Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Bioorg Med Chem ; 85: 117276, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37037115

ABSTRACT

Many non-nucleoside human cytomegalovirus (HCMV) inhibitors have been reported in patent and scientific literature, however, none have reached commercialization despite the urgent need for new HCMV treatments. Herein we report select compounds from different templates that all had low micromolar human ether-à-go-go (hERG) ion channel IC50 values. We also describe a series of pyrroloquinoline derivatives that were designed and synthesized to understand the effect of various substitution on human cytomegalovirus (HCMV) polymerase activity, antiviral activity, and hERG inhibition. These results demonstrated that hERG inhibition can be significantly altered based on the substitution on this template. An HCMV inhibitor with low hERG inhibition and reduced cytotoxicity is also described. The results suggest substitution can be fine tuned for the non-nucleoside polymerase inhibitors to reduce hERG inhibition and maintain HCMV antiviral potency.


Subject(s)
Antiviral Agents , Cytomegalovirus , Humans , Antiviral Agents/pharmacology , Ether/pharmacology , Ether-A-Go-Go Potassium Channels , Cardiotoxicity , Ethyl Ethers/pharmacology , Nucleotidyltransferases , Ethers/pharmacology , ERG1 Potassium Channel , Potassium Channel Blockers/pharmacology
2.
Biotechnol Bioeng ; 118(12): 4687-4698, 2021 12.
Article in English | MEDLINE | ID: mdl-34478150

ABSTRACT

Drug-induced liver injury (DILI) is a leading cause of therapy failure in the clinic and also contributes much to acute liver failure cases. Investigations of predictive sensitivity in animal models have limitations due to interspecies differences. Previously reported in vitro models of liver injury based on primary human hepatocytes (PHHs) cannot meet the requirements of high physiological fidelity, low cost, simple operation, and high throughput with improved sensitivity. Herein, we developed an integrated biomimetic array chip (iBAC) for establishing extracellular matrix (ECM)-based models. A collagen-based 3D PHH model was constructed on the iBAC as a case for the prediction of clinical DILI at throughput. The iBAC has a three-layer structure with a core component of 3D implanting holes. At an initial cell seeding numbers of 5000-10,000, the collagen-based 3D PHH model was optimized with improved and stabilized liver functionality, including cell viability, albumin, and urea production. Moreover, basal activities of most metabolic enzymes on the iBAC were maintained for at least 12 days. Next, a small-scale hepatotoxicity screening indicated that the 3D PHH model on the iBAC was more sensitive for predicting hepatotoxicity than the 2D PHH model on the plate. Finally, a large-scale screening of liver toxicity using 122 clinical drugs further demonstrated that the collagen-based 3D PHH model on the iBAC had superior predictive sensitivity compared to all previously reported in vitro models. These results indicated the importance of 3D collagen for liver physiological functionality and hepatotoxicity prediction. We anticipant it being a promising tool for risk assessment of drug-induced hepatotoxicity with a widespread acceptance in drug industry.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Drug Evaluation, Preclinical , Hepatocytes , Lab-On-A-Chip Devices , Models, Biological , Biomimetics , Cells, Cultured , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects
3.
SLAS Discov ; 26(3): 364-372, 2021 03.
Article in English | MEDLINE | ID: mdl-32914673

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been widely used for the assessment of drug proarrhythmic potential through multielectrode array (MEA). HiPSC-CM cultures beat spontaneously with a wide range of frequencies, however, which could affect drug-induced changes in repolarization. Pacing hiPSC-CMs at a physiological heart rate more closely resembles the state of in vivo ventricular myocytes and permits the standardization of test conditions to improve consistency. In this study, we systematically investigated the time window of stable ion currents in high-purity hiPSC-derived ventricular cardiomyocytes (hiPSC-vCMs) and confirmed that these cells could be used to correctly predict the proarrhythmic risk of Comprehensive In Vitro Proarrhythmia Assay (CiPA) reference compounds. To evaluate drug proarrhythmic potentials at a physiological beating rate, we used a MEA to electrically pace hiPSC-vCMs, and we recorded regular field potential waveforms in hiPSC-vCMs treated with DMSO and 10 CiPA reference drugs. Prolongation of field potential duration was detected in cells after exposure to high- and intermediate-risk drugs; in addition, drug-induced arrhythmia-like events were observed. The results of this study provide a simple and feasible method to investigate drug proarrhythmic potentials in hiPSC-CMs at a physiological beating rate.


