Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.857
Filter
Add more filters

Publication year range
1.
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
2.
Cell ; 185(4): 585-602.e29, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35051368

ABSTRACT

The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8+ T cells, thereby augmenting calcium flux, signal transduction, metabolic reprogramming, immune synapse formation, and, as a consequence, specific cytotoxicity. Accordingly, magnesium-sufficiency sensed via LFA-1 translated to the superior performance of pathogen- and tumor-specific T cells, enhanced effectiveness of bi-specific T cell engaging antibodies, and improved CAR T cell function. Clinically, low serum magnesium levels were associated with more rapid disease progression and shorter overall survival in CAR T cell and immune checkpoint antibody-treated patients. LFA-1 thus directly incorporates information on the composition of the microenvironment as a determinant of outside-in signaling activity. These findings conceptually link co-stimulation and nutrient sensing and point to the magnesium-LFA-1 axis as a therapeutically amenable biologic system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/metabolism , Animals , Bacterial Infections/immunology , Caloric Restriction , Cell Line, Tumor , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Immunologic Memory , Immunological Synapses/metabolism , Immunotherapy , Lymphocyte Activation/immunology , MAP Kinase Signaling System , Magnesium/administration & dosage , Male , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism
3.
Cell ; 183(2): 474-489.e17, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33035451

ABSTRACT

Mg2+ is the most abundant divalent cation in metazoans and an essential cofactor for ATP, nucleic acids, and countless metabolic enzymes. To understand how the spatio-temporal dynamics of intracellular Mg2+ (iMg2+) are integrated into cellular signaling, we implemented a comprehensive screen to discover regulators of iMg2+ dynamics. Lactate emerged as an activator of rapid release of Mg2+ from endoplasmic reticulum (ER) stores, which facilitates mitochondrial Mg2+ (mMg2+) uptake in multiple cell types. We demonstrate that this process is remarkably temperature sensitive and mediated through intracellular but not extracellular signals. The ER-mitochondrial Mg2+ dynamics is selectively stimulated by L-lactate. Further, we show that lactate-mediated mMg2+ entry is facilitated by Mrs2, and point mutations in the intermembrane space loop limits mMg2+ uptake. Intriguingly, suppression of mMg2+ surge alleviates inflammation-induced multi-organ failure. Together, these findings reveal that lactate mobilizes iMg2+ and links the mMg2+ transport machinery with major metabolic feedback circuits and mitochondrial bioenergetics.


Subject(s)
Endoplasmic Reticulum/metabolism , Lactic Acid/metabolism , Magnesium/metabolism , Animals , COS Cells , Calcium/metabolism , Calcium Signaling/physiology , Chlorocebus aethiops , Endoplasmic Reticulum/physiology , Female , HeLa Cells , Hep G2 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism
4.
Cell ; 182(6): 1560-1573.e13, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32783916

ABSTRACT

SARS-CoV-2 is the causative agent of the 2019-2020 pandemic. The SARS-CoV-2 genome is replicated and transcribed by the RNA-dependent RNA polymerase holoenzyme (subunits nsp7/nsp82/nsp12) along with a cast of accessory factors. One of these factors is the nsp13 helicase. Both the holo-RdRp and nsp13 are essential for viral replication and are targets for treating the disease COVID-19. Here we present cryoelectron microscopic structures of the SARS-CoV-2 holo-RdRp with an RNA template product in complex with two molecules of the nsp13 helicase. The Nidovirales order-specific N-terminal domains of each nsp13 interact with the N-terminal extension of each copy of nsp8. One nsp13 also contacts the nsp12 thumb. The structure places the nucleic acid-binding ATPase domains of the helicase directly in front of the replicating-transcribing holo-RdRp, constraining models for nsp13 function. We also observe ADP-Mg2+ bound in the nsp12 N-terminal nidovirus RdRp-associated nucleotidyltransferase domain, detailing a new pocket for anti-viral therapy development.


Subject(s)
Methyltransferases/chemistry , RNA Helicases/chemistry , RNA-Dependent RNA Polymerase/chemistry , Viral Nonstructural Proteins/chemistry , Virus Replication , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Betacoronavirus/genetics , Betacoronavirus/metabolism , Betacoronavirus/ultrastructure , Binding Sites , Coronavirus RNA-Dependent RNA Polymerase , Cryoelectron Microscopy , Holoenzymes/chemistry , Holoenzymes/metabolism , Magnesium/metabolism , Methyltransferases/metabolism , Protein Binding , RNA Helicases/metabolism , RNA, Viral/chemistry , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2 , Viral Nonstructural Proteins/metabolism
5.
Cell ; 177(2): 352-360.e13, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30853217

ABSTRACT

Bacteria exhibit cell-to-cell variability in their resilience to stress, for example, following antibiotic exposure. Higher resilience is typically ascribed to "dormant" non-growing cellular states. Here, by measuring membrane potential dynamics of Bacillus subtilis cells, we show that actively growing bacteria can cope with ribosome-targeting antibiotics through an alternative mechanism based on ion flux modulation. Specifically, we observed two types of cellular behavior: growth-defective cells exhibited a mathematically predicted transient increase in membrane potential (hyperpolarization), followed by cell death, whereas growing cells lacked hyperpolarization events and showed elevated survival. Using structural perturbations of the ribosome and proteomic analysis, we uncovered that stress resilience arises from magnesium influx, which prevents hyperpolarization. Thus, ion flux modulation provides a distinct mechanism to cope with ribosomal stress. These results suggest new approaches to increase the effectiveness of ribosome-targeting antibiotics and reveal an intriguing connection between ribosomes and the membrane potential, two fundamental properties of cells.


Subject(s)
Bacterial Outer Membrane/metabolism , Magnesium/metabolism , Ribosomes/metabolism , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Proteomics , Ribosomal Proteins/metabolism
6.
Cell ; 173(1): 130-139.e10, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29526461

ABSTRACT

Endogenous circadian rhythms are thought to modulate responses to external factors, but mechanisms that confer time-of-day differences in organismal responses to environmental insults/therapeutic treatments are poorly understood. Using a xenobiotic, we find that permeability of the Drosophila "blood"-brain barrier (BBB) is higher at night. The permeability rhythm is driven by circadian regulation of efflux and depends on a molecular clock in the perineurial glia of the BBB, although efflux transporters are restricted to subperineurial glia (SPG). We show that transmission of circadian signals across the layers requires cyclically expressed gap junctions. Specifically, during nighttime, gap junctions reduce intracellular magnesium ([Mg2+]i), a positive regulator of efflux, in SPG. Consistent with lower nighttime efflux, nighttime administration of the anti-epileptic phenytoin is more effective at treating a Drosophila seizure model. These findings identify a novel mechanism of circadian regulation and have therapeutic implications for drugs targeted to the central nervous system.


Subject(s)
Blood-Brain Barrier/metabolism , Circadian Clocks , Drosophila/metabolism , Rhodamines/metabolism , Xenobiotics/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Blood-Brain Barrier/drug effects , Brain/metabolism , Circadian Clocks/drug effects , Connexins/metabolism , Drosophila Proteins/metabolism , Female , Gap Junctions/metabolism , Magnesium/metabolism , Neuroglia/metabolism , Phenytoin/pharmacology , Phenytoin/therapeutic use , Seizures/drug therapy , Seizures/pathology , Seizures/veterinary
7.
Cell ; 164(4): 747-56, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26871634

ABSTRACT

CorA, the major Mg(2+) uptake system in prokaryotes, is gated by intracellular Mg(2+) (KD ∼ 1-2 mM). X-ray crystallographic studies of CorA show similar conformations under Mg(2+)-bound and Mg(2+)-free conditions, but EPR spectroscopic studies reveal large Mg(2+)-driven quaternary conformational changes. Here, we determined cryo-EM structures of CorA in the Mg(2+)-bound closed conformation and in two open Mg(2+)-free states at resolutions of 3.8, 7.1, and 7.1 Å, respectively. In the absence of bound Mg(2+), four of the five subunits are displaced to variable extents (∼ 10-25 Å) by hinge-like motions as large as ∼ 35° at the stalk helix. The transition between a single 5-fold symmetric closed state and an ensemble of low Mg(2+), open, asymmetric conformational states is, thus, the key structural signature of CorA gating. This mechanism is likely to apply to other structurally similar divalent ion channels.


Subject(s)
Bacterial Proteins/ultrastructure , Cation Transport Proteins/ultrastructure , Magnesium/metabolism , Thermotoga maritima/chemistry , Bacterial Proteins/chemistry , Cation Transport Proteins/chemistry , Cryoelectron Microscopy , Models, Molecular , Molecular Dynamics Simulation
8.
Cell ; 164(4): 597-8, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26871624

ABSTRACT

Ligand binding usually moves the target protein from an ensemble of inactive states to a well-defined active conformation. Matthies et al. flip this scheme around, finding that, for the magnesium channel CorA, loss of ligand binding induces an ensemble of conformations that turn the channel on.


Subject(s)
Bacterial Proteins/ultrastructure , Cation Transport Proteins/ultrastructure , Magnesium/metabolism , Thermotoga maritima/chemistry
9.
Nature ; 629(8011): 467-473, 2024 May.
Article in English | MEDLINE | ID: mdl-38471529

ABSTRACT

Prokaryotes have evolved intricate innate immune systems against phage infection1-7. Gabija is a highly widespread prokaryotic defence system that consists of two components, GajA and GajB8. GajA functions as a DNA endonuclease that is inactive in the presence of ATP9. Here, to explore how the Gabija system is activated for anti-phage defence, we report its cryo-electron microscopy structures in five states, including apo GajA, GajA in complex with DNA, GajA bound by ATP, apo GajA-GajB, and GajA-GajB in complex with ATP and Mg2+. GajA is a rhombus-shaped tetramer with its ATPase domain clustered at the centre and the topoisomerase-primase (Toprim) domain located peripherally. ATP binding at the ATPase domain stabilizes the insertion region within the ATPase domain, keeping the Toprim domain in a closed state. Upon ATP depletion by phages, the Toprim domain opens to bind and cleave the DNA substrate. GajB, which docks on GajA, is activated by the cleaved DNA, ultimately leading to prokaryotic cell death. Our study presents a mechanistic landscape of Gabija activation.


Subject(s)
Bacillus cereus , Bacterial Proteins , Bacteriophages , Cryoelectron Microscopy , Immunity, Innate , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/ultrastructure , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Apoproteins/chemistry , Apoproteins/immunology , Apoproteins/metabolism , Apoproteins/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Bacteriophages/immunology , DNA/metabolism , DNA/chemistry , DNA Cleavage , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Protein Binding , Protein Domains , Microbial Viability , Bacillus cereus/chemistry , Bacillus cereus/immunology , Bacillus cereus/metabolism , Bacillus cereus/ultrastructure , Protein Structure, Quaternary , DNA Primase/chemistry , DNA Primase/metabolism , DNA Primase/ultrastructure , DNA Topoisomerases/chemistry , DNA Topoisomerases/metabolism , DNA Topoisomerases/ultrastructure
10.
Mol Cell ; 80(5): 762-763, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33275887

ABSTRACT

Lactate initiates Mg2+ release from the ER and subsequent uptake by the mitochondria.


Subject(s)
Lactic Acid , Magnesium , Biological Transport , Lactic Acid/metabolism , Magnesium/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics
11.
PLoS Biol ; 22(4): e3002560, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38574172

ABSTRACT

In all domains of life, Hsp70 chaperones preserve protein homeostasis by promoting protein folding and degradation and preventing protein aggregation. We now report that the Hsp70 from the bacterial pathogen Salmonella enterica serovar Typhimurium-termed DnaK-independently reduces protein synthesis in vitro and in S. Typhimurium facing cytoplasmic Mg2+ starvation, a condition encountered during infection. This reduction reflects a 3-fold increase in ribosome association with DnaK and a 30-fold decrease in ribosome association with trigger factor, the chaperone normally associated with translating ribosomes. Surprisingly, this reduction does not involve J-domain cochaperones, unlike previously known functions of DnaK. Removing the 74 C-terminal amino acids of the 638-residue long DnaK impeded DnaK association with ribosomes and reduction of protein synthesis, rendering S. Typhimurium defective in protein homeostasis during cytoplasmic Mg2+ starvation. DnaK-dependent reduction in protein synthesis is critical for survival against Mg2+ starvation because inhibiting protein synthesis in a dnaK-independent manner overcame the 10,000-fold loss in viability resulting from DnaK truncation. Our results indicate that DnaK protects bacteria from infection-relevant stresses by coordinating protein synthesis with protein folding capacity.


Subject(s)
Escherichia coli Proteins , Magnesium , Magnesium/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Folding , Bacteria/metabolism , Salmonella
12.
PLoS Biol ; 22(6): e3002694, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38900845

ABSTRACT

Fungi and bacteria coexist in many polymicrobial communities, yet the molecular basis of their interactions remains poorly understood. Here, we show that the fungus Candida albicans sequesters essential magnesium ions from the bacterium Pseudomonas aeruginosa. To counteract fungal Mg2+ sequestration, P. aeruginosa expresses the Mg2+ transporter MgtA when Mg2+ levels are low. Thus, loss of MgtA specifically impairs P. aeruginosa in co-culture with C. albicans, but fitness can be restored by supplementing Mg2+. Using a panel of fungi and bacteria, we show that Mg2+ sequestration is a general mechanism of fungal antagonism against gram-negative bacteria. Mg2+ limitation enhances bacterial resistance to polymyxin antibiotics like colistin, which target gram-negative bacterial membranes. Indeed, experimental evolution reveals that P. aeruginosa evolves C. albicans-dependent colistin resistance via non-canonical means; antifungal treatment renders resistant bacteria colistin-sensitive. Our work suggests that fungal-bacterial competition could profoundly impact polymicrobial infection treatment with antibiotics of last resort.


Subject(s)
Anti-Bacterial Agents , Candida albicans , Colistin , Magnesium , Pseudomonas aeruginosa , Magnesium/pharmacology , Magnesium/metabolism , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/pharmacology , Candida albicans/drug effects , Candida albicans/metabolism , Colistin/pharmacology , Microbial Sensitivity Tests , Polymyxins/pharmacology , Drug Resistance, Bacterial/drug effects , Microbial Interactions/drug effects
13.
Mol Cell ; 76(1): 44-56.e3, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31444105

ABSTRACT

Endonuclease V (EndoV) cleaves the second phosphodiester bond 3' to a deaminated adenosine (inosine). Although highly conserved, EndoV homologs change substrate preference from DNA in bacteria to RNA in eukaryotes. We have characterized EndoV from six different species and determined crystal structures of human EndoV and three EndoV homologs from bacteria to mouse in complex with inosine-containing DNA/RNA hybrid or double-stranded RNA (dsRNA). Inosine recognition is conserved, but changes in several connecting loops in eukaryotic EndoV confer recognition of 3 ribonucleotides upstream and 7 or 8 bp of dsRNA downstream of the cleavage site, and bacterial EndoV binds only 2 or 3 nt flanking the scissile phosphate. In addition to the two canonical metal ions in the active site, a third Mn2+ that coordinates the nucleophilic water appears necessary for product formation. Comparison of EndoV with its homologs RNase H1 and Argonaute reveals the principles by which these enzymes recognize RNA versus DNA.


Subject(s)
Bacterial Proteins/metabolism , DNA Repair , DNA, Bacterial/metabolism , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Evolution, Molecular , Inosine/metabolism , RNA/metabolism , Ribonuclease H/metabolism , Animals , Argonaute Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Deoxyribonuclease (Pyrimidine Dimer)/chemistry , Deoxyribonuclease (Pyrimidine Dimer)/genetics , Humans , Magnesium/metabolism , Manganese/metabolism , Mice , Nucleic Acid Conformation , Protein Conformation , RNA/chemistry , RNA/genetics , Ribonuclease H/chemistry , Ribonuclease H/genetics , Structure-Activity Relationship , Substrate Specificity
14.
Proc Natl Acad Sci U S A ; 121(37): e2407455121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39240971

ABSTRACT

Succinic acid (SA), a dicarboxylic acid of industrial importance, can be efficiently produced by metabolically engineered Mannheimia succiniciproducens. Although the importance of magnesium (Mg2+) ion on SA production has been evident from our previous studies, the role of Mg2+ ion remains largely unexplored. In this study, we investigated the impact of Mg2+ ion on SA production and developed a hyper-SA producing strain of M. succiniciproducens by reconstructing the Mg2+ ion transport system. To achieve this, optimal alkaline neutralizer comprising Mg2+ ion was developed and the physiological effect of Mg2+ ion was analyzed. Subsequently, the Mg2+ ion transport system was reconstructed by introducing an efficient Mg2+ ion transporter from Salmonella enterica. A high-inoculum fed-batch fermentation of the final engineered strain produced 152.23 ± 0.99 g/L of SA, with a maximum productivity of 39.64 ± 0.69 g/L/h. These findings highlight the importance of Mg2+ ions and transportation system optimization in succinic acid production by M. succiniciproducens.


Subject(s)
Fermentation , Magnesium , Mannheimia , Succinic Acid , Succinic Acid/metabolism , Magnesium/metabolism , Mannheimia/metabolism , Mannheimia/genetics , Metabolic Engineering/methods , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics
15.
Proc Natl Acad Sci U S A ; 121(36): e2318527121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190355

ABSTRACT

Catalysis and translocation of multisubunit DNA-directed RNA polymerases underlie all cellular mRNA synthesis. RNA polymerase II (Pol II) synthesizes eukaryotic pre-mRNAs from a DNA template strand buried in its active site. Structural details of catalysis at near-atomic resolution and precise arrangement of key active site components have been elusive. Here, we present the free-electron laser (FEL) structures of a matched ATP-bound Pol II and the hyperactive Rpb1 T834P bridge helix (BH) mutant at the highest resolution to date. The radiation-damage-free FEL structures reveal the full active site interaction network, including the trigger loop (TL) in the closed conformation, bonafide occupancy of both site A and B Mg2+, and, more importantly, a putative third (site C) Mg2+ analogous to that described for some DNA polymerases but not observed previously for cellular RNA polymerases. Molecular dynamics (MD) simulations of the structures indicate that the third Mg2+ is coordinated and stabilized at its observed position. TL residues provide half of the substrate binding pocket while multiple TL/BH interactions induce conformational changes that could allow translocation upon substrate hydrolysis. Consistent with TL/BH communication, a FEL structure and MD simulations of the T834P mutant reveal rearrangement of some active site interactions supporting potential plasticity in active site function and long-distance effects on both the width of the central channel and TL conformation, likely underlying its increased elongation rate at the expense of fidelity.


Subject(s)
Catalytic Domain , Magnesium , Molecular Dynamics Simulation , RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , Magnesium/metabolism , Magnesium/chemistry , Lasers , Protein Conformation , Electrons , Protein Binding , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Binding Sites
16.
Proc Natl Acad Sci U S A ; 121(29): e2408156121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38980907

ABSTRACT

After ATP-actin monomers assemble filaments, the ATP's [Formula: see text]-phosphate is hydrolyzedwithin seconds and dissociates over minutes. We used all-atom molecular dynamics simulations to sample the release of phosphate from filaments and study residues that gate release. Dissociation of phosphate from Mg2+ is rate limiting and associated with an energy barrier of 20 kcal/mol, consistent with experimental rates of phosphate release. Phosphate then diffuses within an internal cavity toward a gate formed by R177, as suggested in prior computational studies and cryo-EM structures. The gate is closed when R177 hydrogen bonds with N111 and is open when R177 forms a salt bridge with D179. Most of the time, interactions of R177 with other residues occlude the phosphate release pathway. Machine learning analysis reveals that the occluding interactions fluctuate rapidly, underscoring the secondary role of backdoor gate opening in Pi release, in contrast with the previous hypothesis that gate opening is the primary event.


Subject(s)
Actin Cytoskeleton , Adenosine Triphosphate , Molecular Dynamics Simulation , Phosphates , Phosphates/metabolism , Phosphates/chemistry , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/chemistry , Adenosine Triphosphate/metabolism , Actins/metabolism , Actins/chemistry , Hydrogen Bonding , Magnesium/metabolism , Magnesium/chemistry , Cryoelectron Microscopy
17.
RNA ; 30(7): 770-778, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38570183

ABSTRACT

30S subunits become inactive upon exposure to low Mg2+ concentration, because of a reversible conformational change that entails nucleotides (nt) in the neck helix (h28) and 3' tail of 16S rRNA. This active-to-inactive transition involves partial unwinding of h28 and repairing of nt 921-923 with nt 1532-1534, which requires flipping of the 3' tail by ∼180°. Growing evidence suggests that immature 30S particles adopt the inactive conformation in the cell, and transition to the active state occurs at a late stage of maturation. Here, we target nucleotides that form the alternative helix (hALT) of the inactive state. Using an orthogonal ribosome system, we find that disruption of hALT decreases translation activity in the cell modestly, by approximately twofold, without compromising ribosome fidelity. Ribosomes carrying substitutions at positions 1532-1533 support the growth of Escherichia coli strain Δ7 prrn (which carries a single rRNA operon), albeit at rates 10%-20% slower than wild-type ribosomes. These mutant Δ7 prrn strains accumulate free 30S particles and precursor 17S rRNA, indicative of biogenesis defects. Analysis of purified control and mutant subunits suggests that hALT stabilizes the inactive state by 1.2 kcal/mol with little-to-no impact on the active state or the transition state of conversion.


Subject(s)
Escherichia coli , Nucleic Acid Conformation , RNA, Ribosomal, 16S , Ribosome Subunits, Small, Bacterial , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Ribosome Subunits, Small, Bacterial/metabolism , Ribosome Subunits, Small, Bacterial/genetics , Protein Biosynthesis , Magnesium/metabolism
18.
RNA ; 30(8): 992-1010, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38777381

ABSTRACT

Residing in the 5' untranslated region of the mRNA, the 2'-deoxyguanosine (2'-dG) riboswitch mRNA element adopts an alternative structure upon binding of the 2'-dG molecule, which terminates transcription. RNA conformations are generally strongly affected by positively charged metal ions (especially Mg2+). We have quantitatively explored the combined effect of ligand (2'-dG) and Mg2+ binding on the energy landscape of the aptamer domain of the 2'-dG riboswitch with both explicit solvent all-atom molecular dynamics simulations (99 µsec aggregate sampling for the study) and selective 2'-hydroxyl acylation analyzed by primer extension (SHAPE) experiments. We show that both ligand and Mg2+ are required for the stabilization of the aptamer domain; however, the two factors act with different modalities. The addition of Mg2+ remodels the energy landscape and reduces its frustration by the formation of additional contacts. In contrast, the binding of 2'-dG eliminates the metastable states by nucleating a compact core for the aptamer domain. Mg2+ ions and ligand binding are required to stabilize the least stable helix, P1 (which needs to unfold to activate the transcription platform), and the riboswitch core formed by the backbone of the P2 and P3 helices. Mg2+ and ligand also facilitate a more compact structure in the three-way junction region.


Subject(s)
Magnesium , Molecular Dynamics Simulation , Nucleic Acid Conformation , RNA, Messenger , Riboswitch , Magnesium/metabolism , Magnesium/chemistry , Magnesium/pharmacology , RNA, Messenger/genetics , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ligands , 5' Untranslated Regions , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics
19.
Cell ; 146(2): 262-76, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21784247

ABSTRACT

The Dicer ribonuclease III (RNase III) enzymes process long double-stranded RNA (dsRNA) into small interfering RNAs (siRNAs) that direct RNA interference. Here, we describe the structure and activity of a catalytically active fragment of Kluyveromyces polysporus Dcr1, which represents the noncanonical Dicers found in budding yeasts. The crystal structure revealed a homodimer resembling that of bacterial RNase III but extended by a unique N-terminal domain, and it identified additional catalytic residues conserved throughout eukaryotic RNase III enzymes. Biochemical analyses showed that Dcr1 dimers bind cooperatively along the dsRNA substrate such that the distance between consecutive active sites determines the length of the siRNA products. Thus, unlike canonical Dicers, which successively remove siRNA duplexes from the dsRNA termini, budding-yeast Dicers initiate processing in the interior and work outward. The distinct mechanism of budding-yeast Dicers establishes a paradigm for natural molecular rulers and imparts substrate preferences with ramifications for biological function.


Subject(s)
Kluyveromyces/enzymology , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Kluyveromyces/metabolism , Magnesium/metabolism , Models, Molecular , Molecular Sequence Data , RNA, Double-Stranded/metabolism , RNA, Small Interfering/metabolism , Saccharomyces/enzymology , Saccharomyces/metabolism , Sequence Alignment
20.
Nucleic Acids Res ; 52(16): 9343-9359, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39019776

ABSTRACT

Cas12a is the immune effector of type V-A CRISPR-Cas systems and has been co-opted for genome editing and other biotechnology tools. The specificity of Cas12a has been the subject of extensive investigation both in vitro and in genome editing experiments. However, in vitro studies have often been performed at high magnesium ion concentrations that are inconsistent with the free Mg2+ concentrations that would be present in cells. By profiling the specificity of Cas12a orthologs at a range of Mg2+ concentrations, we find that Cas12a switches its specificity depending on metal ion concentration. Lowering Mg2+ concentration decreases cleavage defects caused by seed mismatches, while increasing the defects caused by PAM-distal mismatches. We show that Cas12a can bind seed mutant targets more rapidly at low Mg2+ concentrations, resulting in faster cleavage. In contrast, PAM-distal mismatches cause substantial defects in cleavage following formation of the Cas12a-target complex at low Mg2+ concentrations. We observe differences in Cas12a specificity switching between three orthologs that results in variations in the routes of phage escape from Cas12a-mediated immunity. Overall, our results reveal the importance of physiological metal ion conditions on the specificity of Cas effectors that are used in different cellular environments.


CRISPR-Cas systems are commonly used for biotechnology. Their specificity has been studied extensively and has previously been thought to be well understood. In this work, we asked a simple question about the effect of metal ion concentration on CRISPR specificity; the results are surprising and striking. At the actual metal ion concentrations found in cells, Cas12a specificity is inverted in comparison to the higher metal ion conditions that are typically used in test-tube assays. The specificity observed at lower metal ion concentration is more relevant under cellular conditions.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Cas Systems , Magnesium , Magnesium/metabolism , Magnesium/chemistry , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , Gene Editing/methods , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Substrate Specificity , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL