Articles Service
Research article
Amelioration of Astrocytic Dysfunction via AQP4/LRP1 Pathway by Zizania latifolia and Tricin in C6 Cells Exposed to Amyloid β and High-Dose Insulin and in Mice Treated with Scopolamine
1Department of Food Science and Technology, Keimyung University, Daegu 42601, Republic of Korea
2Department of Applied Chemistry, Kumoh National Institute of Technology, Gumi 39177, Republic of Korea
J. Microbiol. Biotechnol. 2025. 35: e2412026
Published February 28, 2025 https://doi.org/10.4014/jmb.2412.12026
Copyright © The Korean Society for Microbiology and Biotechnology.
Abstract
Keywords
Graphical Abstract

Introduction
Astrocytes are the predominant glial cells within the central nervous system (CNS), playing a crucial role in maintaining brain homeostasis. They are integral to metabolic support, nutritional provision, ion and neuro-transmitter balance, blood-brain barrier regulation, and CNS defense mechanisms. Given these homeostatic responsibilities, astrocytes are intimately involved in the pathophysiology of neurological disorders. The balance between neuronal damage, neuroprotection, and regeneration critically influences the progression and outcomes of these diseases. Brain injuries, whether acute or chronic, elicit a specialized glial response known as reactive astrogliosis, characterized by significant morphological and functional changes in astrocytes.
Alzheimer’s disease (AD), an age-associated neurodegenerative condition, is defined by the accumulation of extracellular fibrillar amyloid β (Aβ) plaques and intracellular neurofibrillary tangles of hyperphosphorylated tau protein [1]. Pathological changes in astrocytes during Alzheimer’s disease (AD) highlight distinct temporal patterns. In the later stages of AD, astrogliosis becomes prominent, with reactive astrocytes closely associated with senile plaque formation. Supporting this,
Recent studies have underscored the significant role of astrocytes and brain-derived neurotrophic factor (BDNF) in the development of Alzheimer's disease (AD). BDNF, a crucial neurotrophin, supports neuronal survival and function, and it is essential for synaptic plasticity, which is vital for learning and memory processes. In patients with AD, consistently lower levels of BDNF have been observed, correlating strongly with the severity of cognitive decline [16, 17]. Consequently, therapeutic strategies aimed at preserving or enhancing BDNF levels are being actively explored as potential interventions for AD.
In addition to BDNF, aquaporin-4 (AQP4) has emerged as a critical factor in maintaining normal astrocyte function, as evidenced by recent
Materials and Methods
Reagents
MTT (3-(4,5-dimethylthiazol-2-yl)2-,5-diphenyltetrazolium bromide), and insulin were purchased from Sigma-Aldrich (USA). Antibody against AQP4 (#59678), β-actin (#3700), GFAP (#80788), LRP1 (#64099), horseradish peroxidase-linked anti-mouse secondary antibody (#7076), horseradish peroxidase-linked anti-rabbit secondary antibody (#7074) were purchased from Cell Signaling Technology (USA). Antibody against BDNF (ab108319), matrix metalloproteinase (MMP)-2 (ab92536), MMP-9 (ab76003), and Aβ1-42 (Aβ1-42, ab120301) were purchased from Abcam Biotechnology (UK). IDE (sc-393887) was purchased from Santa Cruz Biotechnology (USA). Fetal bovine serum (FBS), Dulbecco’s modified Eagle’s medium (DMEM) were purchased from the American Type Culture Collection (USA).
Cell Culture
C6 cells were purchased from American Type Culture Collection. Cells were cultured in DMEM supplemented with penicillin (120 units/ml), streptomycin (120 units/ml), and 10% FBS in a 5% CO2 atmosphere at 37°C.
Animals
Balb/cJ mice (11 weeks old, male) were purchased from Hana Bio (Republic of Korea), and were used as the experimental animals after undergoing quarantine and acclimatization for a week at the Dongnam Medical Research Institute Animal Company (Animal Facility Registration Certificate: No. 412). During breeding, the lighting time was set to a 12-h cycle, and food and water were freely available. Group separation and treatment of experimental animals were designed and performed as presented in Table 1. This study was conducted by the Animal Experimental Ethics Committee of Dongnam Medical Research Institute (No. SEMI-21-003). Briefly, scopolamine (3 mg/kg, i.p.) was administered daily along with donepezil (0.5 mg/kg, positive control) for 7 days, after which the experimental animals were euthanized with CO2.
-
Table 1 . Experimental design.
Group Mice Treatment CON 7 Saline (i.p) + saline (p.o) SCO 7 Scopolamine 3 mg/kg (i.p) + saline (p.o) DON 7 Scopolamine 3 mg/kg (i.p) + donepezil 5 mg/kg (p.o) ZLE100 7 Scopolamine 3 mg/kg (i.p) + ZLE1) 100 mg/kg (p.o) ZLE300 7 Scopolamine 3 mg/kg (i.p) + ZLE1) 300 mg/kg (p.o) Tricin 7 Scopolamine 3 mg/kg (i.p) + tricin 0.3 mg/kg (p.o) 1)ZLE,
Zizania latifolia extract
Contents of Acetylcholine (ACh) and Acetylcholine Esterase (AChE)
The brain tissue was harvested and homogenized by adding 0.1 M PBS at a ratio of 1:10, respectively. After centrifugation at 10,000 ×
Hematoxylin and Eosin (H&E) Staining
The mice were sacrificed and brain tissues were harvested for further examination. Each tissue was fixed in formalin solution (4% paraformaldehyde in PBS). Then, the tissues were successively dehydrated in ethanol and xylene, followed by paraffin embedding. Each paraffin block contained a whole-brain sample, and was coronally sectioned to 5 μm thickness using a microtome (RM-2125 RT, Leica, Germany). The sliced sections were stained with H&E and mounted, followed by imaging using an optical microscope (Eclipse 80i, Nikon, Japan).
Western Blot
C6 cells were seeded into a 6-well cell culture plate (1 × 105 cells per well, cell passage; #75) for 24 h, and then treated with ZLE (50, 500 μg/ml) or tricin (1 μg/ml) with insulin (100 μg/ml) and aggregated Aβ1-42 (5 μM) for 48 h. C6 cells were lysed in a membrane protein extraction kit (Thermo Fisher Scientific, Inc., USA) or RIPA lysis buffer. In the
Statistical Analysis
All experiments were repeated at least three times, and each experiment was performed in triplicate. Results are presented as means ± standard deviations (SD). A significant difference from the respective control for each experimental test condition was assessed using Student’s
Results
Effects of ZLE or Tricin on Astrocyte Proteins Expression in Aβ Plus High-Dose Insulin-Treated C6 Cells
First, to investigate the effect of ZLE or tricin on astrocyte dysfunction, the changes in the protein levels of AQP4, GFAP, and BDNF were investigated using Aβ plus high-dose insulin-treated C6 cells. Astrocytic condition of C6 cells for normal control were confirmed by the expression of GFAP and AQP4, together with the normal expression level of BDNF (Fig. 1, left panel, lane 1). Exposure of the cells to high-dose insulin (100 μg/ml) significantly suppressed the protein expressions of AQP4, GFAP, and BDNF in the C6 cells (Fig. 1, lane 2), and treatment with Aβ1-42 further inhibited these expressions (Fig. 1, lane 3). These suppressed expressions of AQP4, GFAP, and BDNF were reversed by the treatment of ZLE (500 μg/ml) about 1.65-, 3.19-, and 2.26-folds, respectively (Fig. 1, lane 5). Tricin (1 μg/ml) also up-regulated these protein expressions about 1.64-, 3.20-, 2.32-folds, respectively (Fig. 1, lane 6).
