Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5944, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013852

ABSTRACT

Loss-of-function mutations in the homotrimeric serine protease HTRA1 cause cerebral vasculopathy. Here, we establish independent approaches to achieve the functional correction of trimer assembly defects. Focusing on the prototypical R274Q mutation, we identify an HTRA1 variant that promotes trimer formation thus restoring enzymatic activity in vitro. Genetic experiments in Htra1R274Q mice further demonstrate that expression of this protein-based corrector in trans is sufficient to stabilize HtrA1-R274Q and restore the proteomic signature of the brain vasculature. An alternative approach employs supramolecular chemical ligands that shift the monomer-trimer equilibrium towards proteolytically active trimers. Moreover, we identify a peptidic ligand that activates HTRA1 monomers. Our findings open perspectives for tailored protein repair strategies.


Subject(s)
High-Temperature Requirement A Serine Peptidase 1 , High-Temperature Requirement A Serine Peptidase 1/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , Animals , Humans , Mice , Protein Conformation , Protein Multimerization , HEK293 Cells , Brain/metabolism , Brain/pathology , Mutation , Loss of Function Mutation
2.
Curr Biol ; 34(11): 2279-2293.e6, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38776902

ABSTRACT

Faithful chromosome segregation requires that sister chromatids establish bi-oriented kinetochore-microtubule attachments. The spindle assembly checkpoint (SAC) prevents premature anaphase onset with incomplete attachments. However, how microtubule attachment and checkpoint signaling are coordinated remains unclear. The conserved kinase Mps1 initiates SAC signaling by localizing transiently to kinetochores in prometaphase and is released upon bi-orientation. Using biochemistry, structure predictions, and cellular assays, we shed light on this dynamic behavior in Saccharomyces cerevisiae. A conserved N-terminal segment of Mps1 binds the neck region of Ndc80:Nuf2, the main microtubule receptor of kinetochores. Mutational disruption of this interface, located at the backside of the paired CH domains and opposite the microtubule-binding site, prevents Mps1 localization, eliminates SAC signaling, and impairs growth. The same interface of Ndc80:Nuf2 binds the microtubule-associated Dam1 complex. We demonstrate that the error correction kinase Ipl1/Aurora B controls the competition between Dam1 and Mps1 for the same binding site. Thus, binding of the Dam1 complex to Ndc80:Nuf2 may release Mps1 from the kinetochore to promote anaphase onset.


Subject(s)
Cell Cycle Proteins , Kinetochores , Microtubules , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Kinetochores/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Microtubules/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Nuclear Proteins
3.
Molecules ; 29(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38675584

ABSTRACT

To understand the biological relevance and mode of action of artificial protein ligands, crystal structures with their protein targets are essential. Here, we describe and investigate all known crystal structures that contain a so-called "molecular tweezer" or one of its derivatives with an attached natural ligand on the respective target protein. The aromatic ring system of these compounds is able to include lysine and arginine side chains, supported by one or two phosphate groups that are attached to the half-moon-shaped molecule. Due to their marked preference for basic amino acids and the fully reversible binding mode, molecular tweezers are able to counteract pathologic protein aggregation and are currently being developed as disease-modifying therapies against neurodegenerative diseases such as Alzheimer's and Parkinson's disease. We analyzed the corresponding crystal structures with 14-3-3 proteins in complex with mono- and diphosphate tweezers. Furthermore, we solved crystal structures of two different tweezer variants in complex with the enzyme Δ1-Pyrroline-5-carboxyl-dehydrogenase (P5CDH) and found that the tweezers are bound to a lysine and methionine side chain, respectively. The different binding modes and their implications for affinity and specificity are discussed, as well as the general problems in crystallizing protein complexes with artificial ligands.


Subject(s)
Protein Binding , Crystallography, X-Ray , Ligands , Humans , Models, Molecular , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/metabolism , Binding Sites , Proteins/chemistry , Protein Conformation
4.
Nat Struct Mol Biol ; 31(6): 861-873, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459128

ABSTRACT

Biorientation of chromosomes during cell division is necessary for precise dispatching of a mother cell's chromosomes into its two daughters. Kinetochores, large layered structures built on specialized chromosome loci named centromeres, promote biorientation by binding and sensing spindle microtubules. One of the outer layer main components is a ten-subunit assembly comprising Knl1C, Mis12C and Ndc80C (KMN) subcomplexes. The KMN is highly elongated and docks on kinetochores and microtubules through interfaces at its opposite extremes. Here, we combine cryogenic electron microscopy reconstructions and AlphaFold2 predictions to generate a model of the human KMN that reveals all intra-KMN interfaces. We identify and functionally validate two interaction interfaces that link Mis12C to Ndc80C and Knl1C. Through targeted interference experiments, we demonstrate that this mutual organization strongly stabilizes the KMN assembly. Our work thus reports a comprehensive structural and functional analysis of this part of the kinetochore microtubule-binding machinery and elucidates the path of connections from the chromatin-bound components to the force-generating components.


Subject(s)
Cryoelectron Microscopy , Kinetochores , Microtubule-Associated Proteins , Models, Molecular , Nuclear Proteins , Humans , Kinetochores/metabolism , Kinetochores/ultrastructure , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/chemistry , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/chemistry , Microtubules/metabolism , Microtubules/ultrastructure , Protein Binding , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/chemistry , HeLa Cells
5.
EMBO J ; 42(13): e112504, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37203876

ABSTRACT

During cell division, kinetochores link chromosomes to spindle microtubules. The Ndc80 complex, a crucial microtubule binder, populates each kinetochore with dozens of copies. Whether adjacent Ndc80 complexes cooperate to promote microtubule binding remains unclear. Here we demonstrate that the Ndc80 loop, a short sequence that interrupts the Ndc80 coiled-coil at a conserved position, folds into a more rigid structure than previously assumed and promotes direct interactions between full-length Ndc80 complexes on microtubules. Mutations in the loop impair these Ndc80-Ndc80 interactions, prevent the formation of force-resistant kinetochore-microtubule attachments, and cause cells to arrest in mitosis for hours. This arrest is not due to an inability to recruit the kinetochore-microtubule stabilizing SKA complex and cannot be overridden by mutations in the Ndc80 tail that strengthen microtubule attachment. Thus, loop-mediated organization of adjacent Ndc80 complexes is crucial for stable end-on kinetochore-microtubule attachment and spindle assembly checkpoint satisfaction.


Subject(s)
Kinetochores , Microtubules , Chromosome Segregation , Kinetochores/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis , Protein Binding , Animals
6.
J Cell Biol ; 221(11)2022 11 07.
Article in English | MEDLINE | ID: mdl-36107127

ABSTRACT

Cytoplasmic Dynein 1, or Dynein, is a microtubule minus end-directed motor. Dynein motility requires Dynactin and a family of activating adaptors that stabilize the Dynein-Dynactin complex and promote regulated interactions with cargo in space and time. How activating adaptors limit Dynein activation to specialized subcellular locales is unclear. Here, we reveal that Spindly, a mitotic Dynein adaptor at the kinetochore corona, exists natively in a closed conformation that occludes binding of Dynein-Dynactin to its CC1 box and Spindly motif. A structure-based analysis identified various mutations promoting an open conformation of Spindly that binds Dynein-Dynactin. A region of Spindly downstream from the Spindly motif and not required for cargo binding faces the CC1 box and stabilizes the intramolecular closed conformation. This region is also required for robust kinetochore localization of Spindly, suggesting that kinetochores promote Spindly activation to recruit Dynein. Thus, our work illustrates how specific Dynein activation at a defined cellular locale may require multiple factors.


Subject(s)
Cell Cycle Proteins , Cytoplasmic Dyneins , Dynactin Complex , Cell Cycle Proteins/metabolism , Cytoplasmic Dyneins/metabolism , Dynactin Complex/metabolism , Kinetochores/metabolism , Protein Conformation
7.
Nat Commun ; 13(1): 4262, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35871249

ABSTRACT

In their GTP-bound (active) form, Rab proteins interact with effector proteins that control downstream signaling. One such Rab15 effector is Rep15, which is known to have a role in receptor recycling from the endocytic recycling compartment but otherwise remains poorly characterized. Here, we report the characterization of the Rep15:Rab15 interaction and identification of Rab3 paralogs and Rab34 as Rep15 interacting partners from a yeast two-hybrid assay. Biochemical validation of the interactions is presented and crystal structures of the Rep15:Rab3B and Rep15:Rab3C complexes provide additional mechanistic insight. We find that Rep15 adopts a globular structure that is distinct from other reported Rab15, Rab3 and Rab34 effectors. Structure-based mutagenesis experiments explain the Rep15:Rab interaction specificity. Rep15 depletion in U138MG glioblastoma cells impairs cell proliferation, cell migration and receptor recycling, underscoring the need for further clarification of the role of Rep15 in cancer.


Subject(s)
rab GTP-Binding Proteins , Protein Binding , Two-Hybrid System Techniques , rab GTP-Binding Proteins/metabolism
8.
Mol Cell ; 82(11): 2113-2131.e8, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35525244

ABSTRACT

Centromeres are specialized chromosome loci that seed the kinetochore, a large protein complex that effects chromosome segregation. A 16-subunit complex, the constitutive centromere associated network (CCAN), connects between the specialized centromeric chromatin, marked by the histone H3 variant CENP-A, and the spindle-binding moiety of the kinetochore. Here, we report a cryo-electron microscopy structure of human CCAN. We highlight unique features such as the pseudo GTPase CENP-M and report how a crucial CENP-C motif binds the CENP-LN complex. The CCAN structure has implications for the mechanism of specific recognition of the CENP-A nucleosome. A model consistent with our structure depicts the CENP-C-bound nucleosome as connected to the CCAN through extended, flexible regions of CENP-C. An alternative model identifies both CENP-C and CENP-N as specificity determinants but requires CENP-N to bind CENP-A in a mode distinct from the classical nucleosome octamer.


Subject(s)
Kinetochores , Nucleosomes , Centromere/metabolism , Centromere Protein A/metabolism , Cryoelectron Microscopy , Humans , Kinetochores/metabolism , Nucleosomes/genetics
9.
EMBO J ; 41(9): e110411, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35373361

ABSTRACT

In metazoans, a ≈1 megadalton (MDa) multiprotein complex comprising the dynein-dynactin adaptor Spindly and the ROD-Zwilch-ZW10 (RZZ) complex is the building block of a fibrous biopolymer, the kinetochore fibrous corona. The corona assembles on mitotic kinetochores to promote microtubule capture and spindle assembly checkpoint (SAC) signaling. We report here a high-resolution cryo-EM structure that captures the essential features of the RZZ complex, including a farnesyl-binding site required for Spindly binding. Using a highly predictive in vitro assay, we demonstrate that the SAC kinase MPS1 is necessary and sufficient for corona assembly at supercritical concentrations of the RZZ-Spindly (RZZS) complex, and describe the molecular mechanism of phosphorylation-dependent filament nucleation. We identify several structural requirements for RZZS polymerization in rings and sheets. Finally, we identify determinants of kinetochore localization and corona assembly of Spindly. Our results describe a framework for the long-sought-for molecular basis of corona assembly on metazoan kinetochores.


Subject(s)
Kinetochores , Spindle Apparatus , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Humans , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism
10.
Sci Adv ; 7(27)2021 06.
Article in English | MEDLINE | ID: mdl-34193424

ABSTRACT

Centromeres are epigenetically determined chromosomal loci that seed kinetochore assembly to promote chromosome segregation during cell division. CENP-A, a centromere-specific histone H3 variant, establishes the foundations for centromere epigenetic memory and kinetochore assembly. It recruits the constitutive centromere-associated network (CCAN), which in turn assembles the microtubule-binding interface. How the specific organization of centromeric chromatin relates to kinetochore assembly and to centromere identity through cell division remains conjectural. Here, we break new ground by reconstituting a functional full-length version of CENP-C, the largest human CCAN subunit and a blueprint of kinetochore assembly. We show that full-length CENP-C, a dimer, binds stably to two nucleosomes and permits further assembly of all other kinetochore subunits in vitro with relative ratios closely matching those of endogenous human kinetochores. Our results imply that human kinetochores emerge from clustering multiple copies of a fundamental module and may have important implications for transgenerational inheritance of centromeric chromatin.


Subject(s)
Histones , Kinetochores , Centromere/metabolism , Centromere Protein A/genetics , Centromere Protein A/metabolism , Humans , Kinetochores/metabolism , Nucleosomes
11.
Acta Crystallogr D Struct Biol ; 77(Pt 5): 674-689, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33950022

ABSTRACT

Vibrio species play a crucial role in maintaining the carbon and nitrogen balance between the oceans and the land through their ability to employ chitin as a sole source of energy. This study describes the structural basis for the action of the GH20 ß-N-acetylglucosaminidase (VhGlcNAcase) in chitin metabolism by Vibrio campbellii (formerly V. harveyi) strain ATCC BAA-1116. Crystal structures of wild-type VhGlcNAcase in the absence and presence of the sugar ligand, and of the unliganded D437A mutant, were determined. VhGlcNAcase contains three distinct domains: an N-terminal carbohydrate-binding domain linked to a small α+ß domain and a C-terminal (ß/α)8 catalytic domain. The active site of VhGlcNAcase has a narrow, shallow pocket that is suitable for accommodating a small chitooligosaccharide. VhGlcNAcase is a monomeric enzyme of 74 kDa, but its crystal structures show two molecules of enzyme per asymmetric unit, in which Gln16 at the dimeric interface of the first molecule partially blocks the entrance to the active site of the neighboring molecule. The GlcNAc unit observed in subsite -1 makes exclusive hydrogen bonds to the conserved residues Arg274, Tyr530, Asp532 and Glu584, while Trp487, Trp546, Trp582 and Trp505 form a hydrophobic wall around the -1 GlcNAc. The catalytic mutants D437A/N and E438A/Q exhibited a drastic loss of GlcNAcase activity, confirming the catalytic role of the acidic pair (Asp437-Glu438).


Subject(s)
Acetylglucosaminidase/chemistry , Chitin/metabolism , Vibrio/enzymology , Protein Binding , Protein Domains , Substrate Specificity
12.
Science ; 371(6524): 67-71, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33384373

ABSTRACT

Open (O) and closed (C) topologies of HORMA-domain proteins are respectively associated with inactive and active states of fundamental cellular pathways. The HORMA protein O-MAD2 converts to C-MAD2 upon binding CDC20. This is rate limiting for assembly of the mitotic checkpoint complex (MCC), the effector of a checkpoint required for mitotic fidelity. A catalyst assembled at kinetochores accelerates MAD2:CDC20 association through a poorly understood mechanism. Using a reconstituted SAC system, we discovered that CDC20 is an impervious substrate for which access to MAD2 requires simultaneous docking on several sites of the catalytic complex. Our analysis indicates that the checkpoint catalyst is substrate assisted and promotes MCC assembly through spatially and temporally coordinated conformational changes in both MAD2 and CDC20. This may define a paradigm for other HORMA-controlled systems.


Subject(s)
Cdc20 Proteins/metabolism , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Mad2 Proteins/metabolism , Multiprotein Complexes/metabolism , Biocatalysis , Cell Cycle Proteins/metabolism , HeLa Cells , Humans , Mad2 Proteins/genetics , Mutation , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Serine-Threonine Kinases , Spindle Apparatus/metabolism
13.
Angew Chem Int Ed Engl ; 60(4): 1813-1820, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33022847

ABSTRACT

The scaffolding protein RbAp48 is part of several epigenetic regulation complexes and is overexpressed in a variety of cancers. In order to develop tool compounds for the study of RbAp48 function, we have developed peptide inhibitors targeting the protein-protein interaction interface between RbAp48 and the scaffold protein MTA1. Based on a MTA1-derived linear peptide with low micromolar affinity and informed by crystallographic analysis, a bicyclic peptide was developed that inhibits the RbAp48/MTA1 interaction with a very low nanomolar KD value of 8.56 nM, and which showed appreciable stability against cellular proteases. Design included exchange of a polar amide cyclization strategy to hydrophobic aromatic linkers enabling mono- and bicyclization by means of cysteine alkylation, which improved affinity by direct interaction of the linkers with a hydrophobic residue on RbAp48. Our results demonstrate that stepwise evolution of a structure-based design is a suitable strategy for inhibitor development targeting PPIs.


Subject(s)
Drug Design , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Retinoblastoma-Binding Protein 4/antagonists & inhibitors , Amino Acid Sequence , Circular Dichroism , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Mutation , Protein Conformation , Thermodynamics
14.
Nat Commun ; 11(1): 4187, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32826901

ABSTRACT

EHBP1 is an adaptor protein that regulates vesicular trafficking by recruiting Rab8 family members and Eps15-homology domain-containing proteins 1/2 (EHD1/2). It also links endosomes to the actin cytoskeleton. However, the underlying molecular mechanism of activation of EHBP1 actin-binding activity is unclear. Here, we show that both termini of EHBP1 have membrane targeting potential. EHBP1 associates with PI(3)P, PI(5)P, and phosphatidylserine via its N-terminal C2 domain. We show that in the absence of Rab8 family members, the C-terminal bivalent Mical/EHBP Rab binding (bMERB) domain forms an intramolecular complex with its central calponin homology (CH) domain and auto-inhibits actin binding. Rab8 binding to the bMERB domain relieves this inhibition. We have analyzed the CH:bMERB auto-inhibited complex and the active bMERB:Rab8 complex biochemically and structurally. Together with structure-based mutational studies, this explains how binding of Rab8 frees the CH domain and allows it to interact with the actin cytoskeleton, leading to membrane tubulation.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/genetics , Microfilament Proteins/genetics , Models, Molecular , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Protein Conformation , Protein Domains , Protein Interaction Domains and Motifs , Protein Transport/physiology , Sequence Alignment , Vesicular Transport Proteins , rab GTP-Binding Proteins/genetics
15.
J Cell Biol ; 219(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31932847

ABSTRACT

Clathrin ensures mitotic spindle stability and efficient chromosome alignment, independently of its vesicle trafficking function. Although clathrin localizes to the mitotic spindle and kinetochore fiber microtubule bundles, the mechanisms by which clathrin stabilizes microtubules are unclear. We show that clathrin adaptor interaction sites on clathrin heavy chain (CHC) are repurposed during mitosis to directly recruit the microtubule-stabilizing protein GTSE1 to the spindle. Structural analyses reveal that these sites interact directly with clathrin-box motifs on GTSE1. Disruption of this interaction releases GTSE1 from spindles, causing defects in chromosome alignment. Surprisingly, this disruption destabilizes astral microtubules, but not kinetochore-microtubule attachments, and chromosome alignment defects are due to a failure of chromosome congression independent of kinetochore-microtubule attachment stability. GTSE1 recruited to the spindle by clathrin stabilizes microtubules by inhibiting the microtubule depolymerase MCAK. This work uncovers a novel role of clathrin adaptor-type interactions to stabilize nonkinetochore fiber microtubules to support chromosome congression, defining for the first time a repurposing of this endocytic interaction mechanism during mitosis.


Subject(s)
Cell Cycle Proteins/genetics , Clathrin Heavy Chains/genetics , Kinesins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Mitosis/genetics , Animals , Chromosome Segregation/genetics , Clathrin/genetics , Humans , Kinetochores/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Spindle Apparatus/genetics
16.
J Med Chem ; 63(1): 425-432, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31841335

ABSTRACT

N-(4-Aminobutyl)-N'-(2-methoxyethyl)guanidine (8a) is a potent inhibitor targeting the hDDAH-1 active site (Ki = 18 µM) and derived from a series of guanidine- and amidine-based inhibitors. Its nonamino acid nature leads to high selectivities toward other enzymes of the nitric oxide-modulating system. Crystallographic data of 8a-bound hDDAH-1 illuminated a unique binding mode. Together with its developed N-hydroxyguanidine prodrug 11, 8a will serve as a most widely applicable, pharmacological tool to target DDAH-1-associated diseases.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Guanidines/chemistry , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Catalytic Domain/drug effects , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Guanidines/chemical synthesis , Guanidines/metabolism , Humans , Protein Binding
17.
Nat Chem Biol ; 15(7): 710-720, 2019 07.
Article in English | MEDLINE | ID: mdl-31222192

ABSTRACT

Autophagy mediates the degradation of damaged proteins, organelles and pathogens, and plays a key role in health and disease. Thus, the identification of new mechanisms involved in the regulation of autophagy is of major interest. In particular, little is known about the role of lipids and lipid-binding proteins in the early steps of autophagosome biogenesis. Using target-agnostic, high-content, image-based identification of indicative phenotypic changes induced by small molecules, we have identified autogramins as a new class of autophagy inhibitor. Autogramins selectively target the recently discovered cholesterol transfer protein GRAM domain-containing protein 1A (GRAMD1A, which had not previously been implicated in autophagy), and directly compete with cholesterol binding to the GRAMD1A StART domain. GRAMD1A accumulates at sites of autophagosome initiation, affects cholesterol distribution in response to starvation and is required for autophagosome biogenesis. These findings identify a new biological function of GRAMD1A and a new role for cholesterol in autophagy.


Subject(s)
Autophagosomes/metabolism , Membrane Proteins/metabolism , Autophagosomes/drug effects , Autophagy/drug effects , Humans , Membrane Proteins/antagonists & inhibitors , Models, Molecular , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured
18.
Elife ; 72018 07 16.
Article in English | MEDLINE | ID: mdl-30010541

ABSTRACT

α-Xenorhabdolysins (Xax) are α-pore-forming toxins (α-PFT) that form 1-1.3 MDa large pore complexes to perforate the host cell membrane. PFTs are used by a variety of bacterial pathogens to attack host cells. Due to the lack of structural information, the molecular mechanism of action of Xax toxins is poorly understood. Here, we report the cryo-EM structure of the XaxAB pore complex from Xenorhabdus nematophila and the crystal structures of the soluble monomers of XaxA and XaxB. The structures reveal that XaxA and XaxB are built similarly and appear as heterodimers in the 12-15 subunits containing pore, classifying XaxAB as bi-component α-PFT. Major conformational changes in XaxB, including the swinging out of an amphipathic helix are responsible for membrane insertion. XaxA acts as an activator and stabilizer for XaxB that forms the actual transmembrane pore. Based on our results, we propose a novel structural model for the mechanism of Xax intoxication.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Bacterial Toxins/chemistry , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/ultrastructure , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Pore Forming Cytotoxic Proteins/metabolism , Protein Conformation , Protein Multimerization
19.
ACS Chem Biol ; 13(5): 1307-1312, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29658704

ABSTRACT

Covalent modifications of nonactive site lysine residues by small molecule probes has recently evolved into an important strategy for interrogating biological systems. Here, we report the discovery of a class of bioreactive compounds that covalently modify lysine residues in DegS, the rate limiting protease of the essential bacterial outer membrane stress response pathway. These modifications lead to an allosteric activation and allow the identification of novel residues involved in the allosteric activation circuit. These findings were validated by structural analyses via X-ray crystallography and cell-based reporter systems. We anticipate that our findings are not only relevant for a deeper understanding of the structural basis of allosteric activation in DegS and other HtrA serine proteases but also pinpoint an alternative use of covalent small molecules for probing essential biochemical mechanisms.


Subject(s)
Lysine/chemistry , Molecular Probes/chemistry , Allosteric Regulation , Bacterial Proteins/chemistry , Catalysis , Crystallography, X-Ray , Protein Conformation
20.
Elife ; 62017 12 27.
Article in English | MEDLINE | ID: mdl-29280735

ABSTRACT

Centromere protein (CENP) A, a histone H3 variant, is a key epigenetic determinant of chromosome domains known as centromeres. Centromeres nucleate kinetochores, multi-subunit complexes that capture spindle microtubules to promote chromosome segregation during mitosis. Two kinetochore proteins, CENP-C and CENP-N, recognize CENP-A in the context of a rare CENP-A nucleosome. Here, we reveal the structural basis for the exquisite selectivity of CENP-N for centromeres. CENP-N uses charge and space complementarity to decode the L1 loop that is unique to CENP-A. It also engages in extensive interactions with a 15-base pair segment of the distorted nucleosomal DNA double helix, in a position predicted to exclude chromatin remodelling enzymes. Besides CENP-A, stable centromere recruitment of CENP-N requires a coincident interaction with a newly identified binding motif on nucleosome-bound CENP-C. Collectively, our studies clarify how CENP-N and CENP-C decode and stabilize the non-canonical CENP-A nucleosome to enforce epigenetic centromere specification and kinetochore assembly.


Subject(s)
Centromere Protein A/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA/metabolism , Cell Line , Centromere/chemistry , Centromere Protein A/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Cryoelectron Microscopy , Crystallography, X-Ray , DNA/chemistry , Humans , Kinetochores/chemistry , Kinetochores/metabolism , Models, Molecular , Protein Binding , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL