Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Eur J Pharmacol ; 978: 176763, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38906239

ABSTRACT

Depression triggered by harmful stress during adolescence is a common problem that can affect mental health. To date, the mechanisms underlying this type of depression remain unclear. One mechanism for the promotion of depression by chronic stress in adulthood is the loss of hippocampal microglia. Since deleterious stress in adolescence also activates microglia, we investigated the dynamic changes of microglia in the hippocampus in mice exposed to chronic unpredictable stress (CUS) in adolescence. Our results showed that 12 days of CUS stimulation in adolescence induced typical depression-like behaviors in adult mice, which were accompanied by a significant decrease and dystrophy of microglia in the dentate gyrus of the hippocampus. Further analysis showed that this decrease in microglia was mediated by the initial response of microglia to unpredictable stress in the dentate gyrus of the hippocampus and their subsequent apoptosis. Blocking the initial response of microglia to unpredictable stress by pretreatment with minocycline was able to prevent apoptosis and microglial decline as well as the development of depression-like behaviors in adult mice induced by adolescent CUS. Moreover, administration of lipopolysaccharide (LPS) or macrophage-colony stimulatory factor (M-CSF), two drugs that reversed microglia decline in the dentate gyrus, ameliorated the depression-like behaviors induced by CUS stimulation in adolescence. These findings reveal a novel mechanism for the development of depression-like behaviors in animals triggered by deleterious stress in adolescence and suggest that reversing microglial decline in the hippocampus may be a hopeful strategy for the treatment of depression triggered by deleterious stress in adolescence.


Subject(s)
Apoptosis , Behavior, Animal , Depression , Hippocampus , Microglia , Stress, Psychological , Animals , Microglia/drug effects , Microglia/pathology , Stress, Psychological/complications , Stress, Psychological/psychology , Apoptosis/drug effects , Mice , Male , Hippocampus/drug effects , Hippocampus/pathology , Behavior, Animal/drug effects , Minocycline/pharmacology , Mice, Inbred C57BL , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Disease Models, Animal , Age Factors , Lipopolysaccharides/pharmacology
2.
Proc Natl Acad Sci U S A ; 120(52): e2307477120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38134195

ABSTRACT

Potassium-ion batteries (PIBs) have attracted ever-increasing interest due to the abundant potassium resources and low cost, which are considered a sustainable energy storage technology. However, the graphite anodes employed in PIBs suffer from low capacity and sluggish reaction kinetics caused by the large radius of potassium ions. Herein, we report nitrogen-doped, defect-rich hollow carbon nanospheres with contact curved interfaces (CCIs) on carbon nanotubes (CNTs), namely CCI-CNS/CNT, to boost both electron transfer and potassium-ion adsorption. Density functional theory calculations validate that engineering CCIs significantly augments the electronic state near the Fermi level, thus promoting electron transfer. In addition, the CCIs exhibit a pronounced affinity for potassium ions, promoting their adsorption and subsequently benefiting potassium storage. As a result, the rationally designed CCI-CNS/CNT anode shows remarkable cyclic stability and rate capability. This work provides a strategy for enhancing the potassium storage performance of carbonaceous materials through CCI engineering, which can be further extended to other battery systems.

3.
Brain Behav Immun ; 91: 451-471, 2021 01.
Article in English | MEDLINE | ID: mdl-33157258

ABSTRACT

Over-activation of the innate immune system constitutes a risk factor for the development of nervous system disorders but may reduce the severity of these disorders by inducing tolerance effect. Here, we studied the tolerance-inducing effect and properties of innate immune stimulation on chronic social defeat stress (CSDS)-induced behavioral abnormalities in mice. A single injection of the innate immune enhancer lipopolysaccharide (LPS) one day before stress exposure prevented CSDS-induced impairment in social interaction and increased immobility time in the tail suspension test and forced swimming test. This effect was observed at varying doses (100, 500, and 1000 µg/kg) and peaked at 100 µg/kg. A single LPS injection (100 µg/kg) either one or five but not ten days before stress exposure prevented CSDS-induced behavioral abnormalities. A second LPS injection ten days after the first LPS injection, or a 2 × or 4 × LPS injections ten days before stress exposure also induced tolerance against stress-induced behavioral abnormalities. Our results furthermore showed that a single LPS injection one day before stress exposure skewed the neuroinflammatory response in the hippocampus and prefrontal cortex of CSDS-exposed mice toward an anti-inflammatory phenotype. Inhibiting the central innate immune response by pretreatment with minocycline or PLX3397 abrogated the tolerance-inducing effect of LPS preconditioning on CSDS-induced behavioral abnormalities and neuroinflammatory responses in the brain. These results provide evidence for a prophylactic effect of innate immune stimulation on stress-induced behavioral abnormalities via changes in microglial activation, which may help develop novel strategies for the prevention of stress-induced psychological disorders.


Subject(s)
Hippocampus , Lipopolysaccharides , Animals , Depression , Immunity, Innate , Inflammation , Mice , Minocycline
4.
Neuropharmacology ; 172: 108132, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32407925

ABSTRACT

Previous studies have reported that macrophage-colony stimulating factor (M-CSF), a drug that is used to treat hematological system disease, can ameliorate chronic stress-induced depressive-like behaviors in mice. This indicates that M-CSF could be developed into a novel antidepressant. Here, we investigated the antidepressive properties of M-CSF, aiming to explore its potential values in depression treatment. Our results showed that a single M-CSF injection at the dose of 75 and 100 µg/kg, but not at 25 or 50 µg/kg, ameliorated chronic unpredictable stress (CUS)-induced depressive-like behaviors in mice at 5 h after the drug treatment. In a time-dependent experiment, a single M-CSF injection (100 µg/kg) was found to ameliorate the CUS-induced depressive-like behaviors in mice at 5 and 8 h, but not at 3 h, after the drug treatment. The antidepressant effect of the single M-CSF injection (100 µg/kg) in chronically-stressed mice persisted at least 10 days and disappeared at 14 days after the drug treatment. Moreover, 14 days after the first injection, a second M-CSF injection (100 µg/kg) still produced antidepressant effects at 5 h after the drug treatment in chronically-stressed mice who re-displayed depressive-like phenotypes. The antidepressant effect of M-CSF appeared to be mediated by the activation of the hippocampal microglia, as pre-inhibition of microglia by minocycline (40 mg/kg) or PLX3397 (290 mg/kg) pretreatment prevented the antidepressant effect of M-CSF in CUS mice. These results demonstrate that M-CSF produces rapid and sustained antidepressant effects via the activation of the microglia in the hippocampus in a dose- and time-dependent manner.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Macrophage Colony-Stimulating Factor/pharmacology , Stress, Psychological/drug therapy , Aminopyridines/pharmacology , Animals , Depression/etiology , Depression/psychology , Dose-Response Relationship, Drug , Hippocampus/drug effects , Macrophage Activation/drug effects , Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Minocycline/pharmacology , Pyrroles/pharmacology , Stress, Psychological/complications , Stress, Psychological/psychology
5.
Int J Neuropsychopharmacol ; 23(10): 673-686, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32453814

ABSTRACT

BACKGROUND: We recently identified neuronal expression of farnesoid X receptor (FXR), a bile acid receptor known to impair autophagy by inhibiting cyclic adenosine monophosphate response element-binding protein (CREB), a protein whose underfunctioning is linked to neuroplasticity and depression. In this study, we hypothesize that FXR may mediate depression via a CREB-dependent mechanism. METHODS: Depression was induced in male C57BL6/J mice via chronic unpredictable stress (CUS). Subjects underwent behavioral testing to identify depression-like behaviors. A variety of molecular biology techniques, including viral-mediated gene transfer, Western blot, co-immunoprecipitation, and immunofluorescence, were used to correlate depression-like behaviors with underlying molecular and physiological events. RESULTS: Overexpression of FXR, whose levels were upregulated by CUS in hippocampal CA1, induced or aggravated depression-like behaviors in stress-naïve and CUS-exposed mice, while FXR short hairpin RNA (shRNA) ameliorated such symptoms in CUS-exposed mice. The behavioral effects of FXR were found to be associated with changes in CREB-brain-derived neurotrophic factor (BDNF) signaling, as FXR overexpression aggravated CUS-induced reduction in BDNF levels while the use of FXR shRNA or disruption of FXR-CREB signaling reversed the CUS-induced reduction in the phosphorylated CREB and BDNF levels. Molecular analysis revealed that FXR shRNA prevented CUS-induced cytoplasmic translocation of CREB-regulated transcription coactivator 2 (CRTC2); CRTC2 overexpression and CRTC2 shRNA abrogated the regulatory effect of FXR overexpression or FXR shRNA on CUS-induced depression-like behaviors. CONCLUSIONS: In stress conditions, increased FXR in the CA1 inhibits CREB by targeting CREB and driving the cytoplasmic translocation of CRTC2. Uncoupling of the FXR-CREB complex may be a novel strategy for depression treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Depression/metabolism , Depression/physiopathology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Behavior, Animal/physiology , Cytoplasm/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Protein Transport/physiology
6.
J Nutr Biochem ; 78: 108323, 2020 04.
Article in English | MEDLINE | ID: mdl-32135404

ABSTRACT

Microglia are the innate immune cells in the nervous system. In the resting state, they display a ramified morphology, while upon disease stimulation their processes would be retracted, along with pro-inflammatory cytokine overproduction. Reversing microglial process retraction may help reduce pro-inflammatory cytokine production and restore microglia's ability to scan surrounding environments, rendering brain function regulation to be more effective. We found that diallyl disulfide (DADS), a major organosulfur compound in garlic oil, administered at different doses and time points, promoted microglial process elongation in both cultured systems and prefrontal cortexes in mice in a reversible manner. Lipopolysaccharide (LPS), a classical activator of microglia, did not affect this pro-elongation effect of DADS at conditions in vitro and in vivo. Functional studies revealed that DADS pre-treatment attenuated LPS-induced decreases in levels of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) mRNA as well as LPS-induced increases in levels of IL-10 and CD206 mRNA in both cultured microglia and prefrontal cortexes in mice. Protein kinase B (Akt) inhibition attenuated the pro-elongation effect of DADS on microglial process and blocked the regulatory effects of DADS on LPS-induced inflammatory responses in both cultured microglia and prefrontal cortexes in mice. In an in vivo model of neuroinflammation, DADS pre-treatment prevented LPS-induced retraction of microglial process in the prefrontal cortex in mice and attenuated LPS-induced increase in immobility time in the tail suspension test and forced swim test. These results indicate that DADS induces an Akt-dependent elongation of microglia process, along with the induction of an anti-inflammatory phenotype.


Subject(s)
Allyl Compounds/pharmacology , Disulfides/pharmacology , Garlic/chemistry , Microglia/drug effects , Microglia/metabolism , Plant Oils/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Behavior, Animal , Cytokines/metabolism , Immunity, Innate , Inflammation , Interleukin-10/metabolism , Lectins, C-Type/metabolism , Lipopolysaccharides/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Neuropharmacology ; 170: 108054, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32217088

ABSTRACT

Depression-alcohol addiction comorbidity is a common clinical phenomenon. Alcohol exposure in adolescence has been shown to induce depression-like behaviors in rodents. However, the mechanism of action for this type of depression remains unclear. Previous studies have reported that several different types of stress, such as chronic unpredictable stress and early social isolation, trigger depression-like symptoms in mice by inducing hippocampal microglial decline, which is mediated by the initial activation of the microglial cells. Since alcohol also activates microglia, we evaluated the dynamic changes in hippocampal microglia in mice receiving adolescent intermittent alcohol exposure (AIE). Our results showed that 14 days of AIE, followed by 21 days period of no treatment, induced behavioral abnormalities as well as a significant loss and dystrophy of hippocampal microglia in mice. We found that this AIE-induced decline in hippocampal microglia was mediated by both microglial activation and apoptosis, as (i) 1 day of alcohol exposure induced a distinct activation of hippocampal microglia followed by their apoptosis, and (ii) blocking the initial activation of hippocampal microglia by pretreatment with minocycline suppressed the AIE-induced apoptosis and loss of hippocampal microglia as well as the AIE-induced depression-like symptoms. Lipopolysaccharide (LPS), a classical activator of microglia, ameliorated the AIE-induced depression-like symptoms by reversing the decline in the hippocampal microglia. These results reveal a possible mechanism for AIE-induced depression and demonstrate that the restoration of hippocampal microglial homeostasis may be a therapeutic strategy for depression induced by alcohol intake and withdrawal.


Subject(s)
Alcohol Drinking/adverse effects , Apoptosis/drug effects , Depression/chemically induced , Ethanol/toxicity , Hippocampus/drug effects , Microglia/drug effects , Age Factors , Alcohol Drinking/pathology , Alcohol Drinking/psychology , Animals , Apoptosis/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Depression/pathology , Depression/psychology , Ethanol/administration & dosage , Hippocampus/pathology , Male , Mice , Microglia/pathology , Minocycline/pharmacology
8.
Article in English | MEDLINE | ID: mdl-32201112

ABSTRACT

The decrease of microglia in the hippocampus is a novel mechanism for depression onset. Reversal of this decrease can ameliorate stress-induced depression-like behaviors in rodents. However, the property of this therapeutic strategy remains unclear. We addressed this issue by designing a series of behavioral experiments. Results showed that a single lipopolysaccharide (LPS) injection at the dose of 75 and 100 µg/kg, but not at 30 or 50 µg/kg, produced obvious antidepressant effects in chronic unpredictable stress (CUS) mice at 5 h after the drug administration. In the time-dependent experiment, a single LPS injection (100 µg/kg) ameliorated the CUS-induced depression-like behaviors in mice at 5 and 8 h, but not at 3 h, after the drug administration. The antidepressant effect of a single LPS injection persisted at least 10 days and disappeared at 14 days after the drug administration. 14 days after the first injection, a second LPS injection (100 µg/kg) still produced antidepressant effects in chronically-stressed mice who re-displayed depression-like behaviors at 5 h after the drug administration. The antidepressant effect of LPS appears to be dependent on microglia, as at 5 h after LPS administration (100 µg/kg), the CUS-induced decrease in microglial numbers and Iba-1 mRNA levels in the hippocampus was reversed markedly, and inhibition of microglia by minocycline (40 mg/kg) or PLX33297 (290 mg/kg) prevented the antidepressant effect of LPS in CUS mice. These results indicate that a single LPS injection displays rapid and sustained antidepressant effects in chronically stressed mice likely through stimulating hippocampal microglia.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/metabolism , Disease Models, Animal , Microglia/metabolism , Stress, Psychological/metabolism , Animals , Antidepressive Agents/pharmacology , Depression/drug therapy , Depression/pathology , Dose-Response Relationship, Drug , Immobilization/methods , Immobilization/psychology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/therapeutic use , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Minocycline/pharmacology , Stress, Psychological/drug therapy , Stress, Psychological/pathology
9.
Pharm Biol ; 58(1): 219-224, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32202179

ABSTRACT

Context: Salvia miltiorrhiza Bge. (Labiatae) (SMB) is applied clinically for management of diabetic osteoporosis in China, and research results has suggested its potential action on renin-angiotensin system (RAS).Objective: This study screens and explores naturally occurring bioactive constituents from the root of SMB acting on renin activity and evaluates its osteoprotective efficacy in diabetic mice.Materials and methods: Human embryonic kidney (HEK) 293 cells, engineered to express human renin, were used as an in vitro model to identify bioactive compound, tanshinone IIA, inhibiting renin activity. The C57BL/6 mice (n = 10 in each group) with diabetes induced by streptozotocin (STZ) were intraperitoneally injected with tanshinone IIA (10 and 30 mg/kg). The mice without STZ treatment and the diabetic mice treated with aliskiren were used as non-diabetic control and positive control, respectively.Results: Tanshinone IIA was found to display inhibitory effects on renin activity of HEK-293 cells; moreover, it down-regulated protein expression of ANG II in human renin-expressed HEK-293 cells. Treatment of diabetic mice with tanshinone IIA with both doses could significantly decrease ANG II level in serum (from 16.56 ± 1.70 to 10.86 ± 0.68 and 9.14 ± 1.31 pg/mL) and reduce ANG II expression in bone, consequently improving trabecular bone mineral density and micro-structure of proximal tibial end and increasing trabecular bone area of distal femoral end in diabetic mice.Conclusions: This study revealed beneficial effects of tanshinone IIA on bone of diabetic mice, and potentially suggested the application of Salvia miltiorrhiza in the treatment of osteoporosis and drug development of tanshinone IIA as a renin inhibitor.


Subject(s)
Abietanes/pharmacology , Anti-Inflammatory Agents/pharmacology , Diabetes Mellitus, Experimental/complications , Osteoporosis/prevention & control , Renin-Angiotensin System/drug effects , Renin/metabolism , Abietanes/isolation & purification , Angiotensin II/blood , Animals , Anti-Inflammatory Agents/isolation & purification , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Osteoporosis/etiology , Osteoporosis/metabolism , Plant Roots/chemistry , Renin/genetics , Salvia miltiorrhiza/chemistry , Transfection
10.
Pharmacology ; 105(9-10): 550-560, 2020.
Article in English | MEDLINE | ID: mdl-32203973

ABSTRACT

Depression is a common disease that afflicts one in 6 people. Numerous hypotheses have been raised in the past decades, but the exact mechanism for depression onset remains obscure. Recently, the neuroinflammatory response and oxidative stress are being attracted more and more attention due to their roles in depression pathogenesis. The inhibition of neuroinflammatory response and oxidative stress is now considered a potential strategy for depression prevention and/or therapy. Sodium butyrate (SB) is a sodium form of the endogenous butyrate. It can inhibit proinflammatory responses and oxidative stress in different models of disease. In the present study, we investigated the effect of SB on lipopolysaccharide (LPS)-induced depression-like behaviors, neuroinflammatory response, and oxido-nitrosative stress in the hippocampus and prefrontal cortex in C57BL6/J mice. Our results showed that 10 days of SB pretreatment at the dose of 300 but not 100 mg/kg markedly ameliorated LPS (0.83 mg/kg)-induced depression-like behaviors in the tail suspension test, forced swimming test, and sucrose preference test. Further analysis showed that 10 days of SB pretreatment not only prevented LPS-induced increases in proinflammatory cytokines, such as interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α, in the hippocampus and prefrontal cortex but also prevented LPS-induced enhancement of oxido-nitrosative stress. Taken together, these results demonstrate that SB is such an agent that could be used to prevent depression onset and/or progression, and inhibition of neuroinflammatory response and oxido-nitrosative stress may be a potential mechanism for its antidepressant actions.


Subject(s)
Antidepressive Agents/pharmacology , Butyric Acid/pharmacology , Depression/prevention & control , Inflammation/metabolism , Inflammation/prevention & control , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Animals , Antidepressive Agents/administration & dosage , Behavior, Animal/drug effects , Butyric Acid/administration & dosage , Cytokines/drug effects , Cytokines/metabolism , Depression/chemically induced , Disease Models, Animal , Hindlimb Suspension , Hippocampus/drug effects , Hippocampus/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Physical Conditioning, Animal , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Swimming
11.
Behav Pharmacol ; 31(4): 322-332, 2020 06.
Article in English | MEDLINE | ID: mdl-31895061

ABSTRACT

ß-hydroxybutyrate, a ketone body metabolite, has been shown to suppress depression-like behavior in rodents. In this study, we examined its antidepressive property in acute and chronic administration modes in mice by using forced swim test and tail suspension test. Results showed that the decrease effect of ß-hydroxybutyrate (300 mg/kg) on immobility time in the tail suspension test and forced swim test in stress-naive mice began to be significant at day 11. In a dose-dependent experiment, ß-hydroxybutyrate treatment (11 days) showed significant antidepressant activities at the dose of 200 and 300 mg/kg. Unlike fluoxetine, ß-hydroxybutyrate treatment (300 mg/kg) showed no antidepressant activities in the acute (1 hour before the test) and three times administration mode within 24 hours (1, 5, and 24 hours before the test). But in a co-administration mode, ß-hydroxybutyrate (100 mg/kg) -fluoxetine (2.5 mg/kg) co-administration exhibited an obvious antidepressant activity in the tail suspension test and forced swim test. Further analysis showed that the antidepressant effects of ß-hydroxybutyrate and fluoxetine were not associated with the change in mouse locomotor activity. Furthermore, both chronic ß-hydroxybutyrate treatment and ß-hydroxybutyrate-fluoxetine co-treatment suppressed chronic unpredictable stress-induced increase in immobility time in the tail suspension test and forced swim test as well as chronic unpredictable stress-induced decrease in mouse body weight. Taken together, these results indicate that ß-hydroxybutyrate (1) needs a relatively long time to show comparable behavioral activity to that of fluoxetine in assays that are sensitive to the behavioral effects of established antidepressant compounds and (2) can augment the antidepressant action of a sub-therapeutic dose of fluoxetine.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Immobility Response, Tonic/drug effects , Animals , Antidepressive Agents/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Fluoxetine/pharmacology , Hindlimb Suspension , Male , Mice , Motor Activity/drug effects
12.
Eur J Pharmacol ; 858: 172500, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31238064

ABSTRACT

Anthocyanins, a class of water soluble flavonoids extracted from plants like berries and soybean seed, have been shown to display obvious anti-oxidative, anti-inflammatory, and anti-apoptotic activities. They are recommended as a supplementation for prevention and/or treatment of disorders ranging from cardiovascular disease, metabolic syndrome, and cancer. In the central nervous system (CNS), anthocyanins and its major component cyanidin-3-O-glucoside (C3G) have been reported to produce preventive and/or therapeutic activities in a wide range of disorders, such as cerebral ischemia, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and glioblastoma. Both anthocyanins and C3G can also affect some important processes in aging, including neuronal apoptosis and death as well as learning and memory impairment. Further, the anthocyanins and C3G have been shown to prevent neuro-toxicities induced by different toxic factors, such as lipopolysaccharide, hydrogen peroxide, ethanol, kainic acid, acrolein, glutamate, and scopolamine. Mechanistic studies have shown that inhibition of oxidative stress and neuroinflammation are two critical mechanisms by which anthocyanins and C3G produce protective effects in CNS disorder prevention and/or treatment. Other mechanisms, including suppression of c-Jun N-terminal kinase (JNK) activation, amelioration of cellular degeneration, activation of the brain-derived neurotrophic factor (BDNF) signaling, and restoration of Ca2+ and Zn2+ homeostasis, may also mediate the neuroprotective effects of anthocyanins and C3G. In this review, we summarize the pharmacological effects of anthocyanins and C3G in CNS disorders as well as their possible mechanisms, aiming to get a clear insight into the role of anthocyanins in the CNS.


Subject(s)
Anthocyanins/pharmacology , Central Nervous System/drug effects , Glucosides/pharmacology , Neuroprotective Agents/pharmacology , Animals , Anthocyanins/therapeutic use , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Central Nervous System Diseases/prevention & control , Glucosides/therapeutic use , Humans , Neuroprotective Agents/therapeutic use
13.
Phytother Res ; 33(2): 442-451, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30456807

ABSTRACT

Tetrahydroxy stilbene glucoside (TSG), an active component from medicinal herb Polygonum multiflorum Thunb, could block the activity of the tissue renin-angiotensin system (RAS), which plays a critical role in development of diabetic osteoporosis. This study aimed to determine if TSG therapy could alleviate bone deteriorations in diabetic mouse model induced by streptozotocin. The diabetic mice showed the loss of trabecular bone mass and the changes of trabecular bone microarchitectural parameters as well as the increase in amount of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts at the distal metaphysis of femur when compared with those of nondiabetic mice. Treatment with TSG significantly elevated calcium content in serum and bone and improved biological parameters of trabecular bone, accompanied by increasing messenger RNA (mRNA) expression of RUNX-2, COL-I, and OCN and protein expression of ß-catenin as well as down-regulating protein expression of RAS components including renin and AT1R. In addition, TSG repressed diabetes-induced decrease in ratio of OPG/RANKL expression and increase in sclerostin expression in bone. The similar effects of TSG on osteoblasts-specific genes were found in MC3T3-E1 cells. Taken together, the present study demonstrated the osteopreserve effects of TSG in diabetic mice, and the underlying mechanism might be attributed to its regulation on osteogenesis and osteoclastogenesis.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Glucosides/pharmacology , Osteoporosis/prevention & control , Stilbenes/pharmacology , Animals , Bone and Bones/drug effects , Diabetes Mellitus, Experimental/complications , Femur/drug effects , Hyperglycemia/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Renin-Angiotensin System/drug effects , Streptozocin , Tartrate-Resistant Acid Phosphatase/metabolism
14.
Neuropharmacology ; 135: 223-233, 2018 06.
Article in English | MEDLINE | ID: mdl-29574097

ABSTRACT

Depression triggered by early-life stress has begun to attract wide attention due to its severe symptoms and poor treatment outcomes. However, the pathophysiological mechanism for this type of depression remains unclear. Recently, we and others reported that different types of chronic stress induce a significant loss of hippocampal microglia, which is mediated by an initial activation of these microglia. Since early-life stress also promotes microglial activation, we investigated the dynamic changes in hippocampal microglia in mice suffering from depression induced by early social isolation (ESI). Results showed that 8 days of ESI induced depressive-like behaviors in a tail suspension test, forced swim test, sucrose preference test, and open field test, and it also induced a loss and dystrophy of hippocampal microglia. We found that this ESI-induced loss of hippocampal microglia was mediated by both microglial activation and apoptosis. This was demonstrated by the following results: (i) 1 day of ESI induced an obvious activation of hippocampal microglia followed by their apoptosis, and (ii) the blockade of the initial activation of hippocampal microglia by minocycline pretreatment suppressed the ESI-induced apoptosis and loss as well as ESI-induced depressive-like behavior. Lipopolysaccharide (LPS) and macrophage colony-stimulating factor (M-CSF), two activators of microglia, almost completely reversed ESI-induced depressive-like behavior by promoting microglial proliferation in the hippocampus. These results reveal an etiological role of hippocampal microglial loss in ESI-induced depression and demonstrate that the restoration of microglial homeostasis in the hippocampus may serve as a therapeutic strategy for depression induced by early-life stress.


Subject(s)
Depression/pathology , Hippocampus/pathology , Microglia/pathology , Social Isolation/psychology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Depression/prevention & control , Depression/psychology , Female , Immobility Response, Tonic , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Microglia/drug effects , Minocycline/pharmacology
15.
Prog Neuropsychopharmacol Biol Psychiatry ; 79(Pt B): 258-267, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28625859

ABSTRACT

Depression is a world-wide disease with no effective therapeutic methods. Increasing evidence indicates that astrocytic pathology contributes to the formation of depression. In this study, we investigated the effects of harmine, a natural ß-carboline alkaloid and potent hallucinogen, known to modulate astrocytic glutamate transporters, on chronic unpredictable stress (CUS)-induced depressive-like behaviors and astrocytic dysfunctions. Results showed that harmine treatment (10, 20mg/kg) protected the mice against the CUS-induced increases in the immobile time in the tail suspension test (TST) and forced swimming test (FST), and also reversed the reduction in sucrose intake in the sucrose preference experiment. Harmine treatment (20mg/kg) prevented the reductions in brain-derived neurotrophic factor (BDNF) protein levels and hippocampal neurogenesis induced by CUS. In addition, harmine treatment (20mg/kg) increased the protein expression levels of glutamate transporter 1 (GLT-1) and prevented the CUS-induced decreases in glial fibrillary acidic protein (GFAP) protein expressions in the prefrontal cortex and hippocampus, suggesting that restoration of astrocytic functions may be a potential mechanism underlying the antidepressant-like effects of harmine. This opinion was proved by the results that administration of mice with l-Alpha-Aminoadipic Acid (L-AAA), a gliotoxin specific for astrocytes, attenuated the antidepressant-like effects of harmine, and prevented the improvement effects of harmine on BDNF protein levels and hippocampal neurogenesis. These results provide further evidence to confirm that astrocytic dysfunction contributes critically to the development of depression and that harmine exerts antidepressant-like effects likely through restoration of astrocytic functions.


Subject(s)
Antidepressive Agents/pharmacology , Astrocytes/drug effects , Depressive Disorder/drug therapy , Harmine/pharmacology , Anhedonia/drug effects , Anhedonia/physiology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain-Derived Neurotrophic Factor/metabolism , Chronic Disease , Depressive Disorder/metabolism , Depressive Disorder/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 2/metabolism , Fluoxetine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Inbred C57BL , Neurogenesis/drug effects , Neurogenesis/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Stress, Psychological/pathology , Uncertainty
16.
Eur J Pharmacol ; 811: 21-29, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28545778

ABSTRACT

2,3,4',5-tetrahydroxystilbene 2-O-ß-D-glucoside (TSG), a resveratrol analog with glucoside, is purified from a traditional Chinese herbal medicine polygonum multiflorum. It has been extensively studied in last decade and known to exert strong anti-inflammatory, anti-oxidative, anti-apoptotic, and free radical scavenging activities, and therefore has been listed as a potential agent for disease therapies. Recent studies extend well-beyond effects of TSG on the injury of neurons, cardiomyocytes and endothelial cells, and report important functions of TSG in a lot of pathophysiological conditions. For example, TSG has been shown to prevent the production of pro-inflammatory cytokines in microglia and macrophages in vitro, and ameliorate pro-inflammatory responses in animal models with neurodegeneration, atherosclerosis, and rat paw or ear oedema. TSG can prevent the proliferation of vascular smooth cells, gastrointestinal dysfunctions, platelet aggregation, osteoblastic injury, diabetic nephropathy and melanogenesis. TSG is also indicated to facilitate long-term potentiation and learning and memory in both normal and pathological conditions. These effects to some extent enrich the understanding about the role of TSG in disease prevention and therapy. However, to date, we still have no outlined knowledges about the pharmacological effects of TSG, though the role of TSG in aging and Alzheimer's disease has been reviewed in recent years. Here, we summarize the current pharmacological developments of TSG as well as its possible mechanisms in disease prevention and therapy, aiming to push the understanding about the protective role of TSG as well as its preclinical assessment of novel applications.


Subject(s)
Glucosides/pharmacology , Stilbenes/pharmacology , Aging/drug effects , Animals , Cardiovascular System/drug effects , Central Nervous System/drug effects , Humans
17.
Neurochem Res ; 42(10): 2698-2711, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28434164

ABSTRACT

Recently, the loss and dystrophy of hippocampal microglia induced by chronic unpredictable stress (CUS) has been reported to mediate the development of major depression in mice whose microglial cells were labeled with enhanced green fluorescent protein-conjuncted-CX3C receptor type 1. However, whether this happens in endogenous microglia with no genetic intervention remains unclear. Here, we addressed this issue in mice treated with different types of chronic stresses, including the CUS, chronic restraint stress (CRS) and chronic social defeat stress (CSDS). Results showed that the cellular numbers, process lengths, soma areas and activation markers of endogenous hippocampal but not cortical microglia, were markedly reduced by CUS, CRS and CSDS treatment. Administration of mice with two classical stimulators of microglia, lipopolysaccharide (LPS) or macrophage colony-stimulating factor (M-CSF), reversed the CUS-, CRS- and CSDS-induced reductions in endogenous hippocampal microglial numbers, and also improved the CUS-, CRS- or CSDS-induced behavioral abnormalities, including the increases in the immobile time in the forced swimming test and tail suspension test, the inhibition of sucrose preference, and the decrease in the time spent in the center of open field. Furthermore, inhibition of the initial activation of hippocampal microglia by minocycline pretreatment also reversed the reduction in hippocampal microglial numbers as well as the behavioral abnormalities induced by CUS, CRS and CSDS treatment. These results provide compelling evidences to show that different types of chronic stresses can trigger the loss of endogenous hippocampal microglia and restoration of microglial numbers may have therapeutic values in major depression.


Subject(s)
Hippocampus/drug effects , Lipopolysaccharides/pharmacology , Microglia/drug effects , Minocycline/pharmacology , Animals , Behavior, Animal/drug effects , Depressive Disorder, Major/etiology , Male , Mice , Neurons/drug effects , Stress, Psychological/physiopathology
18.
Int J Neuropsychopharmacol ; 20(6): 485-497, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28339691

ABSTRACT

Background: Z-guggulsterone, an active compound extracted from the gum resin of the tree Commiphora mukul, has been shown to improve animal memory deficits via activating the brain-derived neurotrophic factor signaling pathway. Here, we investigated the antidepressant-like effect of Z-guggulsterone in a chronic unpredictable stress mouse model of depression. Methods: The effects of Z-guggulsterone were assessed in mice with the tail suspension test and forced swimming test. Z-guggulsterone was also investigated in the chronic unpredictable stress model of depression with fluoxetine as the positive control. Changes in hippocampal neurogenesis as well as the brain-derived neurotrophic factor signaling pathway after chronic unpredictable stress/Z-guggulsterone treatment were investigated. The tryptophan hydroxylase inhibitor and the tyrosine kinase B inhibitor were also used to explore the antidepressant-like mechanisms of Z-guggulsterone. Results: Z-guggulsterone (10, 30 mg/kg) administration protected the mice against the chronic unpredictable stress-induced increases in the immobile time in the tail suspension test and forced swimming test and also reversed the reduction in sucrose intake in sucrose preference experiment. Z-guggulsterone (10, 30 mg/kg) administration prevented the reductions in brain-derived neurotrophic factor protein expression levels as well as the phosphorylation levels of cAMP response element binding protein, extracellular signal-regulated kinase 1/2, and protein kinase B in the hippocampus and cortex induced by chronic unpredictable stress. Z-guggulsterone (10, 30 mg/kg) treatment also improved hippocampal neurogenesis in chronic unpredictable stress-treated mice. Blockade of the brain-derived neurotrophic factor signal, but not the monoaminergic system, attenuated the antidepressant-like effects of Z-guggulsterone. Conclusions: Z-guggulsterone exhibits antidepressant activity via activation of the brain-derived neurotrophic factor signaling pathway and upregulation of hippocampal neurogenesis.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Pregnenediones/pharmacology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluoxetine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Neurogenesis/drug effects , Neurogenesis/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
19.
Int J Biochem Cell Biol ; 84: 75-88, 2017 03.
Article in English | MEDLINE | ID: mdl-28104499

ABSTRACT

Heat shock factor 1 (HSF1) critically contributes to the host defense, and its amount determines the mobilization efficiency of HSF1 under stress conditions. To date, how HSF1 amount is regulated remains largely unknown. We found that methylene blue (MB), an anti-oxidative and anti-inflammatory agent, increased the amount of HSF1 in BV-2 microglia, primary microglia, astrocytes, neurons and vital organs. The increased HSF1 contributed to a more increase in nuclear translocation of HSF1, association of HSF1 with heat shock protein 70 (Hsp70) promoters and Hsp70 expression levels, and also induced a synergistic protection against oxidative stress-induced injuries in MB and heat shock-treated cells. The MB-induced increase in the amount of HSF1 was not associated with light exposition as well as the change in HSF1 gene transcription or macroautophagy, but associated with the proteasome-ubiquitin system. The acetyltransferase p300 was considered to mediate the effect of MB on HSF1, as p300 inhibition or silencing prevented the increase in HSF1-p300 interaction as well as the amount and acetylation level of HSF1 in MB-treated cells. Moreover, inhibition of protein kinase A α (PKAα) was found to attenuate the MB-induced increase in HSF1 amount and HSF1-p300 interaction. These findings were ascertained in primary microglia, astrocytes and neurons where p300 or PKA inhibition prevented the increase in the amount of HSF1 after MB treatment. Taken together, our results showed that MB increases the amount of HSF1 through promotion of PKA-mediated increase in HSF1-p300 interaction, providing evidence to illustrate a new pharmacological effect of MB in clinical application.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/metabolism , Methylene Blue/pharmacology , Microglia/drug effects , Microglia/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/drug effects , Neurons/metabolism , Organ Specificity , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription, Genetic/drug effects
20.
FEBS Lett ; 590(18): 3233-42, 2016 09.
Article in English | MEDLINE | ID: mdl-27545319

ABSTRACT

Farnesoid X receptor (FXR) has been confirmed to sense bile acids in various tissues. However, its expression in brain neurons remains obscure. In this study, we identified FXR mRNA and protein expression in mouse brain neurons and in mouse/human brain tissues. FXR was predominantly localized in the nucleus in cultured neurons, but in neurons in vivo, it mainly appeared in the cytoplasm. In nuclear compartments, the neuronal FXR exhibited a punctate distribution. Activation of FXR increased the small heterodimer partner (SHP) mRNA and protein expression levels in cultured neurons and in brain tissues. These findings will help explore new functions of FXR in the brain.


Subject(s)
Brain/metabolism , Neurons/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Brain/cytology , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...