TOP

Research Article

Split Viewer

Mol. Cells 2023; 46(5): 319-328

Published online April 18, 2023

https://doi.org/10.14348/molcells.2023.2156

© The Korean Society for Molecular and Cellular Biology

Ca2+-Permeable TRPV1 Receptor Mediates Neuroprotective Effects in a Mouse Model of Alzheimer’s Disease via BDNF/CREB Signaling Pathway

Juyong Kim1,2,7 , Sangwoo Seo1,7 , Jung Han Yoon Park3 , Ki Won Lee1,3,4,5 , Jiyoung Kim5,* , and Jin-Chul Kim2,6,*

1Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea, 2Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea, 3Bio-MAX Institute, Seoul National University, Seoul 08826, Korea, 4Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Korea, 5Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea, 6Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Korea, 7These authors contributed equally to this work.

Correspondence to : jckim@kist.re.kr(JCK); jiyoungkim1107@snu.ac.kr(JK)

Received: October 12, 2022; Revised: January 4, 2023; Accepted: January 6, 2023

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.

Transient receptor potential vanilloid 1 (TRPV1) protein is a Ca2+-permeable non-selective cation channel known for its pain modulation pathway. In a previous study, it was discovered that a triple-transgenic Alzheimer’s disease (AD) mouse model (3xTg-AD+/+) has anti-AD effects. The expression of proteins in the brain-derived neurotrophic factor (BDNF)/cAMP response element binding protein (CREB) pathway in a 3xTg-AD/TRPV1 transgenic mice model was investigated to better understand the AD regulatory effect of TRPV1 deficiency. The results show that TRPV1 deficiency leads to CREB activation by increasing BDNF levels and promoting phosphorylation of tyrosine receptor kinase B (TrkB), extracellular signal-regulated kinase (ERK), protein kinase B (Akt), and CREB in the hippocampus. Additionally, TRPV1 deficiency-induced CREB activation increases the antiapoptotic factor B-cell lymphoma 2 (Bcl-2) gene, which consequently downregulates Bcl-2-associated X (Bax) expression and decreases cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase (PARP), which leads to the prevention of hippocampal apoptosis. In conclusion, TRPV1 deficiency exhibits neuroprotective effects by preventing apoptosis through the BDNF/CREB signal transduction pathway in the hippocampus of 3xTg-AD mice.

Keywords Alzheimer’s disease, apoptosis, brain-derived neurotrophic factor, cAMP response element binding protein, transient receptor potential vanilloid 1

Alzheimer’s disease (AD) is one of the most representative age-related neurodegenerative diseases accompanied by memory and cognitive deficits (Hippius and Neundörfer, 2003). Although Alois AD has been known for more than a century, there is still no effective treatment for it. Several factors contribute to AD. The most notable hypotheses for AD development are the amyloid hypothesis and the hyperphosphorylated tau hypothesis (Bloom, 2014; DeTure and Dickson, 2019). The deposition of both amyloid-β peptide (Aβ) and tau neurofibrillary tangles causes several pathological symptoms, including synaptic degradation, neuroinflammation, and neuronal cell loss, which induce memory and cognition impairment and finally develop into severe dementia (LeBlanc, 2005).

In addition to amyloid and hyperphosphorylated tau, calcium (Ca2+) dyshomeostasis is deeply implicated in the pathogenesis of AD (Bojarski et al., 2008; Cascella and Cecchi, 2021; Green and LaFerla, 2008). Ca2+ acts as an intracellular messengers and involves diverse signal transmission activities, including enzyme activation (through phosphorylation or dephosphorylation), exocytosis, gene transcription and programmed cell death (Baker et al., 2013). These actions of intracellular Ca2+ signaling in neurons affect neural growth, memory formation, and synaptic plasticity. In other words, Ca2+ dyshomeostasis causes damage and eventually leads to cell death in the hippocampus.

It has been reported that soluble Aβ aggregates and increases neuronal Ca2+ influx in patients with AD, resulting in Ca2+ overload in neurons (Berridge, 2011). As a result, resting Ca2+ increases lead to Ca2+ dysregulation, which includes several pathological features such as neuronal death and degeneration (Calvo-Rodriguez et al., 2020; Jadiya et al., 2019). Previously, we focused on these aspects and tried mediating AD pathogenesis by regulating Ca2+ levels through transient receptor potential vanilloid 1 (TRPV1), a Ca2+-permeable nonselective cation channel well-known for regulating pain and inflammation. A triple-transgenic AD (3xTg-AD+/+) mouse model was used to characterize the anti-AD effect of TRPV1 deficiency (Kim et al., 2020). In that study, TRPV1 deficiency alleviated Aβ and Tau deposition and consequently improved memory and learning deficits in the 3xTg-AD+/+ mouse model. Additionally, it was shown that Ca2+ was overloaded in 3xTg-AD+/+ mice, resulting in memory and learning deficits. However, in TRPV1 receptor knockout 3xTg-AD mice (3xTg-AD+/+/TRPV1−/−), after normalized Ca2+ levels, memory and learning abilities were restored.

The hippocampus is a curved-shaped region in the middle of the brain, essential for learning and memory. Therefore, damage to the hippocampus can lead to severe memory deficits, including memory formation and consolidation. Decreased hippocampal synaptic density in Patients with AD has been reported, which is highly correlated with cognitive deficits (Scheff and Price, 2006). Additionally, several causes of neuronal apoptosis, such as Aβ accumulation, increased oxidative stress, and DNA fragmentation, have also been reported in the brain of Patients with AD (Calissano et al., 2009). These previous studies showed that apoptosis in the brain is closely related to cognitive ability in patients with AD.

Brain-derived neurotrophic factor (BDNF) is a major neurotrophic factor that regulates neuronal survival, differentiation, and plasticity (Miranda et al., 2019). The expression of BDNF and its receptor, tyrosine receptor kinase B (TrkB), is reduced in Patients with AD (Ferrer et al., 1999; Zheng et al., 2010). There is also evidence that Aβ directly or indirectly reduces BDNF levels, leading to memory and cognition deficits (Corona et al., 2010). Therefore, the expression levels of BDNF and TrkB are important for evaluating the neuroprotective effect of TRPV1 deficiency in AD models.

TrkB activation triggers the activation of several downstream pathways, including phosphorylation of extracellular signal-regulated kinase (ERK) and protein kinase B (Akt), which in turn phosphorylates and activates cAMP response element binding protein (CREB) (Minichiello et al., 2002; Santos et al., 2010). ERK/MAPK (mitogen-activated protein kinase) is related to memory formation (Impey et al., 1999) and antiapoptotic effects (Park and Cho, 2006). Akt also plays an important role in neuroprotection (Zarneshan et al., 2022). CREB is a transcription factor that regulates the proliferation or survival of neurons by binding to cAMP-responsive elements, thereby modulating the transcription of the genes (Finkbeiner et al., 1997).

B-cell lymphoma 2 (Bcl-2) family proteins play a critical role in the apoptotic activity. The Bcl-2 family includes both the antiapoptotic protein Bcl-2 and the proapoptotic protein Bcl-2-associated X (Bax) or Bcl-2 homologous antagonist killer (Bak) (Shacka and Roth, 2005). When CREB is phosphorylated, it directly binds to the Bcl-2 family promoter, activating the signaling cascade (Meller et al., 2005). Bcl-2 forms a heterodimer with the apoptotic protein Bax and inhibits the activation of Bax, which regulates the activation of caspase-3 (Ku et al., 2011; Yin et al., 1994). Cleaved caspase-3 is known to be involved in the death of mammalian neural cells (D’Amelio et al., 2012). Activated caspase-3 cleaves cellular proteins, including poly (ADP-ribose) polymerase (PARP). Because PARP is a crucial enzyme in the repair of DNA breakage, its cleavage causes neurons to undergo apoptosis (Sairanen et al., 2009).

In this study, we aimed to understand how neurological parameters are related to the neuroprotective effect by preventing apoptosis in the hippocampus by using TRPV1 receptor knockout 3xTg-AD mice to explore the effect of TRPV1 deficiency in the hippocampus. It was found that TRPV1 deficiency inhibits hippocampal cell death by promoting the BDNF/TrkB signaling pathway. Furthermore, activating TrkB leads to the activation of CREB, which inhibits the activated caspase-3 dependent apoptosis.

Animals

3xTg-AD mice (kindly given by Professor Frank M. Laferla, UC-Irvine, Irvine, CA, USA) were homozygous for APPswe, TauP301L, and PS1M146V. These 3xTg-AD mice were mated with TRPV1−/− mice (stock #003770; provided from Jackson Laboratories, Bar Harbor, ME, USA) and offspring with nine genotypes (3xTg-AD−/−/TRPV1+/+, 3xTg-AD−/−/TRPV1+/−, 3xTg-AD−/−/TRPV1−/−, 3xTg-AD+/−/TRPV1+/+, 3xTg-AD+/−/TRPV1+/−, 3xTg-AD+/−/TRPV1−/−, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, 3xTg-AD+/+/TRPV1−/−) were generated. Four groups were selected with or without AD pathology and TRPV1 depletion. The selected groups were 3xTg-AD−/−/TRPV1+/+ mice (wild type, WT), 3xTg-AD+/+/TRPV1+/+ mice, 3xTg-AD+/+/TRPV1+/− mice and 3xTg-AD+/+/TRPV1−/− mice. Mice in each group were raised until 12 months. Mice were maintained at 21°C ± 2°C, 12-h light:12-h dark photoperiod with specific pathogen-free conditions in ventilated cages and provided with free choice to consume food and water. All animal testing was performed following protocols approved by the Institutional Animal Care and Use Committee of Seoul National University (SNU-140410-12, SNU-140410-14, SNU-140625-2).

Animal dissection and tissue collection

Zoletil (50 mg/kg) was administered intraperitoneally to anesthetize the mice (n = 5) in each group before normal saline, and a 4% paraformaldehyde solution was transcardially administered. The brains were removed and postfixed in 4% paraformaldehyde overnight at 4°C, and routine-frozen sections (10 μm) were prepared for TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining and immunohistochemistry analysis. The other half of the mice in each group were sacrificed by decapitation, and their brains were rapidly collected and cut on an ice-cooled board. The hippocampus was dissected and stored at −80°C for TUNEL staining and western blotting analysis.

TUNEL staining

TUNEL staining was performed on the frozen-embedded sections using the in-situ cell death detection kit (Promega, USA) according to the standard protocol provided by the manufacturer. Apoptotic nuclei were visualized using the peroxidase-DAB reaction. The sections were counterstained with hematoxylin. TUNEL-positive cells in the hippocampus were analyzed in each of the 3 regions of Cornu Ammonis 3 (CA3), Cornu Ammonis 1 (CA1) (especially the soma of pyramidal neurons), and dentate gyrus (DG) sections per high-power field (×40). TUNEL staining fluorescence signal intensity was measured using the ImageJ analysis program (National Institute of Health, USA). Regions of interest were selected from the hippocampus’s CA3, CA1, and DG regions.

Western blotting

Five mice’s hippocampus tissues were removed, and they were all individually examined. There was no replication. Samples were prepared by homogenizing the hippocampus tissues. The protein concentrations were determined using the Bio-Rad protein assay (Bio-Rad, USA). Western blotting analyses were performed on 50 μg of protein. Samples were briefly separated using 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then transferred to a polyvinylidene fluoride membrane (0.4 μm; Merck Millipore, USA). The membranes were blocked in a 5% bovine serum albumin (Bovogen Biologicals, Australia) in tris-buffered saline and Tween-20 (Junsei Chemical, Japan). Membranes were incubated overnight at 4°C with primary antibodies against BDNF (1:1,000; Santa Cruz Biotechnology, USA), p-TrkB (1:1,000; Cell Signaling Technology, USA), TrkB (80E3, 1:1,000; Cell Signaling Technology), p-Akt (Ser473, 1:1,000; Cell Signaling Technology), Akt (1:1,000; Cell Signaling Technology), p-ERK (E-4, 1:1,000; Santa Cruz Biotechnology), ERK (1:1,000; Santa Cruz Biotechnology), p-CREB (Ser133, D1G6, 1:1,000; Cell Signaling Technology), CREB (48H2, 1:1,000; Cell Signaling Technology), Bax (1:1,000; Santa Cruz Biotechnology), Bcl-2 (1:1,000; Santa Cruz Biotechnology), cleaved caspase-3 (1:500; Cell Signaling Technology), caspase-3 (1:500; Abcam, USA), cleaved PARP (1:1,000; Cell Signaling Technology), GAPDH (1:1,000; Cell Signaling Technology), and β-actin (1:5,000; Sigma, USA). After incubation with horseradish peroxidase-conjugated secondary antibodies (1:5,000; Sigma) for 1 h at room temperature, immunodetection was performed using an enhanced chemiluminescence detection kit (GE Healthcare, USA). The relative density of each band was measured using the ImageJ analysis program (National Institute of Health).

