Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell ; 163(6): 1527-38, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638077

ABSTRACT

The killifish Nothobranchius furzeri is the shortest-lived vertebrate that can be bred in the laboratory. Its rapid growth, early sexual maturation, fast aging, and arrested embryonic development (diapause) make it an attractive model organism in biomedical research. Here, we report a draft sequence of its genome that allowed us to uncover an intra-species Y chromosome polymorphism representing-in real time-different stages of sex chromosome formation that display features of early mammalian XY evolution "in action." Our data suggest that gdf6Y, encoding a TGF-ß family growth factor, is the master sex-determining gene in N. furzeri. Moreover, we observed genomic clustering of aging-related genes, identified genes under positive selection, and revealed significant similarities of gene expression profiles between diapause and aging, particularly for genes controlling cell cycle and translation. The annotated genome sequence is provided as an online resource (http://www.nothobranchius.info/NFINgb).


Subject(s)
Biological Evolution , Killifishes/genetics , Sex Chromosomes , Aging , Animals , Female , Genome , Killifishes/physiology , Male , Molecular Sequence Data , Sex Determination Processes
2.
Proc Natl Acad Sci U S A ; 113(40): 11196-11201, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647913

ABSTRACT

Proprotein convertases (PCs) are highly specific proteases required for the proteolytic modification of many secreted proteins. An unbalanced activity of these enzymes is connected to pathologies like cancer, atherosclerosis, hypercholesterolaemia, and infectious diseases. Novel protein crystallographic structures of the prototypical PC family member furin in different functional states were determined to 1.8-2.0 Å. These, together with biochemical data and modeling by molecular dynamics calculations, suggest essential elements underlying its unusually high substrate specificity. Furin shows a complex activation mechanism and exists in at least four defined states: (i) the "off state," incompatible with substrate binding as seen in the unliganded enzyme; (ii) the active "on state" seen in inhibitor-bound furin; and the respective (iii) calcium-free and (iv) calcium-bound forms. The transition from the off to the on state is triggered by ligand binding at subsites S1 to S4 and appears to underlie the preferential recognition of the four-residue sequence motif of furin. The molecular dynamics simulations of the four structural states reflect the experimental observations in general and provide approximations of the respective stabilities. Ligation by calcium at the PC-specific binding site II influences the active-site geometry and determines the rotamer state of the oxyanion hole-forming Asn295, and thus adds a second level of the activity modulation of furin. The described crystal forms and the observations of different defined functional states may foster the development of new tools and strategies for pharmacological intervention targeting furin.


Subject(s)
Furin/chemistry , Furin/metabolism , Calcium/metabolism , Catalytic Domain , Crystallography, X-Ray , Furin/antagonists & inhibitors , Humans , Ligands , Molecular Dynamics Simulation , Principal Component Analysis , Protein Conformation , Static Electricity , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity
3.
Biochemistry ; 57(6): 925-934, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29314830

ABSTRACT

The proprotein convertase furin is a highly specific serine protease modifying and thereby activating proteins in the secretory pathway by proteolytic cleavage. Its substrates are involved in many diseases, including cancer and infections caused by bacteria and viruses. Understanding furin's substrate specificity is crucially important for the development of pharmacologically applicable inhibitors. Using protein X-ray crystallography, we investigated the extended substrate binding site of furin in complex with three peptide-derived inhibitors at up to 1.9 Å resolution. The structure of the protease bound with a hexapeptide inhibitor revealed molecular details of its S6 pocket, which remained completely unknown so far. The arginine residue at P6 induced an unexpected turnlike conformation of the inhibitor backbone, which is stabilized by intra- and intermolecular H-bonds. In addition, we confirmed the binding of arginine to the previously proposed S5 pocket (S51). An alternative S5 site (S52) could be utilized by shorter side chains as demonstrated for a 4-aminomethyl-phenylacetyl residue, which shows steric properties similar to those of a lysine side chain. Interestingly, we also observed binding of a peptide with citrulline at P4 substituting for the highly conserved arginine. The structural data might indicate an unusual protonation state of Asp264 maintaining the interaction with uncharged citrulline. The herein identified molecular interaction sites at P5 and P6 can be utilized to improve next-generation furin inhibitors. Our data will also help to predict furin substrates more precisely on the basis of the additional specificity determinants observed for P5 and P6.


Subject(s)
Furin/chemistry , Binding Sites , Crystallography, X-Ray , Furin/antagonists & inhibitors , Furin/metabolism , HEK293 Cells , Humans , Hydrogen Bonding , Molecular Docking Simulation , Peptides/chemistry , Peptides/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protein Conformation , Substrate Specificity
4.
Proc Natl Acad Sci U S A ; 110(51): 20587-92, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297905

ABSTRACT

Receptor tyrosine kinases participate in several signaling pathways through small G proteins such as Ras (rat sarcoma). An important component in the activation of these G proteins is Son of sevenless (SOS), which catalyzes the nucleotide exchange on Ras. For optimal activity, a second Ras molecule acts as an allosteric activator by binding to a second Ras-binding site within SOS. This allosteric Ras-binding site is blocked by autoinhibitory domains of SOS. We have reported recently that Ras activation also requires the actin-binding proteins ezrin, radixin, and moesin. Here we report the mechanism by which ezrin modulates SOS activity and thereby Ras activation. Active ezrin enhances Ras/MAPK signaling and interacts with both SOS and Ras in vivo and in vitro. Moreover, in vitro kinetic assays with recombinant proteins show that ezrin also is important for the activity of SOS itself. Ezrin interacts with GDP-Ras and with the Dbl homology (DH)/pleckstrin homology (PH) domains of SOS, bringing GDP-Ras to the proximity of the allosteric site of SOS. These actions of ezrin are antagonized by the neurofibromatosis type 2 tumor-suppressor protein merlin. We propose an additional essential step in SOS/Ras control that is relevant for human cancer as well as all physiological processes involving Ras.


Subject(s)
Cytoskeletal Proteins/metabolism , Guanosine Diphosphate/metabolism , MAP Kinase Signaling System , Neurofibromin 2/metabolism , Oncogene Protein p21(ras)/metabolism , Son of Sevenless Proteins/metabolism , Animals , Cytoskeletal Proteins/genetics , Guanosine Diphosphate/genetics , Humans , Mice , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurofibromin 2/genetics , Oncogene Protein p21(ras)/genetics , Son of Sevenless Proteins/genetics
5.
Biochemistry ; 54(15): 2490-9, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25835329

ABSTRACT

The amyloid precursor protein (APP) and its neurotoxic cleavage product Aß are key players in the development of Alzheimer's disease (AD) and appear to be essential for neuronal development and cell homeostasis. Proteolytic processing of APP and its physiological function depend on its interaction with heparin and are influenced by the binding of metal ions and sorLA. We created various mutations of metal binding site M1 residing within the extracellular E2 domain of APP. Using isothermal titration calorimetry and circular dichroism spectroscopy, we analyzed the binding of Cu(2+) and Zn(2+) to APP E2 and identified two mutations that are most suited for functional studies to dissect ion specific effects of metal binding. The H313A mutation abrogates only copper-based effects, whereas the H382A mutation weakens any metal binding at M1 of APP E2. Subsequently, we tested the effect of Cu(2+) and Zn(2+) on the binding of heparin and sorLA to APP E2 using a chromatographic technique and surface plasmon resonance. We show that Zn(2+) and to a larger degree also Cu(2+) enhance the binding of heparin to APP E2, consistent with an extracellular regulation of the function of APP by both metal ions. In contrast, neither ion seemed to affect the interaction between APP E2 and sorLA. This supports an intracellular interaction between the latter two partners that would not sense extracellular variations of metal ions upon synaptic activity.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Copper/chemistry , Heparin/chemistry , LDL-Receptor Related Proteins/chemistry , Membrane Transport Proteins/chemistry , Mutation, Missense , Zinc/chemistry , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Substitution , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Binding Sites , Copper/metabolism , Heparin/metabolism , Humans , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Protein Binding , Surface Plasmon Resonance , Zinc/metabolism
6.
J Biol Chem ; 289(27): 19019-30, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24855651

ABSTRACT

The amyloid precursor protein (APP) and the APP-like proteins 1 and 2 (APLP1 and APLP2) are a family of multidomain transmembrane proteins possessing homo- and heterotypic contact sites in their ectodomains. We previously reported that divalent metal ions dictate the conformation of the extracellular APP E2 domain (Dahms, S. O., Könnig, I., Roeser, D., Gührs, K.-H., Mayer, M. C., Kaden, D., Multhaup, G., and Than, M. E. (2012) J. Mol. Biol. 416, 438-452), but unresolved is the nature and functional importance of metal ion binding to APLP1 and APLP2. We found here that zinc ions bound to APP and APLP1 E2 domains and mediated their oligomerization, whereas the APLP2 E2 domain interacted more weakly with zinc possessing a less surface-exposed zinc-binding site, and stayed monomeric. Copper ions bound to E2 domains of all three proteins. Fluorescence resonance energy transfer (FRET) analyses examined the effect of metal ion binding to APP and APLPs in the cellular context in real time. Zinc ions specifically induced APP and APLP1 oligomerization and forced APLP1 into multimeric clusters at the plasma membrane consistent with zinc concentrations in the blood and brain. The observed effects were mediated by a novel zinc-binding site within the APLP1 E2 domain as APLP1 deletion mutants revealed. Based upon its cellular localization and its dominant response to zinc ions, APLP1 is mainly affected by extracellular zinc among the APP family proteins. We conclude that zinc binding and APP/APLP oligomerization are intimately linked, and we propose that this represents a novel mechanism for regulating APP/APLP protein function at the molecular level.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Protein Multimerization , Zinc/metabolism , Amino Acid Sequence , Binding Sites , Copper/metabolism , HEK293 Cells , Humans , Protein Multimerization/drug effects , Protein Structure, Quaternary , Protein Structure, Tertiary , Zinc/pharmacology
7.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 3): 494-504, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25760599

ABSTRACT

Beyond the pathology of Alzheimer's disease, the members of the amyloid precursor protein (APP) family are essential for neuronal development and cell homeostasis in mammals. APP and its paralogues APP-like protein 1 (APLP1) and APP-like protein 2 (APLP2) contain the highly conserved heparan sulfate (HS) binding domain E2, which effects various (patho)physiological functions. Here, two crystal structures of the E2 domain of APLP1 are presented in the apo form and in complex with a heparin dodecasaccharide at 2.5 Šresolution. The apo structure of APLP1 E2 revealed an unfolded and hence flexible N-terminal helix αA. The (APLP1 E2)2-(heparin)2 complex structure revealed two distinct binding modes, with APLP1 E2 explicitly recognizing the heparin terminus but also interacting with a continuous heparin chain. The latter only requires a certain register of the sugar moieties that fits to a positively charged surface patch and contributes to the general heparin-binding capability of APP-family proteins. Terminal binding of APLP1 E2 to heparin specifically involves a structure of the nonreducing end that is very similar to heparanase-processed HS chains. These data reveal a conserved mechanism for the binding of APP-family proteins to HS and imply a specific regulatory role of HS modifications in the biology of APP and APP-like proteins.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Heparin/chemistry , Humans , Protein Structure, Secondary , Protein Structure, Tertiary
8.
Chembiochem ; 16(17): 2441-4, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26426719

ABSTRACT

We report the engineering of the monocyclic sunflower trypsin inhibitor (SFTI-1[1,14]) into a potent furin inhibitor. In a rational approach, we converted the native scaffold of this trypsin-like serine protease inhibitor into a subtilisin-like one by substitutions in the canonical and, particularly, in the substrate-binding loop. Although the substrate sequence for furin is Arg-X-Arg/Lys-Arg↓, the most potent inhibitor had a lysine at position P1. C-terminally truncated versions demonstrated the strongest activity, thus suggesting a lack of interaction between this motif and the surface of furin. This observation was further supported by molecular modeling. With an inhibition constant of 0.49 nm, the engineered peptide H-KRCKKSIPPICF-NH2 is a promising compound for further development of furin inhibitors aimed at controlling the activity of this protease in vitro and in vivo.


Subject(s)
Furin/antagonists & inhibitors , Peptides/chemistry , Amino Acid Sequence , Binding Sites , Drug Design , Furin/metabolism , Helianthus/metabolism , Kinetics , Molecular Dynamics Simulation , Peptides/chemical synthesis , Peptides/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Structure, Tertiary
9.
J Struct Biol ; 187(1): 30-37, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24859793

ABSTRACT

The amyloid precursor protein (APP) and its cellular processing are believed to be centrally involved in the etiology of Alzheimer's disease (AD). In addition, many physiological functions have been described for APP, including a role in cell-cell- and cell-ECM-adhesion as well as in axonal outgrowth. We show here the molecular determinants of the oligomerization/dimerization of APP, which is central for its cellular (mis)function. Using size exclusion chromatography (SEC), dynamic light scattering and SEC-coupled static light scattering we demonstrate that the dimerization of APP is energetically induced by a heparin mediated dimerization of the E1 domain, which results in a dimeric interaction of E2. We also show that the acidic domain (AcD) interferes with the dimerization of E1 and propose a model where both, cis- and trans-dimerization occur dependent on cellular localization and function.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Heparin/chemistry , Amyloid beta-Protein Precursor/genetics , Humans , Kinetics , Models, Molecular , Mutation , Protein Binding , Protein Folding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thermodynamics
10.
EMBO J ; 29(16): 2841-57, 2010 Aug 18.
Article in English | MEDLINE | ID: mdl-20606625

ABSTRACT

Mutations in fused in sarcoma (FUS) are a cause of familial amyotrophic lateral sclerosis (fALS). Patients carrying point mutations in the C-terminus of FUS show neuronal cytoplasmic FUS-positive inclusions, whereas in healthy controls, FUS is predominantly nuclear. Cytoplasmic FUS inclusions have also been identified in a subset of frontotemporal lobar degeneration (FTLD-FUS). We show that a non-classical PY nuclear localization signal (NLS) in the C-terminus of FUS is necessary for nuclear import. The majority of fALS-associated mutations occur within the NLS and impair nuclear import to a degree that correlates with the age of disease onset. This presents the first case of disease-causing mutations within a PY-NLS. Nuclear import of FUS is dependent on Transportin, and interference with this transport pathway leads to cytoplasmic redistribution and recruitment of FUS into stress granules. Moreover, proteins known to be stress granule markers co-deposit with inclusions in fALS and FTLD-FUS patients, implicating stress granule formation in the pathogenesis of these diseases. We propose that two pathological hits, namely nuclear import defects and cellular stress, are involved in the pathogenesis of FUS-opathies.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Karyopherins/metabolism , Point Mutation , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cells, Cultured , Cytoplasmic Granules/pathology , Gene Knockdown Techniques , HeLa Cells , Humans , Karyopherins/genetics , Molecular Sequence Data , Neurons/pathology , Protein Structure, Tertiary , RNA-Binding Protein FUS/analysis , RNA-Binding Protein FUS/chemistry , Zebrafish/embryology
11.
Biol Chem ; 395(5): 485-98, 2014 May.
Article in English | MEDLINE | ID: mdl-24516000

ABSTRACT

The amyloid precursor protein (APP) and its processing are widely believed to be central for the etiology of Alzheimer's disease (AD) and appear essential for neuronal development and cell homeostasis in mammals. Many studies show the proteolysis of APP by the proteases α-, ß- and γ-secretase, functional aspects of the protein and the structure of individual domains. It is, however, largely unclear and currently also widely debated of how the structures of individual domains and their interactions determine the observed functionalities of APP and how they are arranged within the three-dimensional architecture of the entire protein. Further unanswered questions relate to the physiologic function of APP, the regulation of its proteolytic processing and the structural and functional effect of its cellular trafficking and processing. In this review, we summarize our current understanding of the structure-function-relationship of the multi-domain protein APP. This type-I transmembrane protein consists of the two folded E1 and E2 segments that are connected to one another and to the single transmembrane helix by flexible segments and likely fulfills several independent functions.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/metabolism , Humans , Membrane Proteins/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
12.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 2): 284-97, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23385464

ABSTRACT

Heavy-atom clusters (HA clusters) containing a large number of specifically arranged electron-dense scatterers are especially useful for experimental phase determination of large complex structures, weakly diffracting crystals or structures with large unit cells. Often, the determination of the exact orientation of the HA cluster and hence of the individual heavy-atom positions proves to be the critical step in successful phasing and subsequent structure solution. Here, it is demonstrated that molecular replacement (MR) with either anomalous or isomorphous differences is a useful strategy for the correct placement of HA cluster compounds. The polyoxometallate cluster hexasodium α-metatungstate (HMT) was applied in phasing the structure of death receptor 6. Even though the HA cluster is bound in alternate partially occupied orientations and is located at a special position, its correct localization and orientation could be determined at resolutions as low as 4.9 Å. The broad applicability of this approach was demonstrated for five different derivative crystals that included the compounds tantalum tetradecabromide and trisodium phosphotungstate in addition to HMT. The correct placement of the HA cluster depends on the length of the intramolecular vectors chosen for MR, such that both a larger cluster size and the optimal choice of the wavelength used for anomalous data collection strongly affect the outcome.


Subject(s)
Crystallography, X-Ray/methods , Muramidase/chemistry , Receptors, Tumor Necrosis Factor/chemistry , Ubiquitin-Activating Enzymes/chemistry , Animals , Crystallography, X-Ray/standards , Databases, Protein/standards , Electrons , Humans , Metals, Heavy/chemistry , Mice , Models, Molecular , Molecular Weight , Muramidase/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Scattering, Radiation , Ubiquitin-Activating Enzymes/metabolism
13.
Proc Natl Acad Sci U S A ; 107(12): 5381-6, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212142

ABSTRACT

The amyloid precursor protein (APP) is the key player in Alzheimer's disease pathology, yet APP and its analogues are also essential for neuronal development and cell homeostasis in mammals. We have determined the crystal structure of the entire N-terminal APP-E1 domain consisting of the growth factor like and the copper binding domains at 2.7-A resolution and show that E1 functions as a rigid functional entity. The two subdomains interact tightly in a pH-dependent manner via an evolutionarily conserved interface area. Two E1 entities dimerize upon their interaction with heparin, requiring 8-12 sugar rings to form the heparin-bridged APP-E1 dimer in an endothermic and pH-dependent process that is characterized by a low micromolar dissociation constant. Limited proteolysis confirms that the heparin-bridged E1 dimers obtained in solution correspond to a dimer contact in our crystal, enabling us to model this heparin-[APP-E1](2) complex. Correspondingly, the APP-based signal transduction, cell-cell- and/or cell-ECM interaction should depend on dimerization induced by heparin, as well as on pH, arguing that APP could fulfill different functions depending on its (sub)cellular localization.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Biophysical Phenomena , Crystallography, X-Ray , Heparin , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Models, Molecular , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
14.
Biochem J ; 433(1): 75-83, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20964629

ABSTRACT

SNX33 (sorting nexin 33) is a homologue of the endocytic protein SNX9 and has been implicated in actin polymerization and the endocytosis of the amyloid precursor protein. SNX33 belongs to the large family of BAR (Bin/amphiphysin/Rvs) domain-containing proteins, which alter cellular protein trafficking by modulating cellular membranes and the cytoskeleton. Some BAR domains engage in homodimerization, whereas other BAR domains also mediate heterodimerization between different BAR domain-containing proteins. The molecular basis for this difference is not yet understood. Using co-immunoprecipitations we report that SNX33 forms homodimers, but not heterodimers, with other BAR domain-containing proteins, such as SNX9. Domain deletion analysis revealed that the BAR domain, but not the SH3 (Src homology 3) domain, was required for homodimerization of SNX33. Additionally, the BAR domain prevented the heterodimerization between SNX9 and SNX33, as determined by domain swap experiments. Molecular modelling of the SNX33 BAR domain structure revealed that key amino acids located at the BAR domain dimer interface of the SNX9 homodimer are not conserved in SNX33. Replacing these amino acids in SNX9 with the corresponding amino acids of SNX33 allowed the mutant SNX9 to heterodimerize with SNX33. Taken together, the present study identifies critical amino acids within the BAR domains of SNX9 and SNX33 as determinants for the specificity of BAR domain-mediated interactions and suggests that SNX9 and SNX33 have distinct molecular functions.


Subject(s)
Protein Multimerization , Sorting Nexins/metabolism , Amino Acids/analysis , Binding Sites , HeLa Cells , Humans , Protein Binding , Protein Structure, Tertiary , Sequence Deletion , Sorting Nexins/genetics
15.
Biol Chem ; 392(11): 973-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21875402

ABSTRACT

In eucaryotes, many secreted proteins and peptides are proteolytically excised from larger precursor proteins by a specific class of serine proteases, the proprotein/prohormone convertases (PCs). This cleavage is essential for substrate activation, making the PCs very interesting pharmacological targets in cancer and infectious disease research. Correspondingly, their structure, function and inhibition are intensely studied - studies that require the respective target proteins in large amounts and at high purity. Here we describe the development of a novel purification protocol of furin, the best-studied member of the PC family. We combined the heterologous expression of furin from CHO cells with a novel purification scheme employing an affinity step that efficiently extracts only active furin from the conditioned medium by using furin-specific inhibitor moieties as bait. Several potential affinity tags were synthesized and their binding to furin characterized. The best compound, Biotin-(Adoa)(2)-Arg-Pro-Arg-4-Amba coupled to streptavidin-Sepharose beads, was used in a three-step chromatographic protocol and routinely resulted in a high yield of a homogeneous furin preparation with a specific activity of ~60 units/mg protein. This purification and the general strategy can easily be adapted to the efficient purification of other PC family members.


Subject(s)
Chromatography, Affinity/methods , Furin/isolation & purification , Animals , CHO Cells , Cricetinae , Furin/genetics , Furin/metabolism , Gene Expression , Mice , Protease Inhibitors/metabolism
16.
Bioorg Med Chem Lett ; 21(2): 836-40, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21168329

ABSTRACT

A novel series of amidinohydrazone-derived furin inhibitors was prepared; the most potent compounds 17 and 21 inhibit furin with K(i) values of 0.46 and 0.59µM, respectively. In contrast to inhibitor 17, which still contains a guanidino residue, compound 21 possesses only weakly basic amidinohydrazone groups.


Subject(s)
Amidines/chemistry , Amidines/pharmacology , Furin/antagonists & inhibitors , Furin/metabolism , Hydrazones/chemistry , Hydrazones/pharmacology , Humans , Molecular Structure
17.
Nat Commun ; 11(1): 5579, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33149111

ABSTRACT

Cell-to-cell communications are critical determinants of pathophysiological phenotypes, but methodologies for their systematic elucidation are lacking. Herein, we propose an approach for the Systematic Elucidation and Assessment of Regulatory Cell-to-cell Interaction Networks (SEARCHIN) to identify ligand-mediated interactions between distinct cellular compartments. To test this approach, we selected a model of amyotrophic lateral sclerosis (ALS), in which astrocytes expressing mutant superoxide dismutase-1 (mutSOD1) kill wild-type motor neurons (MNs) by an unknown mechanism. Our integrative analysis that combines proteomics and regulatory network analysis infers the interaction between astrocyte-released amyloid precursor protein (APP) and death receptor-6 (DR6) on MNs as the top predicted ligand-receptor pair. The inferred deleterious role of APP and DR6 is confirmed in vitro in models of ALS. Moreover, the DR6 knockdown in MNs of transgenic mutSOD1 mice attenuates the ALS-like phenotype. Our results support the usefulness of integrative, systems biology approach to gain insights into complex neurobiological disease processes as in ALS and posit that the proposed methodology is not restricted to this biological context and could be used in a variety of other non-cell-autonomous communication mechanisms.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Cell Communication/physiology , Cell Death/physiology , Motor Neurons/metabolism , Superoxide Dismutase-1/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Animals , Cells, Cultured , Computational Biology , Disease Models, Animal , Gene Knockdown Techniques , Gene Silencing , Humans , Ligands , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Proteomics , RNA, Small Interfering , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Superoxide Dismutase-1/genetics
18.
ChemMedChem ; 14(6): 673-685, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30680958

ABSTRACT

The activation of viral glycoproteins by the host protease furin is an essential step in the replication of numerous pathogenic viruses. Thus, effective inhibitors of furin could serve as broad-spectrum antiviral drugs. A crystal structure of an inhibitory hexapeptide derivative in complex with furin served as template for the rational design of various types of new cyclic inhibitors. Most of the prepared derivatives are relatively potent furin inhibitors with inhibition constants in the low nanomolar or even sub-nanomolar range. For seven derivatives the crystal structures in complex with furin could be determined. In three complexes, electron density was found for the entire inhibitor. In the other cases the structures could be determined only for the P6/P5-P1 segments, which directly interact with furin. The cyclic derivatives together with two non-cyclic reference compounds were tested as inhibitors of the proteolytic activation and replication of respiratory syncytial virus in cells. Significant antiviral activity was found for both linear reference inhibitors, whereas a negligible efficacy was determined for the cyclic derivatives.


Subject(s)
Enzyme Inhibitors/pharmacology , Furin/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Proprotein Convertases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemical synthesis , Humans , Macrocyclic Compounds/chemical synthesis
19.
J Mol Biol ; 357(1): 195-209, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16414069

ABSTRACT

Tryptases alpha and beta are trypsin-like serine proteinases expressed in large amounts by mast cells. Beta-tryptase is a tetramer that has enzymatic activity, but requires heparin binding to maintain functional and structural stability, whereas alpha-tryptase has little, if any, enzymatic activity but is a stable tetramer in the absence of heparin. As shown previously, these differences can be mainly attributed to the different conformations of the 214-220 segment. Interestingly, the replacement of Asp216 by Gly, which is present in beta-tryptase, results in enzymatically active but less stable alpha-tryptase mutants. We have solved the crystal structures of both the single (D216G) and the double (K192Q/D216G) mutant forms of recombinant human alphaI-tryptase in complex with the peptide inhibitor leupeptin, as well as the structure of the non-inhibited single mutant. The inhibited mutants exhibited an open functional substrate binding site, while in the absence of an inhibitor, the open (beta-tryptase-like) and the closed (alpha-tryptase-like) conformations were present simultaneously. This shows that both forms are in a two-state equilibrium, which is influenced by the residues in the vicinity of the active site and by inhibitor/substrate binding. Novel insights regarding the observed stability differences as well as a potential proteolytic activity of wild-type alpha-tryptase, which may possess a cryptic active site, are discussed.


Subject(s)
Cysteine Proteinase Inhibitors/chemistry , Isoenzymes/chemistry , Leupeptins/chemistry , Protein Structure, Quaternary , Serine Endopeptidases/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cattle , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Leupeptins/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Tryptases
20.
ACS Chem Biol ; 12(5): 1211-1216, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28402100

ABSTRACT

Proprotein convertases (PCs) represent highly selective serine proteases that activate their substrates upon proteolytic cleavage. Their inhibition is a promising strategy for the treatment of several pathologies including cancer, atherosclerosis, hypercholesterolaemia, and infectious diseases. Here, we present the first experimental complex of furin with a non-substrate-like small molecule inhibitor, and the X-ray structure of the enzyme complexed to the small molecule inhibitor 1 at 1.9 Å resolution. Two molecules of inhibitor 1 were found to interact with furin. One is anchored at the S4 pocket of the enzyme and interferes directly with the conformation and function of the catalytic triade; the other molecule shows weaker binding and interacts with a distant, less conserved region of furin. The observed binding modes represent a new inhibition strategy of furin and imply the possibility to attain specificity among the PCs providing an innovative starting point of structure guided inhibitor development for furin.


Subject(s)
Catalytic Domain , Enzyme Inhibitors/chemistry , Furin/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Furin/chemistry , Humans , Proprotein Convertases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL