Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biophys J ; 123(12): 1676-1689, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38751114

ABSTRACT

The best-known mode of action of calmodulin (CaM) is binding of Ca2+ to its N- and C-domains, followed by binding to target proteins. An underappreciated facet of this process is that CaM is typically bound to proteins at basal levels of free Ca2+, including the small, intrinsically disordered, neuronal IQ-motif proteins called PEP-19 and neurogranin (Ng). PEP-19 and Ng would not be effective competitive inhibitors of high-affinity Ca2+-dependent CaM targets at equilibrium because they bind to CaM with relatively low affinity, but they could influence the time course of CaM signaling by affecting the rate of association of CaM with high-affinity Ca2+-dependent targets. This mode of regulation may be domain specific because PEP-19 binds to the C-domain of CaM, whereas Ng binds to both N- and C-domains. In this report, we used a model CaM binding peptide (CKIIp) to characterize the preferred pathway of complex formation with Ca2+-CaM at low levels of free Ca2+ (0.25-1.5 µM), and how PEP-19 and Ng affect this process. We show that the dominant encounter complex involves association of CKIIp with the N-domain of CaM, even though the C-domain has a greater affinity for Ca2+. We also show that Ng greatly decreases the rate of association of Ca2+-CaM with CKIIp due to the relatively slow dissociation of Ng from CaM, and to interactions between the Gly-rich C-terminal region of Ng with the N-domain of CaM, which inhibits formation of the preferred encounter complex with CKIIp. These results provide the general mechanistic paradigms that binding CaM to targets can be driven by its N-domain, and that low-affinity regulators of CaM signaling have the potential to influence the rate of activation of high-affinity CaM targets and potentially affect the distribution of limited CaM among multiple targets during Ca2+ oscillations.


Subject(s)
Calmodulin , Neurogranin , Protein Binding , Calmodulin/metabolism , Calmodulin/chemistry , Neurogranin/metabolism , Calcium/metabolism , Peptides/metabolism , Peptides/chemistry , Protein Domains , Kinetics , Amino Acid Sequence , Animals
2.
bioRxiv ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38562851

ABSTRACT

The best-known mode of action of calmodulin (CaM) is binding of Ca 2+ to its N- and C-domains, followed by binding to target proteins. An underappreciated facet of this process is that CaM is typically bound to proteins at basal levels of free Ca 2+ , including the small, intrinsically disordered, neuronal IQ-motif proteins called PEP-19 and neurogranin (Ng). PEP-19 and Ng would not be effective competitive inhibitors of high-affinity Ca 2+ -dependent CaM targets at equilibrium since they bind to CaM with relatively low affinity, but they could influence the time course of CaM signaling by affecting the rate of association of CaM with high-affinity Ca 2+ -dependent targets. This mode of regulation may domain specific since PEP-19 binds to the C-domain of CaM, while Ng binds to both N- and C-domains. In this report, we used a model CaM binding peptide (CKIIp) to characterize the preferred pathway of complex formation with Ca 2+ -CaM at low levels of free Ca 2+ (0.25 to 1.5 µM), and how PEP-19 and Ng affect this process. We show that the dominant encounter complex involves association of CKIIp with the N-domain of CaM, even though the C-domain has a greater affinity for Ca 2+ . We also show that Ng greatly decreases the rate of association of Ca 2+ -CaM with CKIIp due to the relatively slow dissociation of Ng from CaM, and to interactions between the Gly-rich C-terminal region of Ng with the N-domain of CaM, which inhibits formation of the preferred encounter complex with CKIIp. These results provide the general mechanistic paradigms that binding CaM to targets can be driven by its N-domain, and that low-affinity regulators of CaM signaling have the potential to influence the rate of activation of high-affinity CaM targets and potentially affect the distribution of limited CaM among multiple targets during Ca 2+ oscillations. STATEMENT OF SIGNIFICANCE: Calmodulin is a small, essential regulator of multiple cellular processes including growth and differentiation. Its best-known mode of action is to first bind calcium and then bind and regulate the activity of target proteins. Each domain of CaM has distinct calcium binding properties and can interact with targets in distinct ways. We show here that the N-domain of calmodulin can drive its association with targets, and that a small, intrinsically disordered regulator of calmodulin signaling called neurogranin can greatly decrease the rate of association of CaM with high-affinity Ca 2+ -dependent targets. These results demonstrate the potential of neurogranin, and potentially other proteins, to modulate the time course of activation of targets by a limited intracellular supply of calmodulin.

