Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Front Cell Neurosci ; 17: 1152392, 2023.
Article En | MEDLINE | ID: mdl-37124395

Traumatic brain injury (TBI) is one of the main causes of disability and death, especially in plateau areas, where the degree of injury is often more serious than in plain areas. It is likely that high altitude (HA) aggravates neuroinflammation; however, prior studies are limited. This study was designed to evaluate the effects of HA on the degree of TBI and the neuroprotective effects and underlying mechanisms of L-serine against TBI at HA (HA-TBI). In in vivo experiments, wild-type mice and mice with Nfat1 (Nfat1-/- ) deficiency in the C57BL/6 background were kept in a hypobaric chamber for 3 days under simulated conditions of 4,000 m, 6,000 m and 8,000 m above sea level. After leaving the chamber, the standardized TBI model was established immediately. Mice were then intraperitoneally injected with L-serine (342 mg.kg-1) 2 h after TBI and then daily for 5 days. Behavioral tests and histological analysis were assessed at different time points post TBI induction. In vitro, we applied primary cultured microglia for hypoxia treatment (1% O2 for 24 h). The major findings include the following: (1) with increasing altitude, the neurological function of TBI mice decreased, and the damage to cerebral gray matter and white matter became more significant, (2) L-serine significantly improved the sensorimotor function of mice, reversed the increase in brain lesion volume, and promoted the renovation of brain tissue after HA-TBI, (3) L-serine significantly decreased the activation of microglia and promoted microglia polarization toward the protective M2 phenotype both in vivo and in vitro, (4) L-serine significantly suppressed the expression of NFAT1 in mice after HA-TBI and inhibited NFAT1 expression in primary microglia after hypoxia, and (5) knockout of Nfat1 inhibited the inflammatory reaction caused by excessive activation of microglia, and L-serine lost its neuroprotective effect in Nfat1 knockout mice. The present study suggests that HA aggravates brain damage after TBI and that the damage also increases with increasing altitude. As an endogenous amino acid, L-serine may be a neuroprotective agent against HA-TBI, and suppression of NFAT1 in microglia is a potential therapy for neuroinflammation in the future.

2.
Front Mol Neurosci ; 14: 726665, 2021.
Article En | MEDLINE | ID: mdl-34552468

Central nervous system (CNS) lesions are major causes of human death and disability worldwide, and they cause different extents of motor and sensory dysfunction in patients. Thus, it is crucial to develop new effective neuroprotective drugs and approaches targeted to the heterogeneous nature of CNS injury and disease. L-serine is an indispensable neurotrophic factor and a precursor for neurotransmitters. Although L-serine is a native amino acid supplement, its metabolic products have been shown to be essential not only for cell proliferation but also for neuronal development and specific functions in the brain. Growing evidence has suggested that L-serine regulates the release of several cytokines in the brain under some neuropathological conditions to recover cognitive function, improve cerebral blood flow, inhibit inflammation, promote remyelination and exert other neuroprotective effects on neurological injury. L-serine has also been used to treat epilepsy, schizophrenia, psychosis, and Alzheimer's Disease as well as other neurological diseases. Furthermore, the dosing of animals with L-serine and human clinical trials investigating the therapeutic effects of L-serine generally support the safety of L-serine. The high significance of this review lies in its emphasis on the therapeutic potential of using L-serine as a general treatment for numerous CNS diseases and injuries. Because L-serine performs a broad spectrum of functions, it may be clinically used as an effective neuroprotective agent.

3.
Mol Neurobiol ; 57(11): 4810-4824, 2020 Nov.
Article En | MEDLINE | ID: mdl-32803489

Disruption of remyelination contributes to neurodegeneration and cognitive impairment in chronically disabled patients. Valproic acid (VPA) inhibits histone deacetylase (HDAC) function and probably promotes oligodendrocyte progenitor cell (OPC) proliferation and differentiation; however, the relevant molecular mechanisms remain unknown. Here, focal demyelinating lesions (FDLs) were generated in mice by two-point stereotactic injection of lysophosphatidylcholine (LPC) into the corpus callosum. Cognitive functions, sensorimotor abilities and histopathological changes were assessed for up to 28 days post-injury with or without VPA treatment. Primary OPCs were harvested and used to study the effect of VPA on OPC differentiation under inflammatory conditions. VPA dose-dependently attenuated learning and memory deficits and robustly protected white matter after FDL induction, as demonstrated by reductions in SMI-32 and increases in myelin basic protein staining. VPA also promoted OPC proliferation and differentiation and increased subsequent remyelination efficiency by day 28 post-FDL induction. VPA treatment did not affect HDAC1, HDAC2 or HDAC8 expression but reduced HDAC3 protein levels. In vitro, VPA improved the survival of mouse OPCs and promoted their differentiation into oligodendrocytes following lipopolysaccharide (LPS) stimulation. LPS caused OPCs to overexpress HDAC3, which translocated from the cytoplasm into the nucleus, where it directly interacted with the nuclear transcription factor PPAR-γ and negatively regulated PPAR-γ expression. VPA decreased the expression of HDAC3 and promoted remyelination and functional neurological recovery after FDL. These findings may support the use of strategies modulating HDAC3-mediated regulation of protein acetylation for the treatment of demyelination-related cognitive dysfunction.


Cell Differentiation , Demyelinating Diseases/pathology , Histone Deacetylases/metabolism , Oligodendroglia/pathology , PPAR gamma/metabolism , Stem Cells/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cognition/drug effects , Demyelinating Diseases/physiopathology , Male , Mice, Inbred C57BL , Models, Biological , Neuroprotective Agents/pharmacology , Remyelination/drug effects , Valproic Acid/pharmacology , White Matter/drug effects , White Matter/pathology
4.
Brain Behav ; 10(7): e01675, 2020 07.
Article En | MEDLINE | ID: mdl-32475084

OBJECTIVE: Traumatic brain injury (TBI) results not only in gray matter damage, but also in severe white matter injury (WMI). Previous findings support hypoxic preconditioning (HP) could augment the efficacy of bone marrow stromal cell (BMSC) transplantation in a TBI mouse model. However, whether HP-treated BMSCs (H-BMSCs) could overcome remyelination failure after WMI is unclear, and the molecular mechanisms remain to be explored. Here, we focused on the therapeutic benefits of H-BMSC transplantation for treating WMI, as well as its underlying mechanisms. METHODS: In vitro, BMSCs were incubated at passage 4 in the hypoxic preconditioning (1.0% oxygen) for 8 hr. In vivo, a TBI mouse model was established, and DMEM cell culture medium (control), normal cultured BMSCs (N-BMSCs), or H-BMSCs were transplanted to mice 24 hr afterward. Neurobehavioral function, histopathological changes, and oligodendrogenesis were assessed for up to 35 days post-TBI. RESULTS: Compared with the control group, improvement of cognitive functions and smaller lesion volumes was observed in the two BMSC-transplanted groups, especially the H-BMSC group. H-BMSC transplantation resulted in a greater number of neural/glial antigen 2 (NG2)-positive and adenomatous polyposis coli (APC)-positive cells than N-BMSC transplantation in both the corpus callosum and the striatum. In addition, we observed that the expression levels of hypoxia-inducible factor-1a (HIF-1α), phosphorylated mechanistic target of rapamycin (p-mTOR), and vascular endothelial growth factor (VEGF) were all increased in H-BMSC-transplanted mice. Furthermore, the mTOR pathway inhibitor rapamycin attenuated the impact of HP both in vivo and in vitro. CONCLUSION: The results provided mechanistic evidences suggesting that HP-treated BMSCs promoted remyelination partly by modulating the pro-survival mTOR/HIF-1α/VEGF signaling pathway.


Brain Injuries, Traumatic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Brain Injuries, Traumatic/therapy , Cell Differentiation , Hypoxia , Male , Mice , Mice, Inbred C57BL , Oligodendroglia , Rabbits , TOR Serine-Threonine Kinases , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
Micromachines (Basel) ; 10(10)2019 Sep 23.
Article En | MEDLINE | ID: mdl-31547592

Wafer bonding of a silicon carbide (SiC) diaphragm to a patterned SiC substrate coated with aluminum nitride (AlN) film as an insulating layer is a promising choice to fabricate an all-SiC capacitive pressure sensor. To demonstrate the bonding feasibility, a crystalline AlN film with a root-mean-square (RMS) surface roughness less than ~0.70 nm was deposited on a SiC wafer by a pulsed direct current magnetron sputtering method. Room temperature wafer bonding of SiC-AlN by two surface activated bonding (SAB) methods (standard SAB and modified SAB with Si nano-layer sputtering deposition) was studied. Standard SAB failed in the bonding, while the modified SAB achieved the bonding with a bonding energy of ~1.6 J/m2. Both the microstructure and composition of the interface were investigated to understand the bonding mechanisms. Additionally, the surface analyses were employed to confirm the interface investigation. Clear oxidation of the AlN film was found, which is assumed to be the failure reason of direct bonding by standard SAB.

6.
Breast Cancer Res Treat ; 176(2): 291-301, 2019 Jul.
Article En | MEDLINE | ID: mdl-31006103

BACKGROUND: Phosphorylated AKT is highly expressed or overexpressed in chemoresistant tumor samples. However, the precise molecular mechanism involved in AKT phosphorylation-related chemoresistance in breast cancer is still elusive. The present research was designed to estimate the effect of AKT phosphorylation on cell viability and chemoresistance in breast cancer. METHODS: We utilized MCF-7 and MDA-MB468 human breast cancer cell lines and developed multidrug-resistant MCF-7/MDR and cisplatin-resistant MDA-MB-468 cells. Immunofluorescence analysis and Western blotting were employed to test the level of glycogen synthase kinase 3 beta (GSK3ß), phosphorylated phosphatase and tension homologue (p-PTEN) and phosphorylated AKT (p-AKT) in MCF-7/MDR and MDA-MB468 cells. Xenograft assays in nude mice were performed with MCF-7/MDR cells to verify chemoresistance and the signaling pathway upstream of phosphatidylinositide 3-kinase (PI3K)/AKT. RESULTS: An increase in GSK3ß, p-PTEN and p-AKT expression was strongly induced in MCF-7/MDR and cisplatin-resistant MDA-MB-468 cells, and augmented GSK3ß phosphorylation and PTEN inactivation enhanced AKT signaling. The elevation in GSK3ß, p-PTEN and p-AKT was associated with cell viability based on a CCK-8 assay. The results of in vivo and in vitro assays indicated that GSK3ß knockdown with lentiviral shRNA (shRNA-GSK3ß) promoted apoptosis and suppressed the migration of cisplatin-resistant MCF-7/MDR cells, while these effects were reversed by activating p-AKT with the PTEN inhibitor bpV(pic). CONCLUSIONS: AKT phosphorylation mediated by GSK3ß and PTEN were correlated with cell viability, migration and apoptosis, which may promote chemoresistance in breast cancer. Furthermore, GSK3ß can regulate cell viability through the PTEN/PI3K/AKT signaling pathway and induce chemoresistance, serving as a valuable molecular strategy for breast cancer therapy.


Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Glycogen Synthase Kinase 3 beta/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Survival , Cisplatin , Drug Resistance, Multiple , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation
7.
Neuropharmacology ; 146: 39-49, 2019 03 01.
Article En | MEDLINE | ID: mdl-30452956

Demyelination in white matter is the end product of numerous pathological processes. This study was designed to evaluate the neuroprotective effect of l-serine and the underlying mechanisms against the demyelinating injury of white matter. A model of focal demyelinating lesions (FDL) was established using the two-point stereotactic injection of 0.25% lysophosphatidylcholine (LPC, 10 µg per point) into the corpus callosum of mice. Mice were then intraperitoneally injected with one of three doses of l-serine (114, 342, or 1026 mg/kg) 2 h after FDL, and then twice daily for the next five days. Behavior tests and histological analysis were assessed for up to twenty-eight days post-FDL induction. Electron microscopy was used for ultrastructural investigation. In vitro, we applied primary co-cultures of microglia and oligodendrocytes for oxygen glucose deprivation (OGD). After establishing FDL, l-serine treatment: 1) improved spatial learning, memory and cognitive ability in mice, and relieved anxiety for 4 weeks post-FDL induction; 2) reduced abnormally dephosphorylated neurofilament proteins, increased myelin basic protein, and preserved anatomic myelinated axons; 3) inhibited microglia activation and reduced the release of inflammatory factors; 4) promoted recruitment and proliferation of oligodendrocyte progenitor cells, and the efficiency of subsequent remyelination on day twenty-eight post-FDL induction. In vitro experiments, showed that l-serine not only directly protected against oligodendrocytes from OGD damage, but also provided an indirect protective effect by regulating microglia. In our study, l-serine offered long-lasting behavioral and oligodendrocyte protection and promoted remyelination. Therefore, l-serine may be an effective clinical treatment aganist white matter injury.


Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Serine/pharmacology , Animals , Anxiety , Axons/drug effects , Calcium-Binding Proteins/metabolism , Corpus Callosum/drug effects , Corpus Callosum/metabolism , Demyelinating Diseases/chemically induced , Exploratory Behavior/drug effects , Inflammation/metabolism , L-Lactate Dehydrogenase (Cytochrome)/metabolism , Lysophosphatidylcholines/toxicity , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Myelin Basic Protein/metabolism , Myelin Sheath/pathology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Serine/metabolism , Spatial Learning/drug effects , Spatial Memory/drug effects
...