Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Science ; 384(6692): eadn9560, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38603491

ABSTRACT

Formins control the assembly of actin filaments (F-actin) that drive cell morphogenesis and motility in eukaryotes. However, their molecular interaction with F-actin and their mechanism of action remain unclear. In this work, we present high-resolution cryo-electron microscopy structures of F-actin barbed ends bound by three distinct formins, revealing a common asymmetric formin conformation imposed by the filament. Formation of new intersubunit contacts during actin polymerization sterically displaces formin and triggers its translocation. This "undock-and-lock" mechanism explains how actin-filament growth is coordinated with formin movement. Filament elongation speeds are controlled by the positioning and stability of actin-formin interfaces, which distinguish fast and slow formins. Furthermore, we provide a structure of the actin-formin-profilin ring complex, which resolves how profilin is rapidly released from the barbed end during filament elongation.


Subject(s)
Actin Cytoskeleton , Actins , Formins , Actin Cytoskeleton/chemistry , Actins/chemistry , Cryoelectron Microscopy , Formins/chemistry , Formins/genetics , Profilins/chemistry , Mutation , Schizosaccharomyces
2.
Nat Struct Mol Biol ; 30(11): 1774-1785, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37749275

ABSTRACT

The release of inorganic phosphate (Pi) from actin filaments constitutes a key step in their regulated turnover, which is fundamental to many cellular functions. The mechanisms underlying Pi release from the core and barbed end of actin filaments remain unclear. Here, using human and bovine actin isoforms, we combine cryo-EM with molecular-dynamics simulations and in vitro reconstitution to demonstrate how actin releases Pi through a 'molecular backdoor'. While constantly open at the barbed end, the backdoor is predominantly closed in filament-core subunits and opens only transiently through concerted amino acid rearrangements. This explains why Pi escapes rapidly from the filament end but slowly from internal subunits. In a nemaline-myopathy-associated actin variant, the backdoor is predominantly open in filament-core subunits, resulting in accelerated Pi release and filaments with drastically shortened ADP-Pi caps. Our results provide the molecular basis for Pi release from actin and exemplify how a disease-linked mutation distorts the nucleotide-state distribution and atomic structure of the filament.


Subject(s)
Actins , Phosphates , Animals , Cattle , Humans , Actins/metabolism , Phosphates/metabolism , Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Adenosine Diphosphate/metabolism
3.
IUCrdata ; 7(Pt 9): x220852, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36452441

ABSTRACT

Remarkable features are reported in the diffraction pattern produced by a crystal of the second extracellular domain of tetraspanin CD9 (deemed CD9EC2), the structure of which has been described previously [Oosterheert et al. (2020 ▸), Life Sci. Alliance, 3, e202000883]. CD9EC2 crystallized in space group P1 and was twinned. Two types of diffuse streaks are observed. The stronger diffuse streaks are related to the twinning and occur in the direction perpendicular to the twinning interface. It is concluded that the twin domains scatter coherently as both Bragg reflections and diffuse streaks are seen. The weaker streaks along c* are unrelated to the twinning but are caused by intermittent layers of non-crystallographic symmetry related molecules. It is envisaged that the raw diffraction images could be very useful for methods developers trying to remove the diffuse scattering to extract accurate Bragg intensities or using it to model the effect of packing disorder on the molecular structure.

4.
Nature ; 611(7935): 374-379, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36289337

ABSTRACT

The dynamic turnover of actin filaments (F-actin) controls cellular motility in eukaryotes and is coupled to changes in the F-actin nucleotide state1-3. It remains unclear how F-actin hydrolyses ATP and subsequently undergoes subtle conformational rearrangements that ultimately lead to filament depolymerization by actin-binding proteins. Here we present cryo-electron microscopy structures of F-actin in all nucleotide states, polymerized in the presence of Mg2+ or Ca2+ at approximately 2.2 Å resolution. The structures show that actin polymerization induces the relocation of water molecules in the nucleotide-binding pocket, activating one of them for the nucleophilic attack of ATP. Unexpectedly, the back door for the subsequent release of inorganic phosphate (Pi) is closed in all structures, indicating that Pi release occurs transiently. The small changes in the nucleotide-binding pocket after ATP hydrolysis and Pi release are sensed by a key amino acid, amplified and transmitted to the filament periphery. Furthermore, differences in the positions of water molecules in the nucleotide-binding pocket explain why Ca2+-actin shows slower polymerization rates than Mg2+-actin. Our work elucidates the solvent-driven rearrangements that govern actin filament assembly and aging and lays the foundation for the rational design of drugs and small molecules for imaging and therapeutic applications.


Subject(s)
Actin Cytoskeleton , Actins , Aging , Cryoelectron Microscopy , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/chemistry , Actins/metabolism , Actins/ultrastructure , Adenosine Triphosphate/metabolism , Hydrolysis , Nucleotides/chemistry , Nucleotides/metabolism , Water/metabolism , Aging/metabolism , Magnesium , Calcium , Amino Acids , Phosphates
5.
J Immunol ; 208(9): 2207-2219, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35428691

ABSTRACT

Cleavage of the mammalian plasma protein C4 into C4b initiates opsonization, lysis, and clearance of microbes and damaged host cells by the classical and lectin pathways of the complement system. Dysregulated activation of C4 and other initial components of the classical pathway may cause or aggravate pathologies, such as systemic lupus erythematosus, Alzheimer disease, and schizophrenia. Modulating the activity of C4b by small-molecule or protein-based inhibitors may represent a promising therapeutic approach for preventing excessive inflammation and damage to host cells and tissue. Here, we present seven nanobodies, derived from llama (Lama glama) immunization, that bind to human C4b (Homo sapiens) with high affinities ranging from 3.2 nM to 14 pM. The activity of the nanobodies varies from no to complete inhibition of the classical pathway. The inhibiting nanobodies affect different steps in complement activation, in line with blocking sites for proconvertase formation, C3 substrate binding to the convertase, and regulator-mediated inactivation of C4b. For four nanobodies, we determined single-particle cryo-electron microscopy structures in complex with C4b at 3.4-4 Å resolution. The structures rationalize the observed functional effects of the nanobodies and define their mode of action during complement activation. Thus, we characterized seven anti-C4b nanobodies with diverse effects on the classical pathway of complement activation that may be explored for imaging, diagnostic, or therapeutic applications.


Subject(s)
Complement C4b , Single-Domain Antibodies , Animals , Complement Activation , Complement C3-C5 Convertases/metabolism , Cryoelectron Microscopy , Humans , Mammals
6.
Life Sci Alliance ; 3(11)2020 11.
Article in English | MEDLINE | ID: mdl-32958604

ABSTRACT

Tetraspanins are eukaryotic membrane proteins that contribute to a variety of signaling processes by organizing partner-receptor molecules in the plasma membrane. How tetraspanins bind and cluster partner receptors into tetraspanin-enriched microdomains is unknown. Here, we present crystal structures of the large extracellular loop of CD9 bound to nanobodies 4C8 and 4E8 and, the cryo-EM structure of 4C8-bound CD9 in complex with its partner EWI-F. CD9-EWI-F displays a tetrameric arrangement with two central EWI-F molecules, dimerized through their ectodomains, and two CD9 molecules, one bound to each EWI-F transmembrane helix through CD9-helices h3 and h4. In the crystal structures, nanobodies 4C8 and 4E8 bind CD9 at loops C and D, which is in agreement with the 4C8 conformation in the CD9-EWI-F complex. The complex varies from nearly twofold symmetric (with the two CD9 copies nearly anti-parallel) to ca. 50° bent arrangements. This flexible arrangement of CD9-EWI-F with potential CD9 homo-dimerization at either end provides a "concatenation model" for forming short linear or circular assemblies, which may explain the occurrence of tetraspanin-enriched microdomains.


Subject(s)
Tetraspanin 29/metabolism , Tetraspanin 29/ultrastructure , Antigens, CD/chemistry , Antigens, CD/metabolism , Antigens, CD/ultrastructure , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Membrane Glycoproteins/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Tetraspanin 28/metabolism , Tetraspanin 28/ultrastructure , Tetraspanin 29/physiology , Tetraspanins/metabolism , Tetraspanins/physiology , Tetraspanins/ultrastructure , Transcription Factors/metabolism
7.
Acc Chem Res ; 53(9): 1969-1980, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32815713

ABSTRACT

The ferric reductase superfamily comprises several oxidoreductases that use an intracellular electron source to reduce an extracellular acceptor substrate. NADPH oxidases (NOXs) and six-transmembrane epithelial antigen of the prostate enzymes (STEAPs) are iconic members of the superfamily. NOXs produce extracellular reactive oxygen species that exert potent bactericidal activities and trigger redox-signaling cascades that regulate cell division and differentiation. STEAPs catalyze the reduction of extracellular iron and copper which is necessary for the bioavailability of these essential elements. Both NOXs and STEAPs are present as multiple isozymes with distinct regulatory properties and physiological roles. Despite the important roles of NOXs and STEAPs in human physiology and despite their wide involvement in diseases like cancer, their mode of action at the molecular level remained incompletely understood for a long time, in part due to the absence of high-resolution models of the complete enzymes. Our two laboratories have elucidated the three-dimensional structures of NOXs and STEAPs, providing key insight into their mechanisms and evolution. The enzymes share a conserved transmembrane helical domain with an eye-catching hourglass shape. On the extracellular side, a heme prosthetic group is at the bottom of a pocket where the substrate (O2 in NOX, chelated iron or copper in STEAP) is reduced. On the intracellular side, the inner heme of NOX and the FAD of STEAP are bound to topological equivalent sites. This is a rare case where critical amino acid substitutions and local conformational changes enable a cofactor (heme vs FAD) swap between two structurally and functionally conserved scaffolds. The catalytic core of these enzymes is completed by distinct cytosolic NADPH-binding domains that are topologically unrelated (a ferredoxin reductase-like flavoprotein domain in NOX and a F420H2:NADP+-like domain in STEAP), feature different quaternary structures, and underlie specific regulatory mechanisms. Despite their differences, these domains all establish electron-transfer chains that direct the electrons from NADPH to the transmembrane domain. The multistep nature of the process and the chemical nature of the products pose considerable problems in the enzymatic assays. We learned that great care must be exerted in the validation of a candidate inhibitor. Multiple orthogonal assays are required to rule out off-target effects such as ROS-scavenging activities or nonspecific interference with the enzyme redox chain. The structural analysis of STEAP/NOX enzymes led us to further notice that their transmembrane heme-binding topology is shared by other enzymes. We found that the core domain of the cytochrome b subunits of the mitochondrial complex III and photosynthetic cytochrome b6f are closely related to NOXs and STEAPs and likely arose from the same ancestor protein. This observation expands the substrate portfolio of the superfamily since cytochromes b act on ubiquinone. The rigidly packed helices of the NOX/STEAP/cytochrome b domain contrast with the more malleable membrane proteins like ion channels or amino-acid transporters, which undergo large conformational changes to allow passage of relatively large metabolites. This notion of a rigid hourglass scaffold found an unexpected confirmation in the observation, revealed by structural comparisons, that an helical bundle identical to the NOX/STEAP/cytochrome b enzymes is featured by a de novo designed heme-binding protein, PS1. Apparently, nature and protein designers have independently converged to this fold as a versatile scaffold for heme-mediated reactions. The challenge is now to uncover the molecular mechanisms that implement the isozyme-specific regulation of the enzyme functions and develop much needed inhibitors and modulators for chemical biology and drug design studies.


Subject(s)
NADPH Oxidases/metabolism , Oxidoreductases/metabolism , Amino Acid Sequence , Binding Sites , Cyanobacteria/enzymology , Electron Transport , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Helix-Loop-Helix Motifs , Heme/chemistry , Heme/metabolism , Humans , NADP/chemistry , NADP/metabolism , NADPH Oxidases/chemistry , Oxidation-Reduction , Oxidoreductases/chemistry , Protein Structure, Tertiary , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , Sequence Alignment
8.
J Biol Chem ; 295(28): 9502-9512, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32409586

ABSTRACT

Six-transmembrane epithelial antigen of the prostate 1 (STEAP1) is an integral membrane protein that is highly up-regulated on the cell surface of several human cancers, making it a promising therapeutic target to manage these diseases. It shares sequence homology with three enzymes (STEAP2-STEAP4) that catalyze the NADPH-dependent reduction of iron(III). However, STEAP1 lacks an intracellular NADPH-binding domain and does not exhibit cellular ferric reductase activity. Thus, both the molecular function of STEAP1 and its role in cancer progression remain elusive. Here, we present a ∼3.0-Šcryo-EM structure of trimeric human STEAP1 bound to three antigen-binding fragments (Fabs) of the clinically used antibody mAb120.545. The structure revealed that STEAP1 adopts a reductase-like conformation and interacts with the Fabs through its extracellular helices. Enzymatic assays in human cells revealed that STEAP1 promotes iron(III) reduction when fused to the intracellular NADPH-binding domain of its family member STEAP4, suggesting that STEAP1 functions as a ferric reductase in STEAP heterotrimers. Our work provides a foundation for deciphering the molecular mechanisms of STEAP1 and may be useful in the design of new therapeutic strategies to target STEAP1 in cancer.


Subject(s)
Antigens, Neoplasm , Neoplasm Proteins , Neoplasms/enzymology , Oxidoreductases , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/ultrastructure , Antineoplastic Agents, Immunological/chemistry , Cryoelectron Microscopy , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Neoplasm Proteins/ultrastructure , Neoplasms/ultrastructure , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Oxidoreductases/ultrastructure , Protein Domains
9.
Nat Commun ; 9(1): 4337, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337524

ABSTRACT

Enzymes of the six-transmembrane epithelial antigen of the prostate (STEAP) family reduce Fe3+ and Cu2+ ions to facilitate metal-ion uptake by mammalian cells. STEAPs are highly upregulated in several types of cancer, making them potential therapeutic targets. However, the structural basis for STEAP-catalyzed electron transfer through an array of cofactors to metals at the membrane luminal side remains elusive. Here, we report cryo-electron microscopy structures of human STEAP4 in absence and presence of Fe3+-NTA. Domain-swapped, trimeric STEAP4 orients NADPH bound to a cytosolic domain onto axially aligned flavin-adenine dinucleotide (FAD) and a single b-type heme that cross the transmembrane-domain to enable electron transfer. Substrate binding within a positively charged ring indicates that iron gets reduced while in complex with its chelator. These molecular principles of iron reduction provide a basis for exploring STEAPs as therapeutic targets.


Subject(s)
Cryoelectron Microscopy , Iron/metabolism , Membrane Proteins/ultrastructure , Oxidoreductases/ultrastructure , Binding Sites , Biocatalysis , Electrons , Flavin-Adenine Dinucleotide/metabolism , Heme/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , NADP/metabolism , NADPH Oxidases/metabolism , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Domains , Substrate Specificity
10.
Nature ; 538(7623): 66-71, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27626375

ABSTRACT

P2X receptors are trimeric, non-selective cation channels activated by ATP that have important roles in the cardiovascular, neuronal and immune systems. Despite their central function in human physiology and although they are potential targets of therapeutic agents, there are no structures of human P2X receptors. The mechanisms of receptor desensitization and ion permeation, principles of antagonism, and complete structures of the pore-forming transmembrane domains of these receptors remain unclear. Here we report X-ray crystal structures of the human P2X3 receptor in apo/resting, agonist-bound/open-pore, agonist-bound/closed-pore/desensitized and antagonist-bound/closed states. The open state structure harbours an intracellular motif we term the 'cytoplasmic cap', which stabilizes the open state of the ion channel pore and creates lateral, phospholipid-lined cytoplasmic fenestrations for water and ion egress. The competitive antagonists TNP-ATP and A-317491 stabilize the apo/resting state and reveal the interactions responsible for competitive inhibition. These structures illuminate the conformational rearrangements that underlie P2X receptor gating and provide a foundation for the development of new pharmacological agents.


Subject(s)
Ion Channel Gating/drug effects , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X3/chemistry , Receptors, Purinergic P2X3/metabolism , Apoproteins/agonists , Apoproteins/antagonists & inhibitors , Apoproteins/chemistry , Apoproteins/metabolism , Binding Sites/drug effects , Binding, Competitive/drug effects , Crystallization , Crystallography, X-Ray , Humans , Ion Transport , Ligands , Models, Molecular , Porosity , Protein Conformation , Purinergic Agonists/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...