Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.762
Filtrar
1.
Nucleic Acids Res ; 51(6): 2915-2930, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36864821

RESUMO

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.


Assuntos
Acne Vulgar , Antibacterianos , Propionibacterium acnes , Ribossomos , Tetraciclinas , Humanos , Acne Vulgar/tratamento farmacológico , Acne Vulgar/microbiologia , Antibacterianos/química , Propionibacterium acnes/efeitos dos fármacos , Biossíntese de Proteínas , Ribossomos/efeitos dos fármacos , Tetraciclinas/farmacologia
2.
Nature ; 610(7930): 205-211, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36171285

RESUMO

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.


Assuntos
Microscopia Crioeletrônica , Mycoplasma pneumoniae , Biossíntese de Proteínas , Proteínas Ribossômicas , Ribossomos , Antibacterianos/farmacologia , Mycoplasma pneumoniae/citologia , Mycoplasma pneumoniae/efeitos dos fármacos , Mycoplasma pneumoniae/metabolismo , Mycoplasma pneumoniae/ultraestrutura , Elongação Traducional da Cadeia Peptídica/efeitos dos fármacos , Polirribossomos/efeitos dos fármacos , Polirribossomos/metabolismo , Polirribossomos/ultraestrutura , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Ribossômicas/metabolismo , Proteínas Ribossômicas/ultraestrutura , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Ribossomos/ultraestrutura
3.
Science ; 377(6603): 328-335, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35857590

RESUMO

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.


Assuntos
Inflamassomos , MAP Quinase Quinase Quinases , Proteínas NLR , Piroptose , Ribossomos , Estresse Fisiológico , Anisomicina/toxicidade , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Inflamassomos/efeitos da radiação , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , MAP Quinase Quinase Quinases/metabolismo , Mutação , Proteínas NLR/genética , Proteínas NLR/metabolismo , Proteínas de Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Piroptose/efeitos dos fármacos , Piroptose/efeitos da radiação , Ribossomos/efeitos dos fármacos , Ribossomos/efeitos da radiação , Raios Ultravioleta
5.
Nat Struct Mol Biol ; 29(2): 162-171, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35165456

RESUMO

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.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Oxazolidinonas/química , Oxazolidinonas/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Alanina/química , Sítios de Ligação , Microscopia Crioeletrônica , Linezolida/química , Linezolida/farmacologia , Modelos Moleculares , Peptidil Transferases/metabolismo , RNA Ribossômico/química , RNA Ribossômico/metabolismo , RNA de Transferência/química , RNA de Transferência/metabolismo , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Ribossomos/ultraestrutura
6.
Nat Struct Mol Biol ; 29(2): 152-161, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35165455

RESUMO

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.


Assuntos
Cloranfenicol/química , Cloranfenicol/farmacologia , Peptidil Transferases/antagonistas & inibidores , Antibacterianos/química , Antibacterianos/farmacologia , Sítios de Ligação , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Cinética , Modelos Moleculares , Conformação Proteica , Inibidores da Síntese de Proteínas/química , Inibidores da Síntese de Proteínas/farmacologia , Aminoacil-RNA de Transferência/química , Aminoacil-RNA de Transferência/metabolismo , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Eletricidade Estática , Thermus thermophilus/efeitos dos fármacos , Thermus thermophilus/metabolismo
7.
Nat Commun ; 13(1): 173, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013311

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , DNA de Neoplasias/genética , Tolerância a Medicamentos/genética , Fluoruracila/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Receptor IGF Tipo 1/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Replicação do DNA , DNA de Neoplasias/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Células HCT116 , Halogenação , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Receptor IGF Tipo 1/agonistas , Receptor IGF Tipo 1/metabolismo , Ribossomos/efeitos dos fármacos , Ribossomos/genética , Ribossomos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biochem Biophys Res Commun ; 588: 61-67, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34952471

RESUMO

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.


Assuntos
Arabidopsis/crescimento & desenvolvimento , Arabidopsis/metabolismo , Dactinomicina/farmacologia , Biogênese de Organelas , Raízes de Plantas/crescimento & desenvolvimento , Raízes de Plantas/metabolismo , Ribossomos/metabolismo , Arabidopsis/efeitos dos fármacos , Proteínas de Arabidopsis/metabolismo , Morte Celular/efeitos dos fármacos , Dano ao DNA , Ácidos Indolacéticos/metabolismo , Meristema/efeitos dos fármacos , Meristema/crescimento & desenvolvimento , Tamanho do Órgão/efeitos dos fármacos , Raízes de Plantas/efeitos dos fármacos , Ribossomos/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos
9.
Aging (Albany NY) ; 13(19): 23119-23132, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34620733

RESUMO

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.


Assuntos
Nanopartículas Metálicas/química , Ribossomos/efeitos dos fármacos , Análise de Sequência de RNA/métodos , Óxido de Zinco/farmacologia , Células A549 , Estruturas Genéticas , Humanos , Biossíntese de Proteínas , RNA Mensageiro/genética , Ribossomos/genética , Óxido de Zinco/química
10.
Am J Physiol Cell Physiol ; 321(6): C1000-C1009, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34705587

RESUMO

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.


Assuntos
Antineoplásicos/toxicidade , Fibras Musculares Esqueléticas/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Ribossomos/efeitos dos fármacos , Animais , Linhagem Celular , Peróxido de Hidrogênio/toxicidade , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Estresse Oxidativo/fisiologia , Biossíntese de Proteínas/fisiologia , Ribossomos/metabolismo
11.
Nucleic Acids Res ; 49(19): 11134-11144, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34606617

RESUMO

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.


Assuntos
Loci Gênicos/efeitos dos fármacos , Gentamicinas/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Saccharomyces cerevisiae/efeitos dos fármacos , Deleção de Sequência , Transcrição Gênica/efeitos dos fármacos , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Sequência de Bases , Códon , Regulação Fúngica da Expressão Gênica , Técnicas de Inativação de Genes/métodos , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fases de Leitura Aberta , Ribossomos/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Sítio de Iniciação de Transcrição
12.
Toxins (Basel) ; 13(10)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34678982

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Toxinas Shiga/farmacologia , Apoptose , Autofagia , Sistemas de Liberação de Medicamentos , Humanos , Ribossomos/efeitos dos fármacos , Toxinas Shiga/química , Transdução de Sinais/efeitos dos fármacos , Triexosilceramidas/metabolismo
13.
J Med Chem ; 64(20): 15334-15348, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34648707

RESUMO

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.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Ribossomos/efeitos dos fármacos , Ricina/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Sítios de Ligação/efeitos dos fármacos , Chlorocebus aethiops , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Modelos Moleculares , Estrutura Molecular , Ricina/metabolismo , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície , Células Vero
14.
Nature ; 599(7885): 507-512, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34707295

RESUMO

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.


Assuntos
Antibacterianos/síntese química , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Antibacterianos/química , Antibacterianos/classificação , Clindamicina/síntese química , Clindamicina/farmacologia , Descoberta de Drogas , Lincomicina/síntese química , Lincomicina/farmacologia , Metiltransferases/genética , Metiltransferases/metabolismo , Testes de Sensibilidade Microbiana , Modelos Moleculares , Oxepinas , Piranos , RNA Mensageiro/metabolismo , RNA de Transferência/metabolismo , Ribossomos/química , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Thermus thermophilus/efeitos dos fármacos , Thermus thermophilus/enzimologia , Thermus thermophilus/genética
15.
Biomolecules ; 11(9)2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34572607

RESUMO

In the traditional fermentative model yeast Saccharomyces cerevisiae, ScIxr1 is an HMGB (High Mobility Group box B) protein that has been considered as an important regulator of gene transcription in response to external changes like oxygen, carbon source, or nutrient availability. Kluyveromyces lactis is also a useful eukaryotic model, more similar to many human cells due to its respiratory metabolism. We cloned and functionally characterized by different methodologies KlIXR1, which encodes a protein with only 34.4% amino acid sequence similarity to ScIxr1. Our data indicate that both proteins share common functions, including their involvement in the response to hypoxia or oxidative stress induced by hydrogen peroxide or metal treatments, as well as in the control of key regulators for maintenance of the dNTP (deoxyribonucleotide triphosphate) pool and ribosome synthesis. KlIxr1 is able to bind specific regulatory DNA sequences in the promoter of its target genes, which are well conserved between S. cerevisiae and K. lactis. Oppositely, we found important differences between ScIrx1 and KlIxr1 affecting cellular responses to cisplatin or cycloheximide in these yeasts, which could be dependent on specific and non-conserved domains present in these two proteins.


Assuntos
Desoxirribonucleotídeos/metabolismo , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Proteínas HMGB/metabolismo , Kluyveromyces/crescimento & desenvolvimento , Kluyveromyces/genética , Sequência de Bases , Cádmio/toxicidade , Carbono/farmacologia , Ciclo Celular/efeitos dos fármacos , Cisplatino/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Proteínas Fúngicas/química , Deleção de Genes , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Proteínas HMGB/química , Heme/biossíntese , Peróxido de Hidrogênio/toxicidade , Kluyveromyces/efeitos dos fármacos , Mutação/genética , Oxirredução/efeitos dos fármacos , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Ribossômico/genética , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Saccharomyces cerevisiae/metabolismo
16.
Nat Methods ; 18(9): 1068-1074, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34480152

RESUMO

In general, mRNAs are assumed to be loaded with ribosomes instantly upon entry into the cytoplasm. To measure ribosome density (RD) on nascent mRNA, we developed nascent Ribo-Seq by combining Ribo-Seq with progressive 4-thiouridine labeling. In mouse macrophages, we determined experimentally the lag between the appearance of nascent mRNA and its association with ribosomes, which was calculated to be 20-22 min for bulk mRNA. In mouse embryonic stem cells, nRibo-Seq revealed an even stronger lag of 35-38 min in ribosome loading. After stimulation of macrophages with lipopolysaccharide, the lag between cytoplasmic and translated mRNA leads to uncoupling between input and ribosome-protected fragments, which gives rise to distorted RD measurements under conditions where mRNA amounts are far from steady-state expression. As a result, we demonstrate that transcriptional changes affect RD in a passive way.


Assuntos
Biossíntese de Proteínas , Ribossomos/genética , Ribossomos/metabolismo , Análise de Sequência de RNA/métodos , Animais , Citoplasma/genética , Cinética , Lipopolissacarídeos/farmacologia , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/fisiologia , Células RAW 264.7 , RNA Mensageiro/genética , Proteínas Ribossômicas/biossíntese , Proteínas Ribossômicas/genética , Ribossomos/efeitos dos fármacos , Fatores de Tempo
17.
Nature ; 597(7877): 561-565, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34497418

RESUMO

Single-cell sequencing methods have enabled in-depth analysis of the diversity of cell types and cell states in a wide range of organisms. These tools focus predominantly on sequencing the genomes1, epigenomes2 and transcriptomes3 of single cells. However, despite recent progress in detecting proteins by mass spectrometry with single-cell resolution4, it remains a major challenge to measure translation in individual cells. Here, building on existing protocols5-7, we have substantially increased the sensitivity of these assays to enable ribosome profiling in single cells. Integrated with a machine learning approach, this technology achieves single-codon resolution. We validate this method by demonstrating that limitation for a particular amino acid causes ribosome pausing at a subset of the codons encoding the amino acid. Of note, this pausing is only observed in a sub-population of cells correlating to its cell cycle state. We further expand on this phenomenon in non-limiting conditions and detect pronounced GAA pausing during mitosis. Finally, we demonstrate the applicability of this technique to rare primary enteroendocrine cells. This technology provides a first step towards determining the contribution of the translational process to the remarkable diversity between seemingly identical cells.


Assuntos
Ciclo Celular/genética , Códon/genética , Biossíntese de Proteínas , RNA-Seq/métodos , Ribossomos/metabolismo , Análise de Célula Única , Aminoácidos/deficiência , Aminoácidos/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Humanos , Aprendizado de Máquina , Masculino , Camundongos , Elongação Traducional da Cadeia Peptídica , Iniciação Traducional da Cadeia Peptídica , Terminação Traducional da Cadeia Peptídica , Biossíntese de Proteínas/efeitos dos fármacos , Reprodutibilidade dos Testes , Ribossomos/efeitos dos fármacos
18.
Nucleic Acids Res ; 49(16): 9560-9573, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34417608

RESUMO

Macrolides have been effective clinical antibiotics for over 70 years. They inhibit protein biosynthesis in bacterial pathogens by narrowing the nascent protein exit tunnel in the ribosome. The macrolide class of natural products consist of a macrolactone ring linked to one or more sugar molecules. Most of the macrolides used currently are semi-synthetic erythromycin derivatives, composed of a 14- or 15-membered macrolactone ring. Rapidly emerging resistance in bacterial pathogens is among the most urgent global health challenges, which render many antibiotics ineffective, including next-generation macrolides. To address this threat and advance a longer-term plan for developing new antibiotics, we demonstrate how 16-membered macrolides overcome erythromycin resistance in clinically isolated Staphylococcus aureus strains. By determining the structures of complexes of the large ribosomal subunit of Deinococcus radiodurans (D50S) with these 16-membered selected macrolides, and performing anti-microbial studies, we identified resistance mechanisms they may overcome. This new information provides important insights toward the rational design of therapeutics that are effective against drug resistant human pathogens.


Assuntos
Macrolídeos/química , Micromonospora/química , Antibacterianos/química , Antibacterianos/farmacologia , Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Eritromicina/química , Humanos , Macrolídeos/farmacologia , Testes de Sensibilidade Microbiana , Inibidores da Síntese de Proteínas/farmacologia , Ribossomos/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/patogenicidade
19.
Nat Commun ; 12(1): 5094, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34429433

RESUMO

Ribosome profiling measures genome-wide translation dynamics at sub-codon resolution. Cycloheximide (CHX), a widely used translation inhibitor to arrest ribosomes in these experiments, has been shown to induce biases in yeast, questioning its use. However, whether such biases are present in datasets of other organisms including humans is unknown. Here we compare different CHX-treatment conditions in human cells and yeast in parallel experiments using an optimized protocol. We find that human ribosomes are not susceptible to conformational restrictions by CHX, nor does it distort gene-level measurements of ribosome occupancy, measured decoding speed or the translational ramp. Furthermore, CHX-induced codon-specific biases on ribosome occupancy are not detectable in human cells or other model organisms. This shows that reported biases of CHX are species-specific and that CHX does not affect the outcome of ribosome profiling experiments in most settings. Our findings provide a solid framework to conduct and analyze ribosome profiling experiments.


Assuntos
Cicloeximida/farmacologia , Ribossomos/química , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Animais , Viés , Códon/metabolismo , Células HEK293 , Humanos , Camundongos , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Saccharomyces cerevisiae/metabolismo , Especificidade da Espécie
20.
Nat Commun ; 12(1): 4466, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294725

RESUMO

Macrolides and ketolides comprise a family of clinically important antibiotics that inhibit protein synthesis by binding within the exit tunnel of the bacterial ribosome. While these antibiotics are known to interrupt translation at specific sequence motifs, with ketolides predominantly stalling at Arg/Lys-X-Arg/Lys motifs and macrolides displaying a broader specificity, a structural basis for their context-specific action has been lacking. Here, we present structures of ribosomes arrested during the synthesis of an Arg-Leu-Arg sequence by the macrolide erythromycin (ERY) and the ketolide telithromycin (TEL). Together with deep mutagenesis and molecular dynamics simulations, the structures reveal how ERY and TEL interplay with the Arg-Leu-Arg motif to induce translational arrest and illuminate the basis for the less stringent sequence-specific action of ERY over TEL. Because programmed stalling at the Arg/Lys-X-Arg/Lys motifs is used to activate expression of antibiotic resistance genes, our study also provides important insights for future development of improved macrolide antibiotics.


Assuntos
Antibacterianos/farmacologia , Cetolídeos/farmacologia , Macrolídeos/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Antibacterianos/química , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/enzimologia , Bacillus subtilis/genética , Sítios de Ligação/genética , Microscopia Crioeletrônica , Resistência Microbiana a Medicamentos/genética , Eritromicina/química , Eritromicina/farmacologia , Genes Bacterianos , Cetolídeos/química , Cetolídeos/farmacocinética , Macrolídeos/química , Metiltransferases/química , Metiltransferases/genética , Metiltransferases/metabolismo , Simulação de Dinâmica Molecular , Mutagênese Insercional , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/química , Ribossomos/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...