Subject(s)
Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/drug effects , Phenethylamines/adverse effects , Quinidine/adverse effects , Sulfonamides/adverse effects , Action Potentials/physiology , Arrhythmias, Cardiac/prevention & control , Calcium/metabolism , Cations, Divalent , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Ion Transport/drug effects , Microelectrodes , Models, Biological , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Nifedipine/pharmacology , Patch-Clamp Techniques , Primary Cell Culture , Sotalol/adverse effects , Tetrodotoxin/antagonists & inhibitors , Tetrodotoxin/toxicity , Verapamil/pharmacology
4.
Bioorg Med Chem Lett ; 27(22): 4979-4984, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29037948

ABSTRACT

hNav1.7 receives a lot of attention owing to its attractive mechanism of action in pain processing pathway. We have previously reported our design of a novel series of tetrahydropyridine analogues towards hNav1.7 selective inhibitors. Herein, we disclose further efforts to the optimization of hit compound (-)-6, which led to the identification of aminocyclohexene analogues (-)-9 and (-)-17 with good potency, high selectivity, and minimal CYP inhibition. Both compounds (-)-9 and (-)-17 demonstrated improved pharmacokinetic profiles in rats, and robust efficacy in rat formalin-induced nociception and spinal nerve ligation (SNL) models.


Subject(s)
Analgesics/chemistry , Cyclohexenes/chemistry , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Administration, Oral , Analgesics/pharmacokinetics , Analgesics/therapeutic use , Animals , Binding Sites , Cyclohexenes/pharmacokinetics , Cyclohexenes/therapeutic use , Cytochrome P-450 CYP2C9/chemistry , Cytochrome P-450 CYP2C9/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Half-Life , Inhibitory Concentration 50 , Molecular Docking Simulation , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Protein Structure, Tertiary , Rats , Stereoisomerism , Structure-Activity Relationship , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/therapeutic use
5.
Bioorg Med Chem Lett ; 27(10): 2210-2215, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28385504

ABSTRACT

hNav1.7 small molecular inhibitors have attracted lots of attention by its unique analgesic effect. Herein, we report the design and synthesis of a novel series of tetrahydropyridine analogs as hNav1.7 inhibitors for analgesia. Detail structural-activity relationship (SAR) studies were undertaken towards improving hNav1.7 activity, in vitro ADME, and in vivo PK profiles. These efforts resulted in the identification of compound (-)-15h, a highly potent and selective hNav1.7 inhibitor with good ADME and PK profiles.


Subject(s)
Analgesics/chemistry , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pyridines/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Analgesics/chemical synthesis , Analgesics/pharmacokinetics , Animals , Binding Sites , Cytochrome P-450 CYP2C9/chemistry , Cytochrome P-450 CYP2C9/metabolism , Drug Design , Half-Life , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Protein Structure, Tertiary , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Rats , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/chemical synthesis , Voltage-Gated Sodium Channel Blockers/pharmacokinetics
6.
Stem Cell Res ; 19: 94-103, 2017 03.
Article in English | MEDLINE | ID: mdl-28110125

ABSTRACT

Most existing culture media for cardiac differentiation of human pluripotent stem cells (hPSCs) contain significant amounts of albumin. For clinical transplantation applications of hPSC-derived cardiomyocytes (hPSC-CMs), culturing cells in an albumin containing environment raises the concern of pathogen contamination and immunogenicity to the recipient patients. In addition, batch-to-batch variation of albumin may cause the inconsistent of hPSC cardiac differentiation. Here, we demonstrated that antioxidants l-ascorbic acid, trolox, N-acetyl-l-cysteine (NAC) and sodium pyruvate could functionally substitute albumin in the culture medium, and formulated an albumin-free, chemical-defined medium (S12 medium). We showed that S12 medium could support efficient hPSC cardiac differentiation with significantly improved reproducibility, and maintained long-term culture of hPSC-CMs. Furthermore, under chemical-defined and albumin-free conditions, human-induced pluripotent stem cells (hiPSCs) were established, and differentiated into highly homogenous atrial and ventricular myocytes in a scalable fashion with normal electrophysiological properties. Finally, we characterized the activity of three typical cardiac ion channels of those cells, and demonstrated that hPSC-derived ventricular cardiomyocytes (hPSC-vCMs) were suitable for drug cardiac safety evaluation. In summary, this simplified, chemical-defined and albumin-free culture medium supports efficient generation and maintaining of hPSC-CMs and facilitates both research and clinical applications of these cells.


Subject(s)
Culture Media/chemistry , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Action Potentials/drug effects , Antioxidants/pharmacology , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Microscopy, Fluorescence , Myocytes, Cardiac/metabolism , Nifedipine/pharmacology , Patch-Clamp Techniques , Piperidines/pharmacology , Pluripotent Stem Cells/metabolism , Pyridines/pharmacology , Tretinoin/pharmacology
7.
Mol Brain ; 7: 15, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24594013

ABSTRACT

BACKGROUND: Excess expression of acetylcholinesterase (AChE) in the cortex and hippocampus causes a decrease in the number of glutamatergic synapses and alters the expression of neurexin and neuroligin, trans-synaptic proteins that control synaptic stability. The molecular sequence and three-dimensional structure of AChE are homologous to the corresponding aspects of the ectodomain of neuroligin. This study investigated whether excess AChE interacts physically with neurexin to destabilize glutamatergic synapses. RESULTS: The results showed that AChE clusters colocalized with neurexin assemblies in the neurites of hippocampal neurons and that AChE co-immunoprecipitated with neurexin from the lysate of these neurons. Moreover, when expressed in human embryonic kidney 293 cells, N-glycosylated AChE co-immunoprecipitated with non-O-glycosylated neurexin-1ß, with N-glycosylation of the AChE being required for this co-precipitation to occur. Increasing extracellular AChE decreased the association of neurexin with neuroligin and inhibited neuroligin-induced synaptogenesis. The number and activity of excitatory synapses in cultured hippocampal neurons were reduced by extracellular catalytically inactive AChE. CONCLUSIONS: Excessive glycosylated AChE could competitively disrupt a subset of the neurexin-neuroligin junctions consequently impairing the integrity of glutamatergic synapses. This might serve a molecular mechanism of excessive AChE induced neurodegeneration.


Subject(s)
Acetylcholinesterase/metabolism , Glutamates/metabolism , Hippocampus/cytology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cell Membrane/metabolism , Glycosylation , HEK293 Cells , Humans , Immunoprecipitation , Ligands , Models, Biological , Protein Binding , RNA Splicing , Rats , Rats, Wistar
8.
Bioorg Med Chem Lett ; 22(12): 4153-8, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22579422

ABSTRACT

We previously reported the small organic N-type calcium channel blocker NP078585 that while efficacious in animal models for pain, exhibited modest L-type calcium channel selectivity and substantial off-target inhibition against the hERG potassium channel. Structure-activity studies to optimize NP078585 preclinical properties resulted in compound 16, which maintained high potency for N-type calcium channel blockade, and possessed excellent selectivity over the hERG (~120-fold) and L-type (~3600-fold) channels. Compound 16 shows significant anti-hyperalgesic activity in the spinal nerve ligation model of neuropathic pain and is also efficacious in the rat formalin model of inflammatory pain.


Subject(s)
Analgesics/chemical synthesis , Calcium Channel Blockers/chemical synthesis , Calcium Channels, N-Type/metabolism , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Piperazines/chemical synthesis , Spinal Nerves/drug effects , Analgesics/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Disease Models, Animal , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Nerves/metabolism , Structure-Activity Relationship
9.
Assay Drug Dev Technol ; 7(3): 266-80, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19530894

ABSTRACT

T-type voltage-gated Ca(2+) channels have been implicated in contributing to a broad variety of human disorders, including pain, epilepsy, sleep disturbances, cardiac arrhythmias, and certain types of cancer. However, potent and selective T-type Ca(2+) channel modulators are not yet available for clinical use. This may in part be due to their unique biophysical properties that have delayed the development of high-throughput screening (HTS) assays for identifying blockers. One notable challenge is that at the normal resting membrane potential (V(m)) of cell lines commonly utilized for drug screening purposes, T-type Ca(2+) channels are largely inactivated and thus cannot be supported by typical formats of functional HTS assays to both evoke and quantify the Ca(2+) channel signal. Here we describe a simple method that can successfully support a fluorescence-based functional assay for compounds that modulate T-type Ca(2+)channels. The assay functions by exploiting the pore-forming properties of gramicidin to control the cellular V(m) in advance of T-type Ca(2+) channel activation. Using selected ionic conditions in the presence of gramicidin, T-type Ca(2+) channels are converted from the unavailable, inactivated state to the available, resting state, where they can be subsequently activated by application of extracellular K(+). The fidelity of the assay has been pharmacologically characterized with sample T-type Ca(2+) channel blockers whose potency has been determined by conventional manual patch-clamp techniques. This method has the potential for applications in high-throughput fluorometric imaging plate reader (FLIPR(R), Molecular Devices, Sunnyvale, CA) formats with cell lines expressing either recombinant or endogenous T-type Ca(2+) channels.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Algorithms , Calcium Channel Blockers/chemical synthesis , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Channels, R-Type/drug effects , Calcium Channels, R-Type/metabolism , Calcium Channels, T-Type/metabolism , Cation Transport Proteins/drug effects , Cation Transport Proteins/metabolism , Cell Line , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Drug Evaluation, Preclinical , Electrophysiology , Gramicidin/pharmacology , Humans , Membrane Potentials/drug effects , Patch-Clamp Techniques , Pharmaceutical Solutions , Spectrometry, Fluorescence
10.
Int J Physiol Pathophysiol Pharmacol ; 1(1): 64-75, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-21383879

ABSTRACT

Versican is a chondroitin sulfate proteoglycan whose isoforms are differentially expressed, but little is known of their functions in the neuronal system. Here we show that isoforms of versican play different roles in neuronal differentiation and neurite outgrowth. Expression of versican V1 isoform in PC12 cells induced complete neuronal differentiation and increased the expression of nicotinic acetylcholine receptor in NGF-independent manners. The V1-induced neuronal differentiation was different from the NGF-induced differentiation, showing a specific profile of nAChR subunit expression and distinct kinetics of receptor-gated channel activity. Our results have implications for understanding how versican regulates neuronal development, function and repair.

11.
Neurodegener Dis ; 4(2-3): 171-84, 2007.
Article in English | MEDLINE | ID: mdl-17596712

ABSTRACT

BACKGROUND: Peripheral anionic site (PAS) blockade of acetylcholinesterase (AChE) notably affects neuronal activity and cyto-architecture, however, the mechanism(s) involved are incompletely understood. OBJECTIVE: We wished to specify the PAS extracellular effects on specific AChE mRNA splice variants, delineate the consequent cellular remodeling events, and explore the inhibitory effects on interchanging RACK1 interactions. METHODS: We exposed rat hippocampal cultured neurons to BW284C51, the peripheral anionic site inhibitor of AChE, and to the non-selective AChE active site inhibitor, physostigmine for studying the neuronal remodeling of AChE mRNA expression and trafficking. RESULTS: BW284C51 induced overexpression of both AChE splice variants, yet promoted neuritic translocation of the normally rare AChE-R, and retraction of AChE-S mRNA in an antisense-suppressible manner. BW284C51 further caused modest decreases in the expression of the scaffold protein RACK1 (receptor for activated protein kinase betaII), followed by drastic neurite retraction of both RACK1 and the AChE homologue neuroligin1, but not the tubulin-associated MAP2 protein. Accompanying BW284C51 effects involved decreases in the Fyn kinase and membrane insertion of the glutamate receptor NR2B variant and impaired glutamatergic activities of treated cells. Intriguingly, molecular modeling suggested that direct, non-catalytic competition with Fyn binding by the RACK1-interacting AChE-R variant may be involved. CONCLUSIONS: Our findings highlight complex neuronal AChE-R/RACK1 interactions and are compatible with the hypothesis that peripheral site AChE inhibitors induce RACK1-mediated neuronal remodeling, promoting suppressed glutamatergic neurotransmission.


Subject(s)
Acetylcholinesterase/metabolism , Neurons/physiology , Receptors, Cell Surface/metabolism , Acetylcholinesterase/genetics , Alternative Splicing , Animals , Benzenaminium, 4,4'-(3-oxo-1,5-pentanediyl)bis(N,N-dimethyl-N-2-propenyl-), Dibromide/pharmacology , Cells, Cultured , Cholinesterase Inhibitors/pharmacology , Cricetinae , Cricetulus , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Gene Expression Regulation, Enzymologic/drug effects , Hippocampus/cytology , Models, Molecular , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques/methods , Physostigmine/pharmacology , RNA, Messenger/biosynthesis , Rats , Receptors for Activated C Kinase , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
12.
J Cell Physiol ; 211(1): 213-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17219410

ABSTRACT

Versican is a large chondroitin sulfate proteoglycan and belongs to the family of lecticans. Versican possesses two globular domains, G1 and G3 domain, separated by a CS-attachment region. The CS-attachment region present in the middle region is divided into two spliced domains named CSalpha and beta. Alternative splicing of versican generates at least four versican isoforms named V0, V1, V2, and V3. We have successfully cloned the full-length cDNA of chick versican isoforms V1 and V2 and found that versican isoform V1 induced mesenchymal-epithelial transition in NIH3T3 cells. Mesenchymal-epithelial transition induced by V1 in NIH3T3 cells is characterized by expression of E-cadherin and occludin, two epithelial markers, and reduced expression of fibroblastic marker vimentin (Sheng et al., 2006, Mol Biol Cell. 17, 2009-2020). In the present studies, we found that versican V1 isoform not only induced cell transition, but also increased intercellular communication via gap junction channels composed of connexin proteins. Our results showed that V1 induces plasma membrane localization of connexin 43, resulting in increased cell communication. This was further confirmed by blocking assays. Gap junctions mediated the transfer of small cytoplasmic molecules and the diffusion of second messenger molecules between adjacent cells. The ability of versican in regulating gap junction implied a potential role of versican in coordinating functions.


Subject(s)
Cell Communication , Gap Junctions/metabolism , Versicans/metabolism , Animals , Chickens , Connexins/metabolism , Epithelial Cells/cytology , Gene Silencing , Mice , NIH 3T3 Cells , Phosphorylation , Protein Isoforms/metabolism , RNA, Small Interfering/metabolism , Transfection
13.
J Biol Chem ; 281(28): 19358-68, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16648628

ABSTRACT

Versican is one of the major extracellular matrix (ECM) proteins in the brain. ECM molecules and their cleavage products critically regulate the growth and arborization of neurites, hence adjusting the formation of neural networks. Recent findings have revealed that peptide fragments containing the versican C terminus (G3 domain) are present in human brain astrocytoma. The present study demonstrated that a versican G3 domain enhanced cell attachment, neurite growth, and glutamate receptor-mediated currents in cultured embryonic hippocampal neurons. In addition, the G3 domain intensified dendritic spines, increased the clustering of both synaptophysin and the glutamate receptor subunit GluR2, and augmented excitatory synaptic activity. In contrast, a mutated G3 domain lacking the epidermal growth factor (EGF)-like repeats (G3deltaEGF) had little effect on neurite growth and glutamatergic function. Treating the neurons with the G3-conditioned medium rapidly increased the levels of phosphorylated EGF receptor (pEGFR) and phosphorylated extracellular signal-regulated kinase (pERK), indicating an activation of EGFR-mediated signaling pathways. Blockade of EGFR prevented the G3-induced ERK activation and suppressed the G3-provoked enhancement of neurite growth and glutamatergic function but failed to block the G3-mediated enhancement of cell attachment. These combined results indicate that the versican G3 domain regulates neuronal attachment, neurite outgrowth, and synaptic function of hippocampal neurons via EGFR-dependent and -independent signaling pathway(s). Our findings suggest a role for ECM proteolytic products in neural development and regeneration.


Subject(s)
Brain Neoplasms/metabolism , Chondroitin Sulfate Proteoglycans/physiology , ErbB Receptors/metabolism , Hippocampus/metabolism , Lectins, C-Type/physiology , Neurons/metabolism , Synaptic Transmission , Animals , Astrocytoma/metabolism , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/chemistry , Extracellular Matrix/metabolism , Humans , Lectins, C-Type/chemistry , Protein Structure, Tertiary , Rats , Versicans
14.
J Neurosci ; 24(41): 8950-60, 2004 Oct 13.
Article in English | MEDLINE | ID: mdl-15483114

ABSTRACT

Acetylcholinesterase (AChE) exerts noncatalytic activities on neural cell differentiation, adhesion, and neuritogenesis independently of its catalytic function. The noncatalytic functions of AChE have been attributed to its peripheral anionic site (PAS)-mediated protein-protein interactions. Structurally, AChE is highly homologous to the extracellular domain of neuroligin, a postsynaptic transmembrane molecule that interacts with presynaptic beta-neurexins, thus facilitating synaptic formation and maturation. Potential effects of AChE expression on synaptic transmission, however, remain unknown. Using electrophysiology, immunocytochemistry, and molecular biological approaches, this study investigated the role of AChE in the regulation of synaptic formation and functions. We found that AChE was highly expressed in cultured embryonic hippocampal neurons at early culture days, particularly in dendritic compartments including the growth cone. Subsequently, the expression level of AChE declined, whereas synaptic activity and synaptic proteins progressively increased. Chronic blockade of the PAS of AChE with specific inhibitors selectively impaired glutamatergic functions and excitatory synaptic structures independently of cholinergic activation, while inducing AChE overexpression. Moreover, the PAS blockade-induced glutamatergic impairments were associated with a depressed expression of beta-neurexins and an accumulation of other synaptic proteins, including neuroligins, and were mostly preventable by antisense suppression of AChE expression. Our findings demonstrate that interference with the nonenzymatic features of AChE alters AChE expression, which impairs excitatory synaptic structure and functions.


Subject(s)
Acetylcholinesterase/biosynthesis , Glutamic Acid/metabolism , Hippocampus/physiology , Neurons/metabolism , Synapses/physiology , Acetylcholinesterase/drug effects , Acetylcholinesterase/genetics , Animals , Binding Sites/drug effects , Cells, Cultured , Cholinesterase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Growth Cones/enzymology , Growth Cones/physiology , Hippocampus/cytology , Hippocampus/embryology , In Situ Hybridization , Ligands , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Synapses/drug effects , Synapses/enzymology , Time Factors
15.
Mol Biol Cell ; 15(5): 2093-104, 2004 May.
Article in English | MEDLINE | ID: mdl-14978219

ABSTRACT

The chondroitin sulfate proteoglycan versican is one of the major extracellular components in the developing and adult brain. Here, we show that isoforms of versican play different roles in neuronal differentiation and neurite outgrowth. Expression of versican V1 isoform in PC12 cells induced complete differentiation, whereas expression of V2 induced an aborted differentiation accompanied by apoptosis. V1 promoted neurite outgrowth of hippocampal neurons, but V2 failed to do so. V1 transfection enhanced expression of epidermal growth factor receptor and integrins, and facilitated sustained extracellular signal-regulated kinase/MAPK phosphorylation. Blockade of the epidermal growth factor receptor, beta1 integrin, or Src significantly inhibited neuronal differentiation. Finally, we demonstrated that versican V1 isoform also promoted differentiation of neural stem cells into neurons. Our results have implications for understanding how versican regulates neuronal development, function, and repair.


