Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.810
Filter
Add more filters

Publication year range
1.
Cell ; 187(3): 782-796.e23, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38244547

ABSTRACT

The rapid kinetics of biological processes and associated short-lived conformational changes pose a significant challenge in attempts to structurally visualize biomolecules during a reaction in real time. Conventionally, on-pathway intermediates have been trapped using chemical modifications or reduced temperature, giving limited insights. Here, we introduce a time-resolved cryo-EM method using a reusable PDMS-based microfluidic chip assembly with high reactant mixing efficiency. Coating of PDMS walls with SiO2 virtually eliminates non-specific sample adsorption and ensures maintenance of the stoichiometry of the reaction, rendering it highly reproducible. In an operating range from 10 to 1,000 ms, the device allows us to follow in vitro reactions of biological molecules at resolution levels in the range of 3 Å. By employing this method, we show the mechanism of progressive HflX-mediated splitting of the 70S E. coli ribosome in the presence of the GTP via capture of three high-resolution reaction intermediates within 140 ms.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Ribosomes , Cryoelectron Microscopy/methods , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , GTP-Binding Proteins/metabolism , Microfluidics/methods , Ribosomes/metabolism , Silicon Dioxide/analysis
2.
Cell ; 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39173632

ABSTRACT

Horizontal gene transfer is a key driver of bacterial evolution, but it also presents severe risks to bacteria by introducing invasive mobile genetic elements. To counter these threats, bacteria have developed various defense systems, including prokaryotic Argonautes (pAgos) and the DNA defense module DdmDE system. Through biochemical analysis, structural determination, and in vivo plasmid clearance assays, we elucidate the assembly and activation mechanisms of DdmDE, which eliminates small, multicopy plasmids. We demonstrate that DdmE, a pAgo-like protein, acts as a catalytically inactive, DNA-guided, DNA-targeting defense module. In the presence of guide DNA, DdmE targets plasmids and recruits a dimeric DdmD, which contains nuclease and helicase domains. Upon binding to DNA substrates, DdmD transitions from an autoinhibited dimer to an active monomer, which then translocates along and cleaves the plasmids. Together, our findings reveal the intricate mechanisms underlying DdmDE-mediated plasmid clearance, offering fundamental insights into bacterial defense systems against plasmid invasions.

3.
Cell ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39197451

ABSTRACT

DNA polymerases are important drug targets, and many structural studies have captured them in distinct conformations. However, a detailed understanding of the impact of polymerase conformational dynamics on drug resistance is lacking. We determined cryoelectron microscopy (cryo-EM) structures of DNA-bound herpes simplex virus polymerase holoenzyme in multiple conformations and interacting with antivirals in clinical use. These structures reveal how the catalytic subunit Pol and the processivity factor UL42 bind DNA to promote processive DNA synthesis. Unexpectedly, in the absence of an incoming nucleotide, we observed Pol in multiple conformations with the closed state sampled by the fingers domain. Drug-bound structures reveal how antivirals may selectively bind enzymes that more readily adopt the closed conformation. Molecular dynamics simulations and the cryo-EM structure of a drug-resistant mutant indicate that some resistance mutations modulate conformational dynamics rather than directly impacting drug binding, thus clarifying mechanisms that drive drug selectivity.

4.
Cell ; 187(5): 1145-1159.e21, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38428394

ABSTRACT

Chloroplast genes encoding photosynthesis-associated proteins are predominantly transcribed by the plastid-encoded RNA polymerase (PEP). PEP is a multi-subunit complex composed of plastid-encoded subunits similar to bacterial RNA polymerases (RNAPs) stably bound to a set of nuclear-encoded PEP-associated proteins (PAPs). PAPs are essential to PEP activity and chloroplast biogenesis, but their roles are poorly defined. Here, we present cryoelectron microscopy (cryo-EM) structures of native 21-subunit PEP and a PEP transcription elongation complex from white mustard (Sinapis alba). We identify that PAPs encase the core polymerase, forming extensive interactions that likely promote complex assembly and stability. During elongation, PAPs interact with DNA downstream of the transcription bubble and with the nascent mRNA. The models reveal details of the superoxide dismutase, lysine methyltransferase, thioredoxin, and amino acid ligase enzymes that are subunits of PEP. Collectively, these data provide a foundation for the mechanistic understanding of chloroplast transcription and its role in plant growth and adaptation.


Subject(s)
DNA-Directed RNA Polymerases , Plastids , Arabidopsis Proteins/metabolism , Chloroplasts/metabolism , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/chemistry , Gene Expression Regulation, Plant , Plant Proteins/chemistry , Plastids/enzymology , Transcription, Genetic
5.
Cell ; 187(6): 1460-1475.e20, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38428423

ABSTRACT

Apelin is a key hormone in cardiovascular homeostasis that activates the apelin receptor (APLNR), which is regarded as a promising therapeutic target for cardiovascular disease. However, adverse effects through the ß-arrestin pathway limit its pharmacological use. Here, we report cryoelectron microscopy (cryo-EM) structures of APLNR-Gi1 complexes bound to three agonists with divergent signaling profiles. Combined with functional assays, we have identified "twin hotspots" in APLNR as key determinants for signaling bias, guiding the rational design of two exclusive G-protein-biased agonists WN353 and WN561. Cryo-EM structures of WN353- and WN561-stimulated APLNR-G protein complexes further confirm that the designed ligands adopt the desired poses. Pathophysiological experiments have provided evidence that WN561 demonstrates superior therapeutic effects against cardiac hypertrophy and reduced adverse effects compared with the established APLNR agonists. In summary, our designed APLNR modulator may facilitate the development of next-generation cardiovascular medications.


Subject(s)
Apelin Receptors , Cardiovascular Agents , Drug Design , Apelin Receptors/agonists , Apelin Receptors/chemistry , Apelin Receptors/ultrastructure , Cryoelectron Microscopy , GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Humans , Cardiovascular Agents/chemistry
6.
Cell ; 187(9): 2236-2249.e17, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38614100

ABSTRACT

Unlike those of double-stranded DNA (dsDNA), single-stranded DNA (ssDNA), and ssRNA viruses, the mechanism of genome packaging of dsRNA viruses is poorly understood. Here, we combined the techniques of high-resolution cryoelectron microscopy (cryo-EM), cellular cryoelectron tomography (cryo-ET), and structure-guided mutagenesis to investigate genome packaging and capsid assembly of bluetongue virus (BTV), a member of the Reoviridae family of dsRNA viruses. A total of eleven assembly states of BTV capsid were captured, with resolutions up to 2.8 Å, with most visualized in the host cytoplasm. ATPase VP6 was found underneath the vertices of capsid shell protein VP3 as an RNA-harboring pentamer, facilitating RNA packaging. RNA packaging expands the VP3 shell, which then engages middle- and outer-layer proteins to generate infectious virions. These revealed "duality" characteristics of the BTV assembly mechanism reconcile previous contradictory co-assembly and core-filling models and provide insights into the mysterious RNA packaging and capsid assembly of Reoviridae members and beyond.


Subject(s)
Bluetongue virus , Capsid Proteins , Capsid , Cryoelectron Microscopy , RNA, Viral , Viral Genome Packaging , Bluetongue virus/genetics , Bluetongue virus/physiology , Bluetongue virus/metabolism , Capsid/metabolism , Capsid/ultrastructure , Capsid Proteins/metabolism , Capsid Proteins/genetics , Capsid Proteins/chemistry , Animals , RNA, Viral/metabolism , RNA, Viral/genetics , Genome, Viral/genetics , Virus Assembly , Electron Microscope Tomography , Virion/metabolism , Virion/genetics , Virion/ultrastructure , Models, Molecular , Cell Line , Cricetinae
7.
Cell ; 187(14): 3726-3740.e43, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38861993

ABSTRACT

Many growth factors and cytokines signal by binding to the extracellular domains of their receptors and driving association and transphosphorylation of the receptor intracellular tyrosine kinase domains, initiating downstream signaling cascades. To enable systematic exploration of how receptor valency and geometry affect signaling outcomes, we designed cyclic homo-oligomers with up to 8 subunits using repeat protein building blocks that can be modularly extended. By incorporating a de novo-designed fibroblast growth factor receptor (FGFR)-binding module into these scaffolds, we generated a series of synthetic signaling ligands that exhibit potent valency- and geometry-dependent Ca2+ release and mitogen-activated protein kinase (MAPK) pathway activation. The high specificity of the designed agonists reveals distinct roles for two FGFR splice variants in driving arterial endothelium and perivascular cell fates during early vascular development. Our designed modular assemblies should be broadly useful for unraveling the complexities of signaling in key developmental transitions and for developing future therapeutic applications.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors , Receptors, Fibroblast Growth Factor , Signal Transduction , Animals , Humans , Receptors, Fibroblast Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Mice , Ligands , Calcium/metabolism , MAP Kinase Signaling System
8.
Cell ; 187(13): 3249-3261.e14, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38781968

ABSTRACT

Thermostable clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated (Cas9) enzymes could improve genome-editing efficiency and delivery due to extended protein lifetimes. However, initial experimentation demonstrated Geobacillus stearothermophilus Cas9 (GeoCas9) to be virtually inactive when used in cultured human cells. Laboratory-evolved variants of GeoCas9 overcome this natural limitation by acquiring mutations in the wedge (WED) domain that produce >100-fold-higher genome-editing levels. Cryoelectron microscopy (cryo-EM) structures of the wild-type and improved GeoCas9 (iGeoCas9) enzymes reveal extended contacts between the WED domain of iGeoCas9 and DNA substrates. Biochemical analysis shows that iGeoCas9 accelerates DNA unwinding to capture substrates under the magnesium-restricted conditions typical of mammalian but not bacterial cells. These findings enabled rational engineering of other Cas9 orthologs to enhance genome-editing levels, pointing to a general strategy for editing enzyme improvement. Together, these results uncover a new role for the Cas9 WED domain in DNA unwinding and demonstrate how accelerated target unwinding dramatically improves Cas9-induced genome-editing activity.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Cryoelectron Microscopy , DNA , Gene Editing , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , DNA/metabolism , DNA/genetics , Gene Editing/methods , Geobacillus stearothermophilus/genetics , Geobacillus stearothermophilus/metabolism , HEK293 Cells , Protein Domains , Genome, Human , Models, Molecular , Protein Structure, Tertiary , Nucleic Acid Conformation , Biocatalysis , Magnesium/chemistry , Magnesium/metabolism
9.
Cell ; 187(14): 3638-3651.e18, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38838667

ABSTRACT

Telomere maintenance requires the extension of the G-rich telomeric repeat strand by telomerase and the fill-in synthesis of the C-rich strand by Polα/primase. At telomeres, Polα/primase is bound to Ctc1/Stn1/Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Our findings suggest that POT1 hinge phosphorylation is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/primase in an inactive, autoinhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/primase into an active state that completes telomere replication through fill-in synthesis.


Subject(s)
DNA Polymerase I , Shelterin Complex , Telomere-Binding Proteins , Telomere , Humans , Cryoelectron Microscopy , DNA Polymerase I/metabolism , DNA Primase/metabolism , DNA Primase/genetics , Models, Molecular , Phosphorylation , Shelterin Complex/metabolism , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism
10.
Cell ; 187(16): 4213-4230.e19, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39013471

ABSTRACT

Foamy viruses (FVs) are an ancient lineage of retroviruses, with an evolutionary history spanning over 450 million years. Vector systems based on Prototype Foamy Virus (PFV) are promising candidates for gene and oncolytic therapies. Structural studies of PFV contribute to the understanding of the mechanisms of FV replication, cell entry and infection, and retroviral evolution. Here we combine cryoEM and cryoET to determine high-resolution in situ structures of the PFV icosahedral capsid (CA) and envelope glycoprotein (Env), including its type III transmembrane anchor and membrane-proximal external region (MPER), and show how they are organized in an integrated structure of assembled PFV particles. The atomic models reveal an ancient retroviral capsid architecture and an unexpected relationship between Env and other class 1 fusion proteins of the Mononegavirales. Our results represent the de novo structure determination of an assembled retrovirus particle.


Subject(s)
Cryoelectron Microscopy , Spumavirus , Virus Assembly , Virus Internalization , Spumavirus/genetics , Capsid/metabolism , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid Proteins/genetics , Humans , Evolution, Molecular , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Models, Molecular
11.
Cell ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39197449

ABSTRACT

The protein import motor in chloroplasts plays a pivotal role in their biogenesis and homeostasis by driving the translocation of preproteins into chloroplasts. While the Ycf2-FtsHi complex serves as the import motor in land plants, its evolutionary conservation, specialization, and mechanisms across photosynthetic organisms are largely unexplored. Here, we isolated and determined the cryogenic electron microscopy (cryo-EM) structures of the native Ycf2-FtsHi complex from Chlamydomonas reinhardtii, uncovering a complex composed of up to 19 subunits, including multiple green-algae-specific components. The heterohexameric AAA+ ATPase motor module is tilted, potentially facilitating preprotein handover from the translocon at the inner chloroplast membrane (TIC) complex. Preprotein interacts with Ycf2-FtsHi and enhances its ATPase activity in vitro. Integrating Ycf2-FtsHi and translocon at the outer chloroplast membrane (TOC)-TIC supercomplex structures reveals insights into their physical and functional interplay during preprotein translocation. By comparing these findings with those from land plants, our study establishes a structural foundation for understanding the assembly, function, evolutionary conservation, and diversity of chloroplast protein import motors.

12.
Cell ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39197452

ABSTRACT

Chloroplast proteins are imported via the translocon at the outer chloroplast membrane (TOC)-translocon at the inner chloroplast membrane (TIC) supercomplex, driven by an ATPase motor. The Ycf2-FtsHi complex has been identified as the chloroplast import motor. However, its assembly and cooperation with the TIC complex during preprotein translocation remain unclear. Here, we present the structures of the Ycf2-FtsHi and TIC complexes from Arabidopsis and an ultracomplex formed between them from Pisum. The Ycf2-FtsHi structure reveals a heterohexameric AAA+ ATPase motor module with characteristic features. Four previously uncharacterized components of Ycf2-FtsHi were identified, which aid in complex assembly and anchoring of the motor module at a tilted angle relative to the membrane. When considering the structures of the TIC complex and the TIC-Ycf2-FtsHi ultracomplex together, it becomes evident that the tilted motor module of Ycf2-FtsHi enables its close contact with the TIC complex, thereby facilitating efficient preprotein translocation. Our study provides valuable structural insights into the chloroplast protein import process in land plants.

13.
Cell ; 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39208798

ABSTRACT

We use cryoelectron microscopy (cryo-EM) as a sequence- and culture-independent diagnostic tool to identify the etiological agent of an agricultural pandemic. For the past 4 years, American insect-rearing facilities have experienced a distinctive larval pathology and colony collapse of farmed Zophobas morio (superworm). By means of cryo-EM, we discovered the causative agent: a densovirus that we named Zophobas morio black wasting virus (ZmBWV). We confirmed the etiology of disease by fulfilling Koch's postulates and characterizing strains from across the United States. ZmBWV is a member of the family Parvoviridae with a 5,542 nt genome, and we describe intersubunit interactions explaining its expanded internal volume relative to human parvoviruses. Cryo-EM structures at resolutions up to 2.1 Å revealed single-strand DNA (ssDNA) ordering at the capsid inner surface pinned by base-binding pockets in the capsid inner surface. Also, we demonstrated the prophylactic potential of non-pathogenic strains to provide cross-protection in vivo.

14.
Cell ; 187(18): 4996-5009.e14, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38996527

ABSTRACT

Latent membrane protein 1 (LMP1) is the primary oncoprotein of Epstein-Barr virus (EBV) and plays versatile roles in the EBV life cycle and pathogenesis. Despite decades of extensive research, the molecular basis for LMP1 folding, assembly, and activation remains unclear. Here, we report cryo-electron microscopy structures of LMP1 in two unexpected assemblies: a symmetric homodimer and a higher-order filamentous oligomer. LMP1 adopts a non-canonical and unpredicted fold that supports the formation of a stable homodimer through tight and antiparallel intermolecular packing. LMP1 dimers further assemble side-by-side into higher-order filamentous oligomers, thereby allowing the accumulation and specific organization of the flexible cytoplasmic tails for efficient recruitment of downstream factors. Super-resolution microscopy and cellular functional assays demonstrate that mutations at both dimeric and oligomeric interfaces disrupt LMP1 higher-order assembly and block multiple LMP1-mediated signaling pathways. Our research provides a framework for understanding the mechanism of LMP1 and for developing potential therapies targeting EBV-associated diseases.


Subject(s)
Cryoelectron Microscopy , Herpesvirus 4, Human , Protein Multimerization , Viral Matrix Proteins , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Humans , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Models, Molecular , HEK293 Cells , Signal Transduction , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Mutation
15.
Cell ; 187(5): 1296-1311.e26, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38428397

ABSTRACT

Most membrane proteins are modified by covalent addition of complex sugars through N- and O-glycosylation. Unlike proteins, glycans do not typically adopt specific secondary structures and remain very mobile, shielding potentially large fractions of protein surface. High glycan conformational freedom hinders complete structural elucidation of glycoproteins. Computer simulations may be used to model glycosylated proteins but require hundreds of thousands of computing hours on supercomputers, thus limiting routine use. Here, we describe GlycoSHIELD, a reductionist method that can be implemented on personal computers to graft realistic ensembles of glycan conformers onto static protein structures in minutes. Using molecular dynamics simulation, small-angle X-ray scattering, cryoelectron microscopy, and mass spectrometry, we show that this open-access toolkit provides enhanced models of glycoprotein structures. Focusing on N-cadherin, human coronavirus spike proteins, and gamma-aminobutyric acid receptors, we show that GlycoSHIELD can shed light on the impact of glycans on the conformation and activity of complex glycoproteins.


Subject(s)
Glycoproteins , Molecular Dynamics Simulation , Humans , Cryoelectron Microscopy , Glycoproteins/chemistry , Glycosylation , Polysaccharides/chemistry
16.
Cell ; 187(7): 1733-1744.e12, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38552612

ABSTRACT

Mastigonemes, the hair-like lateral appendages lining cilia or flagella, participate in mechanosensation and cellular motion, but their constituents and structure have remained unclear. Here, we report the cryo-EM structure of native mastigonemes isolated from Chlamydomonas at 3.0 Å resolution. The long stem assembles as a super spiral, with each helical turn comprising four pairs of anti-parallel mastigoneme-like protein 1 (Mst1). A large array of arabinoglycans, which represents a common class of glycosylation in plants and algae, is resolved surrounding the type II poly-hydroxyproline (Hyp) helix in Mst1. The EM map unveils a mastigoneme axial protein (Mstax) that is rich in heavily glycosylated Hyp and contains a PKD2-like transmembrane domain (TMD). Mstax, with nearly 8,000 residues spanning from the intracellular region to the distal end of the mastigoneme, provides the framework for Mst1 assembly. Our study provides insights into the complexity of protein and glycan interactions in native bio-architectures.


Subject(s)
Chlamydomonas , Cilia , Chlamydomonas/cytology , Cilia/chemistry , Cilia/ultrastructure , Flagella , Polysaccharides , Proteins
17.
Cell ; 187(8): 1907-1921.e16, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38552624

ABSTRACT

Hydroxyproline-rich glycoproteins (HRGPs) are a ubiquitous class of protein in the extracellular matrices and cell walls of plants and algae, yet little is known of their native structures or interactions. Here, we used electron cryomicroscopy (cryo-EM) to determine the structure of the hydroxyproline-rich mastigoneme, an extracellular filament isolated from the cilia of the alga Chlamydomonas reinhardtii. The structure demonstrates that mastigonemes are formed from two HRGPs (a filament of MST1 wrapped around a single copy of MST3) that both have hyperglycosylated poly(hydroxyproline) helices. Within the helices, O-linked glycosylation of the hydroxyproline residues and O-galactosylation of interspersed serine residues create a carbohydrate casing. Analysis of the associated glycans reveals how the pattern of hydroxyproline repetition determines the type and extent of glycosylation. MST3 possesses a PKD2-like transmembrane domain that forms a heteromeric polycystin-like cation channel with PKD2 and SIP, explaining how mastigonemes are tethered to ciliary membranes.


Subject(s)
Chlamydomonas reinhardtii , Cilia , Glycoproteins , Cilia/chemistry , Glycoproteins/chemistry , Glycosylation , Hydroxyproline/chemistry , Plants/metabolism , Chlamydomonas reinhardtii/chemistry
18.
Cell ; 187(5): 1127-1144.e21, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38428393

ABSTRACT

Chloroplasts are green plastids in the cytoplasm of eukaryotic algae and plants responsible for photosynthesis. The plastid-encoded RNA polymerase (PEP) plays an essential role during chloroplast biogenesis from proplastids and functions as the predominant RNA polymerase in mature chloroplasts. The PEP-centered transcription apparatus comprises a bacterial-origin PEP core and more than a dozen eukaryotic-origin PEP-associated proteins (PAPs) encoded in the nucleus. Here, we determined the cryo-EM structures of Nicotiana tabacum (tobacco) PEP-PAP apoenzyme and PEP-PAP transcription elongation complexes at near-atomic resolutions. Our data show the PEP core adopts a typical fold as bacterial RNAP. Fifteen PAPs bind at the periphery of the PEP core, facilitate assembling the PEP-PAP supercomplex, protect the complex from oxidation damage, and likely couple gene transcription with RNA processing. Our results report the high-resolution architecture of the chloroplast transcription apparatus and provide the structural basis for the mechanistic and functional study of transcription regulation in chloroplasts.


Subject(s)
DNA-Directed RNA Polymerases , Plastids , Chloroplasts/metabolism , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/genetics , Nicotiana/genetics , Photosynthesis , Plastids/enzymology
19.
Cell ; 187(9): 2224-2235.e16, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38614101

ABSTRACT

The membrane protein NINJ1 mediates plasma membrane rupture in pyroptosis and other lytic cell death pathways. Here, we report the cryo-EM structure of a NINJ1 oligomer segmented from NINJ1 rings. Each NINJ1 subunit comprises amphipathic (⍺1, ⍺2) and transmembrane (TM) helices (⍺3, ⍺4) and forms a chain of subunits, mainly by the TM helices and ⍺1. ⍺3 and ⍺4 are kinked, and the Gly residues are important for function. The NINJ1 oligomer possesses a concave hydrophobic side that should face the membrane and a convex hydrophilic side formed by ⍺1 and ⍺2, presumably upon activation. This structural observation suggests that NINJ1 can form membrane disks, consistent with membrane fragmentation by recombinant NINJ1. Live-cell and super-resolution imaging uncover ring-like structures on the plasma membrane that are released into the culture supernatant. Released NINJ1 encircles a membrane inside, as shown by lipid staining. Therefore, NINJ1-mediated membrane disk formation is different from gasdermin-mediated pore formation, resulting in membrane loss and plasma membrane rupture.


Subject(s)
Cell Adhesion Molecules, Neuronal , Cell Membrane , Cryoelectron Microscopy , Cell Membrane/metabolism , Humans , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/chemistry , Animals , Mice , HEK293 Cells , Pyroptosis , Models, Molecular , Membrane Proteins/metabolism , Membrane Proteins/chemistry , Phosphate-Binding Proteins/metabolism
20.
Cell ; 187(5): 1160-1176.e21, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38382524

ABSTRACT

The α7 nicotinic acetylcholine receptor is a pentameric ligand-gated ion channel that plays an important role in cholinergic signaling throughout the nervous system. Its unique physiological characteristics and implications in neurological disorders and inflammation make it a promising but challenging therapeutic target. Positive allosteric modulators overcome limitations of traditional α7 agonists, but their potentiation mechanisms remain unclear. Here, we present high-resolution structures of α7-modulator complexes, revealing partially overlapping binding sites but varying conformational states. Structure-guided functional and computational tests suggest that differences in modulator activity arise from the stable rotation of a channel gating residue out of the pore. We extend the study using a time-resolved cryoelectron microscopy (cryo-EM) approach to reveal asymmetric state transitions for this homomeric channel and also find that a modulator with allosteric agonist activity exploits a distinct channel-gating mechanism. These results define mechanisms of α7 allosteric modulation and activation with implications across the pentameric receptor superfamily.


Subject(s)
alpha7 Nicotinic Acetylcholine Receptor , Humans , alpha7 Nicotinic Acetylcholine Receptor/chemistry , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/ultrastructure , Binding Sites , Cryoelectron Microscopy , Inflammation/drug therapy , Signal Transduction , Allosteric Regulation
SELECTION OF CITATIONS
SEARCH DETAIL