Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Neurooncol Adv ; 3(1): vdaa165, 2021.
Article in English | MEDLINE | ID: mdl-33506204

ABSTRACT

BACKGROUND: Glioma is sensitive to microtubule-targeting agents (MTAs), but most MTAs do not cross the blood brain barrier (BBB). To address this limitation, we developed the new chemical entity, ST-401, a brain-penetrant MTA. METHODS: Synthesis of ST-401. Measures of MT assembly and dynamics. Cell proliferation and viability of patient-derived (PD) glioma in culture. Measure of tumor microtube (TM) parameters using immunofluorescence analysis and machine learning-based workflow. Pharmacokinetics (PK) and experimental toxicity in mice. In vivo antitumor activity in the RCAS/tv-a PDGFB-driven glioma (PDGFB-glioma) mouse model. RESULTS: We discovered that ST-401 disrupts microtubule (MT) function through gentle and reverisible reduction in MT assembly that triggers mitotic delay and cell death in interphase. ST-401 inhibits the formation of TMs, MT-rich structures that connect glioma to a network that promotes resistance to DNA damage. PK analysis of ST-401 in mice shows brain penetration reaching antitumor concentrations, and in vivo testing of ST-401 in a xenograft flank tumor mouse model demonstrates significant antitumor activity and no over toxicity in mice. In the PDGFB-glioma mouse model, ST-401 enhances the therapeutic efficacies of temozolomide (TMZ) and radiation therapy (RT). CONCLUSION: Our study identifies hallmarks of glioma tumorigenesis that are sensitive to MTAs and reports ST-401 as a promising chemical scaffold to develop brain-penetrant MTAs.

2.
J Biol Chem ; 293(5): 1551-1567, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29196604

ABSTRACT

Both long-term potentiation (LTP) and depression (LTD) of excitatory synapse strength require the Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) and its autonomous activity generated by Thr-286 autophosphorylation. Additionally, LTP and LTD are correlated with dendritic spine enlargement and shrinkage that are accompanied by the synaptic accumulation or removal, respectively, of the AMPA-receptor regulatory scaffold protein A-kinase anchoring protein (AKAP) 79/150. We show here that the spine shrinkage associated with LTD indeed requires synaptic AKAP79/150 removal, which in turn requires CaMKII activity. In contrast to normal CaMKII substrates, the substrate sites within the AKAP79/150 N-terminal polybasic membrane-cytoskeletal targeting domain were phosphorylated more efficiently by autonomous compared with Ca2+/CaM-stimulated CaMKII activity. This unusual regulation was mediated by Ca2+/CaM binding to the substrate sites resulting in protection from phosphorylation in the presence of Ca2+/CaM, a mechanism that favors phosphorylation by prolonged, weak LTD stimuli versus brief, strong LTP stimuli. Phosphorylation by CaMKII inhibited AKAP79/150 association with F-actin; it also facilitated AKAP79/150 removal from spines but was not required for it. By contrast, LTD-induced spine removal of AKAP79/150 required its depalmitoylation on two Cys residues within the N-terminal targeting domain. Notably, such LTD-induced depalmitoylation was also blocked by CaMKII inhibition. These results provide a mechanism how CaMKII can indeed mediate not only LTP but also LTD through regulated substrate selection; however, in the case of AKAP79/150, indirect CaMKII effects on palmitoylation are more important than the effects of direct phosphorylation. Additionally, our results provide the first direct evidence for a function of the well-described AKAP79/150 trafficking in regulating LTD-induced spine shrinkage.


Subject(s)
A Kinase Anchor Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Lipoylation , Long-Term Potentiation , Long-Term Synaptic Depression , Protein Processing, Post-Translational , Spine/metabolism , Synapses/metabolism , Animals , Humans , Spine/pathology , Synapses/pathology
3.
Mol Cancer Ther ; 15(9): 2018-29, 2016 09.
Article in English | MEDLINE | ID: mdl-27325686

ABSTRACT

Glioblastoma multiforme is a devastating and intractable type of cancer. Current antineoplastic drugs do not improve the median survival of patients diagnosed with glioblastoma multiforme beyond 14 to 15 months, in part because the blood-brain barrier is generally impermeable to many therapeutic agents. Drugs that target microtubules (MT) have shown remarkable efficacy in a variety of cancers, yet their use as glioblastoma multiforme treatments has also been hindered by the scarcity of brain-penetrant MT-targeting compounds. We have discovered a new alkylindole compound, ST-11, that acts directly on MTs and rapidly attenuates their rate of assembly. Accordingly, ST-11 arrests glioblastoma multiforme cells in prometaphase and triggers apoptosis. In vivo analyses reveal that unlike current antitubulin agents, ST-11 readily crosses the blood-brain barrier. Further investigation in a syngeneic orthotopic mouse model of glioblastoma multiforme shows that ST-11 activates caspase-3 in tumors to reduce tumor volume without overt toxicity. Thus, ST-11 represents the first member of a new class of brain-penetrant antitubulin therapeutic agents. Mol Cancer Ther; 15(9); 2018-29. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Microtubules/metabolism , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Nanoparticles , Pilot Projects , Solubility , Tubulin Modulators/administration & dosage , Tubulin Modulators/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Neuron ; 83(2): 361-371, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-25033180

ABSTRACT

The serine hydrolase α/ß-hydrolase domain 6 (ABHD6) hydrolyzes the most abundant endocannabinoid (eCB) in the brain, 2-arachidonoylglycerol (2-AG), and controls its availability at cannabinoid receptors. We show that ABHD6 inhibition decreases pentylenetetrazole (PTZ)-induced generalized tonic-clonic and myoclonic seizure incidence and severity. This effect is retained in Cnr1(-/-) or Cnr2(-/-) mice, but blocked by addition of a subconvulsive dose of picrotoxin, suggesting the involvement of GABAA receptors. ABHD6 inhibition also blocked spontaneous seizures in R6/2 mice, a genetic model of juvenile Huntington's disease known to exhibit dysregulated eCB signaling. ABHD6 blockade retained its antiepileptic activity over chronic dosing and was not associated with psychomotor or cognitive effects. While the etiology of seizures in R6/2 mice remains unsolved, involvement of the hippocampus is suggested by interictal epileptic discharges, increased expression of vGLUT1 but not vGAT, and reduced Neuropeptide Y (NPY) expression. We conclude that ABHD6 inhibition may represent a novel antiepileptic strategy.


Subject(s)
Anticonvulsants/therapeutic use , Brain/drug effects , Carbamates/therapeutic use , Monoacylglycerol Lipases/antagonists & inhibitors , Seizures/drug therapy , Animals , Anticonvulsants/pharmacology , Behavior, Animal/drug effects , Brain/physiopathology , Carbamates/pharmacology , Male , Mice , Mice, Knockout , Pentylenetetrazole , Receptors, Cannabinoid/genetics , Seizures/chemically induced , Seizures/physiopathology
5.
Eur J Neurosci ; 37(3): 429-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23167744

ABSTRACT

Cannabinoid receptor 1 (CB(1) receptor) controls several neuronal functions, including neurotransmitter release, synaptic plasticity, gene expression and neuronal viability. Downregulation of CB(1) expression in the basal ganglia of patients with Huntington's disease (HD) and animal models represents one of the earliest molecular events induced by mutant huntingtin (mHtt). This early disruption of neuronal CB(1) signaling is thought to contribute to HD symptoms and neurodegeneration. Here we determined whether CB(1) downregulation measured in patients with HD and mouse models was ubiquitous or restricted to specific striatal neuronal subpopulations. Using unbiased semi-quantitative immunohistochemistry, we confirmed previous studies showing that CB(1) expression is downregulated in medium spiny neurons of the indirect pathway, and found that CB(1) is also downregulated in neuropeptide Y (NPY)/neuronal nitric oxide synthase (nNOS)-expressing interneurons while remaining unchanged in parvalbumin- and calretinin-expressing interneurons. CB(1) downregulation in striatal NPY/nNOS-expressing interneurons occurs in R6/2 mice, Hdh(Q150/Q150) mice and the caudate nucleus of patients with HD. In R6/2 mice, CB(1) downregulation in NPY/nNOS-expressing interneurons correlates with diffuse expression of mHtt in the soma. This downregulation also occludes the ability of cannabinoid agonists to activate the pro-survival signaling molecule cAMP response element-binding protein in NPY/nNOS-expressing interneurons. Loss of CB(1) signaling in NPY/nNOS-expressing interneurons could contribute to the impairment of basal ganglia functions linked to HD.


Subject(s)
Basal Ganglia/metabolism , Down-Regulation , Huntington Disease/metabolism , Interneurons/metabolism , Neuropeptide Y/metabolism , Receptor, Cannabinoid, CB1/metabolism , Adult , Aged , Animals , Basal Ganglia/cytology , Calbindin 2 , Cannabinoid Receptor Agonists/pharmacology , Case-Control Studies , Cyclic AMP/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Huntingtin Protein , Interneurons/classification , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nuclear Proteins/genetics , Parvalbumins/genetics , Parvalbumins/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , S100 Calcium Binding Protein G/genetics , S100 Calcium Binding Protein G/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism
6.
J Biol Chem ; 286(33): 28723-28728, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21665953

ABSTRACT

Agonists at cannabinoid receptors, such as the phytocannabinoid Δ(9)-tetrahydrocannabinol, exert a remarkable array of therapeutic effects but are also associated with undesirable psychoactive side effects. Conversely, targeting enzymes that hydrolyze endocannabinoids (eCBs) allows for more precise fine-tuning of cannabinoid receptor signaling, thus providing therapeutic relief with reduced side effects. Here, we report the development and characterization of an inhibitor of eCB hydrolysis, UCM710, which augments both N-arachidonoylethanolamine and 2-arachidonoylglycerol levels in neurons. This compound displays a unique pharmacological profile in that it inhibits fatty acid amide hydrolase and α/ß-hydrolase domain 6 but not monoacylglycerol lipase. Thus, UCM710 represents a novel tool to delineate the therapeutic potential of compounds that manipulate a subset of enzymes that control eCB signaling.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Arachidonic Acids/metabolism , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Enzyme Inhibitors/pharmacology , Glycerides/metabolism , Monoacylglycerol Lipases/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/metabolism , Amidohydrolases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Mice , Monoacylglycerol Lipases/metabolism , Nerve Tissue Proteins/metabolism , Polyunsaturated Alkamides , Receptors, Cannabinoid
7.
Chem Biol ; 18(5): 563-8, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21609837

ABSTRACT

High-throughput screening (HTS) of chemical libraries is often used for the unbiased identification of compounds interacting with G protein-coupled receptors (GPCRs), the largest family of therapeutic targets. However, current HTS methods require removing GPCRs from their native environment, which modifies their pharmacodynamic properties and biases the screen toward false positive hits. Here, we developed and validated a molecular imaging (MI) agent, NIR-mbc94, which emits near infrared (NIR) light and selectively binds to endogenously expressed cannabinoid CB(2) receptors, a recognized target for treating autoimmune diseases, chronic pain and cancer. The precision and ease of this assay allows for the HTS of compounds interacting with CB(2) receptors expressed in their native environment.


Subject(s)
Fluorescent Dyes/chemistry , Norbornanes/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Inverse Agonism , High-Throughput Screening Assays , Mice , Norbornanes/chemistry , Protein Binding , Pyrazoles/chemistry , Receptor, Cannabinoid, CB2/metabolism
8.
Nat Neurosci ; 13(8): 951-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20657592

ABSTRACT

The endocannabinoid 2-arachidonoylglycerol (2-AG) regulates neurotransmission and neuroinflammation by activating CB1 cannabinoid receptors on neurons and CB2 cannabinoid receptors on microglia. Enzymes that hydrolyze 2-AG, such as monoacylglycerol lipase, regulate the accumulation and efficacy of 2-AG at cannabinoid receptors. We found that the recently described serine hydrolase alpha-beta-hydrolase domain 6 (ABHD6) also controls the accumulation and efficacy of 2-AG at cannabinoid receptors. In cells from the BV-2 microglia cell line, ABHD6 knockdown reduced hydrolysis of 2-AG and increased the efficacy with which 2-AG can stimulate CB2-mediated cell migration. ABHD6 was expressed by neurons in primary culture and its inhibition led to activity-dependent accumulation of 2-AG. In adult mouse cortex, ABHD6 was located postsynaptically and its selective inhibition allowed the induction of CB1-dependent long-term depression by otherwise subthreshold stimulation. Our results indicate that ABHD6 is a rate-limiting step of 2-AG signaling and is therefore a bona fide member of the endocannabinoid signaling system.


Subject(s)
Arachidonic Acids/metabolism , Brain/metabolism , Glycerides/metabolism , Monoacylglycerol Lipases/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction/physiology , Animals , COS Cells , Cell Line , Cell Movement , Chlorocebus aethiops , Endocannabinoids , Excitatory Postsynaptic Potentials/physiology , Gene Knockdown Techniques , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microscopy, Electron, Transmission , Neurons/metabolism , Patch-Clamp Techniques , Polymerase Chain Reaction , RNA, Messenger/analysis , Transfection
9.
J Neurosci ; 30(6): 2160-4, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20147543

ABSTRACT

Adenosine A(2A) receptor antagonists are psychomotor stimulants that also hold therapeutic promise for movement disorders. However, the molecular mechanisms underlying their stimulant properties are not well understood. Here, we show that the robust increase in locomotor activity induced by an A(2A) antagonist in vivo is greatly attenuated by antagonizing cannabinoid CB(1) receptor signaling or by administration to CB(1)(-/-) mice. To determine the locus of increased endocannabinoid signaling, we measured the amount of anandamide [AEA (N-arachidonoylethanolamine)] and 2-arachidonoylglycerol (2-AG) in brain tissue from striatum and cortex. We find that 2-AG is selectively increased in striatum after acute blockade of A(2A) receptors, which are highly expressed by striatal indirect-pathway medium spiny neurons (MSNs). Using targeted whole-cell recordings from direct- and indirect-pathway MSNs, we demonstrate that A(2A) receptor antagonists potentiate 2-AG release and induction of long-term depression at indirect-pathway MSNs, but not direct-pathway MSNs. Together, these data outline a molecular mechanism by which A(2A) antagonists reduce excitatory synaptic drive on the indirect pathway through CB(1) receptor signaling, thus leading to increased psychomotor activation.


Subject(s)
Adenosine A2 Receptor Antagonists , Cannabinoid Receptor Modulators/metabolism , Central Nervous System Stimulants/pharmacology , Endocannabinoids , Motor Activity/drug effects , Afferent Pathways/metabolism , Animals , Arachidonic Acids/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Glycerides/metabolism , Long-Term Synaptic Depression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/physiology , Signal Transduction
10.
Nat Methods ; 7(2): 126-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20037591

ABSTRACT

Neurotransmission operates on a millisecond timescale but is changed by normal experience or neuropathology over days to months. Despite the importance of long-term neurotransmitter dynamics, no technique exists to track these changes in a subject from day to day over extended periods of time. Here we describe and characterize a microsensor that can detect the neurotransmitter dopamine with subsecond temporal resolution over months in vivo in rats and mice.


Subject(s)
Behavior, Animal/physiology , Biosensing Techniques/instrumentation , Dopamine/analysis , Electrochemistry/instrumentation , Electrodes, Implanted , Mesencephalon/physiology , Monitoring, Ambulatory/instrumentation , Animals , Equipment Design , Equipment Failure Analysis , Mice , Microelectrodes , Miniaturization , Neurotransmitter Agents/analysis , Rats , Reproducibility of Results , Sensitivity and Specificity
11.
J Neurosci ; 27(13): 3523-34, 2007 Mar 28.
Article in English | MEDLINE | ID: mdl-17392468

ABSTRACT

NMDA receptor (NMDAR)-dependent hippocampal synaptic plasticity underlying learning and memory coordinately regulates dendritic spine structure and AMPA receptor (AMPAR) postsynaptic strength through poorly understood mechanisms. Induction of long-term depression (LTD) activates protein phosphatase 2B/calcineurin (CaN), leading to dendritic spine shrinkage through actin depolymerization and AMPAR depression through receptor dephosphorylation and internalization. The scaffold proteins A-kinase-anchoring protein 79/150 (AKAP79/150) and postsynaptic density 95 (PSD95) form a complex that controls the opposing actions of the cAMP-dependent protein kinase (PKA) and CaN in regulation of AMPAR phosphorylation. The AKAP79/150-PSD95 complex is disrupted in hippocampal neurons during LTD coincident with internalization of AMPARs, decreases in PSD95 levels, and loss of AKAP79/150 and PKA from spines. AKAP79/150 is targeted to spines through binding F-actin and the phospholipid phosphatidylinositol-(4,5)-bisphosphate (PIP2). Previous electrophysiological studies have demonstrated that inhibition of phospholipase C (PLC)-catalyzed hydrolysis of PIP2 inhibits NMDAR-dependent LTD; however, the signaling mechanisms that link PLC activation to alterations in dendritic spine structure and AMPAR function in LTD are unknown. We show here that NMDAR stimulation of PLC in cultured hippocampal neurons is necessary for AKAP79/150 loss from spines and depolymerization of spine actin. Importantly, we demonstrate that NMDAR activation of PLC is also necessary for decreases in spine PSD95 levels and AMPAR internalization. Thus, PLC signaling is required for structural and functional changes in spine actin, PSD scaffolding, and AMPAR trafficking underlying postsynaptic expression of LTD.


Subject(s)
Dendritic Spines/metabolism , Long-Term Synaptic Depression/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , Type C Phospholipases/metabolism , Animals , Biotransformation , Cells, Cultured , Hippocampus/physiology , Neurons/metabolism , Oligopeptides/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Rats , Signal Transduction/physiology , Synaptic Membranes
12.
Eur J Cell Biol ; 85(7): 627-33, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16504338