3.
J Biomol NMR ; 75(6-7): 233-244, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34176062

ABSTRACT

We used NMR to show that the antipsychotic phenothiazine drugs promazine and promethazine bind to GDP-KRAS. Promazine also binds to oncogenic GDP-KRAS(G12D), and to wild type GppNHp-KRAS. A panel of additional phenothiazines bind to GDP-KRAS but with lower affinity than promazine or promethazine. Binding is most dependent on substitutions at C-2 of the tricyclic phenothiazine ring. Promazine was used to generate an NMR-driven HADDOCK model of the drug/GDP-KRAS complex. The structural model shows the tricyclic phenothiazine ring of promazine associates with the hydrophobic pocket p1 that is bordered by the central ß sheet and Switch II in KRAS. Binding appears to stabilize helix 2 in a conformation that is similar to that seen in KRAS bound to other small molecules. Association of phenothiazines with KRAS may affect normal KRAS signaling that could contribute to multiple biological activities of these antipsychotic drugs. Moreover, the phenothiazine ring represents a new core scaffold on which to design modulators of KRAS activity.


Subject(s)
Antipsychotic Agents/chemistry , Models, Molecular , Phenothiazines/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Amino Acid Substitution , Humans , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation, beta-Strand , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
4.
Chem Biol Drug Des ; 94(2): 1441-1456, 2019 08.
Article in English | MEDLINE | ID: mdl-30903639

ABSTRACT

RAS mutations account for >15% of all human tumors, and of these ~85% are due to mutations in a particular RAS gene: KRAS. Recent studies revealed that KRAS harbors four druggable allosteric sites. Here, we have (a) used molecular simulations to generate ensembles of wild type and four major oncogenic KRAS mutants (G12V, G12D, G13D, and Q61H); (b) characterized the druggability of each allosteric pocket in each protein; (c) conducted extensive ensemble-based virtual screening using pocket-tailored ligand libraries; (d) prioritized hits through hierarchical postdocking analysis; and (e) validated predicted hits with NMR. Of the 785 diverse potential hits identified by our in silico analysis, we tested 90 for their ability to bind KRAS using NMR and found that nine cause backbone amide chemical shift perturbations of residues near the functionally responsive switch loops, suggesting potential binding. We conducted detailed biophysical analyses on a novel indole-based compound to demonstrate the potential of our workflow to yield lead compounds. We believe the detailed information documented in this work regarding the druggability profile of each allosteric site and the chemical fingerprints of compounds that target them will serve as vital resources for future structure-based drug design efforts against KRAS, a high-value target for cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Mutation, Missense , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/chemistry , Amino Acid Substitution , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Nuclear Magnetic Resonance, Biomolecular , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
5.
Proc Natl Acad Sci U S A ; 115(24): E5477-E5486, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29844180

