Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
Add more filters










Publication year range
1.
Eur Rev Med Pharmacol Sci ; 26(6): 2201-2214, 2022 03.
Article in English | MEDLINE | ID: mdl-35363371

ABSTRACT

Disruption of intracellular Ca2+ homeostasis plays an important role as an upstream pathology in Alzheimer's disease (AD), and correction of Ca2+ dysregulation has been increasingly proposed as a target of future effective disease-modified drugs for treating AD. Calcium dysregulation is also an upstream pathology for the COVID-19 virus SARS-CoV-2 infection and replication, leading to host cell damage. Clinically available drugs that can inhibit the disturbed intracellular Ca2+ homeostasis have been repurposed to treat COVID-19 patients. This narrative review aims at exploring the underlying mechanism by which lithium, a first line drug for the treatment of bipolar disorder, inhibits Ca2+ dysregulation and associated downstream pathology in both AD and COVID-19. It is suggested that lithium can be repurposed to treat AD patients, especially those afflicted with COVID-19.


Subject(s)
Alzheimer Disease , COVID-19 Drug Treatment , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Lithium/therapeutic use , Lithium Compounds/therapeutic use , SARS-CoV-2
2.
Transl Psychiatry ; 5: e504, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25646593

ABSTRACT

We developed a novel integrative genomic tool called GRANITE (Genetic Regulatory Analysis of Networks Investigational Tool Environment) that can effectively analyze large complex data sets to generate interactive networks. GRANITE is an open-source tool and invaluable resource for a variety of genomic fields. Although our analysis is confined to static expression data, GRANITE has the capability of evaluating time-course data and generating interactive networks that may shed light on acute versus chronic treatment, as well as evaluating dose response and providing insight into mechanisms that underlie therapeutic versus sub-therapeutic doses or toxic doses. As a proof-of-concept study, we investigated lithium (Li) response in bipolar disorder (BD). BD is a severe mood disorder marked by cycles of mania and depression. Li is one of the most commonly prescribed and decidedly effective treatments for many patients (responders), although its mode of action is not yet fully understood, nor is it effective in every patient (non-responders). In an in vitro study, we compared vehicle versus chronic Li treatment in patient-derived lymphoblastoid cells (LCLs) (derived from either responders or non-responders) using both microRNA (miRNA) and messenger RNA gene expression profiling. We present both Li responder and non-responder network visualizations created by our GRANITE analysis in BD. We identified by network visualization that the Let-7 family is consistently downregulated by Li in both groups where this miRNA family has been implicated in neurodegeneration, cell survival and synaptic development. We discuss the potential of this analysis for investigating treatment response and even providing clinicians with a tool for predicting treatment response in their patients, as well as for providing the industry with a tool for identifying network nodes as targets for novel drug discovery.


Subject(s)
Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Lithium Compounds/therapeutic use , MicroRNAs/genetics , Pharmacogenetics/methods , Statistics as Topic/methods , Adult , Female , Genomics/instrumentation , Genomics/methods , Humans , Male , Middle Aged , Pharmacogenetics/instrumentation , Statistics as Topic/instrumentation , Treatment Outcome , Young Adult
3.
Mol Psychiatry ; 20(2): 215-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24468826

ABSTRACT

Fibroblast growth factor-21 (FGF-21) is a new member of the FGF super-family and an important endogenous regulator of glucose and lipid metabolism. It has been proposed as a therapeutic target for diabetes and obesity. Its function in the central nervous system (CNS) remains unknown. Previous studies from our laboratory demonstrated that aging primary neurons are more vulnerable to glutamate-induced excitotoxicity, and that co-treatment with the mood stabilizers lithium and valproic acid (VPA) induces synergistic neuroprotective effects. This study sought to identify molecule(s) involved in these synergistic effects. We found that FGF-21 mRNA was selectively and markedly elevated by co-treatment with lithium and VPA in primary rat brain neurons. FGF-21 protein levels were also robustly increased in neuronal lysates and culture medium following lithium-VPA co-treatment. Combining glycogen synthase kinase-3 (GSK-3) inhibitors with VPA or histone deacetylase (HDAC) inhibitors with lithium synergistically increased FGF-21 mRNA levels, supporting that synergistic effects of lithium and VPA are mediated via GSK-3 and HDAC inhibition, respectively. Exogenous FGF-21 protein completely protected aging neurons from glutamate challenge. This neuroprotection was associated with enhanced Akt-1 activation and GSK-3 inhibition. Lithium-VPA co-treatment markedly prolonged lithium-induced Akt-1 activation and augmented GSK-3 inhibition. Akt-1 knockdown markedly decreased FGF-21 mRNA levels and reduced the neuroprotection induced by FGF-21 or lithium-VPA co-treatment. In addition, FGF-21 knockdown reduced lithium-VPA co-treatment-induced Akt-1 activation and neuroprotection against excitotoxicity. Together, our novel results suggest that FGF-21 is a key mediator of the effects of these mood stabilizers and a potential new therapeutic target for CNS disorders.