ABSTRACT

Central to organization of signaling pathways are scaffolding, anchoring and adaptor proteins that mediate localized assembly of multi-protein complexes containing receptors, second messenger-generating enzymes, kinases, phosphatases, and substrates. At the postsynaptic density (PSD) of excitatory synapses, AMPA (AMPAR) and NMDA (NMDAR) glutamate receptors are linked to signaling proteins, the actin cytoskeleton, and synaptic adhesion molecules on dendritic spines through a network of scaffolding proteins that may play important roles regulating synaptic structure and receptor functions in synaptic plasticity underlying learning and memory. AMPARs are rapidly recruited to dendritic spines through NMDAR activation during induction of long-term potentiation (LTP) through pathways that also increase the size and F-actin content of spines. Phosphorylation of AMPAR-GluR1 subunits by the cAMP-dependent protein kinase (PKA) helps stabilize AMPARs recruited during LTP. In contrast, induction of long-term depression (LTD) leads to rapid calcineurin-protein phosphatase 2B (CaN) mediated dephosphorylation of PKA-phosphorylated GluR1 receptors, endocytic removal of AMPAR from synapses, and a reduction in spine size. However, mechanisms for coordinately regulating AMPAR localization, phosphorylation, and synaptic structure by PKA and CaN are not well understood. A kinase-anchoring protein (AKAP) 79/150 is a PKA- and CaN-anchoring protein that is linked to NMDARs and AMPARs through PSD-95 and SAP97 membrane-associated guanylate kinase (MAGUK) scaffolds. Importantly, disruption of PKA-anchoring in neurons and functional analysis of GluR1-MAGUK-AKAP79 complexes in heterologous cells suggests that AKAP79/150-anchored PKA and CaN may regulate AMPARs in LTD. In the work presented at the "First International Meeting on Anchored cAMP Signaling Pathways" (Berlin-Buch, Germany, October 15-16, 2005), we demonstrate that AKAP79/150 is targeted to dendritic spines by an N-terminal basic region that binds phosphatidylinositol-4,5-bisphosphate (PIP(2)), F-actin, and actin-linked cadherin adhesion molecules. Thus, anchoring of PKA and CaN as well as physical linkage of the AKAP to both cadherin-cytoskeletal and MAGUK-receptor complexes could play roles in coordinating changes in synaptic structure and receptor signaling functions underlying plasticity. Importantly, we provide evidence showing that NMDAR-CaN signaling pathways implicated in AMPAR regulation during LTD lead to a disruption of AKAP79/150 interactions with actin, MAGUKs, and cadherins and lead to a loss of the AKAP and anchored PKA from postsynapses. Our studies thus far indicate that this AKAP79/150 translocation depends on activation of CaN, F-actin reorganization, and possibly Ca(2+)-CaM binding to the N-terminal basic regions. Importantly, this tranlocation of the AKAP79/150-PKA complex from spines may shift the balance of PKA kinase and CaN/PP1 phosphatase activity at the postsynapse in favor of the phosphatases. This loss of PKA could then promote actions of CaN and PP1 during induction of LTD including maintaining AMPAR dephosphorylation, promoting AMPAR endocytosis, and preventing AMPAR recycling. Overall, these findings challenge the accepted notion that AKAPs are static anchors that position signaling proteins near fixed target substrates and instead suggest that AKAPs can function in more dynamic manners to regulate local signaling events.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Neurons/metabolism , Signal Transduction , A Kinase Anchor Proteins , Actins/metabolism , Animals , Cell Adhesion Molecules/metabolism , Humans , Long-Term Potentiation , Long-Term Synaptic Depression , Models, Biological , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Neurotoxins/adverse effects , Nuclear Matrix-Associated Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Tissue Distribution
13.
Nat Cell Biol ; 5(12): 1104-10, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14634666

ABSTRACT

Sensing the osmolarity of the environment is a critical response for all organisms. Whereas bacteria will migrate away from high osmotic conditions, most eukaryotic cells are not motile and use adaptive metabolic responses for survival. The p38 MAPK pathway is a crucial mediator of survival during cellular stress. We have discovered a novel scaffold protein that binds to actin, the GTPase Rac, and the upstream kinases MEKK3 and MKK3 in the p38 MAPK phospho-relay module. RNA interference (RNAi) demonstrates that MEKK3 and the scaffold protein are required for p38 activation in response to sorbitol-induced hyperosmolarity. FRET identifies a cytoplasmic complex of the MEKK3 scaffold protein that is recruited to dynamic actin structures in response to sorbitol treatment. Through its ability to bind actin, relocalize to Rac-containing membrane ruffles and its obligate requirement for p38 activation in response to sorbitol, we have termed this protein osmosensing scaffold for MEKK3 (OSM). The Rac-OSM-MEKK3-MKK3 complex is the mammalian counterpart of the CDC42-STE50-STE11-Pbs2 complex in Saccharomyces cerevisiae that is required for the regulation of p38 activity.


Subject(s)
Cytoskeletal Proteins/isolation & purification , Cytoskeleton/metabolism , MAP Kinase Kinase Kinases/metabolism , Microfilament Proteins/isolation & purification , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Water-Electrolyte Balance/physiology , rac GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Binding Sites/physiology , COS Cells , Cell Surface Extensions/metabolism , Cytoskeletal Proteins/genetics , DNA, Complementary/analysis , DNA, Complementary/genetics , Humans , MAP Kinase Kinase 3 , MAP Kinase Kinase Kinase 3 , Macromolecular Substances , Microfilament Proteins/genetics , Molecular Sequence Data , Osmotic Pressure , Protein Binding/physiology , Sorbitol/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...