ABSTRACT

Covalently cross-linked pilus polymers displayed on the cell surface of Gram-positive bacteria are assembled by class C sortase enzymes. These pilus-specific transpeptidases located on the bacterial membrane catalyze a two-step protein ligation reaction, first cleaving the LPXTG motif of one pilin protomer to form an acyl-enzyme intermediate and then joining the terminal Thr to the nucleophilic Lys residue residing within the pilin motif of another pilin protomer. To date, the determinants of class C enzymes that uniquely enable them to construct pili remain unknown. Here, informed by high-resolution crystal structures of corynebacterial pilus-specific sortase (SrtA) and utilizing a structural variant of the enzyme (SrtA2M), whose catalytic pocket has been unmasked by activating mutations, we successfully reconstituted in vitro polymerization of the cognate major pilin (SpaA). Mass spectrometry, electron microscopy, and biochemical experiments authenticated that SrtA2M synthesizes pilus fibers with correct Lys-Thr isopeptide bonds linking individual pilins via a thioacyl intermediate. Structural modeling of the SpaA-SrtA-SpaA polymerization intermediate depicts SrtA2M sandwiched between the N- and C-terminal domains of SpaA harboring the reactive pilin and LPXTG motifs, respectively. Remarkably, the model uncovered a conserved TP(Y/L)XIN(S/T)H signature sequence following the catalytic Cys, in which the alanine substitutions abrogated cross-linking activity but not cleavage of LPXTG. These insights and our evidence that SrtA2M can terminate pilus polymerization by joining the terminal pilin SpaB to SpaA and catalyze ligation of isolated SpaA domains in vitro provide a facile and versatile platform for protein engineering and bio-conjugation that has major implications for biotechnology.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Corynebacterium/metabolism , Cysteine Endopeptidases/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Catalysis , Cell Wall/metabolism , Crystallography, X-Ray/methods , Peptidyl Transferases/metabolism , Polymerization
6.
Cell Rep ; 21(7): 1968-1981, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29141226

ABSTRACT

Increasing evidence has demonstrated that small nucleolar RNAs (snoRNAs) play important roles in tumorigenesis. We systematically investigated the expression landscape and clinical relevance of snoRNAs in >10,000 samples across 31 cancer types from The Cancer Genome Atlas. We observed overall elevated expression of snoRNAs and their ribonucleoproteins in multiple cancer types. We showed complex regulation of snoRNA expression by their host genes, copy number variation, and DNA methylation. Unsupervised clustering revealed that the snoRNA expression subtype is highly concordant with other molecular/clinical subtypes. We further identified 46 clinically relevant snoRNAs and experimentally demonstrated functional roles of SNORD46 in promoting cell proliferation, migration, and invasion. We developed a user-friendly data portal, SNORic, to benefit the research community. Our study highlights the significant roles of snoRNAs in the development and implementation of biomarkers or therapeutic targets for cancer and provides a valuable resource for cancer research.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA, Small Nucleolar/genetics , Biomarkers, Tumor/metabolism , DNA Methylation , Gene Dosage , Humans , Neoplasms/metabolism , Neoplasms/pathology , RNA, Small Nucleolar/metabolism , Software
7.
Nat Commun ; 7: 13583, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27876793

ABSTRACT

PEP-19 is a small protein that increases the rates of Ca2+ binding to the C-domain of calmodulin (CaM) by an unknown mechanism. Although an IQ motif promotes binding to CaM, an acidic sequence in PEP-19 is required to modulate Ca2+ binding and to sensitize HeLa cells to ATP-induced Ca2+ release. Here, we report the NMR solution structure of a complex between PEP-19 and the C-domain of apo CaM. The acidic sequence of PEP-19 associates between helices E and F of CaM via hydrophobic interactions. This allows the acidic side chains in PEP-19 to extend toward the solvent and form a negatively charged surface that resembles a catcher's mitt near Ca2+ binding loop III of CaM. The topology and gradients of negative electrostatic surface potential support a mechanism by which PEP-19 increases the rate of Ca2+ binding to the C-domain of CaM by 'catching' and electrostatically steering Ca2+ to site III.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Nerve Tissue Proteins/chemistry , Amino Acid Sequence , Calmodulin/chemistry , Gene Expression Regulation , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation
8.
Biophys J ; 109(3): 510-20, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26244733

ABSTRACT

Protein signaling occurs in crowded intracellular environments, and while high concentrations of macromolecules are postulated to modulate protein-protein interactions, analysis of their impact at each step of the reaction pathway has not been systematically addressed. Potential cosolute-induced alterations in target association are particularly important for a signaling molecule like calmodulin (CaM), where competition among >300 targets governs which pathways are selectively activated. To explore how high concentrations of cosolutes influence CaM-target affinity and kinetics, we methodically investigated each step of the CaM-target binding mechanism under crowded or osmolyte-rich environments mimicked by ficoll-70, dextran-10, and sucrose. All cosolutes stabilized compact conformers of CaM and modulated association kinetics by affecting diffusion and rates of conformational change; however, the results showed that differently sized molecules had variable effects to enhance or impede unique steps of the association pathway. On- and off-rates were modulated by all cosolutes in a compensatory fashion, producing little change in steady-state affinity. From this work insights were gained on how high concentrations of inert crowding agents and osmolytes fit into a kinetic framework to describe protein-protein interactions relevant for cellular signaling.


Subject(s)
Calmodulin/chemistry , Molecular Dynamics Simulation , Amino Acid Sequence , Animals , Calmodulin/metabolism , Molecular Sequence Data , Osmolar Concentration , Protein Binding
9.
J Biol Chem ; 289(21): 14644-55, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24713697

ABSTRACT

Neurogranin (Ng) is a member of the IQ motif class of calmodulin (CaM)-binding proteins, and interactions with CaM are its only known biological function. In this report we demonstrate that the binding affinity of Ng for CaM is weakened by Ca(2+) but to a lesser extent (2-3-fold) than that previously suggested from qualitative observations. We also show that Ng induced a >10-fold decrease in the affinity of Ca(2+) binding to the C-terminal domain of CaM with an associated increase in the Ca(2+) dissociation rate. We also discovered a modest, but potentially important, increase in the cooperativity in Ca(2+) binding to the C-lobe of CaM in the presence of Ng, thus sharpening the threshold for the C-domain to become Ca(2+)-saturated. Domain mapping using synthetic peptides indicated that the IQ motif of Ng is a poor mimetic of the intact protein and that the acidic sequence just N-terminal to the IQ motif plays an important role in reproducing Ng-mediated decreases in the Ca(2+) binding affinity of CaM. Using NMR, full-length Ng was shown to make contacts largely with residues in the C-domain of CaM, although contacts were also detected in residues in the N-terminal domain. Together, our results can be consolidated into a model where Ng contacts residues in the N- and C-lobes of both apo- and Ca(2+)-bound CaM and that although Ca(2+) binding weakens Ng interactions with CaM, the most dramatic biochemical effect is the impact of Ng on Ca(2+) binding to the C-terminal lobe of CaM.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Neurogranin/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Binding Sites/genetics , Binding, Competitive , Blotting, Western , Calcium/chemistry , Calmodulin/chemistry , Calorimetry/methods , Humans , Kinetics , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Neurogranin/chemistry , Neurogranin/genetics , Protein Binding
10.
PLoS One ; 8(5): e62861, 2013.
Article in English | MEDLINE | ID: mdl-23658780

ABSTRACT

Calmodulin, an intracellular calcium-binding protein, is thought to regulate ectodomain shedding of many membrane proteins, but the underlying molecular mechanism has remained unclear. Basing on a solution structure of calcium-loaded calmodulin in complex with a L-selectin fragment that contains a portion of its transmembrane domain, Gifford et al. (University of Calgary) recently suggested that calmodulin regulates L-selectin shedding by binding directly to a portion of the L-selectin transmembrane domain in a compact conformation. Using fluorescently labeled calmodulin, we show however that calmodulin adopts a distinctly different and much more extended conformation when it binds to the CLS peptide (i.e. the entire transmembrane and cytoplasmic domains of L-selectin) reconstituted in the phosphatidylcholine liposome with micromolar dissociation constant and in a calcium-independent manner. Calmodulin adopts a similarly extended conformation in a ternary complex with the N-terminal FERM domain of moesin and CLS reconstituted in the phospholipid liposome that mimics the native membrane environment. These results indicate that calmodulin does not bind directly to the transmembrane domain of L-selectin. Understanding the association of calmodulin with L-selectin helps to shed light on the mechanisms underlying regulation of ectodomain shedding.


Subject(s)
Calmodulin/chemistry , Calmodulin/metabolism , Cell Membrane/metabolism , L-Selectin/metabolism , Amino Acid Sequence , Humans , L-Selectin/chemistry , Liposomes/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/metabolism , Phosphatidylcholines/metabolism , Protein Binding , Protein Structure, Tertiary , Water/chemistry
11.
J Biol Chem ; 288(3): 2040-8, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23204517

ABSTRACT

PEP-19 is a small, intrinsically disordered protein that binds to the C-domain of calmodulin (CaM) via an IQ motif and tunes its Ca(2+) binding properties via an acidic sequence. We show here that the acidic sequence of PEP-19 has intrinsic Ca(2+) binding activity, which may modulate Ca(2+) binding to CaM by stabilizing an initial Ca(2+)-CaM complex or by electrostatically steering Ca(2+) to and from CaM. Because PEP-19 is expressed in cells that exhibit highly active Ca(2+) dynamics, we tested the hypothesis that it influences ligand-dependent Ca(2+) release. We show that PEP-19 increases the sensitivity of HeLa cells to ATP-induced Ca(2+) release to greatly increase the percentage of cells responding to sub-saturating doses of ATP and increases the frequency of Ca(2+) oscillations. Mutations in the acidic sequence of PEP-19 that inhibit or prevent it from modulating Ca(2+) binding to CaM greatly inhibit its effect on ATP-induced Ca(2+) release. Thus, this cellular effect of PEP-19 does not depend simply on binding to CaM via the IQ motif but requires its acidic metal binding domain. Tuning the activities of Ca(2+) mobilization pathways places PEP-19 at the top of CaM signaling cascades, with great potential to exert broad effects on downstream CaM targets, thus expanding the biological significance of this small regulator of CaM signaling.


Subject(s)
Calcium Signaling , Calcium/metabolism , Calmodulin/metabolism , Nerve Tissue Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Binding Sites , Calmodulin/chemistry , HeLa Cells , Humans , Kinetics , Ligands , Molecular Imaging , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Static Electricity , Transfection
12.
J Mol Biol ; 411(1): 220-33, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21664913

ABSTRACT

The calmodulin (CaM) hypothesis of ectodomain shedding stipulates that CaM, an intracellular Ca²âº-dependent regulatory protein, associates with the cytoplasmic domain of L-selectin to regulate ectodomain shedding of L-selectin on the other side of the plasma membrane. To understand the underlying molecular mechanism, we have characterized the interactions of CaM with two peptides derived from human L-selectin. The peptide ARR18 corresponds to the entire cytoplasmic domain of L-selectin (residues Ala317-Tyr334 in the mature protein), and CLS corresponds to residues Lys280-Tyr334, which contains the entire transmembrane and cytoplasmic domains of l-selectin. Monitoring the interaction by fluorescence spectroscopy and other biophysical techniques, we found that CaM can bind to ARR18 in aqueous solutions or the L-selectin cytoplasmic domain of CLS reconstituted in the phosphatidylcholine bilayer, both with an affinity of approximately 2 µM. The association is calcium independent and dynamic and involves both lobes of CaM. In a phospholipid bilayer, the positively charged L-selectin cytoplasmic domain of CLS is associated with anionic phosphatidylserine (PS) lipids at the membrane interface through electrostatic interactions. Under conditions where the PS content mimics that in the inner leaflet of the cell plasma membrane, the interaction between CaM and CLS becomes undetectable. These results suggest that the association of CaM with L-selectin in the cell can be influenced by the membrane bilayer and that anionic lipids may modulate ectodomain shedding of transmembrane receptors.


Subject(s)
Calmodulin/metabolism , L-Selectin/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , Humans , Kinetics , Liposomes/metabolism , Molecular Sequence Data , Phosphatidylserines/metabolism , Protein Binding , Protein Interaction Mapping , Spectrometry, Fluorescence , Static Electricity
13.
Biochemistry ; 49(48): 10287-97, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-20973509

ABSTRACT

PEP-19 (Purkinje cell protein 4) is an intrinsically disordered protein with an IQ calmodulin (CaM) binding motif. Expression of PEP-19 was recently shown to protect cells from apoptosis and cell death due to Ca(2+) overload. Our initial studies showed that PEP-19 causes novel and dramatic increases in the rates of association of Ca(2+) with and dissociation of Ca(2+) from the C-domain of CaM. The goal of this work was to study interactions between the C-domain of CaM (C-CaM) and PEP-19 by solution nuclear magnetic resonance (NMR) to identify mechanisms by which PEP-19 regulates binding of Ca(2+) to CaM. Our results show that PEP-19 causes a greater structural change in apo C-CaM than in Ca(2+)-C-CaM, and that the first Ca(2+) binds preferentially to site IV in the presence of PEP-19 with exchange characteristics that are consistent with a decrease in Ca(2+) binding cooperativity. Relatively weak binding of PEP-19 has distinct effects on chemical and conformational exchange on the microsecond to millisecond time scale. In apo C-CaM, PEP-19 binding causes a redistribution of residues that experience conformational exchange, leading to an increase in the number of residues around Ca(2+) binding site IV that undergo conformational exchange on the microsecond to millisecond time scale. This appears to be caused by an allosteric effect because these residues are not localized to the PEP-19 binding site. In contrast, PEP-19 increases the number of residues that exhibit conformational exchange in Ca(2+)-C-CaM. These residues are primarily localized to the PEP-19 binding site but also include Asp93 in site III. These results provide working models for the role of protein dynamics in the regulation of binding of Ca(2+) to CaM by PEP-19.


Subject(s)
Apoproteins/metabolism , Calcium/metabolism , Calmodulin/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Apoproteins/chemistry , Binding Sites , Calmodulin/chemistry , Kinetics , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Reproducibility of Results
14.
J Biol Chem ; 285(11): 8094-103, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20048169

ABSTRACT

The IQ-motif protein PEP-19, binds to the C-domain of calmodulin (CaM) with significantly different k(on) and k(off) rates in the presence and absence of Ca(2+), which could play a role in defining the levels of free CaM during Ca(2+) transients. The initial goal of the current study was to determine whether Ca(2+) binding to sites III or IV in the C-domain of CaM was responsible for affecting the kinetics of binding PEP-19. EF-hand Ca(2+)-binding sites were selectively inactivated by the common strategy of changing Asp to Ala at the X-coordination position. Although Ca(2+) binding to both sites III and IV appeared necessary for native-like interactions with PEP-19, the data also indicated that the mutations caused undesirable structural alterations as evidenced by significant changes in amide chemical shifts for apoCaM. Mutations in the C-domain also affected chemical shifts in the unmodified N-domain, and altered the Ca(2+) binding properties of the N-domain. Conversion of Asp(93) to Ala caused the greatest structural perturbations, possibly due to the loss of stabilizing hydrogen bonds between the side chain of Asp(93) and backbone amides in apo loop III. Thus, although these mutations inhibit binding of Ca(2+), the mutated CaM may not be able to support potentially important native-like activity of the apoprotein. This should be taken into account when designing CaM mutants for expression in cell culture.


Subject(s)
Calcium/metabolism , Calmodulin , Alanine/metabolism , Animals , Aspartic Acid/metabolism , Binding Sites/physiology , Calmodulin/chemistry , Calmodulin/genetics , Calmodulin/metabolism , Fluorescence Resonance Energy Transfer , Hydrogen Bonding , Mammals , Mutagenesis, Site-Directed , Nerve Tissue Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary , Structure-Activity Relationship
15.
J Comput Neurosci ; 27(3): 621-38, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19609660

ABSTRACT

Calmodulin (CaM) is a major Ca(2+) binding protein involved in two opposing processes of synaptic plasticity of CA1 pyramidal neurons: long-term potentiation (LTP) and depression (LTD). The N- and C-terminal lobes of CaM bind to its target separately but cooperatively and introduce complex dynamics that cannot be well understood by experimental measurement. Using a detailed stochastic model constructed upon experimental data, we have studied the interaction between CaM and Ca(2+)-CaM-dependent protein kinase II (CaMKII), a key enzyme underlying LTP. The model suggests that the accelerated binding of one lobe of CaM to CaMKII, when the opposing lobe is already bound to CaMKII, is a critical determinant of the cooperative interaction between Ca(2+), CaM, and CaMKII. The model indicates that the target-bound Ca(2+) free N-lobe has an extended lifetime and may regulate the Ca(2+) response of CaMKII during LTP induction. The model also reveals multiple kinetic pathways which have not been previously predicted for CaM-dissociation from CaMKII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/metabolism , Long-Term Potentiation/physiology , Models, Biological , Stochastic Processes , Algorithms , Animals , Binding Sites , Calcium/metabolism , Calcium/pharmacology , Calcium Signaling/physiology , Dose-Response Relationship, Drug , Time Factors
16.
J Biol Chem ; 284(12): 7455-64, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19106096