-
Fig. 1. Effects of ZLE and tricin on AQP4, GFAP, and BDNF protein expression in C6 cells treated with highdose insulin and Aβ1-42. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. untreated cell and *p < 0.05, *p < 0.005, ***p < 0.001 vs. insulin and Aβ-treated control.
We next examined the effects of the samples on the LRP1/MMPs-mediated Aβ clearance pathway in Aβ plus high-dose insulin-treated C6 cells. The LRP1 expression in astrocytes plays strategic roles in brain Aβ degradation [11], and LRP1 regulates expressions of the Aβ degradation enzymes MMP-2 and -9, eventually alleviating the accumulation of Aβ. Treatment the cells with Aβ plus high-dose insulin almost completely blocked the expressions of LRP1 and MMPs (Fig. 2, left panel, lane 3). The protein expressions of LRP1, MMP-2, and MMP-9 were significantly up-regulated about 2.30-, 32.07-, 3.74-folds, respectively, by ZLE (500 μg/ml, Fig. 2, lane 5), and 2.37-, 47.79-, and 12.37-folds, respectably, by tricin (1 μg/ml, Fig. 2, lane 6). These results suggest that ZLE and tricin could protect brain cells such as astrocytes against damage induced by high-dose insulin and/or Aβ, results in prevention of pathological aging of the brain as well as neurodegenerative disease, including AD.
-
Fig. 2. Effects of ZLE and tricin on LRP1, MMP-2, and MMP-9 protein expression in C6 cells treated with high-dose insulin and Aβ1-42. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. untreated cell and *p < 0.005, ***p < 0.001 vs. insulin and Aβ-treated control.
Effects of ZLE or Tricin on ACh and AChE in Brain Tissues of Scopolamine-Treated Mice
To check that the administration of ZLE or tricin regulated acetylcholine (ACh) and acetylcholinesterase (AChE) in scopolamine-treated mice, ACh level and AChE activity were measured in mice brain tissues. As shown in Fig. 3A, the level of ACh in scopolamine-treated mice was significantly decreased (
-
Fig. 3. Effects of ZLE and tricin on ACh concentration and AChE activity in the hippocampus of scopolamine-treated mice. (A) ACh concentration ; (B) AChE activity. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. normal group and *p < 0.05, *p < 0.005, ***p < 0.001 vs. scopolamine-treated group. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine-treated control group, PC: donepezil-treated positive control group, Z100: ZLE 100 mg/kg treated group, Z300: ZLE 300 mg/kg treated group, T: tricin 0.3 mg/kg treated group.
Effect of ZLE or Tricin on Histopathological Changes of Scopolamine-Treated Mice
The results of the H&E staining for histopathological examination are shown in Fig. 4. The hippocampal neurons of the normal group exhibited well-arranged and a clear structure compared to the scopolamine-treated group. A clear pathological change was observed in the hippocampus of scopolamine-treated negative control group mice. The irregular or altered morphology was clearly prevented by the treatment with ZLE (100, 300 mg/kg) or tricin (0.3 mg/kg; corresponding of 300 mg/kg administration of ZLE group), suggesting the protective effect of ZLE- or tricin-administration against neuronal damage induced by scopolamine.
-
Fig. 4. Histological examination by H&E staining in the hippocampus of scopolamine-treated mice administrated ZLE and tricin. Histological changes were observed at x40 (upper panels) and x100 (lower panels). Black arrows indicate brain tissue necrosis. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine (3 mg/kg) treated control group, PC: scopolamine (3 mg/kg)+donepezil (5 mg/kg) treated positive control group, Z100: scopolamine (3 mg/kg)+ZLE (100 mg/kg) treated group, Z300: scopolamine (3 mg/kg)+ZLE (300 mg/kg) treated group, T: scopolamine (3 mg/kg)+tricin (0.3 mg/kg) treated group.
Effect of ZLE or Tricin on Astrocyte Proteins Expression in Brain Tissues of Scopolamine-Treated Mice
The levels of AQP4, GFAP, and BDNF in hippocampus of scopolamine-treated mice were investigated by Western blot analysis. The expressions of these proteins were dramatically decreased in the scopolamine-treated control group (Fig. 5, left panel lane 2). However, the administration of ZLE at 300 mg/kg or tricin restored these proteins expression compared to the scopolamine-treated mice in the negative control group (
-
Fig. 5. Effects of ZLE and tricin on AQP4, GFAP, and BDNF protein expression in scopolamine-treated mice. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. normal group and *p < 0.05, ***p < 0.001 vs. scopolamine-treated group. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine-treated control group, PC: donepezil-treated positive control group, Z100: ZLE 100 mg/kg treated group, Z300: ZLE 300 mg/kg treated group, T: tricin 0.3 mg/kg treated group.
We next investigated the effects of ZLE or tricin on Aβ clearance pathway, such as LRP1 and Aβ degrading enzymes in scopolamine-treated mice. As shown in Fig. 6, the expression levels of Aβ uptake protein LRP1 and Aβ degradation enzymes (IDE, MMP-2, MMP-9) were suppressed in scopolamine-treated mice, but it was confirmed that these expressions were clearly increased in ZLE (300 mg/kg)- and tricin (0.3 mg/kg)-administrated mice as well as donepezil-administered control group compared with scopolamine-treated mice. On the other hand, the increased expression of ApoE4 protein induced by scopolamine treatment was reduced following sample administration, with a particularly significant reduction observed with Z300 and tricin treatment. These results suggest that in the scopolamine-induced amnesic condition, the generation of apoE4 fragments was promoted, potentially leading to amyloid-beta accumulation, which indicates impaired amyloid clearance.
-
Fig. 6. Effects of ZLE and tricin on Aβ1-42, ApoE4, IDE, LRP1, MMP-2, and MMP-9 protein expression in scopolamine-treated mice. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. normal group and *p < 0.05, *p < 0.005, ***p < 0.001 vs. scopolamine-treated group. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine-treated control group, PC: donepezil-treated positive control group, Z100: ZLE 100 mg/kg treated group, Z300: ZLE 300 mg/kg treated group, T: tricin 0.3 mg/kg treated group.
Discussion
This study aimed to assess the effects of ZLE and its active compound, tricin, on astrocyte dysregulation induced by high-dose insulin combined with Aβ1-42 in C6 astroglial cells. The C6 cell line, which shares characteristics with astrocytes, is commonly utilized
AQP4 plays a pivotal role in the degradation of Aβ and the uptake of glutamate in astrocytes, with its deficiency being strongly associated with cognitive impairments. Research has demonstrated that mice lacking the AQP4 gene exhibit increased deposition of Aβ plaques in the brain, accompanied by memory deficits [3, 4]. In our study, we observed that treatment with ZLE or its major compound, tricin, exerted a protective effect against astroglial dysfunction in C6 cells exposed to high-dose insulin and Aβ1-42. Specifically, we found that the upregulation of astroglial markers GFAP and AQP4 following treatment with ZLE or tricin may help mitigate deficits in the Aβ clearance pathway, as evidenced by the enhanced expression of LRP1 and MMPs, which are critical in the pathogenesis of AD. Tricin, a flavonoid present in the Gramineae family, was identified as the primary active compound in ZLE responsible for its protective effects, including the prevention of allergic responses. Prior research has identified five derivatives of tricin (4',5,7-trihydroxy-3',5'-dimethoxyflavone) in the methanol extract derived from the aerial parts of
In our previous research, we observed that the expression levels of astrocytic markers GFAP and AQP4 increased with successive cell passages. Consequently, for optimal protein expression, we selected cells at passage numbers 75 to 85. To simulate astrodegenerative conditions characterized by reduced GFAP and AQP4 expression, as reported in earlier studies [27], we treated the astroglial cells with a combination of high-dose insulin and Aβ1-42. The downregulation of GFAP and AQP4 in astrocytes may indicate dysfunction, and our findings suggest that ZLE and tricin have the potential to counteract AD progression by preserving astrocyte function, particularly within the Aβ clearance system. Our data demonstrated that ZLE and tricin effectively protected astrocytes from deregulation induced by high-dose insulin and Aβ1-42, leading to the upregulation of key proteins essential for astrocyte function (Fig. 1) and Aβ degradation (Fig. 2). Additionally, existing studies have established that astrocytic low-density lipoprotein receptor-related protein 1 (LRP1) is crucial for Aβ clearance in the brain [11, 12]. Knockdown of LRP1 in primary astrocytes has been shown to reduce cellular Aβ uptake and degradation, while also downregulating several major Aβ-degrading enzymes, including MMP-2, MMP-9, and IDE, further emphasizing the importance of LRP1 in maintaining Aβ homeostasis [44].
