. Animals underwent consecutive six-day trial periods, involving four trials per day using a random set of start locations. The spatial bias of the four quadrants was measured in the probe trial on the seventh day (10). Then the visible platform test was performed in order to measure the time of swimming to the platform (10).

Hippocampal CA1 region long-term potentiation (LTP) in vivo recording

Following the MWM test, the electrophysiological experiment was conducted as described in our previous study (10). In the present study, LTP was evaluated in vivo. The field excitatory postsynaptic potential (fEPSP) of the hippocampal CA1 region was recorded. Baseline fEPSP with test stimuli and LTP with high frequency stimulation (HFS) for at least 60 min were recorded. The averaged value of baseline fEPSP amplitude was taken as 100%.

Bicinchoninic acid (BCA) protein assay

Following the LTP experiment, the rats were sacrificed using an overdose of 25% urethane (5 ml/kg; Sigma-Aldrich) and the hippocampal tissues were dissected and frozen at −80°C. Then the tissues were homogenized with protease lysate, followed by centrifugation at 16,000 × g for 10 min at 4°C, after which the supernatant was collected. The total protein was quantified using a BCA kit (Westang Biotech Co., Ltd., Shanghai, China), according to the manufacturer's instructions. The BCA assay was conducted according to the manufacturer's instructions. Absorbance was recorded at 562 nm using a microplate reader (MK3; Thermo Fisher Scientific, Inc., Waltham, MA, USA). A standard curve was drawn and the total protein concentration was calculated.

Enzyme-linked immunosorbent assay (ELISA)

Subsequently, an ELISA experiment was performed using a commercial kit (cat. no. F15181; Westang Biotech Co., Ltd.) to detect the expression of cyclic adenosine monophosphate (cAMP). The polystyrene ELISA plate was arranged and each group was marked clearly. Next, 100-µl standards of fold dilution were added to 8 wells of ELISA plate successively, then 100-µl samples were added to each well, with mixing and incubating at 37°C for 40 min, then washed 4–6 times and dried. Subsequently 50 µl distilled water and rabbit anti-rat cAMP antibody was added to each well (except the eighth standard well), mixed, incubated at 37°C for 20 min and washed with PBST. Next, 100 µl horse radish peroxidase working solution was added, mixed, incubated at 37°C for 10 min, washed and printed. Next, 100 µl substrate working solution was added at 37°C for 15 min, then subsequently 100 µl stop solution was added. Absorbance was recorded at 450 nm (within 30 min) using a microplate reader (MK3; Thermo Fisher Scientific, Inc.) and used to draw the standard curve. Finally, the content of cAMP was calculated. Absorbance was recorded at 450 nm (within 30 min) using a microplate reader and the content of cAMP was calculated according to the cAMP standard curve that was created with SkanIt Software (Thermo Fisher Scientific, Inc.).

Western blot analysis

The experiment was used to evaluate phosphorylated-cAMP response element binding protein (p-CREB) expression in rat hippocampal tissue. The bilateral hippocampus tissue (100 mg) was lysed in RIPA buffer (Beyotime Institute of Biotechnology, Nanjing, China). Loading sample (40 µg) was selected for separation using 12% SDS-PAGE, then transferred to a PVDF membrane (Whatman; GE Healthcare, Chalfont, UK). The membrane was blocked with 5% non-fat milk powder for 2 h and washed 3 times with 1X TBST buffer for 10 min. Then monoclonal primary antibodies against p-CREB (1:1,000; cat. no. 9198; Cell Signaling Technology, Inc., Danvers, MA, USA) were incubated with the membrane at 4°C overnight. Then the membrane was incubated with the corresponding secondary antibody for 1 h (peroxidase-conjugated goat anti-rat IgG; 1:3,000; cat. no. ZB-2307; Beijing Zhongshan Golden Bridge Biotechnology, Beijing, China). The blot was developed using a super enhanced chemiluminescence detection kit (cat. no. P1030; Applygen Technologies, Inc., Beijing, China), then the membrane was placed in a Kodak Image Station 400 (Kodak Company, Rochester, NY, Japan) for exposure and images of membrane signal bands were obtained. Image J software (imagej.nih.gov/ij/) was used to analyze the western blot bands.

Statistical analysis

Experimental data are presented as the mean ± standard deviation. SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA) was used to perform statistical analysis. One-way analysis of variance and repeated measures analysis of variance were used. Data were considered statistically significant when P<0.05.

Results

Intra-hippocampal injection of Aβ1–42 induced abnormal behavior

The experiment was performed using an MWM test to characterize the changes, if any, in rat spatial learning and memory after intra-hippocampal injection of Aβ1-42. Fig. 1A shows Typical swim trajectories of rats in searching for the hidden platform in all groups in the sixth training day. Fig. 1B-E shows the mean escape latencies and distances in the learning ability test. As shown in Fig. 1, the average escape latencies and distances gradually reduced with the increase of training days, and these indexes remained relatively stable on the fifth day in all groups. The average escape latencies and distances significantly prolonged in the Aβ1–42 group from the second to the sixth training day compared with the control group, although the value of the first day maintained consistent. Fig. 1A shows typical swim trajectories of all groups on the sixth training day.

In order to further investigate the changes in rat spatial memory ability, a probe trial was conducted. Fig. 2A showed typical swim trajectories in all groups in the probe trial. Approximately the same time in the target quadrant and the other quadrants was spent in the Aβ1–42 group; however, the rats in the control group spent more time in the target quadrant. and the time was significantly longer than the other quadrants (Fig. 2B and C).

Exendin-4 mitigated the behavior ability caused by Aβ1–42

The subsequent results showed that there was no significant change of the typical swim trajectories between Exendin-4 and control group in the hidden platform test (Fig. 1A). Firstly, the application of Exendin-4 alone did not change the mean escape latencies and distances on any learning training day (Fig. 1B-E), the time percentage in the target quadrant maintained consistent with control group in probe trial (Fig. 2A-C).

Furthermore, the neuroprotective impact of Exendin-4 on the behavioral ability was investigated. Mean escape latency and distance were decreased after pretreatment with Exendin-4 compared to the Aβ1–42 group in the learning ability test (Fig. 1). In the probe trial, the time in the target quadrant obviously prolonged compared with the Aβ1–42 group (Fig. 2C).

The latency and swimming speed were consistent among all groups during successive six days in the hidden platform and visible platform tests (Fig. 3). These data confirmed that the visual and motion ability of all rats was consistent, and the aforementioned behavioral experimental results indicate the effect of Exendin-4 and Aβ1-42.

LTP was evidently reduced in the Aβ1–42 group

The LTP experiment was conducted following the MWM test. Aβ1–42 significantly suppressed LTP in the rat hippocampus in vivo compared with control group, the average fEPSP amplitudes at 30, 60 and 90 min after HFS showed a significant difference compared to the control group (Fig. 4A and B).

The baseline fEPSP of intra-hippocampal injection of Aβ1–42 was observed. The result indicated that fEPSP without HFS was not significantly different during at least 90 min of recording in the Aβ1–42 group (Fig. 5). The results suggested that Aβ1–42 induced hippocampal LTP damage.

Exendin-4 prevented Aβ1-42-induced impairment of LTP

Similarly, Exendin-4 alone did not impact the baseline fEPSP (Fig. 5). However, Exendin-4 effectively prevented Aβ1-42-induced deficits of LTP. As shown in Fig. 4A and B, Aβ1–42 induced LTP inhibition was significantly antagonized by Exendin-4. The average fEPSP amplitudes at 30, 60 and 90 min post-HFS were significantly higher compared with the Aβ1–42 group.