Subject(s)
Antimanic Agents/pharmacology , Fibroblast Growth Factors/metabolism , Gene Expression Regulation/drug effects , Neurons/drug effects , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Drug Synergism , Excitatory Amino Acid Agonists/toxicity , Fibroblast Growth Factors/pharmacology , Glutamic Acid/toxicity , Glycogen Synthase Kinase 3/metabolism , Hippocampus/cytology , Lithium/pharmacology , Male , Mice , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Rats , Rats, Sprague-Dawley , Transduction, Genetic , Valproic Acid/pharmacology
4.
Mol Psychiatry ; 14(1): 51-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-17925795

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has been strongly implicated in the synaptic plasticity, neuronal survival and pathophysiology of depression. Lithium and valproic acid (VPA) are two primary mood-stabilizing drugs used to treat bipolar disorder. Treatment of cultured rat cortical neurons with therapeutic concentrations of LiCl or VPA selectively increased the levels of exon IV (formerly rat exon III)-containing BDNF mRNA, and the activity of BDNF promoter IV. Surprisingly, lithium- or VPA-responsive element(s) in promoter IV resides in a region upstream from the calcium-responsive elements (CaREs) responsible for depolarization-induced BDNF induction. Moreover, activation of BDNF promoter IV by lithium or VPA occurred in cortical neurons depolarized with KCl, and deletion of these three CaREs did not abolish lithium- or VPA-induced activation. Lithium and VPA are direct inhibitors of glycogen synthase kinase-3 (GSK-3) and histone deacetylase (HDAC), respectively. We showed that lithium-induced activation of promoter IV was mimicked by pharmacological inhibition of GSK-3 or short interfering RNA (siRNA)-mediated gene silencing of GSK-3alpha or GSK-3beta isoforms. Furthermore, treatment with other HDAC inhibitors, sodium butyrate and trichostatin A, or transfection with an HDAC1-specific siRNA also activated BDNF promoter IV. Our study demonstrates for the first time that GSK-3 and HDAC are respective initial targets for lithium and VPA to activate BDNF promoter IV, and that this BDNF induction involves a novel responsive region in promoter IV of the BDNF gene. Our results have strong implications for the therapeutic actions of these two mood stabilizers.


Subject(s)
Antimanic Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Lithium Chloride/pharmacology , Neurons/drug effects , Promoter Regions, Genetic/drug effects , Valproic Acid/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Membrane Potentials/drug effects , Patch-Clamp Techniques , Potassium Chloride/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Time Factors , Transfection
5.
Neuroscience ; 155(3): 567-72, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18640245

ABSTRACT

Lithium and valproic acid (VPA) are two primary drugs used to treat bipolar disorder, and have been shown to have neuroprotective properties in vivo and in vitro. A recent study demonstrated that combined treatment with lithium and VPA elicits synergistic neuroprotective effects against glutamate excitotoxicity in cultured brain neurons, and the synergy involves potentiated inhibition of glycogen synthase kinase-3 (GSK-3) activity through enhanced GSK-3 serine phosphorylation [Leng Y, Liang MH, Ren M, Marinova Z, Leeds P, Chuang DM (2008) Synergistic neuroprotective effects of lithium and valproic acid or other histone deacetylase inhibitors in neurons: roles of glycogen synthase kinase-3 inhibition. J Neurosci 28:2576-2588]. We therefore investigated the effects of lithium and VPA cotreatment on the disease symptom onset, survival time and neurological deficits in cooper zinc superoxide dismutase (SOD1) G93A mutant mice, a commonly used mouse model of amyotrophic lateral sclerosis (ALS). The G93A ALS mice received twice daily i.p. injections with LiCl (60 mg/kg), VPA (300 mg/kg) or lithium plus VPA, starting from the 30(th) day after birth and continuing until death. We found that combined treatment with lithium and VPA produced a greater and more consistent effect in delaying the onset of disease symptoms, prolonging the lifespan and decreasing the neurological deficit scores, compared with the results of monotreatment with lithium or VPA. Moreover, lithium in conjunction with VPA was more effective than lithium or VPA alone in enhancing the immunostaining of phospho-GSK-3beta(Ser9) in brain and lumbar spinal cord sections. To our knowledge, this is the first demonstration of enhanced neuroprotection by a combinatorial approach using mood stabilizers in a mouse ALS model. Our results suggest that clinical trials using lithium and VPA in combination for ALS patients are a rational strategy.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Amyotrophic Lateral Sclerosis/drug therapy , Anticonvulsants/therapeutic use , Lithium Chloride/therapeutic use , Nervous System Diseases/drug therapy , Valproic Acid/therapeutic use , Age Factors , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Drug Therapy, Combination , Glycogen Synthase Kinase 3/metabolism , Hindlimb Suspension/methods , Humans , Mice , Mice, Transgenic , Motor Activity/drug effects , Muscle Strength/drug effects , Mutation , Nervous System Diseases/etiology , Psychomotor Performance/drug effects , Reflex/drug effects , Rotarod Performance Test/methods , Superoxide Dismutase/genetics , Survival Analysis
6.
Neuroscience ; 149(1): 203-12, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17850978

ABSTRACT

Valproic acid (VPA), a widely prescribed drug for seizures and bipolar disorder, has been shown to be an inhibitor of histone deacetylase (HDAC). Our previous study has demonstrated that VPA pretreatment reduces lipopolysaccharide (LPS)-induced dopaminergic (DA) neurotoxicity through the inhibition of microglia over-activation. The aim of this study was to determine the mechanism underlying VPA-induced attenuation of microglia over-activation using rodent primary neuron/glia or enriched glia cultures. Other histone deacetylase inhibitors (HDACIs) were compared with VPA for their effects on microglial activity. We found that VPA induced apoptosis of microglia cells in a time- and concentration-dependent manner. VPA-treated microglial cells showed typical apoptotic hallmarks including phosphatidylserine externalization, chromatin condensation and DNA fragmentation. Further studies revealed that trichostatin A (TSA) and sodium butyrate (SB), two structurally dissimilar HDACIs, also induced microglial apoptosis. The apoptosis of microglia was accompanied by the disruption of mitochondrial membrane potential and the enhancement of acetylation levels of the histone H3 protein. Moreover, pretreatment with SB or TSA caused a robust decrease in LPS-induced pro-inflammatory responses and protected DA neurons from damage in mesencephalic neuron-glia cultures. Taken together, our results shed light on a novel mechanism whereby HDACIs induce neuroprotection and underscore the potential utility of HDACIs in preventing inflammation-related neurodegenerative disorders such as Parkinson's disease.


Subject(s)
Apoptosis/drug effects , Dopamine/metabolism , Enzyme Inhibitors/pharmacology , Lipopolysaccharides/toxicity , Neuroglia/drug effects , Neurons/drug effects , Valproic Acid/pharmacology , Animals , Animals, Newborn , Brain/cytology , Cell Cycle/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , In Situ Nick-End Labeling/methods , Membrane Potential, Mitochondrial/drug effects , Nitrites/metabolism , Pregnancy , Rats , Rats, Inbred F344 , Tumor Necrosis Factor-alpha/metabolism
7.
Mol Psychiatry ; 11(12): 1116-25, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16969367

ABSTRACT

Valproate (VPA), one of the mood stabilizers and antiepileptic drugs, was recently found to inhibit histone deacetylases (HDAC). Increasing reports demonstrate that VPA has neurotrophic effects in diverse cell types including midbrain dopaminergic (DA) neurons. However, the origin and nature of the mediator of the neurotrophic effects are unclear. We have previously demonstrated that VPA prolongs the survival of midbrain DA neurons in lipopolysaccharide (LPS)-treated neuron-glia cultures through the inhibition of the release of pro-inflammatory factors from microglia. In this study, we report that VPA upregulates the expression of neurotrophic factors, including glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) from astrocytes and these effects may play a major role in mediating VPA-induced neurotrophic effects on DA neurons. Moreover, VPA pretreatment protects midbrain DA neurons from LPS or 1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity. Our study identifies astrocyte as a novel target for VPA to induce neurotrophic and neuroprotective actions in rat midbrain and shows a potential new role of cellular interactions between DA neurons and astrocytes. The neurotrophic and neuroprotective effects of VPA also suggest a utility of this drug for treating neurodegenerative disorders including Parkinson's disease. Moreover, the neurotrophic effects of VPA may contribute to the therapeutic action of this drug in treating bipolar mood disorder that involves a loss of neurons and glia in discrete brain areas.


Subject(s)
Anticonvulsants/pharmacology , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Mesencephalon/metabolism , Neurons/metabolism , Valproic Acid/pharmacology , Animals , Astrocytes/cytology , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Cell Communication/drug effects , Cells, Cultured , Coculture Techniques , Female , Humans , Mesencephalon/cytology , Neurons/cytology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Rats, Inbred F344 , Up-Regulation/drug effects
8.
Pharmacogenomics J ; 5(2): 102-11, 2005.
Article in English | MEDLINE | ID: mdl-15668729

ABSTRACT

We investigated the cytoprotective effects of lithium, the mood-stabilizer, on thapsigargin-induced stress on the endoplasmic reticulum (ER) in rat PC12 cells. Protracted lithium pretreatment of PC12 cells elicited cytoprotection against thapsigargin-induced cytotoxicity. Lithium protection was concurrent with inhibition of thapsigargin-induced intracellular calcium increase and with elevated expression of the molecular chaperone GRP78. Moreover, lithium pretreatment upregulated the antiapoptotic protein Bcl-2, and blocked Bcl-2 downregulation elicited by thapsigargin. Prior to the induction of GRP78, lithium treatment alone increased the expression of c-Fos whose induction by ER stress is necessary for GRP78 induction. Curcumin, an inhibitor of transcription factor AP-1, blocked lithium cytoprotection against thapsigargin cytotoxicity. Thus, the induction of GRP78 and Bcl-2, and activation of AP-1 likely contribute to lithium-induced protection against cytotoxicity resulting from ER stress. Additionally, thapsigargin-induced cytotoxicity was suppressed by pretreatment with another mood-stabilizer, valproate, indicating that cytoprotection against ER stress is a common action of mood-stabilizing drugs.


Subject(s)
Calcium/physiology , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/toxicity , Heat-Shock Proteins/physiology , Lithium/pharmacology , Molecular Chaperones/physiology , Neuroprotective Agents , Proto-Oncogene Proteins c-bcl-2/physiology , Thapsigargin/antagonists & inhibitors , Animals , Blotting, Western , Calcium/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Cell Survival/drug effects , Curcumin/pharmacology , Electrophoretic Mobility Shift Assay , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , PC12 Cells , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Thapsigargin/toxicity , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/genetics , Valproic Acid/pharmacology
9.
Neurochem Int ; 46(1): 61-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15567516

ABSTRACT

Neurotrophin-induced neuroprotection against apoptosis was investigated using immature cultured cerebellar granule cells (CGC) from newborn rat pups. Apoptotic cell death induced by treatment with cytosine arabinoside (AraC) was confirmed by DNA fragmentation and quantified by cell survival assays. AraC was most effective in inducing apoptosis when added to CGC on the day of culture preparation, while less or no effect was observed when added at 24 or 48h after plating, respectively. Pretreatment of CGC cultures for 24h with brain-derived neurotrophic factor (BDNF) or neurotrophin-4 (NT-4), but not neurotrophin-3 (NT-3), robustly protected against AraC neurotoxicity. K252a, an inhibitor of the tropomyosin-related kinase (Trk) tyrosine kinase receptor family which showed no toxicity by itself, blocked BDNF protection of AraC-induced apoptosis in a concentration-dependent manner. Neither protein kinase C activation nor inhibition mimicked or affected BDNF protection against AraC neurotoxicity. BDNF, but not NT-3, treatment of immature CGC caused a marked, but transient activation of Akt through phosphatidylinositol (PI) 3-kinase. The neuroprotective effects of BDNF were suppressed by pretreatment with LY 294002 (a PI 3-kinase inhibitor). BDNF neuroprotection was also preceded by activation of mitogen activated protein kinase (MAPK) and suppressed by two MAPK/ERK (MEK)-selective inhibitors, PD 98059 and U-0126. Moreover, inhibitors of PI 3-kinase and MEK potentiated AraC-induced neurotoxicity. These results show that neurotrophins protect against AraC-induced apoptosis, at least in part, through TrkB-mediated activation of the PI 3-kinase/Akt and MEK signaling pathways.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Apoptosis/drug effects , Cerebellum/cytology , Cytarabine/antagonists & inhibitors , Cytarabine/toxicity , Nerve Growth Factors/pharmacology , Neurons/drug effects , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/pharmacology , Cell Survival/drug effects , Cerebellum/drug effects , Cytoplasmic Granules/drug effects , DNA Fragmentation , MAP Kinase Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley
10.
Pharmacogenomics J ; 4(5): 336-44, 2004.
Article in English | MEDLINE | ID: mdl-15289798

ABSTRACT

Valproic acid (VPA), used to treat bipolar mood disorder and seizures, also inhibits histone deacetylase (HDAC). Here, we found that VPA and other HDAC inhibitors, butyrate and trichostatin A, robustly protected mature cerebellar granule cell cultures from excitotoxicity induced by SYM 2081 ((2S, 4R)-4-methylglutamate), an inhibitor of excitatory amino-acid transporters and an agonist of low-affinity kainate receptors. These neuroprotective effects required protracted treatment and were correlated with enhanced acetylated histone levels, indicating HDAC inhibition. SYM-induced excitotoxicity was blocked by MK-801 ((5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate), supporting that the toxicity was largely N-methyl-D-aspartate receptor dependent. SYM excitotoxicity had apoptotic characteristics and was prevented by a caspase inhibitor. SYM-induced apoptosis was associated with a rapid and robust nuclear accumulation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a housekeeping gene previously shown to be proapoptotic. VPA pretreatment suppressed SYM 2081-induced GAPDH nuclear accumulation, concurrent with its neuroprotective effects. Chromatin immunoprecipitation (ChIP) revealed that GAPDH is copresent with acetylated histone H3, including Lys9-acetylated histone, and that VPA treatment caused a time-dependent decrease in the levels of nuclear GAPDH with a concomitant increase in acetylated histones in the ChIP complex. Our results strongly suggest that VPA protects neurons from excitotoxicity through inhibition of HDAC activity and that this protective effect may involve suppression of excitotoxicity-induced accumulation of GAPDH protein in the nucleus.


Subject(s)
Apoptosis/drug effects , Cell Nucleus/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Histone Deacetylase Inhibitors , Valproic Acid/pharmacology , Animals , Apoptosis/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Nucleus/enzymology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists , Glutamates/toxicity , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Histone Deacetylases/metabolism , Neurons/drug effects , Neurons/enzymology , Rats , Rats, Sprague-Dawley
11.
Mol Psychiatry ; 9(4): 371-85, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14702090

ABSTRACT

We assessed the ability of lithium to reduce neurodegeneration and to stimulate cell proliferation in a rat model of Huntington's disease in which quinolinic acid (QA) was unilaterally infused into the striatum. LiCl (0.5-3.0 mEq/kg) was injected subcutaneously 24 h before and 1 h after QA infusion. At 7 days after QA injection, lithium significantly diminished the loss of neurons immunostained for Neuronal Nuclei (NeuN) in the injured striatum, but failed to prevent the reduction of NADPH-diaphorase-positive striatal interneurons. Lithium also reduced the number of neurons showing DNA damage or activated caspase-3. This neuroprotection was associated with an upregulation of Bcl-2 protein levels in the striatal tissue and an increase in the number and density of Bcl-2 immunostaining in striatal neurons. Bromodeoxyuridinie (BrdU) labeling in the lithium-treated injured striatum revealed the presence of large numbers of proliferating cells near the QA-injection site, with a reduction of BrdU-labeled cells in the subventricular zone (SVZ). All BrdU-labeled cells in the SVZ and the majority of BrdU-labeled cells near the QA-injection site were negative for either NeuN or glial fibrillary acidic protein (GFAP), suggesting that they are undifferentiated progenitor cells. However, a small number of BrdU-positive cells found in the QA-injected and lithium-treated striatum site were positive for either NeuN or GFAP. Our results suggest that lithium is neuroprotective in the QA-injection model of Huntington's disease not only due to its ability to inhibit apoptosis but also because it can stimulate neuronal and astroglial progenitor proliferation in the QA-injected striatum or their migration from the SVZ.


Subject(s)
Corpus Striatum/drug effects , Huntington Disease/drug therapy , Huntington Disease/pathology , Lithium/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Corpus Striatum/cytology , Corpus Striatum/enzymology , DNA Damage/drug effects , Dose-Response Relationship, Drug , Huntington Disease/chemically induced , Huntington Disease/metabolism , Interneurons/drug effects , Male , Neurons/enzymology , Neurotoxins , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinolinic Acid , Rats , Rats, Sprague-Dawley , Up-Regulation
12.
Neuroscience ; 117(1): 55-61, 2003.
Article in English | MEDLINE | ID: mdl-12605892

ABSTRACT

The number of neurons in the brain is controlled by production of new neurons and neuronal death. Neural progenitor proliferation in the developing and adult brain plays a prominent role in the production of new neurons. Here, we examined the effects of lithium, a mood-stabilizing drug, on neuronal proliferation in rat primary neuronal cultures. The incorporation of 5-bromo-2'-deoxyuridine (BrdU) into replicating DNA was used to label proliferating cells. BrdU incorporation was detected by immunocytochemistry in cerebellar granule cells prepared from postnatal rats and cerebral cortical cultures prepared from embryonic rats. Quantification of BrdU incorporation into cultures was performed by counting BrdU-positive cells and BrdU-coupled enzyme-linked immunosorbent assay. Both methods revealed that lithium increased BrdU incorporation in cerebellar granule cells and cerebral cortical cultures. Most BrdU-positive cells colocalized with nestin, a neuroblast cell marker, in cerebral cortical cultures. Blockade of DNA replication by cytosine arabinoside almost completely abolished BrdU incorporation, suggesting that lithium-induced BrdU incorporation was mainly due to enhanced DNA replication. Glutamate, glucocorticoids and haloperidol were found to markedly reduce neural progenitor proliferation in cerebellar granule cells. The presence of lithium prevented the loss of proliferation induced by these agents. Lithium-induced neural progenitor proliferation in vitro suggests that similar effects might occur in vivo and this action could also be related to its clinical efficacy. Cultured brain neurons may provide a valuable model for studying the molecular mechanisms underlying lithium-induced up-regulation of neural proliferation.


Subject(s)
Cerebellum/drug effects , Cerebral Cortex/drug effects , Lithium/pharmacology , Neurons/drug effects , Stem Cells/drug effects , Animals , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Dose-Response Relationship, Drug , Neurons/cytology , Rats , Stem Cells/cytology
13.
Neuroscience ; 116(2): 425-35, 2003.
Article in English | MEDLINE | ID: mdl-12559097

ABSTRACT

The cyclic AMP response element binding protein (CREB) has major roles in mediating adaptive responses at glutamatergic synapses and in the neuroprotective effects of neurotrophins. CREB has been implicated as a potential mediator of antidepressant actions. In vitro, chronic lithium treatment has been shown to promote neuronal cell survival. In the present study, we have used cultures of cerebellar granule neurons to analyze the effects of acute and chronic lithium treatment on the response to toxic concentrations of glutamate. Such concentrations of glutamate decrease the phosphorylation of CREB at serine(133) in an N-methyl-D-aspartate (NMDA) receptor-dependent manner. Chronic, but not acute, lithium treatment suppresses glutamate-induced decreases in phosphorylated CREB, and transfection studies indicate that chronic lithium, in the presence of a glutamate stimulus, markedly increases CRE-driven gene expression. Experiments with selected pharmacological reagents indicate that the glutamate-induced decreases in phosphorylated CREB are regulated primarily by protein phosphatase 1. Chronic lithium treatment not only decreases protein phosphatase 1 activity under these circumstances, but also augments glutamate-induced increases in MEK activity. PD 98059, a MEK inhibitor, prevents chronic lithium treatment from increasing phosphorylated CREB levels in glutamate-treated neurons. We conclude from these results that chronic lithium treatment is permissive for maintaining higher phosphorylated CREB levels in the presence of glutamate in part by decreasing protein phosphatase 1 activity and in part by increasing MEK activity. Higher levels of phosphorylated CREB and CRE-responsive genes such as bcl-2 may be responsible for lithium's reported effects on neuronal survival.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Glutamic Acid/pharmacology , Lithium/pharmacology , Neurons/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Cells, Cultured , Cerebellum/cytology , Chromones/pharmacology , Dizocilpine Maleate/pharmacology , Drug Interactions , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Neurons/drug effects , Phosphorylation , Protein Phosphatase 1 , Rats , Rats, Sprague-Dawley
14.
Neuroscience ; 106(3): 603-12, 2001.
Article in English | MEDLINE | ID: mdl-11591460

ABSTRACT

Huntington's disease is a progressive, inherited neurodegenerative disorder characterized by the loss of subsets of neurons primarily in the striatum. In this study, we assessed the neuroprotective effect of lithium against striatal lesion formation in a rat model of Huntington's disease in which quinolinic acid was unilaterally infused into the striatum. For this purpose, we used a dopamine receptor autoradiography and glutamic acid decarboxylase mRNA in situ hybridization analysis, methods previously shown to be adequate for quantitative analysis of the excitotoxin-induced striatal lesion size. Here we demonstrated that subcutaneous injections of LiCl for 16 days prior to quinolinic acid infusion considerably reduced the size of quinolinic acid-induced striatal lesion. Furthermore, these lithium pre-treatments also decreased the number of striatal neurons labeled with the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay. Immunohistochemistry and western blotting demonstrated that lithium-elicited neuroprotection was associated with an increase in Bcl-2 protein levels. Our results raise the possibility that lithium may be considered as a neuroprotective agent in treatment of neurodegenerative diseases such as Huntington's disease.


Subject(s)
Cell Death/drug effects , Huntington Disease/drug therapy , Lithium/pharmacology , Neostriatum/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/antagonists & inhibitors , Animals , Antimanic Agents/pharmacology , Benzazepines/pharmacokinetics , Cell Death/physiology , Cyclin D1/drug effects , Cyclin D1/metabolism , Disease Models, Animal , Dopamine Antagonists/pharmacokinetics , Glutamate Decarboxylase/genetics , Huntington Disease/chemically induced , Huntington Disease/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Isoenzymes/genetics , Lithium Chloride/pharmacology , Male , Neostriatum/pathology , Neostriatum/physiopathology , Neurons/metabolism , Neurons/pathology , Quinolinic Acid/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/metabolism
15.
J Pharmacol Exp Ther ; 297(1): 78-87, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259530

ABSTRACT

Prostaglandin A(1) (PGA1) reportedly inhibits NF-kappaB activation and induces expression of heat shock proteins. Since both these effects could be neuroprotective, the therapeutic potential of PGA1 in neurodegenerative disorders, where excitotoxicity may contribute to pathogenesis, was evaluated in rat striatal neurons exposed to the N-methyl-D-aspartate (NMDA) receptor agonist quinolinic acid (QA). Intrastriatal administration of PGA1 (5-80 nmol) attenuated QA (60 nmol)-induced internucleosomal DNA fragmentation. The inhibitory effects of a single dose of PGA1 (80 nmol) on QA (60 nmol)-induced DNA fragmentation were observed 12 to 48 h after treatment. PGA1 (80 nmol) also attenuated QA-induced DNA fragmentation when administered up to 4 h after QA exposure. PGA1 significantly decreased the loss of D1 dopamine receptors and GAD(67) mRNA in QA-injected striatum as measured by quantitative receptor autoradiography and in situ hybridization histochemistry, suggesting that it reduced the neuronal loss induced by QA. Protection of striatal neurons against QA-induced death by PGA1 was further indicated by Nissl staining 10 days after QA administration. PGA1 (5-80 nmol) significantly inhibited QA-induced NF-kappaB activation by blocking inhibitory kappaB-alpha degradation but had no effect on activator protein-1 binding activity. PGA1 (80 nmol) treatment substantially increased 70- and 72-kDa heat shock protein levels in striatum. These results indicate that PGA1 blunts NMDA receptor-mediated neuronal apoptosis by a mechanism possibly involving the up-regulation of neuroprotective heat shock proteins and inhibition of NF-kappaB activation. In view of its potent neuroprotective activity, PGA1 could prove useful in the treatment of certain neurodegenerative disorders related to excitotoxicity.


