Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.190
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 137(4): 749-60, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450520

ABSTRACT

Temperature compensation of circadian clocks is an unsolved problem with relevance to the general phenomenon of biological compensation. We identify casein kinase 2 (CK2) as a key regulator of temperature compensation of the Neurospora clock by determining that two long-standing clock mutants, chrono and period-3, displaying distinctive alterations in compensation encode the beta1 and alpha subunits of CK2, respectively. Reducing the dose of these subunits, particularly beta1, significantly alters temperature compensation without altering the enzyme's Q(10). By contrast, other kinases and phosphatases implicated in clock function do not play appreciable roles in temperature compensation. CK2 exerts its effects on the clock by directly phosphorylating FREQUENCY (FRQ), and this phosphorylation is compromised in CK2 hypomorphs. Finally, mutation of certain putative CK2 phosphosites on FRQ, shown to be phosphorylated in vivo, predictably alters temperature compensation profiles effectively phenocopying CK2 mutants.


Subject(s)
Casein Kinase II/physiology , Circadian Rhythm , Neurospora crassa/enzymology , Neurospora crassa/physiology , Casein Kinase II/chemistry , Casein Kinase II/genetics , Gene Dosage , Mutation , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/physiology , Temperature
2.
Dev Biol ; 469: 144-159, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33131707

ABSTRACT

Bivalve metamorphosis is a developmental transition from a free-living larva to a benthic juvenile (spat), regulated by a complex interaction of neurotransmitters and neurohormones such as L-DOPA and epinephrine (catecholamine). We recently suggested an N-Methyl-D-aspartate (NMDA) receptor pathway as an additional and previously unknown regulator of bivalve metamorphosis. To explore this theory further, we successfully induced metamorphosis in the Pacific oyster, Crassostrea gigas, by exposing competent larvae to L-DOPA, epinephrine, MK-801 and ifenprodil. Subsequently, we cloned three NMDA receptor subunits CgNR1, CgNR2A and CgNR2B, with sequence analysis suggesting successful assembly of functional NMDA receptor complexes and binding to natural occurring agonists and the channel blocker MK-801. NMDA receptor subunits are expressed in competent larvae, during metamorphosis and in spat, but this expression is neither self-regulated nor regulated by catecholamines. In-situ hybridisation of CgNR1 in competent larvae identified NMDA receptor presence in the apical organ/cerebral ganglia area with a potential sensory function, and in the nervous network of the foot indicating an additional putative muscle regulatory function. Furthermore, phylogenetic analyses identified molluscan-specific gene expansions of key enzymes involved in catecholamine biosynthesis. However, exposure to MK-801 did not alter the expression of selected key enzymes, suggesting that NMDA receptors do not regulate the biosynthesis of catecholamines via gene expression.


Subject(s)
Catecholamines/biosynthesis , Crassostrea/growth & development , Metamorphosis, Biological , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cloning, Molecular , Crassostrea/enzymology , Crassostrea/genetics , Crassostrea/metabolism , Phylogeny , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Sequence Alignment , Sequence Analysis, Protein
3.
Mol Cell ; 56(6): 738-48, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25453762

ABSTRACT

Mammalian circadian rhythms are generated by a negative feedback loop in which PERIOD (PER) proteins accumulate, form a large nuclear complex (PER complex), and bind the transcription factor CLOCK-BMAL1, repressing their own expression. We found that mouse PER complexes include the Mi-2/nucleosome remodelling and deacetylase (NuRD) transcriptional corepressor. Unexpectedly, two NuRD subunits, CHD4 and MTA2, constitutively associate with CLOCK-BMAL1, with CHD4 functioning to promote CLOCK-BMAL1 transcriptional activity. At the onset of negative feedback, the PER complex delivers the remaining complementary NuRD subunits to DNA-bound CLOCK-BMAL1, thereby reconstituting a NuRD corepressor that is important for circadian transcriptional feedback and clock function. The PER complex thus acquires full repressor activity only upon successful targeting of CLOCK-BMAL1. Our results show how specificity is generated in the clock despite its dependence on generic transcriptional regulators and reveal the existence of active communication between the positive and negative limbs of the circadian feedback loop.


Subject(s)
Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Animals , Circadian Clocks , Feedback, Physiological , Liver/metabolism , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Protein Subunits/physiology
4.
Mol Cell ; 53(3): 498-505, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24507717

ABSTRACT

Histone variant H2A.Z-containing nucleosomes exist at most eukaryotic promoters and play important roles in gene transcription and genome stability. The multisubunit nucleosome-remodeling enzyme complex SWR1, conserved from yeast to mammals, catalyzes the ATP-dependent replacement of histone H2A in canonical nucleosomes with H2A.Z. How SWR1 catalyzes the replacement reaction is largely unknown. Here, we determined the crystal structure of the N-terminal region (599-627) of the catalytic subunit Swr1, termed Swr1-Z domain, in complex with the H2A.Z-H2B dimer at 1.78 Å resolution. The Swr1-Z domain forms a 310 helix and an irregular chain. A conserved LxxLF motif in the Swr1-Z 310 helix specifically recognizes the αC helix of H2A.Z. Our results show that the Swr1-Z domain can deliver the H2A.Z-H2B dimer to the DNA-(H3-H4)2 tetrasome to form the nucleosome by a histone chaperone mechanism.


Subject(s)
Adenosine Triphosphatases/chemistry , Histones/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Adenosine Triphosphatases/physiology , Amino Acid Sequence , Chromatin Assembly and Disassembly/genetics , Cloning, Molecular , Crystallography, X-Ray , Dimerization , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/physiology , X-Ray Diffraction
5.
Proc Natl Acad Sci U S A ; 116(1): 158-167, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30584095

ABSTRACT

p97 is an essential hexameric AAA+ ATPase involved in a wide range of cellular processes. Mutations in the enzyme are implicated in the etiology of an autosomal dominant neurological disease in which patients are heterozygous with respect to p97 alleles, containing one copy each of WT and disease-causing mutant genes, so that, in vivo, p97 molecules can be heterogeneous in subunit composition. Studies of p97 have, however, focused on homohexameric constructs, where protomers are either entirely WT or contain a disease-causing mutation, showing that for WT p97, the N-terminal domain (NTD) of each subunit can exist in either a down (ADP) or up (ATP) conformation. NMR studies establish that, in the ADP-bound state, the up/down NTD equilibrium shifts progressively toward the up conformation as a function of disease mutant severity. To understand NTD functional dynamics in biologically relevant p97 heterohexamers comprising both WT and disease-causing mutant subunits, we performed a methyl-transverse relaxation optimized spectroscopy (TROSY) NMR study on a series of constructs in which only one of the protomer types is NMR-labeled. Our results show positive cooperativity of NTD up/down equilibria between neighboring protomers, allowing us to define interprotomer pathways that mediate the allosteric communication between subunits. Notably, the perturbed up/down NTD equilibrium in mutant subunits is partially restored by neighboring WT protomers, as is the two-pronged binding of the UBXD1 adaptor that is affected in disease. This work highlights the plasticity of p97 and how subtle perturbations to its free-energy landscape lead to significant changes in NTD conformation and adaptor binding.


Subject(s)
Valosin Containing Protein/physiology , Humans , Magnetic Resonance Spectroscopy , Mutation , Protein Domains , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/metabolism , Protein Subunits/physiology , Valosin Containing Protein/chemistry , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
6.
Nat Rev Neurosci ; 17(10): 623-35, 2016 10.
Article in English | MEDLINE | ID: mdl-27558536

ABSTRACT

GluN3-containing NMDA receptors (GluN3-NMDARs) are rarer than the 'classical' NMDARs, which are composed solely of GluN1 and GluN2 subunits, and have non-conventional biophysical, trafficking and signalling properties. In the CNS, they seem to have important roles in delaying synapse maturation until the arrival of sensory experience and in targeting non-used synapses for pruning. The reactivation of GluN3A expression at inappropriate ages may underlie maladaptive synaptic rearrangements observed in addiction, neurodegenerative diseases and other major brain disorders. Here, we discuss current evidence for these and other emerging roles for GluN3-NMDARs in the physiology and pathology of the CNS.


Subject(s)
Brain Diseases/physiopathology , Central Nervous System/physiology , Protein Subunits/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain Diseases/metabolism , Central Nervous System/cytology , Central Nervous System/pathology , Humans , Models, Neurological , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction , Synapses/metabolism , Synapses/physiology
7.
Toxicol Appl Pharmacol ; 426: 115643, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34265354

ABSTRACT

The chemical threat agent tetramethylenedisulfotetramine (TETS) is a γ-aminobutyric acid type A receptor (GABA AR) antagonist that causes life threatening seizures. Currently, there is no specific antidote for TETS intoxication. TETS-induced seizures are typically treated with benzodiazepines, which function as nonselective positive allosteric modulators (PAMs) of synaptic GABAARs. The major target of TETS was recently identified as the GABAAR α2ß3γ2 subtype in electrophysiological studies using recombinantly expressed receptor combinations. Here, we tested whether these in vitro findings translate in vivo by comparing the efficacy of GABAAR subunit-selective PAMs in reducing TETS-induced seizure behavior in larval zebrafish. We tested PAMs targeting α1, α2, α2/3/5, α6, ß2/3, ß1/2/3, and δ subunits and compared their efficacy to the benzodiazepine midazolam (MDZ). The data demonstrate that α2- and α6-selective PAMs (SL-651,498 and SB-205384, respectively) were effective at mitigating TETS-induced seizure-like behavior. Combinations of SB-205384 and MDZ or SL-651,498 and 2-261 (ß2/3-selective) mitigated TETS-induced seizure-like behavior at concentrations that did not elicit sedating effects in a photomotor behavioral assay, whereas MDZ alone caused sedation at the concentration required to stop seizure behavior. Isobologram analyses suggested that SB-205384 and MDZ interacted in an antagonistic fashion, while the effects of SL-651,498 and 2-261 were additive. These results further elucidate the molecular mechanism by which TETS induces seizures and provide mechanistic insight regarding specific countermeasures against this chemical convulsant.


Subject(s)
Bridged-Ring Compounds , Convulsants , GABA Modulators/pharmacology , Hypnotics and Sedatives/pharmacology , Protein Subunits/physiology , Receptors, GABA-A/physiology , Seizures/chemically induced , Animals , Behavior, Animal/drug effects , Larva , Locomotion/drug effects , Midazolam/pharmacology , Protein Subunits/genetics , Receptors, GABA-A/genetics , Seizures/physiopathology , Zebrafish
8.
Mol Cell ; 50(2): 200-11, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23499005

ABSTRACT

Glutamine is an essential nutrient for cancer cell survival and proliferation, yet the signaling pathways that sense glutamine levels remain uncharacterized. Here, we report that the protein phosphatase 2A (PP2A)-associated protein, α4, plays a conserved role in glutamine sensing. α4 promotes assembly of an adaptive PP2A complex containing the B55α regulatory subunit via providing the catalytic subunit upon glutamine deprivation. Moreover, B55α is specifically induced upon glutamine deprivation in a ROS-dependent manner to activate p53 and promote cell survival. B55α activates p53 through direct interaction and dephosphorylation of EDD, a negative regulator of p53. Importantly, the B55α-EDD-p53 pathway is essential for cancer cell survival and tumor growth under low glutamine conditions in vitro and in vivo. This study delineates a previously unidentified signaling pathway that senses glutamine levels as well as provides important evidence that protein phosphatase complexes are actively involved in signal transduction.


Subject(s)
Glutamine/deficiency , Protein Phosphatase 2/metabolism , Protein Subunits/metabolism , Stress, Physiological , Tumor Suppressor Protein p53/metabolism , Adaptation, Physiological , Adaptor Proteins, Signal Transducing , Animals , Catalytic Domain , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Expression , Gene Knockdown Techniques , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Nude , Molecular Chaperones , NIH 3T3 Cells , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Multimerization , Protein Phosphatase 2/genetics , Protein Phosphatase 2/physiology , Protein Subunits/genetics , Protein Subunits/physiology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Transcriptional Activation , Tumor Burden , Ubiquitin-Protein Ligases/metabolism
9.
Proc Natl Acad Sci U S A ; 115(33): 8346-8351, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061400

ABSTRACT

In this paper, we report an example of the engineered expression of tetrameric ß-galactosidase (ß-gal) containing varying numbers of active monomers. Specifically, by combining wild-type and single-nucleotide polymorphism plasmids at varying ratios, tetrameric ß-gal was expressed in vitro with one to four active monomers. The kinetics of individual enzyme molecules revealed four distinct populations, corresponding to the number of active monomers in the enzyme. Using single-molecule-level enzyme kinetics, we were able to measure an accurate in vitro mistranslation frequency (5.8 × 10-4 per base). In addition, we studied the kinetics of the mistranslated ß-gal at the single-molecule level.


Subject(s)
Protein Multimerization , Protein Subunits/physiology , beta-Galactosidase/physiology , Kinetics , Plasmids , Polymorphism, Single Nucleotide , Protein Biosynthesis , beta-Galactosidase/biosynthesis , beta-Galactosidase/chemistry , beta-Galactosidase/genetics
10.
Biochem Biophys Res Commun ; 533(3): 613-619, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32981679

ABSTRACT

c-Myc modulator 1 (MM1), also known as PFDN5, is the fifth subunit of prefoldin. It was previously reported that MM1-based prefoldin promotes folding of actin during assembly of cytoskeleton, which plays key roles in cell migration. However, no evidence supports that MM1 affects cell migration. In the present study, we found that MM1 promotes cell migration in multiple cell lines. Further study revealed that MM1 promotes polymerization of ß-actin into filamentous form and increases both density and length of filopodia. Effects of MM1 on filopodia formation and cell migration depend on its prefoldin activity. Though c-Myc is repressed by MM1, simultaneous knock-down of c-Myc fails to rescue migration inhibition induced by MM1 ablation. Taken together, we here, for the first time, report that prefoldin subunit MM1 is involved in cell migration; this involvement of MM1 in cell migration is due to its prefoldin activity to boost polymerization of ß-actin during filopodia formation. Our findings may be helpful to elucidate the mechanism of cell migration and cancer metastasis.


Subject(s)
Cell Movement , Molecular Chaperones/physiology , Pseudopodia/metabolism , Actins/metabolism , Cell Line , Humans , Molecular Chaperones/metabolism , Protein Subunits/metabolism , Protein Subunits/physiology
11.
BMC Plant Biol ; 20(1): 380, 2020 Aug 18.
Article in English | MEDLINE | ID: mdl-32811442

ABSTRACT

BACKGROUND: Glycosylphosphatidylinositol (GPI) addition is one of the several post-translational modifications to proteins that increase their affinity for membranes. In eukaryotes, the GPI transamidase complex (GPI-T) catalyzes the attachment of pre-assembled GPI anchors to GPI-anchored proteins (GAPs) through a transamidation reaction. A mutation in AtGPI8 (gpi8-2), the putative catalytic subunit of GPI-T in Arabidopsis, is transmitted normally through the female gametophyte (FG), indicating the FG tolerates loss of GPI transamidation. In contrast, gpi8-2 almost completely abolishes male gametophyte (MG) function. Still, the unexpected finding that gpi8-2 FGs function normally requires further investigation. Additionally, specific developmental defects in the MG caused by loss of GPI transamidation remain poorly characterized. RESULTS: Here we investigated the effect of loss of AtPIG-S, another GPI-T subunit, in both gametophytes. Like gpi8-2, we showed that a mutation in AtPIG-S (pigs-1) disrupted synergid localization of LORELEI (LRE), a putative GAP critical for pollen tube reception by the FG. Still, pigs-1 is transmitted normally through the FG. Conversely, pigs-1 severely impaired male gametophyte (MG) function during pollen tube emergence and growth in the pistil. A pPIGS:GFP-PIGS transgene complemented these MG defects and enabled generation of pigs-1/pigs-1 seedlings. However, the pPIGS:GFP-PIGS transgene seemingly failed to rescue the function of AtPIG-S in the sporophyte, as pigs-1/pigs-1, pPIGS:GFP-PIGS seedlings died soon after germination. CONCLUSIONS: Characterization of pigs-1 provided further evidence that the FG tolerates loss of GPI transamidation more than the MG and that the MG compared to the FG may be a better haploid system to study the role of GPI-anchoring. Pigs-1 pollen develops normally and thus represent a tool in which GPI anchor biosynthesis and transamidation of GAPs have been uncoupled, offering a potential way to study free GPI in plant development. While previously reported male fertility defects of GPI biosynthesis mutants could have been due either to loss of GPI or GAPs lacking the GPI anchor, our results clarified that the loss of mature GAPs underlie male fertility defects of GPI-deficient pollen grains, as pigs-1 is defective only in the downstream transamidation step.


Subject(s)
Acyltransferases/physiology , Arabidopsis/enzymology , Arabidopsis/growth & development , Pollen Tube/growth & development , Acyltransferases/genetics , Arabidopsis Proteins/metabolism , Cloning, Molecular , Genotyping Techniques , Membrane Glycoproteins/metabolism , Mutation , Pollen/genetics , Protein Subunits/genetics , Protein Subunits/physiology , Real-Time Polymerase Chain Reaction , Nicotiana/genetics
12.
Nat Immunol ; 9(1): 89-96, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18059270

ABSTRACT

CRACM1 (also called Orai1) constitutes the pore subunit of store-operated calcium release-activated calcium channels. A point mutation in the gene encoding CRACM1 is associated with severe combined immunodeficiency disease in humans. Here we generated CRACM1-deficient mice in which beta-galactosidase activity 'reported' CRACM1 expression. CRACM1-deficient mice were smaller in size. Mast cells derived from CRACM1-deficient mice showed grossly defective degranulation and cytokine secretion, and the allergic reactions elicited in vivo were inhibited in CRACM1-deficient mice. We detected robust CRACM1 expression in skeletal muscles and some regions of the brain, heart and kidney but not in the lymphoid regions of thymus and spleen. In contrast, we found CRACM2 expression to be much higher in mouse T cells. In agreement with those findings, the store-operated calcium influx and development and proliferation of CRACM1-deficient T cells was unaffected. Thus, CRACM1 is crucial in mouse mast cell effector function, but mouse T cell calcium release-activated calcium channels are functional in the absence of CRACM1.


Subject(s)
Calcium Channels/physiology , Mast Cells/immunology , Animals , Calcium/metabolism , Calcium Channels/biosynthesis , Cell Degranulation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Mast Cells/metabolism , Mice , Mice, Knockout , ORAI1 Protein , ORAI2 Protein , Organ Specificity , Protein Subunits/biosynthesis , Protein Subunits/physiology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
13.
Cell Biol Int ; 44(3): 830-837, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31814223

