Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
3.
Exp Neurol ; 371: 114585, 2024 01.
Article En | MEDLINE | ID: mdl-37884185

AIMS: Osteopontin (OPN) has demonstrated neuroprotective effects in various stroke models. Its role in neuroinflammation after brain injury remains to be elucidated. This study aims to clarify the effect of OPN on neuroinflammation, particularly on the functional states of microglia after subarachnoid hemorrhage (SAH). METHODS: 77 rats were randomly divided into the following groups: Sham, SAH 24 h, SAH + rOPN, SAH + Vehicle (PBS), SAH + OPN siRNA, and SAH + Scr siRNA, SAH + rOPN+Fib-14 and SAH + rOPN+DMSO. Modified Garcia and beam balance tests were used to evaluate neurobehavioral outcomes. Semi-quantitative immunofluorescence staining was performed to measure expression of myeloperoxidase (MPO) and microglia activation state markers CD16, CD206 after SAH and recombinant OPN treatment. The quantification of microglia activation and functional markers CD16, CD206, TNF-α and IL-10 were further evaluated using Western-blotting. RESULTS: Nasal administration of rOPN improved neurological dysfunction, attenuated neutrophil infiltration, and decreased expression of phenotypic and functional markers of pro-inflammatory microglia CD16 and TNF-α. It also promoted an anti-inflammatory microglial state, as evidenced by increased expression of CD206 and IL-10. Furthermore, after blocking the phosphorylation of FAK signaling, the effects of rOPN on microglial activation states were partially reversed. The downstream pathways of STAT3 and NF-κB also exhibited consistent changes, suggesting the involvement of the STAT3 and NF-κB pathways in OPN's modulation of microglial activation via integrin-FAK signaling. CONCLUSION: OPN attenuates inflammatory responses after SAH by promoting an anti-inflammatory microglial state, potentially mediated through the integrin-FAK-STAT3 and NF-κB signaling pathways.


Osteopontin , Subarachnoid Hemorrhage , Rats , Animals , Osteopontin/therapeutic use , Osteopontin/metabolism , Osteopontin/pharmacology , Rats, Sprague-Dawley , NF-kappa B/metabolism , Interleukin-10 , Microglia/metabolism , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Tumor Necrosis Factor-alpha/metabolism , Neuroinflammatory Diseases , Anti-Inflammatory Agents/pharmacology , Integrins/metabolism , Integrins/therapeutic use , RNA, Small Interfering/pharmacology , Disease Models, Animal
4.
Exp Neurol ; 342: 113752, 2021 08.
Article En | MEDLINE | ID: mdl-33974879

Autophagy is a crucial pathological process in early brain injury (EBI) after subarachnoid hemorrhage (SAH). In this study, we investigated the role of dihydrolipoic acid (DHLA) on enhancing autophagy and alleviating neurological deficits after SAH. SAH was induced by endovascular perforation in male Sprague-Dawley rats. DHLA (30 mg/kg) was administered intraperitoneally 1 h (h) after SAH. Small interfering ribonucleic acid (siRNA) for lysosome-associated membrane protein-1 (LAMP1) was administered through intracerebroventricular (i.c.v) route 48 h before SAH induction. SAH grading score, neurological score, immunofluorescence staining, Fluoro-Jade C (FJC) staining, and Western blot were examined. DHLA treatment increased autophagy-related protein expression and downregulated the apoptosis-related protein expression 24 h after SAH. In addition, the DHLA treatment reduced neuronal cell death and alleviated neurological deficits after SAH. Furthermore, knockdown of LAMP1 abolished the neuroprotective effects of DHLA. These results indicate that LAMP1 may participate in autophagy after SAH. DHLA treatment can enhance autophagy, attenuate apoptosis, and alleviate neurofunctional deficits in EBI after SAH. It may provide an effective alternative method for the treatment of EBI after SAH.


Antioxidants/therapeutic use , Autophagy/drug effects , Nervous System Diseases/drug therapy , Subarachnoid Hemorrhage/drug therapy , Thioctic Acid/analogs & derivatives , Animals , Antioxidants/pharmacology , Autophagy/physiology , Male , Nervous System Diseases/etiology , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/physiopathology , Thioctic Acid/pharmacology , Thioctic Acid/therapeutic use
5.
Front Mol Neurosci ; 14: 750726, 2021.
Article En | MEDLINE | ID: mdl-35002615

In this study, we investigated the role of Exendin-4 (Ex-4), a glucagon-like peptide 1 receptor (GLP-1R) agonist, in blood-brain barrier (BBB) disruption after subarachnoid hemorrhage (SAH) in rats. The endovascular perforation model of SAH was performed in Sprague-Dawley rats. Ex-4 was intraperitoneally injected 1 h after SAH induction. To elucidate the underlying molecular mechanism, small interfering ribonucleic acid (siRNA) for GLP-1R and Dorsomorphin, a specific inhibitor of adenosine monophosphate-activated protein kinase (AMPK), were intracerebroventricularly injected 48 h before induction of SAH correspondingly. Immunofluorescence results supported GLP-1R expressed on the endothelial cells of microvessels in the brain after SAH. Administration of Ex-4 significantly reduced brain water content and Evans blue extravasation in both hemispheres, which improved neurological scores at 24 h after SAH. In the mechanism study, Ex-4 treatment significantly increased the expression of GLP-1R, p-AMPK, IκB-α, Occludin, and Claudin-5, while the expression of p-nuclear factor-kappa B (NF-κB) p65, matrix metalloproteinase-9 (MMP-9), and albumin was significantly decreased. The effects of Ex-4 were reversed by the intervention of GLP-1R siRNA or Dorsomorphin, respectively. In conclusion, Ex-4 could preserve the BBB integrity through GLP-1R/AMPK-dependent NF-κB/MMP-9 inhibition after SAH, which should be further investigated as a potential therapeutic target in SAH.

6.
Exp Neurol ; 326: 113179, 2020 04.
Article En | MEDLINE | ID: mdl-31930990

Subarachnoid hemorrhage (SAH) is the most devastating form of stroke. Reducing neuronal apoptosis is an important countermeasure against early brain injury (EBI) after SAH. Recent evidence indicates that OX40-OX40L coupling is critical for cell survival and proliferation. Current study was performed to detect the role of recombinant OX40 (ReOX40) against neuronal apoptosis after SAH. The endovascular perforation model of SAH was performed on Sprague-Dawley (SD) rats. ReOX40 was injected intracerebroventricularly (i.c.v) 1 h after SAH induction and the following methods were employed: neurological function evaluation, immunofluorescence staining, fluoro-Jade C staining, and western blot. To study the underlying precise molecular mechanism, small interfering ribonucleic acid (siRNA) for OX40L and a specific inhibitor of PI3K, LY294002, were injected i.c.v. into SAH + ReOX40 rats before induction of SAH. When compared with sham rats, the expression of OX40 and OX40L was seen to decrease in the brain at 24 h after SAH induction. Administration of ReOX40 (5 µg/kg) increased expression of the OX40L, reduced the neuronal apoptosis, and improved short and long-term neurological function deficits. Furthermore, ReOx40 heightened activation of OX40L/PI3K/AKT axis, increased the downstream anti-apoptotic protein (Bcl2, Bcl-XL), and depressed the apoptotic protein (cleaved caspase 3, Bax). However, the protective effects of ReOX40 were abolished by the administration of OX40L siRNA and LY294002, respectively. These results demonstrate that ReOX40 attenuates neuronal apoptosis through OX40-OX40L/PI3K/AKT pathway in EBI after SAH.


Apoptosis/drug effects , Neurons/drug effects , Receptors, OX40/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/genetics , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Injections, Intraventricular , Male , Membrane Glycoproteins/antagonists & inhibitors , Oncogene Protein v-akt/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase Inhibitors/therapeutic use , RNA, Small Interfering/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, OX40/pharmacology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Tumor Necrosis Factors
7.
Cells ; 8(9)2019 08 27.
Article En | MEDLINE | ID: mdl-31461955

Osteopontin (OPN) enhances autophagy, reduces apoptosis, and attenuates early brain injury (EBI) after a subarachnoid hemorrhage (SAH). A total of 87 Sprague-Dawley rats were subjected to sham or SAH operations to further investigate the signaling pathway involved in osteopontin-enhanced autophagy during EBI, and the potential effect of recombinant OPN (rOPN) administration to improve long-term outcomes after SAH. Rats were randomly divided into five groups: Sham, SAH + Vehicle (PBS, phosphate-buffered saline), SAH + rOPN (5 µg/rat recombinant OPN), SAH + rOPN + Fib-14 (30 mg/kg of focal adhesion kinase (FAK) inhibitor-14), and SAH + rOPN + DMSO (dimethyl sulfoxide). Short-term and long-term neurobehavior tests were performed, followed by a collection of brain samples for assessment of autophagy markers in neurons, pathway proteins expression, and delayed hippocampal injury. Western blot, double immunofluorescence staining, Nissl staining, and Fluoro-Jade C staining assay were used. Results showed that rOPN administration increased autophagy in neurons and improved neurobehavior in a rat model of SAH. With the administration of FAK inhibitor-14 (Fib-14), neurobehavioral improvement and autophagy enhancement induced by rOPN were abolished, and there were consistent changes in the phosphorylation level of ERK1/2. In addition, early administration of rOPN in rat SAH models improved long-term neurobehavior results, possibly by alleviating hippocampal injury. These results suggest that FAK-ERK signaling may be involved in OPN-enhanced autophagy in the EBI phase after SAH. Early administration of rOPN may be a preventive and therapeutic strategy against delayed brain injury after SAH.


Autophagy , Brain Injuries/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Osteopontin/metabolism , Subarachnoid Hemorrhage/metabolism , Animals , Disease Models, Animal , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Male , Osteopontin/administration & dosage , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism
8.
CNS Neurosci Ther ; 25(10): 1162-1172, 2019 10.
Article En | MEDLINE | ID: mdl-31436915

AIM: To determine the effect of osteopontin (OPN) on autophagy and autophagy-apoptosis interactions after SAH. METHODS: The endovascular perforation model of SAH or sham surgery was performed in a total of 86 Sprague-Dawley male rats. The temporal expressions of endogenous OPN and autophagy-related proteins (Beclin 1, ATG5, LC3 II to I ratio) were measured in sham and SAH rats at different time points (3, 6, 12, 24, and 72 hours). Rats were randomly divided into three groups: Sham, SAH + Vehicle (PBS, phosphate-buffered saline), and SAH + rOPN (5 µg/rat recombinant OPN). Neurobehavioral tests were performed 24 hours after SAH, followed by the collection of brain samples for assessment of autophagy and apoptosis proteins. These tests assessed whether an autophagy-apoptosis relationship existed on the histological level in the brain. RESULTS: Endogenous OPN and autophagy-related proteins all increased after SAH. rOPN administration improved neurological dysfunction, increased the expression of autophagy-related proteins (Beclin 1, ATG5, LC3 II to I ratio) and antiapoptotic protein Bcl-2, while decreasing the expression of proapoptotic proteins (cleaved Caspase-3 and Bax). rOPN also regulated autophagy-apoptosis interactions 24 hours after SAH. CONCLUSION: rOPN attenuates early brain injury and inhibits neuronal apoptosis by activating autophagy and regulating autophagy-apoptosis interactions.


Apoptosis/physiology , Autophagy/physiology , Brain Injuries/metabolism , Brain Injuries/prevention & control , Osteopontin/administration & dosage , Subarachnoid Hemorrhage/metabolism , Administration, Intranasal , Animals , Apoptosis/drug effects , Autophagy/drug effects , Brain Injuries/pathology , Male , Osteopontin/biosynthesis , Random Allocation , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/pathology
9.
Stroke ; 50(4): 978-988, 2019 04.
Article En | MEDLINE | ID: mdl-30890112

Background and Purpose- Mitoquinone has been reported as a mitochondria-targeting antioxidant to promote mitophagy in various chronic diseases. Here, our aim was to study the role of mitoquinone in mitophagy activation and oxidative stress-induced neuronal death reduction after subarachnoid hemorrhage (SAH) in rats. Methods- Endovascular perforation was used for SAH model of male Sprague-Dawley rats. Exogenous mitoquinone was injected intraperitoneally 1 hour after SAH. ML385, an inhibitor of Nrf2 (nuclear factor-E2-related factor 2), was given intracerebroventricularly 24 hours before SAH. Small interfering RNA for PHB2 (prohibitin 2) was injected intracerebroventricularly 48 hours before SAH. Nuclear, mitochondrial, and cytoplasmic fractions were gathered using nucleus and mitochondria isolation kits. SAH grade evaluation, short- and long- term neurological function tests, oxidative stress, and apoptosis measurements were performed. Pathway related proteins were investigated with Western blot and immunofluorescence staining. Results- Expression of Keap1 (Kelch-like epichlorohydrin-associated protein 1, 2.84× at 24 hours), Nrf2 (2.78× at 3 hours), and LC3II (light chain 3-II; 1.94× at 24 hours) increased, whereas PHB2 (0.46× at 24 hours) decreased after SAH compared with sham group. Mitoquinone treatment attenuated oxidative stress and neuronal death, both short-term and long-term. Administration of mitoquinone resulted in a decrease in expression of Keap1 (0.33×), Romo1 (reactive oxygen species modulator 1; 0.24×), Bax (B-cell lymphoma-2 associated X protein; 0.31×), Cleaved Caspase-3 (0.29×) and an increase in Nrf2 (2.13×), Bcl-xl (B-cell lymphoma-extra large; 1.67×), PINK1 (phosphatase and tensin-induced kinase 1; 1.67×), Parkin (1.49×), PHB2 (1.60×), and LC3II (1.67×) proteins compared with SAH+vehicle group. ML385 abolished the treatment effects of mitoquinone on behavior and protein levels. PHB2 small interfering RNA reversed the outcomes of mitoquinone administration through reduction in protein expressions downstream of PHB2. Conclusions- Mitoquinone inhibited oxidative stress-related neuronal death by activating mitophagy via Keap1/Nrf2/PHB2 pathway after SAH. Mitoquinone may serve as a potential treatment to relieve brain injury after SAH.


Kelch-Like ECH-Associated Protein 1/metabolism , Mitophagy/physiology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Repressor Proteins/metabolism , Signal Transduction/physiology , Subarachnoid Hemorrhage/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Male , Organophosphorus Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
10.
Redox Biol ; 20: 75-86, 2019 01.
Article En | MEDLINE | ID: mdl-30296700

Oxidative stress and neuronal apoptosis have been demonstrated to be key features in early brain injury (EBI) after subarachnoid hemorrhage (SAH). Previous studies have indicated that Mas receptor activation initiates an anti-oxidative and anti-apoptotic role in the brain. However, whether Mas activation can attenuate oxidative stress and neuronal apoptosis after SAH remains unknown. To investigate the beneficial effect of Mas on oxidative stress injury and neuronal apoptosis induced by SAH, a total of 196 rats were subjected to an endovascular perforation model of SAH. AVE 0991 (AVE), a selective agonist of Mas, was administered intranasally 1 h after SAH induction. A779, a selective inhibitor of Mas, and small interfering ribonucleic acid (siRNA) for UCP-2 were administered by intracerebroventricular (i.c.v) injection at 1 h and 48 h before SAH induction respectively. Neurological tests, immunofluorescence, TUNEL, Fluoro-Jade C, DHE staining, and Western blot experiments were performed. We found that Mas activation with AVE significantly improved neurobehavioral scores and reduced oxidative stress and neuronal apoptosis in SAH+AVE group compared with SAH+vehicle group. Moreover, AVE treatment significantly promoted phosphorylation of CREB and the expression UCP-2, as well as upregulated expression of Bcl-2 and downregulation of Romo-1 and Bax. The protective effects of AVE were reversed by i.c.v injection of A779 and UCP-2 siRNA in SAH+AVE+A779 and SAH+AVE+UCP-2 siRNA groups, respectively. In conclusion, our data provides evidence that Mas activation with AVE reduces oxidative stress injury and neuronal apoptosis through Mas/PKA/p-CREB/UCP-2 pathway after SAH. Furthermore, our study indicates that Mas may be a novel therapeutic treatment target in early brain injury of SAH.


Apoptosis/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Subarachnoid Hemorrhage/metabolism , Uncoupling Protein 2/metabolism , Animals , Brain/metabolism , Cells, Cultured , Gene Expression , Male , Models, Biological , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Rats , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Subarachnoid Hemorrhage/etiology
11.
J Neuroinflammation ; 15(1): 178, 2018 Jun 09.
Article En | MEDLINE | ID: mdl-29885663

BACKGROUND: Neuroinflammation and blood-brain barrier (BBB) disruption are two critical mechanisms of subarachnoid hemorrhage (SAH)-induced brain injury, which are closely related to patient prognosis. Recently, angiogenic factor with G-patch and FHA domain 1 (Aggf1) was shown to inhibit inflammatory effect and preserve vascular integrity in non-nervous system diseases. This study aimed to determine whether Aggf1 could attenuate neuroinflammation and preserve BBB integrity after experimental SAH, as well as the underlying mechanisms of its protective roles. METHODS: Two hundred forty-nine male Sprague-Dawley rats were subjected to the endovascular perforation model of SAH. Recombinant human Aggf1 (rh-Aggf1) was administered intravenously via tail vein injection at 1 h after SAH induction. To investigate the underlying neuroprotection mechanism, Aggf1 small interfering RNA (Aggf1 siRNA) and PI3K-specific inhibitor LY294002 were administered through intracerebroventricular (i.c.v.) before SAH induction. SAH grade, neurological score, brain water content, BBB permeability, Western blot, and immunohistochemistry were performed. RESULTS: Expression of endogenous Aggf1 was markedly increased after SAH. Aggf1 was primarily expressed in endothelial cells and astrocytes, as well as microglia after SAH. Administration of rh-Aggf1 significantly reduced brain water content and BBB permeability, decreased the numbers of infiltrating neutrophils, and activated microglia in the ipsilateral cerebral cortex following SAH. Furthermore, rh-Aggf1 treatment improved both short- and long-term neurological functions after SAH. Meanwhile, exogenous rh-Aggf1 significantly increased the expression of PI3K, p-Akt, VE-cadherin, Occludin, and Claudin-5, as well as decreased the expression of p-NF-κB p65, albumin, myeloperoxidase (MPO), TNF-α, and IL-1ß. Conversely, knockdown of endogenous Aggf1 aggravated BBB breakdown, inflammatory response and neurological impairments at 24 h after SAH. Additionally, the protective roles of rh-Aggf1 were abolished by LY294002. CONCLUSIONS: Taken together, exogenous Aggf1 treatment attenuated neuroinflammation and BBB disruption, improved neurological deficits after SAH in rats, at least in part through the PI3K/Akt/NF-κB pathway.


Angiogenic Proteins/therapeutic use , Blood-Brain Barrier/drug effects , Encephalitis/drug therapy , Encephalitis/etiology , Signal Transduction/drug effects , Subarachnoid Hemorrhage/complications , Angiogenic Proteins/genetics , Animals , Blood-Brain Barrier/physiology , Brain Edema/etiology , Capillary Permeability/drug effects , Chromones/therapeutic use , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Humans , Injections, Intraventricular , Male , Maze Learning/drug effects , Morpholines/therapeutic use , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/physiology , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/mortality , Subarachnoid Hemorrhage/pathology
12.
Biomed Res Int ; 2017: 8403184, 2017.
Article En | MEDLINE | ID: mdl-29214179

Hemorrhagic stroke is a devastating cerebrovascular disease with significant morbidity and mortality worldwide. Thrombospondins (TSPs), as matricellular proteins, belong to the TSP family which is comprised of five members. All TSPs modulate a variety of cellular functions by binding to various receptors. Recently, TSPs gained attention in the area of hemorrhagic stroke, especially TSP-1. TSP-1 participates in angiogenesis, the inflammatory response, apoptosis, and fibrosis after hemorrhagic stroke through binding to various molecules including but not limited to CD36, CD47, and TGF-ß. In this review, we will discuss the roles of TSPs in hemorrhagic stroke and focus primarily on TSP-1.


Intracranial Hemorrhages/metabolism , Stroke/metabolism , Thrombospondins/metabolism , Animals , Apoptosis/physiology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Intracranial Hemorrhages/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Stroke/pathology
13.
Curr Drug Targets ; 18(12): 1377-1385, 2017.
Article En | MEDLINE | ID: mdl-26648067

BACKGROUND: Intracerebral hemorrhage (ICH), a subtype of stroke, brings high morbidity and mortality to human beings. Multiple studies indicated that neuroinflammation, excitotoxicity, oxidative stress, cytotoxicity resulted from the degradation products of blood clot play vital roles in ICH-induced secondary brain injury, which contributes to deterioration of neurological outcome. Prostaglandin E2 (PGE2), a type of prostanoids commonly up-regulated in these progresses, is proved to modulate numerous cellular and molecular processes by activating EP2 receptor after ICH. OBJECTIVE: This review aim to discuss the PGE2 biosynthesis, downstream signaling pathway of EP2 receptor and the roles of EP2 receptor in ICH-induced brain damage, targeting to provide a potential effective therapeutic strategy. METHODS: A large number of literatures on EP2receptors and intracerebral hemorrhage were searched in PubMed, Medline, and Ebase. RESULTS: Previous studies showed that EP2 receptor mediated double effects in ICH via activation of different signaling pathway. EP2 receptor could induce neuroprotection, spatial learning, and neuroplasticity via cAMP-PKA signaling pathway, while strengthen inflammation mainly through the cAMP-Epac pathway. In addition, the concentration level of cAMP might be the key factor that decides which downstream signaling pathway would be activated. CONCLUSION: In different phase of cerebral hemorrhage, EP2 receptor plays diverse effects in brain damage through different downstream signaling pathways.


Brain Injuries/metabolism , Cerebral Hemorrhage/complications , Dinoprostone/biosynthesis , Receptors, Prostaglandin E/metabolism , Animals , Brain Injuries/etiology , Cerebral Hemorrhage/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Signal Transduction , Up-Regulation
14.
Curr Drug Deliv ; 14(6): 766-772, 2017 Sep 06.
Article En | MEDLINE | ID: mdl-27848883

BACKGROUND: Ischemic stroke is the third leading cause of death and the most frequent cause of permanent disability in adults worldwide. Tremendous advances have been made in understanding of the pathophysiology of cerebral ischemia. Nevertheless, there is still no effective neuroprotectant available in the clinical work. Recently, osteopontin (OPN), a glycophosphoprotein, has attracted more attention due to its various effects in cardiovascular and nervous system diseases. OBJECTIVE: The aim of present review was to summarize recent findings about neuroprotective effects of OPN on ischemic stroke, targeting to provide a novel therapeutic strategy. METHODS: To prepare this review, a pathophysiological and pharmacological literature survey was performed using PubMed, and Web of Science. Also, some statistical and epidemiological literature sources were used. RESULTS: Mounting evidence indicates that OPN attenuates cerebral damage and promotes neurogenesis in ischemic stroke by binding to its receptors to activate diverse signaling pathways. CONCLUSION: The paper highlights the neuroprotective and pro-regenerative effects of OPN after cerebral ischemia, which would demonstrate its therapeutic potential in ischemic stroke and other informs of ischemic brain injury.


Brain Ischemia/metabolism , Osteopontin/metabolism , Stroke/metabolism , Animals , Brain/metabolism , Humans , Neuroprotective Agents/metabolism , Signal Transduction/physiology
15.
J Biochem Mol Biol ; 40(5): 749-56, 2007 Sep 30.
Article En | MEDLINE | ID: mdl-17927909

Phosphorylation on serine/threonine or tyrosine residues of target proteins is an essential and significant regulatory mechanism in signal transduction during many cellular and life processes, including spermatogenesis, oogenesis and fertilization. In the present work, we reported the isolation and characterization of mouse testis-specific serine/threonine kinase 5 (Tssk5), which contains four alternatively spliced variants including, Tssk5alpha, Tssk5beta, Tssk5gamma and Tssk5delta. Moreover, the locus of Tssk5 is on chromosome 14qC3 and the four variants had a similar high expression in the testis and the heart; however, had a low expression in other tissues, except for Tssk5alpha which also had comparably high expression in the spleen. Each variant of Tssk5 expression began in the testis 16 days after birth. Aside from TSSK5alpha, the other isoforms have an insertion of ten amino acid residues (RLTPSLSAAG) in region VIb (HRD domain) (His-Arg-Asp). Moreover, only TSSK5alpha exhibited kinase activity and consistently, a further Luciferase Reporter Assay demonstrated that TSSK5beta, TSSK5gamma and TSSK5delta cannot be stimulated at the CREB/CRE responsive pathway in cmparison to TSSK5alpha. These findings suggest that TSSK5beta, TSSK5gamma, TSSK5delta may be pseudokinases due to the insertion, which may damage the structure responsible for active kinase activity. Pull-down assay experiments indicated that TSSK5beta, TSSK5gamma and TSSK5delta can directly interact with TSSK5alpha. In summary, these four isoforms with similar expression patterns may be involved in spermatogenesis through a coordinative way in testis.


Alternative Splicing , Protein Serine-Threonine Kinases/genetics , Testis/metabolism , Amino Acid Sequence , Animals , Aurora Kinases , Blotting, Western , Catalysis , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed/methods , Plasmids/genetics , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Substrate Specificity , Testis/embryology
16.
Shanghai Kou Qiang Yi Xue ; 13(2): 126-9, 2004 Apr.
Article Zh | MEDLINE | ID: mdl-15133558

PURPOSE: The purpose of this study was to clone human amelogenin gene encoding mature protein, which provides a basis for expressing the recombinant human amelogenin in Escherichia coli. in the future. METHODS: In this study, total RNA was extracted from the dental germ of a legally aborted embryo by Trizol. Using RT-PCR technique we obtained synthesis of cDNA from the total RNA, and the desired DNA products were conducted with PCR from cDNA. The segment was inserted into expression vector PQE30 and the interesting plasmid was transformed into Escherichia coli. host DH5alpha. The double-stranded DNA of positive clone was analyzed by PCR, restriction endonuclease mapping and DNA sequence analysis. RESULTS: The sequence analysis of recombinant plasmid showed that the human amelogenin encoding mature protein was inserted into vector PQE30 accurately. CONCLUSIONS: We conducted human amelogenin encoding mature protein from dental germ of a legally aborted embryo and got the recombinant plasmid which may express amelogenin gene for further research.


Dental Enamel Proteins/genetics , Amelogenin , Cloning, Molecular , Dental Enamel Proteins/biosynthesis , Humans , Molecular Weight , Plasmids , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
17.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 8(2): 97-100, 2000 Jun.
Article Zh | MEDLINE | ID: mdl-12578695

Chronic myeloid leukemia (CML) appears an ideal and exciting immunological target. Novel and rational immunotherapy may therefore play an important adjuvant role in the treatment of CML patients. Peptides derived from the BCR-ABL fusion region have been shown to be immunogenic and are able to stimulate the production of BCR-ABL-specific T cell lines and clones. In this study, A 280 bp multiple epitope region of BCR-ABL fusion antigen was designed and synthesized. This region contains three BCR-ABL antigen epitopes which can bind to HLA-A2, HLA-A3 and HLA-DR11 molecules, respectively, and epitopes of cholera toxin B (CTB) and tetanus toxoid (TT) which are able to elicit vigorous T cell responses. The fusion antigen gene has highly been expressed in E. coli and the purified fusion protein reserved satisfied activity and antigenicity. The results of this investigation provided a basis for further research on the developing specific T cell immunotherapy of CML.

...