Exendin-4 antagonized Aβ1–42 induced decrease of cAMP and p-CREB in rat hippocampus tissue

In order to investigate the molecular mechanism of antagonism of Exendin-4 for Aβ1-42, the levels of cAMP and p-CREB in the rat hippocampus were determined. We found that Aβ1-42-induced the cAMP level was significantly reduced compared with the control group, pretreatment of Exendin-4 antagonized the decrease of cAMP induced by Aβ1-42; however, the level of cAMP was not affected by Exendin-4 application alone (Fig. 6).

Subsequently, p-CREB expression in rat hippocampus tissue was determined in all groups. As shown in Fig. 7, Aβ1–42 evidently reduced the protein expression level of p-CREB. p-CREB protein expression markedly improved after pretreatment with Exendin-4 compared with the application of Aβ1–42 alone. These results suggested that pretreatment with Exendin-4 may reverse the decreased p-CREB protein level induced by Aβ1-42. Furthermore, the protein expression level of p-CREB was not affected by Exendin-4 application alone. Thus, the present results imply that Exendin-4 may antagonize Aβ1–42 toxicity by mediating the cAMP/PKA/CREB pathway.

Discussion

In the present study, intra-hippocampal injection of Aβ1–42 was adopted to generate rat models, then behavioral, electrophysiological and molecular experiments were performed. The results imply that the impairment of learning and memory induced by Aβ1–42 was significantly reversed by Exendin-4 treatment. Furthermore, the electrophysiology and molecular mechanisms underlying the neuroprotective effect of Exendin-4 were investigated.

AD is a degenerative, progressive and incurable neurological disorder which is characterized by the gradual deterioration of cognitive function, neuropsychiatric symptoms and behavioral disturbances, including nutritional disorders and circadian rhythm disruption (15,16). There are three major pathological features associated with AD; neuronal loss, senile plaques formed by the deposition of amyloid β protein and neurofibrillary tangles (17,18). However, the exact pathogenesis of AD remains unclear, and thus a series of hypotheses have been proposed to account for the pathogenesis of AD, such as the Aβ hypothesis, cholinergic hypothesis, tau protein hypothesis and oxidative stress hypothesis, among which the Aβ hypothesis plays the most important role (19,20). The Aβ hypothesis suggests that the pathogenesis of various biological malfunctioning is associated with Aβ deposition, for example, Aβ oligomers extracted from human AD brain can inhibit LTP, reduce dendritic spine density and disrupt memory and learning in vivo when directly injected into a mouse hippocampus (21). Aβ is derived from the precursor protein APP, which generates various polypeptides via sequential cleaving by α-secretase and β-secretase (22). Aβ is a insoluble polypeptide which is generated by the β-secretase pathway, and can induce oxidative stress and Ca2+ uptake. Oxidative stress can in turn active apoptosis and initiate glial cells to produce large quantities of inflammatory mediators and other toxic substances, resulting in irreversible neuronal damage (23). Moreover, Ca2+ may be releases from presynaptic neurotransmitters and influx into the postsynaptic membrane, which is closely associated with learning and memory (24).

By contrast, Aβ peptide includes numerous fragments, including Aβ1-40, Aβ1-42, Aβ31–35 and Aβ25–35 (25,26). Among these, Aβ1–42 exhibits the strongest toxicity owing to its hydrophobicity and aggregation (27). Therefore, Aβ1–42 was adopted for the present experiments. Formerly, intra-cerebroventricular injection was widely used to investigated the toxicity of Aβ1–42 in numerous studies (28,29); however, this delivery method has numerous disadvantages, including drug diffusion, inaccurate positioning and relatively high cost (10,30). Intra-hippocampal administration effectively avoided these shortcomings and shortened delivery time significantly in the present study.

Following intra-hippocampal injection, rats underwent an MWM experiment. Over the course of six consecutive days of spatial learning ability testing, the escape latencies and distances of Aβ1–42 group increased significantly compared with the control group. Approximately equal time was spent in the target quadrant compared with any other quadrant in the Aβ1–42 group in the probe trial. It is thus indicated that the spatial learning and memory impairment were induced by Aβ1-42. Consistent with this, inhibitory avoidance analysis (a fear conditioning-based task) in a previous study suggested that Aβ1–42 caused the damage to learning and memory ability (31). Therefore, protection against neurotoxicity of Aβ is hypothesized to be a therapeutic target for the treatment of AD patients. Several studies have indicated approaches to decreasing Aβ neurotoxicity, such as reducing Aβ generation, increasing Aβ degradation and restraining Aβ toxicity (32,33); however, there is at present no efficacious drug directed against Aβ neurotoxicity. The purpose of the present study was to investigate whether Exendin-4 exhibits neuroprotective effects against Aβ peptide neurotoxicity.

A previous study suggested an association between AD and T2DM, which is another chronic degenerative disease (34). An investigation revealed that 85% of AD patients were diagnosed with T2DM or accompanied by elevated fasting blood glucose (35). Furthermore, the risk of patients with T2DM suffering from AD is twice that of the normal population (36). Notably, Aβ deposition, the typical pathological change in AD patients, is similar to the characteristic accumulation of Aβ in the islet cells of patients with T2DM (37). Therefore, researchers have suggested that AD associated with high blood glucose be considered type 3 diabetes mellitus (38). Thus, pharmacological therapy intended for the treatment of T2DM may additionally help to prevent and improve the symptoms of AD. Insulin, the primary therapy for T2DM, is not applicable as an AD treatment, as insulin-degrading enzyme can recognize and combine the same domain which exists in insulin and Aβ (39). Moreover, insulin injection can reduce the combination between Aβ and IDE, facilitate Aβ deposition in brain, and ultimately exacerbate the neurotoxicity and induce clinical symptoms (40). Furthermore, AD patients with normal blood glucose may sustain glucopenia due to the fact that insulin can lower euglycemia (41). However, GLP-1 and its analogs may promote insulin synthesis and secretion, and do not affect normal blood glucose (42). The polypeptide chain of GLP-1 containing 30 amino acids can be decomposed by DPP-4, therefore the plasma half-life of native GLP-1 is <2 min (6,43). GLP-1 analogs are similar to native GLP-1 as they exhibit similar chemical structure and biological activity (44). Fortunately, certain GLP-1 analogs are not easily biodegraded by DPP-4, which is attributed to the fatty acid side chains, so its plasma half-life is longer than that of native GLP-1 and may cross the blood-brain barrier (45). Exendin-4, derived from American Gila monster's (Heloderma suspectum) saliva, is the main GLP-1 analogue approved for clinical application (46). Approximately 53% homology exists between Exendin-4 and mammalian GLP-1, ad Exendin-4 has high affinity to GLP-1 receptor, whose plasma half-life is 9.57 h (47). Studies have found that Exendin-4 protected the primary human SH-SY5Y cells and rat hippocampal neurons against glutamate excitotoxicity (48), as well as abating the cognitive impairment induced by traumatic brain injury (48). However, whether Exendin-4 can mitigate the damage of spatial learning and memory derived associated with Aβ remains unknown. The results of the present study demonstrated that Exendin-4 was associated with a marked reduction in Aβ1-42-induced learning and memory damage in a rat model. In the behavioral experiment, pretreatment of Exendin-4 decreased mean escape latency and distance travelled by the rats, compared with the Aβ1–42 group in the learning test, and the time in the target quadrant obviously prolonged compared with the Aβ1–42 group in the memory trial. In order to investigate the mechanism underlying the neuroprotective effect of Exendin-4, we conducted further electrophysiological and molecular biological experiments.

As an important electrophysiological mechanism of learning and memory, LTP is a long-lasting increase in synaptic efficacy which follows high-frequency stimulation of afferent fibers (49). Our previous study showed that Aβ1-40-induced damage of late phase LTP, learning and memory were antagonized by Val8-GLP-1 (10). In the present study, we performed electrophysiological experiments and proved that Aβ1-42-induced a reduction of LTP could be reversed by Exendin-4. Baseline fEPSP in the CA1 area of hippocampus was unchanged by Aβ1–42 or Exendin-4 alone, which indicates that Exendin-4 regulates learning and memory impairment of AD model mediated by modifying impaired LTP.

Furthermore, synaptic plasticity, neurogenesis, learning and memory are closely associated with cAMP/PKA/CREB signaling pathway in the central nervous system (50,51). CREB is an important nucleoprotein belonging to activating transcription factor and CREB proteins are activated by phosphorylation at Ser133 (p-CREB) from various kinases that are activated by the increased intracellular cAMP (52). A previous study showed that cAMP/PKA/CREB signaling pathway had a significant effect on memory formation, particularly hippocampal-dependent LTP (53). Increased cAMP, activated MAPK and p-CREB during rapid eye movement sleep ultimately contributed to memory consolidation (54). In addition, a previous study has confirmed that Aβ25-35-treated rats displayed decreased levels of p-CREB (55). The results of the present study indicate that Exendin-4 could reverse Aβ1-42-induced decline of cAMP and p-CREB, which suggested that Exendin-4 ameliorates Aβ1-42-induced damage to learning and memory, potentially by modifying the cAMP/PKA/CREB signaling pathway.

In conclusion, the study demonstrated that Exendin-4 may exert a neuroprotective effect in the experimental rats, and further revealed possible underlying electrophysiological and molecular mechanisms. These findings provide theoretical support for further investigation into the use of Exendin-4 as a drug for the prophylaxis and treatment of AD.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (grant no. 81471343) and Science and Technology Innovation Fund of Shanxi Medical University (grant no. 01201307).

References

1 

Cummings JL: Alzheimer's disease. N Engl J Med. 351:56–67. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Hardy J and Allsop D: Amyloid deposition as the central event in the aetiology of Alzheimer's disease. Trends Pharmacol Sci. 12:383–388. 1991. View Article : Google Scholar : PubMed/NCBI

3 

Hoenicka J: Genes in Alzheimer's disease. Rev Neurol. 42:302–305. 2006.(In Spanish). PubMed/NCBI

4 

Bohm C, Chen F, Sevalle J, Qamar S, Dodd R, Li Y, Schmitt-Ulms G, Fraser PE and St George-Hyslop PH: Current and future implications of basic and translational research on amyloid-β peptide production and removal pathways. Mol Cell Neurosci. 66:3–11. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Ohtake N, Saito M, Eto M and Seki K: Exendin-4 promotes the membrane trafficking of the AMPA receptor GluR1 subunit and ADAM10 in the mouse neocortex. Regul Pept 190–191. 1–11. 2014. View Article : Google Scholar

6 

Vilsbøll T, Agersø H, Krarup T and Holst JJ: Similar elimination rates of glucagon-like peptide-1 in obese type 2 diabetic patients and healthy subjects. J Clin Endocrinol Metab. 88:220–224. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Lupi R, Mancarella R, Del Guerra S, Bugliani M, Del Prato S, Boggi U, Mosca F, Filipponi F and Marchetti P: Effects of exendin-4 on islets from type 2 diabetes patients. Diabetes Obes Metab. 10:515–519. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Bassil F, Fernagut PO, Bezard E and Meissner WG: Insulin, IGF-1 and GLP-1 signaling in neurodegenerative disorders: targets for disease modification? Prog Neurobiol. 118:1–18. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Calsolaro V and Edison P: Novel GLP-1 (glucagon-like peptide-1) analogues and insulin in the treatment for Alzheimer's disease and other neurodegenerative diseases. CNS Drugs. 29:1023–1039. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Wang XH, Li L, Hölscher C, Pan YF, Chen XR and Qi JS: Val8-glucagon-like peptide-1 protects against Aβ1-40-induced impairment of hippocampal late-phase long-term potentiation and spatial learning in rats. Neuroscience. 170:1239–1248. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Wang XH, Yang W, Hölscher C, Wang ZJ, Cai HY, Li QS and Qi JS: Val8-GLP-1 remodels synaptic activity and intracellular calcium homeostasis impaired by amyloid β peptide in rats. J Neurosci Res. 91:568–577. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Gengler S, McClean PL, McCurtin R, Gault VA and Hölscher C: Val (8)GLP-1 rescues synaptic plasticity and reduces dense core plaques in APP/PS1 mice. Neurobiol Aging. 33:265–276. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Xie Z and Xu Z: General anesthetics and β-amyloid protein. Prog Neuropsychopharmacol Biol Psychiatry. 47:140–146. 2013. View Article : Google Scholar : PubMed/NCBI

14 

O'Hare E, Weldon DT, Mantyh PW, Ghilardi JR, Finke MP, Kuskowski MA, Maggio JE, Shephard RA and Cleary J: Delayed behavioral effects following intrahippocampal injection of aggregated A beta (1–42). Brain Res. 815:1–10. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Querfurth HW and LaFerla FM: Alzheimer's disease. N Engl J Med. 362:329–344. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Chiu MJ, Chen TF, Yip PK, Hua MS and Tang LY: Behavioral and psychologic symptoms in different types of dementia. J Formos Med Assoc. 105:556–562. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Sorrentino G and Bonavita V: Neurodegeneration and Alzheimer's disease: The lesson from tauopathies. Neurol Sci. 28:63–71. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Blennow K, de Leon MJ and Zetterberg H: Alzheimer's disease. Lancet. 368:387–403. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Nitta A, Fukuta T, Hasegawa T and Nabeshima T: Continuous infusion of beta-amyloid protein into the rat cerebral ventricle induces learning impairment and neuronal and morphological degeneration. Jpn J Pharmacol. 73:51–57. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Robbins TW, McAlonan G, Muir JL and Everitt BJ: Cognitive enhancers in theory and practice: Studies of the cholinergic hypothesis of cognitive deficits in Alzheimer's disease. Behav Brain Res. 83:15–23. 1997. View Article : Google Scholar : PubMed/NCBI

21 

Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, Lemere CA, et al: Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat Med. 14:837–842. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Glenner GG and Wong CW: Alzheimer's disease: Initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun. 120:885–890. 1984. View Article : Google Scholar : PubMed/NCBI

23 

Guan ZZ: Cross-talk between oxidative stress and modifications of cholinergic and glutaminergic receptors in the pathogenesis of Alzheimer's disease. Acta Pharmacol Sin. 29:773–780. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Chakroborty S, Kim J, Schneider C, Jacobson C, Molgó J and Stutzmann GE: Early presynaptic and postsynaptic calcium signaling abnormalities mask underlying synaptic depression in presymptomatic Alzheimer's disease mice. J Neurosci. 32:8341–8353. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Clementi ME, Marini S, Coletta M, Orsini F, Giardina B and Misiti F: Abeta(31–35) and Abeta(25–35) fragments of amyloid beta-protein induce cellular death through apoptotic signals: Role of the redox state of methionine-35. FEBS Lett. 579:2913–2918. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Kanai M, Matsubara E, Isoe K, Urakami K, Nakashima K, Arai H, Sasaki H, Abe K, Iwatsubo T, Kosaka T, et al: Longitudinal study of cerebrospinal fluid levels of tau, A beta1-40, and A beta1-42(43) in Alzheimer's disease: a study in Japan. Ann Neurol. 44:17–26. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, Morgan TE, Rozovsky I, Trommer B, Viola KL, et al: Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci USA. 95:6448–6453. 1998. View Article : Google Scholar : PubMed/NCBI

28 

Bernardi A, Frozza RL, Meneghetti A, Hoppe JB, Battastini AM, Pohlmann AR, Guterres SS and Salbego CG: Indomethacin-loaded lipid-core nanocapsules reduce the damage triggered by Aβ1-42 in Alzheimer's disease models. Int J Nanomedicine. 7:4927–4942. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Yamada K, Tanaka T, Han D, Senzaki K, Kameyama T and Nabeshima T: Protective effects of idebenone and alpha-tocopherol on beta-amyloid-(1–42)-induced learning and memory deficits in rats: implication of oxidative stress in beta-amyloid-induced neurotoxicity in vivo. Eur J Neurosci. 11:83–90. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Kheirbakhsh R, Chinisaz M, Khodayari S, Amanpour S, Dehpour AR, Muhammadnejad A, Larijani B and Ebrahim-Habibi A: Injection of insulin amyloid fibrils in the hippocampus of male Wistar rats: Report on memory impairment and formation of amyloid plaques. Neurol Sci. 36:1411–1416. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Garcia-Osta A and Alberini CM: Amyloid beta mediates memory formation. Learn Mem. 16:267–272. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Nalivaeva NN, Beckett C, Belyaev ND and Turner AJ: Are amyloid-degrading enzymes viable therapeutic targets in Alzheimer's disease? J Neurochem. 120:(Suppl 1). S167–S185. 2012. View Article : Google Scholar

33 

Soininen H and Hiltunen M: Targeting ApoE4/ApoE receptor LRP1 in Alzheimer's disease. Expert Opin Ther Targets. 17:781–794. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Jones A, Kulozik P, Ostertag A and Herzig S: Common pathological processes and transcriptional pathways in Alzheimer's disease and type 2 diabetes. J Alzheimers Dis. 16:787–808. 2009.PubMed/NCBI

35 

Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E, O'Brien PC and Palumbo PJ: Risk of dementia among persons with diabetes mellitus: A population-based cohort study. Am J Epidemiol. 145:301–308. 1997. View Article : Google Scholar : PubMed/NCBI

36 

Janson J, Laedtke T, Parisi JE, O'Brien P, Petersen RC and Butler PC: Increased risk of type 2 diabetes in Alzheimer disease. Diabetes. 53:474–481. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Alafuzoff I, Aho L, Helisalmi S, Mannermaa A and Soininen H: Beta-amyloid deposition in brains of subjects with diabetes. Neuropathol Appl Neurobiol. 35:60–68. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Accardi G, Caruso C, Colonna-Romano G, Camarda C, Monastero R and Candore G: Can Alzheimer disease be a form of type 3 diabetes? Rejuvenation Res. 15:217–221. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Farris W, Mansourian S, Chang Y, Lindsley L, Eckman EA, Frosch MP, Eckman CB, Tanzi RE, Selkoe DJ and Guenette S: Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo. Proc Natl Acad Sci. 100:4162–4167. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Frank-Cannon TC, Alto LT, McAlpine FE and Tansey MG: Does neuroinflammation fan the flame in neurodegenerative diseases? Mol Neurodegener. 4:472009. View Article : Google Scholar : PubMed/NCBI

41 

Kahn SE, Hull RL and Utzschneider KM: Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 444:840–846. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Nadkarni P, Chepurny OG and Holz GG: Regulation of glucose homeostasis by GLP-1. Prog Mol Biol Transl Sci. 121:23–65. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Baggio LL and Drucker DJ: Biology of incretins: GLP-1 and GIP. Gastroenterology. 132:2131–2157. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Manandhar B and Ahn JM: Glucagon-like peptide-1 (GLP-1) analogs: Recent advances, new possibilities, and therapeutic implications. J Med Chem. 58:1020–1037. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Hunter K and Hölscher C: Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 13:332012. View Article : Google Scholar : PubMed/NCBI