ABSTRACT

Glioblastoma is the most common primary brain tumor. Glioblastoma cells secrete a significant amount of glutamate, which serve as a potential growth factor in glioma pathobiology through their specific receptor subtypes including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR). Glioblastoma express AMPAR subunits; however, its regulation and activation with downstream intracellular signaling are not well-understood. Phosphorylated-extracellular signaling-regulated kinase (ERK)1/2 is known to regulate the ionotropic glutamate receptors in cortical neurons. The mitogen-activated protein kinase cascade is frequently activated in several tumors, including glioma. Nonetheless, the association of ERK signaling with AMPAR subunits in glioblastoma is undetermined. Here, we demonstrated potential role of AMPAR in invasion, and the modulation of AMPAR subunits at transcript level by ERK signaling in glioblastoma cells. Inhibition of ERK signaling specifically downregulated the expression of calcium-permeable AMPAR subunits, GluA1 and GluA4, and upregulated calcium-impermeable AMPAR subunit GluA2 implying differential regulation of the expression of calcium-permeable AMPAR subunits of glioblastoma. Concomitantly, it significantly decreased the invasion of U87MG cells. Taken together, these findings suggest that the AMPAR enhances invasion of glioblastoma, and ERK signaling modulates the differential expression of calcium-permeable AMPAR phenotype that might play a crucial role in the invasive propensity of glioblastoma cells.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MAP Kinase Signaling System/physiology , Receptors, Glutamate/physiology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , Protein Subunits/physiology
14.
Biol Pharm Bull ; 43(7): 1118-1122, 2020.
Article in English | MEDLINE | ID: mdl-32612074

ABSTRACT

Large conductance Ca2+-activated K+ (BKCa) channels are ubiquitously expressed in plasma membrane of both excitable and non-excitable cells and possess significant physiological functions. A tetrameric complex of α subunit (BKα) forms a functional pore of BKCa channel. The properties of BKCa channel, such as voltage-dependence, Ca2+ sensitivity and pharmacological responses, are extensively modulated by co-expressing accessory ß subunits (BKß), which can associate with BKα in one to one manner. Although the functional significance of newly identified γ subunits (BKγ) has been revealed, the stoichiometry between BKα and BKγ1 remains unclear. In the present study, we utilized a single molecule fluorescence imaging with a total internal reflection fluorescence (TIRF) microscope to directly count the number of green fluorescent protein (GFP)-tagged BKγ1 (BKγ1-GFP) within a single BKCa channel complex in HEK293 cell expression system. BKγ1-GFP significantly enhanced the BK channel activity even when the intracellular Ca2+ concentration was kept lower, i.e., 10 nM, than the physiological resting level. BKγ1-GFP stably formed molecular complexes with BKα-mCherry in the plasma membrane. Counting of GFP bleaching steps revealed that a BKCa channel can contain up to four BKγ1 per channel at the maximum. These results suggest that BKγ1 forms a BKCa channel complex with BKα in a 1 : 1 stoichiometry in a human cell line.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Protein Subunits/physiology , HEK293 Cells , Humans , Optical Imaging , Single Molecule Imaging
15.
Mol Cell ; 46(5): 606-15, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22521690

ABSTRACT

In bacterial and archaeal CRISPR immune pathways, DNA sequences from invading bacteriophage or plasmids are integrated into CRISPR loci within the host genome, conferring immunity against subsequent infections. The ribonucleoprotein complex Cascade utilizes RNAs generated from these loci to target complementary "nonself" DNA sequences for destruction, while avoiding binding to "self" sequences within the CRISPR locus. Here we show that CasA, the largest protein subunit of Cascade, is required for nonself target recognition and binding. Combining a 2.3 Å crystal structure of CasA with cryo-EM structures of Cascade, we have identified a loop that is required for viral defense. This loop contacts a conserved three base pair motif that is required for nonself target selection. Our data suggest a model in which the CasA loop scans DNA for this short motif prior to target destabilization and binding, maximizing the efficiency of DNA surveillance by Cascade.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/immunology , Protein Subunits/physiology , Binding Sites , DNA/chemistry , Escherichia coli/genetics , Escherichia coli/virology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/immunology , Models, Immunological , Models, Molecular , Nucleic Acid Conformation , Protein Subunits/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Bacterial/physiology , Ribonucleoproteins/chemistry , Ribonucleoproteins/immunology , Ribonucleoproteins/physiology
16.
Bull Exp Biol Med ; 168(4): 419-422, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32146633

ABSTRACT

Whole-cell patch-clamp technique was employed to record chloride ionic current IGABA evoked by fast (600 msec) application of GABA to hippocampal pyramidal neurons and cerebellar Purkinje cells isolated from rat brain. GABA solution in the application pipette was either neutral (pH 7.4) or acidic (pH 7.0 or 6.0). Application of protons to neurons causes a rapid, reversible, and dose-dependent decrease in the amplitude of IGABA; the effect was more pronounced on hippocampal neurons (carrying both synaptic and extrasynaptic GABAA receptors) than in cerebellar Purkinje cells (predominantly equipped with synaptic GABAA receptors). In hippocampal neurons, pharmacological isolation of extrasynaptic component from total IGABA was performed with GABAA receptor antagonist gabazine (50 nM). The extrasynaptic component of IGABA was stronger blocked by protons than total IGABA. It was concluded that acidic medium produced more potent blocking effect on extrasynaptic GABAA receptors than on synaptic ones.


Subject(s)
Evoked Potentials/drug effects , Protons , Purkinje Cells/drug effects , Pyramidal Cells/drug effects , Receptors, GABA-A/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/physiology , Dose-Response Relationship, Drug , Evoked Potentials/physiology , GABA Antagonists/pharmacology , Hydrogen-Ion Concentration , Patch-Clamp Techniques , Primary Cell Culture , Protein Subunits/physiology , Purkinje Cells/cytology , Purkinje Cells/physiology , Pyramidal Cells/cytology , Pyramidal Cells/physiology , Pyridazines/pharmacology , Rats , Rats, Wistar
17.
Development ; 143(22): 4249-4260, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27729411

ABSTRACT

The brain ventricular system is essential for neurogenesis and brain homeostasis. Its neuroepithelial lining effects these functions, but the underlying molecular pathways remain to be understood. We found that the potassium channels expressed in neuroepithelial cells determine the formation of the ventricular system. The phenotype of a novel zebrafish mutant characterized by denudation of neuroepithelial lining of the ventricular system and hydrocephalus is mechanistically linked to Kcng4b, a homologue of the 'silent' voltage-gated potassium channel α-subunit Kv6.4. We demonstrated that Kcng4b modulates proliferation of cells lining the ventricular system and maintains their integrity. The gain of Kcng4b function reduces the size of brain ventricles. Electrophysiological studies suggest that Kcng4b mediates its effects via an antagonistic interaction with Kcnb1, the homologue of the electrically active delayed rectifier potassium channel subunit Kv2.1. Mutation of kcnb1 reduces the size of the ventricular system and its gain of function causes hydrocephalus, which is opposite to the function of Kcng4b. This demonstrates the dynamic interplay between potassium channel subunits in the neuroepithelium as a novel and crucial regulator of ventricular development in the vertebrate brain.


Subject(s)
Brain/embryology , Cerebral Ventricles/embryology , Organogenesis , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/physiology , Voltage-Dependent Anion Channels/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Brain/metabolism , Cell Proliferation/genetics , Cerebral Ventricles/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Hydrocephalus/embryology , Hydrocephalus/genetics , Neuroepithelial Cells/metabolism , Neuroepithelial Cells/physiology , Organogenesis/genetics , Potassium Channels, Voltage-Gated/genetics , Protein Subunits/antagonists & inhibitors , Protein Subunits/physiology , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/physiology , Zebrafish
18.
Alcohol Clin Exp Res ; 43(5): 791-802, 2019 05.
Article in English | MEDLINE | ID: mdl-30861153

ABSTRACT

BACKGROUND: Previous studies have investigated α1GABAA and α5GABAA receptor mechanisms in the behavioral effects of ethanol (EtOH) in monkeys. However, genetic studies in humans and preclinical studies with mutant mice suggest a role for α2GABAA and/or α3GABAA receptors in the effects of EtOH. The development of novel positive allosteric modulators (PAMs) with functional selectivity (i.e., selective efficacy) at α2GABAA and α3GABAA receptors allows for probing of these subtypes in preclinical models of the discriminative stimulus and reinforcing effects of EtOH in rhesus macaques. METHODS: In discrimination studies, subjects were trained to discriminate EtOH (2 g/kg, intragastrically) from water under a fixed-ratio (FR) schedule of food delivery. In oral self-administration studies, subjects were trained to self-administer EtOH (2% w/v) or sucrose (0.3 to 1% w/v) under an FR schedule of solution availability. RESULTS: In discrimination studies, functionally selective PAMs at α2GABAA and α3GABAA (HZ-166) or α3GABAA (YT-III-31) receptors substituted fully (maximum percentage of EtOH-lever responding ≥80%) for the discriminative stimulus effects of EtOH without altering response rates. Full substitution for EtOH also was engendered by a nonselective PAM (triazolam), an α5GABAA -preferring PAM (QH-ii-066) and a PAM at α2GABAA , α3GABAA , and α5GABAA receptors (L-838417). A partial (MRK-696) or an α1GABAA -preferring (zolpidem) PAM only engendered partial substitution (i.e., ~50 to 60% EtOH-lever responding). In self-administration studies, pretreatments with the functionally selective PAMs at α2GABAA and α3GABAA (XHe-II-053 and HZ-166) or α3GABAA (YT-III-31 and YT-III-271) receptors increased EtOH, but not sucrose, drinking at doses that had few, or no, observable sedative-motor effects. CONCLUSIONS: Our results confirm prior findings regarding the respective roles of α1GABAA and α5GABAA receptors in the discriminative stimulus effects of EtOH and, further, suggest a key facilitatory role for α3GABAA and potentially α2GABAA receptors in several abuse-related effects of EtOH in monkeys. Moreover, they reveal a potential role for these latter subtypes in EtOH's sedative effects.


Subject(s)
Alcoholism/psychology , Discrimination Learning/physiology , Ethanol/administration & dosage , Protein Subunits/physiology , Receptors, GABA-A/physiology , Alcoholism/drug therapy , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Discrimination Learning/drug effects , Dose-Response Relationship, Drug , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Antagonists/administration & dosage , Macaca mulatta , Male , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Self Administration
19.
Neurobiol Learn Mem ; 154: 78-86, 2018 10.
Article in English | MEDLINE | ID: mdl-29474957

ABSTRACT

Reconsolidation, a process by which long-term memories are rendered malleable following retrieval, has been shown to occur across many different species and types of memory. However, there are conditions under which memories do not reconsolidate, and the reasons for this are poorly understood. One emerging theory is that these boundary conditions are mediated by a form of metaplasticity: cellular changes through which experience can affect future synaptic plasticity. We review evidence that N-methyl-D-aspartate receptors (NMDARs) might contribute to this phenomenon, and hypothesize that resistance to memory destabilization may be mediated by the ratio of GluN2A/GluN2B subunits that make up these receptors. Qualities such as memory strength and the age of the memory may increase the GluN2A/GluN2B ratio, reducing the ability of reactivation cues to induce destabilization, thereby preventing reconsolidation. Other examples of experience-dependent learning and evolutionary perspectives of reconsolidation are also discussed.


Subject(s)
Memory Consolidation/physiology , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cues , Humans , Models, Neurological , Protein Subunits/physiology
20.
Nat Rev Neurosci ; 14(6): 383-400, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23686171

ABSTRACT

NMDA receptors (NMDARs) are glutamate-gated ion channels and are crucial for neuronal communication. NMDARs form tetrameric complexes that consist of several homologous subunits. The subunit composition of NMDARs is plastic, resulting in a large number of receptor subtypes. As each receptor subtype has distinct biophysical, pharmacological and signalling properties, there is great interest in determining whether individual subtypes carry out specific functions in the CNS in both normal and pathological conditions. Here, we review the effects of subunit composition on NMDAR properties, synaptic plasticity and cellular mechanisms implicated in neuropsychiatric disorders. Understanding the rules and roles of NMDAR diversity could provide new therapeutic strategies against dysfunctions of glutamatergic transmission.


Subject(s)
Mental Disorders/physiopathology , Neuronal Plasticity/physiology , Protein Subunits/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Humans , Models, Biological , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL