Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2022

Open Access 01.12.2022 | Research

Neuroprotective effects of ex vivo-expanded regulatory T cells on trimethyltin-induced neurodegeneration in mice

verfasst von: Seon-Young Park, HyeJin Yang, Minsook Ye, Xiao Liu, Insop Shim, Young-Tae Chang, Hyunsu Bae

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2022

Abstract

Background

Trimethyltin (TMT) is a potent neurotoxicant that leads to hippocampal neurodegeneration. Regulatory T cells (Tregs) play an important role in maintaining the immune balance in the central nervous system (CNS), but their activities are impaired in neurodegenerative diseases. In this study, we aimed to determine whether adoptive transfer of Tregs, as a living drug, ameliorates hippocampal neurodegeneration in TMT-intoxicated mice.

Methods

CD4+CD25+ Tregs were expanded in vitro and adoptively transferred to TMT-treated mice. First, we explored the effects of Tregs on behavioral deficits using the Morris water maze and elevated plus maze tests. Biomarkers related to memory formation, such as cAMP response element-binding protein (CREB), protein kinase C (PKC), neuronal nuclear protein (NeuN), nerve growth factor (NGF), and ionized calcium binding adaptor molecule 1 (Iba1) in the hippocampus were examined by immunohistochemistry after killing the mouse. To investigate the neuroinflammatory responses, the polarization status of microglia was examined in vivo and in vitro using real-time reverse transcription polymerase chain reaction (rtPCR) and Enzyme-linked immunosorbent assay (ELISA). Additionally, the inhibitory effects of Tregs on TMT-induced microglial activation were examined using time-lapse live imaging in vitro with an activation-specific fluorescence probe, CDr20.

Results

Adoptive transfer of Tregs improved spatial learning and memory functions and reduced anxiety in TMT-intoxicated mice. Additionally, adoptive transfer of Tregs reduced neuronal loss and recovered the expression of neurogenesis enhancing molecules in the hippocampi of TMT-intoxicated mice. In particular, Tregs inhibited microglial activation and pro-inflammatory cytokine release in the hippocampi of TMT-intoxicated mice. The inhibitory effects of TMT were also confirmed via in vitro live time-lapse imaging in a Treg/microglia co-culture system.

Conclusions

These data suggest that adoptive transfer of Tregs ameliorates disease progression in TMT-induced neurodegeneration by promoting neurogenesis and modulating microglial activation and polarization.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
Amyloid-β
AD
Alzheimer’s disease
ALS
Amyotrophic lateral sclerosis
Arg1
Arginase 1
BDNF
Brain-derived neurotrophic factor
BM
Bone marrow
BSA
Bovine serum albumin
bvPLA2
Bee venom phospholipase A2
CNS
Central nervous system
CREB
CAMP response element-binding protein
DAB
Diaminobenzidine
DCs
Dendritic cells
DG
Dentate gyrus
DMSO
Dimethyl sulfoxide
EPM
Elevated plus maze
GM-CSF
Granulocyte-macrophage colony-stimulating factor
Iba1
Ionized calcium binding adaptor molecule 1
IL
Interleukin
Mrc1
Mannose receptor 1
MWM
Morris water maze
NOS2
Nitric oxide synthase 2
NeuN
Neuronal nuclear protein
NGF
Nerve growth factor
PBS
Phosphate-buffered saline
PD
Parkinson’s disease
PKC
Protein kinase C
TGFβ
Transforming growth factor β
TMT
Trimethyltin
Tregs
Regulatory T cells
TNFα
Tumor necrosis factor α

Background

The hippocampus, an area under the medial temporal lobe of the mammalian brain, plays a pivotal role in the neurobiology of learning and memory. It is one of the first regions damaged in Alzheimer’s disease (AD) [1, 2]. Hippocampal neurodegeneration accounts for the cognitive impairments observed in neurodegenerative disorders, such as AD [3]. There is a clinical association between hippocampal neurogenesis and cognition and microglia are important effectors of hippocampal neurogenesis. Activated pro-inflammatory microglia have a negative effect on hippocampal neurogenesis and cognitive processes [4].
Trimethyltin (TMT) is an organotin compound that is considered a potent neurotoxicant and causes behavioral alterations as well as learning and memory impairment in mammals [5, 6]. Cognitive impairment, including memory loss and learning impairment, developed in experimental animals exposed to TMT, indicating severe hippocampal damage [7]. It was reported that the levels of activated microglia and pro-inflammatory factors, such as TNFα, IL-1β, and NO were elevated in the hippocampus prior to neuronal death by TMT treatment in rodents. Consistently, previous studies have indicated that microglial activation by TMT exacerbates neuronal death in vivo and in vitro [810].
Regulatory T cells (Tregs) act as immune suppressors, playing a role in self-tolerance and immune homeostasis. Immune balance in the central nervous system (CNS) is tightly controlled by Tregs. Previous studies suggested that the Tregs suppress the microglial inflammation by promoting polarization toward anti-inflammatory M2 rather than pro-inflammatory M1 phenotype [1113]. However, the suppressive activity of Tregs is dysregulated in neurodegenerative diseases, leading to neuroinflammation in these diseases. For these reasons, Tregs are emerging as an attractive therapeutic strategy against neurodegenerative diseases [14, 15]. Therefore, adoptive cell therapy using Tregs has attracted attention as an individualized medicine for inflammatory diseases [16]. Treg cell therapy has been attempted in mouse models of neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and Parkinson’s disease (PD) to evaluate its neuroprotective effects [17, 18]. In our previous study, we adoptively transferred Tregs into 3×Tg-AD mice containing three mutations associated with familial Alzheimer’s disease (APP Swedish, MAPT P301L, and PSEN1 M146V) and demonstrated the inhibitory effects of Tregs on the accumulation of amyloid-beta (Aβ) and activation of microglia in the hippocampus [19]. Since TMT-treated animal models are used to study hippocampus-specific neurodegeneration that accompanies microglial activation, similar to that seen in AD, we aimed to confirm that Treg cell therapy is also effective in TMT-induced hippocampal neurodegeneration. Based on the effects of Treg therapy in other neurodegenerative diseases, it is expected that TMT-induced neuronal loss and behavior disorders will prevent through microglial activation by Treg transfer.
There is some evidence indicating that antigen-specific Tregs may be more efficient, so the generation and expansion of antigen-specific Tregs are important in Treg cell therapy [20]. To generate antigen-specific Tregs, we presented fibrillar Aβ to bone marrow-derived dendritic cells (Aβ-DCs) and performed ex vivo Treg expansion in the presence of Aβ-DCs. In addition, to increase the efficiency of Treg expansion, we treated cells with bee venom phospholipase A2 (bvPLA2), a Treg expansion inducer [21]. We previously demonstrated that bvPLA2 induced the Treg population by suppressing apoptosis [22]. Moreover, we reported that administration of bvPLA2 had neuroprotective effects on AD and PD mouse model [23, 24].
In the present study, we attempted to expand Aβ-specific Tregs and examine the effects of the adoptive transfer of these Tregs on behavioral deficits, memory formation, and neuronal loss in TMT-induced neurodegenerative mice. Furthermore, we sought to determine whether the effects of Tregs are associated with microglial activation, which induces pro-inflammatory responses. Our findings would be helpful in developing a new treatment strategy for neurodegenerative diseases.

Materials and methods

Animals

Seven-week-old male C57BL/6 mice were purchased from Taconic Farms, Inc. (Samtako Bio Korea, Kyunggi, Korea) and Deahan Biolink (Chungbuk, Korea). The mice were maintained under a 12-h light/dark cycle and temperature-controlled conditions, with food and water ad libitum. All experiments were performed in accordance with the approved animal protocols and guidelines established by Kyung Hee University (KHUAP(SE)-18-073).

Regulatory T cell preparation

To prepare fibrillary Aβ, 5 mM Aβ1–42 peptide (Genscript, NJ, USA) in dimethyl sulfoxide (DMSO) was diluted with 10 mM HCl to a final concentration of 100 µM Aβ and incubated overnight (O/N) at 37 °C. Bone marrow (BM)-leukocytes from femurs and tibiae of mice were resuspended in a medium containing 20 ng/mL granulocyte-macrophage colony-stimulating factor (GM-CSF; R&D Systems, Minneapolis, MN, USA) [25]. After 7 days, BM-leukocytes were washed with magnetic-activated cell sorting buffer (Miltenyi Biotec Inc., CA, USA) and dendritic cells (DCs) were isolated using CD11c+ MicroBeads (Miltenyi). The DCs were resuspended at a density of 2 × 105/mL and seeded in 96-well U-bottom plates. For antigen presentation, DCs were treated with 0.5 µM fibrillated Aβ for 24 h. CD4+ T cells from splenocytes were isolated using CD4 (L3T4) MicroBeads (Miltenyi), resuspended at a density of 2 × 106/mL, and added to the DC culture at a ratio of 10: 1 (CD4+ T cells: DCs) with 0.4 µg/mL bvPLA2 (Sigma-Aldrich, MO, USA). Four days after CD4 T cell–DC co-culture, CD4+CD25+ T cells (Tregs) were isolated using MACS, according to the manufacturer’s protocol (CD4+CD25+ Regulatory T Cell Isolation Kit; Miltenyi). CD4+CD25+ regulatory T cells were stimulated using the Treg Expansion Kit (Miltenyi) for 2 weeks. To confirm the purity of isolated cells and the change in phenotype, cells were stained with fluorescently labeled antibodies and analyzed using flow cytometry. The following antibodies were used (1:1000): PE-CD11c (eBioscience, San Diego, CA, USA) for DC purity, PE-CD4 (BD Pharmingen, CA, USA) for CD4 T cell purity, and PE-CD127 (eBioscience), PE-Cy7-CD4 (Invitrogen, CA, USA), APC-CD62L (Invitrogen), and APC-Cy7-CD25 (BD Pharmingen) for Treg phenotype. Samples were washed with the BD FACS stain buffer (BD Bioscience, CA, USA) and stained for 30 min at 4 °C in the dark. After staining, the cells were washed 2 times with the stain buffer. The data were acquired using a BD FACSlyric™ flow cytometer (BD Bioscience) and analyzed using BD FACSuite software (BD Bioscience).

BV2 microglia and Treg co-culture

BV2 microglia were incubated at 37 °C with 95% humidity and 5% CO2 for all experiments. To examine the effects of Tregs on microglial polarization, 1 × 106 BV2 microglia in Dulbecco’s modified Eagle’s medium (DMEM; Welgene Daegu, Korea) 500 µL were seeded into 12-well plates. After 2–3 h, Tregs were co-cultured with BV2 cells (BV2:Treg = 10:1) and the cells were immediately stimulated with 3 µM TMT for 24 h according to previous study [26]. The cell culture supernatants were collected for ELISA, and the remaining adherent cells were harvested for mRNA extraction.

Animal experiments

For TMT (Sigma-Aldrich, Steinheim, Germany) treatment, the mice were intraperitoneally (i.p.) administered TMT (2.6 mg/kg) and randomly divided into five groups of 18 to 25 mice, except for the control group (n = 21) that did not receive TMT. After 7 days, Treg cells (4 × 104, 2 × 105, or 1 × 106) were intravenously injected (i.v.) into the tail vein of TMT-treated mice. Aricept (3 mg/kg; Eisai Co. Ltd, Tokyo, Japan) was orally administered once daily for 2 weeks from day 7.

Behavior tests

Ten days after Treg injection, spatial learning and memory were examined in mice using the Morris water maze (MWM) test with minor modifications [27]. The water maze was a circular pool with a 90-cm diameter and was filled with opaque water containing 1 kg of powdered skim milk (maintained at 22 ± 2 °C). During training, a 6-cm hidden platform was fixed 1 cm below the water surface. The pool was surrounded by different extra-maze cues. The maximal trial duration was 60 s, with 30 s on the platform at the end of the first trial. Each animal was trained for one of the different starting positions and swimming paths once per day for 4 days. All mice were subjected to three trials per day at intervals of 15 min for 4 consecutive days. For the probe trial, the platform was removed from the pool, and the mice were allowed to swim freely for 60 s to search for the previous location of the platform. Escape latency, time spent in the platform quadrant, and the number of platform crossings were recorded for each mouse.
The elevated plus maze (EPM) test was performed after the first MWM training to measure the anxiety levels in mice. The EPM equipment was a cross-shaped maze that was elevated to a height of 50 cm above the floor. It consisted of two opposite open arms and two closed arms. Mice were positioned on the central platform and allowed to explore the maze for 3 min.
Data were collected using a video camera connected to a video recorder and a tracking device (S-MART, Pan-Lab).

Immunohistochemistry

After the behavioral test, mice were anesthetized by pentobarbital (50 mg/kg, i.p.) and transcardially perfused with formalin and PBS. The brain was transferred into a 30% sucrose solution, and frozen-sectioned on a sliding microtome into 30-μm-thick coronal sections. The brain sections (3–5 sections/mice) were washed with phosphate-buffered saline (PBS) and incubated for 10 min with 3% hydrogen peroxide (Sigma-Aldrich) to quench endogenous peroxidase activity. Nonspecific binding was reduced by blocking the sections with 1.5% bovine serum albumin (BSA; Millipore, MA, USA) in PBS for 1 h. The sections were incubated with antibodies (1:500) for mouse CREB (Cell Signaling Technology, MA, USA), Iba1 (WAKO, Osaka, Japan), PKC (Abcam, MA, USA), NeuN (Abcam), or NGF (Invitrogen) for 24 h at RT. Brain sections were washed with PBS, incubated with a biotinylated secondary antibody (Vectastain ABC kit; Vector Laboratories, CA, USA) for 2 h, and processed using an avidin–biotin peroxidase complex kit (Vectastain ABC kit; Vector Laboratories) for 1 h. Each marker was visualized by incubation with 0.05% diaminobenzidine–HCl (DAB; Vector Laboratories). The labeled sections were mounted and analyzed under a bright-field microscope (Nikon) and the intensities were quantified using the ImageJ software (US National Institutes of Health; available at http://​rsb.​info.​nih.​gov/​ij/​) as previously describe [28, 29]. Data were analyzed under the same conditions by two observers for each experiment in blinded conditions to avoid the bias. Images were calibrated into an array of 512 × 512 pixels corresponding to a tissue area. Each pixel resolution had 256 Gy levels, and the intensity of immunoreactivity was evaluated based on the ROD, which was obtained after transformation of the mean gray level using the following formula: ROD = log10 (256/mean gray level).

RT-PCR assay

Mice were transcardially perfused with PBS after anesthetization. RNA was isolated from the brain and BV2 cells using the easy-BLUE RNA extraction kit (iNtRON Biotechnology, Seoul, Korea), and cDNA was synthesized using Cyclescript reverse transcriptase (Bioneer, Seoul, Korea). The samples were prepared for real-time PCR using the SensiFAST SYBR no-Rox kit (Bioline, OH, USA). Real-time quantitative PCR was performed using CFX Connect (Bio-Rad, WA, USA) and the data were analyzed using CFX Maestro Software (Bio-Rad). The amplification conditions were 95 °C for 30 s, followed by 50 cycles at 95 °C for 10 s and 55 °C for 30 s. The expression levels of each target mRNAs, 2−dCt values, were normalized to those of mouse β-actin, a housekeeping gene used as an endogenous control [30]. Then the relative mRNA expression values were calculated as a fold change in which the mean value of the control group considered 1. The base sequences of the primers are shown in Table 1.
Table 1
The base sequence of primers for rtPCR
Primer name
Forward primer sequence (5′-3′)
Reverse primer sequence (5′-3′)
β-actin
GTG CTA TGT TGC TCT AGA CTT CG
ATG CCA CAG GAT TCC ATA CC
NOS2
CAG CTG GGC TGT ACA AAC CTT
CAT TGG AAG TGA AGC GTT TCG
IL-1β
AAG CCT CGT GCT GTC GGA CC
TGA GGC CCA AGG CCA CAG G
IL-6
TTC CAT CCA GTT GCC TTC TTG
GGG AGT GGT ATC CTC TGT GAA GTC
TNFα
GGC AGG TTC TGT CCC TTT CAC
TTC TGT GCT CAT GGT GTC TTT TCT
TGFβ
GAG GTC ACC CGC GTG CTA
TGT GTG AGA TGT CTT TGG TTT TCT C
BDNF
GGA ATT CGA GTG ATG ACC ATC CTT TTC CTT AC
CGG ATC CCT ATC TTC CCC TTT TAA TGG TCA GTG
Mrc1
TTC GGT GGA CTG TGG ACG AGC
ATA AGC CAC CTG CCA CTC CGG
Ym1
TGG AGG ATG GAA GTT TGG AC
GAG TAG CAG CCT TGG AAT GT
Arg1
CTC CAA GCC AAA GTC CTT AGA G
AGG AGC TGT CAT TAG GGA CAT C

ELISA

After anesthetization, mice were transcardially perfused with PBS. Total protein was isolated from the brain using RIPA buffer (Biosesang, Seoul, Korea) with protease and phosphatase inhibitors (Thermo Fisher Scientific, CA, USA). Levels of pro-inflammatory cytokines were quantified using TNF-α, IL-1β, and IL-6 DuoSet ELISA (R&D Systems) and normalized to the levels of BSA. The cytokines secreted by BV2 cells were measured using BV2 cell culture media and TNFα and TGFβ DuoSet ELISA (R&D Systems). The optical density was measured at 450 nm using a microplate reader (Versamax Microplate Reader, USA). All fold changes were expressed relative to those in the control group.

Live cell imaging

CDr20 is a microglia-specific biofluorescence probe with high performance for visualizing live microglia both in vitro and in vivo [31]. For time-lapse imaging, 5 × 104 BV2 microglia in 1 mL were seeded into 4-well chambers and cultured for 2–3 h before live-cell imaging was performed. Approximately 5 × 104 mouse Tregs were seeded onto each chamber containing microglia. Microglia were continuously observed from pre-activation to post-activation with TMT (3 µM) treatment in the presence of 0.5 mM CDr20 (1 µM) every 3 min for a total of 30 min under the red fluorescent channel (excitation at 570 nm and emission at 600 nm). The change in the region of intensity (ROI) of each cell was measured for 30 min. All observations were performed using a DeltaVision imaging system (GE, Boston, MA, USA). To assess the intensity of fluorescence live-cell imaging, the SoftWorX software (v.6.1.3, GE) was used. CDr20 was kindly provided by Dr. YT Chang (Pohang University of Science and Technology, Pohang, Korea).

Statistical analysis

All data were analyzed using GraphPad Prism 5.01 (GraphPad Software Inc., CA, USA). The data are presented as the mean and standard error of the mean (SEM) where indicated. All statistical significance of each variable was evaluated by one-way analysis of variance (ANOVA), followed by Tukey multiple comparison test for multiple comparisons except the intensity of PKC and time-lapse live imaging: *p < 0.05, **p < 0.01, ***p < 0.001. The intensity of PKC and time-lapse live imaging were analyzed using two-tailed Student’s t-test and two-way ANOVA followed by Bonferroni post-tests, respectively. All experiments were performed in a blinded manner and repeated independently under identical conditions.

Results

Isolation and ex vivo expansion of Tregs

To prepare Aβ-specific Tregs, CD4+CD25+ Tregs were isolated after 4 days of co-culture of CD11c+ DCs and CD4+ T cells. After ex vivo expansion, Tregs were injected into TMT-intoxicated mice (Fig. 1A). The isolated cells were analyzed using flow cytometry. FACS analysis demonstrated that CD11c+ DCs and CD4+ T cells were more than 90% enriched for these subsets. The purity of CD4+CD25+ Tregs was greater than 97% (Fig. 1B). During expansion, the changes in phenotypes were analyzed for 2 weeks. Various subsets depend on the phenotype of Tregs. For example, CD62L is highly expressed in the naïve phenotype, and CD127, the IL-7 receptor α chain, is considered a memory marker [32, 33]. Some studies have suggested the importance of the Treg phenotype, especially the CD62L+ naïve phenotype, for clinical manipulation. The CD62L+ Treg subset is an optimal suppressor that expands far more easily in culture [34]. Therefore, the phenotypes of Tregs were divided into CD62LhiCD127low naïve, CD62LlowCD127low effector, and CD62LlowCD127hi memory phenotypes (Fig. 1C). At week 2, the transferred Tregs were mainly effector phenotypes (96.69%).

Regulatory T cells prevent cognitive impairments in TMT-intoxicated mice

To measure the effect of regulatory T cell transfer on spatial learning and memory ability in TMT-induced mice, the MWM test was conducted. TMT–intoxicated mice exhibited longer latency times than control mice on days 2, 3, and 4. However, the latency times of Treg groups (2 × 105, 1 × 106) were decreased compared to those of the TMT group (Fig. 2A). The time spent in the target quadrant was also significantly increased in the 2 × 105 and 1 × 106 Treg groups compared with those in the TMT group (Fig. 2B). The elevated plus maze test was used to measure anxiety in TMT-intoxicated mice. The number of entries into the closed or open arms was recorded. The number of open arm entries in the TMT group was significantly decreased compared with that of the control group, whereas that of the 1 × 106 Treg group was significantly increased compared with that of the TMT group (Fig. 2C, D). These data indicate that adoptive transfer of Tregs reverts cognitive deficits in TMT-intoxicated mice.

Regulatory T cells improve synaptic strengthening and memory function in TMT- intoxicated mice

CREB immunostaining was performed on sections of the mouse brain (Fig. 3A). Remarkable losses of CREB-positive cells in both CA1 and CA3 were observed in the TMT group compared to the control group. However, adoptive transfer of 1 × 106 Tregs resulted in a significantly greater number of CREB-positive cells (Fig. 3B). The expression of PKC, an upstream protein kinase that activates CREB, was measured in the CA1 and CA3 regions (Fig. 3C). TMT intoxication reduced the intensity of PKC in both CA1 and CA3 compared to that in the control (Fig. 3D). The intensity of PKC in CA1 was not altered significantly following adoptive transfer of Tregs; however, adoptive transfer of Tregs increased the intensity of PKC in CA3, especially at doses of 4 × 104 and 1 × 106 cells/mouse.

Tregs inhibit neuronal loss in TMT-intoxicated mice

To evaluate the effect of Tregs on TMT-induced neuronal loss, mouse brain sections were stained for NeuN, a neuronal biomarker (Fig. 4A). The number of NeuN-positive cells was significantly reduced in TMT-treated mice compared with that in the control in both CA1 and CA3 (Fig. 4B). All groups adoptively transferred Tregs showed more NeuN-positive cells than the TMT group. Additionally, the expression of NGF, a neurotrophic factor, was assessed (Fig. 4C). Similar to that for NeuN, the intensity of NGF staining that was significantly decreased in CA3 upon TMT intoxication was recovered upon Treg transfer (Fig. 4D). These results suggest that adoptive transfer of Tregs inhibits TMT-induced neuronal loss.

Tregs reduce pro-inflammatory microglial activation in TMT-intoxicated mice

To assess microglial activation, mouse brain sections were stained for Iba1, an activated microglial marker (Fig. 5A). The number of Iba1-positive cells in the TMT group was significantly increased compared with that in the control group, whereas that in all Treg-transfer groups was significantly decreased compared with that in the TMT group in CA3. Similar tendencies were observed in CA1, but the difference was not significant (Fig. 5B). The protein levels of pro-inflammatory cytokines, such as TNFα, IL-1β, and IL-6, were measured using ELISA (Fig. 5C). The expression of these cytokines was significantly increased in TMT-intoxicated mice compared with that in the control. When Tregs were adoptively transferred, the levels of these cytokines showed decreasing tendencies compared with those in the TMT group. Next, mRNA levels in the brain were measured using real-time PCR (Fig. 5D). The mRNA levels of pro-inflammatory microglial markers TNFα, IL-1β, IL-6, and NOS2 in the TMT group were significantly increased compared with those in the control group. In all Treg-transfer groups, the mRNA levels of NOS2 and IL-6 were significantly decreased compared with those in the TMT group. The mRNA levels of TNFα and IL-1β showed similar trends. However, the mRNA levels of TGFβ, Mrc1, Arg1, and BDNF showed tendencies opposite to those shown by the pro-inflammatory microglial markers. Collectively, the results indicate that adoptive transfer of Tregs inhibits TMT-induced microglial activation, especially that of pro-inflammatory M1 microglia.

Tregs induce transition of M1–M2 phenotypes in TMT-treated microglia

To confirm the direct impact of Tregs on microglial activation, we utilized murine BV2 microglia under TMT-induced inflammatory conditions in vitro. For live imaging, live BV2 cells were imaged using CDr20, a high-performance fluorogenic chemical probe for activated microglia [31], during which, the intensity of the labeled cells was recorded for 30 min (Fig. 6A). As BV2 cells were activated upon TMT treatment, the intensity gradually increased for 30 min. However, co-culture with Tregs showed inhibitory effects on the TMT-induced activation of microglia. To further confirm the effect of Tregs on microglial polarization, the release of the pro-inflammatory cytokine TNFα and anti-inflammatory cytokine TGFβ was measured using ELISA (Fig. 6B). TNFα levels were higher in TMT-treated BV2 cells than in normal BV2 cells. Furthermore, in co-culture with Tregs, the level of TNFα significantly decreased, whereas that of TGFβ increased compared with that in TMT-treated BV2 cells. The mRNA expression of the M1 microglial markers TNFα, NOS2, and IL-1β and M2 microglial markers TGFβ, Mrc1, and Ym1 were also measured (Fig. 6C). As expected, the levels of the pro-inflammatory M1 microglial markers were increased in TMT-treated BV2 cells, and co-culture with Tregs substantially decreased the mRNA expression of these markers. Conversely, the expression of the M2 microglial markers increased upon co-culture with Tregs. Taken together, these data suggest that Tregs modulate microglial polarization upon TMT treatment.

Discussion

In this study, we investigated the effects of Tregs on TMT-induced hippocampal neurodegeneration. We found that Tregs not only improved cognitive function, but also reduced anxiety in TMT-intoxicated mice. Moreover, Tregs inhibited neuronal loss, and the neuroprotective effects of Tregs could potentially be attributed to suppression of microglia-mediated neuroinflammation. Compared with Aricept, a drug used for AD, adoptive transfer of Tregs was found to be similarly or more effective. Our study supports the potential of Treg therapy for hippocampal neurodegeneration.
Tregs are considered attractive therapeutic targets for attenuating inflammation. Tregs play roles in inhibiting pro-inflammatory cytokines and inducing neurotrophic factors and apoptosis of pro-inflammatory microglia, ultimately promoting neuroprotection [35]. In a previous study from our laboratory, adoptive transfer of Tregs was attempted in 3×Tg-AD mice, upon which a clear delay in the onset of AD neuropathology was observed. In addition, the neuroprotective effect of Tregs was demonstrated, including reduction in Aβ deposition and microglial activation in the hippocampus. However, Treg adoptive transfer has never been attempted in the TMT-induced neurodegenerative model, although it has been considered as a model of AD-like disease in rats [36]. Therefore, in this study, we transplanted Tregs into TMT-intoxicated mice to alleviate TMT-induced hippocampal neurodegeneration.
For clinical application, various strategies were proposed to improve the effects of Treg therapy. The most common method is developing antigen-specific Tregs instead polyclonal Tregs which may lead to off-target suppression. Antigen presentation could enhance the therapeutic utility of T cell transfer to induce target sites [37, 38]. Aβ is also present in the normal brain; however, it is misfolded and deposited in the hippocampus in several pathological conditions such as AD. Therefore, it is regarded as one of the characteristics of these diseases [39]. Moreover, since Aβ accumulation was detected in TMT-intoxicated mice, we chose Aβ as an antigen for presentation to adoptively transfer Tregs [40]. Additionally, we treated bvPLA2 during antigen presentation to expand the Treg population. It was reported that bvPLA2 induces Treg population both in vivo and in vitro and significantly suppresses apoptosis in Tregs [21, 22]. The combination of antigen presentation via DCs and bvPLA2 treatment for the generation and expansion of Aβ-specific Tregs is an important attempt of this study. The effects and mechanism of action of Aβ presentation and bvPLA2 treatment on the efficacy of Tregs remain unclear. This will be investigated in a future study.
For decades, TMT-induced neurodegenerative models, especially rats and mice, have been used as good research tools. In the rat model, TMT administration induces a progressive cell death accompanied by microglial activation in CA1 and CA3 like AD [36, 41]. Notably, TMT injection into mice can also cause dentate gyrus (DG) granular cell apoptosis. Many studies on TMT-induced mouse model focused on neuropathology in DG [4246]. DG is the site where adult hippocampus neurogenesis occurs and most information of DG is sent to CA3 to CA1 according to the tri-synaptic pathway in the hippocampus [47]. Some studies reported neuronal self-repair following TMT-induced neuronal loss in DG. These data indicated that neuronal regeneration occurs in DG approximately 7–10 days after TMT intoxication [4851]. Therefore, TMT has been considered as a toxicant that acts exclusively on DG in mice. However, there are several reports that TMT induced neurodegeneration was not restricted in DG. KR Reuhl and colleagues reported extensive degenerative and necrotic changes in CA3 after TMT intoxication [52]. In another report, IB4, a microglial marker, and Fas, an apoptotic molecule, were increased in CA1 after TMT intoxication [53]. Nevertheless, degenerative change in neurons of CA was unremarkable than DG, TMT has been considered as a neurotoxicant that selectively affects in DG [54]. However, studies on TMT-induced cognitive dysfunction have emerged [55, 56]. In recent studies, it was reported that memory impairment accompanied by neurodegeneration in CA induced by TMT intoxication in mice. According to these studies, neuronal loss was observed in CA1 even after 7 days of TMT intoxication, unlike in DG [5760]. This implied that the timing of TMT-induced neurotoxicity on DG and CA is different, probably it occurs later in CA than in DG. It is a very interesting topic that these events could affect the tri-synaptic circuit related disorders and will be revealed in further studies. In the present study, we focus on TMT-induced cognitive disorders and molecular change in CA1 and CA3.
It is reported that TMT-intoxicated animals developed cognitive impairment and hyperactivity [7]. The results of MWM and EPM test showed that high dose of Tregs improved these behavior changes. But Aricept, a positive control, showed no significant effect. It is probably because Aricept is not an effective drug for long-term treatment. In fact, it has been reported that Aricept treatment for 16 weeks did not improve cognitive function in APPswe/PS1dE9 mice [61]. These results imply that Treg possess a sufficient potential as a more effective treatment option than Aricept.
CREB is a key molecule in synaptic strengthening, memory formation, and neurogenesis. It controls the transcription of genes involved in neuronal growth and survival and the lack of CREB gene results in neurodegeneration. Indeed, disruption of the CREB phosphorylation mechanism results in a reduction in CREB activation following memory impairment in AD [6264]. Likewise, TMT-induced memory impairment was observed upon inhibition of CREB activation and was alleviated by regulation of the CREB-signaling pathway in the hippocampus [65]. Since CREB plays a critical role in short- to long-term memory, drugs targeting CREB itself have been proposed for memory modification [66]. One of the molecules present the upstream of CREB and regulating it is PKC. Therefore, activation of PKC leads to CREB phosphorylation [67, 68]. In addition, PKC itself performs neurogenesis-related functions, including cell differentiation and proliferation and immune-related processes. In a previous study, Bacopa monnieri (L.) Wettst. extract prevented TMT-induced hippocampal damage via PKC [69]. NGF is also a neurotrophic factor that enhances neurogenesis [70]. We showed that TMT intoxication induced neuronal cell death, represented by the expression of NeuN, in both CA1 and CA3, whereas there was no difference in DG (data not shown). These results are consistent with the possibility of different TMT toxicity timing on DG and CA mentioned above. And Tregs, especially at high dosage, increased the expression of CREB, PKC, and NGF as well as NeuN. This suggests that adoptively transferred Tregs not only prevent neuronal loss, but also induce neurogenesis in the hippocampus.
Microglia are phagocytic macrophages that comprise 10–15% of the total cells in the CNS. Since they can be either beneficial or harmful depending on their activation status, their polarization is considered a potential therapeutic target in neurodegenerative diseases such as AD. Classically activated “M1” microglia contribute to inflammation by secreting free radicals, NOS2, and pro-inflammatory cytokines such as IL-1, IL-6, and TNFα. Neuroinflammation amplifies microglial activation and further worsens the disease. In contrast, alternatively activated “M2” microglia promote tissue repair by releasing neuroprotective cytokines such as IL-10, TGFβ, and IGF1. Therefore, microglial polarization is considered an attractive therapeutic strategy against cognitive disorders [71, 72]. Indeed, there have been many studies that treat neurodegenerative diseases by shifting microglial phenotypes. Some studies reported behavior recovery following enhancing M2 microglia in not only AD, but also traumatic brain injury and spinal cord injury [7375]. It is well known that pro-inflammatory microglial activation and cytokine secretion are associated with TMT intoxication [7679]. Therefore, we confirmed the activation and polarization of microglia in vivo and in vitro. As expected, microglial activation and pro-inflammatory marker expression were increased upon TMT intoxication. However, Tregs inhibited the activation of M1 but enhanced M2 microglia in vivo. Additionally, we observed microglial activation over time using time-lapse live imaging in vitro. TMT treatment activated microglia for 30 min, but co-culture with Tregs suppressed this activation. ELISA and RT-PCR data showed that this inhibition by Tregs targeted M1 microglia. Based on these in vivo and in vitro results, Tregs could effectively inhibit microglial activations and covert microglial phenotype from M1 to M2. These changes in microglia phenotype lead to neurogenesis and ultimately improve cognitive impairment. It is in line with those of previous studies showing that Tregs modulate microglia and alleviate neurodegenerative disorders [11, 72, 80, 81].

Conclusions

Taken together, as shown in Fig. 7, we report that adoptive transfer of Tregs reduces behavioral deficits in TMT-intoxicated mice. Tregs inhibit neuronal loss and increase the expression of factors that enhance neurogenesis. In particular, Tregs dramatically modulated the activation and polarization of microglia upon TMT intoxication both in vivo and in vitro. These findings support the potential of Treg therapy in hippocampal neurodegeneration.

Acknowledgements

Not applicable.

Declarations

All animal experiments were performed in accordance with the approved animal protocols and guidelines established by Kyung Hee University (KHUAP(SE)-18-073).
Not applicable.

Competing interests

The authors declare that there were no commercial or financial relationships that could be construed as a potential conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Akinyemi RO, Allan LM, Oakley A, Kalaria RN. Hippocampal neurodegenerative pathology in post-stroke dementia compared to other dementias and aging controls. Front Neurosci. 2017;11:717.PubMedPubMedCentralCrossRef Akinyemi RO, Allan LM, Oakley A, Kalaria RN. Hippocampal neurodegenerative pathology in post-stroke dementia compared to other dementias and aging controls. Front Neurosci. 2017;11:717.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Spanswick SC, Lehmann H, Sutherland RJ. A novel animal model of hippocampal cognitive deficits, slow neurodegeneration, and neuroregeneration. J Biomed Biotechnol. 2011;2011:527201.PubMedPubMedCentralCrossRef Spanswick SC, Lehmann H, Sutherland RJ. A novel animal model of hippocampal cognitive deficits, slow neurodegeneration, and neuroregeneration. J Biomed Biotechnol. 2011;2011:527201.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Ryan SM, Nolan YM. Neuroinflammation negatively affects adult hippocampal neurogenesis and cognition: can exercise compensate? Neurosci Biobehav Rev. 2016;61:121–31.PubMedCrossRef Ryan SM, Nolan YM. Neuroinflammation negatively affects adult hippocampal neurogenesis and cognition: can exercise compensate? Neurosci Biobehav Rev. 2016;61:121–31.PubMedCrossRef
5.
Zurück zum Zitat Ishida N, Akaike M, Tsutsumi S, Kanai H, Masui A, Sadamatsu M, et al. Trimethyltin syndrome as a hippocampal degeneration model: temporal changes and neurochemical features of seizure susceptibility and learning impairment. Neuroscience. 1997;81(4):1183–91.PubMedCrossRef Ishida N, Akaike M, Tsutsumi S, Kanai H, Masui A, Sadamatsu M, et al. Trimethyltin syndrome as a hippocampal degeneration model: temporal changes and neurochemical features of seizure susceptibility and learning impairment. Neuroscience. 1997;81(4):1183–91.PubMedCrossRef
6.
Zurück zum Zitat Cristofol RM, Gasso S, Vilchez D, Pertusa M, Rodriguez-Farre E, Sanfeliu C. Neurotoxic effects of trimethyltin and triethyltin on human fetal neuron and astrocyte cultures: a comparative study with rat neuronal cultures and human cell lines. Toxicol Lett. 2004;152(1):35–46.PubMedCrossRef Cristofol RM, Gasso S, Vilchez D, Pertusa M, Rodriguez-Farre E, Sanfeliu C. Neurotoxic effects of trimethyltin and triethyltin on human fetal neuron and astrocyte cultures: a comparative study with rat neuronal cultures and human cell lines. Toxicol Lett. 2004;152(1):35–46.PubMedCrossRef
7.
Zurück zum Zitat Geloso MC, Corvino V, Michetti F. Trimethyltin-induced hippocampal degeneration as a tool to investigate neurodegenerative processes. Neurochem Int. 2011;58(7):729–38.PubMedCrossRef Geloso MC, Corvino V, Michetti F. Trimethyltin-induced hippocampal degeneration as a tool to investigate neurodegenerative processes. Neurochem Int. 2011;58(7):729–38.PubMedCrossRef
8.
Zurück zum Zitat Eskes C, Juillerat-Jeanneret L, Leuba G, Honegger P, Monnet-Tschudi F. Involvement of microglia-neuron interactions in the tumor necrosis factor-alpha release, microglial activation, and neurodegeneration induced by trimethyltin. J Neurosci Res. 2003;71(4):583–90.PubMedCrossRef Eskes C, Juillerat-Jeanneret L, Leuba G, Honegger P, Monnet-Tschudi F. Involvement of microglia-neuron interactions in the tumor necrosis factor-alpha release, microglial activation, and neurodegeneration induced by trimethyltin. J Neurosci Res. 2003;71(4):583–90.PubMedCrossRef
9.
Zurück zum Zitat McPherson CA, Kraft AD, Harry GJ. Injury-induced neurogenesis: consideration of resident microglia as supportive of neural progenitor cells. Neurotox Res. 2011;19(2):341–52.PubMedCrossRef McPherson CA, Kraft AD, Harry GJ. Injury-induced neurogenesis: consideration of resident microglia as supportive of neural progenitor cells. Neurotox Res. 2011;19(2):341–52.PubMedCrossRef
10.
Zurück zum Zitat Maier WE, Brown HW, Tilson HA, Luster MI, Harry GJ. Trimethyltin increases interleukin (IL)-1 alpha, IL-6 and tumor necrosis factor alpha mRNA levels in rat hippocampus. J Neuroimmunol. 1995;59(1–2):65–75.PubMedCrossRef Maier WE, Brown HW, Tilson HA, Luster MI, Harry GJ. Trimethyltin increases interleukin (IL)-1 alpha, IL-6 and tumor necrosis factor alpha mRNA levels in rat hippocampus. J Neuroimmunol. 1995;59(1–2):65–75.PubMedCrossRef
11.
Zurück zum Zitat Zhou K, Zhong Q, Wang YC, Xiong XY, Meng ZY, Zhao T, et al. Regulatory T cells ameliorate intracerebral hemorrhage-induced inflammatory injury by modulating microglia/macrophage polarization through the IL-10/GSK3beta/PTEN axis. J Cereb Blood Flow Metabol. 2017;37(3):967–79.CrossRef Zhou K, Zhong Q, Wang YC, Xiong XY, Meng ZY, Zhao T, et al. Regulatory T cells ameliorate intracerebral hemorrhage-induced inflammatory injury by modulating microglia/macrophage polarization through the IL-10/GSK3beta/PTEN axis. J Cereb Blood Flow Metabol. 2017;37(3):967–79.CrossRef
12.
Zurück zum Zitat Wang S, Zhang H, Xu Y. Crosstalk between microglia and T cells contributes to brain damage and recovery after ischemic stroke. Neurol Res. 2016;38(6):495–503.PubMedCrossRef Wang S, Zhang H, Xu Y. Crosstalk between microglia and T cells contributes to brain damage and recovery after ischemic stroke. Neurol Res. 2016;38(6):495–503.PubMedCrossRef
13.
Zurück zum Zitat Zhao W, Beers DR, Appel SH. Immune-mediated mechanisms in the pathoprogression of amyotrophic lateral sclerosis. J Neuroimmune Pharmacol. 2013;8(4):888–99.PubMedPubMedCentralCrossRef Zhao W, Beers DR, Appel SH. Immune-mediated mechanisms in the pathoprogression of amyotrophic lateral sclerosis. J Neuroimmune Pharmacol. 2013;8(4):888–99.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat He F, Balling R. The role of regulatory T cells in neurodegenerative diseases. Wiley Interdiscip Rev Syst Biol Med. 2013;5(2):153–80.PubMedCrossRef He F, Balling R. The role of regulatory T cells in neurodegenerative diseases. Wiley Interdiscip Rev Syst Biol Med. 2013;5(2):153–80.PubMedCrossRef
15.
Zurück zum Zitat Xie L, Choudhury GR, Winters A, Yang SH, Jin K. Cerebral regulatory T cells restrain microglia/macrophage-mediated inflammatory responses via IL-10. Eur J Immunol. 2015;45(1):180–91.PubMedCrossRef Xie L, Choudhury GR, Winters A, Yang SH, Jin K. Cerebral regulatory T cells restrain microglia/macrophage-mediated inflammatory responses via IL-10. Eur J Immunol. 2015;45(1):180–91.PubMedCrossRef
16.
Zurück zum Zitat Jiang S, Lechler RI. CD4+CD25+ regulatory T-cell therapy for allergy, autoimmune disease and transplant rejection. Inflamm Allergy Drug Targets. 2006;5(4):239–42.PubMedCrossRef Jiang S, Lechler RI. CD4+CD25+ regulatory T-cell therapy for allergy, autoimmune disease and transplant rejection. Inflamm Allergy Drug Targets. 2006;5(4):239–42.PubMedCrossRef
17.
Zurück zum Zitat Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson’s disease. J Leukoc Biol. 2007;82(5):1083–94.PubMedCrossRef Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson’s disease. J Leukoc Biol. 2007;82(5):1083–94.PubMedCrossRef
18.
Zurück zum Zitat Beers DR, Henkel JS, Zhao W, Wang J, Huang A, Wen S, et al. Endogenous regulatory T lymphocytes ameliorate amyotrophic lateral sclerosis in mice and correlate with disease progression in patients with amyotrophic lateral sclerosis. Brain J Neurol. 2011;134(Pt 5):1293–314.CrossRef Beers DR, Henkel JS, Zhao W, Wang J, Huang A, Wen S, et al. Endogenous regulatory T lymphocytes ameliorate amyotrophic lateral sclerosis in mice and correlate with disease progression in patients with amyotrophic lateral sclerosis. Brain J Neurol. 2011;134(Pt 5):1293–314.CrossRef
19.
Zurück zum Zitat Baek H, Ye M, Kang GH, Lee C, Lee G, Choi DB, et al. Neuroprotective effects of CD4+CD25+Foxp3+ regulatory T cells in a 3xTg-AD Alzheimer’s disease model. Oncotarget. 2016;7(43):69347–57.PubMedPubMedCentralCrossRef Baek H, Ye M, Kang GH, Lee C, Lee G, Choi DB, et al. Neuroprotective effects of CD4+CD25+Foxp3+ regulatory T cells in a 3xTg-AD Alzheimer’s disease model. Oncotarget. 2016;7(43):69347–57.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Selck C, Dominguez-Villar M. Antigen-specific regulatory T cell therapy in autoimmune diseases and transplantation. Front Immunol. 2021;12:1748.CrossRef Selck C, Dominguez-Villar M. Antigen-specific regulatory T cell therapy in autoimmune diseases and transplantation. Front Immunol. 2021;12:1748.CrossRef
21.
Zurück zum Zitat Kim H, Keum DJ, Kwak J, Chung HS, Bae H. Bee venom phospholipase A2 protects against acetaminophen-induced acute liver injury by modulating regulatory T cells and IL-10 in mice. PLoS ONE. 2014;9(12):e114726.PubMedPubMedCentralCrossRef Kim H, Keum DJ, Kwak J, Chung HS, Bae H. Bee venom phospholipase A2 protects against acetaminophen-induced acute liver injury by modulating regulatory T cells and IL-10 in mice. PLoS ONE. 2014;9(12):e114726.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Baek H, Park SY, Ku SJ, Ryu K, Kim Y, Bae H, et al. Bee venom phospholipase A2 induces regulatory T cell populations by suppressing apoptotic signaling pathway. Toxins. 2020;12(3):198.PubMedCentralCrossRef Baek H, Park SY, Ku SJ, Ryu K, Kim Y, Bae H, et al. Bee venom phospholipase A2 induces regulatory T cell populations by suppressing apoptotic signaling pathway. Toxins. 2020;12(3):198.PubMedCentralCrossRef
23.
Zurück zum Zitat Chung ES, Lee G, Lee C, Ye M, Chung HS, Kim H, et al. Bee venom phospholipase A2, a novel Foxp3+ regulatory T cell inducer, protects dopaminergic neurons by modulating neuroinflammatory responses in a mouse model of Parkinson’s disease. J Immunol. 2015;195(10):4853–60.PubMedCrossRef Chung ES, Lee G, Lee C, Ye M, Chung HS, Kim H, et al. Bee venom phospholipase A2, a novel Foxp3+ regulatory T cell inducer, protects dopaminergic neurons by modulating neuroinflammatory responses in a mouse model of Parkinson’s disease. J Immunol. 2015;195(10):4853–60.PubMedCrossRef
24.
Zurück zum Zitat Ye M, Chung HS, Lee C, Yoon MS, Yu AR, Kim JS, et al. Neuroprotective effects of bee venom phospholipase A2 in the 3xTg AD mouse model of Alzheimer’s disease. J Neuroinflamm. 2016;13:10.CrossRef Ye M, Chung HS, Lee C, Yoon MS, Yu AR, Kim JS, et al. Neuroprotective effects of bee venom phospholipase A2 in the 3xTg AD mouse model of Alzheimer’s disease. J Neuroinflamm. 2016;13:10.CrossRef
25.
Zurück zum Zitat Lutz MB, Kukutsch N, Ogilvie AL, Rossner S, Koch F, Romani N, et al. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J Immunol Methods. 1999;223(1):77–92.PubMedCrossRef Lutz MB, Kukutsch N, Ogilvie AL, Rossner S, Koch F, Romani N, et al. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J Immunol Methods. 1999;223(1):77–92.PubMedCrossRef
26.
Zurück zum Zitat Kim DJ, Kim YS. Trimethyltin-induced microglial activation via NADPH oxidase and MAPKs pathway in BV-2 microglial cells. Mediators Inflamm. 2015;2015:729509.PubMedPubMedCentral Kim DJ, Kim YS. Trimethyltin-induced microglial activation via NADPH oxidase and MAPKs pathway in BV-2 microglial cells. Mediators Inflamm. 2015;2015:729509.PubMedPubMedCentral
27.
Zurück zum Zitat Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984;11(1):47–60.PubMedCrossRef Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984;11(1):47–60.PubMedCrossRef
28.
Zurück zum Zitat Jung HY, Kwon HJ, Kim W, Nam SM, Kim JW, Hahn KR, et al. Phosphoglycerate mutase 1 promotes cell proliferation and neuroblast differentiation in the dentate gyrus by facilitating the phosphorylation of cAMP response element-binding protein. Neurochem Res. 2019;44(2):323–32.PubMedCrossRef Jung HY, Kwon HJ, Kim W, Nam SM, Kim JW, Hahn KR, et al. Phosphoglycerate mutase 1 promotes cell proliferation and neuroblast differentiation in the dentate gyrus by facilitating the phosphorylation of cAMP response element-binding protein. Neurochem Res. 2019;44(2):323–32.PubMedCrossRef
29.
Zurück zum Zitat Yoo DY, Lee KY, Park JH, Jung HY, Kim JW, Yoon YS, et al. Glucose metabolism and neurogenesis in the gerbil hippocampus after transient forebrain ischemia. Neural Regen Res. 2016;11(8):1254–9.PubMedPubMedCentralCrossRef Yoo DY, Lee KY, Park JH, Jung HY, Kim JW, Yoon YS, et al. Glucose metabolism and neurogenesis in the gerbil hippocampus after transient forebrain ischemia. Neural Regen Res. 2016;11(8):1254–9.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Ruijter JM, Pfaffl MW, Zhao S, Spiess AN, Boggy G, Blom J, et al. Evaluation of qPCR curve analysis methods for reliable biomarker discovery: bias, resolution, precision, and implications. Methods. 2013;59(1):32–46.PubMedCrossRef Ruijter JM, Pfaffl MW, Zhao S, Spiess AN, Boggy G, Blom J, et al. Evaluation of qPCR curve analysis methods for reliable biomarker discovery: bias, resolution, precision, and implications. Methods. 2013;59(1):32–46.PubMedCrossRef
31.
Zurück zum Zitat Kim B, Fukuda M, Lee JY, Su D, Sanu S, Silvin A, et al. Visualizing microglia with a fluorescence turn-on Ugt1a7c substrate. Angew Chem. 2019;58(24):7972–6.CrossRef Kim B, Fukuda M, Lee JY, Su D, Sanu S, Silvin A, et al. Visualizing microglia with a fluorescence turn-on Ugt1a7c substrate. Angew Chem. 2019;58(24):7972–6.CrossRef
32.
Zurück zum Zitat Anderson BE, McNiff J, Yan J, Doyle H, Mamula M, Shlomchik MJ, et al. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest. 2003;112(1):101–8.PubMedPubMedCentralCrossRef Anderson BE, McNiff J, Yan J, Doyle H, Mamula M, Shlomchik MJ, et al. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest. 2003;112(1):101–8.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Huster KM, Busch V, Schiemann M, Linkemann K, Kerksiek KM, Wagner H, et al. Selective expression of IL-7 receptor on memory T cells identifies early CD40L-dependent generation of distinct CD8+ memory T cell subsets. Proc Natl Acad Sci USA. 2004;101(15):5610–5.PubMedPubMedCentralCrossRef Huster KM, Busch V, Schiemann M, Linkemann K, Kerksiek KM, Wagner H, et al. Selective expression of IL-7 receptor on memory T cells identifies early CD40L-dependent generation of distinct CD8+ memory T cell subsets. Proc Natl Acad Sci USA. 2004;101(15):5610–5.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Fu S, Yopp AC, Mao X, Chen D, Zhang N, Chen D, et al. CD4+ CD25+ CD62+ T-regulatory cell subset has optimal suppressive and proliferative potential. Am J Transplant. 2004;4(1):65–78.PubMedCrossRef Fu S, Yopp AC, Mao X, Chen D, Zhang N, Chen D, et al. CD4+ CD25+ CD62+ T-regulatory cell subset has optimal suppressive and proliferative potential. Am J Transplant. 2004;4(1):65–78.PubMedCrossRef
36.
Zurück zum Zitat Lattanzi W, Corvino V, Di Maria V, Michetti F, Geloso MC. Gene expression profiling as a tool to investigate the molecular machinery activated during hippocampal neurodegeneration induced by trimethyltin (TMT) administration. Int J Mol Sci. 2013;14(8):16817–35.PubMedPubMedCentralCrossRef Lattanzi W, Corvino V, Di Maria V, Michetti F, Geloso MC. Gene expression profiling as a tool to investigate the molecular machinery activated during hippocampal neurodegeneration induced by trimethyltin (TMT) administration. Int J Mol Sci. 2013;14(8):16817–35.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Qiao J, Wang H, Kottke T, Diaz RM, Willmon C, Hudacek A, et al. Loading of oncolytic vesicular stomatitis virus onto antigen-specific T cells enhances the efficacy of adoptive T-cell therapy of tumors. Gene Ther. 2008;15(8):604–16.PubMedPubMedCentralCrossRef Qiao J, Wang H, Kottke T, Diaz RM, Willmon C, Hudacek A, et al. Loading of oncolytic vesicular stomatitis virus onto antigen-specific T cells enhances the efficacy of adoptive T-cell therapy of tumors. Gene Ther. 2008;15(8):604–16.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Bluestone JA. Regulatory T-cell therapy: is it ready for the clinic? Nat Rev Immunol. 2005;5(4):343–9.PubMedCrossRef Bluestone JA. Regulatory T-cell therapy: is it ready for the clinic? Nat Rev Immunol. 2005;5(4):343–9.PubMedCrossRef
39.
Zurück zum Zitat Rajasekhar K, Chakrabarti M, Govindaraju T. Function and toxicity of amyloid beta and recent therapeutic interventions targeting amyloid beta in Alzheimer’s disease. Chem Commun. 2015;51(70):13434–50.CrossRef Rajasekhar K, Chakrabarti M, Govindaraju T. Function and toxicity of amyloid beta and recent therapeutic interventions targeting amyloid beta in Alzheimer’s disease. Chem Commun. 2015;51(70):13434–50.CrossRef
40.
Zurück zum Zitat Park SK, Kang JY, Kim JM, Yoo SK, Han HJ, Chung DH, et al. Fucoidan-rich substances from ecklonia cava improve trimethyltin-induced cognitive dysfunction via down-regulation of amyloid beta production/tau hyperphosphorylation. Mar Drugs. 2019;17(10):591.PubMedCentralCrossRef Park SK, Kang JY, Kim JM, Yoo SK, Han HJ, Chung DH, et al. Fucoidan-rich substances from ecklonia cava improve trimethyltin-induced cognitive dysfunction via down-regulation of amyloid beta production/tau hyperphosphorylation. Mar Drugs. 2019;17(10):591.PubMedCentralCrossRef
41.
Zurück zum Zitat Lee S, Yang M, Kim J, Kang S, Kim J, Kim JC, et al. Trimethyltin-induced hippocampal neurodegeneration: a mechanism-based review. Brain Res Bull. 2016;125:187–99.PubMedCrossRef Lee S, Yang M, Kim J, Kang S, Kim J, Kim JC, et al. Trimethyltin-induced hippocampal neurodegeneration: a mechanism-based review. Brain Res Bull. 2016;125:187–99.PubMedCrossRef
42.
Zurück zum Zitat Chang LW, Tiemeyer TM, Wenger GR, McMillan DE. Neuropathology of mouse hippocampus in acute trimethyltin intoxication. Neurobehav Toxicol Teratol. 1982;4(2):149–56.PubMed Chang LW, Tiemeyer TM, Wenger GR, McMillan DE. Neuropathology of mouse hippocampus in acute trimethyltin intoxication. Neurobehav Toxicol Teratol. 1982;4(2):149–56.PubMed
43.
Zurück zum Zitat Bruccoleri A, Brown H, Harry GJ. Cellular localization and temporal elevation of tumor necrosis factor-alpha, interleukin-1 alpha, and transforming growth factor-beta 1 mRNA in hippocampal injury response induced by trimethyltin. J Neurochem. 1998;71(4):1577–87.PubMedCrossRef Bruccoleri A, Brown H, Harry GJ. Cellular localization and temporal elevation of tumor necrosis factor-alpha, interleukin-1 alpha, and transforming growth factor-beta 1 mRNA in hippocampal injury response induced by trimethyltin. J Neurochem. 1998;71(4):1577–87.PubMedCrossRef
44.
Zurück zum Zitat Fiedorowicz A, Figiel I, Kaminska B, Zaremba M, Wilk S, Oderfeld-Nowak B. Dentate granule neuron apoptosis and glia activation in murine hippocampus induced by trimethyltin exposure. Brain Res. 2001;912(2):116–27.PubMedCrossRef Fiedorowicz A, Figiel I, Kaminska B, Zaremba M, Wilk S, Oderfeld-Nowak B. Dentate granule neuron apoptosis and glia activation in murine hippocampus induced by trimethyltin exposure. Brain Res. 2001;912(2):116–27.PubMedCrossRef
45.
Zurück zum Zitat Reuhl KR, Cranmer JM. Developmental neuropathology of organotin compounds. Neurotoxicology. 1984;5(2):187–204.PubMed Reuhl KR, Cranmer JM. Developmental neuropathology of organotin compounds. Neurotoxicology. 1984;5(2):187–204.PubMed
46.
Zurück zum Zitat Dey PM, Graff RD, Lagunowich LA, Reuhl KR. Selective loss of the 180-kDa form of the neural cell adhesion molecule in hippocampus and cerebellum of the adult mouse following trimethyltin administration. Toxicol Appl Pharmacol. 1994;126(1):69–74.PubMedCrossRef Dey PM, Graff RD, Lagunowich LA, Reuhl KR. Selective loss of the 180-kDa form of the neural cell adhesion molecule in hippocampus and cerebellum of the adult mouse following trimethyltin administration. Toxicol Appl Pharmacol. 1994;126(1):69–74.PubMedCrossRef
47.
Zurück zum Zitat Li Y, Mu Y, Gage FH. Development of neural circuits in the adult hippocampus. Curr Top Dev Biol. 2009;87:149–74.PubMedCrossRef Li Y, Mu Y, Gage FH. Development of neural circuits in the adult hippocampus. Curr Top Dev Biol. 2009;87:149–74.PubMedCrossRef
48.
Zurück zum Zitat Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4(11):1313–7.PubMedCrossRef Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, Nordborg C, Peterson DA, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4(11):1313–7.PubMedCrossRef
49.
Zurück zum Zitat Ogita K, Nishiyama N, Sugiyama C, Higuchi K, Yoneyama M, Yoneda Y. Regeneration of granule neurons after lesioning of hippocampal dentate gyrus: evaluation using adult mice treated with trimethyltin chloride as a model. J Neurosci Res. 2005;82(5):609–21.PubMedCrossRef Ogita K, Nishiyama N, Sugiyama C, Higuchi K, Yoneyama M, Yoneda Y. Regeneration of granule neurons after lesioning of hippocampal dentate gyrus: evaluation using adult mice treated with trimethyltin chloride as a model. J Neurosci Res. 2005;82(5):609–21.PubMedCrossRef
50.
Zurück zum Zitat Yoneyama M, Kawada K, Ogita K. Enhanced neurogenesis in the olfactory bulb in adult mice after injury induced by acute treatment with trimethyltin. J Neurosci Res. 2010;88(6):1242–51.PubMed Yoneyama M, Kawada K, Ogita K. Enhanced neurogenesis in the olfactory bulb in adult mice after injury induced by acute treatment with trimethyltin. J Neurosci Res. 2010;88(6):1242–51.PubMed
51.
Zurück zum Zitat Kikuta M, Shiba T, Yoneyama M, Kawada K, Yamaguchi T, Hinoi E, et al. In vivo and in vitro treatment with edaravone promotes proliferation of neural progenitor cells generated following neuronal loss in the mouse dentate gyrus. J Pharmacol Sci. 2013;121(1):74–83.PubMedCrossRef Kikuta M, Shiba T, Yoneyama M, Kawada K, Yamaguchi T, Hinoi E, et al. In vivo and in vitro treatment with edaravone promotes proliferation of neural progenitor cells generated following neuronal loss in the mouse dentate gyrus. J Pharmacol Sci. 2013;121(1):74–83.PubMedCrossRef
52.
Zurück zum Zitat Reuhl KR, Smallridge EA, Chang LW, Mackenzie BA. Developmental effects of trimethyltin intoxication in the neonatal mouse. I. Light microscopic studies. Neurotoxicology. 1983;4(1):19–28.PubMed Reuhl KR, Smallridge EA, Chang LW, Mackenzie BA. Developmental effects of trimethyltin intoxication in the neonatal mouse. I. Light microscopic studies. Neurotoxicology. 1983;4(1):19–28.PubMed
53.
Zurück zum Zitat Harry GJ, Lefebvre d’Hellencourt C, McPherson CA, Funk JA, Aoyama M, Wine RN. Tumor necrosis factor p55 and p75 receptors are involved in chemical-induced apoptosis of dentate granule neurons. J Neurochem. 2008;106(1):281–98.PubMedCrossRef Harry GJ, Lefebvre d’Hellencourt C, McPherson CA, Funk JA, Aoyama M, Wine RN. Tumor necrosis factor p55 and p75 receptors are involved in chemical-induced apoptosis of dentate granule neurons. J Neurochem. 2008;106(1):281–98.PubMedCrossRef
54.
Zurück zum Zitat Chang LW. Neuropathology of trimethyltin: a proposed pathogenetic mechanism. Fundam Appl Toxicol. 1986;6(2):217–32.PubMedCrossRef Chang LW. Neuropathology of trimethyltin: a proposed pathogenetic mechanism. Fundam Appl Toxicol. 1986;6(2):217–32.PubMedCrossRef
55.
Zurück zum Zitat Kim J, Yang M, Kim SH, Kim JC, Wang H, Shin T, et al. Possible role of the glycogen synthase kinase-3 signaling pathway in trimethyltin-induced hippocampal neurodegeneration in mice. PLoS ONE. 2013;8(8):e70356.PubMedPubMedCentralCrossRef Kim J, Yang M, Kim SH, Kim JC, Wang H, Shin T, et al. Possible role of the glycogen synthase kinase-3 signaling pathway in trimethyltin-induced hippocampal neurodegeneration in mice. PLoS ONE. 2013;8(8):e70356.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Kaur S, Chhabra R, Nehru B. Ginkgo biloba extract attenuates hippocampal neuronal loss and cognitive dysfunction resulting from trimethyltin in mice. Phytomedicine. 2013;20(2):178–86.PubMedCrossRef Kaur S, Chhabra R, Nehru B. Ginkgo biloba extract attenuates hippocampal neuronal loss and cognitive dysfunction resulting from trimethyltin in mice. Phytomedicine. 2013;20(2):178–86.PubMedCrossRef
57.
Zurück zum Zitat Thong-Asa W, Prasartsri S, Klomkleaw N, Thongwan N. The neuroprotective effect of betanin in trimethyltin-induced neurodegeneration in mice. Metab Brain Dis. 2020;35(8):1395–405.PubMedCrossRef Thong-Asa W, Prasartsri S, Klomkleaw N, Thongwan N. The neuroprotective effect of betanin in trimethyltin-induced neurodegeneration in mice. Metab Brain Dis. 2020;35(8):1395–405.PubMedCrossRef
58.
Zurück zum Zitat Kwon OY, Lee SH. Ishige okamurae suppresses trimethyltin-induced neurodegeneration and glutamate-mediated excitotoxicity by regulating MAPKs/Nrf2/HO-1 antioxidant pathways. Antioxidants. 2021;10(3):440.PubMedPubMedCentralCrossRef Kwon OY, Lee SH. Ishige okamurae suppresses trimethyltin-induced neurodegeneration and glutamate-mediated excitotoxicity by regulating MAPKs/Nrf2/HO-1 antioxidant pathways. Antioxidants. 2021;10(3):440.PubMedPubMedCentralCrossRef
59.
60.
Zurück zum Zitat Pham HTN, Phan SV, Tran HN, Phi XT, Le XT, Nguyen KM, et al. Bacopa monnieri (L.) ameliorates cognitive deficits caused in a trimethyltin-induced neurotoxicity model mice. Biol Pharm Bull. 2019;42(8):1384–93.PubMedCrossRef Pham HTN, Phan SV, Tran HN, Phi XT, Le XT, Nguyen KM, et al. Bacopa monnieri (L.) ameliorates cognitive deficits caused in a trimethyltin-induced neurotoxicity model mice. Biol Pharm Bull. 2019;42(8):1384–93.PubMedCrossRef
61.
Zurück zum Zitat Wang DM, Yang YJ, Zhang L, Zhang X, Guan FF, Zhang LF. Naringin enhances CaMKII activity and improves long-term memory in a mouse model of Alzheimer’s disease. Int J Mol Sci. 2013;14(3):5576–86.PubMedPubMedCentralCrossRef Wang DM, Yang YJ, Zhang L, Zhang X, Guan FF, Zhang LF. Naringin enhances CaMKII activity and improves long-term memory in a mouse model of Alzheimer’s disease. Int J Mol Sci. 2013;14(3):5576–86.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Teich AF, Nicholls RE, Puzzo D, Fiorito J, Purgatorio R, Fa M, et al. Synaptic therapy in Alzheimer’s disease: a CREB-centric approach. Neurotherapeutics. 2015;12(1):29–41.PubMedPubMedCentralCrossRef Teich AF, Nicholls RE, Puzzo D, Fiorito J, Purgatorio R, Fa M, et al. Synaptic therapy in Alzheimer’s disease: a CREB-centric approach. Neurotherapeutics. 2015;12(1):29–41.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Merz K, Herold S, Lie DC. CREB in adult neurogenesis–master and partner in the development of adult-born neurons? Eur J Neurosci. 2011;33(6):1078–86.PubMedCrossRef Merz K, Herold S, Lie DC. CREB in adult neurogenesis–master and partner in the development of adult-born neurons? Eur J Neurosci. 2011;33(6):1078–86.PubMedCrossRef
64.
Zurück zum Zitat Lonze BE, Ginty DD. Function and regulation of CREB family transcription factors in the nervous system. Neuron. 2002;35(4):605–23.PubMedCrossRef Lonze BE, Ginty DD. Function and regulation of CREB family transcription factors in the nervous system. Neuron. 2002;35(4):605–23.PubMedCrossRef
65.
Zurück zum Zitat Park HJ, Shim HS, Ahn YH, Kim KS, Park KJ, Choi WK, et al. Tremella fuciformis enhances the neurite outgrowth of PC12 cells and restores trimethyltin-induced impairment of memory in rats via activation of CREB transcription and cholinergic systems. Behav Brain Res. 2012;229(1):82–90.PubMedCrossRef Park HJ, Shim HS, Ahn YH, Kim KS, Park KJ, Choi WK, et al. Tremella fuciformis enhances the neurite outgrowth of PC12 cells and restores trimethyltin-induced impairment of memory in rats via activation of CREB transcription and cholinergic systems. Behav Brain Res. 2012;229(1):82–90.PubMedCrossRef
66.
Zurück zum Zitat Jackson T, Ramaswami M. Prospects of memory-modifying drugs that target the CREB pathway. Curr Opin Drug Discov Dev. 2003;6(5):712–9. Jackson T, Ramaswami M. Prospects of memory-modifying drugs that target the CREB pathway. Curr Opin Drug Discov Dev. 2003;6(5):712–9.
67.
Zurück zum Zitat Wang H, Xu J, Lazarovici P, Quirion R, Zheng W. cAMP response element-binding protein (CREB): a possible signaling molecule link in the pathophysiology of schizophrenia. Front Mol Neurosci. 2018;11:255.PubMedPubMedCentralCrossRef Wang H, Xu J, Lazarovici P, Quirion R, Zheng W. cAMP response element-binding protein (CREB): a possible signaling molecule link in the pathophysiology of schizophrenia. Front Mol Neurosci. 2018;11:255.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Pham HTN, Tran HN, Nguyen PT, Le XT, Nguyen KM, Phan SV, et al. Bacopa monnieri (L.) Wettst. Extract improves memory performance via promotion of neurogenesis in the hippocampal dentate gyrus of adolescent mice. Int J Mol Sci. 2020;21(9):3365.PubMedCentralCrossRef Pham HTN, Tran HN, Nguyen PT, Le XT, Nguyen KM, Phan SV, et al. Bacopa monnieri (L.) Wettst. Extract improves memory performance via promotion of neurogenesis in the hippocampal dentate gyrus of adolescent mice. Int J Mol Sci. 2020;21(9):3365.PubMedCentralCrossRef
71.
Zurück zum Zitat Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann Transl Med. 2015;3(10):136.PubMedPubMedCentral Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann Transl Med. 2015;3(10):136.PubMedPubMedCentral
72.
Zurück zum Zitat Yao K, Zu HB. Microglial polarization: novel therapeutic mechanism against Alzheimer’s disease. Inflammopharmacology. 2020;28(1):95–110.PubMedCrossRef Yao K, Zu HB. Microglial polarization: novel therapeutic mechanism against Alzheimer’s disease. Inflammopharmacology. 2020;28(1):95–110.PubMedCrossRef
73.
Zurück zum Zitat Xiao Q, Yu W, Tian Q, Fu X, Wang X, Gu M, et al. Chitinase1 contributed to a potential protection via microglia polarization and Abeta oligomer reduction in D-galactose and aluminum-induced rat model with cognitive impairments. Neuroscience. 2017;355:61–70.PubMedCrossRef Xiao Q, Yu W, Tian Q, Fu X, Wang X, Gu M, et al. Chitinase1 contributed to a potential protection via microglia polarization and Abeta oligomer reduction in D-galactose and aluminum-induced rat model with cognitive impairments. Neuroscience. 2017;355:61–70.PubMedCrossRef
74.
Zurück zum Zitat Li Y, Yang YY, Ren JL, Xu F, Chen FM, Li A. Exosomes secreted by stem cells from human exfoliated deciduous teeth contribute to functional recovery after traumatic brain injury by shifting microglia M1/M2 polarization in rats. Stem Cell Res Ther. 2017;8(1):198.PubMedPubMedCentralCrossRef Li Y, Yang YY, Ren JL, Xu F, Chen FM, Li A. Exosomes secreted by stem cells from human exfoliated deciduous teeth contribute to functional recovery after traumatic brain injury by shifting microglia M1/M2 polarization in rats. Stem Cell Res Ther. 2017;8(1):198.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Liu W, Rong Y, Wang J, Zhou Z, Ge X, Ji C, et al. Exosome-shuttled miR-216a-5p from hypoxic preconditioned mesenchymal stem cells repair traumatic spinal cord injury by shifting microglial M1/M2 polarization. J Neuroinflamm. 2020;17(1):47.CrossRef Liu W, Rong Y, Wang J, Zhou Z, Ge X, Ji C, et al. Exosome-shuttled miR-216a-5p from hypoxic preconditioned mesenchymal stem cells repair traumatic spinal cord injury by shifting microglial M1/M2 polarization. J Neuroinflamm. 2020;17(1):47.CrossRef
76.
Zurück zum Zitat Brabeck C, Michetti F, Geloso MC, Corvino V, Goezalan F, Meyermann R, et al. Expression of EMAP-II by activated monocytes/microglial cells in different regions of the rat hippocampus after trimethyltin-induced brain damage. Exp Neurol. 2002;177(1):341–6.PubMedCrossRef Brabeck C, Michetti F, Geloso MC, Corvino V, Goezalan F, Meyermann R, et al. Expression of EMAP-II by activated monocytes/microglial cells in different regions of the rat hippocampus after trimethyltin-induced brain damage. Exp Neurol. 2002;177(1):341–6.PubMedCrossRef
77.
Zurück zum Zitat Harry GJ, Funk JA, Lefebvre d’Hellencourt C, McPherson CA, Aoyama M. The type 1 interleukin 1 receptor is not required for the death of murine hippocampal dentate granule cells and microglia activation. Brain Res. 2008;1194:8–20.PubMedCrossRef Harry GJ, Funk JA, Lefebvre d’Hellencourt C, McPherson CA, Aoyama M. The type 1 interleukin 1 receptor is not required for the death of murine hippocampal dentate granule cells and microglia activation. Brain Res. 2008;1194:8–20.PubMedCrossRef
78.
Zurück zum Zitat Reali C, Scintu F, Pillai R, Donato R, Michetti F, Sogos V. S100b counteracts effects of the neurotoxicant trimethyltin on astrocytes and microglia. J Neurosci Res. 2005;81(5):677–86.PubMedCrossRef Reali C, Scintu F, Pillai R, Donato R, Michetti F, Sogos V. S100b counteracts effects of the neurotoxicant trimethyltin on astrocytes and microglia. J Neurosci Res. 2005;81(5):677–86.PubMedCrossRef
79.
Zurück zum Zitat Rohl C, Sievers J. Microglia is activated by astrocytes in trimethyltin intoxication. Toxicol Appl Pharmacol. 2005;204(1):36–45.PubMedCrossRef Rohl C, Sievers J. Microglia is activated by astrocytes in trimethyltin intoxication. Toxicol Appl Pharmacol. 2005;204(1):36–45.PubMedCrossRef
80.
Zurück zum Zitat Anderson KM, Olson KE, Estes KA, Flanagan K, Gendelman HE, Mosley RL. Dual destructive and protective roles of adaptive immunity in neurodegenerative disorders. Transl Neurodegener. 2014;3(1):25.PubMedPubMedCentralCrossRef Anderson KM, Olson KE, Estes KA, Flanagan K, Gendelman HE, Mosley RL. Dual destructive and protective roles of adaptive immunity in neurodegenerative disorders. Transl Neurodegener. 2014;3(1):25.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Motataianu A, Barcutean L, Balasa R. Neuroimmunity in amyotrophic lateral sclerosis: focus on microglia. Amyotrophic Lateral Sclerosis Frontotemporal Degener. 2020;21(3–4):159–66.CrossRef Motataianu A, Barcutean L, Balasa R. Neuroimmunity in amyotrophic lateral sclerosis: focus on microglia. Amyotrophic Lateral Sclerosis Frontotemporal Degener. 2020;21(3–4):159–66.CrossRef
Metadaten
Titel
Neuroprotective effects of ex vivo-expanded regulatory T cells on trimethyltin-induced neurodegeneration in mice
verfasst von
Seon-Young Park
HyeJin Yang
Minsook Ye
Xiao Liu
Insop Shim
Young-Tae Chang
Hyunsu Bae
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2022
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02512-z

Weitere Artikel der Ausgabe 1/2022

Journal of Neuroinflammation 1/2022 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Prämenstruelle Beschwerden mit Suizidrisiko assoziiert

04.06.2024 Suizidalität Nachrichten

Manche Frauen, die regelmäßig psychische und körperliche Symptome vor ihrer Menstruation erleben, haben ein deutlich erhöhtes Suizidrisiko. Jüngere Frauen sind besonders gefährdet.

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Demenzkranke durch Antipsychotika vielfach gefährdet

Demenz Nachrichten

Der Einsatz von Antipsychotika gegen psychische und Verhaltenssymptome in Zusammenhang mit Demenzerkrankungen erfordert eine sorgfältige Nutzen-Risiken-Abwägung. Neuen Erkenntnissen zufolge sind auf der Risikoseite weitere schwerwiegende Ereignisse zu berücksichtigen.

Schlaganfall: frühzeitige Blutdrucksenkung im Krankenwagen ohne Nutzen

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.