Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Neurochem Res ; 42(6): 1847-1863, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28478595

ABSTRACT

Cell migration is identified as a highly orchestrated process. It is a fundamental and essential phenomenon underlying tissue morphogenesis, wound healing, and immune response. Under dysregulation, it contributes to cancer metastasis. Brain is considered to be the most complex organ in human body containing many types of neural cells with astrocytes playing crucial roles in monitoring both physiological and pathological functions. Astrocytoma originates from astrocytes and its most malignant type is glioblastoma multiforme (WHO Grade IV astrocytoma), which is capable to infiltrate widely into the neighboring brain tissues making a complete resection of tumors impossible. Very recently, we have reviewed the mechanisms for astrocytes in migration. Given the fact that astrocytoma shares many histological features with astrocytes, we therefore attempt to review the mechanisms for glioma cells in migration and compare them to normal astrocytes, hoping to obtain a better insight into the dysregulation of migratory mechanisms contributing to their metastasis in the brain.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Cell Movement/physiology , Glioma/pathology , Animals , Astrocytes/pathology , Brain/metabolism , Brain/surgery , Brain Neoplasms/metabolism , Brain Neoplasms/surgery , Glioma/metabolism , Glioma/surgery , Humans
2.
Neurochem Res ; 42(1): 272-282, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27837318

ABSTRACT

Cell migration is a fundamental phenomenon that underlies tissue morphogenesis, wound healing, immune response, and cancer metastasis. Great progresses have been made in research methodologies, with cell migration identified as a highly orchestrated process. Brain is considered the most complex organ in the human body, containing many types of neural cells with astrocytes playing crucial roles in monitoring normal functions of the central nervous system. Astrocytes are mostly quiescent under normal physiological conditions in the adult brain but become migratory after injury. Under most known pathological conditions in the brain, spinal cord and retina, astrocytes are activated and become hypertrophic, hyperplastic, and up-regulating GFAP based on the grades of severity. These three observations are the hallmark in glia scar formation-astrogliosis. The reactivation process is initiated with structural changes involving cell process migration and ended with cell migration. Detailed mechanisms in astrocyte migration have not been studied extensively and remain largely unknown. Here, we therefore attempt to review the mechanisms in migration of astrocytes.


Subject(s)
Astrocytes/metabolism , Cell Movement/physiology , Animals , Brain/cytology , Brain/metabolism , Cell Adhesion/physiology , Cells, Cultured , Humans , Spinal Cord/cytology , Spinal Cord/metabolism
3.
Sheng Li Xue Bao ; 69(5): 703-714, 2017 Oct 25.
Article in Zh | MEDLINE | ID: mdl-29063118

ABSTRACT

DREAM (downstream regulatory element antagonist modulator), Calsenilin and KChIP3 (potassium channel interacting protein 3) belong to the neuronal calcium sensor (NCS) superfamily, which transduces the intracellular calcium signaling into a variety of activities. They are encoded by the same gene locus, but have distinct subcellular locations. DREAM was first found to interact with DRE (downstream regulatory element) site in the vicinity of the promoter of prodynorphin gene to suppress gene transcription. Calcium can disassemble this interaction by binding reversibly to DREAM protein on its four EF-hand motifs. Apart from having calcium dependent DRE site binding, DREAM can also interact with other transcription factors, such as cAMP responsive element binding protein (CREB), CREB-binding protein (CBP) and cAMP responsive element modulator (CREM), by this concerted actions, DREAM extends the gene pool under its control. DREAM is predominantly expressed in central nervous system with its highest level in cerebellum, and accumulating evidence demonstrated that DREAM might play important roles in pain sensitivity. Novel findings have shown that DREAM is also involved in learning and memory processes, Alzheimer's disease and stroke. This mini-review provides a brief introduction of its discovery history and protein structure properties, focusing on the mechanism of DREAM nuclear translocation and gene transcription regulation functions.


Subject(s)
Gene Expression Regulation , Kv Channel-Interacting Proteins/physiology , Repressor Proteins/physiology , Animals , Calcium Signaling/physiology , Humans , Kv Channel-Interacting Proteins/genetics , Pain Threshold , Repressor Proteins/genetics
4.
J Neurooncol ; 128(2): 217-23, 2016 06.
Article in English | MEDLINE | ID: mdl-26983952

ABSTRACT

Phloretin, a flavonoid present in various plants, has been reported to exert anticarcinogenic effects. However, the mechanism of its chemo-preventive effect on human glioblastoma cells is not fully understood. This study aimed to investigate the molecular mechanism of phloretin and its associated chemo-preventive effect in human glioblastoma cells. The results indicate that phloretin inhibited cell proliferation by inducing cell cycle arrest at the G0-G1 phase and induced apoptosis of human glioblastoma cells. Phloretin-induced cell cycle arrest was associated with increased expression of p27 and decreased expression of cdk2, cdk4, cdk6, cyclinD and cyclinE. Moreover, the PI3K/AKT/mTOR signaling cascades were suppressed by phloretin in a dose-dependent manner. In addition, phloretin triggered the mitochondrial apoptosis pathway and generated reactive oxygen species (ROS). This was accompanied by the up-regulation of Bax, Bak and c-PARP and the down-regulation of Bcl-2. The antioxidant agents N-acetyl-L-cysteine and glutathione weakened the effect of phloretin on glioblastoma cells. In conclusion, these results demonstrate that phloretin exerts potent chemo-preventive activity in human glioblastoma cells through the generation of ROS.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Glioblastoma/drug therapy , Phloretin/pharmacology , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Caspase 9/metabolism , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Glioblastoma/metabolism , Humans , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
5.
J Neurosci Res ; 93(10): 1507-18, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25711139

ABSTRACT

Stroke is a leading cause of death and disability, and new strategies are required to reduce neuronal injury and improve prognosis. Ischemia preconditioning (IPC) is an intrinsic phenomenon that protects cells from subsequent ischemic injury and might provide promising mechanisms for clinical treatment. In this study, primary astrocytes exhibited significantly less cell death than control when exposed to different durations of IPC (15, 30, 60, or 120 min). A 15-min duration was the most effective IPC to protect astrocytes from 8-hr-ischemia injury. The protective mechanisms of IPC involve the upregulation of protective proteins, including 14-3-3γ, and attenuation of malondialdehyde (MDA) content and ATP depletion. 14-3-3γ is an antiapoptotic intracellular protein that was significantly upregulated for up to 84 hr after IPC. In addition, IPC promoted activation of the c-Jun N-terminal kinase (JNK), extracellular signal-related kinase (ERK)-1/2, p38, and protein kinase B (Akt) signaling pathways. When JNK was specifically inhibited with SP600125, the upregulation of 14-3-3γ induced by IPC was almost completely abolished; however, there was no effect on ATP or MDA levels. This suggests that, even though both energy preservation and 14-3-3γ up-regulation were turned on by IPC, they were controlled by different pathways. The ERK1/2, p38, and Akt signaling pathways were not involved in the 14-3-3γ upregulation and energy preservation. These results indicate that IPC could protect astrocytes from ischemia injury by inducing 14-3-3γ and by alleviating energy depletion through different pathways, suggesting multiple protection of IPC and providing new insights into potential stroke therapies.


Subject(s)
14-3-3 Proteins/metabolism , Astrocytes/metabolism , Gene Expression Regulation/physiology , Ischemic Preconditioning , Signal Transduction/physiology , Up-Regulation/physiology , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Cell Count , Cell Death/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/cytology , Gene Expression Regulation/drug effects , Ischemia/prevention & control , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Mice , Mice, Inbred ICR , Signal Transduction/drug effects , Time Factors
6.
J Neurosci Res ; 93(2): 253-67, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25250856

ABSTRACT

The superfamily of importin-ß-related proteins is the largest class of nuclear transport receptors and can be generally divided into importins and exportins according to their transport directions. Eleven importins and seven exportins have been identified, and the expression patterns of both classes are important for their functions in nucleocytoplasmic transport activities. This study demonstrates that all of the importins (importin-ß; transportin-1, -2, and -3; and importin-4, -5, -7, -8, -9, -11, and -13) and all the exportins (exportin-1, -2, -4, -5, -6, -7, and -t) are differentially expressed in the cerebral cortex, cerebellum, hippocampus, and brainstem and in primary cultures of cerebral cortical astrocytes and neurons. For astrocytes, we observed that different importins and exportins displayed different expression changes during 0-6 hr of ischemia treatment, especially an increase of both the mRNA and the protein of exportin-7. Immunostaining showed that exportin-7 accumulated inside the nucleus and around the nuclear envelope. In addition, we noticed an increased cytoplasmic distribution of one of the cargo proteins of exportin-7, LKB1, an important element in maintaining energy homeostasis. This increased cytoplasmic distribution was accompanied by an increased expression of exportin-7 under ischemia in astrocytes. We demonstrate that exportin-7 responds to ischemia in astrocytes and that this response involves translocation of LKB1, a protein that plays important roles during metabolic stress, from the nucleus to the cytoplasm.


Subject(s)
Astrocytes/metabolism , Astrocytes/ultrastructure , Brain Ischemia/pathology , Gene Expression Regulation/physiology , Protein Serine-Threonine Kinases/metabolism , ran GTP-Binding Protein/metabolism , AMP-Activated Protein Kinases , Animals , Cell Hypoxia/physiology , Cell Nucleus/metabolism , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation/genetics , Karyopherins/genetics , Karyopherins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred ICR , Neurons/cytology , Neurons/metabolism , Photobleaching , Protein Serine-Threonine Kinases/genetics , Protein Transport/genetics , Protein Transport/physiology , RNA, Messenger/metabolism , beta Karyopherins/genetics , beta Karyopherins/metabolism , ran GTP-Binding Protein/genetics
7.
Neurochem Res ; 40(9): 1929-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26248512

ABSTRACT

Cadmium (Cd), a highly ubiquitous toxic heavy metal, can contaminate the environment, including agricultural soil, water and air, via industrial runoff and other sources of pollution. Cd accumulated in the body via direct exposure or through the food chain results in neurodegeneration and many other diseases. Previous studies on its toxicity in the central nervous system (CNS) focused mainly on neurons. To obtain a more comprehensive understanding of Cd toxicity for the CNS, we investigated how astrocytes respond to acute and chronic Cd exposure and its toxic molecular mechanisms. When primary cultures of cerebral cortical astrocytes incubated with 1-300 µM CdCl2, morphological changes, LDH release and cell death were observed in a time and dose-dependent manner. Further studies demonstrated that acute and chronic Cd treatment phosphorylated JNK, p38 and Akt to different degrees, while ERK1/2 was only phosphorylated under low doses of Cd (10 µM) exposure. Inhibition of JNK and PI3K/Akt, but not of p38, could partially protect astrocyte from cytotoxicity in chronic and acute Cd exposure. Moreover, Cd also induced a strong calcium signal, while BAPTA, a specific intracellular calcium (Ca(2+)) chelator, prevented Cd-induced intracellular increase of calcium levels in astrocytes; inhibited the Cd-induced activation of ERK1/2, JNK, p38 and Akt; and also significantly reduced astrocyte cell death. All of these results suggested that the Cd-Ca(2+)-MAPK and PI3K/Akt signaling pathways were involved in Cd-induced toxicity in astrocytes. This toxicity involvement indicates that these pathways may be exploited as a target for the prevention of Cd-induced neurodegenerative diseases.


Subject(s)
Astrocytes/drug effects , Cadmium/toxicity , Calcium Signaling , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Astrocytes/enzymology , Dose-Response Relationship, Drug , Mice , Mice, Inbred ICR
8.
Cell Mol Neurobiol ; 34(6): 881-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24913968

ABSTRACT

Stem cell transplantation therapy has provided promising hope for the treatment of a variety of neurodegenerative disorders. Among challenges in developing disease-specific stem cell therapies, identification of key regulatory signals for neuronal differentiation is an essential and critical issue that remains to be resolved. Several lines of evidence suggest that JNK, also known as SAPK, is involved in neuronal differentiation and neural plasticity. It may also play a role in neurite outgrowth during neuronal development. In cultured mouse embryonic stem (ES) cells, we test the hypothesis that the JNK pathway is required for neuronal differentiation. After neural induction, the cells were plated and underwent differentiation for up to 5 days. Western blot analysis showed a dramatic increase in phosphorylated JNKs at 1-5 days after plating. The phosphorylation of JNK subsequently induced activation of STAT1 and STAT3 that lead to expressions of GAP-43, neurofilament, ßIII-tubulin, and synaptophysin. NeuN-colabelled with DCX, a marker for neuroblast, was enhanced by JNK signaling. Neuronal differentiation of ES cells was attenuated by treatment with SP600125, which inhibited the JNK activation and decreased the activation of STAT1 and STAT3, and consequently suppressed the expressions of GAP-43, neurofilament, ßIII-tubulin, and the secretion of VEGF. Data from immunocytochemistry indicated that the nuclear translocation of STAT3 was reduced, and neurites of ES-derived neurons were shorter after treatment with SP600125 compared with control cells. These results suggest that the JNK-STAT3 pathway is a key regulator required for early neuronal differentiation of mouse ES cells. Further investigation on expression of JNK isoforms showed that JNK-3 was significantly upregulated during the differentiation stage, while JNK-1 and JNK-2 levels decreased. Our study provided interesting information on JNK functions during ES cell neuronal differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , MAP Kinase Signaling System/physiology , Neurons/cytology , STAT3 Transcription Factor/metabolism , Animals , Anthracenes/pharmacology , Cells, Cultured , Doublecortin Protein , Mice , Phosphorylation
9.
Glia ; 61(12): 2063-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123203

ABSTRACT

Astrocyte activation is a hallmark of central nervous system injuries resulting in glial scar formation (astrogliosis). The activation of astrocytes involves metabolic and morphological changes with complex underlying mechanisms, which should be defined to provide targets for astrogliosis intervention. Astrogliosis is usually accompanied by an upregulation of glial fibrillary acidic protein (GFAP). Using an in vitro scratch injury model, we scratched primary cultures of cerebral cortical astrocytes and observed an influx of calcium in the form of waves spreading away from the wound through gap junctions. Using the calcium blocker BAPTA-AM and the JNK inhibitor SP600125, we demonstrated that the calcium wave triggered the activation of JNK, which then phosphorylated the transcription factor c-Jun to facilitate the binding of AP-1 to the GFAP gene promoter to switch on GFAP upregulation. Blocking calcium mobilization with BAPTA-AM in an in vivo stab wound model reduced GFAP expression and glial scar formation, showing that the calcium signal, and the subsequent regulation of downstream signaling molecules, plays an essential role in brain injury response. Our findings demonstrated that traumatic scratch injury to astrocytes triggered a calcium influx from the extracellular compartment and activated the JNK/c-Jun/AP-1 pathway to switch on GFAP expression, identifying a previously unreported signaling cascade that is important in astrogliosis and the physiological response following brain injury.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Genes, jun/physiology , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , MAP Kinase Signaling System/physiology , Transcription Factor AP-1/metabolism , Animals , Astrocytes/cytology , Calcium Signaling/genetics , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gliosis/genetics , Mice , Mice, Inbred ICR , Transcription Factor AP-1/genetics , Transcriptional Activation
10.
Biochem Biophys Res Commun ; 437(1): 87-93, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23796709

ABSTRACT

Piperlongumine (PL), a natural alkaloid isolated from the long pepper, may have anti-cancer properties. It selectively targets and kills cancer cells but leaves normal cells intact. Here, we reported that PL selectively killed glioblastoma multiforme (GBM) cells via accumulating reactive oxygen species (ROS) to activate JNK and p38. PL at 20µM could induce severe cell death in three GBM cell lines (LN229, U87 and 8MG) but not astrocytes in cultures. PL elevated ROS prominently and reduced glutathione levels in LN229 and U87 cells. Antioxidant N-acetyl-L-cysteine (NAC) completely reversed PL-induced ROS accumulation and prevented cell death in LN229 and U87 cells. In LN229 and U87 cells, PL-treatment activated JNK and p38 but not Erk and Akt, in a dosage-dependent manner. These activations could be blocked by NAC pre-treatment. JNK and p38 specific inhibitors, SB203580 and SP600125 respectively, significantly blocked the cytotoxic effects of PL in LN229 and U87 cells. Our data first suggests that PL may have therapeutic potential for one of the most malignant and refractory tumors GBM.


Subject(s)
Dioxolanes/pharmacology , Glioblastoma/enzymology , Glioblastoma/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Cell Death/drug effects , Cell Line, Tumor , Dioxolanes/chemistry , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Humans
11.
Cells ; 12(16)2023 08 17.
Article in English | MEDLINE | ID: mdl-37626895

ABSTRACT

This study comprehensively addresses the involvement of the protein CKLF-like Marvel transmembrane domain-containing family member 5 (CMTM5) in the context of demyelination and cytodegenerative autoimmune diseases, particularly multiple Sclerosis (MS). An observed reduction in CMTM5 expression in post-mortem MS lesions prompted further investigations in both in vitro and in vivo animal models. In the cuprizone animal model, we detected a decrease in CMTM5 expression in oligodendrocytes that is absent in other members of the CMTM protein family. Our findings also confirm these results in the experimental autoimmune encephalomyelitis (EAE) model with decreased CMTM5 expression in both cerebellum and spinal cord white matter. We also examined the effects of a Cmtm5 knockdown in vitro in the oligodendroglial Oli-neu mouse cell line using the CRISPR interference technique. Interestingly, we found no effects on cell response to thapsigargin-induced endoplasmic reticulum (ER) stress as determined by Atf4 activity, an indicator of cellular stress responses. Overall, these results substantiate previous findings suggesting that CMTM5, rather than contributing to myelin biogenesis, is involved in maintaining axonal integrity. Our study further demonstrates that the knockdown of Cmtm5 in vitro does not modulate oligodendroglial responses to ER stress. These results warrant further investigation into the functional role of CMTM5 during axonal degeneration in the context of demyelinating conditions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Mice , Multiple Sclerosis/genetics , Myelin Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Autopsy , Oligodendroglia
12.
Cells ; 11(16)2022 08 18.
Article in English | MEDLINE | ID: mdl-36010640

ABSTRACT

Astrocytes have distinctive morphological and functional characteristics, and are found throughout the central nervous system. Astrocytes are now known to be far more than just housekeeping cells in the brain. Their functions include contributing to the formation of the blood-brain barrier, physically and metabolically supporting and communicating with neurons, regulating the formation and functions of synapses, and maintaining water homeostasis and the microenvironment in the brain. Aquaporins (AQPs) are transmembrane proteins responsible for fast water movement across cell membranes. Various subtypes of AQPs (AQP1, AQP3, AQP4, AQP5, AQP8 and AQP9) have been reported to be expressed in astrocytes, and the expressions and subcellular localizations of AQPs in astrocytes are highly correlated with both their physiological and pathophysiological functions. This review describes and summarizes the recent advances in our understanding of astrocytes and AQPs in regard to controlling water homeostasis in the brain. Findings regarding the features of different AQP subtypes, such as their expression, subcellular localization, physiological functions, and the pathophysiological roles of astrocytes are presented, with brain edema and glioma serving as two representative AQP-associated pathological conditions. The aim is to provide a better insight into the elaborate "water distribution" system in cells, exemplified by astrocytes, under normal and pathological conditions.


Subject(s)
Aquaporins , Astrocytes , Aquaporins/metabolism , Astrocytes/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Water/metabolism
13.
J Neurosci Res ; 89(12): 2041-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21544851

ABSTRACT

We observed nuclear swelling in glutamate (Glu)-treated astrocytes that was concomitant with but independent of astrocytic cell swelling. We confirmed Glu-induced nuclear swelling with nuclei isolated from astrocytes. Ammonia is metabolically related to Glu and could induce a nuclear swelling in intact astrocytes but shrinkage in isolated nuclei. Other compounds such as glutamine, aspartate, taurine, glycine, and ATP did not cause any nuclear swelling in isolated nuclei of astrocytes. Surprisingly, Glu and ammonia did not induce nuclear swelling in microglia, C6, HEK 293, or Hep G2 cell lines in cultures and their isolated nuclei. The Glu- and ammonia-induced nuclear size changes appear to be a specific response of astrocytes to these two closely related metabolic compounds.


Subject(s)
Ammonia/pharmacology , Astrocytes/ultrastructure , Cell Nucleus/ultrastructure , Glutamic Acid/pharmacology , Ammonia/metabolism , Animals , Astrocytes/drug effects , Cell Nucleus/drug effects , Cells, Cultured , Glutamic Acid/metabolism , Mice , Mice, Inbred ICR , Microscopy, Atomic Force
14.
Neurochem Res ; 35(12): 2124-34, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21116713

ABSTRACT

The expression of the N-methyl-D-aspartate receptor (NMDA-R) in astrocytes is controversial. The receptor is commonly considered neuron-specific. We showed that astrocytes in primary cultures differentially expressed mRNA of NMDA-R subunits, NR1, NR2A and NR2B, in development, ischemia and post-ischemia. One-week-old cultures expressed detectable NR1 mRNA, which fell significantly at 2 weeks and became barely detectable at 4 weeks. NR2A and NR2B mRNA were both significantly up-regulated from 1 to 2 weeks. In 4 weeks, 2 h of ischemia caused a significant up-regulation of NR1 and NR2B mRNA; while 6 h caused down-regulation of NR2A mRNA. Under 3 h of post-ischemia, only NR1 mRNA was increased. Ischemia induced the expression of major NMDA-R effecter, nitric oxide synthase 1, which was unaffected by AMPA-R antagonist CNQX, but dose-dependently inhibited by NMDA-R specific antagonist MK-801. These findings reflected that astrocyte could express inducible functional NMDA receptors without the presence of neurons.


Subject(s)
Astrocytes/metabolism , Brain Ischemia/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain Ischemia/pathology , Down-Regulation , Fluorescent Antibody Technique , Glutamic Acid/pharmacology , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred ICR , Nitric Oxide Synthase Type I/metabolism , RNA, Messenger/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Up-Regulation
15.
J Neurochem ; 109 Suppl 1: 182-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19393026

ABSTRACT

Ischemia occurs in the brain as the result of stroke and other related injuries and few therapies are effective. If more is understood then potential treatments could be investigated. It was previously reported that 14-3-3gamma could be up-regulated by ischemia in astrocyte to protect cells from ischemia-induced apoptosis. In this study, we attempted to uncover the mechanism responsible for this 14-3-3gamma up-regulation in primary culture of astrocytes under ischemic-like conditions. It was found that in vitro ischemia may activate PI3K/Akt and MAPK signaling pathways. Astrocyte cultures were treated with LY294002 (PI3K inhibitor), U0126 (ERK inhibitor), SB203580 (p38 inhibitor) and SP600125 (JNK inhibitor). Only SP600125 could inhibit the ischemia-induced 14-3-3gamma up-regulation in astrocytes. At the same time, we observed an ischemia-induced nuclear translocation of p-c-Jun, a major downstream component of JNK. Inhibition of AP-1 with curcumin also inhibited 14-3-3gamma up-regulation indicating that ischemia-induced up-regulation of 14-3-3gamma in astrocyte involves activation of the JNK/p-c-Jun/AP-1 pathway.


Subject(s)
14-3-3 Proteins/biosynthesis , Astrocytes/metabolism , Brain Ischemia/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/physiology , Transcription Factor AP-1/metabolism , Astrocytes/enzymology , Atmosphere Exposure Chambers , Brain Ischemia/enzymology , Cells, Cultured , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , RNA/biosynthesis , RNA/isolation & purification , Signal Transduction/drug effects , Transcription Factor AP-1/antagonists & inhibitors , Up-Regulation
16.
Sheng Li Ke Xue Jin Zhan ; 40(3): 197-202, 2009 Jul.
Article in Zh | MEDLINE | ID: mdl-19803421

ABSTRACT

Polyphosphate (poly P) has been widely identified in both inorganic environment and living organisms. Research shows that poly P in bacteria enhances their resistance to severe environment, triggers their protective responses, increases biofilm formation and involves in predation and bacterial virulence. In eukaryotes, poly P has been found to enhance the proliferation of fibroblast and many tumor cell lines, induce the calcification of osteoblast and be involved in calcium ion release. Based on the existing information, we attempt to discuss the possible functions of poly P in the nervous system.


Subject(s)
Polyphosphates/metabolism , Bacteria/metabolism , Calcium/metabolism , Cell Line, Tumor , Fibroblasts/cytology , Humans , Nervous System/metabolism , Osteoblasts/cytology
17.
Clin Infect Dis ; 47(12): 1575-8, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18990065

ABSTRACT

The mechanism of systemic spread of H5N1 virus in patients with avian influenza is unknown. Here, H5N1 nucleoprotein and hemagglutinin were identified by immunohistochemistry in the nucleus and cytoplasm of neutrophils in the placental blood of a pregnant woman. Viral RNA was detected in neutrophils by in situ hybridization and enhanced real-time polymerase chain reaction. Therefore, neutrophils may serve as a vehicle for viral replication and transportation in avian influenza.


Subject(s)
Influenza A Virus, H5N1 Subtype/growth & development , Influenza, Human/virology , Neutrophils/virology , Adult , Antigens, Viral/analysis , Humans , Immunohistochemistry , In Situ Hybridization , Neutrophils/chemistry , Pregnant Women , RNA, Viral/analysis
18.
Lancet ; 370(9593): 1137-45, 2007 Sep 29.
Article in English | MEDLINE | ID: mdl-17905166

ABSTRACT

BACKGROUND: Human infection with avian influenza H5N1 is an emerging infectious disease characterised by respiratory symptoms and a high fatality rate. Previous studies have shown that the human infection with avian influenza H5N1 could also target organs apart from the lungs. METHODS: We studied post-mortem tissues of two adults (one man and one pregnant woman) infected with H5N1 influenza virus, and a fetus carried by the woman. In-situ hybridisation (with sense and antisense probes to haemagglutinin and nucleoprotein) and immunohistochemistry (with monoclonal antibodies to haemagglutinin and nucleoprotein) were done on selected tissues. Reverse-transcriptase (RT) PCR, real-time RT-PCR, strand-specific RT-PCR, and nucleic acid sequence-based amplification (NASBA) detection assays were also undertaken to detect viral RNA in organ tissue samples. FINDINGS: We detected viral genomic sequences and antigens in type II epithelial cells of the lungs, ciliated and non-ciliated epithelial cells of the trachea, T cells of the lymph node, neurons of the brain, and Hofbauer cells and cytotrophoblasts of the placenta. Viral genomic sequences (but no viral antigens) were detected in the intestinal mucosa. In the fetus, we found viral sequences and antigens in the lungs, circulating mononuclear cells, and macrophages of the liver. The presence of viral sequences in the organs and the fetus was also confirmed by RT-PCR, strand-specific RT-PCR, real-time RT-PCR, and NASBA. INTERPRETATION: In addition to the lungs, H5N1 influenza virus infects the trachea and disseminates to other organs including the brain. The virus could also be transmitted from mother to fetus across the placenta.


Subject(s)
Fetus/pathology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/pathology , Respiratory System/pathology , Adult , Female , Genome, Viral , Humans , Immunohistochemistry , In Situ Hybridization/methods , Infectious Disease Transmission, Vertical , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza, Human/transmission , Influenza, Human/virology , Male , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Self-Sustained Sequence Replication/methods
19.
Front Biosci ; 13: 7123-31, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508721

ABSTRACT

Focal adhesion associated protein (FAAP), encoded by murine D10Wsu52e gene, is highly homologous to human HSPC117, which interacts with vinculin and talin. HeLa cells transfected with FAAP exhibited normal adhesion incorporation but showed impaired cell spreading, and restrained focal adhesion translocation. Moreover, FAAP facilitated vinculin-paxillin association, decreased interaction of paxillin-focal adhesion kinase and inhibited the phosphorylation of extracellular signal-regulated kinase. Together, these results suggest that FAAP, by virtue of modulating interaction of adhesion molecules, regulates cell adhesion dynamics.


Subject(s)
Cell Adhesion/physiology , Focal Adhesion Kinase 1/physiology , Paxillin/physiology , Vinculin/physiology , Animals , Cell Adhesion/genetics , Cell Movement/physiology , Focal Adhesion Kinase 1/genetics , Genetic Markers , HeLa Cells/cytology , HeLa Cells/physiology , Humans , Mice , Transfection
20.
Neurochem Res ; 33(12): 2480-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18563562

ABSTRACT

The unexpected finding of astrocytes to release glutamate as gliotransmitter challenges the traditional concepts on astrocyte being "passive" in CNS communications. Glutamate is the major excitatory transmitter in transferring information between neurons, but is now also known to activate astrocyte through transporters and receptors. Together with the sensitive swelling response, astrocytes could respond directly to glutamate and neuronal activity. Other new functions of astrocytes include modulation of synaptic plasticity and cerebral blood flow (CBF). The classic glutamate deplenishment through glutamine synthesis and CO(2) production does not account for the total glutamate internalized into astrocytes. This leads us to speculate there are many hidden functions of glutamate in neurons and astrocytes waiting to be discovered. In this review, we attempted to reexamine some of these new and older functions of glutamate and to reevaluate the roles of glutamate intoning these two cell types.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Animals , Astrocytes/cytology , Brain/blood supply , Brain/cytology , Humans , Neuronal Plasticity , Neurons/cytology
SELECTION OF CITATIONS
SEARCH DETAIL