Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Neuroscience ; 536: 131-142, 2024 Jan 09.
Article En | MEDLINE | ID: mdl-37993087

OBJECTIVE: GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPARs) inserted into postsynaptic membranes are key to the process of long-term potentiation (LTP). Some evidence has shown that 4.1N plays a critical role in the membrane trafficking of AMPARs. However, the underlying mechanism behind this is still unclear. We investigated the role of 4.1N-mediated membrane trafficking of AMPARs during theta-burst stimulation long-term potentiation (TBS-LTP), to illustrate the molecular mechanism behind LTP. METHODS: LTP was induced by TBS in rat hippocampal CA1 neuron. Tat-GluA1 (MPR), which disrupts the association of 4.1N-GluA1, and autocamtide-2-inhibitory peptide, myristoylated (Myr-AIP), a CaMKII antagonist, were used to explore the role of 4.1N in the AMPARs trafficking during TBS-induced LTP. Immunoprecipitation (IP) and immunoblotting (IB)were used to detect protein expression, phosphorylation, and the interaction of p-CaMKII-4.1N-GluA1. RESULTS: We found that Myr-AIP attenuated increases of p-CaMKII (T286), p-GluA1 (ser831), and 4.1N phosphorylation after TBS-LTP, and decreased the association of p-CaMKII-4.1N-GluA1, along with the expression of GluA1, at postsynaptic densities during TBS-LTP. We also designed interfering peptides to disrupt the interaction between 4.1N and GluA1, which showed that Tat-GluA1 (MPR) or Myr-AIP inhibited TBS-LTP and attenuated increases of GluA1 at postsynaptic sites, while Tat-GluA1 (MPR) or Myr-AIP had no effects on miniature excitatory postsynaptic currents (mEPSCs) in non-stimulated hippocampal CA1 neurons. CONCLUSION: Active CaMKII enhanced the phosphorylation of 4.1N and facilitated the association of p-CaMKII with 4.1N-GluA1, which in turn resulted in GluA1 trafficking during TBS-LTP. The association of 4.1N-GluA1 is required for LTP, but not for basal synaptic transmission.


Long-Term Potentiation , Receptors, AMPA , Animals , Rats , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Phosphorylation , Receptors, AMPA/metabolism , Synapses/metabolism
2.
Neural Regen Res ; 18(6): 1286-1292, 2023 Jun.
Article En | MEDLINE | ID: mdl-36453413

Although bone marrow mesenchymal stem cells (BMSCs) might have therapeutic potency in ischemic stroke, the benefits are limited. The current study investigated the effects of BMSCs engineered to overexpress vascular endothelial growth factor (VEGF) on behavioral defects in a rat model of transient cerebral ischemia, which was induced by middle cerebral artery occlusion. VEGF-BMSCs or control grafts were injected into the left striatum of the infarcted hemisphere 24 hours after stroke. We found that compared with the stroke-only group and the vehicle- and BMSCs-control groups, the VEGF-BMSCs treated animals displayed the largest benefits, as evidenced by attenuated behavioral defects and smaller infarct volume 7 days after stroke. Additionally, VEGF-BMSCs greatly inhibited destruction of the blood-brain barrier, increased the regeneration of blood vessels in the region of ischemic penumbra, and reducedneuronal degeneration surrounding the infarct core. Further mechanistic studies showed that among all transplant groups, VEGF-BMSCs transplantation induced the highest level of brain-derived neurotrophic factor. These results suggest that BMSCs transplantation with vascular endothelial growth factor has the potential to treat ischemic stroke with better results than are currently available.

3.
Colloids Surf B Biointerfaces ; 213: 112419, 2022 May.
Article En | MEDLINE | ID: mdl-35227994

In native bone tissue regeneration, blood vessels, providing oxygen and nutrition for tissues, can promote the regeneration of bone and accelerate the repair of a defected area. In this study, Poly(D, L-lactic-co-glycolic acid) (PLGA) inverse opal scaffolds with high pore interconnectivity were fabricated and further modified with vascular endothelial growth factor (VEGF). The rat bone marrow derived mesenchymal stem cells (rMSCs) and human umbilical vein endothelial cells (HUVECs) were co-cultured onto the scaffolds to enhance vascularization for bone tissue regeneration. Cell attachment, viability, proliferation, and morphology were detected by cell counting kit-8 (CCK-8) assay, live and dead staining and scanning electron microscopy (SEM). Hydrostatic pressure with 0-279 KPa and 1 Hz one hour per day for 7 days was applied to tissue engineered bone constructs to investigate whether the loading stimulation can promote osteogenesis and angiogenesis mutually evaluated in parallel by multiple in vitro assays and in an in vivo chicken chorioallantoic membrane (CAM) model. The results indicated that the immobilization of VEGF can improve biocompatibility of PLGA scaffolds and promote cell attachment and proliferation. The cell-scaffold constructs showed higher CD31 expression because of the angiogenic differentiation of rMSCs in hydrostatic loading culture condition in vitro. The in vivo CAM model experiment demonstrated that hydrostatic loading stimulated angiogenic differentiation of rMSCs can accelerate tubulogenesis. Furthermore, the new capillaries formed in cell-scaffold constructs were conducive to calcium deposition in vivo.


Osteogenesis , Vascular Endothelial Growth Factor A , Animals , Coculture Techniques , Human Umbilical Vein Endothelial Cells , Humans , Hydrostatic Pressure , Lactic Acid , Neovascularization, Pathologic , Porosity , Rats , Tissue Engineering/methods , Tissue Scaffolds , Vascular Endothelial Growth Factor A/pharmacology
4.
Free Radic Res ; 55(11-12): 1104-1118, 2021 Dec.
Article En | MEDLINE | ID: mdl-34979841

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are the production of renal ischemia/reperfusion (I/R). The current study is to elucidate a mechanism of SIRT2 tyrosine nitration to accelerate the cell apoptosis induced by peroxynitrite (ONOO‾), the most reactive and deleterious RNS type in renal ischemia/reperfusion (I/R) injury. Our results demonstrate that there is a significant enhancement of the 3-nitrotyrosine levels in renal tissues of Acute Kidney Injury (AKI) patients and rats that underwent renal I/R, and a positive correlation between the 3-nitrotyrosine level and renal function impairment, indicative of an accumulation of peroxynitrite. Notably, peroxynitrite-evoked nitration of SIRT2 destroyed its enzymatic activity and the capability to deacetylate FOXO3a, and enhanced expression of Bim and caspase3, facilitating renal cell apoptosis in renal ischemia/reperfusion and SIN-1(peroxynitrite donor) treatment in vitro, and these effects were reversed by FeTMPyP, a peroxynitrite decomposition scavenger. Importantly, we identified that the tyrosine 86 is responsible for SIRT2 nitration and inactivation using site-mutation assay and Mass Spectrography analysis. Altogether, these findings point to a novel protective mechanism that an inhibition of SIRT2 tyrosine nitration can be a promising strategy to prevent ischemic renal diseases involving AKI.


Acute Kidney Injury , Reperfusion Injury , Animals , Humans , Ischemia , Kidney/physiology , Peroxynitrous Acid/pharmacology , Rats , Reperfusion Injury/metabolism , Sirtuin 2 , Tyrosine
5.
Comput Math Methods Med ; 2020: 8741739, 2020.
Article En | MEDLINE | ID: mdl-32908583

Neonatal sepsis is one of the most prevalent causes of death of the neonates. However, the mechanisms underlying neonatal sepsis remained unclear. The present study identified a total of 1128 upregulated mRNAs and 1008 downregulated mRNAs, 28 upregulated lncRNAs, and 61 downregulated lncRNAs in neonatal sepsis. Then, we constructed PPI networks to identify key regulators in neonatal sepsis, including ITGAM, ITGAX, TLR4, ITGB2, SRC, ELANE, RPLP0, RPS28, RPL26, and RPL27. lncRNA coexpression analysis showed HS.294603, LOC391811, C12ORF47, LOC729021, HS.546375, HNRPA1L-2, LOC158345, and HS.495041 played important roles in the progression of neonatal sepsis. Bioinformatics analysis showed DEGs were involved in the regulation cellular extravasation, acute inflammatory response, macrophage activation of NF-kappa B signaling pathway, TNF signaling pathway, HIF-1 signaling pathway, Toll-like receptor signaling pathway, and ribosome, RNA transport, and spliceosome. lncRNAs were involved in regulating ribosome, T cell receptor signaling pathway, RNA degradation, insulin resistance, ribosome biogenesis in eukaryotes, and hematopoietic cell lineage. We thought this study provided useful information for identifying novel therapeutic markers for neonatal sepsis.


Neonatal Sepsis/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Computational Biology , Disease Progression , Gene Expression Profiling , Genetic Markers , Humans , Infant, Newborn , Mathematical Concepts , Protein Interaction Maps/genetics
6.
J Biomater Sci Polym Ed ; 30(2): 150-161, 2019 02.
Article En | MEDLINE | ID: mdl-30556784

Having advantageous biocompatibility and osteoconductive properties known to enhance the osteogenic differentiation of mesenchymal stem cells (MSCs), hydroxyapatite (HA) is a commonly used material for bone tissue engineering. What remains unclear, however, is whether HA holds a similar potential for stimulating the osteogenic differentiation of MSCs to that of a more frequently used osteogenic-inducing medium (OIM). To that end, we used PHBV electrospun nanofibrous scaffolds to directly compare the osteogenic capacities of HA with OIM over MSCs. Through the observation of cellular morphology, the staining of osteogenic markers, and the quantitative measuring of osteogenic-related genes, as well as microRNA analyses, we not only found that HA was as capable as OIM for differentiating MSCs down an osteogenic lineage; albeit, at a significantly slower rate, but also that numerous microRNAs are involved in the osteogenic differentiation of MSCs through multiple pathways involving the inhibition of cellular proliferation and stemness, chondrogenesis and adipogenesis, and the active promotion of osteogenesis. Taken together, we have shown for the first time that PHBV electrospun nanofibrous scaffolds combined with HA have a similar osteogenic-inducing potential as OIM and may therefore be used as a viable replacement for OIM for alternative in vivo-mimicking bone tissue engineering applications.


Cell Differentiation/drug effects , Durapatite/metabolism , Mesenchymal Stem Cells/drug effects , Nanofibers/chemistry , Osteogenesis/drug effects , Polyesters/chemistry , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Durapatite/chemistry , Extracellular Matrix/drug effects , Gene Expression Regulation/drug effects , Humans , MicroRNAs/metabolism , Polyesters/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Tissue Engineering , Tissue Scaffolds/chemistry
7.
Neurosci Lett ; 666: 85-91, 2018 02 14.
Article En | MEDLINE | ID: mdl-29274438

This study aimed to evaluate the effect of bone mesenchymal stem cells (BMSCs) and BMSC neural-like cells (BMSC-Ns) on the spinal cord injury (SCI) in the rat model of SCI. BMSC-Ns were prepared from the third passage of BMSCs by induction of healthy cerebrospinal fluid (CSF) of an adult human. The SCI rat model was established through a surgical procedure, and after 7 days the rats were randomly divided into 3 (A, B and C) groups. Groups A (BMSC-Ns) and B (BMSCs) were treated with 1 × 106/20 µl cells, while group C (saline) was treated with saline, all via intracerebroventricular injection. After transplantation, the BBB score of group A was significantly higher than that of group B, which in turn was significantly higher than that of group C (P < .05). The levels of Bdnf, Ngf, Ntf3 were statistically significantly higher in group A than those in groups B and C (P < .05). The levels of 5-HT, NA, Ach, DA, GABA in group A were significantly higher than those in groups B and C, whereas the level of Glu was significantly lower in group A than that in groups B and C (P < .05). The histopathological data showed remarkably less necrosis of the spinal cord in group A, compared to that in groups B and C. Transplanting BMSC-Ns or BMSCs into the lateral ventricles improved the neurological function of rats with SCI. Moreover, BMSC-Ns were significantly more effective than BMSCs, which provides a possible approach for the treatment of SCI.


Bone Marrow Cells/pathology , Brain-Derived Neurotrophic Factor/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries/therapy , Animals , Bone Marrow/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Transplantation/methods , Cerebrospinal Fluid/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord Injuries/pathology
8.
Neuromolecular Med ; 19(4): 541-554, 2017 Dec.
Article En | MEDLINE | ID: mdl-28916896

The pro-inflammatory activity of interleukin 17, which is produced by the IL-23/IL-17 axis, has been associated with the pathogenesis of traumatic brain injury (TBI). The study investigated the potential role of IL-17 in secondary brain injury of TBI in a rat model. Our data showed that the levels of IL-17 increased from 6 h to 7 days and peaked at 3 days, in both the CNS and serum, which were consistent with the severity of secondary brain injury. The IL-23 inhibitor suberoylanilide hydroxamic acid (SAHA) treatment markedly decreased the expressions of IL-17 and apoptosis-associated proteins cleaved caspase-3 and increased the protein ratio of Bcl-2 (B cell lymphoma/leukemia-2)/Bax (Bcl-2-associated X protein). Meanwhile, neuronal apoptosis was reduced, and neural function was improved after SAHA treatment. This study suggests that IL-17 is involved in secondary brain injury after TBI. Administering an IL-23 inhibitor and thereby blocking the IL-23/IL-17 axis may be beneficial in the treatment of TBI.