Statistical analysis

IBM SPSS Statistics for Windows (ver. 19.0; IBM, USA) was used for the statistical analyzes. The results were presented as mean ± SEM values. All the data were analyzed using the Fisher’s LSD (least significant difference).

TRPV1 deficiency reduces apoptosis in the hippocampus of 3xTg-AD mice

To investigate the neuroprotective effect of TRPV1 deficiency, we performed a TUNEL assay to detect hippocampal apoptosis in WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. CA3, CA1, and DG regions of the hippocampus were subjected. Nuclei of apoptotic cells in each region are shown in green fluorescence. The immunofluorescence results showed that TUNEL-positive apoptotic cells were remarkably increased in all hippocampus regions of 3xTg-AD+/+/TRPV1+/+ mice compared with WT mice. However, apoptotic cells tremendously decreased in all 3 regions of 3xTg-AD+/+/TRPV1−/− mice compared with 3xTg-AD+/+/TRPV1+/+ mice (Fig. 1A). Quantitative analysis also showed that TRPV1 deficiency significantly reduced apoptotic cells in CA3 (Fig. 1B), CA1 (Fig. 1C), and DG (Fig. 1D) regions by 67%, 67%, and 62%, respectively, compared with the same region in 3xTg-AD+/+/TRPV1+/+ mice. These results indicate that TRPV1 deficiency protects hippocampal neurons and neuroglial cells from apoptosis due to AD pathogenesis.

TRPV1 deficiency increases BDNF expression levels and promotes TrkB receptor phosphorylation in the hippocampus of 3xTg-AD mice

Western blotting analyses were conducted to examine whether TRPV1 deficiency increases the levels of BDNF and activation of its receptor protein TrkB. BDNF expression levels significantly decreased in 3xTg-AD+/+/TRPV1+/+ mice by 26% compared with 3xTg-AD−/−/TRPV1+/+ mice. However, BDNF expression levels significantly increased in 3xTg-AD+/+/TRPV1−/− mice by 625% compared with 3xTg-AD+/+/TRPV1+/+ mice (Figs. 2A and 2B). Similarly, p-TrkB expression levels proportional to TrkB and β-actin significantly decreased in 3xTg-AD+/+/TRPV1+/+ mice by 40% and 29%, respectively compared with 3xTg-AD−/−/TRPV1+/+ mice. p-TrkB expression levels also significantly increased in 3xTg-AD+/+/TRPV1−/− mice by 268% and 310% respectively compared with 3xTg-AD+/+/TRPV1+/+ mice (Figs. 2C and 2D). These results indicate that TRPV1 deficiency increases the levels of BDNF and promotes the phosphorylation of TrkB in the hippocampus of 3xTg-AD mice. As previously mentioned, BDNF is responsible for synaptic plasticity and neuronal survival and is reported to be impaired in patients with AD. In this respect, BDNF-signaling recovery is considered a promising therapeutic approach to treating AD (Fumagalli et al., 2006). Memantine, a well-known AD drug, has also been reported to reduce preclinical AD pathology by increasing BDNF protein levels, supporting the usefulness of our results (Folch et al., 2018).

TRPV1 deficiency increases the phosphorylation of ERK, Akt, and CREB

To investigate whether the activation of TrkB leads to activation of the downstream signaling of BDNF/TrkB, we measured the phosphorylation level of Akt and ERK. Akt and ERK independently mediate the BDNF/CREB pathway, where the downstream protein CREB plays an important role in memory formation by regulating BDNF (Saura and Valero, 2011). Western blotting revealed that the levels of p-Akt and p-ERK were decreased in the hippocampus of 3xTg-AD+/+/TRPV1+/+ mice compared to 3xTg-AD−/−/TRPV1+/+ mice, and the difference disappeared in 3xTg-AD+/+/TRPV1+/− and 3xTg-AD+/+/TRPV1−/− mice (Fig. 3A). The densitometry results show that TRPV1 deficiency induces the phosphorylation of Akt (Fig. 3B) and ERK (Fig. 3C) whereas the basal levels of Akt and ERK did not change. We then investigated the expression and phosphorylation level of CREB, which plays an important role in mediating the apoptosic pathway. In 3xTg-AD+/+/TRPV1+/+ mice, CREB phosphorylation levels decreased compare with WT. However, CREB phosphorylation levels in 3xTg-AD+/+/TRPV1−/− mice were significantly increased, similar to those in WT mice. Previous studies have reported that Akt and ERK mediate signal transduction in various neurodegenerative diseases, including AD (Rai et al., 2019). Similarly, our results indicate that TRPV1 deficiency in the hippocampus of 3xTg-AD mice has a neuroprotective effect by phosphorylating and activating Akt and ERK, thereby activating the CREB pathway (Figs. 3D-3F).

TRPV1 deficiency reduces the cleavage of caspase-3 and PARP in 3xTg-AD mice

As shown in Fig. 1, TRPV1 deficiency reduces apoptosis in the hippocampus of 3xTg-AD mice. As an executioner of apoptosis, caspase-3 is an essential regulator in AD progression (Asadi et al., 2022). In this regard, we investigated how TRPV1 deficiency reduces apoptosis via the regulation of caspase-3 in the hippocampus of 3xTg-AD mice. Western blotting revealed that the level of Bcl-2 was decreased, whereas Bax, cleaved-caspase-3, and cleaved PARP increased in the hippocampus of 3xTg-AD+/+/TRPV1+/+ mice compared with 3xTg-AD−/−/TRPV1+/+ mice. These differences disappeared in 3xTg-AD+/+/TRPV1+/− and 3xTg-AD+/+/TRPV1−/− mice (Fig. 4A).

The densitometry results showed that Bcl-2 expression levels significantly increased in 3xTg-AD+/+/TRPV1−/− mice compared with 3xTg-AD+/+/TRPV1+/+ mice. Consequently, expression levels of apoptotic factors Bcl-2 (Fig. 4B), Bax (Fig. 4C), cleaved caspase-3 (Fig. 4D), and cleaved PARP (Fig. 4E) decreased in 3xTg-AD+/+/TRPV1−/− mice, which indicates that TRPV1 deficiency blocks apoptosis by activating Bcl-2 and downregulating Bax, cleaved caspase-3 and cleaved PARP expression in the hippocampus.

Memory and cognition deficits are well-known symptoms of AD, and apoptosis in the brain contributes to most of these deficits. Aβ and tau neurofibrillary tangles play a major role in memory and learning deficits. However, Ca2+ dyshomeostasis is reported to be another major cause of AD pathogenesis (Wang et al., 2017b). Ca2+ plays a critical role as a signal carrier regulating synaptic signaling and neurotransmission. It has been reported that Ca2+ is overloaded in Patients with AD (Ichimiya et al., 1988) and the 3xTg-AD mouse model (Jadiya et al., 2019), leading to memory and behavior disorders.

In a previous study, TRPV1 deficiency effectively restored Ca2+ levels in the hippocampus and consequently decreased Aβ and Tau deposition, resulting in anti-AD effects (Kim et al., 2020). Interestingly, there are reports regarding the effect of TRPV1 activation on AD pathology in which TRPV1 activation was shown to be beneficial in other AD mouse models by alleviating cognitive impairments in APP23/PS45 double transgenic AD model mice at the age of 4 months (Du et al., 2020) and alleviating microglia dysfunction in APP/PS1 mice at the age of 7-8 months (Lu et al., 2021). As calcium is an important secondary messanger for cellular growth and death through signal transduction, adequate influx through TRPV1 activation might have positive effects up to a moderate level. However, we used more severe AD-state animal model with 3xTg-AD mice at the age of 12 months. In this model, Aβ aggregation occurs more severely and moreover, tau pathology also occurs. Accordingly, calcium dyshomeostasis was already highly formed and as a result we assumed that TRPV1 dificiency, not activation, has positive effects in the hippocampus.

TRPV1 induces proliferation through Ca2+ entry, ATP release, membrane purinoceptor 2 (P2Y2) receptor activation and EGFR (epidermal growth factor receptor) activity. Activation of TRPV1 results in phosphorylation and activation of ATM serine-threonine kinase, which induces Fas signal. Fas is a subgroup of the TNF-R (tumor necrosis factor receptor) family which trigger apoptotic cell death. As a result, this causes mitochondrial damage and induces endoplasmic reticulum stress, resulting in apoptosis in the hippocampus (Zhai et al., 2020).

Memantine, an N-methyl-D-aspartate receptor (NMDAR) antagonist and treatment for AD, has also been reported to have neuroprotective effects by blocking excessive calcium influx into cells (Robinson and Keating, 2006). NMDAR is a ion channel with a high calcium permeability, in which overexpression of NMDAR accelerates cognitive dysfunction. Interestingly, NMDAR and TRPV1 both mediate signal pathway through Ca2+ influx. The activation of TRPV1 increases Ca2+ influx and activates CaMKII/protein kinase C (PKC) pathway. NMDAR consequently activated by PKC, which induces additional Ca2+ influx, which result in apoptosis in the brain. Similary, TRPV1 is also activated by the increased Ca2+ influx due to NMDAR activation, which inferred that NMDAR and TRPV1 affect each other, not in one direction. These relationship is also shown when the treatment of TRPV1 agonist capsaicin promotes NMDAR activation, whereas treatment of TRPV1 antagonist capsazepine blocks NMDAR activation by inhibiting phosphorylation of NMDAR subunit 2B, which suppresses neuroinflammation (Lee et al., 2012).

Subsequent studies also supported this neuroprotective effect of memantine in reducing hippocampal apoptosis (Wang et al., 2018). Additionally, these studies showed that memantine reduced caspase-3 activity (Wang et al., 2017a) by preventing the loss of BDNF function (Tanqueiro et al., 2018) and activating the BDNF/TrkB/Akt/ERK/CREB pathway (Jantas et al., 2009; Takahashi et al., 2018; Zhu et al., 2015).

Apoptosis is a form of cell death that occurs through a series of molecular steps within the cell (Gorman et al., 2000). TUNEL assay results show that TUNEL-positive cells were significantly increased in the hippocampus’s CA3, CA1, and DG regions in 3xTg-AD+/+/TRPV1+/+ mice. This increase was almost completely prevented by TRPV1 deficiency in all 3 regions of the hippocampus (Fig. 1). These results indicate that TRPV1 deficiency exerts a neuroprotective effect by preventing apoptosis in the hippocampus.

It has been reported that Aβ induces NMDAR activation, leading to intracellular Ca2+ overload, which activates calpain and reduces BDNF levels (Tanqueiro et al., 2018; Vanhoutte and Bading, 2003). This study hypothesized that TRPV1 deficiency protects neurons by inhibiting apoptosis in the hippocampus through Ca2+ normalization. Western blotting revealed that TRPV1 deficiency restores the protein expression of BDNF and the phosphorylation level of its membrane receptor TrkB (Fig. 2), suggesting that BDNF is involved in TRPV1 deficiency-mediated neuroprotective effects. Subsequent western blotting results show that TRPV1 deficiency increases the BDNF downstream of the intracellular proteins Akt and ERK phosphorylation, which activates the CREB (Fig. 3).

When CREB is phosphorylated, p-CREB binds to the Bcl-2 promoter and increases the Bcl-2 expression (Meller et al., 2005). Bcl-2 then binds directly to Bax and inhibits activation and oligomerization of Bax (Fletcher et al., 2008). Therefore, the apoptotic effect of Bax is blocked by the forming pores in the mitochondria (Antonsson et al., 1997). Additionally, Bax plays an essential role in activating caspase-3 by releasing cytochrome c from mitochondria, which interacts with the APAF-1 (apoptotic protease activating factor-1) and activates caspase-3 (Cregan et al., 1999; Elena-Real et al., 2018; Wang, 2001; Yuan et al., 2003). When caspase-3 is cleaved and activated, it cleaves cellular proteins, including PARP, that can repair damaged DNA (Sairanen et al., 2009). Thus, CREB phosphorylation upregulates Bcl-2 and downregulates Bax expression, preventing apoptosis (Eimer and Vassar, 2013; Porter and Jänicke, 1999). In this study, expression levels of Bcl-2 increased, whereas expression levels of the apoptotic factors Bax, cleaved caspase-3, and cleaved PARP decreased in 3xTg-AD+/+/TRPV1−/− mice compared to 3xTg-AD+/+/TRPV1+/+ mice (Fig. 4).

Generally, these findings suggest that TRPV1 deficiency has neuroprotective effects through BDNF/CREB signaling that mediates the Bcl-2 family-related antiapoptotic pathway in the hippocampus (Fig. 5).

In conclusion, a proposed molecular mechanism of neuroprotection by TRPV1 deficiency in 3xTg-AD mice is shown in Fig. 5. TRPV1 deficiency normalizes Ca2+ levels in the 3xTg-AD mouse model (Kim et al., 2020). Consequently, TRPV1 deficiency upregulates the BDNF/TrkB and downstream Akt/ERK/CREB signaling pathways. This signaling upregulates the antiapoptotic factor Bcl-2, which causes the levels of the apoptotic factors Bax, cleaved caspase-3, and cleaved PARP to decrease. In conclusion, TRPV1 deficiency protects hippocampal neurons from hippocampal apoptosis due to AD pathogenesis in the 3xTg-AD mouse model via the BDNF/CREB signaling pathway. Additionally, the normalization of Ca2+ levels by TRPV1 deficiency may be a therapeutic target to suppress memory loss and learning deficits in AD pathogenesis.

This research was supported by intramural grants (2E31881) from the Korea Institute of Science and Technology (KIST) and the Basic Science Research Program (2018R1D1A1B07050182) from the Ministry of Education in Korea.

J.K. (Juyong Kim) and S.S. contributed equally to this work and were responsible for the study design and performing the experiments. J.K. (Juyong Kim) interpreted the results, and S.S. wrote the initial draft of the manuscript. J.K. (Jiyoung Kim) designed this study to investigate the role of cell membrane-bound Ca2+ transporting-TRPV1 deficiency on 3xTg-AD and helped generate multiple genotypes of TRPV1 and 3xTg-AD. J.K. (Jiyoung Kim) also helped identify the involvement of BDNF and apoptosis markers as key factors for TRPV1-deficient 3xTg-AD, which showed improved memory in a previous study. J.H.Y.P. revised the manuscript. J.-C.K. contributed to the interpretation of the results. K.W.L. was responsible for the concept and design of the study, and supervised the work.

Fig. 1. The effect of TRPV1 deficiency on apoptosis in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−and 3xTg-AD+/+/TRPV1−/− mice. , (A) Representative photographs of TUNEL staining in the CA3, CA1, and DG regions. Scale bars = 2.5 μm. (B-D) Quantification of TUNEL staining fluorescence intensity in the (B) CA3, (C) CA1, and (D) DG regions. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 in compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; WT, wild type; AD, Alzheimer’s disease; CA, cornu ammonis; DG, dentate gyrus.
Fig. 2. The effect of TRPV1 deficiency on the expression level of BDNF and p-TrkB protein in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing expression levels of BDNF, p-TrkB, TrkB, and β-actin proteins. (B-D) Western blot densitometry results for (B) BDNF/β-actin, (C) p-TrkB/TrkB, and (D) p-TrkB/β-actin protein expression-level analysis. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; BDNF, brain-derived neurotrophic factor; TrkB, tyrosine receptor kinase B; WT, wild type; AD, Alzheimer’s disease.
Fig. 3. The effect of TRPV1 deficiency on p-Akt, p-ERK, and p-CREB protein expression level in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing p-Akt, Akt, p-ERK, ERK, and β-actin expression levels. (B and C) Western blot densitometry results for (B) p-Akt/Akt and (C) p-ERK/ERK. (D) Representative Western blot bands showing the expression level of p-CREB and CREB. (E and F) Western blot densitometry results for (E) p-CREB/CREB and (F) p-CREB/β-actin. The results are presented as mean ± SEM, #P < 0.05 compared with the WT, *P < 0.05 compared with the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; ERK, extracellular signal-regulated kinase; CREB, cAMP response element binding protein; WT, wild type; AD, Alzheimer’s disease.
Fig. 4. The effect of TRPV1 deficiency on the expression of apoptosis-related proteins in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing expression levels of Bcl-2, Bax, cleaved caspase-3, and cleaved PARP. (B-E) Western blot densitometry results for (B) Bcl-2/GAPDH, (C) Bax/GAPDH, (D) c-Caspase-3/GAPDH, and (E) cleaved PARP/GAPDH. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; WT, wild type; AD, Alzheimer’s disease; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X; PARP, poly (ADP-ribose) polymerase; c-Caspase-3, cleaved caspase-3; c-PARP, cleaved PARP.
Fig. 5. Molecular pathway of neuroprotection by TRPV1 deficiency in 3xTg-AD mice. Ca2+ is overloaded in 3xTg-AD+/+/TRPV1+/+ and as a result, apoptosis occurs (left). In 3xTg-AD+/+/TRPV1−/− mice, the Ca2+ level is normalized and apoptosis is prevented by activation of the BDNF/CREB pathway (right). TRPV1, transient receptor potential vanilloid 1; AD, Alzheimer’s disease; BDNF, brain-derived neurotrophic factor; CREB, cAMP response element binding protein; NMDA, N-methyl-D-aspartate; TrkB, tyrosine receptor kinase B; ERK, extracellular signal-regulated kinase; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X; PARP, poly (ADP-ribose) polymerase. Created with BioRender.com.
  1. Antonsson B., Conti F., Ciavatta A., Montessuit S., Lewis S., Martinou I., Bernasconi L., Bernard A., Mermod J.J., and Mazzei G., et al. (1997). Inhibition of Bax channel-forming activity by Bcl-2. Science 277, 370-372.
    Pubmed CrossRef
  2. Asadi M., Taghizadeh S., Kaviani E., Vakili O., Taheri-Anganeh M., Tahamtan M., and Savardashtaki A. (2022). Caspase‐3: structure, function, and biotechnological aspects. Biotechnol. Appl. Biochem. 69, 1633-1645.
    Pubmed CrossRef
  3. Baker K.D., Edwards T.M., and Rickard N.S. (2013). The role of intracellular calcium stores in synaptic plasticity and memory consolidation. Neurosci. Biobehav. Rev. 37, 1211-1239.
    Pubmed CrossRef
  4. Berridge M.J. (2011). Calcium signalling and Alzheimer's disease. Neurochem. Res. 36, 1149-1156.
    Pubmed CrossRef
  5. Bloom G.S. (2014). Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 71, 505-508.
    Pubmed CrossRef
  6. Bojarski L., Herms J., and Kuznicki J. (2008). Calcium dysregulation in Alzheimer's disease. Neurochem. Int. 52, 621-633.
    Pubmed CrossRef
  7. Calissano P., Matrone C., and Amadoro G. (2009). Apoptosis and in vitro Alzheimer's disease neuronal models. Commun. Integr. Biol. 2, 163-169.
    Pubmed KoreaMed CrossRef
  8. Calvo-Rodriguez M., Hou S.S., Snyder A.C., Kharitonova E.K., Russ A.N., Das S., Fan Z., Muzikansky A., Garcia-Alloza M., and Serrano-Pozo A., et al. (2020). Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer's disease. Nat. Commun. 11, 2146.
    Pubmed KoreaMed CrossRef
  9. Cascella R. and Cecchi C. (2021). Calcium dyshomeostasis in Alzheimer's disease pathogenesis. Int. J. Mol. Sci. 22, 4914.
    Pubmed KoreaMed CrossRef
  10. Corona C., Masciopinto F., Silvestri E., Viscovo A.D., Lattanzio R., Sorda R.L., Ciavardelli D., Goglia F., Piantelli M., and Canzoniero L.M., et al. (2010). Dietary zinc supplementation of 3xTg-AD mice increases BDNF levels and prevents cognitive deficits as well as mitochondrial dysfunction. Cell Death Dis. 1, e91.
    Pubmed KoreaMed CrossRef
  11. Cregan S.P., MacLaurin J.G., Craig C.G., Robertson G.S., Nicholson D.W., Park D.S., and Slack R.S. (1999). Bax-dependent caspase-3 activation is a key determinant in p53-induced apoptosis in neurons. J. Neurosci. 19, 7860-7869.
    Pubmed KoreaMed CrossRef
  12. D'Amelio M., Sheng M., and Cecconi F. (2012). Caspase-3 in the central nervous system: beyond apoptosis. Trends Neurosci. 35, 700-709.
    Pubmed CrossRef
  13. DeTure M.A. and Dickson D.W. (2019). The neuropathological diagnosis of Alzheimer's disease. Mol. Neurodegener. 14, 32.
    Pubmed KoreaMed CrossRef
  14. Du Y., Fu M., Huang Z., Tian X., Li J., Pang Y., Song W., Tian Wang Y., and Dong Z. (2020). TRPV1 activation alleviates cognitive and synaptic plasticity impairments through inhibiting AMPAR endocytosis in APP23/PS45 mouse model of Alzheimer's disease. Aging Cell 19, e13113.
    Pubmed KoreaMed CrossRef
  15. Eimer W.A. and Vassar R. (2013). Neuron loss in the 5XFAD mouse model of Alzheimer's disease correlates with intraneuronal Aβ42 accumulation and Caspase-3 activation. Mol. Neurodegener. 8, 2.
    Pubmed KoreaMed CrossRef
  16. Elena-Real C.A., Díaz-Quintana A., González-Arzola K., Velázquez-Campoy A., Orzáez M., López-Rivas A., Gil-Caballero S., De, la Rosa M.Á., and Díaz-Moreno I. (2018). Cytochrome c speeds up caspase cascade activation by blocking 14-3-3ε-dependent Apaf-1 inhibition. Cell Death Dis. 9, 365.
    Pubmed KoreaMed CrossRef
  17. Ferrer I., Marín C., Rey M.J., Ribalta T., Goutan E., Blanco R., Tolosa E., and Martí E. (1999). BDNF and full-length and truncated TrkB expression in Alzheimer disease. Implications in therapeutic strategies. J. Neuropathol. Exp. Neurol. 58, 729-739.
    Pubmed CrossRef
  18. Finkbeiner S., Tavazoie S.F., Maloratsky A., Jacobs K.M., Harris K.M., and Greenberg M.E. (1997). CREB: a major mediator of neuronal neurotrophin responses. Neuron 19, 1031-1047.
    Pubmed CrossRef
  19. Fletcher J.I., Meusburger S., Hawkins C.J., Riglar D.T., Lee E.F., Fairlie W.D., Huang D.C., and Adams J.M. (2008). Apoptosis is triggered when prosurvival Bcl-2 proteins cannot restrain Bax. Proc. Natl. Acad. Sci. U. S. A. 105, 18081-18087.
    Pubmed KoreaMed CrossRef
  20. Folch J., Busquets O., Ettcheto M., Sánchez-López E., Castro-Torres R.D., Verdaguer E., Garcia M.L., Olloquequi J., Casadesús G., and Beas-Zarate C., et al. (2018). Memantine for the treatment of dementia: a review on its current and future applications. J. Alzheimers Dis. 62, 1223-1240.
    Pubmed KoreaMed CrossRef
  21. Fumagalli F., Racagni G., and Riva M. (2006). The expanding role of BDNF: a therapeutic target for Alzheimer's disease? Pharmacogenomics J. 6, 8-15.
    Pubmed CrossRef
  22. Gorman A.M., Ceccatelli S., and Orrenius S. (2000). Role of mitochondria in neuronal apoptosis. Dev. Neurosci. 22, 348-358.
    Pubmed CrossRef
  23. Green K.N. and LaFerla F.M. (2008). Linking calcium to Abeta and Alzheimer's disease. Neuron 59, 190-194.
    Pubmed CrossRef
  24. Hippius H. and Neundörfer G. (2003). The discovery of Alzheimer's disease. Dialogues Clin. Neurosci. 5, 101-108.
    Pubmed KoreaMed CrossRef
  25. Ichimiya Y., Emson P.C., Mountjoy C.Q., Lawson D.E.M., and Heizmann C.W. (1988). Loss of calbindin-28K immunoreactive neurones from the cortex in Alzheimer-type dementia. Brain Res. 475, 156-159.
    Pubmed CrossRef
  26. Impey S., Obrietan K., and Storm D.R. (1999). Making new connections: role of ERK/MAP kinase signaling in neuronal plasticity. Neuron 23, 11-14.
    Pubmed CrossRef
  27. Jadiya P., Kolmetzky D.W., Tomar D., Di Meco A., Lombardi A.A., Lambert J.P., Luongo T.S., Ludtmann M.H., Pratico D., and Elrod J.W. (2019). Impaired mitochondrial calcium efflux contributes to disease progression in models of Alzheimer's disease. Nat. Commun. 10, 3885.
    Pubmed KoreaMed CrossRef
  28. Jantas D., Szymanska M., Budziszewska B., and Lason W. (2009). An involvement of BDNF and PI3-K/Akt in the anti-apoptotic effect of memantine on staurosporine-evoked cell death in primary cortical neurons. Apoptosis 14, 900-912.
    Pubmed CrossRef
  29. Kim J., Lee S., Kim J., Ham S., Park J.H.Y., Han S., Jung Y.K., Shim I., Han J.S., and Lee K.W., et al. (2020). Ca2+-permeable TRPV1 pain receptor knockout rescues memory deficits and reduces amyloid-β and tau in a mouse model of Alzheimer's disease. Hum. Mol. Genet. 29, 228-237.
    Pubmed CrossRef
  30. Ku B., Liang C., Jung J.U., and Oh B.H. (2011). Evidence that inhibition of BAX activation by BCL-2 involves its tight and preferential interaction with the BH3 domain of BAX. Cell Res. 21, 627-641.
    Pubmed KoreaMed CrossRef
  31. LeBlanc A.C. (2005). The role of apoptotic pathways in Alzheimer's disease neurodegeneration and cell death. Curr. Alzheimer Res. 2, 389-402.
    Pubmed CrossRef
  32. Lee J., Saloman J.L., Weiland G., Auh Q.S., Chung M.K., and Ro J.Y. (2012). Functional interactions between NMDA receptors and TRPV1 in trigeminal sensory neurons mediate mechanical hyperalgesia in the rat masseter muscle. Pain 153, 1514-1524.
    Pubmed KoreaMed CrossRef
  33. Lu J., Zhou W., Dou F., Wang C., and Yu Z. (2021). TRPV1 sustains microglial metabolic reprogramming in Alzheimer's disease. EMBO Rep. 22, e52013.
    Pubmed KoreaMed CrossRef
  34. Meller R., Minami M., Cameron J.A., Impey S., Chen D., Lan J.Q., Henshall D.C., and Simon R.P. (2005). CREB-mediated Bcl-2 protein expression after ischemic preconditioning. J. Cereb. Blood Flow Metab. 25, 234-246.
    Pubmed CrossRef
  35. Minichiello L., Calella A.M., Medina D.L., Bonhoeffer T., Klein R., and Korte M. (2002). Mechanism of TrkB-mediated hippocampal long-term potentiation. Neuron 36, 121-137.
    Pubmed CrossRef
  36. Miranda M., Morici J.F., Zanoni M.B., and Bekinschtein P. (2019). Brain-derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain. Front. Cell. Neurosci. 13, 363.
    Pubmed KoreaMed CrossRef
  37. Park E.M. and Cho S. (2006). Enhanced ERK dependent CREB activation reduces apoptosis in staurosporine-treated human neuroblastoma SK-N-BE (2) C cells. Neurosci. Lett. 402, 190-194.
    Pubmed CrossRef
  38. Porter A.G. and Jänicke R.U. (1999). Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 6, 99-104.
    Pubmed CrossRef
  39. Rai S.N., Dilnashin H., Birla H., Singh S.S., Zahra W., Rathore A.S., Singh B.K., and Singh S.P. (2019). The role of PI3K/Akt and ERK in neurodegenerative disorders. Neurotox. Res. 35, 775-795.
    Pubmed CrossRef
  40. Robinson D.M. and Keating G.M. (2006). Memantine: a review of its use in Alzheimer's disease. Drugs 66, 1515-1534.
    Pubmed CrossRef
  41. Sairanen T., Szepesi R., Karjalainen-Lindsberg M.L., Saksi J., Paetau A., and Lindsberg P.J. (2009). Neuronal caspase-3 and PARP-1 correlate differentially with apoptosis and necrosis in ischemic human stroke. Acta Neuropathol. 118, 541-552.
    Pubmed CrossRef
  42. Santos A.R., Comprido D., and Duarte C.B. (2010). Regulation of local translation at the synapse by BDNF. Prog. Neurobiol. 92, 505-516.
    Pubmed CrossRef
  43. Saura C.A. and Valero J. (2011). The role of CREB signaling in Alzheimer's disease and other cognitive disorders. Rev. Neurosci. 22, 153-169.
    Pubmed CrossRef
  44. Scheff S.W. and Price D.A. (2006). Alzheimer's disease-related alterations in synaptic density: neocortex and hippocampus. J. Alzheimers Dis. 9((3 Suppl)), 101-115.
    Pubmed CrossRef
  45. Shacka J.J. and Roth K.A. (2005). Regulation of neuronal cell death and neurodegeneration by members of the Bcl-2 family: therapeutic implications. Curr. Drug Targets CNS Neurol. Disord. 4, 25-39.
    Pubmed CrossRef
  46. Takahashi K., Nakagawasai O., Nemoto W., Kadota S., Isono J., Odaira T., Sakuma W., Arai Y., Tadano T., and Tan-No K. (2018). Memantine ameliorates depressive-like behaviors by regulating hippocampal cell proliferation and neuroprotection in olfactory bulbectomized mice. Neuropharmacology 137, 141-155.
    Pubmed CrossRef
  47. Tanqueiro S.R., Ramalho R.M., Rodrigues T.M., Lopes L.V., Sebastião A.M., and Diógenes M.J. (2018). Inhibition of NMDA receptors prevents the loss of BDNF function induced by amyloid β. Front. Pharmacol. 9, 237.
    Pubmed KoreaMed CrossRef
  48. Vanhoutte P. and Bading H. (2003). Opposing roles of synaptic and extrasynaptic NMDA receptors in neuronal calcium signalling and BDNF gene regulation. Curr. Opin. Neurobiol. 13, 366-371.
    Pubmed CrossRef
  49. Wang X. (2001). The expanding role of mitochondria in apoptosis. Genes Dev. 15, 2922-2933.
    Pubmed
  50. Wang X., Chen J., Wang H., Yu H., Wang C., You J., Wang P., Feng C., Xu G., and Wu X., et al. (2017a). Memantine can reduce ethanol-induced caspase-3 activity and apoptosis in H4 cells by decreasing intracellular calcium. J. Mol. Neurosci. 62, 402-411.
    Pubmed CrossRef
  51. Wang X., Yu H., You J., Wang C., Feng C., Liu Z., Li Y., Wei R., Xu S., and Zhao R., et al. (2018). Memantine can improve chronic ethanol exposure-induced spatial memory impairment in male C57BL/6 mice by reducing hippocampal apoptosis. Toxicology 406-407, 21-32.
    Pubmed CrossRef
  52. Wang Y., Shi Y., and Wei H. (2017b). Calcium dysregulation in Alzheimer's disease: a target for new drug development. J. Alzheimers Dis. Parkinsonism 7, 374.
    Pubmed KoreaMed CrossRef
  53. Yin X.M., Oltvai Z.N., and Korsmeyer S.J. (1994). BH1 and BH2 domains of Bcl-2 are required for inhibition of apoptosis and heterodimerization with Bax. Nature 369, 321-323.
    Pubmed CrossRef
  54. Yuan J., Murrell G.A., Trickett A., and Wang M.X. (2003). Involvement of cytochrome c release and caspase-3 activation in the oxidative stress-induced apoptosis in human tendon fibroblasts. Biochim. Biophys. Acta 1641, 35-41.
    Pubmed CrossRef
  55. Zarneshan S.N., Fakhri S., and Khan H. (2022). Targeting Akt/CREB/BDNF signaling pathway by ginsenosides in neurodegenerative diseases: a mechanistic approach. Pharmacol. Res. 177, 106099.
    Pubmed CrossRef
  56. Zhai K., Liskova A., Kubatka P., and Büsselberg D. (2020). Calcium entry through TRPV1: a potential target for the regulation of proliferation and apoptosis in cancerous and healthy cells. Int. J. Mol. Sci. 21, 4177.
    Pubmed KoreaMed CrossRef
  57. Zheng Z., Sabirzhanov B., and Keifer J. (2010). Oligomeric amyloid-β inhibits the proteolytic conversion of brain-derived neurotrophic factor (BDNF), AMPA receptor trafficking, and classical conditioning. J. Biol. Chem. 285, 34708-34717.
    Pubmed KoreaMed CrossRef
  58. Zhu G., Li J., He L., Wang X., and Hong X. (2015). MPTP‐induced changes in hippocampal synaptic plasticity and memory are prevented by memantine through the BDNF‐TrkB pathway. Br. J. Pharmacol. 172, 2354-2368.
    Pubmed KoreaMed CrossRef

Article

Research Article

Mol. Cells 2023; 46(5): 319-328

Published online May 31, 2023 https://doi.org/10.14348/molcells.2023.2156

Copyright © The Korean Society for Molecular and Cellular Biology.

Ca2+-Permeable TRPV1 Receptor Mediates Neuroprotective Effects in a Mouse Model of Alzheimer’s Disease via BDNF/CREB Signaling Pathway

Juyong Kim1,2,7 , Sangwoo Seo1,7 , Jung Han Yoon Park3 , Ki Won Lee1,3,4,5 , Jiyoung Kim5,* , and Jin-Chul Kim2,6,*

1Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea, 2Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea, 3Bio-MAX Institute, Seoul National University, Seoul 08826, Korea, 4Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Korea, 5Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea, 6Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Korea, 7These authors contributed equally to this work.

Correspondence to:jckim@kist.re.kr(JCK); jiyoungkim1107@snu.ac.kr(JK)

Received: October 12, 2022; Revised: January 4, 2023; Accepted: January 6, 2023

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.

Abstract

Transient receptor potential vanilloid 1 (TRPV1) protein is a Ca2+-permeable non-selective cation channel known for its pain modulation pathway. In a previous study, it was discovered that a triple-transgenic Alzheimer’s disease (AD) mouse model (3xTg-AD+/+) has anti-AD effects. The expression of proteins in the brain-derived neurotrophic factor (BDNF)/cAMP response element binding protein (CREB) pathway in a 3xTg-AD/TRPV1 transgenic mice model was investigated to better understand the AD regulatory effect of TRPV1 deficiency. The results show that TRPV1 deficiency leads to CREB activation by increasing BDNF levels and promoting phosphorylation of tyrosine receptor kinase B (TrkB), extracellular signal-regulated kinase (ERK), protein kinase B (Akt), and CREB in the hippocampus. Additionally, TRPV1 deficiency-induced CREB activation increases the antiapoptotic factor B-cell lymphoma 2 (Bcl-2) gene, which consequently downregulates Bcl-2-associated X (Bax) expression and decreases cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase (PARP), which leads to the prevention of hippocampal apoptosis. In conclusion, TRPV1 deficiency exhibits neuroprotective effects by preventing apoptosis through the BDNF/CREB signal transduction pathway in the hippocampus of 3xTg-AD mice.

Keywords: Alzheimer’s disease, apoptosis, brain-derived neurotrophic factor, cAMP response element binding protein, transient receptor potential vanilloid 1

INTRODUCTION

Alzheimer’s disease (AD) is one of the most representative age-related neurodegenerative diseases accompanied by memory and cognitive deficits (Hippius and Neundörfer, 2003). Although Alois AD has been known for more than a century, there is still no effective treatment for it. Several factors contribute to AD. The most notable hypotheses for AD development are the amyloid hypothesis and the hyperphosphorylated tau hypothesis (Bloom, 2014; DeTure and Dickson, 2019). The deposition of both amyloid-β peptide (Aβ) and tau neurofibrillary tangles causes several pathological symptoms, including synaptic degradation, neuroinflammation, and neuronal cell loss, which induce memory and cognition impairment and finally develop into severe dementia (LeBlanc, 2005).

In addition to amyloid and hyperphosphorylated tau, calcium (Ca2+) dyshomeostasis is deeply implicated in the pathogenesis of AD (Bojarski et al., 2008; Cascella and Cecchi, 2021; Green and LaFerla, 2008). Ca2+ acts as an intracellular messengers and involves diverse signal transmission activities, including enzyme activation (through phosphorylation or dephosphorylation), exocytosis, gene transcription and programmed cell death (Baker et al., 2013). These actions of intracellular Ca2+ signaling in neurons affect neural growth, memory formation, and synaptic plasticity. In other words, Ca2+ dyshomeostasis causes damage and eventually leads to cell death in the hippocampus.

It has been reported that soluble Aβ aggregates and increases neuronal Ca2+ influx in patients with AD, resulting in Ca2+ overload in neurons (Berridge, 2011). As a result, resting Ca2+ increases lead to Ca2+ dysregulation, which includes several pathological features such as neuronal death and degeneration (Calvo-Rodriguez et al., 2020; Jadiya et al., 2019). Previously, we focused on these aspects and tried mediating AD pathogenesis by regulating Ca2+ levels through transient receptor potential vanilloid 1 (TRPV1), a Ca2+-permeable nonselective cation channel well-known for regulating pain and inflammation. A triple-transgenic AD (3xTg-AD+/+) mouse model was used to characterize the anti-AD effect of TRPV1 deficiency (Kim et al., 2020). In that study, TRPV1 deficiency alleviated Aβ and Tau deposition and consequently improved memory and learning deficits in the 3xTg-AD+/+ mouse model. Additionally, it was shown that Ca2+ was overloaded in 3xTg-AD+/+ mice, resulting in memory and learning deficits. However, in TRPV1 receptor knockout 3xTg-AD mice (3xTg-AD+/+/TRPV1−/−), after normalized Ca2+ levels, memory and learning abilities were restored.

The hippocampus is a curved-shaped region in the middle of the brain, essential for learning and memory. Therefore, damage to the hippocampus can lead to severe memory deficits, including memory formation and consolidation. Decreased hippocampal synaptic density in Patients with AD has been reported, which is highly correlated with cognitive deficits (Scheff and Price, 2006). Additionally, several causes of neuronal apoptosis, such as Aβ accumulation, increased oxidative stress, and DNA fragmentation, have also been reported in the brain of Patients with AD (Calissano et al., 2009). These previous studies showed that apoptosis in the brain is closely related to cognitive ability in patients with AD.

Brain-derived neurotrophic factor (BDNF) is a major neurotrophic factor that regulates neuronal survival, differentiation, and plasticity (Miranda et al., 2019). The expression of BDNF and its receptor, tyrosine receptor kinase B (TrkB), is reduced in Patients with AD (Ferrer et al., 1999; Zheng et al., 2010). There is also evidence that Aβ directly or indirectly reduces BDNF levels, leading to memory and cognition deficits (Corona et al., 2010). Therefore, the expression levels of BDNF and TrkB are important for evaluating the neuroprotective effect of TRPV1 deficiency in AD models.

TrkB activation triggers the activation of several downstream pathways, including phosphorylation of extracellular signal-regulated kinase (ERK) and protein kinase B (Akt), which in turn phosphorylates and activates cAMP response element binding protein (CREB) (Minichiello et al., 2002; Santos et al., 2010). ERK/MAPK (mitogen-activated protein kinase) is related to memory formation (Impey et al., 1999) and antiapoptotic effects (Park and Cho, 2006). Akt also plays an important role in neuroprotection (Zarneshan et al., 2022). CREB is a transcription factor that regulates the proliferation or survival of neurons by binding to cAMP-responsive elements, thereby modulating the transcription of the genes (Finkbeiner et al., 1997).

B-cell lymphoma 2 (Bcl-2) family proteins play a critical role in the apoptotic activity. The Bcl-2 family includes both the antiapoptotic protein Bcl-2 and the proapoptotic protein Bcl-2-associated X (Bax) or Bcl-2 homologous antagonist killer (Bak) (Shacka and Roth, 2005). When CREB is phosphorylated, it directly binds to the Bcl-2 family promoter, activating the signaling cascade (Meller et al., 2005). Bcl-2 forms a heterodimer with the apoptotic protein Bax and inhibits the activation of Bax, which regulates the activation of caspase-3 (Ku et al., 2011; Yin et al., 1994). Cleaved caspase-3 is known to be involved in the death of mammalian neural cells (D’Amelio et al., 2012). Activated caspase-3 cleaves cellular proteins, including poly (ADP-ribose) polymerase (PARP). Because PARP is a crucial enzyme in the repair of DNA breakage, its cleavage causes neurons to undergo apoptosis (Sairanen et al., 2009).

In this study, we aimed to understand how neurological parameters are related to the neuroprotective effect by preventing apoptosis in the hippocampus by using TRPV1 receptor knockout 3xTg-AD mice to explore the effect of TRPV1 deficiency in the hippocampus. It was found that TRPV1 deficiency inhibits hippocampal cell death by promoting the BDNF/TrkB signaling pathway. Furthermore, activating TrkB leads to the activation of CREB, which inhibits the activated caspase-3 dependent apoptosis.

MATERIALS AND METHODS

Animals

3xTg-AD mice (kindly given by Professor Frank M. Laferla, UC-Irvine, Irvine, CA, USA) were homozygous for APPswe, TauP301L, and PS1M146V. These 3xTg-AD mice were mated with TRPV1−/− mice (stock #003770; provided from Jackson Laboratories, Bar Harbor, ME, USA) and offspring with nine genotypes (3xTg-AD−/−/TRPV1+/+, 3xTg-AD−/−/TRPV1+/−, 3xTg-AD−/−/TRPV1−/−, 3xTg-AD+/−/TRPV1+/+, 3xTg-AD+/−/TRPV1+/−, 3xTg-AD+/−/TRPV1−/−, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, 3xTg-AD+/+/TRPV1−/−) were generated. Four groups were selected with or without AD pathology and TRPV1 depletion. The selected groups were 3xTg-AD−/−/TRPV1+/+ mice (wild type, WT), 3xTg-AD+/+/TRPV1+/+ mice, 3xTg-AD+/+/TRPV1+/− mice and 3xTg-AD+/+/TRPV1−/− mice. Mice in each group were raised until 12 months. Mice were maintained at 21°C ± 2°C, 12-h light:12-h dark photoperiod with specific pathogen-free conditions in ventilated cages and provided with free choice to consume food and water. All animal testing was performed following protocols approved by the Institutional Animal Care and Use Committee of Seoul National University (SNU-140410-12, SNU-140410-14, SNU-140625-2).

Animal dissection and tissue collection

Zoletil (50 mg/kg) was administered intraperitoneally to anesthetize the mice (n = 5) in each group before normal saline, and a 4% paraformaldehyde solution was transcardially administered. The brains were removed and postfixed in 4% paraformaldehyde overnight at 4°C, and routine-frozen sections (10 μm) were prepared for TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining and immunohistochemistry analysis. The other half of the mice in each group were sacrificed by decapitation, and their brains were rapidly collected and cut on an ice-cooled board. The hippocampus was dissected and stored at −80°C for TUNEL staining and western blotting analysis.

TUNEL staining

TUNEL staining was performed on the frozen-embedded sections using the in-situ cell death detection kit (Promega, USA) according to the standard protocol provided by the manufacturer. Apoptotic nuclei were visualized using the peroxidase-DAB reaction. The sections were counterstained with hematoxylin. TUNEL-positive cells in the hippocampus were analyzed in each of the 3 regions of Cornu Ammonis 3 (CA3), Cornu Ammonis 1 (CA1) (especially the soma of pyramidal neurons), and dentate gyrus (DG) sections per high-power field (×40). TUNEL staining fluorescence signal intensity was measured using the ImageJ analysis program (National Institute of Health, USA). Regions of interest were selected from the hippocampus’s CA3, CA1, and DG regions.

Western blotting

Five mice’s hippocampus tissues were removed, and they were all individually examined. There was no replication. Samples were prepared by homogenizing the hippocampus tissues. The protein concentrations were determined using the Bio-Rad protein assay (Bio-Rad, USA). Western blotting analyses were performed on 50 μg of protein. Samples were briefly separated using 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then transferred to a polyvinylidene fluoride membrane (0.4 μm; Merck Millipore, USA). The membranes were blocked in a 5% bovine serum albumin (Bovogen Biologicals, Australia) in tris-buffered saline and Tween-20 (Junsei Chemical, Japan). Membranes were incubated overnight at 4°C with primary antibodies against BDNF (1:1,000; Santa Cruz Biotechnology, USA), p-TrkB (1:1,000; Cell Signaling Technology, USA), TrkB (80E3, 1:1,000; Cell Signaling Technology), p-Akt (Ser473, 1:1,000; Cell Signaling Technology), Akt (1:1,000; Cell Signaling Technology), p-ERK (E-4, 1:1,000; Santa Cruz Biotechnology), ERK (1:1,000; Santa Cruz Biotechnology), p-CREB (Ser133, D1G6, 1:1,000; Cell Signaling Technology), CREB (48H2, 1:1,000; Cell Signaling Technology), Bax (1:1,000; Santa Cruz Biotechnology), Bcl-2 (1:1,000; Santa Cruz Biotechnology), cleaved caspase-3 (1:500; Cell Signaling Technology), caspase-3 (1:500; Abcam, USA), cleaved PARP (1:1,000; Cell Signaling Technology), GAPDH (1:1,000; Cell Signaling Technology), and β-actin (1:5,000; Sigma, USA). After incubation with horseradish peroxidase-conjugated secondary antibodies (1:5,000; Sigma) for 1 h at room temperature, immunodetection was performed using an enhanced chemiluminescence detection kit (GE Healthcare, USA). The relative density of each band was measured using the ImageJ analysis program (National Institute of Health).

Statistical analysis

IBM SPSS Statistics for Windows (ver. 19.0; IBM, USA) was used for the statistical analyzes. The results were presented as mean ± SEM values. All the data were analyzed using the Fisher’s LSD (least significant difference).

RESULTS

TRPV1 deficiency reduces apoptosis in the hippocampus of 3xTg-AD mice

To investigate the neuroprotective effect of TRPV1 deficiency, we performed a TUNEL assay to detect hippocampal apoptosis in WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. CA3, CA1, and DG regions of the hippocampus were subjected. Nuclei of apoptotic cells in each region are shown in green fluorescence. The immunofluorescence results showed that TUNEL-positive apoptotic cells were remarkably increased in all hippocampus regions of 3xTg-AD+/+/TRPV1+/+ mice compared with WT mice. However, apoptotic cells tremendously decreased in all 3 regions of 3xTg-AD+/+/TRPV1−/− mice compared with 3xTg-AD+/+/TRPV1+/+ mice (Fig. 1A). Quantitative analysis also showed that TRPV1 deficiency significantly reduced apoptotic cells in CA3 (Fig. 1B), CA1 (Fig. 1C), and DG (Fig. 1D) regions by 67%, 67%, and 62%, respectively, compared with the same region in 3xTg-AD+/+/TRPV1+/+ mice. These results indicate that TRPV1 deficiency protects hippocampal neurons and neuroglial cells from apoptosis due to AD pathogenesis.

TRPV1 deficiency increases BDNF expression levels and promotes TrkB receptor phosphorylation in the hippocampus of 3xTg-AD mice

Western blotting analyses were conducted to examine whether TRPV1 deficiency increases the levels of BDNF and activation of its receptor protein TrkB. BDNF expression levels significantly decreased in 3xTg-AD+/+/TRPV1+/+ mice by 26% compared with 3xTg-AD−/−/TRPV1+/+ mice. However, BDNF expression levels significantly increased in 3xTg-AD+/+/TRPV1−/− mice by 625% compared with 3xTg-AD+/+/TRPV1+/+ mice (Figs. 2A and 2B). Similarly, p-TrkB expression levels proportional to TrkB and β-actin significantly decreased in 3xTg-AD+/+/TRPV1+/+ mice by 40% and 29%, respectively compared with 3xTg-AD−/−/TRPV1+/+ mice. p-TrkB expression levels also significantly increased in 3xTg-AD+/+/TRPV1−/− mice by 268% and 310% respectively compared with 3xTg-AD+/+/TRPV1+/+ mice (Figs. 2C and 2D). These results indicate that TRPV1 deficiency increases the levels of BDNF and promotes the phosphorylation of TrkB in the hippocampus of 3xTg-AD mice. As previously mentioned, BDNF is responsible for synaptic plasticity and neuronal survival and is reported to be impaired in patients with AD. In this respect, BDNF-signaling recovery is considered a promising therapeutic approach to treating AD (Fumagalli et al., 2006). Memantine, a well-known AD drug, has also been reported to reduce preclinical AD pathology by increasing BDNF protein levels, supporting the usefulness of our results (Folch et al., 2018).

TRPV1 deficiency increases the phosphorylation of ERK, Akt, and CREB

To investigate whether the activation of TrkB leads to activation of the downstream signaling of BDNF/TrkB, we measured the phosphorylation level of Akt and ERK. Akt and ERK independently mediate the BDNF/CREB pathway, where the downstream protein CREB plays an important role in memory formation by regulating BDNF (Saura and Valero, 2011). Western blotting revealed that the levels of p-Akt and p-ERK were decreased in the hippocampus of 3xTg-AD+/+/TRPV1+/+ mice compared to 3xTg-AD−/−/TRPV1+/+ mice, and the difference disappeared in 3xTg-AD+/+/TRPV1+/− and 3xTg-AD+/+/TRPV1−/− mice (Fig. 3A). The densitometry results show that TRPV1 deficiency induces the phosphorylation of Akt (Fig. 3B) and ERK (Fig. 3C) whereas the basal levels of Akt and ERK did not change. We then investigated the expression and phosphorylation level of CREB, which plays an important role in mediating the apoptosic pathway. In 3xTg-AD+/+/TRPV1+/+ mice, CREB phosphorylation levels decreased compare with WT. However, CREB phosphorylation levels in 3xTg-AD+/+/TRPV1−/− mice were significantly increased, similar to those in WT mice. Previous studies have reported that Akt and ERK mediate signal transduction in various neurodegenerative diseases, including AD (Rai et al., 2019). Similarly, our results indicate that TRPV1 deficiency in the hippocampus of 3xTg-AD mice has a neuroprotective effect by phosphorylating and activating Akt and ERK, thereby activating the CREB pathway (Figs. 3D-3F).

TRPV1 deficiency reduces the cleavage of caspase-3 and PARP in 3xTg-AD mice

As shown in Fig. 1, TRPV1 deficiency reduces apoptosis in the hippocampus of 3xTg-AD mice. As an executioner of apoptosis, caspase-3 is an essential regulator in AD progression (Asadi et al., 2022). In this regard, we investigated how TRPV1 deficiency reduces apoptosis via the regulation of caspase-3 in the hippocampus of 3xTg-AD mice. Western blotting revealed that the level of Bcl-2 was decreased, whereas Bax, cleaved-caspase-3, and cleaved PARP increased in the hippocampus of 3xTg-AD+/+/TRPV1+/+ mice compared with 3xTg-AD−/−/TRPV1+/+ mice. These differences disappeared in 3xTg-AD+/+/TRPV1+/− and 3xTg-AD+/+/TRPV1−/− mice (Fig. 4A).

The densitometry results showed that Bcl-2 expression levels significantly increased in 3xTg-AD+/+/TRPV1−/− mice compared with 3xTg-AD+/+/TRPV1+/+ mice. Consequently, expression levels of apoptotic factors Bcl-2 (Fig. 4B), Bax (Fig. 4C), cleaved caspase-3 (Fig. 4D), and cleaved PARP (Fig. 4E) decreased in 3xTg-AD+/+/TRPV1−/− mice, which indicates that TRPV1 deficiency blocks apoptosis by activating Bcl-2 and downregulating Bax, cleaved caspase-3 and cleaved PARP expression in the hippocampus.

DISCUSSION

Memory and cognition deficits are well-known symptoms of AD, and apoptosis in the brain contributes to most of these deficits. Aβ and tau neurofibrillary tangles play a major role in memory and learning deficits. However, Ca2+ dyshomeostasis is reported to be another major cause of AD pathogenesis (Wang et al., 2017b). Ca2+ plays a critical role as a signal carrier regulating synaptic signaling and neurotransmission. It has been reported that Ca2+ is overloaded in Patients with AD (Ichimiya et al., 1988) and the 3xTg-AD mouse model (Jadiya et al., 2019), leading to memory and behavior disorders.

In a previous study, TRPV1 deficiency effectively restored Ca2+ levels in the hippocampus and consequently decreased Aβ and Tau deposition, resulting in anti-AD effects (Kim et al., 2020). Interestingly, there are reports regarding the effect of TRPV1 activation on AD pathology in which TRPV1 activation was shown to be beneficial in other AD mouse models by alleviating cognitive impairments in APP23/PS45 double transgenic AD model mice at the age of 4 months (Du et al., 2020) and alleviating microglia dysfunction in APP/PS1 mice at the age of 7-8 months (Lu et al., 2021). As calcium is an important secondary messanger for cellular growth and death through signal transduction, adequate influx through TRPV1 activation might have positive effects up to a moderate level. However, we used more severe AD-state animal model with 3xTg-AD mice at the age of 12 months. In this model, Aβ aggregation occurs more severely and moreover, tau pathology also occurs. Accordingly, calcium dyshomeostasis was already highly formed and as a result we assumed that TRPV1 dificiency, not activation, has positive effects in the hippocampus.

TRPV1 induces proliferation through Ca2+ entry, ATP release, membrane purinoceptor 2 (P2Y2) receptor activation and EGFR (epidermal growth factor receptor) activity. Activation of TRPV1 results in phosphorylation and activation of ATM serine-threonine kinase, which induces Fas signal. Fas is a subgroup of the TNF-R (tumor necrosis factor receptor) family which trigger apoptotic cell death. As a result, this causes mitochondrial damage and induces endoplasmic reticulum stress, resulting in apoptosis in the hippocampus (Zhai et al., 2020).

Memantine, an N-methyl-D-aspartate receptor (NMDAR) antagonist and treatment for AD, has also been reported to have neuroprotective effects by blocking excessive calcium influx into cells (Robinson and Keating, 2006). NMDAR is a ion channel with a high calcium permeability, in which overexpression of NMDAR accelerates cognitive dysfunction. Interestingly, NMDAR and TRPV1 both mediate signal pathway through Ca2+ influx. The activation of TRPV1 increases Ca2+ influx and activates CaMKII/protein kinase C (PKC) pathway. NMDAR consequently activated by PKC, which induces additional Ca2+ influx, which result in apoptosis in the brain. Similary, TRPV1 is also activated by the increased Ca2+ influx due to NMDAR activation, which inferred that NMDAR and TRPV1 affect each other, not in one direction. These relationship is also shown when the treatment of TRPV1 agonist capsaicin promotes NMDAR activation, whereas treatment of TRPV1 antagonist capsazepine blocks NMDAR activation by inhibiting phosphorylation of NMDAR subunit 2B, which suppresses neuroinflammation (Lee et al., 2012).

Subsequent studies also supported this neuroprotective effect of memantine in reducing hippocampal apoptosis (Wang et al., 2018). Additionally, these studies showed that memantine reduced caspase-3 activity (Wang et al., 2017a) by preventing the loss of BDNF function (Tanqueiro et al., 2018) and activating the BDNF/TrkB/Akt/ERK/CREB pathway (Jantas et al., 2009; Takahashi et al., 2018; Zhu et al., 2015).

Apoptosis is a form of cell death that occurs through a series of molecular steps within the cell (Gorman et al., 2000). TUNEL assay results show that TUNEL-positive cells were significantly increased in the hippocampus’s CA3, CA1, and DG regions in 3xTg-AD+/+/TRPV1+/+ mice. This increase was almost completely prevented by TRPV1 deficiency in all 3 regions of the hippocampus (Fig. 1). These results indicate that TRPV1 deficiency exerts a neuroprotective effect by preventing apoptosis in the hippocampus.

It has been reported that Aβ induces NMDAR activation, leading to intracellular Ca2+ overload, which activates calpain and reduces BDNF levels (Tanqueiro et al., 2018; Vanhoutte and Bading, 2003). This study hypothesized that TRPV1 deficiency protects neurons by inhibiting apoptosis in the hippocampus through Ca2+ normalization. Western blotting revealed that TRPV1 deficiency restores the protein expression of BDNF and the phosphorylation level of its membrane receptor TrkB (Fig. 2), suggesting that BDNF is involved in TRPV1 deficiency-mediated neuroprotective effects. Subsequent western blotting results show that TRPV1 deficiency increases the BDNF downstream of the intracellular proteins Akt and ERK phosphorylation, which activates the CREB (Fig. 3).

When CREB is phosphorylated, p-CREB binds to the Bcl-2 promoter and increases the Bcl-2 expression (Meller et al., 2005). Bcl-2 then binds directly to Bax and inhibits activation and oligomerization of Bax (Fletcher et al., 2008). Therefore, the apoptotic effect of Bax is blocked by the forming pores in the mitochondria (Antonsson et al., 1997). Additionally, Bax plays an essential role in activating caspase-3 by releasing cytochrome c from mitochondria, which interacts with the APAF-1 (apoptotic protease activating factor-1) and activates caspase-3 (Cregan et al., 1999; Elena-Real et al., 2018; Wang, 2001; Yuan et al., 2003). When caspase-3 is cleaved and activated, it cleaves cellular proteins, including PARP, that can repair damaged DNA (Sairanen et al., 2009). Thus, CREB phosphorylation upregulates Bcl-2 and downregulates Bax expression, preventing apoptosis (Eimer and Vassar, 2013; Porter and Jänicke, 1999). In this study, expression levels of Bcl-2 increased, whereas expression levels of the apoptotic factors Bax, cleaved caspase-3, and cleaved PARP decreased in 3xTg-AD+/+/TRPV1−/− mice compared to 3xTg-AD+/+/TRPV1+/+ mice (Fig. 4).

Generally, these findings suggest that TRPV1 deficiency has neuroprotective effects through BDNF/CREB signaling that mediates the Bcl-2 family-related antiapoptotic pathway in the hippocampus (Fig. 5).

In conclusion, a proposed molecular mechanism of neuroprotection by TRPV1 deficiency in 3xTg-AD mice is shown in Fig. 5. TRPV1 deficiency normalizes Ca2+ levels in the 3xTg-AD mouse model (Kim et al., 2020). Consequently, TRPV1 deficiency upregulates the BDNF/TrkB and downstream Akt/ERK/CREB signaling pathways. This signaling upregulates the antiapoptotic factor Bcl-2, which causes the levels of the apoptotic factors Bax, cleaved caspase-3, and cleaved PARP to decrease. In conclusion, TRPV1 deficiency protects hippocampal neurons from hippocampal apoptosis due to AD pathogenesis in the 3xTg-AD mouse model via the BDNF/CREB signaling pathway. Additionally, the normalization of Ca2+ levels by TRPV1 deficiency may be a therapeutic target to suppress memory loss and learning deficits in AD pathogenesis.

ACKNOWLEDGMENTS

This research was supported by intramural grants (2E31881) from the Korea Institute of Science and Technology (KIST) and the Basic Science Research Program (2018R1D1A1B07050182) from the Ministry of Education in Korea.

AUTHOR CONTRIBUTIONS

J.K. (Juyong Kim) and S.S. contributed equally to this work and were responsible for the study design and performing the experiments. J.K. (Juyong Kim) interpreted the results, and S.S. wrote the initial draft of the manuscript. J.K. (Jiyoung Kim) designed this study to investigate the role of cell membrane-bound Ca2+ transporting-TRPV1 deficiency on 3xTg-AD and helped generate multiple genotypes of TRPV1 and 3xTg-AD. J.K. (Jiyoung Kim) also helped identify the involvement of BDNF and apoptosis markers as key factors for TRPV1-deficient 3xTg-AD, which showed improved memory in a previous study. J.H.Y.P. revised the manuscript. J.-C.K. contributed to the interpretation of the results. K.W.L. was responsible for the concept and design of the study, and supervised the work.

CONFLICT OF INTEREST

The authors have no potential conflicts of interest to disclose.

Fig. 1.The effect of TRPV1 deficiency on apoptosis in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−and 3xTg-AD+/+/TRPV1−/− mice. , (A) Representative photographs of TUNEL staining in the CA3, CA1, and DG regions. Scale bars = 2.5 μm. (B-D) Quantification of TUNEL staining fluorescence intensity in the (B) CA3, (C) CA1, and (D) DG regions. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 in compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; WT, wild type; AD, Alzheimer’s disease; CA, cornu ammonis; DG, dentate gyrus.
Fig. 2.The effect of TRPV1 deficiency on the expression level of BDNF and p-TrkB protein in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing expression levels of BDNF, p-TrkB, TrkB, and β-actin proteins. (B-D) Western blot densitometry results for (B) BDNF/β-actin, (C) p-TrkB/TrkB, and (D) p-TrkB/β-actin protein expression-level analysis. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; BDNF, brain-derived neurotrophic factor; TrkB, tyrosine receptor kinase B; WT, wild type; AD, Alzheimer’s disease.
Fig. 3.The effect of TRPV1 deficiency on p-Akt, p-ERK, and p-CREB protein expression level in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing p-Akt, Akt, p-ERK, ERK, and β-actin expression levels. (B and C) Western blot densitometry results for (B) p-Akt/Akt and (C) p-ERK/ERK. (D) Representative Western blot bands showing the expression level of p-CREB and CREB. (E and F) Western blot densitometry results for (E) p-CREB/CREB and (F) p-CREB/β-actin. The results are presented as mean ± SEM, #P < 0.05 compared with the WT, *P < 0.05 compared with the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; ERK, extracellular signal-regulated kinase; CREB, cAMP response element binding protein; WT, wild type; AD, Alzheimer’s disease.
Fig. 4.The effect of TRPV1 deficiency on the expression of apoptosis-related proteins in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing expression levels of Bcl-2, Bax, cleaved caspase-3, and cleaved PARP. (B-E) Western blot densitometry results for (B) Bcl-2/GAPDH, (C) Bax/GAPDH, (D) c-Caspase-3/GAPDH, and (E) cleaved PARP/GAPDH. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; WT, wild type; AD, Alzheimer’s disease; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X; PARP, poly (ADP-ribose) polymerase; c-Caspase-3, cleaved caspase-3; c-PARP, cleaved PARP.
Fig. 5.Molecular pathway of neuroprotection by TRPV1 deficiency in 3xTg-AD mice. Ca2+ is overloaded in 3xTg-AD+/+/TRPV1+/+ and as a result, apoptosis occurs (left). In 3xTg-AD+/+/TRPV1−/− mice, the Ca2+ level is normalized and apoptosis is prevented by activation of the BDNF/CREB pathway (right). TRPV1, transient receptor potential vanilloid 1; AD, Alzheimer’s disease; BDNF, brain-derived neurotrophic factor; CREB, cAMP response element binding protein; NMDA, N-methyl-D-aspartate; TrkB, tyrosine receptor kinase B; ERK, extracellular signal-regulated kinase; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X; PARP, poly (ADP-ribose) polymerase. Created with BioRender.com.

Fig 1.

Figure 1.The effect of TRPV1 deficiency on apoptosis in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−and 3xTg-AD+/+/TRPV1−/− mice. , (A) Representative photographs of TUNEL staining in the CA3, CA1, and DG regions. Scale bars = 2.5 μm. (B-D) Quantification of TUNEL staining fluorescence intensity in the (B) CA3, (C) CA1, and (D) DG regions. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 in compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; WT, wild type; AD, Alzheimer’s disease; CA, cornu ammonis; DG, dentate gyrus.
Molecules and Cells 2023; 46: 319-328https://doi.org/10.14348/molcells.2023.2156

Fig 2.

Figure 2.The effect of TRPV1 deficiency on the expression level of BDNF and p-TrkB protein in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing expression levels of BDNF, p-TrkB, TrkB, and β-actin proteins. (B-D) Western blot densitometry results for (B) BDNF/β-actin, (C) p-TrkB/TrkB, and (D) p-TrkB/β-actin protein expression-level analysis. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; BDNF, brain-derived neurotrophic factor; TrkB, tyrosine receptor kinase B; WT, wild type; AD, Alzheimer’s disease.
Molecules and Cells 2023; 46: 319-328https://doi.org/10.14348/molcells.2023.2156

Fig 3.

Figure 3.The effect of TRPV1 deficiency on p-Akt, p-ERK, and p-CREB protein expression level in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing p-Akt, Akt, p-ERK, ERK, and β-actin expression levels. (B and C) Western blot densitometry results for (B) p-Akt/Akt and (C) p-ERK/ERK. (D) Representative Western blot bands showing the expression level of p-CREB and CREB. (E and F) Western blot densitometry results for (E) p-CREB/CREB and (F) p-CREB/β-actin. The results are presented as mean ± SEM, #P < 0.05 compared with the WT, *P < 0.05 compared with the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; ERK, extracellular signal-regulated kinase; CREB, cAMP response element binding protein; WT, wild type; AD, Alzheimer’s disease.
Molecules and Cells 2023; 46: 319-328https://doi.org/10.14348/molcells.2023.2156

Fig 4.

Figure 4.The effect of TRPV1 deficiency on the expression of apoptosis-related proteins in the hippocampus of WT, 3xTg-AD+/+/TRPV1+/+, 3xTg-AD+/+/TRPV1+/−, and 3xTg-AD+/+/TRPV1−/− mice. (A) Representative Western blot bands showing expression levels of Bcl-2, Bax, cleaved caspase-3, and cleaved PARP. (B-E) Western blot densitometry results for (B) Bcl-2/GAPDH, (C) Bax/GAPDH, (D) c-Caspase-3/GAPDH, and (E) cleaved PARP/GAPDH. The results are presented as mean ± SEM, #P < 0.05 compared to the WT, *P < 0.05 compared to the 3xTg-AD+/+, one-way ANOVA followed by Fisher’s LSD (least significant difference); n = 5 per group. TRPV1, transient receptor potential vanilloid 1; WT, wild type; AD, Alzheimer’s disease; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X; PARP, poly (ADP-ribose) polymerase; c-Caspase-3, cleaved caspase-3; c-PARP, cleaved PARP.
Molecules and Cells 2023; 46: 319-328https://doi.org/10.14348/molcells.2023.2156

Fig 5.

Figure 5.Molecular pathway of neuroprotection by TRPV1 deficiency in 3xTg-AD mice. Ca2+ is overloaded in 3xTg-AD+/+/TRPV1+/+ and as a result, apoptosis occurs (left). In 3xTg-AD+/+/TRPV1−/− mice, the Ca2+ level is normalized and apoptosis is prevented by activation of the BDNF/CREB pathway (right). TRPV1, transient receptor potential vanilloid 1; AD, Alzheimer’s disease; BDNF, brain-derived neurotrophic factor; CREB, cAMP response element binding protein; NMDA, N-methyl-D-aspartate; TrkB, tyrosine receptor kinase B; ERK, extracellular signal-regulated kinase; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X; PARP, poly (ADP-ribose) polymerase. Created with BioRender.com.
Molecules and Cells 2023; 46: 319-328https://doi.org/10.14348/molcells.2023.2156

References

  1. Antonsson B., Conti F., Ciavatta A., Montessuit S., Lewis S., Martinou I., Bernasconi L., Bernard A., Mermod J.J., and Mazzei G., et al. (1997). Inhibition of Bax channel-forming activity by Bcl-2. Science 277, 370-372.
    Pubmed CrossRef
  2. Asadi M., Taghizadeh S., Kaviani E., Vakili O., Taheri-Anganeh M., Tahamtan M., and Savardashtaki A. (2022). Caspase‐3: structure, function, and biotechnological aspects. Biotechnol. Appl. Biochem. 69, 1633-1645.
    Pubmed CrossRef
  3. Baker K.D., Edwards T.M., and Rickard N.S. (2013). The role of intracellular calcium stores in synaptic plasticity and memory consolidation. Neurosci. Biobehav. Rev. 37, 1211-1239.
    Pubmed CrossRef
  4. Berridge M.J. (2011). Calcium signalling and Alzheimer's disease. Neurochem. Res. 36, 1149-1156.
    Pubmed CrossRef
  5. Bloom G.S. (2014). Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 71, 505-508.
    Pubmed CrossRef
  6. Bojarski L., Herms J., and Kuznicki J. (2008). Calcium dysregulation in Alzheimer's disease. Neurochem. Int. 52, 621-633.
    Pubmed CrossRef
  7. Calissano P., Matrone C., and Amadoro G. (2009). Apoptosis and in vitro Alzheimer's disease neuronal models. Commun. Integr. Biol. 2, 163-169.
    Pubmed KoreaMed CrossRef
  8. Calvo-Rodriguez M., Hou S.S., Snyder A.C., Kharitonova E.K., Russ A.N., Das S., Fan Z., Muzikansky A., Garcia-Alloza M., and Serrano-Pozo A., et al. (2020). Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer's disease. Nat. Commun. 11, 2146.
    Pubmed KoreaMed CrossRef
  9. Cascella R. and Cecchi C. (2021). Calcium dyshomeostasis in Alzheimer's disease pathogenesis. Int. J. Mol. Sci. 22, 4914.
    Pubmed KoreaMed CrossRef
  10. Corona C., Masciopinto F., Silvestri E., Viscovo A.D., Lattanzio R., Sorda R.L., Ciavardelli D., Goglia F., Piantelli M., and Canzoniero L.M., et al. (2010). Dietary zinc supplementation of 3xTg-AD mice increases BDNF levels and prevents cognitive deficits as well as mitochondrial dysfunction. Cell Death Dis. 1, e91.
    Pubmed KoreaMed CrossRef
  11. Cregan S.P., MacLaurin J.G., Craig C.G., Robertson G.S., Nicholson D.W., Park D.S., and Slack R.S. (1999). Bax-dependent caspase-3 activation is a key determinant in p53-induced apoptosis in neurons. J. Neurosci. 19, 7860-7869.
    Pubmed KoreaMed CrossRef
  12. D'Amelio M., Sheng M., and Cecconi F. (2012). Caspase-3 in the central nervous system: beyond apoptosis. Trends Neurosci. 35, 700-709.
    Pubmed CrossRef
  13. DeTure M.A. and Dickson D.W. (2019). The neuropathological diagnosis of Alzheimer's disease. Mol. Neurodegener. 14, 32.
    Pubmed KoreaMed CrossRef
  14. Du Y., Fu M., Huang Z., Tian X., Li J., Pang Y., Song W., Tian Wang Y., and Dong Z. (2020). TRPV1 activation alleviates cognitive and synaptic plasticity impairments through inhibiting AMPAR endocytosis in APP23/PS45 mouse model of Alzheimer's disease. Aging Cell 19, e13113.
    Pubmed KoreaMed CrossRef
  15. Eimer W.A. and Vassar R. (2013). Neuron loss in the 5XFAD mouse model of Alzheimer's disease correlates with intraneuronal Aβ42 accumulation and Caspase-3 activation. Mol. Neurodegener. 8, 2.
    Pubmed KoreaMed CrossRef
  16. Elena-Real C.A., Díaz-Quintana A., González-Arzola K., Velázquez-Campoy A., Orzáez M., López-Rivas A., Gil-Caballero S., De, la Rosa M.Á., and Díaz-Moreno I. (2018). Cytochrome c speeds up caspase cascade activation by blocking 14-3-3ε-dependent Apaf-1 inhibition. Cell Death Dis. 9, 365.
    Pubmed KoreaMed CrossRef
  17. Ferrer I., Marín C., Rey M.J., Ribalta T., Goutan E., Blanco R., Tolosa E., and Martí E. (1999). BDNF and full-length and truncated TrkB expression in Alzheimer disease. Implications in therapeutic strategies. J. Neuropathol. Exp. Neurol. 58, 729-739.
    Pubmed CrossRef
  18. Finkbeiner S., Tavazoie S.F., Maloratsky A., Jacobs K.M., Harris K.M., and Greenberg M.E. (1997). CREB: a major mediator of neuronal neurotrophin responses. Neuron 19, 1031-1047.
    Pubmed CrossRef
  19. Fletcher J.I., Meusburger S., Hawkins C.J., Riglar D.T., Lee E.F., Fairlie W.D., Huang D.C., and Adams J.M. (2008). Apoptosis is triggered when prosurvival Bcl-2 proteins cannot restrain Bax. Proc. Natl. Acad. Sci. U. S. A. 105, 18081-18087.
    Pubmed KoreaMed CrossRef
  20. Folch J., Busquets O., Ettcheto M., Sánchez-López E., Castro-Torres R.D., Verdaguer E., Garcia M.L., Olloquequi J., Casadesús G., and Beas-Zarate C., et al. (2018). Memantine for the treatment of dementia: a review on its current and future applications. J. Alzheimers Dis. 62, 1223-1240.
    Pubmed KoreaMed CrossRef
  21. Fumagalli F., Racagni G., and Riva M. (2006). The expanding role of BDNF: a therapeutic target for Alzheimer's disease? Pharmacogenomics J. 6, 8-15.
    Pubmed CrossRef
  22. Gorman A.M., Ceccatelli S., and Orrenius S. (2000). Role of mitochondria in neuronal apoptosis. Dev. Neurosci. 22, 348-358.
    Pubmed CrossRef
  23. Green K.N. and LaFerla F.M. (2008). Linking calcium to Abeta and Alzheimer's disease. Neuron 59, 190-194.
    Pubmed CrossRef
  24. Hippius H. and Neundörfer G. (2003). The discovery of Alzheimer's disease. Dialogues Clin. Neurosci. 5, 101-108.
    Pubmed KoreaMed CrossRef
  25. Ichimiya Y., Emson P.C., Mountjoy C.Q., Lawson D.E.M., and Heizmann C.W. (1988). Loss of calbindin-28K immunoreactive neurones from the cortex in Alzheimer-type dementia. Brain Res. 475, 156-159.
    Pubmed CrossRef
  26. Impey S., Obrietan K., and Storm D.R. (1999). Making new connections: role of ERK/MAP kinase signaling in neuronal plasticity. Neuron 23, 11-14.
    Pubmed CrossRef
  27. Jadiya P., Kolmetzky D.W., Tomar D., Di Meco A., Lombardi A.A., Lambert J.P., Luongo T.S., Ludtmann M.H., Pratico D., and Elrod J.W. (2019). Impaired mitochondrial calcium efflux contributes to disease progression in models of Alzheimer's disease. Nat. Commun. 10, 3885.
    Pubmed KoreaMed CrossRef
  28. Jantas D., Szymanska M., Budziszewska B., and Lason W. (2009). An involvement of BDNF and PI3-K/Akt in the anti-apoptotic effect of memantine on staurosporine-evoked cell death in primary cortical neurons. Apoptosis 14, 900-912.
    Pubmed CrossRef
  29. Kim J., Lee S., Kim J., Ham S., Park J.H.Y., Han S., Jung Y.K., Shim I., Han J.S., and Lee K.W., et al. (2020). Ca2+-permeable TRPV1 pain receptor knockout rescues memory deficits and reduces amyloid-β and tau in a mouse model of Alzheimer's disease. Hum. Mol. Genet. 29, 228-237.
    Pubmed CrossRef
  30. Ku B., Liang C., Jung J.U., and Oh B.H. (2011). Evidence that inhibition of BAX activation by BCL-2 involves its tight and preferential interaction with the BH3 domain of BAX. Cell Res. 21, 627-641.
    Pubmed KoreaMed CrossRef
  31. LeBlanc A.C. (2005). The role of apoptotic pathways in Alzheimer's disease neurodegeneration and cell death. Curr. Alzheimer Res. 2, 389-402.
    Pubmed CrossRef
  32. Lee J., Saloman J.L., Weiland G., Auh Q.S., Chung M.K., and Ro J.Y. (2012). Functional interactions between NMDA receptors and TRPV1 in trigeminal sensory neurons mediate mechanical hyperalgesia in the rat masseter muscle. Pain 153, 1514-1524.
    Pubmed KoreaMed CrossRef
  33. Lu J., Zhou W., Dou F., Wang C., and Yu Z. (2021). TRPV1 sustains microglial metabolic reprogramming in Alzheimer's disease. EMBO Rep. 22, e52013.
    Pubmed KoreaMed CrossRef
  34. Meller R., Minami M., Cameron J.A., Impey S., Chen D., Lan J.Q., Henshall D.C., and Simon R.P. (2005). CREB-mediated Bcl-2 protein expression after ischemic preconditioning. J. Cereb. Blood Flow Metab. 25, 234-246.
    Pubmed CrossRef
  35. Minichiello L., Calella A.M., Medina D.L., Bonhoeffer T., Klein R., and Korte M. (2002). Mechanism of TrkB-mediated hippocampal long-term potentiation. Neuron 36, 121-137.
    Pubmed CrossRef
  36. Miranda M., Morici J.F., Zanoni M.B., and Bekinschtein P. (2019). Brain-derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain. Front. Cell. Neurosci. 13, 363.
    Pubmed KoreaMed CrossRef
  37. Park E.M. and Cho S. (2006). Enhanced ERK dependent CREB activation reduces apoptosis in staurosporine-treated human neuroblastoma SK-N-BE (2) C cells. Neurosci. Lett. 402, 190-194.
    Pubmed CrossRef
  38. Porter A.G. and Jänicke R.U. (1999). Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 6, 99-104.
    Pubmed CrossRef
  39. Rai S.N., Dilnashin H., Birla H., Singh S.S., Zahra W., Rathore A.S., Singh B.K., and Singh S.P. (2019). The role of PI3K/Akt and ERK in neurodegenerative disorders. Neurotox. Res. 35, 775-795.
    Pubmed CrossRef
  40. Robinson D.M. and Keating G.M. (2006). Memantine: a review of its use in Alzheimer's disease. Drugs 66, 1515-1534.
    Pubmed CrossRef
  41. Sairanen T., Szepesi R., Karjalainen-Lindsberg M.L., Saksi J., Paetau A., and Lindsberg P.J. (2009). Neuronal caspase-3 and PARP-1 correlate differentially with apoptosis and necrosis in ischemic human stroke. Acta Neuropathol. 118, 541-552.
    Pubmed CrossRef
  42. Santos A.R., Comprido D., and Duarte C.B. (2010). Regulation of local translation at the synapse by BDNF. Prog. Neurobiol. 92, 505-516.
    Pubmed CrossRef
  43. Saura C.A. and Valero J. (2011). The role of CREB signaling in Alzheimer's disease and other cognitive disorders. Rev. Neurosci. 22, 153-169.
    Pubmed CrossRef
  44. Scheff S.W. and Price D.A. (2006). Alzheimer's disease-related alterations in synaptic density: neocortex and hippocampus. J. Alzheimers Dis. 9((3 Suppl)), 101-115.
    Pubmed CrossRef
  45. Shacka J.J. and Roth K.A. (2005). Regulation of neuronal cell death and neurodegeneration by members of the Bcl-2 family: therapeutic implications. Curr. Drug Targets CNS Neurol. Disord. 4, 25-39.
    Pubmed CrossRef
  46. Takahashi K., Nakagawasai O., Nemoto W., Kadota S., Isono J., Odaira T., Sakuma W., Arai Y., Tadano T., and Tan-No K. (2018). Memantine ameliorates depressive-like behaviors by regulating hippocampal cell proliferation and neuroprotection in olfactory bulbectomized mice. Neuropharmacology 137, 141-155.
    Pubmed CrossRef
  47. Tanqueiro S.R., Ramalho R.M., Rodrigues T.M., Lopes L.V., Sebastião A.M., and Diógenes M.J. (2018). Inhibition of NMDA receptors prevents the loss of BDNF function induced by amyloid β. Front. Pharmacol. 9, 237.
    Pubmed KoreaMed CrossRef
  48. Vanhoutte P. and Bading H. (2003). Opposing roles of synaptic and extrasynaptic NMDA receptors in neuronal calcium signalling and BDNF gene regulation. Curr. Opin. Neurobiol. 13, 366-371.
    Pubmed CrossRef
  49. Wang X. (2001). The expanding role of mitochondria in apoptosis. Genes Dev. 15, 2922-2933.
    Pubmed
  50. Wang X., Chen J., Wang H., Yu H., Wang C., You J., Wang P., Feng C., Xu G., and Wu X., et al. (2017a). Memantine can reduce ethanol-induced caspase-3 activity and apoptosis in H4 cells by decreasing intracellular calcium. J. Mol. Neurosci. 62, 402-411.
    Pubmed CrossRef
  51. Wang X., Yu H., You J., Wang C., Feng C., Liu Z., Li Y., Wei R., Xu S., and Zhao R., et al. (2018). Memantine can improve chronic ethanol exposure-induced spatial memory impairment in male C57BL/6 mice by reducing hippocampal apoptosis. Toxicology 406-407, 21-32.
    Pubmed CrossRef
  52. Wang Y., Shi Y., and Wei H. (2017b). Calcium dysregulation in Alzheimer's disease: a target for new drug development. J. Alzheimers Dis. Parkinsonism 7, 374.
    Pubmed KoreaMed CrossRef
  53. Yin X.M., Oltvai Z.N., and Korsmeyer S.J. (1994). BH1 and BH2 domains of Bcl-2 are required for inhibition of apoptosis and heterodimerization with Bax. Nature 369, 321-323.
    Pubmed CrossRef
  54. Yuan J., Murrell G.A., Trickett A., and Wang M.X. (2003). Involvement of cytochrome c release and caspase-3 activation in the oxidative stress-induced apoptosis in human tendon fibroblasts. Biochim. Biophys. Acta 1641, 35-41.
    Pubmed CrossRef
  55. Zarneshan S.N., Fakhri S., and Khan H. (2022). Targeting Akt/CREB/BDNF signaling pathway by ginsenosides in neurodegenerative diseases: a mechanistic approach. Pharmacol. Res. 177, 106099.
    Pubmed CrossRef
  56. Zhai K., Liskova A., Kubatka P., and Büsselberg D. (2020). Calcium entry through TRPV1: a potential target for the regulation of proliferation and apoptosis in cancerous and healthy cells. Int. J. Mol. Sci. 21, 4177.
    Pubmed KoreaMed CrossRef
  57. Zheng Z., Sabirzhanov B., and Keifer J. (2010). Oligomeric amyloid-β inhibits the proteolytic conversion of brain-derived neurotrophic factor (BDNF), AMPA receptor trafficking, and classical conditioning. J. Biol. Chem. 285, 34708-34717.
    Pubmed KoreaMed CrossRef
  58. Zhu G., Li J., He L., Wang X., and Hong X. (2015). MPTP‐induced changes in hippocampal synaptic plasticity and memory are prevented by memantine through the BDNF‐TrkB pathway. Br. J. Pharmacol. 172, 2354-2368.
    Pubmed KoreaMed CrossRef
Mol. Cells
Nov 30, 2023 Vol.46 No.11, pp. 655~725
COVER PICTURE
Kim et al. (pp. 710-724) demonstrated that a pathogen-derived Ralstonia pseudosolanacearum type III effector RipL delays flowering time and enhances susceptibility to bacterial infection in Arabidopsis thaliana. Shown is the RipL-expressing Arabidopsis plant, which displays general dampening of the transcriptional program during pathogen infection, grown in long-day conditions.

Share this article on

  • line

Related articles in Mol. Cells

Molecules and Cells

eISSN 0219-1032
qr-code Download