Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 24(2): 371-82, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25180020

ABSTRACT

The deposition of amyloid-beta (Aß) aggregates in the brain is a major pathological hallmark of Alzheimer's disease (AD). Aß is generated from the cleavage of C-terminal fragments of the amyloid precursor protein (APP-CTFs) by γ-secretase, an intramembrane-cleaving protease with multiple substrates, including the Notch receptors. Endogenous modulation of γ-secretase is pointed to be implicated in the sporadic, age-dependent form of AD. Moreover, specifically modulating Aß production has become a priority for the safe treatment of AD because the inhibition of γ-secretase results in adverse effects that are related to impaired Notch cleavage. Here, we report the identification of the adipocyte differentiation protein APMAP as a novel endogenous suppressor of Aß generation. We found that APMAP interacts physically with γ-secretase and its substrate APP. In cells, the partial depletion of APMAP drastically increased the levels of APP-CTFs, as well as uniquely affecting their stability, with the consequence being increased secretion of Aß. In wild-type and APP/ presenilin 1 transgenic mice, partial adeno-associated virus-mediated APMAP knockdown in the hippocampus increased Aß production by ∼20 and ∼55%, respectively. Together, our data demonstrate that APMAP is a negative regulator of Aß production through its interaction with APP and γ-secretase. All observed APMAP phenotypes can be explained by an impaired degradation of APP-CTFs, likely caused by an altered substrate transport capacity to the lysosomal/autophagic system.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Brain/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Animals , Brain/enzymology , Cell Line , Female , Humans , Male , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Protein Binding
2.
J Neurochem ; 133(3): 409-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25458374

ABSTRACT

An important pathological hallmark of Alzheimer's disease (AD) is the deposition of amyloid-beta (Aß) peptides in the brain parenchyma, leading to neuronal death and impaired learning and memory. The protease γ-secretase is responsible for the intramembrane proteolysis of the amyloid-ß precursor protein (APP), which leads to the production of the toxic Aß peptides. Thus, an attractive therapeutic strategy to treat AD is the modulation of the γ-secretase activity, to reduce Aß42 production. Because phosphorylation of proteins is a post-translational modification known to modulate the activity of many different enzymes, we used electrospray (LC-MS/MS) mass spectrometry to identify new phosphosites on highly purified human γ-secretase. We identified 11 new single or double phosphosites in two well-defined domains of Presenilin-1 (PS1), the catalytic subunit of the γ-secretase complex. Next, mutagenesis and biochemical approaches were used to investigate the role of each phosphosite in the maturation and activity of γ-secretase. Together, our results suggest that the newly identified phosphorylation sites in PS1 do not modulate γ-secretase activity and the production of the Alzheimer's Aß peptides. Individual PS1 phosphosites shall probably not be considered therapeutic targets for reducing cerebral Aß plaque formation in AD. In this study, we identified 11 new phosphosites in Presenilin-1 (PS1), the catalytic subunit of the Alzheimer's γ-secretase complex. By combining a mutagenesis approach with cell-based and cell-free γ-secretase assays, we demonstrate that the new phosphosites do not modulate the maturation and activity of γ-secretase. Individual PS1 phosphosites shall thus not be considered therapeutic targets for reducing cerebral Aß plaque formation in Alzheimer's Disease. Aß, amyloid beta.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Presenilin-1/genetics , Presenilin-1/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Cell Line, Tumor , Enzyme Activation/physiology , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation/physiology
3.
Breast Cancer Res Treat ; 148(2): 455-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25248409

ABSTRACT

The goal of targeted cancer therapies is to specifically block oncogenic signalling, thus maximising efficacy, while reducing side-effects to patients. The gamma-secretase (GS) complex is an attractive therapeutic target in haematological malignancies and solid tumours with major pharmaceutical activity to identify optimal inhibitors. Within GS, nicastrin (NCSTN) offers an opportunity for therapeutic intervention using blocking monoclonal antibodies (mAbs). Here we explore the role of anti-nicastrin monoclonal antibodies, which we have developed as specific, multi-faceted inhibitors of proliferation and invasive traits of triple-negative breast cancer cells. We use 3D in vitro proliferation and invasion assays as well as an orthotopic and tail vail injection triple-negative breast cancer in vivo xenograft model systems. RNAScope assessed nicastrin in patient samples. Anti-NCSTN mAb clone-2H6 demonstrated a superior anti-tumour efficacy than clone-10C11 and the RO4929097 small molecule GS inhibitor, acting by inhibiting GS enzymatic activity and Notch signalling in vitro and in vivo. Confirming clinical relevance of nicastrin as a target, we report evidence of increased NCSTN mRNA levels by RNA in situ hybridization (RNAScope) in a large cohort of oestrogen receptor negative breast cancers, conferring independent prognostic significance for disease-free survival, in multivariate analysis. We demonstrate here that targeting NCSTN using specific mAbs may represent a novel mode of treatment for invasive triple-negative breast cancer, for which there are few targeted therapeutic options. Furthermore, we propose that measuring NCSTN in patient samples using RNAScope technology may serve as companion diagnostic for anti-NCSTN therapy in the clinic.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Membrane Glycoproteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Nat Commun ; 4: 2246, 2013.
Article in English | MEDLINE | ID: mdl-23907250

