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

Open Access 01.12.2022 | Research

Social isolation induces neuroinflammation and microglia overactivation, while dihydromyricetin prevents and improves them

verfasst von: Alzahra J. Al Omran, Amy S. Shao, Saki Watanabe, Zeyu Zhang, Jifeng Zhang, Chen Xue, Junji Watanabe, Daryl L. Davies, Xuesi M. Shao, Jing Liang

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2022

Abstract

Background

Anxiety disorders are the most prevalent mental illnesses in the U.S. and are estimated to consume one-third of the country’s mental health treatment cost. Although anxiolytic therapies are available, many patients still exhibit treatment resistance, relapse, or substantial side effects. Further, due to the COVID-19 pandemic and stay-at-home order, social isolation, fear of the pandemic, and unprecedented times, the incidence of anxiety has dramatically increased. Previously, we have demonstrated dihydromyricetin (DHM), the major bioactive flavonoid extracted from Ampelopsis grossedentata, exhibits anxiolytic properties in a mouse model of social isolation-induced anxiety. Because GABAergic transmission modulates the immune system in addition to the inhibitory signal transmission, we investigated the effects of short-term social isolation on the neuroimmune system.

Methods

Eight-week-old male C57BL/6 mice were housed under absolute social isolation for 4 weeks. The anxiety-like behaviors after DHM treatment were examined using elevated plus-maze and open field behavioral tests. Gephyrin protein expression, microglial profile changes, NF-κB pathway activation, cytokine level, and serum corticosterone were measured.

Results

Socially isolated mice showed increased anxiety levels, reduced exploratory behaviors, and reduced gephyrin levels. Also, a dynamic alteration in hippocampal microglia were detected illustrated as a decline in microglia number and overactivation as determined by significant morphological changes including decreases in lacunarity, perimeter, and cell size and increase in cell density. Moreover, social isolation induced an increase in serum corticosterone level and activation in NF-κB pathway. Notably, DHM treatment counteracted these changes.

Conclusion

The results suggest that social isolation contributes to neuroinflammation, while DHM has the ability to improve neuroinflammation induced by anxiety.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12974-021-02368-9.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
DHM
Dihydromyricetin
NF-κB
Nuclear factor-κB
CNS
Central nervous system
PTSD
Posttraumatic stress disorder
GABAARs
GABAA receptors
PAM
Positive allosteric modulator
HPA
Hypothalamic–pituitary–adrenal
MAPK
Mitogen-activated protein kinase
BSA
Bovine serum albumin
PBS
Phosphate buffer saline
TBST
Tris-buffered saline with Tween 20
ELISA
Enzyme linked immunosorbent assay
ANOVA
Analysis of variance
EPM
Elevated plus-maze
OF
Open field
GAD67
Decarboxylase 67
AD
Alzheimer’s disease

Background

Anxiety disorders are the largest class of mental health diseases in the U.S. and are estimated to consume one-third of the country’s mental health spending. Considering the ineffective therapeutics in the clinic due to treatment resistance, inconsistent patient adherence, and other exogenous factors, the prevalence of chronic anxiety continues to be on the rise [13]. The rise in individuals suffering anxiety has been particularly noticeable during the ongoing COVID-19 pandemic that has resulted in quarantine and social isolation [4]. The pandemic also has led to an increase in posttraumatic stress disorder (PTSD) [5]. The pandemic is bringing up similar fears and mental distress, due to loss of family member(s) and loved ones [5, 6]. The ongoing increase in cases of individuals suffering from anxiety disorders, coupled with the lack of effective medications for many, highlight the need for new treatment strategies to prevent or manage anxiety disorders.
Clinical evidence indicates that GABAergic neurotransmission alteration involves in pathophysiology of anxiety disorders in humans [7]. Therefore, modifying GABAA receptor (GABAARs) activity is a target for regulating anxiety [7, 8]. We have demonstrated that dihydromyricetin (DHM) [(2R,3R)-3,5,7-trihydroxy-2-(3,4,5-trihydroxyphenyl)-2,3-dihydrochromen-4-one], a major bioactive flavonoid extracted from Ampelopsis grossedentata, is a positive allosteric modulator (PAM) of GABAergic transmission, and thus has the potential to regulate anxiety-like behavior via its action on GABAergic receptors [9, 10]. In addition, we found that DHM antagonizes the acute and chronic effects of alcohol on GABAARs [9, 10]. Thus, the activity of DHM on GABAARs provides one possible mechanism for its role in anxiolysis [10]. Because GABAergic transmission modulates the immune system in addition to the inhibitory signal transmission, we investigate the effects of social isolation on the neuroimmune system. To date, the role of neuroinflammation in anxiety pathogenesis is not well established. Several studies had highlighted the regulatory role of GABA in neuroimmune functions. GABA involves in inflammation regulation by modulating the production of proinflammatory cytokines through activating inflammatory signaling pathways such as nuclear factor-κB (NFκB) and mitogen-activated protein kinase (MAPK) [11, 12]. Indeed, GABAergic system components, including GABA enzyme, transports, and receptors, are expressed in the immune cells [13, 14].
The extended psychological stress that occurs due to social isolation disrupts the hypothalamic–pituitary–adrenal (HPA) axis, which is considered the primary stress adaption pathway in the body [15]. This disruption has the potential to increase proinflammatory cytokines, persistent microglia and astrocytes activation, and reduce synaptic plasticity [16]. Additionally, several studies indicated that GABAergic deficits during stress contribute to HPA hyperactivity that correlated with anxiety pathogenesis [17, 18]. The outcome of these immunoendocrine dysregulations could lead to the occurrence of neuroinflammation and anxiety.
Microglia, the resident macrophages of the brain’s innate system, are a key player in modulating the neuroinflammatory response in the CNS. Microglia are not only involved in the nervous system infection and debris phagocytosis, but also play a crucial role in the physiological development of the brain by engaging in the shaping process of neuronal circuits and synapse plasticity [19, 20]. During various psychological stressors such as social isolation, microglia undergo several changes that compromise their functions and further lead to neurological and neurodegenerative disorders [21]. To understand the role of microglial in social isolation-induced anxiety, we utilized a social isolation mouse model that induces anxiety via reduction of social interaction as a stressor. The primary goal of this study was to determine whether social isolation-induced anxiety leads to neuroinflammation, and to understand the pharmacological mechanisms of DHM as an alternative therapy for preventing and reducing neuroinflammation.

Methods

Animals and treatments

Overview

Eight-week-old male C57BL/6 mice (Charles River Laboratories, Hollister, CA) were housed in the vivarium under a 12 h light/dark cycle with direct bedding and free access to food and water. All animal experiments were performed according to the protocols approved by the University of California, Los Angeles (UCLA) and the University of Southern California (USC) Institutional Animal Care and Use Committees, and all methods were carried out in accordance with relevant guidelines and regulations. Animals were habituated to the vivarium for 2 days before beginning experimentation.

Social isolation

Social isolation is reported to elicit anxious and depressive behaviors in rodents [2225]. For the present study, we modified these methods to induce stress associated with social isolation by using opaque-walled cages, thus depriving the animals of environmental enhancers (e.g., toys, objects, etc.). We investigated anxiety-like behaviors after 4 and 6 weeks post-social isolation to determine behavioral responses comparable to the established 4–6 weeks isolation that results in anxiety [24]. We used these time points to determine the potential therapeutic effects of DHM. Group-housed mice were housed with the standard 3–4 mice per cage. Isolated mice were singly housed with opaque walls without human handling except to change cages once per week.
The mice were divided as follows:
1.
Group-housed mice without any drug administration for 2 weeks, and then given daily sucrose agar as a vehicle for an additional 2 weeks (G2 + Veh2).
 
2.
Group-housed mice without any drug administration for 2 weeks, and then given daily DHM in sucrose + agar for an additional 2 weeks (G2 + D2, 2 mg/kg DHM).
 
3.
Isolated mice without any drug administration for 2 weeks, and then given daily sucrose agar as a vehicle for an additional 2 weeks for a total isolation period of 4 weeks (Iso2 + Veh2).
 
4.
Isolated mice without any drug administration for 2 weeks, and then given daily DHM for an additional 2 weeks for a total isolation period of 4 weeks (Iso2 + D2).
 
5.
Isolated mice without any drug administration for 4 weeks, and then given daily vehicle for an additional 2 weeks for a total isolation period of 6 weeks (Iso4 + Veh2).
 
6.
Isolated mice without any drug administration for 4 weeks, and then given daily oral administration of DHM for an additional 2 weeks for a total isolation period of 6 weeks (Iso4 + D2).
 

Drug preparations

DHM (HPLC purified ≥ 98%, Master Herbs Inc., Pomona, CA) was given orally as agar cube once per day (2 mg/kg) for 2 weeks [10]. To prepare the DHM or vehicle agar cube, 3% agar was prepared with water, heated to ~ 90 °C to dissolve the agar, then DHM + 5% sucrose or 5% sucrose only were added and mixed until cooled and solidified. Agar was prepared for the mice by cutting it into cubes of 0.5 × 0.5 × 0.5 cm each.

Drug administration

Every evening (2 PM), all food from the cages of each mouse was removed, and an agar cube was placed in the cage for each mouse. The mouse was observed to ensure it ate the agar cube, which ranged from 30 to 90 min. Afterward, 4 g of regular rodent food (the recommended daily amount for an adult mouse; was placed in the cage for each mouse for the rest of the day [26]. To ensure each mouse of the group housing mice received one cube, they were isolated, fed, and then returned to group housing.

Behavioral testing

Anxiety-like behavior was tested 24 h after the last treatment, at the end of the 4-week or 6-week time points in the following evening (8 PM; in the dark phase of 12/12-h light/dark cycle). Anxiety tests were reliant on ethologically appropriate behavior and sensitive to ‘state’ anxiety measurements. Because mice are nocturnal animals, behavior tests were conducted during their active time to ensure accurate behavioral responses and minimize interference of their circadian cycle. Behavioral tests were performed under indirect red lighting and recorded with a video camera. Indirect red lighting was used to better assess parameters of anxiety without influencing mouse behavior (i.e., reduced activity in the open field test) and stress [27]. Investigators were blind to experimental groups when conducting behavioral analyses.

Elevated plus-maze

The elevated plus-maze was conducted following a previously published protocol [28]. The elevated plus-maze apparatus was made of opaque, 0.6-cm-thick plastic. It comprised two open arms 25 × 8 cm across from each other and perpendicular to two closed arms 25 × 8 cm with a center platform of 8 × 8 cm. The closed arms had a 20-cm-high wall that enclosed the arms. The walls and floors of the closed arms were black, while the open arms were white. The apparatus was elevated 50 cm above the floor. Throughout the test, each animal was placed in the center of the maze facing an open arm and allowed to explore for 5 min. The behaviors were recorded by a ceiling-mounted camera. Entry into an arm of the maze was defined by the placement of at least 3 paws into that compartment. The following measures were physiological scoring system: number of entries into open arms, closed arms, or center platform and time spent in each of these areas. All scoring was performed offline in a double-blind manner.

Open field

The open field test was conducted following a previously published protocol [10, 29]. The open field chamber measured 50 cm (length) × 50 cm (width) × 38 cm (height) and was made from a white acrylic plastic sheet. 4 × 4 grid lines were drawn to divide the floor into 10 × 10 cm squares, and an additional 20 × 20 cm square zone was drawn in the center. Mouse activity was assessed as previously reported in Chen et al., 2004 for 10 min. The following parameters were summed for each animal during the 10-min test: the time spent in the central zone, time spent in the 4-corner square grid, pathlength (cm) traveled in the apparatus (determined by measuring the distance of the nose of the mouse relative to the 10 × 10 cm square grid lines on the floor of the open field chamber), and the numbers of times the animal reared. All scoring was conducted manually in a double-blind manner, with each recording being observed three times to minimize error.

Immunohistochemistry analysis

Mouse brain was fixed in 4% formaldehyde for 24 h, and then incubated in 30% sucrose until tissues are sink. Fixed brain was flash frozen using pre-cooled isopentane (− 78 °C), sectioned at 30 μm using Microm HM525 Cryostat (Thermo) and picked up on Superfrost Plus slides (VWR, 48311-703). Sections were blocked with 5% normal goat serum and washed in PBS with 1% bovine serum albumin (BSA) and incubated with rabbit- anti-mouse Iba-1 primary antibody (FUJIFILM Wako Pure Chemical Corporation 019-19741, 1:500) or rabbit- anti-mouse Iba-1 primary antibody Alexa Fluor® 594 conjugate (Cell Signaling, 48934, 1:50) overnight. Sections were washed with phosphate buffer with 1% Tween 20 (PBS-T), and then incubated in goat anti-rabbit IgG (H + L) secondary antibody (Vector laboratory CY-1300, 1:250) at room temperature for 1 h when unconjugated antibody was used. Afterword, sections were washed three times with PBS-T followed by mounting on coverslip using Vectashield DAPI (4′6-diamidino-2-phenylindole 2HCl, Vector Labs, Burlingame, U.S.) mounting media to detect nuclei.

Imaging and analysis

The immune-stained sections were scanned for high-resolution images by Cytation 5 cell imaging multi-mode reader (BioTek, Winooski, VT, USA) or super-resolution images by Zeiss LSM880 with Airyscan confocal laser scanning microscope (Carl Zeiss Microscopy, White Plains, NY). The images from the CA1 and CA2 of the brain hippocampus were processed by Zen black and blue imaging analysis software (Carl Zeiss Microscopy) and analyzed by FracLac box counting and convex hull analysis to evaluate the morphological changes of microglia cells using ImageJ software by following the steps published in Young and Morrison, 2018 [30]. Microglia cells count, within the region of interest ROI (0.3 × 0.4 mm) in the CA1 and CA2 hippocampus area, was done manually blinded to the treatment group using ImageJ cell count command and presented in cells per 1 mm2.

Western blot analysis

Hippocampus was homogenized in pre-cooled Tris–EDTA with 1% PMSF (0.1 M Tris–acetate buffer + 2 mM EDTA, pH 7.75). The homogenate was centrifuged at 10,000×g for 15 min at 4 °C and the supernatant was collected. The supernatant protein was quantified using the BCA Protein Assay kit (Pierce Biotechnology, Rockford, IL) according to the manufacturer’s instructions. 50 μg of protein was separated on a 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis and then transferred to PVDF membranes (Bio-Rad Laboratories, Hercules, CA). The transferred membrane was blocked with a blocking buffer containing 5% skim milk in 1X Tris-buffered saline with Tween 20 (TBST) for 1 h at room temperature. The membrane was incubated in the following primary antibody for either: rabbit anti-mouse gephyrin (Cell Signaling 14304, 1:1000), mouse anti-mouse β-actin (Cell Signaling 4970, 1:1000), rabbit anti-mouse NF-κB p65 (Cell Signaling 8242, 1:1000) or rabbit anti-mouse phospho-NF-κB p65 (Cell Signaling 3033, 1:1000) in 1X TBST overnight at 4 °C with gentle agitation. The membrane was washed three times with 1X TBST for 10 min each and incubated with a secondary antibody goat anti-rabbit IgG or goat anti-mouse (Bio-Rad 1706515 and 1706516) in 1X TBST for 1 h. Finally, the membrane was visualized with enhanced chemiluminescence detection reagent (Bio-Rad 1705061) and Chemi-Doc (Bio-Rad) imaging device.

Cytokines profile arrays

Serum samples were pooled from 4 to 5 mice/group (n = 4 vehicle grouped housed, n = 4 DHM grouped housed, n = 5 vehicle social isolated, n = 5 DHM in social isolated group). According to the manufacturer’s instructions, pooled sera were tested by Profiler Mouse Cytokine Array Kit, Panel A (ARY006, R&D Systems, Minneapolis, MN, USA) for a quantitative measure of the peripheral cytokines and chemokines level to determine changes in response to the social isolation-induced anxiety. The membrane immunoreactivity was detected after adding chemiluminescence reagent mix. Array images were obtained using (Chemi-Doc imager system (Bio-Rad). The results were analyzed using ImageJ software and expressed as a mean change in the gray value relative to the vehicle grouped housed groups.

ELISA analysis

Fresh blood was collected from the right atrium quickly after the animals were killed. serum samples were collected by centrifugation at 1000×g for 10 min at 4 °C. A competitive ELISA kit (Abcam, ab108821) was used to determine the corticosterone levels according to the manufacturer’s protocol. The intensity of the color developed was measured using Synergy H1 Hybrid Multi-Mode Reader (BioTek).

Statistical analysis

All assays were performed at least three times. All behavioral records were observed and analyzed in a double-blind manner. The data were presented as the mean ± standard error of the mean using GraphPad Prism 9 (GraphPad Software, Inc., La Jolla, CA). One or two-way analysis of variance (ANOVA) followed by Holm–Sidak multiple comparison tests were performed, the significance level is set at p ≤ 0.05.

Results

DHM treatment ameliorates social isolation-induced anxiety-like behaviors

Using a mouse model of social isolation stress, we examined social isolation stress induced anxiety levels and the effects of DHM with the elevated plus-maze (EPM) and open field (OF) tests [3133]. Group-housed control mice (G2 + Veh2) spent 2.31 ± 0.27 (min) of a total 5 min in open arms of the elevated plus-maze (Fig. 1A) and 2.24 ± 0.31 min in the closed arm. Mice socially isolated for 2 weeks, followed by 2 weeks of vehicle treatment (Iso2 + Veh2) spent substantially less time (1.26 ± 0.17 min) in the open arms compared to the closed arms (3.31 ± 0.27 min). Socially isolated mice treated with DHM (Iso2 + D2) resulted in greater time spent in the open arm (2.07 ± 0.22 min) when compared with untreated socially isolated mice (Additional file 1: Table S1). These results suggest that social isolation increased anxiety levels. Furthermore, DHM administration ameliorates isolation-induced anxiety behaviors, as observed with increased entry into and staying in the open arms.
To further examine anxiety-like behavior, locomotor activity, and exploration behavior were analyzed by measuring the distance traveled of the mice in the open field test (Fig. 1B). During the 10 min observation trial, group-housed mice (G2 + Veh2) traveled 2765 ± 161 cm, while isolated mice (Iso2 + Veh2) traveled a shorter (2176 ± 145 cm) pathlength suggesting that social isolation decreased motor activity of these mice. In contrast, isolated mice treated with DHM (Iso2 + D2) resulted in greater running distance (2398 ± 147 cm). The number of rearing and time spent in the center of the open field was significantly decreased in mice housed in isolation compared to that of group-housed mice (46.6 ± 1.5 times vs 28.3 ± 2.1 times) (19.6 ± 0.7 vs 7.6 ± 0.86). Social isolation mice spent more time in the corners compared to group-housed mice (73 ± 4.2 s vs 28.2 ± 2.06 s). Administration of DHM in Iso2 + D2 increased the number of rearing (40.7 ± 1.03 times) and the time in the center (18.3 ± 0.49 s) while decreasing stay in the corner duration (38.5 ± 3.5 s) (Additional file 1: Table S1). Collectively, these results suggest that isolation decreased exploratory/locomotor activity in adult male C57BL/6J mice and that DHM treatment ameliorates these behavioral responses in socially isolated mice.

Social isolation down-regulates gephyrin protein expression, while DHM treatment improves it

Gephyrin is a scaffold protein essential for GABAAR clustering via several mechanisms [34, 35]. The changes in the expression of gephyrin can partially explain the changes in GABAergic neurotransmission. Gephyrin protein expression was measured in extracted hippocampi and evaluated by Western blot. Gephyrin expression was 40% lower in isolated mice compared to group-housed mice (Fig. 2) (Additional file 1: Table S2). DHM treatment upregulated gephyrin expression at Iso4 + D2 group relative to isolated mice without DHM. Collectively, the behavioral assay and the gephyrin results confirm the phenotype of the social isolation animal model. The results also suggest that DHM improves gephyrin expression levels in hippocampi and subsequent GABAAR function.

Social isolation induces loss and dystrophy in the hippocampus microglia, while DHM improves it

Microglia number was reduced after short-term social isolation in the CA hippocampus brain region in addition to alteration in morphological characteristics of microglia cells. The changes appeared due to the activation of cells toward more dystrophic morphology and apoptosis leading to decrease in microglia number compared to the basal state. Microglia counts in social isolation group (Iso2 + Veh2) was lower (28.50 ± 1.64 cells/mm2) compared to the control group G2 + Veh2 (37.27 ± 2.1) (Fig. 3C). Moreover, microglia cells in Iso2 + Veh2 show a significant decrease in the lacunarity value from 0.353 ± 0.018 to 0.302 ± 0.009 (Fig. 4B) as an evidence of microglia activation that was restored after DHM treatment. Lacunarity measures cell shape heterogenicity and changes in the soma [36]. Moreover, a decrease in microglial cell area was observed in the social isolation group (Iso2 + Veh2) illustrated in the reduction of the perimeter of the individual cell outline, where social isolation for 2 weeks followed by 2 weeks of DHM treatment (Iso2 + D2) showed an increase in cell perimeter from 278.7 ± 16.76 to 382.9 ± 21.56 µm [F (2, 28) = 17.72, p ≤ 0.0001] (Fig. 4C). Iso2 + Veh2 microglia cells demonstrated a more compact shape illustrated in significantly higher cell density [F (2, 27) = 3.388, p = 0.0387] (Fig. 3D]. In contrast, the DHM isolated group (Iso2 + D2) showed higher cell density which was statistically similar to the control grouped house (G2 + Veh2). In addition, we evaluated the maximum distance between two points across the convex hull and found that DHM administered social isolated group (Iso2 + D2) had an average distance relatively similar to the control group house (G2 + Veh2) (Fig. 4E), while the socially isolated group (Iso2 + Veh2) had a substantially smaller average distance across the convex hull from 172.5 ± 12.04 to 103.2 ± 6.40 µm [F (2, 28) = 14.36, p ≤ 0.0001] (Additional file 1: Table S3). These results suggest a modulation in several microglia morphological parameters related to the microglia activation after social isolation, indicating an acute response to social isolation challenges, which were ameliorated by DHM administration.

