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.
Neural Regen Res ; 13(6): 1019-1025, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29926829

ABSTRACT

Synapses are key structures in neural networks, and are involved in learning and memory in the central nervous system. Investigating synaptogenesis and synaptic aging is important in understanding neural development and neural degeneration in diseases such as Alzheimer disease and Parkinson's disease. Our previous study found that synaptogenesis and synaptic maturation were harmonized with brain development and maturation. However, synaptic damage and loss in the aging cerebellum are not well understood. This study was designed to investigate the occurrence of synaptic aging in the cerebellum by observing the ultrastructural changes of dendritic spines and synapses in cerebellar Purkinje cells of aging mice. Immunocytochemistry, DiI diolistic assays, and transmission electron microscopy were used to visualize the morphological characteristics of synaptic buttons, dendritic spines and synapses of Purkinje cells in mice at various ages. With synaptic aging in the cerebellum, dendritic spines and synaptic buttons were lost, and the synaptic ultrastructure was altered, including a reduction in the number of synaptic vesicles and mitochondria in presynaptic termini and smaller thin specialized zones in pre- and post-synaptic membranes. These findings confirm that synaptic morphology and function is disrupted in aging synapses, which may be an important pathological cause of neurodegenerative diseases.

2.
Int J Mol Med ; 40(6): 1699-1708, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039446

ABSTRACT

Autophagy is a highly conserved process of self-digestion to promote cell survival in response to nutrient starvation and other metabolic stresses. However, whether ischemic-hypoxic (IH) injury-induced autophagy acts as a neuroprotective mechanism or leads to neuroinjury is a subject of debate. It is known that autophagy is regulated by signaling pathways, including the mammalian target of rapamycin pathway. However, in neural IH injury, whether other signaling pathways are involved in the regulation of autophagy remains to be fully elucidated. In the present study, using the autophagy agonist (rampycin), autophagy antagonist [3-methyl adenine (3-MA)] and lysosome antagonist (MHY1485), autophagy was intervened with at oxygen-glucose deprivation (OGD) 6 h, in order to elucidate the regulatory mechanisms of autophagy. Using immunocytochemistry and western blot analysis, the expression levels of stress-related proteins, such as hypoxia-inducible factor-1α (HIF-1α) (a key regulator in hypoxia) and cyclooxygenase 2 (COX2; inflammatory indicator), were analyzed. In addition, the upstream proteins (Wnt1 and Wnt3a), downstream proteins (Dvl2, ß-catenin) and target proteins (C-myc and cyclin D) in the Wnt/ß-catenin signaling pathway were examined by immunocytochemistry and western blot analysis. The present study revealed that autophagy was activated with the upregulation of autophagic flux in IH injury; it was demonstrated that autophagy had a protective role in IH injury. The Wnt/ß-catenin pathway was involved in IH injury regulation, and the upstream proteins in the Wnt/ß-catenin signaling pathway were upregulated, whereas downstream proteins were downregulated by the activity of autophagy accordingly.


Subject(s)
Autophagy/physiology , Hypoxia-Ischemia, Brain/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Models, Biological , Morpholines/pharmacology , PC12 Cells , Rats , Sirolimus/pharmacology , Triazines/pharmacology , Wnt Signaling Pathway/drug effects
3.
Neurochem Res ; 42(10): 2841-2849, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28508993

ABSTRACT

Neuronal apoptosis mediated by the mitochondrial apoptosis pathway is an important pathological process in cerebral ischemia-reperfusion injury. 14,15-EET, an intermediate metabolite of arachidonic acid, can promote cell survival during ischemia/reperfusion. However, whether the mitochondrial apoptotic pathway is involved this survival mechanism is not fully understood. In this study, we observed that infarct size in ischemia-reperfusion injury was reduced in sEH gene knockout mice. In addition, Caspase 3 activation, cytochrome C release and AIF nuclear translocation were also inhibited. In this study, 14,15-EET pretreatment reduced neuronal apoptosis in the oxygen-glucose deprivation and re-oxygenation group in vitro. The mitochondrial apoptosis pathway was also inhibited, as evidenced by AIF translocation from the mitochondria to nucleus and the reduction in the expressions of cleaved-caspase 3 and cytochrome C in the cytoplasm. 14,15-EET could reduce neuronal apoptosis through upregulation of the ratio of Bcl-2 (anti-apoptotic protein) to Bax (apoptosis protein) and inhibition of Bax aggregation onto mitochondria. PI3K/AKT pathway is also probably involved in the reduction of neuronal apoptosis by EET. Our study suggests that 14,15-EET could suppress neuronal apoptosis and reduce infarct volume through the mitochondrial apoptotic pathway. Furthermore, the PI3K/AKT pathway also appears to be involved in the neuroprotection against ischemia-reperfusion by 14,15-EET.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Apoptosis/drug effects , Mitochondria/drug effects , Reperfusion Injury/drug therapy , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Apoptosis/physiology , Cytochromes c/drug effects , Cytochromes c/metabolism , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reperfusion Injury/metabolism , Signal Transduction/physiology
4.
Neural Regen Res ; 12(3): 440-446, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28469659

ABSTRACT

Cold exposure is an external stress factor that causes skin frostbite as well as a variety of diseases. Estrogen might participate in neuroprotection after cold exposure, but its precise mechanism remains unclear. In this study, mice were exposed to 10°C for 7 days and 0-4°C for 30 days to induce a model of chronic cold exposure. Results showed that oxidative stress-related c-fos and cyclooxygenase 2 expressions, MAP1LC3-labeled autophagic cells, Iba1-labeled activated microglia, and interleukin-1ß-positive pyramidal cells were increased in the hippocampal CA1 area. Chronic cold exposure markedly elevated the levels of estrogen in the blood and the estrogen receptor, G protein-coupled receptor 30. These results indicate that neuroimmunoreactivity is involved in chronic cold exposure-induced pathological alterations, including oxidative stress, neuronal autophagy, and neuroimmunoreactivity. Moreover, estrogen exerts a neuroprotective effect on cold exposure.

5.
Neural Regen Res ; 11(2): 312-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27073386

ABSTRACT

To investigate the pattern of neural differentiation and synaptogenesis in the mouse retina, immunolabeling, BrdU assay and transmission electron microscopy were used. We show that the neuroblastic cell layer is the germinal zone for neural differentiation and retinal lamination. Ganglion cells differentiated initially at embryonic day 13 (E13), and at E18 horizontal cells appeared in the neuroblastic cell layer. Neural stem cells in the outer neuroblastic cell layer differentiated into photoreceptor cells as early as postnatal day 0 (P0), and neural stem cells in the inner neuroblastic cell layer differentiated into bipolar cells at P7. Synapses in the retina were mainly located in the outer and inner plexiform layers. At P7, synaptophysin immunostaining appeared in presynaptic terminals in the outer and inner plexiform layers with button-like structures. After P14, presynaptic buttons were concentrated in outer and inner plexiform layers with strong staining. These data indicate that neural differentiation and synaptogenesis in the retina play important roles in the formation of retinal neural circuitry. Our study showed that the period before P14, especially between P0 and P14, represents a critical period during retinal development. Mouse eye opening occurs during that period, suggesting that cell differentiation and synaptic formation lead to the attainment of visual function.

6.
J Neurosci Res ; 92(4): 496-505, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24474045

ABSTRACT

Recently, cold-adaptation medicine has gotten more and more attention because of its specific significance to health care, military activities, sports performance, and so on. Although numerous studies have focused on respiratory, immune, and circulatory systems as well as skin damage upon cold exposure, the impacts on central nervous system are not well understood. This study explores the effects of chronic cold exposure on the murine central nervous system. To establish a chronic cold-exposure animal model, adult male mice from postnatal days 40-50 (P40-50) were housed at 0-4°C for 20 days. During the study period, estrogen receptors were labeled via immunohistochemistry, the dendritic spines of visual cortical pyramidal cells were labeled with DiI diolistic assay, and synaptic ultrastructure was observed by transmission electron microscopy. The results showed that cold exposure could inhibit neural proliferation significantly, with an increase of G-protein-coupled receptor 30 (GPR30) expression. Chronic cold exposure could also induce a decrease in the dendritic spines of pyramidal cells in visual cortex, along with a decrease in the number of synaptic formations. The ultrastructure of synapses after cold exposure was observed. It was found that pre- and postsynaptic membranes were fused, with a vague synaptic cleft. Furthermore, neuronal cytoplasmic and organelle swellings were also observed, along with microtubule disintegration. In conclusion, chronic cold exposure can cause structural and functional changes in the mouse central nervous system, possibly by direct participation of estrogen and its receptor, GPR30, in response to chronic cold exposure.


Subject(s)
Adaptation, Physiological/physiology , Central Nervous System/physiology , Cold Temperature , Gene Expression Regulation/physiology , Neurons/metabolism , Animals , Behavior, Animal , Bromodeoxyuridine/metabolism , Cell Proliferation , Central Nervous System/cytology , Dendritic Spines/physiology , Male , Mice , Microscopy, Electron, Transmission , Neurons/ultrastructure , Receptors, Estrogen , Receptors, G-Protein-Coupled/metabolism , Synapses/physiology
7.
Alcohol Alcohol ; 47(4): 380-5, 2012.
Article in English | MEDLINE | ID: mdl-22414921

ABSTRACT

AIMS: Our aim is to investigate the effects of prenatal alcohol exposure (PAE) on the development of retinal bipolar and horizontal cells. METHODS: The alterations of the retinal bipolar and horizontal cells in P7, P14 and P30 mice were observed after PAE, with immunofluorescent labeling and DiI diolistic assay. RESULTS: The retinal development of filial pups was affected by PAE in a dose-dependent and long-term manner. The number of bipolar cells of alcohol groups was significantly lower than that of the control, and the dendritic receptive field of horizontal cells was also significantly smaller than those of the control groups (P < 0.01). CONCLUSION: PAE was able to cause retarded development of pup retinal neural cells.


Subject(s)
Ethanol/adverse effects , Fetal Alcohol Spectrum Disorders/pathology , Prenatal Exposure Delayed Effects/pathology , Retina/abnormalities , Retinal Bipolar Cells/drug effects , Retinal Horizontal Cells/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Ethanol/blood , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy , Retinal Bipolar Cells/pathology , Retinal Horizontal Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...