ABSTRACT

Pathological amino-acid substitutions in the amyloid precursor protein (APP) and chemical γ-secretase modulators affect the processing of APP by the γ-secretase complex and the production of the amyloid-beta peptide Aß42, the accumulation of which is considered causative of Alzheimer's disease. Here we demonstrate that mutations in the transmembrane domain of APP causing aggressive early-onset familial Alzheimer's disease affect both γ- and ε-cleavage sites, by raising the Aß42/40 ratio and inhibiting the production of AICD50-99, one of the two physiological APP intracellular domains (ICDs). This is in sharp contrast to γ-secretase modulators, which shift Aß42 production towards the shorter Aß38, but unequivocally spare the ε-site and APP- and Notch-ICDs production. Molecular simulations suggest that familial Alzheimer's disease mutations modulate the flexibility of the APP transmembrane domain and the presentation of its γ-site, modifying at the same time, the solvation of the ε-site.


Subject(s)
Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Mutation/genetics , Amino Acid Sequence , Amyloid beta-Protein Precursor/metabolism , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Piperidines/chemistry , Piperidines/pharmacology , Protein Structure, Tertiary , Proteolysis/drug effects , Receptors, Notch/metabolism
5.
Biotechnol Bioeng ; 110(7): 1995-2005, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23359429

ABSTRACT

Inefficient production of membrane-embedded multi-protein complexes by conventional methods has largely prevented the generation of high-resolution structural information and the performance of high-throughput drug discovery screens for this class of proteins. Not exempt from this rule is γ-secretase, an intramembrane-cleaving protease complex regulating a multitude of signaling pathways and biological processes by influencing gene transcription. γ-Secretase is also implicated in the pathogenesis of Alzheimer's disease and several types of cancer. As an additional challenge, the reconstitution of the protease complex in its active form requires an intricate assembly and maturation process, including a highly regulated endoproteolytic processing of its catalytic component. In this article we report the application of a transposon-mediated multigene stable integration technology to produce active γ-secretase in mammalian cells in amounts adequate for crystallization studies and drug screening. Our strategy is expected to help elucidate the molecular mechanisms of intramembrane proteolysis. It is further expected to be widely used for the production of other multi-protein complexes for applications in structural biology and drug development.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Animals , Biotechnology/methods , CHO Cells , Cricetulus , DNA Transposable Elements , Genetic Vectors , Metabolic Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic
6.
J Biol Chem ; 288(4): 2521-31, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23209290

ABSTRACT

γ-Secretase is a large enzyme complex comprising presenilin, nicastrin, presenilin enhancer 2, and anterior pharynx-defective 1 that mediates the intramembrane proteolysis of a large number of proteins including amyloid precursor protein and Notch. Recently, a novel γ-secretase activating protein (GSAP) was identified that interacts with γ-secretase and the C-terminal fragment of amyloid precursor protein to selectively increase amyloid-ß production. In this study we have further characterized the role of endogenous and exogenous GSAP in the regulation of γ-secretase activity and amyloid-ß production in vitro. Knockdown of GSAP expression in N2a cells decreased amyloid-ß levels. In contrast, overexpression of GSAP in HEK cells expressing amyloid precursor protein or in N2a cells had no overt effect on amyloid-ß generation. Likewise, purified recombinant GSAP had no effect on amyloid-ß generation in two distinct in vitro γ-secretase assays. In subsequent cellular studies with imatinib, a kinase inhibitor that reportedly prevents the interaction of GSAP with the C-terminal fragment of amyloid precursor protein, a concentration-dependent decrease in amyloid-ß levels was observed. However, no interaction between GSAP and the C-terminal fragment of amyloid precursor protein was evident in co-immunoprecipitation studies. In addition, subchronic administration of imatinib to rats had no effect on brain amyloid-ß levels. In summary, these findings suggest the roles of GSAP and imatinib in the regulation of γ-secretase activity and amyloid-ß generation are uncertain.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Gene Expression Regulation , Piperazines/pharmacology , Proteins/chemistry , Pyrimidines/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Benzamides , Brain/metabolism , Cell Line, Tumor , Humans , Imatinib Mesylate , Male , Mice , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism
7.
Biochemistry ; 51(44): 8779-90, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23066899

ABSTRACT

A detailed understanding of γ-secretase structure is crucially needed to elucidate its unique properties of intramembrane protein cleavage and to design therapeutic compounds for the safe treatment of Alzheimer's disease. γ-Secretase is an enzyme complex composed of four membrane proteins, and the scarcity of its supply associated with the challenges of crystallizing membrane proteins is a major hurdle for the determination of its high-resolution structure. This study addresses some of these issues, first by adapting CHO cells overexpressing γ-secretase to growth in suspension, thus yielding multiliter cultures and milligram quantities of highly purified, active γ-secretase. Next, the amounts of γ-secretase were sufficient for immunization of mice and allowed generation of Nicastrin- and Aph-1-specific monoclonal antibodies, from which Fab fragments were proteolytically prepared and subsequently purified. The amounts of γ-secretase produced are compatible with robot-assisted crystallogenesis using nanoliter technologies. In addition, our Fab fragments bind exposed regions of native γ-secretase in a dose-dependent manner without interfering with its catalytic properties and can therefore be used as specific tools to facilitate crystal formation.


Subject(s)
Amyloid Precursor Protein Secretases/chemistry , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/immunology , Amyloid Precursor Protein Secretases/isolation & purification , Animals , Antibodies, Monoclonal , CHO Cells , Cholic Acids , Cricetinae , Cricetulus , Crystallization , Humans , Immunoglobulin Fab Fragments , Mice , Presenilin-1
8.
FASEB J ; 25(7): 2287-95, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21415381

ABSTRACT

Prenatal exposure to mercury causes neurodevelopmental disorders and neurological pathologies in infants, such as microcephaly and mental retardation. Despite critical importance, the molecular interactions leading to mercury toxicity are yet to be elucidated. We first used a cell-free assay to investigate mercury effects on purified γ-secretase activity. Next, we treated adult Drosophila melanogaster with mercury and collected control and mercury-treated embryos, which were subjected to mild hypotonic protein extraction, or immunostained to reveal nervous system morphology. Embryos left to develop into adults were examined for wing phenotypes. Relative to control metals, we found that mercury strongly inhibits in vitro γ-secretase processing of both amyloid-ß precursor protein (APP) and Notch. Mercury inhibited APP and Notch cleavage in a dose-dependent manner, with IC(50) values of 50-125 nM, and is therefore comparable in potency to benchmark γ-secretase inhibitors. Immunoblot analysis of embryonic protein extracts showed that mercury inhibits Notch cleavage by γ-secretase in vivo. This is accompanied by severe neurodevelopmental abnormalities in embryos and adult wing-notching phenotypes. Our findings provide first evidence that mercury is a direct and potent γ-secretase inhibitor and suggest that inhibition of γ-secretase and disruption of the Notch developmental pathway potentially contribute to mercury-induced toxicity in the nervous system.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Mercury/toxicity , Receptors, Notch/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Animals , Blotting, Western , Dipeptides/toxicity , Dose-Response Relationship, Drug , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Immunohistochemistry , Male , Methylmercury Compounds/toxicity , Nervous System Diseases/chemically induced , Nervous System Diseases/embryology , Nervous System Diseases/metabolism , Wings, Animal/abnormalities , Wings, Animal/drug effects , Wings, Animal/metabolism
9.
Proc Natl Acad Sci U S A ; 104(44): 17394-9, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17954913

ABSTRACT

Acid beta-glucosidase (GCase) is a soluble lysosomal enzyme responsible for the hydrolysis of glucose from glucosylceramide and requires activation by the small nonenzymatic protein saposin C (sapC) to gain access to the membrane-embedded glycosphingolipid substrate. We have used in situ atomic force microscopy (AFM) with simultaneous confocal and epifluorescence microscopies to investigate the interactions of GCase and sapC with lipid bilayers. GCase binds to sites on membranes transformed by sapC, and enzyme activity occurs at loci containing both GCase and sapC. Using FRET, we establish the presence of GCase/sapC and GCase/product contacts in the bilayer. These data support a mechanism in which sapC locally alters regions of bilayer for subsequent attack by the enzyme in stably bound protein complexes.


Subject(s)
Lipid Bilayers/metabolism , Saposins/pharmacology , beta-Glucosidase/metabolism , Enzyme Activation/drug effects , Microscopy, Atomic Force , Protein Binding , Saposins/genetics , Saposins/metabolism
10.
J Mol Biol ; 362(5): 943-53, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-16949605

ABSTRACT

Saposins A, B, C and D are soluble, non-enzymatic proteins that interact with lysosomal membranes to activate the breakdown and transfer of glycosphingolipids. The mechanisms of hydrolase activation and lipid transfer by saposins remain unknown. We have used in situ atomic force microscopy (AFM) with simultaneous confocal fluorescence microscopy to investigate the interactions of saposins with lipid membranes. AFM images of the effect of saposins A, B and C on supported lipid bilayers showed a time and concentration-dependent nucleated spread of membrane transformation. Saposin B produced deep gaps that ultimately filled with granular material, while saposins A and C lead to localized areas of membrane that were reduced in height by approximately 1.5 nm. Fluorescence-labeled saposin C co-localized with the transformed areas of the bilayer, indicating stable binding to the membrane. Fluorescence resonance energy transfer confirmed a direct interaction between saposin C and lipid. Under certain conditions of membrane lipid composition and saposin concentration, extensive bilayer lipid removal was observed. We propose a multi-step mechanism that integrates the structural features and amphipathic properties of the saposin proteins.


Subject(s)
Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Saposins/chemistry , Saposins/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Maleimides , Microscopy, Atomic Force , Microscopy, Confocal , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Rhodamines , Saposins/genetics , Saposins/ultrastructure
11.
Protein Sci ; 15(8): 1849-57, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16823039

ABSTRACT

Saposins A and C are sphingolipid activator proteins required for the lysosomal breakdown of galactosylceramide and glucosylceramide, respectively. The saposins interact with lipids, leading to an enhanced accessibility of the lipid headgroups to their cognate hydrolases. We have determined the crystal structures of human saposins A and C to 2.0 Angstroms and 2.4 Angstroms, respectively, and both reveal the compact, monomeric saposin fold. We confirmed that these two proteins were monomeric in solution at pH 7.0 by analytical centrifugation. However, at pH 4.8, in the presence of the detergent C(8)E(5), saposin A assembled into dimers, while saposin C formed trimers. Saposin B was dimeric under all conditions tested. The self-association of the saposins is likely to be relevant to how these small proteins interact with lipids, membranes, and hydrolase enzymes.


Subject(s)
Saposins/chemistry , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Detergents , Dimerization , Ethers , Humans , Models, Molecular , Molecular Sequence Data , Polyethylene Glycols , Protein Structure, Quaternary , Sequence Alignment , Ultracentrifugation
12.
J Struct Biol ; 154(1): 42-58, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16459101

ABSTRACT

We report here on an in situ atomic force microscopy study of the interaction of indolicidin, a tryptophan-rich antimicrobial peptide, with phase-segregated zwitterionic DOPC/DSPC supported planar bilayers. By varying the peptide concentration and bilayer composition through the inclusion of anionic lipids (DOPG or DSPG), we found that indolicidin interacts with these model membranes in one of two concentration-dependent manners. At low peptide concentrations, indolicidin forms an amorphous layer on the fluid domains when these domains contain anionic lipids. At high peptide concentrations, indolicidin appears to initiate a lowering of the gel-phase domains independent of the presence of an anionic lipid. Similar studies performed using membrane-raft mimetic bilayers comprising 30mol% cholesterol/1:1 DOPC/egg sphingomyelin revealed that indolicidin does not form a carpet-like layer on the zwitterionic DOPC domains at low peptide concentrations and does not induce membrane lowering of the liquid-ordered sphingomyelin/cholesterol-rich domains at high peptide concentration. Simultaneous AFM-confocal microscopy imaging did however reveal that indolicidin preferentially inserts into the fluid-phase DOPC domains. These data suggest that the indolicidin-membrane association is influenced greatly by specific electrostatic interactions, lipid fluidity, and peptide concentration. These insights provide a glimpse into the mechanism of the membrane selectivity of antibacterial peptides and suggest a powerful correlated approach for characterizing peptide-membrane interactions.


Subject(s)
Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Lipid Bilayers/chemistry , Cell Membrane/ultrastructure , Cholesterol/chemistry , Gels/chemistry , Liposomes/chemistry , Membrane Fluidity , Membrane Microdomains/chemistry , Membrane Microdomains/ultrastructure , Microscopy, Atomic Force , Microscopy, Confocal , Phosphatidylglycerols/chemistry , Solubility , Sphingomyelins/chemistry
13.
J Lipid Res ; 46(10): 2254-64, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16061947

ABSTRACT

A mass spectrometric method is described for monitoring cerebrosides in the presence of excess concentrations of alkali metal salts. This method has been adapted for use in the assay of arylsulfatase A (ASA) and the cerebroside sulfate activator protein (CSAct or saposin B). Detection of the neutral glycosphingolipid cerebroside product was achieved via enhancement of ionization efficiency in the presence of lithium ions. Assay samples were extracted into the chloroform phase as for the existing assays, dried, and diluted in methanol-chloroform-containing lithium chloride. Samples were analyzed by electrospray ionization mass spectrometry with a triple quadrupole mass spectrometer in the multiple reaction monitoring tandem mass spectrometric mode. The assay has been used to demonstrate several previously unknown or ambiguous aspects of the coupled ASA/CSAct reaction, including an absolute in vitro preference for CSAct over the other saposins (A, C, and D) and a preference for the non-hydroxylated species of the sulfatide substrate over the corresponding hydroxylated species. The modified assay for the coupled ASA/CSAct reaction could find applicability in settings in which the assay could not be performed previously because of the need for radiolabeled substrate, which is now not required.


Subject(s)
Cerebroside-Sulfatase/analysis , Saposins/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Cattle , Lithium/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Nature ; 420(6916): 696-700, 2002 Dec 12.
Article in English | MEDLINE | ID: mdl-12442173

ABSTRACT

In a variety of cells, the Ca2+ signalling process is mediated by the endoplasmic-reticulum-membrane-associated Ca2+ release channel, inositol 1,4,5-trisphosphate (InsP3) receptor (InsP3R). Being ubiquitous and present in organisms ranging from humans to Caenorhabditis elegans, InsP3R has a vital role in the control of cellular and physiological processes as diverse as cell division, cell proliferation, apoptosis, fertilization, development, behaviour, memory and learning. Mouse type I InsP3R (InsP3R1), found in high abundance in cerebellar Purkinje cells, is a polypeptide with three major functionally distinct regions: the amino-terminal InsP3-binding region, the central modulatory region and the carboxy-terminal channel region. Here we present a 2.2-A crystal structure of the InsP3-binding core of mouse InsP3R1 in complex with InsP3. The asymmetric, boomerang-like structure consists of an N-terminal beta-trefoil domain and a C-terminal alpha-helical domain containing an 'armadillo repeat'-like fold. The cleft formed by the two domains exposes a cluster of arginine and lysine residues that coordinate the three phosphoryl groups of InsP3. Putative Ca2+-binding sites are identified in two separate locations within the InsP3-binding core.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Inositol 1,4,5-Trisphosphate Receptors , Ligands , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary
16.
J Biol Chem ; 277(52): 50573-8, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12370172

ABSTRACT

Yellow emission variants of green fluorescent protein (GFP) have been found useful in a variety of applications in biological systems due to their red-shifted emission spectrum and sensitivity to environmental parameters, such as pH and ionic strength. However, slow maturation properties and new requirements for more intense fluorescence necessitated further mutagenesis studies of these proteins. Venus, a new variant with improved maturation and brightness, as well as reduced environmental dependence, was recently developed by introducing five mutations into the well characterized variant, enhanced yellow fluorescent protein (EYFP). In this paper, we present the crystal structure of Venus at 2.2 A resolution, which enabled us to correlate its novel features with these mutation points. The rearrangement of several side chains near the chromophore, initiated by the F46L mutation, was found to improve maturation at 37 degrees C by removing steric and energetic constraints, which may hinder folding of the polypeptide chain, and by accelerating the oxidation of the Calpha-Cbeta bond of Tyr(66) during chromophore formation. M153T, V163A, and S175G were also found to improve the rate of maturation by creating regions of greater flexibility. F64L induced large conformational changes in the molecule, leading to the removal of halide sensitivity by preventing ion access to the binding site.


Subject(s)
Bacterial Proteins/chemistry , Luminescent Proteins/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Crystallography, X-Ray , Genetic Variation , Luminescent Proteins/genetics , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...