ABSTRACT

PEP-19 is a small calmodulin (CaM)-binding protein that greatly increases the rates of association and dissociation of Ca(2+) from the C-domain of CaM, an effect that is mediated by an acidic/IQ sequence in PEP-19. We show here using NMR that PEP-19 is an intrinsically disordered protein, but with residual structure localized to its acidic/IQ motif. We also show that the k(on) and k(off) rates for binding PEP-19 to apo-CaM are at least 50-fold slower than for binding to Ca(2+)-CaM. These data indicate that intrinsic disorder confers plasticity that allows PEP-19 to bind to either apo- or Ca(2+)-CaM via different structural modes, and that complex formation may be facilitated by conformational selection of residual structure in the acidic/IQ sequence.


Subject(s)
Calmodulin/chemistry , Nerve Tissue Proteins/chemistry , Signal Transduction/physiology , Amino Acid Motifs/physiology , Animals , Calcium/chemistry , Calcium/metabolism , Calmodulin/metabolism , Humans , Nerve Tissue Proteins/metabolism , Protein Binding/physiology , Protein Structure, Tertiary/physiology
17.
J Neurophysiol ; 99(1): 264-76, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17959737

ABSTRACT

Calmodulin (CaM) is most recognized for its role in activating Ca(2+)-CaM-dependent enzymes following increased intracellular Ca(2+). However, CaM's high intracellular concentration indicates CaM has the potential to play a significant role as a Ca(2+) buffer. Neurogranin (Ng) is a small neuronal IQ-motif-containing protein that accelerates Ca(2+) dissociation from CaM. In cells that contain high concentrations of both Ng and CaM, like CA1 pyramidal neurons, we hypothesize that the accelerated Ca(2+) dissociation from CaM by Ng decreases the buffering capacity of CaM and thereby shapes the transient dynamics of intracellular free Ca(2+). We examined this hypothesis using a mathematical model constructed on the known biochemistry of Ng and confirmed the simulation results with Ca(2+) imaging data in the literature. In a single-compartment model that contains no Ca(2+) extrusion mechanism, Ng increased the steady-state free Ca(2+). However, in the presence of a Ca(2+) extrusion mechanism, Ng accelerated the decay rate of free Ca(2+) through its ability to increase the Ca(2+) dissociation from CaM, which in turn becomes subject to Ca(2+) extrusion. Interestingly, PEP-19, another neuronal IQ-motif protein that accelerates both Ca(2+) association and dissociation from CaM, appears to have the opposite impact than that of Ng on free Ca(2+). As such, Ng may regulate, in addition to the Ca(2+)-CaM-dependent process, Ca(2+)-sensitive enzymes by influencing the buffering capacity of CaM and subsequently free Ca(2+) levels. We examined the relative impact of these Ng-induced effects in the induction of synaptic plasticity.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Hippocampus/metabolism , Intracellular Fluid/metabolism , Neurogranin/metabolism , Neurons/metabolism , Action Potentials/physiology , Amino Acid Motifs/physiology , Animals , Buffers , Calcium Signaling/physiology , Computer Simulation , Cytoplasm/metabolism , Humans , Mice , Mice, Knockout , Models, Neurological , Nerve Tissue Proteins/metabolism , Neurogranin/chemistry , Neurogranin/genetics , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Up-Regulation/physiology
18.
J Biol Chem ; 283(3): 1401-1410, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-17991744