Additionally, we investigated the impact of ZLE and tricin on the expression of astrocyte marker proteins and the Aβ-degrading pathway in a scopolamine-induced mouse model. Scopolamine, a muscarinic acetylcholine receptor antagonist, disrupts cholinergic neurotransmission, leading to cognitive and memory impairments in animal models. These impairments mimic the memory dysfunction observed in Alzheimer's disease patients, making scopolamine a widely used agent for studying neurodegenerative processes and potential therapeutic interventions [45]. Scopolamine-induced elevation of AChE activity is linked to a reduction in ACh levels, a process that can result in memory deficits and is implicated in the development of AD pathology [46]. In this study, the administration of
In conclusion, this study demonstrated the protective effects of ZLE and tricin on astrocyte function within an
Acknowledgment
This research was supported by the Bisa Research Grant of Keimyung University in 2023 (Project No: 20230313).
Author Contributions
Seun-Ah Yang: Conceptualization; Funding acquisition; Methodology; Visualization; Writing - original draft; Writing - review & editing. Se-Ho Park: Data curation; Formal analysis; Investigation; Validation; Visualization; Writing – original draft. Eun-Hye Kim: Investigation; Validation. Won-Bin Bae : Investigation; Validation. Kwang-Hwan Jhee : Validation; Writing – review & editing. All authors have read and agreed to the published version of this manuscript.
Conflict of Interest
The authors have no financial conflicts of interest to declare.
References
- Rodriguez J, Olabarria M, Chvatal A, Verkhratsky A. 2009. Astroglia in dementia and Alzheimer's disease.
Cell Death Difer. 16 : 378-385. - Hou L, Liu Y, Wang X, Ma H, He J, Zhang Y,
et al . 2011. The effects of amyloid-β 42 oligomer on the proliferation and activation of astrocytesin vitro .In vitro Cell. Dev. Biol. Anim. 47 : 573-580. - Beauquis J, Pavía P, Pomilio C, Vinuesa A, Podlutskaya N, Galvan V,
et al . 2013. Environmental enrichment prevents astroglial pathological changes in the hippocampus of APP transgenic mice, model of Alzheimer's disease.Exp. Neurol. 239 : 28-37. - Beauquis J, Vinuesa A, Pomilio C, Pavía P, Galván V, Saravia F. 2014. Neuronal and glial alterations, increased anxiety, and cognitive impairment before hippocampal amyloid deposition in PDAPP mice, model of Alzheimer's disease.
Hippocampus 24 : 257-269. - Braun K, Antemano R, Helmeke C, Büchner M, Poeggel G. 2009. Juvenile separation stress induces rapid region-and layer-specific changes in S100β-and glial fibrillary acidic protein-immunoreactivity in astrocytes of the rodent medial prefrontal cortex.
Neurosci. 160 : 629-638. - Rajkowska G, A Stockmeier C. 2013. Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue.
Curr. Drug Targets 14 : 1225-1236. - Xu Z, Xiao N, Chen Y, Huang H, Marshall C, Gao J,
et al . 2015. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Aβ accumulation and memory deficits.Mol. Neurodegener. 10 : 58. - Lan YL, Zhao J, Ma T, Li S. 2016. The potential roles of aquaporin 4 in Alzheimer's disease.
Mol. Neurobiol. 53 : 5300-5309. - Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA,
et al . 2012. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β.Sci. Transl. Med. 4 : 147ra111. - Verghese JP, Terry A, de Natale ER, Politis M. 2022. Research evidence of the role of the glymphatic system and its potential pharmacological modulation in neurodegenerative diseases.
J. Clin. Med. 11 : 6964. - Ries M, Sastre M. 2016. Mechanisms of Aβ clearance and degradation by glial cells.
Front. Aging Neurosci. 8 : 160. - Kanekiyo T, Liu CC, Shinohara M, Li J, Bu G. 2012. LRP1 in brain vascular smooth muscle cells mediates local clearance of Alzheimer's amyloid-β.
J. Neurosci. 32 : 16458-16465. - Eckman EA, Reed DK, Eckman CB. 2001. Degradation of the Alzheimer's amyloid β peptide by endothelin-converting enzyme.
J. Biol. Chem. 276 : 24540-24548. - Bernstein HG, Lendeckel U, Bukowska A, Ansorge S, Ernst T, Stauch R,
et al . 2008. Regional and cellular distribution patterns of insulin-degrading enzyme in the adult human brain and pituitary.J. Chem. Neuroanat. 35 : 216-224. - Hashimoto T, Serrano-Pozo A, Hori Y, Adams KW, Takeda S, Banerji AO,
et al . 2012. Apolipoprotein E, especially apolipoprotein E4, increases the oligomerization of amyloid β peptide.J. Neurosci. 32 : 15181-15192. - Murer MG, Yan Q, Raisman-Vozari R. 2001. Brain-derived neurotrophic factor in the control human brain, and in Alzheimer's disease and Parkinson's disease.
Prog. Neurobiol. 63 : 71-124. - Zuccato C, Cattaneo E. 2009. Brain-derived neurotrophic factor in neurodegenerative diseases.
Nat. Rev. Neurol. 5 : 311-322. - Guo S, Wang S, Kim WJ, Lee S-R, Frosch MP, Bacskai BJ,
et al . 2006. Effects of apoE isoforms on beta-amyloid-induced matrix metalloproteinase-9 in rat astrocytes.Brain Res. 1111 : 222-226. - Gottschall PE, Deb S. 1996. Regulation of matrix metalloproteinase expression in astrocytes, microglia and neurons.
Neuroimmunomodulation 3 : 69-75. - de Pins B, Cifuentes-Díaz C, Farah AT, López-Molina L, Montalban E, Sancho-Balsells A,
et al . 2019. Conditional BDNF delivery from astrocytes rescues memory deficits, spine density, and synaptic properties in the 5xFAD mouse model of Alzheimer disease.J. Neurosci. 39 : 2441-2458. - Capoccia E, Cirillo C, Marchetto A, Tiberi S, Sawikr Y, Pesce M,
et al . 2015. S100B-p53 disengagement by pentamidine promotes apoptosis and inhibits cellular migration via aquaporin-4 and metalloproteinase-2 inhibition in C6 glioma cells.Oncol. Lett. 9 : 2864-2870. - Dolman D, Drndarski S, Abbott NJ, Rattray M. 2005. Induction of aquaporin 1 but not aquaporin 4 messenger RNA in rat primary brain microvessel endothelial cells in culture.
J. Neurochem. 93 : 825-833. - Yoneda K, Yamamoto N, Asai K, Sobue K, Fujita Y, Fujita M,
et al . 2001. Regulation of aquaporin-4 expression in astrocytes.Mol. Brain Res. 89 : 94-102. - Grobben B, De Deyn P, Slegers H. 2002. Rat C6 glioma as experimental model system for the study of glioblastoma growth and invasion.
Cell Tissue Res. 310 : 257-270. - Galland F, Seady M, Taday J, Smaili SS, Gonçalves CA, Leite MC. 2019. Astrocyte culture models: molecular and function characterization of primary culture, immortalized astrocytes and C6 glioma cells.
Neurochem. Int. 131 : 104538. - Goya L, Feng PT, Aliabadi S, Timiras PS. 1996. Effect of growth factors on the
in vitro growth and differentiation of early and late passage C6 glioma cells.Int. J. Dev. Neurosci. 14 : 409-417. - Park SH, Lee JY, Jhee KH, Yang SA. 2020. Amyloid-ß peptides inhibit the expression of AQP4 and glutamate transporter EAAC1 in insulin-treated C6 glioma cells.
Toxicol. Rep. 7 : 1083-1089. - Schäfer M, Erdö SL. 1991. Development of glutamate neurotoxicity in cortical cultures: induction of vulnerability by insulin.
Dev. Brain Res. 62 : 293-296. - Schäfer M, Erdö SL. 1992. Insulin-specific sensitization of cultured cerebrocortical neurons to glutamate excitotoxicity.
Brain Res. 580 : 331-333. - Cai W, Xue C, Sakaguchi M, Konishi M, Shirazian A, Ferris HA,
et al . 2018. Insulin regulates astrocyte gliotransmission and modulates behavior.J. Clin. Invest. 128 : 2914-2926. - Qiu WQ, Ye Z, Kholodenko D, Seubert P, Selkoe DJ. 1997. Degradation of amyloid β-protein by a metalloprotease secreted by microglia and other neural and non-neural cells.
J. Biol. Chem. 272 : 6641-6646. - Farris W, Mansourian S, Chang Y, Lindsley L, Eckman EA, Frosch MP,
et al . 2003. Insulin-degrading enzyme regulates the levels of insulin, amyloid β-protein, and the β-amyloid precursor protein intracellular domainin vivo .Proc. Natl. Acad. Sci. USA 100 : 4162-4167. - Miller BC, Eckman EA, Sambamurti K, Dobbs N, Chow KM, Eckman CB,
et al . 2003. Amyloid-β peptide levels in brain are inversely correlated with insulysin activity levelsin vivo .Proc. Natl. Acad. Sci. USA 100 : 6221-6226. - Lee JY, Park SH, Jhee KH, Yang SA. 2022.
Zizania latifolia and its major compound tricin regulate immune responses in OVA-treated mice.Molecules 27 : 3978. - Lee JY, Park SH, Jhee KH, Yang SA. 2020. Tricin isolated from enzyme-treated
Zizania latifolia extract inhibits IgE-mediated allergic reactions in RBL-2H3 cells by targeting the Lyn/Syk pathway.Molecules 25 : 2084. - Park SH, Lee SS, Bang MH, Jo SK, Jhee KH, Yang SA. 2019. Protection against UVB-induced damages in human dermal fibroblasts: efficacy of tricin isolated from enzyme-treated
Zizania latifolia extract.Biosci. Biotechnol. Biochem. 83 : 551-560. - Gan RY, Lui WY, Wu K, Chan CL, Dai SH, Sui ZQ,
et al . 2017. Bioactive compounds and bioactivities of germinated edible seeds and sprouts: an updated review.Trends Food Sci. Technol. 59 : 1-14. - Vallejo R, Platt DC, Rink JA, Jones MA, Kelley CA, Gupta A,
et al . 2019. Electrical stimulation of C6 glia-precursor cellsin vitro differentially modulates gene expression related to chronic pain pathways.Brain Sci. 9 : 303. - Preato AM, Pinheiro EdS, Rosenstock TR, Glezer I. 2024. The relevance of astrocytic cell culture models for neuroinflammation in neurodegeneration research.
Neuroglia 5 : 27-49. - Prajapat M, Kaur G, Choudhary G, Pahwa P, Bansal S, Joshi R,
et al . 2023. A systematic review for the development of Alzheimer's disease inin vitro models: a focus on different inducing agents.Front. Aging Neurosci. 15 : 1296919. - Wozniak M, Rydzewski B, Baker SP, Raizada MK. 1993. The cellular and physiological actions of insulin in the central nervous system.
Neurochem. Int. 22 : 1-10. - Freude S, Plum L, Schnitker J, Leeser U, Udelhoven M, Krone W,
et al . 2005. Peripheral hyperinsulinemia promotes tau phosphorylationin vivo .Diabetes 54 : 3343-3348. - Lee SS, Baek YS, Eun CS, Yu MH, Baek NI, Chung Dk,
et al . 2015. Tricin derivatives as anti-inflammatory and anti-allergic constituents from the aerial part ofZizania latifolia .Biosci. Biotechnol. Biochem. 79 : 700-706. - Liu CC, Hu J, Zhao N, Wang J, Wang N, Cirrito JR,
et al . 2017. Astrocytic LRP1 mediates brain Aβ clearance and impacts amyloid deposition.J. Neurosci. 37 : 4023-4031. - Talebi M, Kakouri E, Talebi M, Tarantilis PA, Farkhondeh T, İlgün S,
et al . 2021. Nutraceuticals-based therapeutic approach: recent advances to combat pathogenesis of Alzheimer's disease.Expert Rev. Neurother. 21 : 625-642. - Bajo R, Pusil S, Lopez ME, Canuet L, Pereda E, Osipova D,
et al . 2015. Scopolamine effects on functional brain connectivity: a pharmacological model of Alzheimer's disease.Sci. Rep. 5 : 9748. - Konar A, Shah N, Singh R, Saxena N, Kaul SC, Wadhwa R,
et al . 2011. Protective role of Ashwagandha leaf extract and its component withanone on scopolamine-induced changes in the brain and brain-derived cells.PLoS One 6 : e27265.
Related articles in JMB

Article
Research article
J. Microbiol. Biotechnol. 2025; 35(): 1-9
Published online February 28, 2025 https://doi.org/10.4014/jmb.2412.12026
Copyright © The Korean Society for Microbiology and Biotechnology.
Amelioration of Astrocytic Dysfunction via AQP4/LRP1 Pathway by Zizania latifolia and Tricin in C6 Cells Exposed to Amyloid β and High-Dose Insulin and in Mice Treated with Scopolamine
Seun-Ah Yang1†*, Se-Ho Park1†, Eun-Hye Kim2, Won-Bin Bae1, and Kwang-Hwan Jhee2
1Department of Food Science and Technology, Keimyung University, Daegu 42601, Republic of Korea
2Department of Applied Chemistry, Kumoh National Institute of Technology, Gumi 39177, Republic of Korea
Correspondence to:Seun-Ah Yang, seunahy@kmu.ac.kr
†These authors contributed equally to this study.