Subject(s)
Carrier Proteins/physiology , Nerve Tissue Proteins/physiology , Neurites/ultrastructure , Neurons/cytology , Animals , Benzoquinones , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Differentiation , Cyclins/genetics , Cyclins/metabolism , ErbB Receptors , Gene Expression , Genetic Vectors , Glycoproteins/metabolism , Hippocampus/cytology , Integrin beta1/pharmacology , Integrins/metabolism , Lactams, Macrocyclic , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Growth Factors/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurites/metabolism , Neurons/metabolism , PC12 Cells , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/physiology , Quinones/pharmacology , Rats , Rifabutin/analogs & derivatives , Transfection , Versicans
16.
Nat Genet ; 31(2): 184-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11992121

ABSTRACT

Although many genes that predispose for epilepsy in humans have been determined, those that underlie the classical syndromes of idiopathic generalized epilepsy (IGE) have yet to be identified. We report that an Ala322Asp mutation in GABRA1, encoding the alpha1 subunit of the gamma-aminobutyric acid receptor subtype A (GABA(A)), is found in affected individuals of a large French Canadian family with juvenile myoclonic epilepsy. Compared with wildtype receptors, GABA(A) receptors that contain the mutant subunit show a lesser amplitude of GABA-activated currents in vitro, indicating that seizures may result from loss of function of this inhibitory ligand-gated channel. Our results confirm that mutation of GABRA1 predisposes towards a common idiopathic generalized epilepsy syndrome in humans.


Subject(s)
Mutation, Missense , Myoclonic Epilepsy, Juvenile/genetics , Receptors, GABA-A/genetics , Adolescent , Adult , Amino Acid Sequence , Amino Acid Substitution/genetics , Child , Child, Preschool , Epilepsy, Generalized/etiology , Epilepsy, Generalized/genetics , Genes, Dominant , Genetic Predisposition to Disease , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Myoclonic Epilepsy, Juvenile/etiology , Pedigree
17.
Auton Neurosci ; 95(1-2): 24-31, 2002 Jan 10.
Article in English | MEDLINE | ID: mdl-11871782

ABSTRACT

Distention of the rat distal esophagus evokes an arterial pressor and a cardioaccelerator response that depends upon activation of a vagal afferent projection to the nucleus tractus solitarii (NTS). The present study aimed to determine in urethane-anesthetized rats if the afferent limb of this reflex (a) relays in the NTS subdivision known ro receive esophageal afferents, and (b) utilizes glutamatergic synapses. To this end, tetrodotoxin or the glutamate antagonists gamma-D-glutamyl-glycine, 6-7-dinitroquinoxaline-2,3-dione (DNQX) and 2-amino-5-phosphonovaleric acid (AP-5) were applied to the NTS extraventricular surface rostral to the obex. All four agents inhibited both components of the reflex. DNQX or AP-5 produced a similar reversible inhibition upon pressure ejection in the vagal esophageal afferent projection area. Application of tetrodotoxin to the dorsomedial medulla oblongata caudal to the area postrema (AP) was ineffective. Basal heart rate (HR) (except in the case of AP-5) and blood pressure increased upon NTS surface application of the antagonists but not after intra-NTS ejection. At corresponding dorsal NTS sites, focal excitation of solitarial neurons with glutamate evoked hypotension and cardiac slowing. At adjacent ventral sites in the NTS subnucleus centralis (NTSc) and/or its immediate vicinity, glutamate elicited an arterial pressor response that coincided with an esophageal contraction in most but not all cases. In conclusion, afferent fibers of the esophago-cardiovascular reflex (ECVR) probably (1) terminate in the vicinity of esophageal premotor neurons comprising the NTSc and (2) activate second-order neurons via glutamate receptors of both the NMDA and non-NMDA subtype.


Subject(s)
Cardiovascular Physiological Phenomena/drug effects , Esophagus/innervation , Glutamic Acid/metabolism , Solitary Nucleus/metabolism , Synaptic Transmission/physiology , Vagus Nerve/metabolism , Visceral Afferents/metabolism , Anesthetics, Local/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Esophagus/physiology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/pharmacology , Heart Rate/drug effects , Heart Rate/physiology , Male , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Reflex/drug effects , Reflex/physiology , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Synaptic Transmission/drug effects , Tetrodotoxin/pharmacology , Vagus Nerve/cytology , Vagus Nerve/drug effects , Visceral Afferents/cytology , Visceral Afferents/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL