Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
EMBO J ; 31(17): 3635-46, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-22850675

ABSTRACT

Stomatin proteins oligomerize at membranes and have been implicated in ion channel regulation and membrane trafficking. To obtain mechanistic insights into their function, we determined three crystal structures of the conserved stomatin domain of mouse stomatin that assembles into a banana-shaped dimer. We show that dimerization is crucial for the repression of acid-sensing ion channel 3 (ASIC3) activity. A hydrophobic pocket at the inside of the concave surface is open in the presence of an internal peptide ligand and closes in the absence of this ligand, and we demonstrate a function of this pocket in the inhibition of ASIC3 activity. In one crystal form, stomatin assembles via two conserved surfaces into a cylindrical oligomer, and these oligomerization surfaces are also essential for the inhibition of ASIC3-mediated currents. The assembly mode of stomatin uncovered in this study might serve as a model to understand oligomerization processes of related membrane-remodelling proteins, such as flotillin and prohibitin.


Subject(s)
Acid Sensing Ion Channels/metabolism , Blood Proteins/metabolism , Membrane Proteins/metabolism , Acid Sensing Ion Channels/chemistry , Animals , Blood Proteins/chemistry , Blood Proteins/genetics , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dimerization , Fibroblasts , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Transgenic , Protein Structure, Tertiary , Rats
2.
Nature ; 465(7297): 502-6, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20428112

ABSTRACT

The interferon-inducible dynamin-like myxovirus resistance protein 1 (MxA; also called MX1) GTPase is a key mediator of cell-autonomous innate immunity against pathogens such as influenza viruses. MxA partially localizes to COPI-positive membranes of the smooth endoplasmic reticulum-Golgi intermediate compartment. At the point of infection, it redistributes to sites of viral replication and promotes missorting of essential viral constituents. It has been proposed that the middle domain and the GTPase effector domain of dynamin-like GTPases constitute a stalk that mediates oligomerization and transmits conformational changes from the G domain to the target structure; however, the molecular architecture of this stalk has remained elusive. Here we report the crystal structure of the stalk of human MxA, which folds into a four-helical bundle. This structure tightly oligomerizes in the crystal in a criss-cross pattern involving three distinct interfaces and one loop. Mutations in each of these interaction sites interfere with native assembly, oligomerization, membrane binding and antiviral activity of MxA. On the basis of these results, we propose a structural model for dynamin oligomerization and stimulated GTP hydrolysis that is consistent with previous structural predictions and has functional implications for all members of the dynamin family.


Subject(s)
Dynamins/chemistry , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Protein Multimerization , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Binding Sites , Cell Line , Crystallography, X-Ray , Dynamins/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/pharmacology , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/physiology , Models, Molecular , Myxovirus Resistance Proteins , Protein Conformation , Virus Replication/drug effects
3.
J Biol Chem ; 287(11): 8633-40, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22262864

ABSTRACT

In yeast, the membrane-bound HMG-CoA reductase degradation (HRD) ubiquitin-ligase complex is a key player of the ER-associated protein degradation pathway that targets misfolded proteins for proteolysis. Yos9, a component of the luminal submodule of the ligase, scans proteins for specific oligosaccharide modifications, which constitute a critical determinant of the degradation signal. Here, we report the crystal structure of the Yos9 domain that was previously suggested to confer binding to Hrd3, another component of the HRD complex. We observe an αß-roll domain architecture and a dimeric assembly which are confirmed by analytical ultracentrifugation of both the crystallized domain and full-length Yos9. Our binding studies indicate that, instead of this domain, the N-terminal part of Yos9 including the mannose 6-phosphate receptor homology domain mediates the association with Hrd3 in vitro. Our results support the model of a dimeric state of the HRD complex and provide first-time evidence of self-association on its luminal side.


Subject(s)
Carrier Proteins/chemistry , Hydroxymethylglutaryl CoA Reductases/chemistry , Models, Molecular , Multienzyme Complexes/chemistry , Protein Multimerization , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Ubiquitin-Protein Ligases/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallography, X-Ray , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Protein Structure, Tertiary , Proteolysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response/physiology
4.
Proc Natl Acad Sci U S A ; 107(47): 20299-304, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059949

ABSTRACT

GTPases of immunity-associated proteins (GIMAPs) are a distinctive family of GTPases, which control apoptosis in lymphocytes and play a central role in lymphocyte maturation and lymphocyte-associated diseases. To explore their function and mechanism, we determined crystal structures of a representative member, GIMAP2, in different nucleotide-loading and oligomerization states. Nucleotide-free and GDP-bound GIMAP2 were monomeric and revealed a guanine nucleotide-binding domain of the TRAFAC (translation factor associated) class with a unique amphipathic helix α7 packing against switch II. In the absence of α7 and the presence of GTP, GIMAP2 oligomerized via two distinct interfaces in the crystal. GTP-induced stabilization of switch I mediates dimerization across the nucleotide-binding site, which also involves the GIMAP specificity motif and the nucleotide base. Structural rearrangements in switch II appear to induce the release of α7 allowing oligomerization to proceed via a second interface. The unique architecture of the linear oligomer was confirmed by mutagenesis. Furthermore, we showed a function for the GIMAP2 oligomer at the surface of lipid droplets. Although earlier studies indicated that GIMAPs are related to the septins, the current structure also revealed a strikingly similar nucleotide coordination and dimerization mode as in the dynamin GTPase. Based on this, we reexamined the relationships of the septin- and dynamin-like GTPases and demonstrate that these are likely to have emerged from a common membrane-associated dimerizing ancestor. This ancestral property appears to be critical for the role of GIMAPs as nucleotide-regulated scaffolds on intracellular membranes.


Subject(s)
GTP Phosphohydrolases/chemistry , Membrane Proteins/chemistry , Models, Molecular , Phylogeny , Protein Conformation , Protein Structure, Tertiary , Calorimetry , Chromatography, High Pressure Liquid , Crystallization , Dimerization , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Hydrolysis , Jurkat Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutagenesis , Nucleotides/metabolism , Ultracentrifugation
5.
J Neurosci ; 30(8): 2897-910, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181587

ABSTRACT

The coxsackievirus-adenovirus receptor (CAR) is a member of the Ig superfamily strongly expressed in the developing nervous system. Our histological investigations during development reveal an initial uniform distribution of CAR on all neural cells with a concentration on membranes that face the margins of the nervous system (e.g., the basal laminae and the ventricular side). At more advanced stages, CAR becomes downregulated and restricted to specific regions including areas rich in axonal and dendritic surfaces. To study the function of CAR on neural cells, we used the fiber knob of the adenovirus, extracellular CAR domains, blocking antibodies to CAR, as well as CAR-deficient neural cells. Blocking antibodies were found to inhibit neurite extension in retina organ and retinal explant cultures, whereas the application of the recombinant fiber knob of the adenovirus subtype Ad2 or extracellular CAR domains promoted neurite extension and adhesion to extracellular matrices. We observed a promiscuous interaction of CAR with extracellular matrix glycoproteins, which was deduced from analytical ultracentrifugation experiments, affinity chromatography, and adhesion assays. The membrane proximal Ig domain of CAR, termed D2, was found to bind to a fibronectin fragment, including the heparin-binding domain 2, which promotes neurite extension of wild type, but not of CAR-deficient neural cells. In contrast to heterophilic interactions, homophilic association of CAR involves both Ig domains, as was revealed by ultracentrifugation, chemical cross-linking, and adhesion studies. The results of these functional and binding studies are correlated to a U-shaped homodimer of the complete extracellular domains of CAR detected by x-ray crystallography.


Subject(s)
Cell Differentiation/genetics , Central Nervous System/embryology , Central Nervous System/metabolism , Gene Expression Regulation, Developmental/genetics , Neurogenesis/genetics , Neurons/metabolism , Receptors, Virus/metabolism , Animals , Antibodies, Blocking/pharmacology , CHO Cells , Cell Adhesion/genetics , Cells, Cultured , Central Nervous System/cytology , Chick Embryo , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Cricetinae , Cricetulus , Crystallography, X-Ray , Dimerization , Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Neurites/metabolism , Neurites/ultrastructure , Neurons/cytology , Organ Culture Techniques , Protein Structure, Tertiary/physiology , Receptors, Virus/chemistry , Receptors, Virus/genetics , Retina/cytology , Retina/embryology , Retina/metabolism
6.
Biochem Biophys Res Commun ; 405(3): 473-9, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21256114

