1.G protein-coupled estrogen receptor alleviates cerebral ischemia-reperfusion injury through inhibiting endoplasmic reticulum stress.
Zi-Wei HAN ; Li-Cang ZHU ; Yue-Chen CHANG ; Ying ZHOU ; Jia-An ZONG ; Ke-Tao MA ; Jun-Qiang SI ; Li LI
Acta Physiologica Sinica 2019;71(4):527-536
The aim of this study was to investigate whether G protein-coupled estrogen receptor (GPER) could alleviate hippocampal neuron injury under cerebral ischemia-reperfusion injury (CIRI) by acting on endoplasmic reticulum stress (ERS). The CIRI animal model was established by middle cerebral artery occlusion (MCAO). Female ovariectomized (OVX) Sprague-Dawley (SD) female rats were randomly divided into 4 groups: control, ischemia-reperfusion injury (MCAO), vehicle (MCAO+DMSO), and GPER-specific agonist G1 (MCAO+G1) groups. The neurobehavioral score was assessed by the Longa score method, the morphological changes of the neurons were observed by the Nissl staining, the cerebral infarction was detected by the TTC staining, and the neural apoptosis in the hippocampal CA1 region was detected by TUNEL staining. The distribution and expression of GRP78 (78 kDa glucose-regulated protein 78) in the hippocampal CA1 region were observed by immunofluorescent staining. The protein expression levels of GRP78, Caspase-12, CHOP and Caspase-3 were detected by Western blot, and the mRNA expression levels of GRP78, Caspase-12, and CHOP were detected by the real-time PCR. The results showed that the neurobehavioral score, cerebral infarct volume, cellular apoptosis index, as well as GRP78, Caspase-12 and CHOP protein and mRNA expression levels in the MCAO group were significantly higher than those of control group. And G1 reversed the above-mentioned changes in the MCAO+G1 group. These results suggest that the activation of GPER can decrease the apoptosis of hippocampal neurons and relieve CIRI, and its mechanism may involve the inhibition of ERS.
Animals
;
Apoptosis
;
Brain Ischemia
;
CA1 Region, Hippocampal
;
cytology
;
Caspase 12
;
metabolism
;
Caspase 3
;
metabolism
;
Endoplasmic Reticulum Stress
;
Female
;
Heat-Shock Proteins
;
metabolism
;
Neurons
;
cytology
;
Random Allocation
;
Rats
;
Rats, Sprague-Dawley
;
Receptors, Estrogen
;
physiology
;
Receptors, G-Protein-Coupled
;
agonists
;
Reperfusion Injury
;
Transcription Factor CHOP
;
metabolism
2.Hierarchical Control of Drosophila Sleep, Courtship, and Feeding Behaviors by Male-Specific P1 Neurons.
Wenxuan ZHANG ; Chao GUO ; Dandan CHEN ; Qionglin PENG ; Yufeng PAN
Neuroscience Bulletin 2018;34(6):1105-1110
Animals choose among sleep, courtship, and feeding behaviors based on the integration of both external sensory cues and internal states; such choices are essential for survival and reproduction. These competing behaviors are closely related and controlled by distinct neural circuits, but whether they are also regulated by shared neural nodes is unclear. Here, we investigated how a set of male-specific P1 neurons controls sleep, courtship, and feeding behaviors in Drosophila males. We found that mild activation of P1 neurons was sufficient to affect sleep, but not courtship or feeding, while stronger activation of P1 neurons labeled by four out of five independent drivers induced courtship, but only the driver that targeted the largest number of P1 neurons affected feeding. These results reveal a common neural node that affects sleep, courtship, and feeding in a threshold-dependent manner, and provide insights into how competing behaviors can be regulated by a shared neural node.
Animals
;
Animals, Genetically Modified
;
Brain
;
cytology
;
Courtship
;
Drosophila
;
Drosophila Proteins
;
genetics
;
metabolism
;
Feeding Behavior
;
physiology
;
Locomotion
;
Male
;
Neural Inhibition
;
physiology
;
Neural Pathways
;
physiology
;
Neurons
;
physiology
;
Sex Factors
;
Sleep
;
physiology
3.Whole-Brain Mapping of Direct Inputs to and Axonal Projections from GABAergic Neurons in the Parafacial Zone.
Yun-Ting SU ; Meng-Yang GU ; Xi CHU ; Xiang FENG ; Yan-Qin YU
Neuroscience Bulletin 2018;34(3):485-496
The GABAergic neurons in the parafacial zone (PZ) play an important role in sleep-wake regulation and have been identified as part of a sleep-promoting center in the brainstem, but the long-range connections mediating this function remain poorly characterized. Here, we performed whole-brain mapping of both the inputs and outputs of the GABAergic neurons in the PZ of the mouse brain. We used the modified rabies virus EnvA-ΔG-DsRed combined with a Cre/loxP gene-expression strategy to map the direct monosynaptic inputs to the GABAergic neurons in the PZ, and found that they receive inputs mainly from the hypothalamic area, zona incerta, and parasubthalamic nucleus in the hypothalamus; the substantia nigra, pars reticulata and deep mesencephalic nucleus in the midbrain; and the intermediate reticular nucleus and medial vestibular nucleus (parvocellular part) in the pons and medulla. We also mapped the axonal projections of the PZ GABAergic neurons with adeno-associated virus, and defined the reciprocal connections of the PZ GABAergic neurons with their input and output nuclei. The newly-found inputs and outputs of the PZ were also listed compared with the literature. This cell-type-specific neuronal whole-brain mapping of the PZ GABAergic neurons may reveal the circuits underlying various functions such as sleep-wake regulation.
Animals
;
Axons
;
physiology
;
Brain
;
anatomy & histology
;
Brain Mapping
;
Brain Stem
;
cytology
;
GABAergic Neurons
;
physiology
;
Green Fluorescent Proteins
;
genetics
;
metabolism
;
Mice
;
Mice, Inbred C57BL
;
Mice, Transgenic
;
Neural Pathways
;
physiology
;
Peptide Elongation Factor 1
;
genetics
;
metabolism
;
Rabies virus
;
genetics
;
metabolism
;
Transduction, Genetic
;
Vesicular Inhibitory Amino Acid Transport Proteins
;
genetics
;
metabolism
4.Nr2e1 Downregulation Is Involved in Excess Retinoic Acid-induced Developmental Abnormality in the Mouse Brain.
Juan YU ; Qian GUO ; Jian Bing MU ; Ting ZHANG ; Ren Ke LI ; Jun XIE
Biomedical and Environmental Sciences 2017;30(3):185-193
OBJECTIVEThis study aimed to investigate the expression pattern and function of Nuclear receptor subfamily 2 group E member 1 (Nr2e1) in retinoic acid (RA)-induced brain abnormality.
METHODSThe mouse model of brain abnormality was established by administering 28 mg/kg RA, and neural stem cells (NSCs) were isolated from the mouse embryo and cultured in vitro. Nr2e1 expression was detected by whole mount in situ hybridization, RT-PCR, and Western blotting. Nr2e1 function was determined by transducing Nr2e1 shRNA into NSCs, and the effect on the sonic hedgehog (Shh) signaling pathway was assessed in the cells. In addition, the regulation of Nr2e1 expression by RA was also determined in vitro.
RESULTSNr2e1 expression was significantly downregulated in the brain and NSCs of RA-treated mouse embryos, and knockdown of Nr2e1 affected the proliferation of NSCs in vitro. In addition, a similar expression pattern of Nr2e1 and RA receptor (RAR) α was observed after treatment of NSCs with different concentrations of RA.
CONCLUSIONOur study demonstrated that Nr2e1 could be regulated by RA, which would aid a better understanding of the mechanism underlying RA-induced brain abnormality.
Animals ; Brain ; cytology ; embryology ; Cell Proliferation ; Down-Regulation ; Gene Expression Regulation ; Gene Expression Regulation, Developmental ; drug effects ; Mice ; Mice, Inbred C57BL ; Neural Stem Cells ; drug effects ; physiology ; Receptors, Cytoplasmic and Nuclear ; genetics ; metabolism ; Tretinoin ; pharmacology
5.Possible Mechanism of Therapeutic Effect of 3-Methyl-1-phenyl-2-pyrazolin-5-one and Bone Marrow Stromal Cells Combination Treatment in Rat Ischemic Stroke Model.
Li-Hua SHEN ; Jin CHEN ; Hua-Chao SHEN ; Min YE ; Xiao-Fei LIU ; Wen-Sen DING ; Ya-Feng SHENG ; Xin-Sheng DING ;
Chinese Medical Journal 2016;129(12):1471-1476
BACKGROUNDThe functional improvement following bone marrow stromal cells (BMSCs) transplantation after stroke is directly related to the number of engrafted cells and neurogenesis in the injured brain. Here, we tried to evaluate whether 3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186), a free radical scavenger, might influence BMSCs migration to ischemic brain, which could promote neurogenesis and thereby enhance treatment effects after stroke.
METHODSRat transient middle cerebral artery occlusion (MCAO) model was established. Two separate MCAO groups were administered with either MCI-186 or phosphate-buffered saline (PBS) solution to evaluate the expression of stromal cell-derived factor-1 (SDF-1) in ischemic brain, and compared to that in sham group (n = 5/ group/time point[at 1, 3, and 7 days after operation]). The content of chemokine receptor-4 (CXCR4, a main receptor of SDF-1) at 7 days after operation was also observed on cultured BMSCs. Another four MCAO groups were intravenously administered with either PBS, MCI-186, BMSCs (2 × 106), or a combination of MCI-186 and BMSCs (n = 10/group). 5-bromo-2-deoxyuridine (BrdU) and Nestin double-immunofluorescence staining was performed to identify the engrafted BMSCs and neuronal differentiation. Adhesive-removal test and foot-fault evaluation were used to test the neurological outcome.
RESULTSMCI-186 upregulated the expression of SDF-1 in ischemic brain and CXCR4 content in BMSCs was enhanced after hypoxic stimulation. When MCAO rats were treated with either MCI-186, BMSCs, or a combination of MCI-186 and BMSCs, the neurologic function was obviously recovered as compared to PBS control group (P < 0.01 or 0.05, respectively). Combination therapy represented a further restoration, increased the number of BMSCs and Nestin+ cells in ischemic brain as compared with BMSCs monotherapy (P < 0.01). The number of engrafted-BMSCs was correlated with the density of neuronal cells in ischemic brain (r = 0.72 , P < 0.01) and the improvement of foot-fault (r = 0.70, P < 0.01).
CONCLUSIONMCI-186 might promote BMSCs migration to the ischemic brain, amplify the neurogenesis, and improve the effects of cell therapy.
Animals ; Antipyrine ; analogs & derivatives ; therapeutic use ; Bone Marrow Cells ; cytology ; physiology ; Brain Ischemia ; drug therapy ; metabolism ; therapy ; Chemokine CXCL12 ; metabolism ; Disease Models, Animal ; Infarction, Middle Cerebral Artery ; drug therapy ; metabolism ; therapy ; Male ; Mesenchymal Stromal Cells ; physiology ; Neurogenesis ; physiology ; Rats ; Rats, Sprague-Dawley ; Stroke ; drug therapy ; metabolism ; therapy
6.Role of TRPV4 channels in regulation of eNOS expression in brain microvascular endothelial cells under the condition of mechanical stretch.
Jiujun PU ; Zhiming WANG ; Hui ZHOU ; Ailing ZHONG ; Lunliang RUAN ; Kai JIN ; Gang YANG
Journal of Central South University(Medical Sciences) 2015;40(9):960-966
OBJECTIVE:
To identify the subtype of transient receptor potential (TRPs) channel involved in stretch-induced injury of human brain microvascular endothelial cells (HBMEC) and to explore the mechanism responsible for eNOS expression.
METHODS:
TRPs expression was examined by Western blot and immunocytofluoresence in the cultured HBMEC. Mechanical stretch was performed by mini-type multi-functional bio-impact machine. The levels of free calcium ion in cells were examined by the flow cytometry. The eNOS expression was detected by Western blot.
RESULTS:
The mRNA and protein expression of TRPV4 was detected in HBMEC by qRT-PCR, Western blot and immunocytofluoresence. The levels of free calcium ion in the stretch-treated HBMEC was significantly decreased in the presence of TRPV4 specific inhibitor (P<0.001), but there was no difference in calcium levels between the stretch and the control or unspecific inhibitor group (P=0.072 or 0.308). The levels of eNOS protein in the stretch-treated HBMEC were reduced in the presence of TRPV4 specific inhibitor or NOS inhibitor (P<0.05), but it was not changed compared with that in the control group (P>0.05).
CONCLUSION
The eNOS expression is up-regulated under the condition of mechanic stretch, which is related to the activation of TRPV4, resulting in the influx of calcium.
Brain
;
cytology
;
Calcium
;
metabolism
;
Cells, Cultured
;
Endothelial Cells
;
physiology
;
Humans
;
Nitric Oxide Synthase Type III
;
metabolism
;
Stress, Mechanical
;
TRPV Cation Channels
;
physiology
7.Proliferation and differentiation of neural stem cells co-cultured with cerebral microvascular endothelial cells after oxygen-glucose deprivation.
Yong-jie XIONG ; Bo YIN ; Lian-chen XIAO ; Qian WANG ; Li GAN ; Yi-chi ZHANG ; Su-ming ZHANG
Journal of Huazhong University of Science and Technology (Medical Sciences) 2013;33(1):63-68
Various stem cells, including neural stem cells (NSCs), have been extensively studied in stroke models, but how to increase neuronal differentiation rate of NSCs remains unresolved, particularly in a damaged environment. The purpose of this study was to investigate the effects of cerebral microvascular endothelial cells (CMECs) on the neurogenesis of NSCs with or without oxygen-glucose deprivation (OGD). The NSCs acquired from primary culture were immunostained to prove cell purity. Survival and proliferation of NSCs were determined after the co-culture with CMECs for 7 days. After removing the CMECs, NSCs were randomly divided into two groups as follows: OGD and non-OGD groups. Both groups were maintained in differentiation culture for 4 days to evaluate the differentiation rate. Mouse embryo fibroblast (MEF) cells co-cultured with NSCs served as control group. NSCs co-cultured with CMECs had an increase in size (on the 7th day: 89.80±26.12 μm vs. 73.08±15.01 μm, P<0.001) (n=12) and number [on the 7th day: 6.33±5.61/high power objective (HP) vs. 2.23±1.61/HP, P<0.001] (n=12) as compared with those co-cultured with MEF cells. After further differentiation culture for 4 days, NSCs co-cultured with CMECs had an increase in neuronal differentiation rate in OGD and non-OGD groups, but not in the control group (15.16% and 16.07% vs. 8.81%; both P<0.001) (n=6). This study provided evidence that OGD could not alter the effects of CMECs in promoting the neuronal differentiation potential of NSCs. These findings may have important implications for the development of new cell therapies for cerebral vascular diseases.
Animals
;
Animals, Newborn
;
Brain
;
blood supply
;
Cell Differentiation
;
physiology
;
Cell Proliferation
;
Cells, Cultured
;
Coculture Techniques
;
methods
;
Endothelial Cells
;
cytology
;
metabolism
;
Glucose
;
metabolism
;
Mice
;
Mice, Inbred C57BL
;
Microvessels
;
cytology
;
metabolism
;
Neural Stem Cells
;
cytology
;
metabolism
;
Oxygen
;
metabolism
8.Influence of acute ethanol intoxication on neuronal apoptosis and Bcl-2 protein expression after severe traumatic brain injury in rats.
Min HE ; Wei-Guo LIU ; Liang WEN ; Hang-Gen DU ; Li-Chun YIN ; Li CHEN
Chinese Journal of Traumatology 2013;16(3):136-139
OBJECTIVETo study the influence and mechanism of acute ethanol intoxication (AEI) on rat neuronal apoptosis after severe traumatic brain injury (TBI).
METHODSNinety-six Sprague-Dawley rats were randomly divided into four groups: normal control, AEI-only, TBI-only and TBI+AEI (n equal to 24 for each). Severe TBI model was developed according to Feeney's method. Rats in TBI+AEI group were firstly subjected to AEI, and then suffered head trauma. In each group, animals were sacrificed at 6 h, 24 h, 72 h, and 168 h after TBI. The level of neuronal apoptosis and the expression of Bcl-2 protein were determined by TUNEL assay and immunohistochemical method, respectively.
RESULTSApoptotic cells mainly distributed in the cortex and white matter around the damaged area. Neuronal apoptosis significantly increased at 6 h after trauma and peaked at 72 h. Both the level of neuronal apoptosis and expression of Bcl-2 protein in TBI-only group and TBI+AEI group were higher than those in control group (P less than 0.05). Compared with TBI-only group, the two indexes were much higher in TBI+AEI group at all time points (P less than 0.05).
CONCLUSIONOur findings suggest that AEI can increase neuronal apoptosis after severe TBI.
Animals ; Apoptosis ; drug effects ; Brain Injuries ; Cerebral Cortex ; cytology ; Disease Models, Animal ; Ethanol ; poisoning ; Immunohistochemistry ; In Situ Nick-End Labeling ; Male ; Neurons ; physiology ; Prosencephalon ; cytology ; Proto-Oncogene Proteins c-bcl-2 ; metabolism ; Rats ; Rats, Sprague-Dawley
9.Research progress of the relationship between microglia and cerebral ischemia.
Wen-Jiao TAI ; Xuan YE ; Xiu-Qi BAO ; Xiao-Liang WANG ; Dan ZHANG
Acta Pharmaceutica Sinica 2012;47(3):346-353
Microglia are the principal immune effectors in brain and participate in a series ofneurodegenerative diseases. The microglial shapes are highly plastic. The morphology is closely related with their activation status and biological functions. Cerebral ischemia could induce microglial activation, and microglial activation is subjected to precise regulation. Microglia could play either protective or neurotoxic roles in cerebral ischemia. Therefore, regulating the expression of receptors or protein molecules on microglia, inhibiting the excessive activation of microglia and production of pro-inflammatory factors, promoting the release of neuroprotective substances might be beneficial to the treatment of cerebral ischemia. The study about relationship between microglia and cerebral ischemia will shed a light on the treatment of cerebral ischemia. This paper is a review of microglial activation and regulation during cerebral ischemia as well as related therapeutic methods.
Animals
;
Brain Ischemia
;
metabolism
;
pathology
;
Class Ib Phosphatidylinositol 3-Kinase
;
metabolism
;
Humans
;
Inflammation
;
metabolism
;
Microglia
;
cytology
;
drug effects
;
metabolism
;
physiology
;
Neuroprotective Agents
;
pharmacology
;
Nitric Oxide Synthase
;
metabolism
;
Receptors, Purinergic P2X7
;
metabolism
;
Regeneration
;
TNF-Related Apoptosis-Inducing Ligand
;
metabolism
;
Toll-Like Receptors
;
metabolism
10.Increased leakage of brain antigens after traumatic brain injury and effect of immune tolerance induced by cells on traumatic brain injury.
Hua YAN ; Hong-Wei ZHANG ; Qiao-Li WU ; Guo-Bin ZHANG ; Kui LIU ; Da-Shi ZHI ; Zhen-Bo HU ; Xian-Wei ZENG
Chinese Medical Journal 2012;125(9):1618-1626
BACKGROUNDAlthough traumatic brain injury can lead to opening the blood-brain barrier and leaking of blood substances (including water) into brain tissue, few studies of brain antigens leaking into the blood and the pathways have been reported. Brain antigens result in damage to brain tissues by stimulating the immune system to produce anti-brain antibodies, but no treatment has been reported to reduce the production of anti-brain antibodies and protect the brain tissue. The aim of the study is to confirm the relationship between immune injury and arachnoid granulations following traumatic brain injury, and provide some new methods to inhibit the immune injury.
METHODSIn part one, methylene blue was injected into the rabbits' cisterna magna after traumatic brain injury, and concentrations of methylene blue and tumor necrosis factor (TNF)-α in blood were detected to determine the permeability of arachnoid granulations. In part two, umbilical cord mesenchymal stem cells and immature dendritic cells were injected into veins, and concentrations of interleukin 1 (IL-1), IL-10, interferon (IFN)-γ, transforming growth factor (TGF)-β, anti-brain antibodies (ABAb), and IL-12 were measured by ELISA on days 1, 3, 7, 14 and 21 after injury, and the numbers of leukocytes in the blood were counted. Twenty-one days after injury, expression of glutamate in brain tissue was determined by immunohistochemical staining, and neuronal degeneration was detected by H&E staining.
RESULTSIn part one, blood concentrations of methylene blue and TNF-α in the traumatic brain injury group were higher than in the control group (P < 0.05). Concentrations of methylene blue and TNF-α in the trauma cerebrospinal fluid (CSF) injected group were higher than in the control cerebrospinal fluid injected group (P < 0.05). In part two, concentrations of IL-1, IFN-γ, ABAb, IL-12, expression of glutamate (Glu), neuronal degeneration and number of peripheral blood leukocytes were lower in the group with cell treatment compared to the control group. IL-10 and TGF-β were elevated compared to the control group.
CONCLUSIONSTraumatic brain injury can lead to stronger arachnoid granulations (AGs) permeability; umbilical cord mesenchymal stem cells and immature dendritic cells can induce immune tolerance and reduce inflammation and anti-brain antibodies to protect the brain tissue.
Adipocytes ; cytology ; Animals ; Antigens ; blood ; metabolism ; Brain Injuries ; blood ; cerebrospinal fluid ; metabolism ; Cell Differentiation ; physiology ; Cells, Cultured ; Dendritic Cells ; metabolism ; Enzyme-Linked Immunosorbent Assay ; Interleukin-1 ; blood ; cerebrospinal fluid ; Interleukin-10 ; blood ; cerebrospinal fluid ; Interleukin-12 ; blood ; cerebrospinal fluid ; Mesenchymal Stromal Cells ; cytology ; Methylene Blue ; metabolism ; Osteoblasts ; cytology ; Rabbits ; Transforming Growth Factor beta ; blood ; cerebrospinal fluid ; Tumor Necrosis Factor-alpha ; blood ; cerebrospinal fluid

Result Analysis
Print
Save
E-mail