Brain Damage, Chronic/physiopathology , Brain Injuries, Traumatic/physiopathology , Interleukin-17/physiology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/prevention & control , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Down-Regulation/drug effects , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Inflammation , Interleukin-17/blood , Interleukin-17/cerebrospinal fluid , Interleukin-17/genetics , Interleukin-23/antagonists & inhibitors , Interleukin-23/physiology , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/blood , Nerve Tissue Proteins/cerebrospinal fluid , Nerve Tissue Proteins/genetics , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors , Vorinostat
9.
Front Neurol ; 7: 183, 2016.
Article En | MEDLINE | ID: mdl-27833584

Bone marrow mesenchymal stem cells (BMSCs) represent a promising tool for stem cell-based therapies. However, the majority of BMSC transplants only allow for limited recovery of the lost functions. We previously found that human cerebrospinal fluid (hCSF) is more potent than growth factors in differentiating human BMSCs into neuron-like cells in vitro. In this study, we studied the effect of transplantation of rat BMSC-derived neuron-like cells (BMSC-Ns) induced by hCSF into rat brain with middle cerebral artery occlusion (MCAO). The survival and differentiation of the transplanted cells were determined using immunofluorescence staining of bromodeoxyuridine. The recovery of neurological function were observed by the modified neurological severity score (modified NSS) at 4, 15, and 32 days after cell transplantation, HE staining for determination of the infarct volume at day 32 after cell transplantation. Transplantation of BMSC-Ns or BMSCs significantly improved indexes of neurological function and reduced infarct size in rats previously subjected to MCAO compared with those in the control group. Remarkably, 32 days after transplantation, rats treated with BMSC-Ns presented a smaller infarct size, higher number of neuron-specific, enolase-positive, and BrdU-positive cells, and improved neurological function compared with BMSC group. Our results demonstrate that transplantation of hCSF-treated BMSC-Ns significantly improves neurological function and reduces infarct size in rats subjected to MCAO. This study may pave a new avenue for the treatment of MCAO.

10.
Neurosci Lett ; 597: 90-6, 2015 Jun 15.
Article En | MEDLINE | ID: mdl-25929187

Previous studies have demonstrated that activation of proline-rich tyrosine kinase 2 (PYK2) in cerebral ischemia is involved in the modulation of N-methyl-d-aspartate-type (NMDA) glutamate receptor activity and Ca(2+) dynamics, resulting in ischemic neuron death ultimately. A number of reports indicate that PYK2 is a redox sensitive kinase that must be activated by an estrogen-induced reactive oxygen species (ROS). However, the mechanism of PYK2 activation remains incompletely illustrated. Accumulating attention is focused on nitric oxide (NO, a free radical) which plays a critical role in cellular signal transduction through stimulus-coupled S-nitrosylation of cysteine residues. Here we reported that PYK2 over-expressed in human embryonic kidney (HEK293) cells was S-nitrosylated (forming SNO-PYK2) by reacting with GSNO, an exogenous NO donor, at one critical cysteine residue (Cys534) with a biotin switch assay. Moreover, our results showed that S-nitrosylation and phosphorylation of PYK2 over-expressed in SH-SY5Y cells was significantly increased after oxygen-glucose deprivation (OGD). We further investigated whether the activation (phosphorylation) of PYK2 was associated with S-nitrosylation following SH-SY5Y cells OGD. Our results showed that the cysteine534 residue (site of S-nitrosylation) mutant PYK2 over-expressed in SH-SY5Y cells diminished S-nitrosylation of PYK2 and inhibited its phosphorylation induced by OGD. In addition, overexpression of the mutant PYK2 protein could prevent nuclear accumulation and abrogate neuronal cell death compared to wild type PYK2 in SH-SY5Y cells induced by OGD. These data suggest that the activation of PYK2 following OGD may be modulated by S-nitrosylation, which provides a new avenue for stroke therapy by targeting the post-translational modification machinery.


Focal Adhesion Kinase 2/metabolism , Glucose/deficiency , Nitric Oxide/metabolism , Oxygen/metabolism , Cell Hypoxia , Cell Line, Tumor , Enzyme Activation , Humans , Nitric Oxide Donors/metabolism , Nitric Oxide Synthase Type I/metabolism , S-Nitrosoglutathione/metabolism
11.
Mol Cell Biochem ; 365(1-2): 363-77, 2012 Jun.
Article En | MEDLINE | ID: mdl-22422045

Previous studies suggested that activated c-Src promote the tyrosine phosphorylation of NMDA receptor subunit NR2A, and thus aggravate the injury induced by transient cerebral ischemia/reperfusion (I/R) in rat hippocampus CA1 region. In this study, we examined the effect of nitric oxide (NO) on the activation of c-Src and the tyrosine phosphorylation of NMDA receptor NR2A subunit. The results show that S-nitrosylation and the phosphorylation of c-Src were induced after cerebral I/R in rats, and administration of nNOS inhibitor 7-NI, nNOS antisense oligonucleotides and exogenous NO donor sodium nitroprusside diminished the increased S-nitrosylation and phosphorylation of c-Src during cerebral I/R. The cysteine residues of c-Src modified by S-nitrosylation are Cys489, Cys498, and Cys500. On the other hand, NMDAR antagonist MK-801 could attenuate the S-nitrosylation and activation of c-Src. Taken together, the S-nitrosylation of c-Src is provoked by NO derived from endogenous nNOS, which is activated by Ca(2+) influx from NMDA receptors, and promotes the auto-phosphorylation at tyrosines and further phosphorylates NR2A. The molecular mechanism we outlined here is a novel postsynaptic NMDAR-nNOS/c-Src-mediated signaling amplification, the 'NMDAR-nNOS â†’ NO â†’ SNO-c-Src â†’ p-c-Src â†’ NMDAR-nNOS' cycle, which presents the possibility as a potential therapeutic target for stroke treatment.


Brain Ischemia/enzymology , Enzyme Activation , Nitric Oxide Synthase Type I/metabolism , Protein Processing, Post-Translational , Receptors, N-Methyl-D-Aspartate/metabolism , Reperfusion Injury/enzymology , src-Family Kinases/metabolism , Amino Acid Motifs , Animals , Apoptosis , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cysteine/metabolism , Dizocilpine Maleate/pharmacology , HEK293 Cells , Hippocampus/blood supply , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Humans , Indazoles/pharmacology , Male , Neuroprotective Agents/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitroprusside/pharmacology , Phosphorylation , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , S-Nitrosoglutathione/pharmacology , src-Family Kinases/chemistry
12.
J Biol Chem ; 287(4): 2364-77, 2012 Jan 20.
Article En | MEDLINE | ID: mdl-22123824

Previous studies in our laboratory have shown that mixed lineage kinase 3 (MLK3) can be activated following global ischemia. In addition, other laboratories have reported that the activation of MLK3 may be linked to the accumulation of free radicals. However, the mechanism of MLK3 activation remains incompletely understood. We report here that MLK3, overexpressed in HEK293 cells, is S-nitrosylated (forming SNO-MLK3) via a reaction with S-nitrosoglutathione, an exogenous nitric oxide (NO) donor, at one critical cysteine residue (Cys-688). We further show that the S-nitrosylation of MLK3 contributes to its dimerization and activation. We also investigated whether the activation of MLK3 is associated with S-nitrosylation following rat brain ischemia/reperfusion. Our results show that the administration of 7-nitroindazole, an inhibitor of neuronal NO synthase (nNOS), or nNOS antisense oligodeoxynucleotides diminished the S-nitrosylation of MLK3 and inhibited its activation induced by cerebral ischemia/reperfusion. In contrast, 2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine (an inhibitor of inducible NO synthase) or nNOS missense oligodeoxynucleotides did not affect the S-nitrosylation of MLK3. In addition, treatment with sodium nitroprusside (an exogenous NO donor) and S-nitrosoglutathione or MK801, an antagonist of the N-methyl-D-aspartate receptor, also diminished the S-nitrosylation and activation of MLK3 induced by cerebral ischemia/reperfusion. The activation of MLK3 facilitated its downstream protein kinase kinase 4/7 (MKK4/7)-JNK signaling module and both nuclear and non-nuclear apoptosis pathways. These data suggest that the activation of MLK3 during the early stages of ischemia/reperfusion is modulated by S-nitrosylation and provides a potential new approach for stroke therapy whereby the post-translational modification machinery is targeted.


Brain Ischemia/enzymology , MAP Kinase Kinase Kinases/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Dizocilpine Maleate/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/genetics , Male , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , S-Nitrosoglutathione/metabolism , Thiazines/pharmacology , Mitogen-Activated Protein Kinase Kinase Kinase 11
13.
PLoS One ; 7(12): e52788, 2012.
Article En | MEDLINE | ID: mdl-23285183

Our laboratory once reported that neuronal nitric oxide synthase (nNOS) S-nitrosylation was decreased in rat hippocampus during cerebral ischemia-reperfusion, but the underlying mechanism was unclear. In this study, we show that nNOS activity is dynamically regulated by S-nitrosylation. We found that overexpressed nNOS in HEK293 (human embryonic kidney) cells could be S-nitrosylated by exogenous NO donor GSNO and which is associated with the enzyme activity decrease. Cys(331), one of the zinc-tetrathiolate cysteines, was identified as the key site of nNOS S-nitrosylation. In addition, we also found that nNOS is highly S-nitrosylated in resting rat hippocampal neurons and the enzyme undergos denitrosylation during the process of rat brain ischemia/reperfusion. Intrestingly, the process of nNOS denitrosylation is coupling with the decrease of nNOS phosphorylation at Ser(847), a site associated with nNOS activation. Further more, we document that nNOS denitrosylation could be suppressed by pretreatment of neurons with MK801, an antagonist of NMDAR, GSNO, EGTA, BAPTA, W-7, an inhibitor of calmodulin as well as TrxR1 antisense oligonucleotide (AS-ODN) respectively. Taken together, our data demonstrate that the denitrosylation of nNOS induced by calcium ion influx is a NMDAR-dependent process during the early stage of ischemia/reperfusion, which is majorly mediated by thioredoxin-1 (Trx1) system. nNOS dephosphorylation may be induced by the enzyme denitrosylation, which suggest that S-nitrosylation/denitrosylation of nNOS may be an important mechanism in regulating the enzyme activity.


Nitric Oxide Synthase Type I/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Apoptosis , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Calcium/metabolism , Cystine/metabolism , Enzyme Activation , HEK293 Cells , Humans , Male , Neurons/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/chemistry , Phosphorylation , Rats , Reperfusion Injury/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism
14.
Brain Res ; 1328: 162-70, 2010 Apr 30.
Article En | MEDLINE | ID: mdl-20197063

Over-activation of NMDA receptor has been widely believed to be the main signal resulting in ischemic cell injury. We recently reported that the triplicate complex NR2A-PSD-95-Src is a signaling module to facilitate NMDA receptor over-activation. In addition, over-activation of NMDA receptor can activate another signaling molecule nNOS, which is also mediated by PSD-95 after cerebral ischemia. Here, we examined whether overexpression of the PDZ1 domain of PSD-95 could disrupt the functional interaction between NMDA receptor and PSD-95 in rat hippocampal CA1 region, and whether or not it could exert a neuroprotective effect against cerebral ischemia. Our results showed that overexpression of PDZ1 domain not only decreased the assembly of NR2A-PSD-95-Src signaling module and the auto-phosphorylation of Src, which mediates NMDA receptor phosphorylation, but also enhanced nNOS (Ser847) phosphorylation. Most importantly, overexpression of PDZ1 domain protected rat hippocampal CA1 neurons against cerebral ischemia injury. These results suggest that overexpression of the PDZ1 domain can perturb the binding of PSD-95 to NMDA receptor, suppress the activity of both NMDA receptor and nNOS, and thus have a neuroprotective effect.


Brain Ischemia/metabolism , Cytoprotection/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , src-Family Kinases/metabolism , Amino Acid Sequence/physiology , Animals , Brain Ischemia/physiopathology , Catalytic Domain/physiology , Disks Large Homolog 4 Protein , Down-Regulation/physiology , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Membrane Proteins/chemistry , Neurons/metabolism , Nitric Oxide/biosynthesis , Phosphorylation/physiology , Protein Structure, Tertiary/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism , Signal Transduction/physiology , Up-Regulation/physiology
15.
Hippocampus ; 20(1): 65-77, 2010 Jan.
Article En | MEDLINE | ID: mdl-19330848

alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are responsible for excitotoxicity induced by ischemic injury in hippocampal CA1 neurons, whereas the molecular mechanisms responsible for their neurotrophic activities are much less studied. Here, we examined the neuroprotective effect of positive modeulation of AMPARs by coapplication of AMPA with PEPA, an allosteric potentiator of AMPARs. We showed that coapplication of AMPA with PEPA protected hippocampal CA1 neurons from brain ischemia-induced death. Coapplication of AMPA with PEPA could prevent downregulated expression of GluR2 subunit caused by ischemia and increase BDNF expression via Lyn-ERK1/2-CREB signaling. Furthermore, TrkB receptor-mediated PI3K/Akt signal pathway was activated after coapplication of AMPA with PEPA, which was related to MAPK pathway and protected CA1 neurons against ischemic insults through depression of JNK3 activity, release of cytochrome c to cytosol and depression of capase-3 activity. Our results revealed that positive modulation of AMPARs could exert neuroprotective effects and the possible signaling pathways underlied.


Brain Ischemia/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, AMPA/metabolism , src-Family Kinases/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Gene Expression Regulation , MAP Kinase Signaling System , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Receptors, AMPA/genetics , Signal Transduction
16.
Redox Rep ; 14(6): 243-50, 2009.
Article En | MEDLINE | ID: mdl-20003709

Previous studies have reported that selenite, a known antioxidant, protects brain against ischemia/reperfusion injury, which is mediated by oxidative stress. The aim of this study was to investigate whether selenite can protect kidney against ischemic injury by reducing activation of the apoptosis signal regulating kinase 1 (ASK1)/mitogen-activated protein kinase kinase 3 (MKK3)/p38 mitogen-activated protein kinase signaling pathway. The activation and expression of ASK1, MKK3, p38, caspase 3 and cleaved PARP were analyzed by Western blot. Apoptosis of renal tubular epithelial cells was assessed by the terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling method. Malondialdehyde (MDA) levels were measured by the thiobarbituric acid reaction. Blood serum creatinine and blood urea nitrogen level were measured with an Olympus automatic multi-analyzer. We found that selenite attenuated significantly ASK1, MKK3, and p38 phosphorylation at 3 h after renal ischemia. Furthermore, selenite decreased significantly renal epithelial tubular cell apoptosis. In addition, selenite reduced the MDA level. These findings suggest that the protective action of selenite on ischemia renal injury is associated closely with reducing activation of the ASK1-MKK3-p38 signal pathway.


Kidney/drug effects , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Sodium Selenite/therapeutic use , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/physiology , Caspase 3/metabolism , Enzyme Activation , Humans , Kidney/metabolism , Kidney/pathology , Lipid Peroxidation/drug effects , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Male , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/physiology , Sodium Selenite/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Neurosci Res ; 87(16): 3626-38, 2009 Dec.
Article En | MEDLINE | ID: mdl-19610093

Recent studies have shown that kainate (KA) receptors are involved in neuronal cell death induced by seizure, which is mediated by the GluR6.PSD-95.MLK3 signaling module and subsequent JNK activation. In our previous studies, we demonstrated the neuroprotective role of a GluR6 c-terminus containing peptide against KA or cerebral ischemia-induced excitotoxicity in vitro and in vivo. Here, we first report that overexpression of the PDZ1 domain of PSD-95 protein exerts a protective role against neuronal death induced by cerebral ischemia-reperfusion in vivo and can prevent neuronal cell death induced by oxygen-glucose deprivation. Further studies show that overexpression of PDZ1 can perturb the interaction of GluR6 with PSD-95 and suppress the assembly of the GluR6.PSD-95.MLK3 signaling module and therefore inhibit JNK activation. Thus, it not only inhibits phosphorylation of c-Jun and down-regulates Fas ligand expression but also inhibits phosphorylation of 14-3-3 and decreases Bax translocation to mitochondria, decreases the release of cytochrome c, and decreases caspase-3 activation. Overall, the essential role of the PDZ1 domain of PSD-95 in apoptotic cell death in neurons provides an experimental foundation for gene therapy of neurodegenerative diseases with overexpression of the PDZ1 domain.


Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Neurons/metabolism , Reperfusion Injury/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Death/genetics , Cell Fractionation , Cell Line , Cells, Cultured , Cytochromes c/metabolism , Disks Large Homolog 4 Protein , Glucose/deficiency , Hippocampus/pathology , Humans , Hypoxia/genetics , Hypoxia/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Neurons/pathology , Phosphorylation/genetics , Protein Transport/genetics , Rats , Rats, Sprague-Dawley , Receptors, Kainic Acid/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction/physiology , Subcellular Fractions/metabolism , Subcellular Fractions/pathology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , GluK2 Kainate Receptor
18.
Neurosci Lett ; 460(2): 133-7, 2009 Aug 28.
Article En | MEDLINE | ID: mdl-19477222

In our previous studies, Tat-GluR6-9c (a glutamate receptor 6 C-terminus peptide fused the TAT protein transduction sequence) not only inhibited the activation of MLK3 (mixed lineage kinase 3) and JNK (c-Jun N-terminal kinase) via the GluR6.PSD-95 (postsynaptic density protein 95).MLK3 signaling module but also diminished neuronal death induced by kainic acid or transient cerebral ischemia in rat hippocampus. Here, we investigate whether overexpression of the PDZ1 domain of PSD-95 protein could suppress the binding of GluR6 with PSD-95 and the activation of MLK3, MKK7 (mitogen-activated kinase kinase 7) and JNK1/2, and rescused neuronal cell death induced by kainic acid. Our results showed that overexpression of the PDZ1 domain of PSD-95 protein could prevent nuclear accumulation and abrogate neuronal cell death in SD (Sprague-Dawley) rat hippocampal neuronal cells. Further studies indicated that overexpression of PDZ1 could inhibit the enhancement of binding of GluR6 to PSD-95 and prevent the activation of MLK3, MKK7 and JNK1/2 induced by kainic acid. Taken together, the essential role of the PDZ1 domain of PSD-95 in apoptotic cell death in neurons provides an experimental foundation for gene therapy of neurodegenerative diseases with overexpression of the PDZ1 domain.


Apoptosis/drug effects , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/cytology , Kainic Acid/pharmacology , Neurons/drug effects , PDZ Domains/physiology , Animals , Animals, Newborn , Apoptosis/genetics , Cells, Cultured , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Humans , MAP Kinase Kinase 7/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , PDZ Domains/genetics , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Time Factors , Transfection/methods , Mitogen-Activated Protein Kinase Kinase Kinase 11
19.
Mol Med Rep ; 2(6): 931-6, 2009.
Article En | MEDLINE | ID: mdl-21475923

This study was designed to investigate the repair of point mutations in the low density lipoprotein receptor (LDLR) gene mediated by single-stranded oligonucleotides (SSOs) in vivo. Mutations in the LDLR gene are known to be the prime cause of familial hypercholesterolemia (FH). SSOs result in sequence-specific alterations leading to the correction of mutations. In the present study, the LDLR gene with a nonsense mutation (c660x) was fused to a luciferase reporter gene (p660-LDLR-luc) and introduced into mouse liver by hydrodynamic gene transfer. These mice were then injected via the tail vein with different SSOs complexed with polyethylenimine. Firefly luciferase activity present in hepatic cell lysate was measured to analyze repair efficiency. Restriction fragment length polymorphism analysis and direct sequencing were performed to affirm that the LDLR mutation was corrected. The results indicate that the LDLR mutation was corrected in the liver in vivo only in the presence of antisense SSOs (anti-SSOs). Our findings provide initial evidence that the point mutation in p660-LDLR-luc can be corrected by anti-SSO targeted repair in vivo. This may be a potential strategy for the treatment of FH.

20.
Sheng Li Xue Bao ; 60(6): 730-6, 2008 Dec 25.
Article Zh | MEDLINE | ID: mdl-19082428

To detect the effect of PDZ1, domain of postsynaptic density 95 (PSD-95), on apoptosis of hippocampal neurons induced by oxygen-glucose deprivation (OGD), Sprague-Dawley rat hippocampal neurons were infected with PDZ1-viruses after 21 days of plating. Twenty-four hours after infection, cells were treated with OGD for 1.5 h, then were incubated with DAPI and apoptosis-like cells were characterized, or were collected for co-immunoprecipitation and Western blot analyses. The results showed that: (1) PDZ1 overexpression was observed in hippocampal neurons; (2) Apoptosis induced by OGD was obviously decreased in neurons overexpressing PDZ1 (P<0.05); (3) Overexpression of PDZ1 prevented the binding of GluR6 to PSD-95; (4) Overexpression of PDZ1 inhibited MLK3 and JNK1/2 activation induced by OGD. These results indicate that overexpression of PDZ1 may prevent hippocampal neurons from apoptosis induced by OGD.


Apoptosis , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurons/cytology , PDZ Domains , Animals , Cells, Cultured , Culture Media/chemistry , Disks Large Homolog 4 Protein , Glucose/chemistry , Hippocampus/cytology , Neurons/pathology , Oxygen/chemistry , Rats , Rats, Sprague-Dawley
...