ABSTRACT

Myomesin plays an important structural and functional role in the M-band of striated muscles. The C-terminal domain 13 of myomesin dimerises and forms antiparallel strands which cross-link neighboring Myosin filaments and titin in the M-line of the sarcomeres. These interactions stabilise the contractile apparatus during striated muscle contraction. Since myomesin is an important component of the M-band we screened the myomesin gene for genetic variants in patients with hypertrophic cardiomyopathy (HCM). We identified the missense mutation V1490I in domain 12 of myomesin in a family with inherited HCM. Analytical ultracentrifugation experiments, circular dichroism spectra, and surface plasmon resonance spectroscopy of myomesin fragments were carried out to investigate the effects of the mutation V1490I on structure and function of myomesin domains 11-13 and 12-13. Both the wild type and mutated myomesin domains My11-13 revealed similar secondary structures and formed stable dimers. Mutated myomesin domains My11-13 and My12-13 dimers revealed a reduced thermal stability and a significantly decreased dimerisation affinity, showing disturbed functional properties of V1490I mutated myomesin. However, monomeric myomesin domains My11-12, i.e. without dimerisation domain 13 showed no difference in thermal stability between wild type and V1490I mutated myomesin. In conclusion, the V1490I mutation associated with HCM lead to myomesin proteins with abnormal functional properties which affect dimerisation properties of myomesin domain 13. These effects may contribute to the pathogenesis of HCM.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Adult , Connectin , Female , Humans , Male , Middle Aged , Muscle Proteins/chemistry , Mutation, Missense , Pedigree , Protein Conformation , Protein Multimerization , Surface Plasmon Resonance
7.
Pflugers Arch ; 460(4): 719-30, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20607281

ABSTRACT

Ahnak1, a giant 700 kDa protein, has been implicated in Ca(2+) signalling in various cells. Previous work suggested that the interaction between ahnak1 and Cavbeta(2) subunit plays a role in L-type Ca(2+) current (I (CaL)) regulation. Here, we performed structure-function studies with the most C-terminal domain of ahnak1 (188 amino acids) containing a PxxP consensus motif (designated as 188-PSTP) using ventricular cardiomyocytes isolated from rats, wild-type (WT) mice and ahnak1-deficient mice. In vitro binding studies revealed that 188-PSTP conferred high-affinity binding to Cavbeta(2) (K (d) approximately 60 nM). Replacement of proline residues by alanines (188-ASTA) decreased Cavbeta(2) affinity about 20-fold. Both 188-PSTP and 188-ASTA were functional in ahnak1-expressing rat and mouse cardiomyocytes during whole-cell patch clamp. Upon intracellular application, they increased the net Ca(2+) influx by enhancing I (CaL) density and/or increasing I (CaL) inactivation time course without altering voltage dependency. Specifically, 188-ASTA, which failed to affect I (CaL) density, markedly slowed I (CaL) inactivation resulting in a 50-70% increase in transported Ca(2+) during a 0 mV depolarising pulse. Both ahnak1 fragments also slowed current inactivation with Ba(2+) as charge carrier. By contrast, neither 188-PSTP nor 188-ASTA affected any I (CaL) characteristics in ahnak1-deficient mouse cardiomyocytes. Our results indicate that the presence of endogenous ahnak1 is required for tuning the voltage-dependent component of I (CaL) inactivation by ahnak1 fragments. We suggest that ahnak1 modulates the accessibility of molecular determinants in Cavbeta(2) and/or scaffolds selectively different beta-subunit isoforms in the heart.


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Neoplasm Proteins/metabolism , Amino Acid Motifs , Animals , Blotting, Western , Calcium Signaling/physiology , Male , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neoplasm Proteins/chemistry , Patch-Clamp Techniques , Rats , Rats, Wistar
8.
Biochem Biophys Res Commun ; 396(4): 939-43, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20460111

ABSTRACT

The denuded IQ2 domain, i.e. myosin heavy chain not associated with regulatory light chains, exerts an inhibitory effect on myosin ATPase activity. In this study, we elaborated a structural explanation for this auto-inhibitory effect of IQ2 on myosin function. We employed analytical ultracentrifugation, circular dichroism, and surface plasmon resonance spectroscopy to investigate structural and functional properties of a myosin heavy chain (MYH) head-rod fragment aa664-915. MYH(664-915) was monomeric, adopted a closed shape, and bound essential myosin light chains (HIS-MLC-1) with low affinity to IQ1. Deletion of IQ2, however opened MYH(664-915). Four amino acids present in IQ2 could be identified to be responsible for this auto-inhibitory structural effect: alanine mutagenesis of I814, Q815, R819, and W827 stretched MYH(664-915) and increased 30-fold the binding affinity of HIS-MLC-1 to IQ1. In this study we show, that denuded IQ2 favours a closed conformation of myosin with a low HIS-MLC-1 binding affinity. The collapsed structure of myosin with denuded IQ2 could explain the auto-inhibitory effects of IQ2 on enzymatic activity of myosin.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Myosin Heavy Chains/chemistry , Myosin Type II/antagonists & inhibitors , Animals , Circular Dichroism , Myosin Heavy Chains/genetics , Myosin Light Chains/chemistry , Protein Structure, Tertiary/genetics , Rats , Sequence Deletion , Surface Plasmon Resonance , Ultracentrifugation
9.
Eur Biophys J ; 39(3): 449-55, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19266192

ABSTRACT

Sedimentation velocity experiments can be used to identify two or more independent non-interacting macromolecules, which differ in their size by only a few percent. The procedure requires the extrapolation of differential apparent sedimentation coefficient distributions obtained at different running time to t --> infinity and works because it eliminates or greatly reduces diffusion effects. Here, we present an improved time extrapolation function of sedimentation distribution profiles originally presented by Stafford (In: Harding, Rowe, Horton (eds.) Analytical ultracentrifugation in biochemistry and polymer science, 1992). We describe a computing procedure with the program LAMM: to analyze concentration profiles obtained by absorbance or interference optics that utilizes suitable smoothing methods for noisy data sets and present examples which include time invariant noises.


Subject(s)
Centrifugation/methods , Signal Processing, Computer-Assisted , Algorithms , Computers , Macromolecular Substances/chemistry , Molecular Weight , Software , Time Factors
10.
Structure ; 16(5): 809-17, 2008 May.
Article in English | MEDLINE | ID: mdl-18462685

ABSTRACT

Human saposins are essential proteins required for degradation of sphingolipids and lipid antigen presentation. Despite the conserved structural organization of saposins, their distinct modes of interaction with biological membranes are not fully understood. We describe two crystal structures of human saposin C in an "open" configuration with unusual domain swapped homodimers. This form of SapC dimer supports the "clip-on" model for SapC-induced vesicle fusion. In addition, we present the crystal structure of SapD in two crystal forms. They reveal the monomer-monomer interface for the SapD dimer, which was confirmed in solution by analytical ultracentrifugation. The crystal structure of SapD suggests that side chains of Lys10 and Arg17 are involved in initial association with the preferred anionic biological membranes by forming salt bridges with sulfate or phosphate lipid headgroups.


Subject(s)
Saposins/chemistry , Amino Acid Sequence , Cell Membrane/metabolism , Cloning, Molecular , Conserved Sequence , Crystallography, X-Ray , Cysteine/chemistry , Dimerization , Humans , Hydrophobic and Hydrophilic Interactions , Lipid Metabolism , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutagenesis , Pichia/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Saposins/genetics , Saposins/isolation & purification , Sequence Homology, Amino Acid
11.
FEBS J ; 275(18): 4627-40, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18699778

ABSTRACT

Iba2 is a homolog of ionized calcium-binding adapter molecule 1 (Iba1), a 17-kDa protein that binds and cross-links filamentous actin (F-actin) and localizes to membrane ruffles and phagocytic cups. Here, we present the crystal structure of human Iba2 and its homodimerization properties, F-actin cross-linking activity, cellular localization and recruitment upon bacterial invasion in comparison with Iba1. The Iba2 structure comprises two central EF-hand motifs lacking bound Ca2+. Iba2 crystallized as a homodimer stabilized by a disulfide bridge and zinc ions. Analytical ultracentrifugation revealed a different mode of dimerization under reducing conditions that was independent of Ca2+. Furthermore, no binding of Ca2+ up to 0.1 mM was detected by equilibrium dialysis. Correspondingly, Iba EF-hand motifs lack residues essential for strong Ca2+ coordination. Sedimentation experiments and microscopy detected pronounced, indistinguishable F-actin binding and cross-linking activity of Iba1 and Iba2 with induction of F-actin bundles. Fluorescent Iba fusion proteins were expressed in HeLa cells and co-localized with F-actin. Iba1 was recruited into cellular projections to a larger extent than Iba2. Additionally, we studied Iba recruitment in a Shigella invasion model that induces cytoskeletal rearrangements. Both proteins were recruited into the bacterial invasion zone and Iba1 was again concentrated slightly higher in the cellular extensions.


Subject(s)
Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/chemistry , Microfilament Proteins/chemistry , Actins/metabolism , Amino Acid Sequence , Calcium/metabolism , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/metabolism , Crystallography, X-Ray , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Dimerization , HeLa Cells , Humans , Microfilament Proteins/analysis , Microfilament Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Shigella/pathogenicity
12.
FASEB J ; 20(7): 865-73, 2006 May.
Article in English | MEDLINE | ID: mdl-16675844

ABSTRACT

In this study we investigated whether the expression of N-terminal myosin light chain-1 (MLC-1) peptides could improve the intrinsic contractility of the whole heart. We generated transgenic rats (TGR) that overexpressed minigenes encoding the N-terminal 15 amino acids of human atrial MLC-1 (TGR/hALC-1/1-15, lines 7475 and 3966) or human ventricular MLC-1 (TGR/hVLC-1/1-15, lines 6113 and 6114) isoforms in cardiomyocytes. Synthetic N-terminal peptides revealed specific actin binding, with a significantly (P<0.01) lower dissociation constant (K(D)) for the hVLC-1/1-15-actin complex compared with the K(D) value of the hALC-1/1-15-actin complex. Using synthetic hVLC-1/1-15 as a TAT fusion peptide labeled with the fluorochrome TAMRA, we observed specific accumulation of the N-terminal MLC-1 peptide at the sarcomere predominantly within the actin-containing I-band, but also within the actin-myosin overlap zone (A-band) in intact adult cardiomyocytes. For the first time we show that the expression of N-terminal human MLC-1 peptides in TGR (range: 3-6 muM) correlated positively with significant (P<0.001) improvements of the intrinsic contractile state of the isolated perfused heart (Langendorff mode): systolic force generation, as well as the rates of both force generation and relaxation, rose in TGR lines that expressed the transgenic human MLC-1 peptide, but not in a TGR line with undetectable transgene expression levels. The positive inotropic effect of MLC-1 peptides occurred in the absence of a hypertrophic response. Thus, expression of N-terminal domains of MLC-1 represent a valuable tool for the treatment of the failing heart.


Subject(s)
Heart/physiology , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Myosin Light Chains/chemistry , Myosin Light Chains/metabolism , Actins , Animals , Animals, Genetically Modified , Gene Expression Regulation , Genetic Therapy , Humans , Male , Myocytes, Cardiac/metabolism , Myosin Light Chains/genetics , Protein Binding , Rats , Rats, Inbred WKY
13.
Mol Cell Biol ; 23(6): 2029-41, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12612076

ABSTRACT

The IkappaB kinase (IKK) complex mediates activation of transcription factor NF-kappaB by phosphorylation of IkappaB proteins. Its catalytic subunits, IKKalpha and IKKbeta, require association with the regulatory IKKgamma (NEMO) component to gain full basal and inducible kinase activity. However, the oligomeric composition of the IKK complex and its regulation by IKKgamma are poorly understood. We show here that IKKgamma predominantly forms tetramers and interacts with IKKalpha or IKKbeta in this state. We propose that tetramerization is accomplished by a prerequisite dimerization through a C-terminal coiled-coil minimal oligomerization domain (MOD). This is followed by dimerization of the dimers with their N-terminal sequences. Tetrameric IKKgamma sequesters four kinase molecules, yielding a gamma(4)(alpha/beta)(4) stoichiometry. Deletion of the MOD leads to loss of tetramerization and of phosphorylation of IKKbeta and IKKgamma, although the kinase can still interact with the resultant IKKgamma monomers and dimers. Likewise, MOD-mediated IKKgamma tetramerization is required to enhance IKKbeta kinase activity when overexpressed in 293 cells and to reconstitute a lipopolysaccharide-responsive IKK complex in pre-B cells. These data thus suggest that IKKgamma tetramerization enforces a spatial positioning of two kinase dimers to facilitate transautophosphorylation and activation.


Subject(s)
NF-kappa B/metabolism , Protein Serine-Threonine Kinases/chemistry , Biopolymers , Cell Line , HeLa Cells , Humans , I-kappa B Kinase , I-kappa B Proteins/metabolism , Kidney , Macromolecular Substances , Models, Molecular , NF-KappaB Inhibitor alpha , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/physiology , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Sequence Deletion , Structure-Activity Relationship , Zinc Fingers/physiology
14.
Biophys Chem ; 125(1): 159-65, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16916573

ABSTRACT

The mitochondrial steroid hydroxylase system of vertebrates utilizes adrenodoxin (Adx), a small iron-sulfur cluster protein of about 14 kDa as an electron carrier between a reductase and cytochrome P450. Although the crystal structure of this protein has been elucidated, the solution structure of Adx was discussed contrary in the literature [I.A. Pikuleva, K. Tesh, M.R. Waterman, Y. Kim, The tertiary structure of full-length bovine adrenodoxin suggests functional dimers, Arch. Biochem. Biophys. 373 (2000) 44-55; D. Beilke, R. Weiss, F. Löhr, P. Pristovsek, F. Hannemann, R. Bernhardt, H. Rüterjans, A new electron mechanism in mitochondrial steroid hydroxylase systems based on structural changes upon the reduction of adrenodoxin, Biochemistry 41 (2002) 7969-7978]. Therefore, it was necessary to study the self-association of this protein by using analytical ultracentrifugation over a larger concentration range. As could be demonstrated in sedimentation velocity experiments, as well as sedimentation equilibrium runs with explicit consideration of thermodynamic non-ideality, the full-length protein (residues 1-128) in the oxidized state resulted in a monomer-dimer equilibrium (K(a) approximately 3 x 10(2) M(-1)). For truncated Adx (1-108), as well as the reduced Adx, the association behavior was strongly reduced. The consequences of this behavior are discussed with respect to the physiological meaning for the Adx system.


Subject(s)
Adrenodoxin/chemistry , Protein Structure, Quaternary , Oxidation-Reduction , Thermodynamics , Ultracentrifugation/methods
15.
FASEB J ; 19(14): 1969-77, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16319140

ABSTRACT

Defective L-type Ca2+ channel (I(CaL)) regulation is one major cause for contractile dysfunction in the heart. The I(CaL) is enhanced by sympathetic nervous stimulation: via the activation of beta-adrenergic receptors, PKA phosphorylates the alpha1C(Ca(V)1.2)- and beta2-channel subunits and ahnak, an associated 5643-amino acid (aa) protein. In this study, we examined the role of a naturally occurring, genetic variant Ile5236Thr-ahnak on I(CaL). Binding experiments with ahnak fragments (wild-type, Ile5236Thr mutated) and patch clamp recordings revealed that Ile5236Thr-ahnak critically affected both beta2 subunit interaction and I(CaL) regulation. Binding affinity between ahnak-C1 (aa 4646-5288) and beta2 subunit decreased by approximately 50% after PKA phosphorylation or in the presence of Ile5236Thr-ahnak peptide. On native cardiomyocytes, intracellular application of this mutated ahnak peptide mimicked the PKA-effects on I(CaL) increasing the amplitude by approximately 60% and slowing its inactivation together with a leftward shift of its voltage dependency. Both mutated Ile5236Thr-peptide and Ile5236Thr-fragment (aa 5215-5288) prevented specifically the further up-regulation of I(CaL) by isoprenaline. Hence, we suggest the ahnak-C1 domain serves as physiological brake on I(CaL). Relief from this inhibition is proposed as common pathway used by sympathetic signaling and Ile5236Thr-ahnak fragments to increase I(CaL). This genetic ahnak variant might cause individual differences in I(CaL) regulation upon physiological challenges or therapeutic interventions.