Abstract
Zizania latifolia and its bioactive compound tricin have been recognized for their anti-inflammatory, anti-allergic, and anti-aging properties. However, the impact of Z. latifolia extract (ZLE) and tricin on astrocyte dysfunction, particularly related to disruptions in the amyloid β (Aβ) clearance pathway, has not been extensively studied. This research aims to explore the regulatory effects of ZLE and tricin on astroglial dysfunction, utilizing astrocytic differentiated C6 cells (passages 75~85) subjected to Aβ and high-dose insulin, as well as scopolamine-induced mice. Results revealed that ZLE (500 μg/ml) and tricin (1 μg/ml) significantly upregulated the expression of astrocyte proteins GFAP and AQP4, brain-derived neurotrophic factor (BDNF), low-density lipoprotein receptor-related protein 1 (LRP1), and matrix metalloproteinases (MMPs) in C6 cells treated with Aβ and high-dose insulin. Furthermore, oral administration of ZLE (100 and 300 mg/kg) and tricin (0.3 mg/kg) in mice led to an increase in acetylcholine (ACh) levels and upregulation of insulin-degrading enzyme (IDE), LRP1, and MMPs, while reducing the levels of acetylcholinesterase (AChE), Aβ and ApoE4. These findings suggest that ZLE and tricin may ameliorate Aβ and high-dose insulin-induced astrocyte dysfunction in C6 cells and scopolamine-treated mice, potentially through the AQP4/LRP1 pathway.
Keywords: C6 astroglial cell, amyloid &beta,, GFAP, AQP4, LRP1
Introduction
Astrocytes are the predominant glial cells within the central nervous system (CNS), playing a crucial role in maintaining brain homeostasis. They are integral to metabolic support, nutritional provision, ion and neuro-transmitter balance, blood-brain barrier regulation, and CNS defense mechanisms. Given these homeostatic responsibilities, astrocytes are intimately involved in the pathophysiology of neurological disorders. The balance between neuronal damage, neuroprotection, and regeneration critically influences the progression and outcomes of these diseases. Brain injuries, whether acute or chronic, elicit a specialized glial response known as reactive astrogliosis, characterized by significant morphological and functional changes in astrocytes.
Alzheimer’s disease (AD), an age-associated neurodegenerative condition, is defined by the accumulation of extracellular fibrillar amyloid β (Aβ) plaques and intracellular neurofibrillary tangles of hyperphosphorylated tau protein [1]. Pathological changes in astrocytes during Alzheimer’s disease (AD) highlight distinct temporal patterns. In the later stages of AD, astrogliosis becomes prominent, with reactive astrocytes closely associated with senile plaque formation. Supporting this,
Recent studies have underscored the significant role of astrocytes and brain-derived neurotrophic factor (BDNF) in the development of Alzheimer's disease (AD). BDNF, a crucial neurotrophin, supports neuronal survival and function, and it is essential for synaptic plasticity, which is vital for learning and memory processes. In patients with AD, consistently lower levels of BDNF have been observed, correlating strongly with the severity of cognitive decline [16, 17]. Consequently, therapeutic strategies aimed at preserving or enhancing BDNF levels are being actively explored as potential interventions for AD.
In addition to BDNF, aquaporin-4 (AQP4) has emerged as a critical factor in maintaining normal astrocyte function, as evidenced by recent
Materials and Methods
Reagents
MTT (3-(4,5-dimethylthiazol-2-yl)2-,5-diphenyltetrazolium bromide), and insulin were purchased from Sigma-Aldrich (USA). Antibody against AQP4 (#59678), β-actin (#3700), GFAP (#80788), LRP1 (#64099), horseradish peroxidase-linked anti-mouse secondary antibody (#7076), horseradish peroxidase-linked anti-rabbit secondary antibody (#7074) were purchased from Cell Signaling Technology (USA). Antibody against BDNF (ab108319), matrix metalloproteinase (MMP)-2 (ab92536), MMP-9 (ab76003), and Aβ1-42 (Aβ1-42, ab120301) were purchased from Abcam Biotechnology (UK). IDE (sc-393887) was purchased from Santa Cruz Biotechnology (USA). Fetal bovine serum (FBS), Dulbecco’s modified Eagle’s medium (DMEM) were purchased from the American Type Culture Collection (USA).
Cell Culture
C6 cells were purchased from American Type Culture Collection. Cells were cultured in DMEM supplemented with penicillin (120 units/ml), streptomycin (120 units/ml), and 10% FBS in a 5% CO2 atmosphere at 37°C.
Animals
Balb/cJ mice (11 weeks old, male) were purchased from Hana Bio (Republic of Korea), and were used as the experimental animals after undergoing quarantine and acclimatization for a week at the Dongnam Medical Research Institute Animal Company (Animal Facility Registration Certificate: No. 412). During breeding, the lighting time was set to a 12-h cycle, and food and water were freely available. Group separation and treatment of experimental animals were designed and performed as presented in Table 1. This study was conducted by the Animal Experimental Ethics Committee of Dongnam Medical Research Institute (No. SEMI-21-003). Briefly, scopolamine (3 mg/kg, i.p.) was administered daily along with donepezil (0.5 mg/kg, positive control) for 7 days, after which the experimental animals were euthanized with CO2.
-
Table 1 . Experimental design..
Group Mice Treatment CON 7 Saline (i.p) + saline (p.o) SCO 7 Scopolamine 3 mg/kg (i.p) + saline (p.o) DON 7 Scopolamine 3 mg/kg (i.p) + donepezil 5 mg/kg (p.o) ZLE100 7 Scopolamine 3 mg/kg (i.p) + ZLE1) 100 mg/kg (p.o) ZLE300 7 Scopolamine 3 mg/kg (i.p) + ZLE1) 300 mg/kg (p.o) Tricin 7 Scopolamine 3 mg/kg (i.p) + tricin 0.3 mg/kg (p.o) 1)ZLE,
Zizania latifolia extract.
Contents of Acetylcholine (ACh) and Acetylcholine Esterase (AChE)
The brain tissue was harvested and homogenized by adding 0.1 M PBS at a ratio of 1:10, respectively. After centrifugation at 10,000 ×
Hematoxylin and Eosin (H&E) Staining
The mice were sacrificed and brain tissues were harvested for further examination. Each tissue was fixed in formalin solution (4% paraformaldehyde in PBS). Then, the tissues were successively dehydrated in ethanol and xylene, followed by paraffin embedding. Each paraffin block contained a whole-brain sample, and was coronally sectioned to 5 μm thickness using a microtome (RM-2125 RT, Leica, Germany). The sliced sections were stained with H&E and mounted, followed by imaging using an optical microscope (Eclipse 80i, Nikon, Japan).
Western Blot
C6 cells were seeded into a 6-well cell culture plate (1 × 105 cells per well, cell passage; #75) for 24 h, and then treated with ZLE (50, 500 μg/ml) or tricin (1 μg/ml) with insulin (100 μg/ml) and aggregated Aβ1-42 (5 μM) for 48 h. C6 cells were lysed in a membrane protein extraction kit (Thermo Fisher Scientific, Inc., USA) or RIPA lysis buffer. In the
Statistical Analysis
All experiments were repeated at least three times, and each experiment was performed in triplicate. Results are presented as means ± standard deviations (SD). A significant difference from the respective control for each experimental test condition was assessed using Student’s
Results
Effects of ZLE or Tricin on Astrocyte Proteins Expression in Aβ Plus High-Dose Insulin-Treated C6 Cells
First, to investigate the effect of ZLE or tricin on astrocyte dysfunction, the changes in the protein levels of AQP4, GFAP, and BDNF were investigated using Aβ plus high-dose insulin-treated C6 cells. Astrocytic condition of C6 cells for normal control were confirmed by the expression of GFAP and AQP4, together with the normal expression level of BDNF (Fig. 1, left panel, lane 1). Exposure of the cells to high-dose insulin (100 μg/ml) significantly suppressed the protein expressions of AQP4, GFAP, and BDNF in the C6 cells (Fig. 1, lane 2), and treatment with Aβ1-42 further inhibited these expressions (Fig. 1, lane 3). These suppressed expressions of AQP4, GFAP, and BDNF were reversed by the treatment of ZLE (500 μg/ml) about 1.65-, 3.19-, and 2.26-folds, respectively (Fig. 1, lane 5). Tricin (1 μg/ml) also up-regulated these protein expressions about 1.64-, 3.20-, 2.32-folds, respectively (Fig. 1, lane 6).
-
Figure 1. Effects of ZLE and tricin on AQP4, GFAP, and BDNF protein expression in C6 cells treated with highdose insulin and Aβ1-42. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. untreated cell and *p < 0.05, *p < 0.005, ***p < 0.001 vs. insulin and Aβ-treated control.
We next examined the effects of the samples on the LRP1/MMPs-mediated Aβ clearance pathway in Aβ plus high-dose insulin-treated C6 cells. The LRP1 expression in astrocytes plays strategic roles in brain Aβ degradation [11], and LRP1 regulates expressions of the Aβ degradation enzymes MMP-2 and -9, eventually alleviating the accumulation of Aβ. Treatment the cells with Aβ plus high-dose insulin almost completely blocked the expressions of LRP1 and MMPs (Fig. 2, left panel, lane 3). The protein expressions of LRP1, MMP-2, and MMP-9 were significantly up-regulated about 2.30-, 32.07-, 3.74-folds, respectively, by ZLE (500 μg/ml, Fig. 2, lane 5), and 2.37-, 47.79-, and 12.37-folds, respectably, by tricin (1 μg/ml, Fig. 2, lane 6). These results suggest that ZLE and tricin could protect brain cells such as astrocytes against damage induced by high-dose insulin and/or Aβ, results in prevention of pathological aging of the brain as well as neurodegenerative disease, including AD.
-
Figure 2. Effects of ZLE and tricin on LRP1, MMP-2, and MMP-9 protein expression in C6 cells treated with high-dose insulin and Aβ1-42. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. untreated cell and *p < 0.005, ***p < 0.001 vs. insulin and Aβ-treated control.
Effects of ZLE or Tricin on ACh and AChE in Brain Tissues of Scopolamine-Treated Mice
To check that the administration of ZLE or tricin regulated acetylcholine (ACh) and acetylcholinesterase (AChE) in scopolamine-treated mice, ACh level and AChE activity were measured in mice brain tissues. As shown in Fig. 3A, the level of ACh in scopolamine-treated mice was significantly decreased (
-
Figure 3. Effects of ZLE and tricin on ACh concentration and AChE activity in the hippocampus of scopolamine-treated mice. (A) ACh concentration ; (B) AChE activity. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. normal group and *p < 0.05, *p < 0.005, ***p < 0.001 vs. scopolamine-treated group. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine-treated control group, PC: donepezil-treated positive control group, Z100: ZLE 100 mg/kg treated group, Z300: ZLE 300 mg/kg treated group, T: tricin 0.3 mg/kg treated group.
Effect of ZLE or Tricin on Histopathological Changes of Scopolamine-Treated Mice
The results of the H&E staining for histopathological examination are shown in Fig. 4. The hippocampal neurons of the normal group exhibited well-arranged and a clear structure compared to the scopolamine-treated group. A clear pathological change was observed in the hippocampus of scopolamine-treated negative control group mice. The irregular or altered morphology was clearly prevented by the treatment with ZLE (100, 300 mg/kg) or tricin (0.3 mg/kg; corresponding of 300 mg/kg administration of ZLE group), suggesting the protective effect of ZLE- or tricin-administration against neuronal damage induced by scopolamine.
-
Figure 4. Histological examination by H&E staining in the hippocampus of scopolamine-treated mice administrated ZLE and tricin. Histological changes were observed at x40 (upper panels) and x100 (lower panels). Black arrows indicate brain tissue necrosis. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine (3 mg/kg) treated control group, PC: scopolamine (3 mg/kg)+donepezil (5 mg/kg) treated positive control group, Z100: scopolamine (3 mg/kg)+ZLE (100 mg/kg) treated group, Z300: scopolamine (3 mg/kg)+ZLE (300 mg/kg) treated group, T: scopolamine (3 mg/kg)+tricin (0.3 mg/kg) treated group.
Effect of ZLE or Tricin on Astrocyte Proteins Expression in Brain Tissues of Scopolamine-Treated Mice
The levels of AQP4, GFAP, and BDNF in hippocampus of scopolamine-treated mice were investigated by Western blot analysis. The expressions of these proteins were dramatically decreased in the scopolamine-treated control group (Fig. 5, left panel lane 2). However, the administration of ZLE at 300 mg/kg or tricin restored these proteins expression compared to the scopolamine-treated mice in the negative control group (
-
Figure 5. Effects of ZLE and tricin on AQP4, GFAP, and BDNF protein expression in scopolamine-treated mice. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. normal group and *p < 0.05, ***p < 0.001 vs. scopolamine-treated group. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine-treated control group, PC: donepezil-treated positive control group, Z100: ZLE 100 mg/kg treated group, Z300: ZLE 300 mg/kg treated group, T: tricin 0.3 mg/kg treated group.
We next investigated the effects of ZLE or tricin on Aβ clearance pathway, such as LRP1 and Aβ degrading enzymes in scopolamine-treated mice. As shown in Fig. 6, the expression levels of Aβ uptake protein LRP1 and Aβ degradation enzymes (IDE, MMP-2, MMP-9) were suppressed in scopolamine-treated mice, but it was confirmed that these expressions were clearly increased in ZLE (300 mg/kg)- and tricin (0.3 mg/kg)-administrated mice as well as donepezil-administered control group compared with scopolamine-treated mice. On the other hand, the increased expression of ApoE4 protein induced by scopolamine treatment was reduced following sample administration, with a particularly significant reduction observed with Z300 and tricin treatment. These results suggest that in the scopolamine-induced amnesic condition, the generation of apoE4 fragments was promoted, potentially leading to amyloid-beta accumulation, which indicates impaired amyloid clearance.
-
Figure 6. Effects of ZLE and tricin on Aβ1-42, ApoE4, IDE, LRP1, MMP-2, and MMP-9 protein expression in scopolamine-treated mice. β-actin was used as the internal control for Western blot analysis. Results are presented as the means ± SD of percentages calculated with respect to control levels, of three independent experiments. #
p < 0.05, vs. normal group and *p < 0.05, *p < 0.005, ***p < 0.001 vs. scopolamine-treated group. Donepezil (5 mg/kg) was used as the positive control. N: normal group, C: scopolamine-treated control group, PC: donepezil-treated positive control group, Z100: ZLE 100 mg/kg treated group, Z300: ZLE 300 mg/kg treated group, T: tricin 0.3 mg/kg treated group.
Discussion
This study aimed to assess the effects of ZLE and its active compound, tricin, on astrocyte dysregulation induced by high-dose insulin combined with Aβ1-42 in C6 astroglial cells. The C6 cell line, which shares characteristics with astrocytes, is commonly utilized
AQP4 plays a pivotal role in the degradation of Aβ and the uptake of glutamate in astrocytes, with its deficiency being strongly associated with cognitive impairments. Research has demonstrated that mice lacking the AQP4 gene exhibit increased deposition of Aβ plaques in the brain, accompanied by memory deficits [3, 4]. In our study, we observed that treatment with ZLE or its major compound, tricin, exerted a protective effect against astroglial dysfunction in C6 cells exposed to high-dose insulin and Aβ1-42. Specifically, we found that the upregulation of astroglial markers GFAP and AQP4 following treatment with ZLE or tricin may help mitigate deficits in the Aβ clearance pathway, as evidenced by the enhanced expression of LRP1 and MMPs, which are critical in the pathogenesis of AD. Tricin, a flavonoid present in the Gramineae family, was identified as the primary active compound in ZLE responsible for its protective effects, including the prevention of allergic responses. Prior research has identified five derivatives of tricin (4',5,7-trihydroxy-3',5'-dimethoxyflavone) in the methanol extract derived from the aerial parts of
In our previous research, we observed that the expression levels of astrocytic markers GFAP and AQP4 increased with successive cell passages. Consequently, for optimal protein expression, we selected cells at passage numbers 75 to 85. To simulate astrodegenerative conditions characterized by reduced GFAP and AQP4 expression, as reported in earlier studies [27], we treated the astroglial cells with a combination of high-dose insulin and Aβ1-42. The downregulation of GFAP and AQP4 in astrocytes may indicate dysfunction, and our findings suggest that ZLE and tricin have the potential to counteract AD progression by preserving astrocyte function, particularly within the Aβ clearance system. Our data demonstrated that ZLE and tricin effectively protected astrocytes from deregulation induced by high-dose insulin and Aβ1-42, leading to the upregulation of key proteins essential for astrocyte function (Fig. 1) and Aβ degradation (Fig. 2). Additionally, existing studies have established that astrocytic low-density lipoprotein receptor-related protein 1 (LRP1) is crucial for Aβ clearance in the brain [11, 12]. Knockdown of LRP1 in primary astrocytes has been shown to reduce cellular Aβ uptake and degradation, while also downregulating several major Aβ-degrading enzymes, including MMP-2, MMP-9, and IDE, further emphasizing the importance of LRP1 in maintaining Aβ homeostasis [44].
Additionally, we investigated the impact of ZLE and tricin on the expression of astrocyte marker proteins and the Aβ-degrading pathway in a scopolamine-induced mouse model. Scopolamine, a muscarinic acetylcholine receptor antagonist, disrupts cholinergic neurotransmission, leading to cognitive and memory impairments in animal models. These impairments mimic the memory dysfunction observed in Alzheimer's disease patients, making scopolamine a widely used agent for studying neurodegenerative processes and potential therapeutic interventions [45]. Scopolamine-induced elevation of AChE activity is linked to a reduction in ACh levels, a process that can result in memory deficits and is implicated in the development of AD pathology [46]. In this study, the administration of
In conclusion, this study demonstrated the protective effects of ZLE and tricin on astrocyte function within an
Acknowledgment
This research was supported by the Bisa Research Grant of Keimyung University in 2023 (Project No: 20230313).
Author Contributions
Seun-Ah Yang: Conceptualization; Funding acquisition; Methodology; Visualization; Writing - original draft; Writing - review & editing. Se-Ho Park: Data curation; Formal analysis; Investigation; Validation; Visualization; Writing – original draft. Eun-Hye Kim: Investigation; Validation. Won-Bin Bae : Investigation; Validation. Kwang-Hwan Jhee : Validation; Writing – review & editing. All authors have read and agreed to the published version of this manuscript.
Conflict of Interest
The authors have no financial conflicts of interest to declare.
Fig 1.

Fig 2.

Fig 3.

Fig 4.

Fig 5.

Fig 6.

-
Table 1 . Experimental design..
Group Mice Treatment CON 7 Saline (i.p) + saline (p.o) SCO 7 Scopolamine 3 mg/kg (i.p) + saline (p.o) DON 7 Scopolamine 3 mg/kg (i.p) + donepezil 5 mg/kg (p.o) ZLE100 7 Scopolamine 3 mg/kg (i.p) + ZLE1) 100 mg/kg (p.o) ZLE300 7 Scopolamine 3 mg/kg (i.p) + ZLE1) 300 mg/kg (p.o) Tricin 7 Scopolamine 3 mg/kg (i.p) + tricin 0.3 mg/kg (p.o) 1)ZLE,
Zizania latifolia extract.
References
- Rodriguez J, Olabarria M, Chvatal A, Verkhratsky A. 2009. Astroglia in dementia and Alzheimer's disease.
Cell Death Difer. 16 : 378-385. - Hou L, Liu Y, Wang X, Ma H, He J, Zhang Y,
et al . 2011. The effects of amyloid-β 42 oligomer on the proliferation and activation of astrocytesin vitro .In vitro Cell. Dev. Biol. Anim. 47 : 573-580. - Beauquis J, Pavía P, Pomilio C, Vinuesa A, Podlutskaya N, Galvan V,
et al . 2013. Environmental enrichment prevents astroglial pathological changes in the hippocampus of APP transgenic mice, model of Alzheimer's disease.Exp. Neurol. 239 : 28-37. - Beauquis J, Vinuesa A, Pomilio C, Pavía P, Galván V, Saravia F. 2014. Neuronal and glial alterations, increased anxiety, and cognitive impairment before hippocampal amyloid deposition in PDAPP mice, model of Alzheimer's disease.
Hippocampus 24 : 257-269. - Braun K, Antemano R, Helmeke C, Büchner M, Poeggel G. 2009. Juvenile separation stress induces rapid region-and layer-specific changes in S100β-and glial fibrillary acidic protein-immunoreactivity in astrocytes of the rodent medial prefrontal cortex.
Neurosci. 160 : 629-638. - Rajkowska G, A Stockmeier C. 2013. Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue.
Curr. Drug Targets 14 : 1225-1236. - Xu Z, Xiao N, Chen Y, Huang H, Marshall C, Gao J,
et al . 2015. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Aβ accumulation and memory deficits.Mol. Neurodegener. 10 : 58. - Lan YL, Zhao J, Ma T, Li S. 2016. The potential roles of aquaporin 4 in Alzheimer's disease.
Mol. Neurobiol. 53 : 5300-5309. - Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA,
et al . 2012. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β.Sci. Transl. Med. 4 : 147ra111. - Verghese JP, Terry A, de Natale ER, Politis M. 2022. Research evidence of the role of the glymphatic system and its potential pharmacological modulation in neurodegenerative diseases.
J. Clin. Med. 11 : 6964. - Ries M, Sastre M. 2016. Mechanisms of Aβ clearance and degradation by glial cells.
Front. Aging Neurosci. 8 : 160. - Kanekiyo T, Liu CC, Shinohara M, Li J, Bu G. 2012. LRP1 in brain vascular smooth muscle cells mediates local clearance of Alzheimer's amyloid-β.
J. Neurosci. 32 : 16458-16465. - Eckman EA, Reed DK, Eckman CB. 2001. Degradation of the Alzheimer's amyloid β peptide by endothelin-converting enzyme.
J. Biol. Chem. 276 : 24540-24548. - Bernstein HG, Lendeckel U, Bukowska A, Ansorge S, Ernst T, Stauch R,
et al . 2008. Regional and cellular distribution patterns of insulin-degrading enzyme in the adult human brain and pituitary.J. Chem. Neuroanat. 35 : 216-224. - Hashimoto T, Serrano-Pozo A, Hori Y, Adams KW, Takeda S, Banerji AO,
et al . 2012. Apolipoprotein E, especially apolipoprotein E4, increases the oligomerization of amyloid β peptide.J. Neurosci. 32 : 15181-15192. - Murer MG, Yan Q, Raisman-Vozari R. 2001. Brain-derived neurotrophic factor in the control human brain, and in Alzheimer's disease and Parkinson's disease.
Prog. Neurobiol. 63 : 71-124. - Zuccato C, Cattaneo E. 2009. Brain-derived neurotrophic factor in neurodegenerative diseases.
Nat. Rev. Neurol. 5 : 311-322. - Guo S, Wang S, Kim WJ, Lee S-R, Frosch MP, Bacskai BJ,
et al . 2006. Effects of apoE isoforms on beta-amyloid-induced matrix metalloproteinase-9 in rat astrocytes.Brain Res. 1111 : 222-226. - Gottschall PE, Deb S. 1996. Regulation of matrix metalloproteinase expression in astrocytes, microglia and neurons.
Neuroimmunomodulation 3 : 69-75. - de Pins B, Cifuentes-Díaz C, Farah AT, López-Molina L, Montalban E, Sancho-Balsells A,
et al . 2019. Conditional BDNF delivery from astrocytes rescues memory deficits, spine density, and synaptic properties in the 5xFAD mouse model of Alzheimer disease.J. Neurosci. 39 : 2441-2458. - Capoccia E, Cirillo C, Marchetto A, Tiberi S, Sawikr Y, Pesce M,
et al . 2015. S100B-p53 disengagement by pentamidine promotes apoptosis and inhibits cellular migration via aquaporin-4 and metalloproteinase-2 inhibition in C6 glioma cells.Oncol. Lett. 9 : 2864-2870. - Dolman D, Drndarski S, Abbott NJ, Rattray M. 2005. Induction of aquaporin 1 but not aquaporin 4 messenger RNA in rat primary brain microvessel endothelial cells in culture.
J. Neurochem. 93 : 825-833. - Yoneda K, Yamamoto N, Asai K, Sobue K, Fujita Y, Fujita M,
et al . 2001. Regulation of aquaporin-4 expression in astrocytes.Mol. Brain Res. 89 : 94-102. - Grobben B, De Deyn P, Slegers H. 2002. Rat C6 glioma as experimental model system for the study of glioblastoma growth and invasion.
Cell Tissue Res. 310 : 257-270. - Galland F, Seady M, Taday J, Smaili SS, Gonçalves CA, Leite MC. 2019. Astrocyte culture models: molecular and function characterization of primary culture, immortalized astrocytes and C6 glioma cells.
Neurochem. Int. 131 : 104538. - Goya L, Feng PT, Aliabadi S, Timiras PS. 1996. Effect of growth factors on the
in vitro growth and differentiation of early and late passage C6 glioma cells.Int. J. Dev. Neurosci. 14 : 409-417. - Park SH, Lee JY, Jhee KH, Yang SA. 2020. Amyloid-ß peptides inhibit the expression of AQP4 and glutamate transporter EAAC1 in insulin-treated C6 glioma cells.
Toxicol. Rep. 7 : 1083-1089. - Schäfer M, Erdö SL. 1991. Development of glutamate neurotoxicity in cortical cultures: induction of vulnerability by insulin.
Dev. Brain Res. 62 : 293-296. - Schäfer M, Erdö SL. 1992. Insulin-specific sensitization of cultured cerebrocortical neurons to glutamate excitotoxicity.
Brain Res. 580 : 331-333. - Cai W, Xue C, Sakaguchi M, Konishi M, Shirazian A, Ferris HA,
et al . 2018. Insulin regulates astrocyte gliotransmission and modulates behavior.J. Clin. Invest. 128 : 2914-2926. - Qiu WQ, Ye Z, Kholodenko D, Seubert P, Selkoe DJ. 1997. Degradation of amyloid β-protein by a metalloprotease secreted by microglia and other neural and non-neural cells.
J. Biol. Chem. 272 : 6641-6646. - Farris W, Mansourian S, Chang Y, Lindsley L, Eckman EA, Frosch MP,
et al . 2003. Insulin-degrading enzyme regulates the levels of insulin, amyloid β-protein, and the β-amyloid precursor protein intracellular domainin vivo .Proc. Natl. Acad. Sci. USA 100 : 4162-4167. - Miller BC, Eckman EA, Sambamurti K, Dobbs N, Chow KM, Eckman CB,
et al . 2003. Amyloid-β peptide levels in brain are inversely correlated with insulysin activity levelsin vivo .Proc. Natl. Acad. Sci. USA 100 : 6221-6226. - Lee JY, Park SH, Jhee KH, Yang SA. 2022.
Zizania latifolia and its major compound tricin regulate immune responses in OVA-treated mice.Molecules 27 : 3978. - Lee JY, Park SH, Jhee KH, Yang SA. 2020. Tricin isolated from enzyme-treated
Zizania latifolia extract inhibits IgE-mediated allergic reactions in RBL-2H3 cells by targeting the Lyn/Syk pathway.Molecules 25 : 2084. - Park SH, Lee SS, Bang MH, Jo SK, Jhee KH, Yang SA. 2019. Protection against UVB-induced damages in human dermal fibroblasts: efficacy of tricin isolated from enzyme-treated
Zizania latifolia extract.Biosci. Biotechnol. Biochem. 83 : 551-560. - Gan RY, Lui WY, Wu K, Chan CL, Dai SH, Sui ZQ,
et al . 2017. Bioactive compounds and bioactivities of germinated edible seeds and sprouts: an updated review.Trends Food Sci. Technol. 59 : 1-14. - Vallejo R, Platt DC, Rink JA, Jones MA, Kelley CA, Gupta A,
et al . 2019. Electrical stimulation of C6 glia-precursor cellsin vitro differentially modulates gene expression related to chronic pain pathways.Brain Sci. 9 : 303. - Preato AM, Pinheiro EdS, Rosenstock TR, Glezer I. 2024. The relevance of astrocytic cell culture models for neuroinflammation in neurodegeneration research.
Neuroglia 5 : 27-49. - Prajapat M, Kaur G, Choudhary G, Pahwa P, Bansal S, Joshi R,
et al . 2023. A systematic review for the development of Alzheimer's disease inin vitro models: a focus on different inducing agents.Front. Aging Neurosci. 15 : 1296919. - Wozniak M, Rydzewski B, Baker SP, Raizada MK. 1993. The cellular and physiological actions of insulin in the central nervous system.
Neurochem. Int. 22 : 1-10. - Freude S, Plum L, Schnitker J, Leeser U, Udelhoven M, Krone W,
et al . 2005. Peripheral hyperinsulinemia promotes tau phosphorylationin vivo .Diabetes 54 : 3343-3348. - Lee SS, Baek YS, Eun CS, Yu MH, Baek NI, Chung Dk,
et al . 2015. Tricin derivatives as anti-inflammatory and anti-allergic constituents from the aerial part ofZizania latifolia .Biosci. Biotechnol. Biochem. 79 : 700-706. - Liu CC, Hu J, Zhao N, Wang J, Wang N, Cirrito JR,
et al . 2017. Astrocytic LRP1 mediates brain Aβ clearance and impacts amyloid deposition.J. Neurosci. 37 : 4023-4031. - Talebi M, Kakouri E, Talebi M, Tarantilis PA, Farkhondeh T, İlgün S,
et al . 2021. Nutraceuticals-based therapeutic approach: recent advances to combat pathogenesis of Alzheimer's disease.Expert Rev. Neurother. 21 : 625-642. - Bajo R, Pusil S, Lopez ME, Canuet L, Pereda E, Osipova D,
et al . 2015. Scopolamine effects on functional brain connectivity: a pharmacological model of Alzheimer's disease.Sci. Rep. 5 : 9748. - Konar A, Shah N, Singh R, Saxena N, Kaul SC, Wadhwa R,
et al . 2011. Protective role of Ashwagandha leaf extract and its component withanone on scopolamine-induced changes in the brain and brain-derived cells.PLoS One 6 : e27265.