ABSTRACT

The small IQ motif proteins PEP-19 (62 amino acids) and RC3 (78 amino acids) greatly accelerate the rates of Ca(2+) binding to sites III and IV in the C-domain of calmodulin (CaM). We show here that PEP-19 decreases the degree of cooperativity of Ca(2+) binding to sites III and IV, and we present a model showing that this could increase Ca(2+) binding rate constants. Comparative sequence analysis showed that residues 28 to 58 from PEP-19 are conserved in other proteins. This region includes the IQ motif (amino acids 39-62), and an adjacent acidic cluster of amino acids (amino acids 28-40). A synthetic peptide spanning residues 28-62 faithfully mimics intact PEP-19 with respect to increasing the rates of Ca(2+) association and dissociation, as well as binding preferentially to the C-domain of CaM. In contrast, a peptide encoding only the core IQ motif does not modulate Ca(2+) binding, and binds to multiple sites on CaM. A peptide that includes only the acidic region does not bind to CaM. These results show that PEP-19 has a novel acidic/IQ CaM regulatory motif in which the IQ sequence provides a targeting function that allows binding of PEP-19 to CaM, whereas the acidic residues modify the nature of this interaction, and are essential for modulating Ca(2+) binding to the C-domain of CaM.


Subject(s)
Calmodulin/metabolism , Peptides/metabolism , Amides , Amino Acid Motifs , Amino Acid Sequence , Calcium/metabolism , Calmodulin/chemistry , Consensus Sequence , Kinetics , Molecular Sequence Data , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Interaction Mapping , Sequence Alignment
19.
Biophys J ; 93(11): 3848-59, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17704141

ABSTRACT

Neurogranin (Ng) is a postsynaptic IQ-motif containing protein that accelerates Ca(2+) dissociation from calmodulin (CaM), a key regulator of long-term potentiation and long-term depression in CA1 pyramidal neurons. The exact physiological role of Ng, however, remains controversial. Two genetic knockout studies of Ng showed opposite outcomes in terms of the induction of synaptic plasticity. To understand its function, we test the hypothesis that Ng could regulate the spatial range of action of Ca(2+)/CaM based on its ability to accelerate the dissociation of Ca(2+) from CaM. Using a mathematical model constructed on the known biochemistry of Ng, we calculate the cycle time that CaM molecules alternate between the fully Ca(2+) saturated state and the Ca(2+) unbound state. We then use these results and include diffusion of CaM to illustrate the impact that Ng has on modulating the spatial profile of Ca(2+)-saturated CaM within a model spine compartment. Finally, the first-passage time of CaM to transition from the Ca(2+)-free state to the Ca(2+)-saturated state was calculated with or without Ng present. These analyses suggest that Ng regulates the encounter rate between Ca(2+) saturated CaM and its downstream targets during postsynaptic Ca(2+) transients.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Calmodulin/metabolism , Excitatory Postsynaptic Potentials/physiology , Models, Neurological , Neurogranin/metabolism , Pyramidal Cells/physiology , Animals , Calcium/metabolism , Computer Simulation , Humans , Neurotransmitter Agents/metabolism
20.
Structure ; 14(10): 1547-56, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17027503

ABSTRACT

Calmodulin regulates ryanodine receptor-mediated Ca(2+) release through a conserved binding site. The crystal structure of Ca(2+)-calmodulin bound to this conserved site reveals that calmodulin recognizes two hydrophobic anchor residues at a novel "1-17" spacing that brings the calmodulin lobes close together but prevents them from contacting one another. NMR residual dipolar couplings demonstrate that the detailed structure of each lobe is preserved in solution but also show that the lobes experience domain motions within the complex. FRET measurements confirm the close approach of the lobes in binding the 1-17 target and show that calmodulin binds with one lobe to a peptide lacking the second anchor. We suggest that calmodulin regulates the Ca(2+) channel by switching between the contiguous binding mode seen in our crystal structure and a state where one lobe of calmodulin contacts the conserved binding site while the other interacts with a noncontiguous site on the channel.


Subject(s)
Calcium/chemistry , Calmodulin/chemistry , Models, Molecular , Ryanodine Receptor Calcium Release Channel/chemistry , Amino Acid Sequence , Binding Sites , Fluorescence Resonance Energy Transfer , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL
...