Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.768
1.
Nat Commun ; 15(1): 4783, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38839776

Ribosomes translate the genetic code into proteins. Recent technical advances have facilitated in situ structural analyses of ribosome functional states inside eukaryotic cells and the minimal bacterium Mycoplasma. However, such analyses of Gram-negative bacteria are lacking, despite their ribosomes being major antimicrobial drug targets. Here we compare two E. coli strains, a lab E. coli K-12 and human gut isolate E. coli ED1a, for which tetracycline exhibits bacteriostatic and bactericidal action, respectively. Using our approach for close-to-native E. coli sample preparation, we assess the two strains by cryo-ET and visualize their ribosomes at high resolution in situ. Upon tetracycline treatment, these exhibit virtually identical drug binding sites, yet the conformation distribution of ribosomal complexes differs. While K-12 retains ribosomes in a translation-competent state, tRNAs are lost in the vast majority of ED1a ribosomes. These structural findings together with the proteome-wide abundance and thermal stability assessments indicate that antibiotic responses are complex in cells and can differ between different strains of a single species, thus arguing that all relevant bacterial strains should be analyzed in situ when addressing antibiotic mode of action.


Anti-Bacterial Agents , Escherichia coli , Ribosomes , Tetracycline , Ribosomes/metabolism , Ribosomes/drug effects , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Tetracycline/pharmacology , Cryoelectron Microscopy , RNA, Transfer/metabolism , RNA, Transfer/genetics , Humans , Binding Sites , Protein Biosynthesis/drug effects , Escherichia coli K12/drug effects , Escherichia coli K12/genetics , Escherichia coli K12/metabolism
2.
Sci Rep ; 14(1): 14253, 2024 06 20.
Article En | MEDLINE | ID: mdl-38902339

The antibiotic fusidic acid (FA) is used to treat Staphylococcus aureus infections. It inhibits protein synthesis by binding to elongation factor G (EF-G) and preventing its release from the ribosome after translocation. While FA, due to permeability issues, is only effective against gram-positive bacteria, the available structures of FA-inhibited complexes are from gram-negative model organisms. To fill this knowledge gap, we solved cryo-EM structures of the S. aureus ribosome in complex with mRNA, tRNA, EF-G and FA to 2.5 Å resolution and the corresponding complex structures with the recently developed FA derivative FA-cyclopentane (FA-CP) to 2.0 Å resolution. With both FA variants, the majority of the ribosomal particles are observed in chimeric state and only a minor population in post-translocational state. As expected, FA binds in a pocket between domains I, II and III of EF-G and the sarcin-ricin loop of 23S rRNA. FA-CP binds in an identical position, but its cyclopentane moiety provides additional contacts to EF-G and 23S rRNA, suggesting that its improved resistance profile towards mutations in EF-G is due to higher-affinity binding. These high-resolution structures reveal new details about the S. aureus ribosome, including confirmation of many rRNA modifications, and provide an optimal starting point for future structure-based drug discovery on an important clinical drug target.


Cryoelectron Microscopy , Cyclopentanes , Fusidic Acid , Peptide Elongation Factor G , Ribosomes , Staphylococcus aureus , Fusidic Acid/pharmacology , Fusidic Acid/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Ribosomes/metabolism , Ribosomes/drug effects , Cyclopentanes/pharmacology , Cyclopentanes/chemistry , Peptide Elongation Factor G/metabolism , Peptide Elongation Factor G/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Models, Molecular , RNA, Transfer/metabolism , RNA, Transfer/chemistry
3.
Zool Res ; 45(3): 663-678, 2024 May 18.
Article En | MEDLINE | ID: mdl-38766748

A growing number of studies have demonstrated that repeated exposure to sevoflurane during development results in persistent social abnormalities and cognitive impairment. Davunetide, an active fragment of the activity-dependent neuroprotective protein (ADNP), has been implicated in social and cognitive protection. However, the potential of davunetide to attenuate social deficits following sevoflurane exposure and the underlying developmental mechanisms remain poorly understood. In this study, ribosome and proteome profiles were analyzed to investigate the molecular basis of sevoflurane-induced social deficits in neonatal mice. The neuropathological basis was also explored using Golgi staining, morphological analysis, western blotting, electrophysiological analysis, and behavioral analysis. Results indicated that ADNP was significantly down-regulated following developmental exposure to sevoflurane. In adulthood, anterior cingulate cortex (ACC) neurons exposed to sevoflurane exhibited a decrease in dendrite number, total dendrite length, and spine density. Furthermore, the expression levels of Homer, PSD95, synaptophysin, and vglut2 were significantly reduced in the sevoflurane group. Patch-clamp recordings indicated reductions in both the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs). Notably, davunetide significantly ameliorated the synaptic defects, social behavior deficits, and cognitive impairments induced by sevoflurane. Mechanistic analysis revealed that loss of ADNP led to dysregulation of Ca 2+ activity via the Wnt/ß-catenin signaling, resulting in decreased expression of synaptic proteins. Suppression of Wnt signaling was restored in the davunetide-treated group. Thus, ADNP was identified as a promising therapeutic target for the prevention and treatment of neurodevelopmental toxicity caused by general anesthetics. This study provides important insights into the mechanisms underlying social and cognitive disturbances caused by sevoflurane exposure in neonatal mice and elucidates the regulatory pathways involved.


Animals, Newborn , Cognitive Dysfunction , Proteome , Sevoflurane , Social Behavior , Animals , Sevoflurane/adverse effects , Mice , Cognitive Dysfunction/chemically induced , Ribosomes/drug effects , Ribosomes/metabolism , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/toxicity , Anesthetics, Inhalation/pharmacology , Nerve Tissue Proteins/metabolism , Male , Behavior, Animal/drug effects
4.
Elife ; 122024 Apr 29.
Article En | MEDLINE | ID: mdl-38682900

The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the 'WIN' site that tethers WDR5 to chromatin. WIN site inhibitors (WINi) are active against multiple cancer cell types in vitro, the most notable of which are those derived from MLL-rearranged (MLLr) leukemias. Peptidomimetic WINi were originally proposed to inhibit MLLr cells via dysregulation of genes connected to hematopoietic stem cell expansion. Our discovery and interrogation of small-molecule WINi, however, revealed that they act in MLLr cell lines to suppress ribosome protein gene (RPG) transcription, induce nucleolar stress, and activate p53. Because there is no precedent for an anticancer strategy that specifically targets RPG expression, we took an integrated multi-omics approach to further interrogate the mechanism of action of WINi in human MLLr cancer cells. We show that WINi induce depletion of the stock of ribosomes, accompanied by a broad yet modest translational choke and changes in alternative mRNA splicing that inactivate the p53 antagonist MDM4. We also show that WINi are synergistic with agents including venetoclax and BET-bromodomain inhibitors. Together, these studies reinforce the concept that WINi are a novel type of ribosome-directed anticancer therapy and provide a resource to support their clinical implementation in MLLr leukemias and other malignancies.


Intracellular Signaling Peptides and Proteins , Myeloid-Lymphoid Leukemia Protein , Nuclear Proteins , Ribosomes , Tumor Suppressor Protein p53 , Humans , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Peptidomimetics/pharmacology
5.
Bioorg Chem ; 147: 107338, 2024 Jun.
Article En | MEDLINE | ID: mdl-38583253

Macrozones are novel conjugates of azithromycin and thiosemicarbazones, which exhibit very good in vitro antibacterial activities against susceptible and some resistant bacterial strains thus showing a potential for further development. A combination of spectrometric (fluorimetry, STD and WaterLOGSY NMR) and molecular docking studies provided insights into atomic details of interactions between selected macrozones and biological receptors such as E. coli ribosome and bovine serum albumin. Fluorimetric measurements revealed binding constants in the micro-molar range while NMR experiments provided data on binding epitopes. It has been demonstrated that both STD and WaterLOGSY gave comparable and consistent results unveiling atoms in intimate contacts with biological receptors. Docking studies pointed towards main interactions between macrozones and E. coli ribosome which included specific π - π stacking and hydrogen bonding interactions with thiosemicarbazone part extending down the ribosome exit tunnel. The results of the docking experiments were in fine correlation with those obtained by NMR and fluorimetry. Our investigation pointed towards a two-site binding mechanism of interactions between macrozones and E. coli ribosome which is the most probable reason for their activity against azithromycin-resistant strains. Much better activity of macrozone-nickel coordinated compound against E. coli ribosome compared to other macrozones has been attributed to the higher polarity which enabled better bacterial membrane penetration and binding of the two thiosemicarbazone units thus additionally contributing to the overall binding energy. The knowledge gained in this study should play an important role in anti-infective macrolide design in the future.


Anti-Bacterial Agents , Escherichia coli , Fluorometry , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Docking Simulation , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Binding Sites , Molecular Structure , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Structure-Activity Relationship , Ribosomes/metabolism , Ribosomes/drug effects , Dose-Response Relationship, Drug , Animals , Cattle , Azithromycin/pharmacology , Azithromycin/chemistry , Azithromycin/metabolism
6.
Toxins (Basel) ; 16(4)2024 Apr 10.
Article En | MEDLINE | ID: mdl-38668610

Ribosome-inactivating proteins (RIPs) are a group of proteins with rRNA N-glycosylase activity that irreversibly inhibit protein synthesis and consequently cause cell death. Recently, an RIP called ledodin has been found in shiitake; it is cytotoxic, strongly inhibits protein synthesis, and shows rRNA N-glycosylase activity. In this work, we isolated and characterized a 50 kDa cytotoxic protein from shiitake that we named edodin. Edodin inhibits protein synthesis in a mammalian cell-free system, but not in insect-, yeast-, and bacteria-derived systems. It exhibits rRNA N-glycosylase and DNA-nicking activities, which relate it to plant RIPs. It was also shown to be toxic to HeLa and COLO 320 cells. Its structure is not related to other RIPs found in plants, bacteria, or fungi, but, instead, it presents the characteristic structure of the fold type I of pyridoxal phosphate-dependent enzymes. Homologous sequences have been found in other fungi of the class Agaricomycetes; thus, edodin could be a new type of toxin present in many fungi, some of them edible, which makes them of great interest in health, both for their involvement in food safety and for their potential biomedical and biotechnological applications.


Ribosomes , Shiitake Mushrooms , Humans , Ribosomes/drug effects , Ribosomes/metabolism , Shiitake Mushrooms/chemistry , HeLa Cells , Animals , Mycotoxins/toxicity , Mycotoxins/chemistry , Ribosome Inactivating Proteins/chemistry , Ribosome Inactivating Proteins/pharmacology , Fungal Proteins/chemistry , Fungal Proteins/toxicity , Fungal Proteins/pharmacology , Fungal Proteins/metabolism , Cell Line, Tumor
7.
Nucleic Acids Res ; 51(6): 2915-2930, 2023 04 11.
Article En | MEDLINE | ID: mdl-36864821

Acne vulgaris is a chronic disfiguring skin disease affecting ∼1 billion people worldwide, often having persistent negative effects on physical and mental health. The Gram-positive anaerobe, Cutibacterium acnes is implicated in acne pathogenesis and is, therefore, a main target for antibiotic-based acne therapy. We determined a 2.8-Šresolution structure of the 70S ribosome of Cutibacterium acnes by cryogenic electron microscopy and discovered that sarecycline, a narrow-spectrum antibiotic against Cutibacterium acnes, may inhibit two active sites of this bacterium's ribosome in contrast to the one site detected previously on the model ribosome of Thermus thermophilus. Apart from the canonical binding site at the mRNA decoding center, the second binding site for sarecycline exists at the nascent peptide exit tunnel, reminiscent of the macrolides class of antibiotics. The structure also revealed Cutibacterium acnes-specific features of the ribosomal RNA and proteins. Unlike the ribosome of the Gram-negative bacterium Escherichia coli, Cutibacterium acnes ribosome has two additional proteins, bS22 and bL37, which are also present in the ribosomes of Mycobacterium smegmatis and Mycobacterium tuberculosis. We show that bS22 and bL37 have antimicrobial properties and may be involved in maintaining the healthy homeostasis of the human skin microbiome.


Acne Vulgaris , Anti-Bacterial Agents , Propionibacterium acnes , Ribosomes , Tetracyclines , Humans , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Anti-Bacterial Agents/chemistry , Propionibacterium acnes/drug effects , Protein Biosynthesis , Ribosomes/drug effects , Tetracyclines/pharmacology
8.
Nature ; 610(7930): 205-211, 2022 10.
Article En | MEDLINE | ID: mdl-36171285

Translation is the fundamental process of protein synthesis and is catalysed by the ribosome in all living cells1. Here we use advances in cryo-electron tomography and sub-tomogram analysis2,3 to visualize the structural dynamics of translation inside the bacterium Mycoplasma pneumoniae. To interpret the functional states in detail, we first obtain a high-resolution in-cell average map of all translating ribosomes and build an atomic model for the M. pneumoniae ribosome that reveals distinct extensions of ribosomal proteins. Classification then resolves 13 ribosome states that differ in their conformation and composition. These recapitulate major states that were previously resolved in vitro, and reflect intermediates during active translation. On the basis of these states, we animate translation elongation inside native cells and show how antibiotics reshape the cellular translation landscapes. During translation elongation, ribosomes often assemble in defined three-dimensional arrangements to form polysomes4. By mapping the intracellular organization of translating ribosomes, we show that their association into polysomes involves a local coordination mechanism that is mediated by the ribosomal protein L9. We propose that an extended conformation of L9 within polysomes mitigates collisions to facilitate translation fidelity. Our work thus demonstrates the feasibility of visualizing molecular processes at atomic detail inside cells.


Cryoelectron Microscopy , Mycoplasma pneumoniae , Protein Biosynthesis , Ribosomal Proteins , Ribosomes , Anti-Bacterial Agents/pharmacology , Mycoplasma pneumoniae/cytology , Mycoplasma pneumoniae/drug effects , Mycoplasma pneumoniae/metabolism , Mycoplasma pneumoniae/ultrastructure , Peptide Chain Elongation, Translational/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Polyribosomes/ultrastructure , Protein Biosynthesis/drug effects , Ribosomal Proteins/metabolism , Ribosomal Proteins/ultrastructure , Ribosomes/drug effects , Ribosomes/metabolism , Ribosomes/ultrastructure
9.
Science ; 377(6603): 328-335, 2022 07 15.
Article En | MEDLINE | ID: mdl-35857590

Human NLRP1 (NACHT, LRR, and PYD domain-containing protein 1) is an innate immune sensor predominantly expressed in the skin and airway epithelium. Here, we report that human NLRP1 senses the ultraviolet B (UVB)- and toxin-induced ribotoxic stress response (RSR). Biochemically, RSR leads to the direct hyperphosphorylation of a human-specific disordered linker region of NLRP1 (NLRP1DR) by MAP3K20/ZAKα kinase and its downstream effector, p38. Mutating a single ZAKα phosphorylation site in NLRP1DR abrogates UVB- and ribotoxin-driven pyroptosis in human keratinocytes. Moreover, fusing NLRP1DR to CARD8, which is insensitive to RSR by itself, creates a minimal inflammasome sensor for UVB and ribotoxins. These results provide insight into UVB sensing by human skin keratinocytes, identify several ribotoxins as NLRP1 agonists, and establish inflammasome-driven pyroptosis as an integral component of the RSR.


Inflammasomes , MAP Kinase Kinase Kinases , NLR Proteins , Pyroptosis , Ribosomes , Stress, Physiological , Anisomycin/toxicity , CARD Signaling Adaptor Proteins/metabolism , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Inflammasomes/radiation effects , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , MAP Kinase Kinase Kinases/metabolism , Mutation , NLR Proteins/genetics , NLR Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Pyroptosis/drug effects , Pyroptosis/radiation effects , Ribosomes/drug effects , Ribosomes/radiation effects , Ultraviolet Rays
11.
Nat Struct Mol Biol ; 29(2): 162-171, 2022 02.
Article En | MEDLINE | ID: mdl-35165456

The antibiotic linezolid, the first clinically approved member of the oxazolidinone class, inhibits translation of bacterial ribosomes by binding to the peptidyl transferase center. Recent work has demonstrated that linezolid does not inhibit peptide bond formation at all sequences but rather acts in a context-specific manner, namely when alanine occupies the penultimate position of the nascent chain. However, the molecular basis for context-specificity has not been elucidated. Here we show that the second-generation oxazolidinone radezolid also induces stalling with a penultimate alanine, and we determine high-resolution cryo-EM structures of linezolid- and radezolid-stalled ribosome complexes to explain their mechanism of action. These structures reveal that the alanine side chain fits within a small hydrophobic crevice created by oxazolidinone, resulting in improved ribosome binding. Modification of the ribosome by the antibiotic resistance enzyme Cfr disrupts stalling due to repositioning of the modified nucleotide. Together, our findings provide molecular understanding for the context-specificity of oxazolidinones.


Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Oxazolidinones/chemistry , Oxazolidinones/pharmacology , Protein Biosynthesis/drug effects , Alanine/chemistry , Binding Sites , Cryoelectron Microscopy , Linezolid/chemistry , Linezolid/pharmacology , Models, Molecular , Peptidyl Transferases/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Ribosomes/ultrastructure
12.
Nat Struct Mol Biol ; 29(2): 152-161, 2022 02.
Article En | MEDLINE | ID: mdl-35165455

Ribosome-targeting antibiotics serve as powerful antimicrobials and as tools for studying the ribosome, the catalytic peptidyl transferase center (PTC) of which is targeted by many drugs. The classic PTC-acting antibiotic chloramphenicol (CHL) and the newest clinically significant linezolid (LZD) were considered indiscriminate inhibitors of protein synthesis that cause ribosome stalling at every codon of every gene being translated. However, recent discoveries have shown that CHL and LZD preferentially arrest translation when the ribosome needs to polymerize particular amino acid sequences. The molecular mechanisms that underlie the context-specific action of ribosome inhibitors are unknown. Here we present high-resolution structures of ribosomal complexes, with or without CHL, carrying specific nascent peptides that support or negate the drug action. Our data suggest that the penultimate residue of the nascent peptide directly modulates antibiotic affinity to the ribosome by either establishing specific interactions with the drug or by obstructing its proper placement in the binding site.


Chloramphenicol/chemistry , Chloramphenicol/pharmacology , Peptidyl Transferases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Models, Molecular , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Static Electricity , Thermus thermophilus/drug effects , Thermus thermophilus/metabolism
13.
Nat Commun ; 13(1): 173, 2022 01 10.
Article En | MEDLINE | ID: mdl-35013311

Mechanisms of drug-tolerance remain poorly understood and have been linked to genomic but also to non-genomic processes. 5-fluorouracil (5-FU), the most widely used chemotherapy in oncology is associated with resistance. While prescribed as an inhibitor of DNA replication, 5-FU alters all RNA pathways. Here, we show that 5-FU treatment leads to the production of fluorinated ribosomes exhibiting altered translational activities. 5-FU is incorporated into ribosomal RNAs of mature ribosomes in cancer cell lines, colorectal xenografts, and human tumors. Fluorinated ribosomes appear to be functional, yet, they display a selective translational activity towards mRNAs depending on the nature of their 5'-untranslated region. As a result, we find that sustained translation of IGF-1R mRNA, which encodes one of the most potent cell survival effectors, promotes the survival of 5-FU-treated colorectal cancer cells. Altogether, our results demonstrate that "man-made" fluorinated ribosomes favor the drug-tolerant cellular phenotype by promoting translation of survival genes.


Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/drug therapy , DNA, Neoplasm/genetics , Drug Tolerance/genetics , Fluorouracil/pharmacology , Protein Biosynthesis/drug effects , Receptor, IGF Type 1/genetics , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Replication , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm/genetics , HCT116 Cells , Halogenation , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Receptor, IGF Type 1/agonists , Receptor, IGF Type 1/metabolism , Ribosomes/drug effects , Ribosomes/genetics , Ribosomes/metabolism , Xenograft Model Antitumor Assays
14.
Biochem Biophys Res Commun ; 588: 61-67, 2022 01 15.
Article En | MEDLINE | ID: mdl-34952471

Actinomycin D has been reported to selectively inhibit rRNA synthesis and ribosome biogenesis, induce G2 checkpoint of cell cycle arrest in HeLa cells. In Arabidopsis, actinomycin D was also used as agent to preferentially inhibit the ribosome biosynthesis and ribosomal function. However, the function of actinomycin D on Arabidopsis root development remains to be elucidated. In this study, we exposed Arabidopsis seedlings to actinomycin D with the aim of evaluating the effects of ribosome biogenesis on root development. The results demonstrated that actinomycin D inhibited Arabidopsis root growth by reduced meristematic activity in a dose dependent manner. Exposure to actinomycin D decreased the expression of WOX5 and key stem cell niche-defining transcription factors SHR and PLT1, thus the loss function of QC identity and stem cell niche maintenance. In addition, dead cells were observed after actinomycin D treatment in root stele initials and DNA damage response was constitutively activated. Collectively, we propose that ribosome biogenesis plays key role in primary root growth through maintenance of root stem cell niche and DNA damage response in Arabidopsis.


Arabidopsis/growth & development , Arabidopsis/metabolism , Dactinomycin/pharmacology , Organelle Biogenesis , Plant Roots/growth & development , Plant Roots/metabolism , Ribosomes/metabolism , Arabidopsis/drug effects , Arabidopsis Proteins/metabolism , Cell Death/drug effects , DNA Damage , Indoleacetic Acids/metabolism , Meristem/drug effects , Meristem/growth & development , Organ Size/drug effects , Plant Roots/drug effects , Ribosomes/drug effects , Stem Cell Niche/drug effects
15.
J Med Chem ; 64(20): 15334-15348, 2021 10 28.
Article En | MEDLINE | ID: mdl-34648707

Ricin toxin A subunit (RTA) is the catalytic subunit of ricin, which depurinates an adenine from the sarcin/ricin loop in eukaryotic ribosomes. There are no approved inhibitors against ricin. We used a new strategy to disrupt RTA-ribosome interactions by fragment screening using surface plasmon resonance. Here, using a structure-guided approach, we improved the affinity and inhibitory activity of small-molecular-weight lead compounds and obtained improved compounds with over an order of magnitude higher efficiency. Four advanced compounds were characterized by X-ray crystallography. They bind at the RTA-ribosome binding site as the original compound but in a distinctive manner. These inhibitors bind remotely from the catalytic site and cause local conformational changes with no alteration of the catalytic site geometry. Yet they inhibit depurination by ricin holotoxin and inhibit the cytotoxicity of ricin in mammalian cells. They are the first agents that protect against ricin holotoxin by acting directly on RTA.


Drug Design , Enzyme Inhibitors/pharmacology , Ribosomes/drug effects , Ricin/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Binding Sites/drug effects , Chlorocebus aethiops , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Ricin/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Surface Plasmon Resonance , Vero Cells
16.
Toxins (Basel) ; 13(10)2021 09 28.
Article En | MEDLINE | ID: mdl-34678982

Shiga toxins (Stxs), also known as Shiga-like toxins (SLT) or verotoxins (VT), constitute a family of structurally and functionally related cytotoxic proteins produced by the enteric pathogens Shigella dysenteriae type 1 and Stx-producing Escherichia coli (STEC). Infection with these bacteria causes bloody diarrhea and other pathological manifestations that can lead to HUS (hemolytic and uremic syndrome). At the cellular level, Stxs bind to the cellular receptor Gb3 and inhibit protein synthesis by removing an adenine from the 28S rRNA. This triggers multiple cellular signaling pathways, including the ribotoxic stress response (RSR), unfolded protein response (UPR), autophagy and apoptosis. Stxs cause several pathologies of major public health concern, but their specific targeting of host cells and efficient delivery to the cytosol could potentially be exploited for biomedical purposes. Moreover, high levels of expression have been reported for the Stxs receptor, Gb3/CD77, in Burkitt's lymphoma (BL) cells and on various types of solid tumors. These properties have led to many attempts to develop Stxs as tools for biomedical applications, such as cancer treatment or imaging, and several engineered Stxs are currently being tested. We provide here an overview of these studies.


Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Shiga Toxins/pharmacology , Apoptosis , Autophagy , Drug Delivery Systems , Humans , Ribosomes/drug effects , Shiga Toxins/chemistry , Signal Transduction/drug effects , Trihexosylceramides/metabolism
17.
Am J Physiol Cell Physiol ; 321(6): C1000-C1009, 2021 12 01.
Article En | MEDLINE | ID: mdl-34705587

Chemotherapeutic agents (CAs) are first-line antineoplastic treatments against a wide variety of cancers. Despite their effectiveness in halting tumor progression, side effects associated with CAs promote muscle loss by incompletely understood mechanisms. To address this problem, we first identified how oxidative stress impairs protein synthesis in C2C12 myotubes. Transient elevations in reactive oxygen species (ROS) resulted in protein synthesis deficits and reduced ribosomal (r)RNA levels. Oxidative stress did not reduce rRNA gene (rDNA) transcription, but it caused an increase in rRNA and protein oxidation. To determine whether CAs affect protein synthesis independent of oxidative stress, we exposed myotubes to Paclitaxel (PTX), Doxorubicin (DXR), or Marizomib (Mzb) at doses that did result in elevated ROS levels (sub-ROS). Exposure to CAs reduced protein synthesis and rRNA levels, but unlike oxidative stress, sub-ROS exposures impaired rDNA transcription. These results indicate that although oxidative stress disrupts protein synthesis by compromising ribosomal quantity and quality, CAs at sub-ROS doses compromise protein synthesis and ribosomal capacity, at least in part, by reducing rDNA transcription. Therefore, CAs negatively impact protein synthesis by causing oxidative stress in addition to directly reducing the ribosomal capacity of myotubes in a ROS-independent manner.


Antineoplastic Agents/toxicity , Muscle Fibers, Skeletal/drug effects , Oxidative Stress/drug effects , Protein Biosynthesis/drug effects , Ribosomes/drug effects , Animals , Cell Line , Hydrogen Peroxide/toxicity , Mice , Muscle Fibers, Skeletal/metabolism , Oxidative Stress/physiology , Protein Biosynthesis/physiology , Ribosomes/metabolism
18.
Nature ; 599(7885): 507-512, 2021 11.
Article En | MEDLINE | ID: mdl-34707295

The dearth of new medicines effective against antibiotic-resistant bacteria presents a growing global public health concern1. For more than five decades, the search for new antibiotics has relied heavily on the chemical modification of natural products (semisynthesis), a method ill-equipped to combat rapidly evolving resistance threats. Semisynthetic modifications are typically of limited scope within polyfunctional antibiotics, usually increase molecular weight, and seldom permit modifications of the underlying scaffold. When properly designed, fully synthetic routes can easily address these shortcomings2. Here we report the structure-guided design and component-based synthesis of a rigid oxepanoproline scaffold which, when linked to the aminooctose residue of clindamycin, produces an antibiotic of exceptional potency and spectrum of activity, which we name iboxamycin. Iboxamycin is effective against ESKAPE pathogens including strains expressing Erm and Cfr ribosomal RNA methyltransferase enzymes, products of genes that confer resistance to all clinically relevant antibiotics targeting the large ribosomal subunit, namely macrolides, lincosamides, phenicols, oxazolidinones, pleuromutilins and streptogramins. X-ray crystallographic studies of iboxamycin in complex with the native bacterial ribosome, as well as with the Erm-methylated ribosome, uncover the structural basis for this enhanced activity, including a displacement of the [Formula: see text] nucleotide upon antibiotic binding. Iboxamycin is orally bioavailable, safe and effective in treating both Gram-positive and Gram-negative bacterial infections in mice, attesting to the capacity for chemical synthesis to provide new antibiotics in an era of increasing resistance.


Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/classification , Clindamycin/chemical synthesis , Clindamycin/pharmacology , Drug Discovery , Lincomycin/chemical synthesis , Lincomycin/pharmacology , Methyltransferases/genetics , Methyltransferases/metabolism , Microbial Sensitivity Tests , Models, Molecular , Oxepins , Pyrans , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Ribosomes/chemistry , Ribosomes/drug effects , Ribosomes/metabolism , Thermus thermophilus/drug effects , Thermus thermophilus/enzymology , Thermus thermophilus/genetics
19.
Aging (Albany NY) ; 13(19): 23119-23132, 2021 10 07.
Article En | MEDLINE | ID: mdl-34620733

The anticancer effect of zinc oxide nanoparticles (ZnO NPs) largely relies on cellular responses such as alteration of gene expression. Although ZnO NPs have been reported to induce transcriptional changes, the potential of ZnO NPs to affect cellular translatome remains largely unknown. Using ribosome profiling, we demonstrated that the transcription of 78 genes and the translation of 1,448 genes are affected during one hour of ZnO NPs exposure in A549 human lung cancer cells. The mitogen-activated protein kinase (MAPK) pathway is up-regulated upon ZnO NP treatment. The upstream open reading frame (uORF) plays a pervasive role in the induction of up-regulated genes, including TLNRD1 and CCNB1IP1. Knockdown of TLNRD1 or CCNB1IP1 reduces ZnO NP-induced cytotoxicity. Together, our study characterizes the landscape of translational alteration under ZnO NPs treatment and provides potential targets to augment the anticancer effect of ZnO NPs.


Metal Nanoparticles/chemistry , Ribosomes/drug effects , Sequence Analysis, RNA/methods , Zinc Oxide/pharmacology , A549 Cells , Genetic Structures , Humans , Protein Biosynthesis , RNA, Messenger/genetics , Ribosomes/genetics , Zinc Oxide/chemistry
20.
Nucleic Acids Res ; 49(19): 11134-11144, 2021 11 08.
Article En | MEDLINE | ID: mdl-34606617

The Saccharomyces cerevisiae gene deletion collection is widely used for functional gene annotation and genetic interaction analyses. However, the standard G418-resistance cassette used to produce knockout mutants delivers strong regulatory elements into the target genetic loci. To date, its side effects on the expression of neighboring genes have never been systematically assessed. Here, using ribosome profiling data, RT-qPCR, and reporter expression, we investigated perturbations induced by the KanMX module. Our analysis revealed significant alterations in the transcription efficiency of neighboring genes and, more importantly, severe impairment of their mRNA translation, leading to changes in protein abundance. In the 'head-to-head' orientation of the deleted and neighboring genes, knockout often led to a shift of the transcription start site of the latter, introducing new uAUG codon(s) into the expanded 5' untranslated region (5' UTR). In the 'tail-to-tail' arrangement, knockout led to activation of alternative polyadenylation signals in the neighboring gene, thus altering its 3' UTR. These events may explain the so-called neighboring gene effect (NGE), i.e. false genetic interactions of the deleted genes. We estimate that in as much as ∼1/5 of knockout strains the expression of neighboring genes may be substantially (>2-fold) deregulated at the level of translation.


Genetic Loci/drug effects , Gentamicins/pharmacology , Protein Biosynthesis/drug effects , Saccharomyces cerevisiae/drug effects , Sequence Deletion , Transcription, Genetic/drug effects , 3' Untranslated Regions , 5' Untranslated Regions , Base Sequence , Codon , Gene Expression Regulation, Fungal , Gene Knockout Techniques/methods , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Open Reading Frames , Ribosomes/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Initiation Site
...