Social isolation anxiety increases serum corticosterone level, activates NF-κB signaling pathway, and increases proinflammatory cytokines, while DHM attenuates these changes

Corticosterone is the primary hormone of the pituitary adrenocortical axis in response to environmental challenges, and it has an essential function in stress [37]. Corticosterone level was evaluated after social isolation to illustrate the effect of social isolation on the hypothalamic–pituitary–adrenal (HPA) axis. Our results revealed a significant increase in the serum corticosterone level from 285.8 ± 26.65 ng/ml in G2 + Veh2 to 466.0 ± 60.51 ng/ml in Iso2 + Veh2 group [F (3, 15) = 7.897], (p = 0.002) (Fig. 5A). Administration of DHM at the dose of 2 mg/kg in the isolation group decreased the corticosterone level to 283.3 ± 27.43 ng/ml closed to the basal level of G2 + Veh2 (Additional file 1: Table S4). This observation suggests that corticosterone responds to social isolation-induced stress challenges, and DHM attenuates the stress (anxiety) following social isolation and reduces corticosterone levels.
To determine the effect of social isolation on inflammation, we examined the protein expression level of NF-κB p65 in extracted hippocampi utilizing Western blot. NF-κB transcription factor is an essential inflammatory pathway that plays a crucial role in regulating immune and inflammatory responses by inducing the expression of cytokines and chemokines genes [38]. The active phosphorylated NF-κB p65 expression was significantly higher in the Iso2 + Veh2 group compared to the control. DHM treatment group (Iso2 + D2) shows a comparable phospho-NF-κBp65 protein level to the control (G2 + Veh2) and treatment control (G2 + D2) (Fig. 5B) (p = 0.574). An increase in the expression of several numbers of proinflammatory cytokines and chemokines was observed using cytokines profile proteome assay in the isolated group (Iso2 + Veh2) compared to the control and DHM group-housed (G2 + Veh2) (G2 + D2) (Fig. 5C). The DHM treated isolated group (Iso2 + D2) showed lower cytokines and chemokines expression levels compared to (Iso2 + Veh2) (Additional file 1: Table S2). These data suggest that social isolation activates the NF-κB signaling pathway leading to an increase in the transcription of proinflammatory cytokines while DHM treatment counteract them.

Discussion

The primary goal of this study was to determine the effects of anxiety induced by short-term social isolation on neuroinflammation and to understand pharmacological mechanisms underlying the therapeutic effects of DHM. In this study, we found that social isolation induced anxiety-like behaviors in mice, increased corticosterone level, down-regulated gephyrin expression, enhanced the activation of NF-kB p65 pathway, decrease hippocampal microglial cell number, and increase the reactive microglia. Importantly, these pathological changes and behavioral deficits were ameliorated by DHM treatment. Findings from the present work suggest that short-term social isolation leads to changes in the HPA axis, disrupts GABAergic neurotransmission, and provokes neuroinflammation. DHM was found to restore these molecular and cellular changes (Fig. 6).
Gephyrin is a key protein that anchors, clusters, and stabilizes GABAergic synapses [39]. Mounting evidence has indicated the critical role of GABA transmission in the progression of anxiety disorder [40]. In our previous study, we found that changes in gephyrin expression play a role in changes in GABAAR amount and GABAergic function [10, 33]. Additionally, we found a reduction in GABAAR-mediated extra-synaptic, pre-, and post-synaptic currents in the social isolation anxiety model, which was restored after DHM treatment [33]. To elucidate the mechanism in which DHM modulates GABAARs in the socially isolated model, gephyrin protein expression was assessed utilizing Western blot. Gephyrin protein expression was reduced 40% in the isolation group and upregulated after DHM treatment (Fig. 1A). Taking into consideration that GABAAR expression is ubiquitous in the brain and the primary role of GABA neurotransmitter in anxiety pathogenesis, a reduction in gephyrin protein level might provide a partial explanation of the behavioral changes shown in (Fig. 1B). It is essential to emphasize the interconnection between GABA and neuroinflammation. Neuroinflammation decreases GABA synthesis by reducing glutamate acid decarboxylase 67 (GAD67) enzyme, downregulating GABAAR protein expression, and inhibiting GABA current by decrease GABA neurons density [41]. Therefore, studies of neuroinflammation will further clarify the underlining mechanisms of DHM effects on improving anxiety-like behaviors.
As part of the innate immune system, microglia play a significant role in initiating and mediating neuroinflammation in the brain [42]. Microglia are not only involved in brain infection and debris phagocytosis, but also play a crucial role in the physiological development of the brain by engaging in the shaping process of neuronal circuits and synapse plasticity [19, 20]. Microglia activation occurred after various types of stress [21, 43, 44]. We assessed microgliosis after short-term social isolation using immunohistochemistry staining of Iba1, one of the most common markers of microglia [4547]. These data indicate that social isolation-induced anxiety led to a decline in microglia cell number (Fig. 3C). Several evidence demonstrate a tight connection between the microglial loss mediated by overactivation and social isolation and other form of stress [4850]. Additionally, social isolation triggers microglia activation, manifested as changes in the cell morphology. High lacunarity value indicates the heterogeneity of the cells, meaning the single-cell image of microglia has different gap sizes. On the other hand, a low lacunarity value indicates homogeneity, suggesting the cell shape has less variance [36]. Under the influence of social isolation, the microglial cell showed a lower lacunarity value compared to the control. This implies the transformation of the microglial cell to a more homogenous state. This change generally suggests activation of the microglia from its surveillant state [51]. Furthermore, the activated microglia cells in socially isolated mice showed a reduction in cell size, based on the measurement of the perimeter of single cell outlines. Reduction of cell perimeter is an illustration of fewer ramifications, shorter and thicker branches, and a larger soma size of the cell that manifested during activation [52]. Microglia density is another morphometric parameter that was altered after social isolation and DHM treatment. Higher cell density demonstrates the transformation of the cells to a more compact form during activation [53, 54]. The maximum span across the hull in each microglial cell was significantly smaller in the social isolation group than the control, reflecting the size of the overall cells and the length of the processes. Microglia in less ramified form is considered activated or intermediate active, which explains the reduction in the hull span size [51, 55]. These results are consistent with the findings from several groups that demonstrated the activated microglia contribute to anxiety induced by social isolation [56, 57]. In addition, several studies showed that microgliosis mediate synapses loss that is considered the earliest manifestation of AD pathology [5860].
Accumulated evidence suggests a link between neuroinflammation and the pathology of anxiety and other psychological disorders [44, 61]. Neuroinflammation is involved in several pathological changes in the nervous system and the neuroendocrine system, and it is initiated as a result of the modulation of the HPA axis [62]. Chronic exposure to stress leads to disruption in HPA axis and an increase in the cortisol level in humans and corticosterone in rodents [63]. Animal studies of the social isolation model have shown that the corticosterone level is higher after exposure to stress than the group-housed control [64]. There was an increase in serum corticosterone level after 4 weeks of social isolation; and our results demonstrate that DHM treatment at doses of 2 mg/kg reduces corticosterone levels.
The transcription factor NF-κB plays a key function in regulating immune and inflammatory responses by inducing the expression of cytokines and chemokines genes [38]. Growing evidence suggests that stress-induced behavioral deficits are mediated by NF-κB signaling activation, which increases the production of the proinflammatory cytokine and chemokines leading to microglia activation and ultimately neuroinflammation [65, 66]. Our results indicate an increase in the phospho-NF-kB p65 protein level compared to the nonactive NF-kB p65, which might explain the increase of proinflammatory cytokines expression, and the microglial activation observed in the social isolation group. Additionally, DHM treatment attenuates the activation of NF-κB pathway induced by social isolation, and this finding is consistent with a previous work that demonstrated a reduction in NF-κB protein expression after DHM treatment [67].

Study limitations

A previous study from our team showed no difference in GABAARs-mediated neurotransmission and gephyrin level between 2-week and 4-week social isolation [33]. The current study in neuroinflammation focused only on 2 weeks isolation. However, future studies might be necessary to include more different time points.
The gender choice in the current study was based on a previous publication from our team [33], which was conducted only on males. Future research on the effects of DHM in socially isolated female mice is necessary to gain insight into the therapeutics role of DHM in both genders. Despite the limitations, the current study provides important data for understanding possible mechanisms of actions of DHM in ameliorating anxiety-like behaviors.

Conclusion

There has been a growing body of evidence supporting the association of anxiety with cognitive decline leading researchers to conclude that anxiety symptoms could be predictive for the progression of Alzheimer’s disease (AD). This work provided insights into the mechanisms that anxiety induces neuroinflammation and DHM reverses the neuropathology resulted from anxiety as a component of preclinical AD, as well as the utility of DHM as a novel therapy for anxiety. DHM reduces neuroinflammation, restores GABAergic function by upregulating gephyrin levels, and decreases serum cortisol levels, therefore, improves the social isolation induced anxiety and the early onset of AD. DHM could provide an early intervention to manage anxiety and to reduce the risk of cognitive decline and AD development.

Acknowledgements

Carefree Biotechnology Foundation, and Army Health Professions Scholarship Program (HPSP) (to A.S.S.), Saudi Arabian Cultural Mission (SACM) (to A.J.A).

Declarations

All animal experiments were performed according to the protocols approved by the University of California, Los Angeles (UCLA) and the University of Southern California (USC) Institutional Animal Care and Use Committees, and all methods were carried out in accordance with relevant guidelines and regulations.
Not applicable.

Competing interests

The authors declare no conflict of interest and competing interests.
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 Devane CL, Chiao E, Franklin M, Kruep EJ. Anxiety disorders in the 21st century: status, challenges, opportunities, and comorbidity with depression. Am J Manag Care. 2005;11(12 Suppl):S344–53.PubMed Devane CL, Chiao E, Franklin M, Kruep EJ. Anxiety disorders in the 21st century: status, challenges, opportunities, and comorbidity with depression. Am J Manag Care. 2005;11(12 Suppl):S344–53.PubMed
2.
Zurück zum Zitat Konnopka A, Leichsenring F, Leibing E, König HH. Cost-of-illness studies and cost-effectiveness analyses in anxiety disorders: a systematic review. J Affect Disord. 2009;114(1–3):14–31.PubMedCrossRef Konnopka A, Leichsenring F, Leibing E, König HH. Cost-of-illness studies and cost-effectiveness analyses in anxiety disorders: a systematic review. J Affect Disord. 2009;114(1–3):14–31.PubMedCrossRef
3.
4.
Zurück zum Zitat Twenge JM, Joiner TE. U.S. Census Bureau-assessed prevalence of anxiety and depressive symptoms in 2019 and during the 2020 COVID-19 pandemic. Depress Anxiety. 2020. Twenge JM, Joiner TE. U.S. Census Bureau-assessed prevalence of anxiety and depressive symptoms in 2019 and during the 2020 COVID-19 pandemic. Depress Anxiety. 2020.
5.
Zurück zum Zitat Yuan K, Gong YM, Liu L, Sun YK, Tian SS, Wang YJ, et al. Prevalence of posttraumatic stress disorder after infectious disease pandemics in the twenty-first century, including COVID-19: a meta-analysis and systematic review. Mol Psychiatry. 2021. Yuan K, Gong YM, Liu L, Sun YK, Tian SS, Wang YJ, et al. Prevalence of posttraumatic stress disorder after infectious disease pandemics in the twenty-first century, including COVID-19: a meta-analysis and systematic review. Mol Psychiatry. 2021.
6.
7.
Zurück zum Zitat Rudolph U, Knoflach F. Beyond classical benzodiazepines: novel therapeutic potential of GABAA receptor subtypes. Nat Rev Drug Discov. 2011;10(9):685–97.PubMedPubMedCentralCrossRef Rudolph U, Knoflach F. Beyond classical benzodiazepines: novel therapeutic potential of GABAA receptor subtypes. Nat Rev Drug Discov. 2011;10(9):685–97.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Shekhar A, Hingtgen JN, DiMicco JA. GABA receptors in the posterior hypothalamus regulate experimental anxiety in rats. Brain Res. 1990;512(1):81–8.PubMedCrossRef Shekhar A, Hingtgen JN, DiMicco JA. GABA receptors in the posterior hypothalamus regulate experimental anxiety in rats. Brain Res. 1990;512(1):81–8.PubMedCrossRef
9.
Zurück zum Zitat Shen Y, Lindemeyer AK, Gonzalez C, Shao XM, Spigelman I, Olsen RW, et al. Dihydromyricetin as a novel anti-alcohol intoxication medication. J Neurosci. 2012;32(1):390–401.PubMedPubMedCentralCrossRef Shen Y, Lindemeyer AK, Gonzalez C, Shao XM, Spigelman I, Olsen RW, et al. Dihydromyricetin as a novel anti-alcohol intoxication medication. J Neurosci. 2012;32(1):390–401.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Liang J, López-Valdés HE, Martínez-Coria H, Lindemeyer AK, Shen Y, Shao XM, et al. Dihydromyricetin ameliorates behavioral deficits and reverses neuropathology of transgenic mouse models of Alzheimer’s disease. Neurochem Res. 2014;39(6):1171–81.PubMedCrossRef Liang J, López-Valdés HE, Martínez-Coria H, Lindemeyer AK, Shen Y, Shao XM, et al. Dihydromyricetin ameliorates behavioral deficits and reverses neuropathology of transgenic mouse models of Alzheimer’s disease. Neurochem Res. 2014;39(6):1171–81.PubMedCrossRef
11.
Zurück zum Zitat Crowley T, Fitzpatrick JM, Kuijper T, Cryan JF, O’Toole O, O’Leary OF, et al. Modulation of TLR3/TLR4 inflammatory signaling by the GABAB receptor agonist baclofen in glia and immune cells: relevance to therapeutic effects in multiple sclerosis. Front Cell Neurosci. 2015;9:284.PubMedPubMedCentralCrossRef Crowley T, Fitzpatrick JM, Kuijper T, Cryan JF, O’Toole O, O’Leary OF, et al. Modulation of TLR3/TLR4 inflammatory signaling by the GABAB receptor agonist baclofen in glia and immune cells: relevance to therapeutic effects in multiple sclerosis. Front Cell Neurosci. 2015;9:284.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Bhat R, Axtell R, Mitra A, Miranda M, Lock C, Tsien RW, et al. Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci U S A. 2010;107(6):2580–5.PubMedPubMedCentralCrossRef Bhat R, Axtell R, Mitra A, Miranda M, Lock C, Tsien RW, et al. Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci U S A. 2010;107(6):2580–5.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Schleimer SB, Hinton T, Dixon G, Johnston GA. GABA transporters GAT-1 and GAT-3 in the human dorsolateral prefrontal cortex in schizophrenia. Neuropsychobiology. 2004;50(3):226–30.PubMedCrossRef Schleimer SB, Hinton T, Dixon G, Johnston GA. GABA transporters GAT-1 and GAT-3 in the human dorsolateral prefrontal cortex in schizophrenia. Neuropsychobiology. 2004;50(3):226–30.PubMedCrossRef
14.
Zurück zum Zitat Jin XT, Galvan A, Wichmann T, Smith Y. Localization and function of GABA transporters GAT-1 and GAT-3 in the Basal Ganglia. Front Syst Neurosci. 2011;5:63.PubMedPubMedCentralCrossRef Jin XT, Galvan A, Wichmann T, Smith Y. Localization and function of GABA transporters GAT-1 and GAT-3 in the Basal Ganglia. Front Syst Neurosci. 2011;5:63.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Mumtaz F, Khan MI, Zubair M, Dehpour AR. Neurobiology and consequences of social isolation stress in animal model-A comprehensive review. Biomed Pharmacother. 2018;105:1205–22.PubMedCrossRef Mumtaz F, Khan MI, Zubair M, Dehpour AR. Neurobiology and consequences of social isolation stress in animal model-A comprehensive review. Biomed Pharmacother. 2018;105:1205–22.PubMedCrossRef
17.
Zurück zum Zitat Shen Q, Lal R, Luellen BA, Earnheart JC, Andrews AM, Luscher B. gamma-Aminobutyric acid-type A receptor deficits cause hypothalamic-pituitary-adrenal axis hyperactivity and antidepressant drug sensitivity reminiscent of melancholic forms of depression. Biol Psychiatry. 2010;68(6):512–20.PubMedPubMedCentralCrossRef Shen Q, Lal R, Luellen BA, Earnheart JC, Andrews AM, Luscher B. gamma-Aminobutyric acid-type A receptor deficits cause hypothalamic-pituitary-adrenal axis hyperactivity and antidepressant drug sensitivity reminiscent of melancholic forms of depression. Biol Psychiatry. 2010;68(6):512–20.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Mody I, Maguire J. The reciprocal regulation of stress hormones and GABA(A) receptors. Front Cell Neurosci. 2011;6:4.PubMed Mody I, Maguire J. The reciprocal regulation of stress hormones and GABA(A) receptors. Front Cell Neurosci. 2011;6:4.PubMed
19.
Zurück zum Zitat Salter MW, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell. 2014;158(1):15–24.PubMedCrossRef Salter MW, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell. 2014;158(1):15–24.PubMedCrossRef
20.
Zurück zum Zitat Wake H, Moorhouse AJ, Miyamoto A, Nabekura J. Microglia: actively surveying and shaping neuronal circuit structure and function. Trends Neurosci. 2013;36(4):209–17.PubMedCrossRef Wake H, Moorhouse AJ, Miyamoto A, Nabekura J. Microglia: actively surveying and shaping neuronal circuit structure and function. Trends Neurosci. 2013;36(4):209–17.PubMedCrossRef
21.
Zurück zum Zitat Calcia MA, Bonsall DR, Bloomfield PS, Selvaraj S, Barichello T, Howes OD. Stress and neuroinflammation: a systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology. 2016;233(9):1637–50.PubMedPubMedCentralCrossRef Calcia MA, Bonsall DR, Bloomfield PS, Selvaraj S, Barichello T, Howes OD. Stress and neuroinflammation: a systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology. 2016;233(9):1637–50.PubMedPubMedCentralCrossRef
22.
23.
Zurück zum Zitat Hershenberg R, Gros DF, Brawman-Mintzer O. Role of atypical antipsychotics in the treatment of generalized anxiety disorder. CNS Drugs. 2014;28(6):519–33.PubMedCrossRef Hershenberg R, Gros DF, Brawman-Mintzer O. Role of atypical antipsychotics in the treatment of generalized anxiety disorder. CNS Drugs. 2014;28(6):519–33.PubMedCrossRef
24.
Zurück zum Zitat Pinna G, Agis-Balboa RC, Zhubi A, Matsumoto K, Grayson DR, Costa E, et al. Imidazenil and diazepam increase locomotor activity in mice exposed to protracted social isolation. Proc Natl Acad Sci U S A. 2006;103(11):4275–80.PubMedPubMedCentralCrossRef Pinna G, Agis-Balboa RC, Zhubi A, Matsumoto K, Grayson DR, Costa E, et al. Imidazenil and diazepam increase locomotor activity in mice exposed to protracted social isolation. Proc Natl Acad Sci U S A. 2006;103(11):4275–80.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Pinna G, Costa E, Guidotti A. Fluoxetine and norfluoxetine stereospecifically facilitate pentobarbital sedation by increasing neurosteroids. Proc Natl Acad Sci U S A. 2004;101(16):6222–5.PubMedPubMedCentralCrossRef Pinna G, Costa E, Guidotti A. Fluoxetine and norfluoxetine stereospecifically facilitate pentobarbital sedation by increasing neurosteroids. Proc Natl Acad Sci U S A. 2004;101(16):6222–5.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Bachmanov AA, Reed DR, Beauchamp GK, Tordoff MG. Food intake, water intake, and drinking spout side preference of 28 mouse strains. Behav Genet. 2002;32(6):435–43.PubMedPubMedCentralCrossRef Bachmanov AA, Reed DR, Beauchamp GK, Tordoff MG. Food intake, water intake, and drinking spout side preference of 28 mouse strains. Behav Genet. 2002;32(6):435–43.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Liang J, Lopez-Valdes HE, Martinez-Coria H, Lindemeyer AK, Shen Y, Shao XM, et al. Dihydromyricetin ameliorates behavioral deficits and reverses neuropathology of transgenic mouse models of Alzheimer’s disease. Neurochem Res. 2014;39(6):1171–81.PubMedCrossRef Liang J, Lopez-Valdes HE, Martinez-Coria H, Lindemeyer AK, Shen Y, Shao XM, et al. Dihydromyricetin ameliorates behavioral deficits and reverses neuropathology of transgenic mouse models of Alzheimer’s disease. Neurochem Res. 2014;39(6):1171–81.PubMedCrossRef
29.
Zurück zum Zitat Chen K, Holschneider DP, Wu W, Rebrin I, Shih JC. A spontaneous point mutation produces monoamine oxidase A/B knock-out mice with greatly elevated monoamines and anxiety-like behavior. J Biol Chem. 2004;279(38):39645–52.PubMedCrossRef Chen K, Holschneider DP, Wu W, Rebrin I, Shih JC. A spontaneous point mutation produces monoamine oxidase A/B knock-out mice with greatly elevated monoamines and anxiety-like behavior. J Biol Chem. 2004;279(38):39645–52.PubMedCrossRef
30.
Zurück zum Zitat Young K, Morrison H. Quantifying microglia morphology from photomicrographs of immunohistochemistry prepared tissue using imageJ. J Vis Exp. 2018(136). Young K, Morrison H. Quantifying microglia morphology from photomicrographs of immunohistochemistry prepared tissue using imageJ. J Vis Exp. 2018(136).
31.
Zurück zum Zitat Dityatev AE, Bolshakov VY. Amygdala, long-term potentiation, and fear conditioning. Neuroscientist. 2005;11(1):75–88.PubMedCrossRef Dityatev AE, Bolshakov VY. Amygdala, long-term potentiation, and fear conditioning. Neuroscientist. 2005;11(1):75–88.PubMedCrossRef
32.
Zurück zum Zitat Ieraci A, Mallei A, Popoli M. Social isolation stress induces anxious-depressive-like behavior and alterations of neuroplasticity-related genes in adult male mice. Neural Plast. 2016;2016:6212983.PubMedPubMedCentralCrossRef Ieraci A, Mallei A, Popoli M. Social isolation stress induces anxious-depressive-like behavior and alterations of neuroplasticity-related genes in adult male mice. Neural Plast. 2016;2016:6212983.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Silva J, Shao AS, Shen Y, Davies DL, Olsen RW, Holschneider DP, et al. Modulation of hippocampal GABAergic neurotransmission and gephyrin levels by dihydromyricetin improves anxiety. Front Pharmacol. 2020;11:1008.PubMedPubMedCentralCrossRef Silva J, Shao AS, Shen Y, Davies DL, Olsen RW, Holschneider DP, et al. Modulation of hippocampal GABAergic neurotransmission and gephyrin levels by dihydromyricetin improves anxiety. Front Pharmacol. 2020;11:1008.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Machado P, Rostaing P, Guigonis JM, Renner M, Dumoulin A, Samson M, et al. Heat shock cognate protein 70 regulates gephyrin clustering. J Neurosci. 2011;31(1):3–14.PubMedPubMedCentralCrossRef Machado P, Rostaing P, Guigonis JM, Renner M, Dumoulin A, Samson M, et al. Heat shock cognate protein 70 regulates gephyrin clustering. J Neurosci. 2011;31(1):3–14.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Parra LA, Baust TB, Smith AD, Jaumotte JD, Zigmond MJ, Torres S, et al. The molecular chaperone Hsc70 interacts with tyrosine hydroxylase to regulate enzyme activity and synaptic vesicle localization. J Biol Chem. 2016;291(34):17510–22.PubMedPubMedCentralCrossRef Parra LA, Baust TB, Smith AD, Jaumotte JD, Zigmond MJ, Torres S, et al. The molecular chaperone Hsc70 interacts with tyrosine hydroxylase to regulate enzyme activity and synaptic vesicle localization. J Biol Chem. 2016;291(34):17510–22.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Fernández-Arjona MDM, Grondona JM, Granados-Durán P, Fernández-Llebrez P, López-Ávalos MD. Microglia morphological categorization in a rat model of neuroinflammation by hierarchical cluster and principal components analysis. Front Cell Neurosci. 2017;11:235.PubMedPubMedCentralCrossRef Fernández-Arjona MDM, Grondona JM, Granados-Durán P, Fernández-Llebrez P, López-Ávalos MD. Microglia morphological categorization in a rat model of neuroinflammation by hierarchical cluster and principal components analysis. Front Cell Neurosci. 2017;11:235.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Kinlein SA, Phillips DJ, Keller CR, Karatsoreos IN. Role of corticosterone in altered neurobehavioral responses to acute stress in a model of compromised hypothalamic-pituitary-adrenal axis function. Psychoneuroendocrinology. 2019;102:248–55.PubMedCrossRef Kinlein SA, Phillips DJ, Keller CR, Karatsoreos IN. Role of corticosterone in altered neurobehavioral responses to acute stress in a model of compromised hypothalamic-pituitary-adrenal axis function. Psychoneuroendocrinology. 2019;102:248–55.PubMedCrossRef
38.
Zurück zum Zitat Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2. Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2.
39.
40.
Zurück zum Zitat Tasan RO, Bukovac A, Peterschmitt YN, Sartori SB, Landgraf R, Singewald N, et al. Altered GABA transmission in a mouse model of increased trait anxiety. Neuroscience. 2011;183:71–80.PubMedCrossRef Tasan RO, Bukovac A, Peterschmitt YN, Sartori SB, Landgraf R, Singewald N, et al. Altered GABA transmission in a mouse model of increased trait anxiety. Neuroscience. 2011;183:71–80.PubMedCrossRef
41.
Zurück zum Zitat Crowley T, Cryan JF, Downer EJ, O’Leary OF. Inhibiting neuroinflammation: the role and therapeutic potential of GABA in neuro-immune interactions. Brain Behav Immun. 2016;54:260–77.PubMedCrossRef Crowley T, Cryan JF, Downer EJ, O’Leary OF. Inhibiting neuroinflammation: the role and therapeutic potential of GABA in neuro-immune interactions. Brain Behav Immun. 2016;54:260–77.PubMedCrossRef
43.
Zurück zum Zitat Wohleb ES, Delpech JC. Dynamic cross-talk between microglia and peripheral monocytes underlies stress-induced neuroinflammation and behavioral consequences. Prog Neuropsychopharmacol Biol Psychiatry. 2017;79(Pt A):40–8.PubMedCrossRef Wohleb ES, Delpech JC. Dynamic cross-talk between microglia and peripheral monocytes underlies stress-induced neuroinflammation and behavioral consequences. Prog Neuropsychopharmacol Biol Psychiatry. 2017;79(Pt A):40–8.PubMedCrossRef
44.
Zurück zum Zitat Wang YL, Han QQ, Gong WQ, Pan DH, Wang LZ, Hu W, et al. Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats. J Neuroinflammation. 2018;15(1):21.PubMedPubMedCentralCrossRef Wang YL, Han QQ, Gong WQ, Pan DH, Wang LZ, Hu W, et al. Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats. J Neuroinflammation. 2018;15(1):21.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Hinwood M, Tynan RJ, Charnley JL, Beynon SB, Day TA, Walker FR. Chronic stress induced remodeling of the prefrontal cortex: structural re-organization of microglia and the inhibitory effect of minocycline. Cereb Cortex. 2013;23(8):1784–97.PubMedCrossRef Hinwood M, Tynan RJ, Charnley JL, Beynon SB, Day TA, Walker FR. Chronic stress induced remodeling of the prefrontal cortex: structural re-organization of microglia and the inhibitory effect of minocycline. Cereb Cortex. 2013;23(8):1784–97.PubMedCrossRef
46.
Zurück zum Zitat Frank MG, Baratta MV, Sprunger DB, Watkins LR, Maier SF. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav Immun. 2007;21(1):47–59.PubMedCrossRef Frank MG, Baratta MV, Sprunger DB, Watkins LR, Maier SF. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav Immun. 2007;21(1):47–59.PubMedCrossRef
47.
Zurück zum Zitat Lehmann ML, Weigel TK, Poffenberger CN, Herkenham M. The behavioral sequelae of social defeat require microglia and are driven by oxidative stress in mice. J Neurosci. 2019;39(28):5594–605.PubMedPubMedCentralCrossRef Lehmann ML, Weigel TK, Poffenberger CN, Herkenham M. The behavioral sequelae of social defeat require microglia and are driven by oxidative stress in mice. J Neurosci. 2019;39(28):5594–605.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Tong L, Gong Y, Wang P, Hu W, Wang J, Chen Z, et al. Microglia loss contributes to the development of major depression induced by different types of chronic stresses. Neurochem Res. 2017;42(10):2698–711.PubMedCrossRef Tong L, Gong Y, Wang P, Hu W, Wang J, Chen Z, et al. Microglia loss contributes to the development of major depression induced by different types of chronic stresses. Neurochem Res. 2017;42(10):2698–711.PubMedCrossRef
49.
Zurück zum Zitat Kreisel T, Frank MG, Licht T, Reshef R, Ben-Menachem-Zidon O, Baratta MV, et al. Dynamic microglial alterations underlie stress-induced depressive-like behavior and suppressed neurogenesis. Mol Psychiatry. 2014;19(6):699–709.PubMedCrossRef Kreisel T, Frank MG, Licht T, Reshef R, Ben-Menachem-Zidon O, Baratta MV, et al. Dynamic microglial alterations underlie stress-induced depressive-like behavior and suppressed neurogenesis. Mol Psychiatry. 2014;19(6):699–709.PubMedCrossRef
50.
Zurück zum Zitat Gong Y, Tong L, Yang R, Hu W, Xu X, Wang W, et al. Dynamic changes in hippocampal microglia contribute to depressive-like behavior induced by early social isolation. Neuropharmacology. 2018;135:223–33.PubMedCrossRef Gong Y, Tong L, Yang R, Hu W, Xu X, Wang W, et al. Dynamic changes in hippocampal microglia contribute to depressive-like behavior induced by early social isolation. Neuropharmacology. 2018;135:223–33.PubMedCrossRef
51.
52.
Zurück zum Zitat Stein DJ, Vasconcelos MF, Albrechet-Souza L, Ceresér KMM, de Almeida RMM. Microglial over-activation by social defeat stress contributes to anxiety- and depressive-like behaviors. Front Behav Neurosci. 2017;11:207.PubMedPubMedCentralCrossRef Stein DJ, Vasconcelos MF, Albrechet-Souza L, Ceresér KMM, de Almeida RMM. Microglial over-activation by social defeat stress contributes to anxiety- and depressive-like behaviors. Front Behav Neurosci. 2017;11:207.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Hinwood M, Morandini J, Day TA, Walker FR. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb Cortex. 2012;22(6):1442–54.PubMedCrossRef Hinwood M, Morandini J, Day TA, Walker FR. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb Cortex. 2012;22(6):1442–54.PubMedCrossRef
54.
Zurück zum Zitat Tynan RJ, Naicker S, Hinwood M, Nalivaiko E, Buller KM, Pow DV, et al. Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain Behav Immun. 2010;24(7):1058–68.PubMedCrossRef Tynan RJ, Naicker S, Hinwood M, Nalivaiko E, Buller KM, Pow DV, et al. Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain Behav Immun. 2010;24(7):1058–68.PubMedCrossRef
55.
Zurück zum Zitat Fernández-Arjona MDM, Grondona JM, Fernández-Llebrez P, López-Ávalos MD. Microglial morphometric parameters correlate with the expression level of IL-1β, and allow identifying different activated morphotypes. Front Cell Neurosci. 2019;13:472.PubMedPubMedCentralCrossRef Fernández-Arjona MDM, Grondona JM, Fernández-Llebrez P, López-Ávalos MD. Microglial morphometric parameters correlate with the expression level of IL-1β, and allow identifying different activated morphotypes. Front Cell Neurosci. 2019;13:472.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Du Preez A, Law T, Onorato D, Lim YM, Eiben P, Musaelyan K, et al. The type of stress matters: repeated injection and permanent social isolation stress in male mice have a differential effect on anxiety- and depressive-like behaviours, and associated biological alterations. Transl Psychiatry. 2020;10(1):325.PubMedPubMedCentralCrossRef Du Preez A, Law T, Onorato D, Lim YM, Eiben P, Musaelyan K, et al. The type of stress matters: repeated injection and permanent social isolation stress in male mice have a differential effect on anxiety- and depressive-like behaviours, and associated biological alterations. Transl Psychiatry. 2020;10(1):325.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Haj-Mirzaian A, Khosravi A, Rahbar A, Ramezanzadeh K, Nikbakhsh R, Pirri F, et al. The potential role of very small embryonic-like stem cells in the neuroinflammation induced by social isolation stress: introduction of a new paradigm. Brain Res Bull. 2020;163:21–30.PubMedCrossRef Haj-Mirzaian A, Khosravi A, Rahbar A, Ramezanzadeh K, Nikbakhsh R, Pirri F, et al. The potential role of very small embryonic-like stem cells in the neuroinflammation induced by social isolation stress: introduction of a new paradigm. Brain Res Bull. 2020;163:21–30.PubMedCrossRef
58.
Zurück zum Zitat Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74(4):691–705.PubMedPubMedCentralCrossRef Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74(4):691–705.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53(3):337–51.PubMedCrossRef Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53(3):337–51.PubMedCrossRef
61.
Zurück zum Zitat Liu HY, Yue J, Hu LN, Cheng LF, Wang XS, Wang XJ, et al. Chronic minocycline treatment reduces the anxiety-like behaviors induced by repeated restraint stress through modulating neuroinflammation. Brain Res Bull. 2018;143:19–26.PubMedCrossRef Liu HY, Yue J, Hu LN, Cheng LF, Wang XS, Wang XJ, et al. Chronic minocycline treatment reduces the anxiety-like behaviors induced by repeated restraint stress through modulating neuroinflammation. Brain Res Bull. 2018;143:19–26.PubMedCrossRef
62.
Zurück zum Zitat Hannibal KE, Bishop MD. Chronic stress, cortisol dysfunction, and pain: a psychoneuroendocrine rationale for stress management in pain rehabilitation. Phys Ther. 2014;94(12):1816–25.PubMedPubMedCentralCrossRef Hannibal KE, Bishop MD. Chronic stress, cortisol dysfunction, and pain: a psychoneuroendocrine rationale for stress management in pain rehabilitation. Phys Ther. 2014;94(12):1816–25.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Chrousos GP. The role of stress and the hypothalamic-pituitary-adrenal axis in the pathogenesis of the metabolic syndrome: neuro-endocrine and target tissue-related causes. Int J Obes Relat Metab Disord. 2000;24(Suppl 2):S50–5.PubMedCrossRef Chrousos GP. The role of stress and the hypothalamic-pituitary-adrenal axis in the pathogenesis of the metabolic syndrome: neuro-endocrine and target tissue-related causes. Int J Obes Relat Metab Disord. 2000;24(Suppl 2):S50–5.PubMedCrossRef
64.
Zurück zum Zitat Serra M, Pisu MG, Floris I, Biggio G. Social isolation-induced changes in the hypothalamic-pituitary-adrenal axis in the rat. Stress. 2005;8(4):259–64.PubMedCrossRef Serra M, Pisu MG, Floris I, Biggio G. Social isolation-induced changes in the hypothalamic-pituitary-adrenal axis in the rat. Stress. 2005;8(4):259–64.PubMedCrossRef
65.
Zurück zum Zitat Koo JW, Russo SJ, Ferguson D, Nestler EJ, Duman RS. Nuclear factor-kappaB is a critical mediator of stress-impaired neurogenesis and depressive behavior. Proc Natl Acad Sci U S A. 2010;107(6):2669–74.PubMedPubMedCentralCrossRef Koo JW, Russo SJ, Ferguson D, Nestler EJ, Duman RS. Nuclear factor-kappaB is a critical mediator of stress-impaired neurogenesis and depressive behavior. Proc Natl Acad Sci U S A. 2010;107(6):2669–74.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Zlatković J, Bernardi RE, Filipović D. Protective effect of Hsp70i against chronic social isolation stress in the rat hippocampus. J Neural Transm (Vienna). 2014;121(1):3–14.CrossRef Zlatković J, Bernardi RE, Filipović D. Protective effect of Hsp70i against chronic social isolation stress in the rat hippocampus. J Neural Transm (Vienna). 2014;121(1):3–14.CrossRef
67.
Zurück zum Zitat Jing N, Li X. Dihydromyricetin attenuates inflammation through TLR4/NF-kappaB Pathway. Open Med (Wars). 2019;14:719–25.CrossRef Jing N, Li X. Dihydromyricetin attenuates inflammation through TLR4/NF-kappaB Pathway. Open Med (Wars). 2019;14:719–25.CrossRef
Metadaten
Titel
Social isolation induces neuroinflammation and microglia overactivation, while dihydromyricetin prevents and improves them
verfasst von
Alzahra J. Al Omran
Amy S. Shao
Saki Watanabe
Zeyu Zhang
Jifeng Zhang
Chen Xue
Junji Watanabe
Daryl L. Davies
Xuesi M. Shao
Jing Liang
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/s12974-021-02368-9

Weitere Artikel der Ausgabe 1/2022

Journal of Neuroinflammation 1/2022 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Fehlerkultur in der Medizin – Offenheit zählt!

28.05.2024 Fehlerkultur Podcast

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.