46 

Shirazi RH, Dickson SL and Skibicka KP: Gut peptide GLP-1 and its analogue, Exendin-4, decrease alcohol intake and reward. PLoS One. 8:e619652013. View Article : Google Scholar : PubMed/NCBI

47 

Kiesewetter DO, Gao H, Ma Y, Niu G, Quan Q, Guo N and Chen X: 18F-radiolabeled analogs of exendin-4 for PET imaging of GLP-1 in insulinoma. Eur J Nucl Med Mol Imaging. 39:463–473. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Eakin K, Li Y, Chiang YH, Hoffer BJ, Rosenheim H, Greig NH and Miller JP: Exendin-4 ameliorates traumatic brain injury-induced cognitive impairment in rats. PLoS One. 8:e820162013. View Article : Google Scholar : PubMed/NCBI

49 

Baudry M, Zhu G, Liu Y, Wang Y, Briz V and Bi X: Multiple cellular cascades participate in long-term potentiation and in hippocampus-dependent learning. Brain Res. 1621:73–81. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Li YF, Cheng YF, Huang Y, Conti M, Wilson SP, O'Donnell JM and Zhang HT: Phosphodiesterase-4D knock-out and RNA interference-mediated knock-down enhance memory and increase hippocampal neurogenesis via increased cAMP signaling. J Neurosci. 31:172–183. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Brightwell JJ, Smith CA, Neve RL and Colombo PJ: Long-term memory for place learning is facilitated by expression of cAMP response element-binding protein in the dorsal hippocampus. Learn Mem. 14:195–199. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Gonzalez GA and Montminy MR: Cyclic AMP stimulates somatostatin gene transcription by phosphorylation of CREB at serine 133. Cell. 59:675–680. 1989. View Article : Google Scholar : PubMed/NCBI

53 

Xiong WX, Zhou GX, Wang B, Xue ZG, Wang L, Sun HC and Ge SJ: Impaired spatial learning and memory after sevoflurane-nitrous oxide anesthesia in aged rats is associated with down-regulated cAMP/CREB signaling. PLoS One. 8:e794082013. View Article : Google Scholar : PubMed/NCBI

54 

Luo J, Phan TX, Yang Y, Garelick MG and Storm DR: Increases in cAMP, MAPK activity and CREB phosphorylation during REM sleep: Implications for REM sleep and memory consolidation. J Neurosci. 33:6460–6468. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Wang C, Yang XM, Zhuo YY, Zhou H, Lin HB, Cheng YF, Xu JP and Zhang HT: The phosphodiesterase-4 inhibitor rolipram reverses Aβ-induced cognitive impairment and neuroinflammatory and apoptotic responses in rats. Int J Neuropsychopharmacol. 15:749–766. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 12 Issue 5

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
Wang X, Wang L, Jiang R, Xu Y, Zhao X and Li Y: Exendin‑4 antagonizes Aβ1‑42‑induced attenuation of spatial learning and memory ability. Exp Ther Med 12: 2885-2892, 2016
APA
Wang, X., Wang, L., Jiang, R., Xu, Y., Zhao, X., & Li, Y. (2016). Exendin‑4 antagonizes Aβ1‑42‑induced attenuation of spatial learning and memory ability. Experimental and Therapeutic Medicine, 12, 2885-2892. https://doi.org/10.3892/etm.2016.3742
MLA
Wang, X., Wang, L., Jiang, R., Xu, Y., Zhao, X., Li, Y."Exendin‑4 antagonizes Aβ1‑42‑induced attenuation of spatial learning and memory ability". Experimental and Therapeutic Medicine 12.5 (2016): 2885-2892.
Chicago
Wang, X., Wang, L., Jiang, R., Xu, Y., Zhao, X., Li, Y."Exendin‑4 antagonizes Aβ1‑42‑induced attenuation of spatial learning and memory ability". Experimental and Therapeutic Medicine 12, no. 5 (2016): 2885-2892. https://doi.org/10.3892/etm.2016.3742