Subject(s)
Calcium Channels, L-Type/physiology , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Receptors, Adrenergic, beta/metabolism , Animals , Calcium/chemistry , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Female , Gene Expression Regulation , Genetic Variation , Glutathione Transferase/metabolism , Heterozygote , Humans , Isoleucine/chemistry , Male , Membrane Proteins/genetics , Models, Biological , Models, Genetic , Mutation , Mutation, Missense , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Neoplasm Proteins/genetics , Peptides/chemistry , Phosphorylation , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational , Protein Binding , Protein Structure, Tertiary , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Software , Temperature , Threonine/chemistry , Ultracentrifugation , Up-Regulation
16.
Nucleic Acids Res ; 30(9): 2052-60, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11972345

ABSTRACT

Plasmid pIP501 encoded transcriptional repressor CopR is one of the two regulators of plasmid copy number. Previous data suggested that CopR is a HTH protein belonging to a family of 578 HTH proteins (termed HTH 3-family). Only a very limited number of proteins in this family, among them lambda c1 repressor, 434 c1 repressor and P22 c2 repressor, have been characterized in detail so far. Previously, a CopR structural model was built based on structural homologies to the 434 c1 and P22 c2 repressor and used to identify amino acids involved in DNA binding and dimerization. Site-directed mutagenesis in combination with electrophoretic mobility shift assay (EMSA), dimerization studies and circular dichroism (CD) measurements verified the model predictions. In this study we used hydroxyl radical footprinting and fluorescence resonance energy transfer (FRET) measurements to obtain detailed information about the structure of the DNA in the CopR-DNA complex. Our results show that the DNA is bent gently around the protein, comparable to the bending angle of 20-25 degrees observed in the 434 c1 repressor-DNA complex and the lambda c1 repressor-DNA complex. The shape of CopR dimers as determined by sedimentation velocity experiments is extended and accounts for the relatively large area of protection observed with hydroxyl radical footprinting.


Subject(s)
Bacterial Proteins , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Trans-Activators/metabolism , Base Sequence , Binding Sites , Binding, Competitive , DNA Footprinting , Dimerization , Fluorescent Dyes/chemistry , Hydroxyl Radical/chemistry , Nucleic Acid Conformation , Repressor Proteins/metabolism , Spectrometry, Fluorescence , Ultracentrifugation , Viral Proteins , Viral Regulatory and Accessory Proteins
17.
BMC Struct Biol ; 5: 21, 2005 Dec 14.
Article in English | MEDLINE | ID: mdl-16354304

ABSTRACT

BACKGROUND: Human Aortic Preferentially Expressed Protein-1 (APEG-1) is a novel specific smooth muscle differentiation marker thought to play a role in the growth and differentiation of arterial smooth muscle cells (SMCs). RESULTS: Good quality crystals that were suitable for X-ray crystallographic studies were obtained following the truncation of the 14 N-terminal amino acids of APEG-1, a region predicted to be disordered. The truncated protein (termed DeltaAPEG-1) consists of a single immunoglobulin (Ig) like domain which includes an Arg-Gly-Asp (RGD) adhesion recognition motif. The RGD motif is crucial for the interaction of extracellular proteins and plays a role in cell adhesion. The X-ray structure of DeltaAPEG-1 was determined and was refined to sub-atomic resolution (0.96 A). This is the best resolution for an immunoglobulin domain structure so far. The structure adopts a Greek-key beta-sandwich fold and belongs to the I (intermediate) set of the immunoglobulin superfamily. The residues lying between the beta-sheets form a hydrophobic core. The RGD motif folds into a 310 helix that is involved in the formation of a homodimer in the crystal which is mainly stabilized by salt bridges. Analytical ultracentrifugation studies revealed a moderate dissociation constant of 20 microM at physiological ionic strength, suggesting that APEG-1 dimerisation is only transient in the cell. The binding constant is strongly dependent on ionic strength. CONCLUSION: Our data suggests that the RGD motif might play a role not only in the adhesion of extracellular proteins but also in intracellular protein-protein interactions. However, it remains to be established whether the rather weak dimerisation of APEG-1 involving this motif is physiologically relevant.


Subject(s)
Muscle Proteins/physiology , Amino Acid Motifs , Amino Acid Sequence , Arteries/metabolism , Biophysics/methods , Cell Adhesion , Cloning, Molecular , Crystallography, X-Ray , Databases, Protein , Dimerization , Escherichia coli/metabolism , Humans , Immunoglobulins/chemistry , Kinetics , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Muscle Proteins/chemistry , Myocytes, Smooth Muscle/metabolism , Oligopeptides/chemistry , Protein Binding , Protein Conformation , Protein Engineering , Protein Serine-Threonine Kinases , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Ultracentrifugation
18.
FASEB J ; 16(10): 1205-16, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12153988

ABSTRACT

Ahnak is a ubiquitously expressed giant protein of 5643 amino acids implicated in cell differentiation and signal transduction. In a recent study, we demonstrated the association of ahnak with the regulatory beta2 subunit of the cardiac L-type Ca2+ channel. Here we identify the most carboxyl-terminal ahnak region (aa 5262-5643) to interact with recombinant beta2a as well as with beta2 and beta1a isoforms of native muscle Ca2+ channels using a panel of GST fusion proteins. Equilibrium sedimentation analysis revealed Kd values of 55 +/- 11 nM and 328 +/- 24 nM for carboxyl-terminal (aa 195-606) and amino-terminal (aa 1-200) truncates of the beta2a subunit, respectively. The same carboxyl-terminal ahnak region (aa 5262-5643) bound to G-actin and cosedimented with F-actin. Confocal microscopy of human left ventricular tissue localized the carboxyl-terminal ahnak portion to the sarcolemma including the T-tubular system and the intercalated disks of cardiomyocytes. These results suggest that ahnak provides a structural basis for the subsarcolemmal cytoarchitecture and confers the regulatory role of the actin-based cytoskeleton to the L-type Ca2+ channel.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium Channels, L-Type/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Myocardium/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels, L-Type/genetics , Humans , Membrane Proteins/genetics , Models, Biological , Muscle, Skeletal , Myocardium/chemistry , Neoplasm Proteins/genetics , Protein Subunits , Recombinant Fusion Proteins/metabolism , Sarcolemma/chemistry , Sequence Deletion , Swine
19.
Z Naturforsch C J Biosci ; 60(11-12): 927-31, 2005.
Article in English | MEDLINE | ID: mdl-16402555

ABSTRACT

The influence of phosphate, ionic strength, temperature and enzyme concentration on the oligomeric structure of calf spleen purine nucleoside phosphorylase (PNP) in solution was studied by analytical ultracentrifugation methods. Sedimentation equilibrium analysis used to directly determine the enzyme molecular mass revealed a trimeric molecule with Mr = (90.6 +/- 2.1) kDa, regardless the conditions investigated: protein concentration in the range 0.02-1.0 mg/ml, presence of up to 100 mM phosphate and up to 200 mM NaCl, temperature in the range 4-25 degrees C. The sedimentation coefficient (6.04 +/- 0.02) S, together with the diffusion coefficient (6.15 +/- 0.11) 10(-7) cm2/s, both values obtained from the classic sedimentation velocity method at 1.0 mg/ml PNP concentration in 20 mM Hepes, pH 7.0, yielded a molecular mass of (90.2 +/- 1.6) kDa as expected for the trimeric enzyme molecule. Moreover, as shown by active enzyme sedimentation, calf spleen PNP remained trimeric even at low protein concentrations (1 microg/ml). Hence in solution, similar like in the crystalline state, calf spleen PNP is a homotrimer and previous suggestions for dissociation of this enzyme into more active monomers, upon dilution of the enzyme or addition of phosphate, are incorrect.


Subject(s)
Purine-Nucleoside Phosphorylase/chemistry , Animals , Cattle , Kinetics , Macromolecular Substances , Mammals , Molecular Weight , Purine-Nucleoside Phosphorylase/isolation & purification , Purine-Nucleoside Phosphorylase/metabolism , Spleen/enzymology
20.
Biophys Chem ; 95(1): 59-68, 2002 Jan 23.
Article in English | MEDLINE | ID: mdl-11880173

ABSTRACT

Sedimentation velocity is one of the best-suited physical methods for determining the size and shape of macromolecular substances or their complexes in the range from 1 to several thousand kDa. The moving boundary in sedimentation velocity runs can be described by the Lamm differential equation. Fitting of suitable model functions or solutions of the Lamm equation to the moving boundary is used to obtain directly sedimentation and diffusion coefficients, thus allowing quick determination of size, shape and other parameters of macromolecules. Here we present a new approximate whole boundary solution of the Lamm equation that simultaneously allows the specification of sedimentation and diffusion coefficients with deviations smaller than 1% from the expected values.


Subject(s)
Ultracentrifugation/statistics & numerical data , Algorithms , Diffusion , Software , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL