Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Neurosci Lett ; 810: 137350, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37327854

ABSTRACT

Depression is a mental disease that seriously affects the quality of life. Its pathophysiology is complex and includes neuroinflammation and apoptosis. Virgin coconut oil (VCO) is a natural food that has been found to have remarkable anti-inflammatory and antiapoptotic properties. We assessed the effects of VCO on depression and the related mechanisms by performing network pharmacology analysis and evaluating depressive-like behaviors in rat model and found that VCO-treatment alleviated the depressive-like behaviors, inhibited microglial and astrocytic activation and reduced neuronal loss in the hippocampus, possibly by decreasing neuronal apoptosis. In addition, network pharmacology analysis and western blotting showed that VCO might exert neuroprotective effects by activating Protein Kinase B (AKT)-related pathway. Taken together, our results revealed the previously unrecognized effects of VCO on depression, and further explored the underlying mechanism of depression.


Subject(s)
Depression , Lipopolysaccharides , Rats , Animals , Coconut Oil/pharmacology , Lipopolysaccharides/pharmacology , Depression/chemically induced , Depression/drug therapy , Network Pharmacology , Quality of Life
2.
Glia ; 71(5): 1197-1216, 2023 05.
Article in English | MEDLINE | ID: mdl-36617748

ABSTRACT

The homeostasis of glutamate is mainly regulated by the excitatory amino acid transporters (EAATs), especially by EAAT2 in astrocytes. Excessive glutamate in the synaptic cleft caused by dysfunction or dysregulation of EAAT2 can lead to excitotoxicity, neuronal death and cognitive dysfunction. However, it remains unclear about the detailed regulation mechanism of expression and function of astrocytic EAAT2. In this study, first, we found increased neuronal death and impairment of cognitive function in YAPGFAP -CKO mice (conditionally knock out Yes-associated protein [YAP] in astrocytes), and identified EAAT2 as a downstream target of YAP through RNA sequencing. Second, the expression of EAAT2 was decreased in cultured YAP-/- astrocytes and the hippocampus of YAPGFAP -CKO mice, and glutamate uptake was reduced in YAP-/- astrocytes, but increased in YAP-upregulated astrocytes. Third, further investigation of the mechanism showed that the mRNA and protein levels of ß-catenin were decreased in YAP-/- astrocytes and increased in YAP-upregulated astrocytes. Wnt3a activated YAP signaling and up-regulated EAAT2 through ß-catenin. Furthermore, over-expression or activation of ß-catenin partially restored the downregulation of EAAT2, the impairment of glutamate uptake, neuronal death and cognitive decline that caused by YAP deletion. Finally, activation of EAAT2 also rescued neuronal death and cognitive decline in YAPGFAP -CKO mice. Taken together, our study identifies an unrecognized role of YAP signaling in the regulation of glutamate homeostasis through the ß-catenin/EAAT2 pathway in astrocytes, which may provide novel insights into the pathogenesis of brain diseases that closely related to the dysfunction or dysregulation of EAAT2, and promote the development of clinical strategy.


Subject(s)
Astrocytes , YAP-Signaling Proteins , Animals , Mice , Astrocytes/metabolism , beta Catenin/metabolism , Glutamic Acid/metabolism , Homeostasis , Amino Acid Transport Systems/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism
3.
Cell Death Dis ; 13(7): 638, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35869039

ABSTRACT

Autism spectrum disorder (ASD), a group of neurodevelopmental disorder diseases, is characterized by social deficits, communication difficulties, and repetitive behaviors. Sterile alpha and TIR motif-containing 1 protein (SARM1) is known as an autism-associated protein and is enriched in brain tissue. Moreover, SARM1 knockdown mice exhibit autism-like behaviors. However, its specific mechanism in ASD pathogenesis remains unclear. Here we generated parvalbumin-positive interneurons (PVI)-specific conditional SARM1 knockout (SARM1PV-CKO) mice. SARM1PV-CKO male mice showed autism-like behaviors, such as mild social interaction deficits and repetitive behaviors. Moreover, we found that the expression level of parvalbumin was reduced in SARM1PV-CKO male mice, together with upregulated apoptosis-related proteins and more cleaved-caspase-3-positive PVIs, suggesting that knocking out SARM1 may cause a reduction in the number of PVIs due to apoptosis. Furthermore, the expression of c-fos was shown to increase in SARM1PV-CKO male mice, in combination with upregulation of excitatory postsynaptic proteins such as PSD-95 or neuroligin-1, indicating enhanced excitatory synaptic input in mutant mice. This notion was further supported by the partial rescue of autism-like behavior deficits by the administration of GABA receptor agonists in SARM1PV-CKO male mice. In conclusion, our findings suggest that SARM1 deficiency in PVIs may be involved in the pathogenesis of ASD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Armadillo Domain Proteins/genetics , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Autistic Disorder/pathology , Cytoskeletal Proteins/genetics , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Parvalbumins/metabolism
4.
Front Aging Neurosci ; 13: 592845, 2021.
Article in English | MEDLINE | ID: mdl-33935680

ABSTRACT

OBJECTIVE: There is growing evidence that testosterone may be implicated in the pathogenesis of Alzheimer's disease (AD). We aimed to examine the relationship between plasma total testosterone levels and change in brain glucose metabolism over time among non-demented older people. METHODS: The association of plasma total testosterone levels with change in brain glucose metabolism among non-demented older people was investigated cross-sectionally and longitudinally. Given a significant difference in levels of plasma total testosterone between gender, we performed our analysis in a sex-stratified way. At baseline, 228 non-demented older people were included: 152 males and 76 females. RESULTS: In the cross-sectional analysis, no significant relationship between plasma total testosterone levels and brain glucose metabolism was found in males or females. In the longitudinal analysis, we found a significant association of plasma total testosterone levels with change in brain glucose metabolism over time in males, but not in females. More specifically, in males, higher levels of total testosterone in plasma at baseline were associated with slower decline in brain glucose metabolism. CONCLUSION: We found that higher levels of total testosterone in plasma at baseline were associated with slower decline in brain glucose metabolism in males without dementia, indicating that testosterone may have beneficial effects on brain function.

5.
J Neurosci ; 40(13): 2644-2662, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32066583

ABSTRACT

Yes-associated protein (YAP) transcriptional coactivator is negatively regulated by the Hippo pathway and functions in controlling the size of multiple organs, such as liver during development. However, it is not clear whether YAP signaling participates in the process of the formation of glia scars after spinal cord injury (SCI). In this study, we found that YAP was upregulated and activated in astrocytes of C57BL/6 male mice after SCI in a Hippo pathway-dependent manner. Conditional knockout (KO) of yap in astrocytes significantly inhibited astrocytic proliferation, impaired the formation of glial scars, inhibited the axonal regeneration, and impaired the behavioral recovery of C57BL/6 male mice after SCI. Mechanistically, the bFGF was upregulated after SCI and induced the activation of YAP through RhoA pathways, thereby promoting the formation of glial scars. Additionally, YAP promoted bFGF-induced proliferation by negatively controlling nuclear distribution of p27Kip1 mediated by CRM1. Finally, bFGF or XMU-MP-1 (an inhibitor of Hippo kinase MST1/2 to activate YAP) injection indeed activated YAP signaling and promoted the formation of glial scars and the functional recovery of mice after SCI. These findings suggest that YAP promotes the formation of glial scars and neural regeneration of mice after SCI, and that the bFGF-RhoA-YAP-p27Kip1 pathway positively regulates astrocytic proliferation after SCI.SIGNIFICANCE STATEMENT Glial scars play critical roles in neuronal regeneration of CNS injury diseases, such as spinal cord injury (SCI). Here, we provide evidence for the function of Yes-associated protein (YAP) in the formation of glial scars after SCI through regulation of astrocyte proliferation. As a downstream of bFGF (which is upregulated after SCI), YAP promotes the proliferation of astrocytes through negatively controlling nuclear distribution of p27Kip1 mediated by CRM1. Activation of YAP by bFGF or XMU-MP-1 injection promotes the formation of glial scar and the functional recovery of mice after SCI. These results suggest that the bFGF-RhoA-YAP-p27Kip1 axis for the formation of glial scars may be a potential therapeutic strategy for SCI patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Astrocytes/metabolism , Cell Cycle Proteins/metabolism , Gliosis/metabolism , Nerve Regeneration/physiology , Spinal Cord Injuries/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gliosis/genetics , Gliosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Recovery of Function/physiology , Signal Transduction/physiology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , YAP-Signaling Proteins
6.
Glia ; 68(9): 1757-1774, 2020 09.
Article in English | MEDLINE | ID: mdl-32057144

ABSTRACT

Olfactory ensheathing cells (OECs) are unique glial cells with axonal growth-promoting properties in the olfactory epithelium and olfactory bulb, covering the entire length of the olfactory nerve. The proliferation of OECs is necessary for the formation of the presumptive olfactory nerve layer (ONL) during development and OECs transplantation. However, the molecular mechanism underlying the regulation of OEC proliferation in the ONL still remains unknown. In the present study, we examined the role of sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs) on OEC proliferation. Initially, reverse transcription-PCR (RT-PCR), western blot and immunostaining revealed that S1PRs were highly expressed in the OECs in vitro and in vivo. Furthermore, we found that S1P treatment promoted the proliferation of primary cultured OECs mediated by S1PR1. Mechanistically, yes-associated protein (YAP) was required for S1P-induced OEC proliferation through RhoA signaling. Finally, conditional knockout of YAP in OECs reduced OEC proliferation in ONL, which impaired the axonal projection and growth of olfactory sensory neurons, and olfactory functions. Taken together, these results reveal a previously unrecognized function of S1P/RhoA/YAP pathway in the proliferation of OECs, contributing to the formation of ONL and the projection, growth, and function of olfactory sensory neurons during development.


Subject(s)
Neuroglia , Olfactory Nerve , Cell Proliferation , Cells, Cultured , Lysophospholipids , Olfactory Bulb , Sphingosine/analogs & derivatives
7.
Mol Cell Neurosci ; 92: 27-39, 2018 10.
Article in English | MEDLINE | ID: mdl-29940213

ABSTRACT

Olfactory ensheathing cells (OECs) migrate from olfactory epithelium towards olfactory bulb (OB), contributing to formation of the presumptive olfactory nerve layer during development. However, it remains unclear that molecular mechanism of regulation of OEC migration in OB. In the present study, we found that OECs highly expressed the receptors of semaphorin 3A (Sema3A) in vitro and in vivo, whereas Sema3A displayed a gradient expression pattern with higher in inner layer of OB and lower in outer layer of OB. Furthermore, the collapse assays, Boyden chamber migration assays and single-cell migration assays showed that Sema3A induced the collapse of leading front of OECs and inhibited OEC migration. Thirdly, the leading front of OECs exhibited adaptation in a protein synthesis-independent manner, and endocytosis-dependent manner during Sema3A-induced OEC migration. Finally, Sema3A-induced collapse of leading front was required the decrease of focal adhesion and a retrograde F-actin flow in a cofilin activation-dependent manner. Taken together, these results demonstrate that Sema3A as an inhibitive migratory factor for OEC migration through cofilin activation is involved in the formation of olfactory nerve layer.


Subject(s)
Cell Movement , Olfactory Nerve/cytology , Semaphorin-3A/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Cells, Cultured , Male , Neurogenesis , Neuroglia/cytology , Neuroglia/metabolism , Olfactory Nerve/metabolism , Rats , Rats, Sprague-Dawley , Semaphorin-3A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...