Subject(s)
Corpus Striatum/drug effects , Prostaglandins A/pharmacology , Quinolinic Acid/toxicity , Animals , Apoptosis/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , DNA Fragmentation/drug effects , HSP70 Heat-Shock Proteins/biosynthesis , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nucleosomes/drug effects , Rats , Rats, Sprague-Dawley
16.
J Neurochem ; 75(1): 81-90, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10854250

ABSTRACT

We have studied neurotoxicity induced by pharmacological concentrations of 3-hydroxykynurenine (3-HK), an endogenous toxin implicated in certain neurodegenerative diseases, in cerebellar granule cells, PC12 pheochromocytoma cells, and GT1-7 hypothalamic neurosecretory cells. In all three cell types, the toxicity was induced in a dose-dependent manner by 3-HK at high micromolar concentrations and had features characteristic of apoptosis, including chromatin condensation and internucleosomal DNA cleavage. In cerebellar granule cells, the 3-HK neurotoxicity was unaffected by xanthine oxidase inhibitors but markedly potentiated by superoxide dismutase and its hemelike mimetic, MnTBAP [manganese(III) tetrakis(benzoic acid)porphyrin chloride]. Catalase blocked 3-HK neurotoxicity in the absence and presence of superoxide dismutase or MnTBAP. The formation of H(2)O(2) was demonstrated in PC12 and GT1-7 cells treated with 3-HK, by measuring the increase in the fluorescent product, 2',7'-dichlorofluorescein. In both PC12 and cerebellar granule cells, inhibitors of the neutral amino acid transporter that mediates the uptake of 3-HK failed to block 3-HK toxicity. However, their toxicity was slightly potentiated by the iron chelator, deferoxamine. Taken together, our results suggest that neurotoxicity induced by pharmacological concentrations of 3-HK in these cell types is mediated primarily by H(2)O(2), which is formed most likely by auto-oxidation of 3-HK in extracellular compartments. 3-HK-induced death of PC12 and GT1-7 cells was protected by dantrolene, an inhibitor of calcium release from the endoplasmic reticulum. The protection by dantrolene was associated with a marked increase in the protein level of Bcl-2, a prominent antiapoptotic gene product. Moreover, overexpression of Bcl-2 in GT1-7 cells elicited by gene transfection suppressed 3-HK toxicity. Thus, dantrolene may elicit its neuroprotective effects by mechanisms involving up-regulation of the level and function of Bcl-2 protein.


Subject(s)
Apoptosis/drug effects , Dantrolene/pharmacology , Gene Expression , Kynurenine/analogs & derivatives , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Cerebellum/cytology , Cerebellum/metabolism , Chromatin/ultrastructure , DNA Fragmentation , Drug Synergism , Enzyme Inhibitors/pharmacology , Free Radical Scavengers , Iron Chelating Agents/pharmacology , Kynurenine/pharmacology , Metalloporphyrins/pharmacology , PC12 Cells , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/pharmacology , Xanthine Oxidase/antagonists & inhibitors
18.
J Neurosci ; 19(21): 9654-62, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10531467

ABSTRACT

We recently reported that cytosine arabinoside (AraC)-induced apoptosis of cerebellar neurons involves the overexpression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The present study was undertaken to investigate whether p53 and/or Bax overexpression participates in the AraC-induced apoptosis of cerebellar granule cells and, if so, the relationship between p53 induction and GAPDH overexpression in these cells. AraC-induced apoptosis of cerebellar granule cells was preceded by an increase in levels of p53 mRNA and protein detected between 1 and 8 hr after treatment. The mRNA level for a p53 target gene, Bax, was also increased. The increase in GAPDH mRNA lasted longer than that of either p53 or Bax, and the level of GAPDH protein in the particulate fraction increased after induction of GAPDH mRNA. The antisense oligonucleotide to p53 protected granule cells from AraC-induced chromatin condensation, internucleosomal cleavage, and apoptotic death. The inhibition of p53 expression by the p53 antisense oligonucleotide not only blocked the expression of Bax but also partially suppressed the increased GAPDH mRNA and protein levels. Conversely, the suppression of GAPDH expression and subsequent attenuation of apoptosis of granule cells by GAPDH antisense oligonucleotide did not influence the expression of p53 or Bax. Cerebellar granule cells prepared from p53 knock-out mice were resistant to AraC toxicity, and the p53 gene knock-out suppressed AraC-upregulated GAPDH expression. Moreover, infection of PC12 cells with an adenoviral vector containing p53 gene dramatically increased GAPDH expression and triggered cell apoptosis. These results suggest that AraC-induced apoptosis of cerebellar granule cells involves the expression of both GAPDH and p53 and that, similar to Bax, GAPDH is upregulated by p53 after exposure to the apoptotic insult.


Subject(s)
Apoptosis/physiology , Cerebellum/cytology , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Neurons/cytology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/metabolism , Animals , Cerebellum/physiology , Cytarabine/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation, Enzymologic , Genes, p53/drug effects , Mice , Mice, Knockout , Oligodeoxyribonucleotides, Antisense/pharmacology , PC12 Cells , Proto-Oncogene Proteins/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Transfection , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
19.
Proc Natl Acad Sci U S A ; 96(15): 8745-50, 1999 Jul 20.
Article in English | MEDLINE | ID: mdl-10411946

ABSTRACT

This report describes a modulatory action of lithium and glutamate on the activity of serine/threonine kinase Akt-1. Lithium is most commonly used to treat bipolar disorder, but the mechanism of its therapeutic action remains unknown. We have recently demonstrated that lithium protects against glutamate-induced excitotoxicity in cultured brain neurons and in an animal model of cerebral ischemia. This study was undertaken to investigate the role of Akt-1, activated by the phosphatidylinositol 3-kinase (PI 3-K) signaling pathway, in mediating glutamate excitotoxicity and lithium protection in cerebellar granule cells. High levels of phosphorylation and activity of Akt-1 were detected in cerebellar neurons cultured in the presence of serum. Protracted treatment with selective PI 3-K inhibitors, wortmannin and LY294002, abolished Akt-1 activity and induced neuronal death that could be reduced by long-term lithium pretreatment. Exposure of cells to glutamate induced a rapid and reversible loss of Akt-1 phosphorylation and kinase activity. These effects were closely correlated with excitotoxicity and caspase 3 activation and were prevented by phosphatase inhibitors, okadaic acid and caliculin A. Long-term lithium pretreatment suppressed glutamate-induced loss of Akt-1 activity and accelerated its recovery toward the control levels. Lithium treatment alone induced rapid increase in PI 3-K activity, and Akt-1 phosphorylation with accompanying kinase activation, which was blocked by PI 3-K inhibitors. Lithium also increased the phosphorylation of glycogen synthase kinase-3 (GSK-3), a downstream physiological target of Akt. Thus, modulation of Akt-1 activity appears to play a key role in the mechanism of glutamate excitotoxicity and lithium neuroprotection.


Subject(s)
Cerebellum/metabolism , Glutamic Acid/pharmacology , Lithium/pharmacology , Neurons/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Androstadienes/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Chromones/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Wortmannin
20.
Biol Psychiatry ; 46(2): 247-55, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10418700

ABSTRACT

BACKGROUND: A number of investigators have reported finding elevated basal and stimulated intracellular calcium levels in the platelets or lymphocytes of bipolar disorder patients. METHODS: Intracellular calcium was measured by a micro fura-2 fluorometric method in the platelets and lymphocytes of 30 affective disorder patients and 14 control subjects. RESULTS: We observed significantly elevated basal calcium concentrations in bipolar patient platelets and lymphocytes compared to control subjects. Bipolar patient platelet calcium responses to thrombin, serotonin, and thapsigargin were also significantly greater than control subjects. The peak calcium levels of lymphocytes of bipolar patients were greater than control subjects only when stimulated by thapsigargin. There were significant differences between bipolar and unipolar patients in basal and thapsigargin-stimulated calcium measures but not between bipolar I and bipolar II patients. Unmedicated versus medicated calcium measures were not significantly different. We also found little correlation between calcium measures and the severity of mood rating. CONCLUSIONS: Using this method, we were able to confirm and extend the work of others, indicating altered intracellular calcium homeostasis in the blood cells of bipolar disorder patients. In addition, our data suggest that storage operated calcium channels may be the source of the elevated intracellular calcium in platelets and lymphocytes of bipolar patients.


Subject(s)
Basal Metabolism/physiology , Bipolar Disorder/blood , Blood Platelets/metabolism , Calcium/blood , Enzyme Inhibitors/pharmacokinetics , Fluorometry/methods , Lymphocytes/metabolism , Thapsigargin/pharmacokinetics , Adult , Calcium Channels/metabolism , Equipment Design , Female , Humans , Ion Transport/physiology , Male , Platelet Activation/drug effects , Prospective Studies , Retrospective Studies , Thapsigargin/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...