Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 9(10)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38775150

RESUMEN

This study lays the groundwork for future lentivirus-mediated gene therapy in patients with Diamond Blackfan anemia (DBA) caused by mutations in ribosomal protein S19 (RPS19), showing evidence of a new safe and effective therapy. The data show that, unlike patients with Fanconi anemia (FA), the hematopoietic stem cell (HSC) reservoir of patients with DBA was not significantly reduced, suggesting that collection of these cells should not constitute a remarkable restriction for DBA gene therapy. Subsequently, 2 clinically applicable lentiviral vectors were developed. In the former lentiviral vector, PGK.CoRPS19 LV, a codon-optimized version of RPS19 was driven by the phosphoglycerate kinase promoter (PGK) already used in different gene therapy trials, including FA gene therapy. In the latter one, EF1α.CoRPS19 LV, RPS19 expression was driven by the elongation factor alpha short promoter, EF1α(s). Preclinical experiments showed that transduction of DBA patient CD34+ cells with the PGK.CoRPS19 LV restored erythroid differentiation, and demonstrated the long-term repopulating properties of corrected DBA CD34+ cells, providing evidence of improved erythroid maturation. Concomitantly, long-term restoration of ribosomal biogenesis was verified using a potentially novel method applicable to patients' blood cells, based on ribosomal RNA methylation analyses. Finally, in vivo safety studies and proviral insertion site analyses showed that lentivirus-mediated gene therapy was nontoxic.


Asunto(s)
Anemia de Diamond-Blackfan , Terapia Genética , Vectores Genéticos , Células Madre Hematopoyéticas , Lentivirus , Proteínas Ribosómicas , Anemia de Diamond-Blackfan/terapia , Anemia de Diamond-Blackfan/genética , Humanos , Terapia Genética/métodos , Lentivirus/genética , Proteínas Ribosómicas/genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Ratones , Masculino , Femenino , Ribosomas/metabolismo , Ribosomas/genética , Regiones Promotoras Genéticas , Mutación , Trasplante de Células Madre Hematopoyéticas/métodos
2.
EMBO Rep ; 25(6): 2786-2811, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38654122

RESUMEN

Ribosome biogenesis is initiated in the nucleolus, a multiphase biomolecular condensate formed by liquid-liquid phase separation. The nucleolus is a powerful disease biomarker and stress biosensor whose morphology reflects function. Here we have used digital holographic microscopy (DHM), a label-free quantitative phase contrast microscopy technique, to detect nucleoli in adherent and suspension human cells. We trained convolutional neural networks to detect and quantify nucleoli automatically on DHM images. Holograms containing cell optical thickness information allowed us to define a novel index which we used to distinguish nucleoli whose material state had been modulated optogenetically by blue-light-induced protein aggregation. Nucleoli whose function had been impacted by drug treatment or depletion of ribosomal proteins could also be distinguished. We explored the potential of the technology to detect other natural and pathological condensates, such as those formed upon overexpression of a mutant form of huntingtin, ataxin-3, or TDP-43, and also other cell assemblies (lipid droplets). We conclude that DHM is a powerful tool for quantitatively characterizing nucleoli and other cell assemblies, including their material state, without any staining.


Asunto(s)
Nucléolo Celular , Holografía , Humanos , Nucléolo Celular/metabolismo , Holografía/métodos , Redes Neurales de la Computación , Microscopía/métodos , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/genética , Ataxina-3/metabolismo , Ataxina-3/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Microscopía de Contraste de Fase/métodos , Imágenes de Fase Cuantitativa
3.
Mol Cell ; 83(18): 3268-3282.e7, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37689068

RESUMEN

Heritable non-genetic information can regulate a variety of complex phenotypes. However, what specific non-genetic cues are transmitted from parents to their descendants are poorly understood. Here, we perform metabolic methyl-labeling experiments to track the heritable transmission of methylation from ancestors to their descendants in the nematode Caenorhabditis elegans (C. elegans). We find heritable methylation in DNA, RNA, proteins, and lipids. We find that parental starvation elicits reduced fertility, increased heat stress resistance, and extended longevity in fed, naïve progeny. This intergenerational hormesis is accompanied by a heritable increase in N6'-dimethyl adenosine (m6,2A) on the 18S ribosomal RNA at adenosines 1735 and 1736. We identified DIMT-1/DIMT1 as the m6,2A and BUD-23/BUD23 as the m7G methyltransferases in C. elegans that are both required for intergenerational hormesis, while other rRNA methyltransferases are dispensable. This study labels and tracks heritable non-genetic material across generations and demonstrates the importance of rRNA methylation for regulating epigenetic inheritance.


Asunto(s)
Caenorhabditis elegans , Hormesis , Animales , ARN Ribosómico 18S , Caenorhabditis elegans/genética , Metiltransferasas/genética , Adenosina
4.
RNA Biol ; 20(1): 652-665, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-37635368

RESUMEN

Ribosomal RNAs are decorated by numerous post-transcriptional modifications whose exact roles in ribosome biogenesis, function, and human pathophysiology remain largely unknown. Here, we report a targeted direct rRNA sequencing approach involving a substrate selection step and demonstrate its suitability to identify differential modification sites in combination with the JACUSA2 software. We compared JACUSA2 to other tools designed for RNA modification detection and show that JACUSA2 outperforms other software with regard to detection of base modifications such as methylation, acetylation and aminocarboxypropylation. To illustrate its widespread usability, we applied our method to a collection of CRISPR-Cas9 engineered colon carcinoma cells lacking specific enzymatic activities responsible for particular rRNA modifications and systematically compared them to isogenic wild-type RNAs. Besides the numerous 2'-O methylated riboses and pseudouridylated residues, our approach was suitable to reliably identify differential base methylation and acetylation events. Importantly, our method does not require any prior knowledge of modification sites or the need to train complex models. We further report for the first time detection of human rRNA modifications by direct RNA-sequencing on Flongle flow cells, the smallest-scale nanopore flow cell available to date. The use of these smaller flow cells reduces RNA input requirements, making our workflow suitable for the analysis of samples with limited availability and clinical work.


Asunto(s)
Nanoporos , ARN , Humanos , ARN/genética , Ribosomas/genética , ARN Ribosómico/genética , Procesamiento Postranscripcional del ARN
5.
Nat Commun ; 13(1): 3706, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35764642

RESUMEN

Ribosome biogenesis is an energetically expensive program that is dictated by nutrient availability. Here we report that nutrient deprivation severely impairs precursor ribosomal RNA (pre-rRNA) processing and leads to the accumulation of unprocessed rRNAs. Upon nutrient restoration, pre-rRNAs stored under starvation are processed into mature rRNAs that are utilized for ribosome biogenesis. Failure to accumulate pre-rRNAs under nutrient stress leads to perturbed ribosome assembly upon nutrient restoration and subsequent apoptosis via uL5/uL18-mediated activation of p53. Restoration of glutamine alone activates p53 by triggering uL5/uL18 translation. Induction of uL5/uL18 protein synthesis by glutamine is dependent on the translation factor eukaryotic elongation factor 2 (eEF2), which is in turn dependent on Raf/MEK/ERK signaling. Depriving cells of glutamine prevents the activation of p53 by rRNA synthesis inhibitors. Our data reveals a mechanism that tumor cells can exploit to suppress p53-mediated apoptosis during fluctuations in environmental nutrient availability.


Asunto(s)
Glutamina , Neoplasias , Glutamina/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico , Precursores del ARN/metabolismo , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Ribosomas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Nucleic Acids Res ; 47(15): 7719-7733, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31328227

RESUMEN

N6-methyladenosine (m6A) has recently been found abundantly on messenger RNA and shown to regulate most steps of mRNA metabolism. Several important m6A methyltransferases have been described functionally and structurally, but the enzymes responsible for installing one m6A residue on each subunit of human ribosomes at functionally important sites have eluded identification for over 30 years. Here, we identify METTL5 as the enzyme responsible for 18S rRNA m6A modification and confirm ZCCHC4 as the 28S rRNA modification enzyme. We show that METTL5 must form a heterodimeric complex with TRMT112, a known methyltransferase activator, to gain metabolic stability in cells. We provide the first atomic resolution structure of METTL5-TRMT112, supporting that its RNA-binding mode differs distinctly from that of other m6A RNA methyltransferases. On the basis of similarities with a DNA methyltransferase, we propose that METTL5-TRMT112 acts by extruding the adenosine to be modified from a double-stranded nucleic acid.


Asunto(s)
Adenosina/química , Regulación Neoplásica de la Expresión Génica , Metiltransferasas/química , ARN Mensajero/química , ARN Ribosómico 18S/química , Adenosina/genética , Adenosina/metabolismo , Secuencia de Bases , Sitios de Unión , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Cristalografía por Rayos X , Eliminación de Gen , Células HCT116 , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Ribosómico 18S/genética , ARN Ribosómico 18S/metabolismo , Transducción de Señal , Especificidad por Sustrato
7.
Biochem Pharmacol ; 159: 74-81, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30468711

RESUMEN

Ribosomes are nanomachines essential for protein production in all living cells. Ribosome synthesis increases in cancer cells to cope with a rise in protein synthesis and sustain unrestricted growth. This increase in ribosome biogenesis is reflected by severe morphological alterations of the nucleolus, the cell compartment where the initial steps of ribosome biogenesis take place. Ribosome biogenesis has recently emerged as an effective target in cancer therapy, and several compounds that inhibit ribosome production or function, killing preferentially cancer cells, have entered clinical trials. Recent research indicates that cells express heterogeneous populations of ribosomes and that the composition of ribosomes may play a key role in tumorigenesis, exposing novel therapeutic opportunities. Here, we review recent data demonstrating that ribosome biogenesis is a promising druggable pathway in cancer therapy, and discuss future research perspectives.


Asunto(s)
Antineoplásicos/farmacología , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo , Humanos , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , ARN Ribosómico/metabolismo
8.
Structure ; 26(3): 416-425.e4, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29429877

RESUMEN

Alkaloids isolated from the Amaryllidaceae plants have potential as therapeutics for treating human diseases. Haemanthamine has been studied as a novel anticancer agent due to its ability to overcome cancer cell resistance to apoptosis. Biochemical experiments have suggested that hemanthamine targets the ribosome. However, a structural characterization of its mechanism has been missing. Here we present the 3.1 Å resolution X-ray structure of haemanthamine bound to the Saccharomyces cerevisiae 80S ribosome. This structure reveals that haemanthamine targets the A-site cleft on the large ribosomal subunit rearranging rRNA to halt the elongation phase of translation. Furthermore, we provide evidence that haemanthamine and other Amaryllidaceae alkaloids also inhibit specifically ribosome biogenesis, triggering nucleolar stress response and leading to p53 stabilization in cancer cells. Together with a computer-aided interpretation of existing structure-activity relationships of Amaryllidaceae alkaloids congeners, we provide a rationale for designing molecules with enhanced potencies and reduced toxicities.


Asunto(s)
Alcaloides de Amaryllidaceae/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/metabolismo , Fenantridinas/farmacología , Ribosomas/metabolismo , Saccharomyces cerevisiae/metabolismo , Alcaloides de Amaryllidaceae/química , Antineoplásicos/química , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Cristalografía por Rayos X , Células HCT116 , Humanos , Modelos Moleculares , Conformación Molecular , Fenantridinas/química , ARN Ribosómico/química , ARN Ribosómico/metabolismo , Ribosomas/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Relación Estructura-Actividad , Proteína p53 Supresora de Tumor/metabolismo
9.
Mol Biol Cell ; 26(11): 2080-95, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25851604

RESUMEN

At the heart of the ribosome lie rRNAs, whose catalytic function in translation is subtly modulated by posttranscriptional modifications. In the small ribosomal subunit of budding yeast, on the 18S rRNA, two adjacent adenosines (A1781/A1782) are N(6)-dimethylated by Dim1 near the decoding site, and one guanosine (G1575) is N(7)-methylated by Bud23-Trm112 at a ridge between the P- and E-site tRNAs. Here we establish human DIMT1L and WBSCR22-TRMT112 as the functional homologues of yeast Dim1 and Bud23-Trm112. We report that these enzymes are required for distinct pre-rRNA processing reactions leading to synthesis of 18S rRNA, and we demonstrate that in human cells, as in budding yeast, ribosome biogenesis requires the presence of the modification enzyme rather than its RNA-modifying catalytic activity. We conclude that a quality control mechanism has been conserved from yeast to human by which binding of a methyltransferase to nascent pre-rRNAs is a prerequisite to processing, so that all cleaved RNAs are committed to faithful modification. We further report that 18S rRNA dimethylation is nuclear in human cells, in contrast to yeast, where it is cytoplasmic. Yeast and human ribosome biogenesis thus have both conserved and distinctive features.


Asunto(s)
Metiltransferasas/metabolismo , Procesamiento Postranscripcional del ARN , ARN Ribosómico 18S/metabolismo , Ribosomas/metabolismo , Humanos , Metilación , Precursores del ARN/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(51): E5518-26, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25489090

RESUMEN

The eukaryotic small ribosomal subunit carries only four ribosomal (r) RNA methylated bases, all close to important functional sites. N(7)-methylguanosine (m(7)G) introduced at position 1575 on 18S rRNA by Bud23-Trm112 is at a ridge forming a steric block between P- and E-site tRNAs. Here we report atomic resolution structures of Bud23-Trm112 in the apo and S-adenosyl-L-methionine (SAM)-bound forms. Bud23 and Trm112 interact through formation of a ß-zipper involving main-chain atoms, burying an important hydrophobic surface and stabilizing the complex. The structures revealed that the coactivator Trm112 undergoes an induced fit to accommodate its methyltransferase (MTase) partner. We report important structural similarity between the active sites of Bud23 and Coffea canephora xanthosine MTase, leading us to propose and validate experimentally a model for G1575 coordination. We identify Bud23 residues important for Bud23-Trm112 complex formation and recruitment to pre-ribosomes. We report that though Bud23-Trm112 binds precursor ribosomes at an early nucleolar stage, m(7)G methylation occurs at a late step of small subunit biogenesis, implying specifically delayed catalytic activation. Finally, we show that Bud23-Trm112 interacts directly with the box C/D snoRNA U3-associated DEAH RNA helicase Dhr1 supposedly involved in central pseudoknot formation; this suggests that Bud23-Trm112 might also contribute to controlling formation of this irreversible and dramatic structural reorganization essential to overall folding of small subunit rRNA. Our study contributes important new elements to our understanding of key molecular aspects of human ribosomopathy syndromes associated with WBSCR22 (human Bud23) malfunction.


Asunto(s)
Metiltransferasas/metabolismo , ARN Ribosómico 18S/metabolismo , Ribosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , ARNt Metiltransferasas/metabolismo , Catálisis , Metilación , Metiltransferasas/química , Modelos Moleculares , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/química , ARNt Metiltransferasas/química
11.
Mol Cell ; 51(4): 539-51, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973377

RESUMEN

Mature ribosomal RNAs (rRNAs) are produced from polycistronic precursors following complex processing. Precursor (pre)-rRNA processing has been extensively characterized in yeast and was assumed to be conserved in humans. We functionally characterized 625 nucleolar proteins in HeLa cells and identified 286 required for processing, including 74 without a yeast homolog. For selected candidates, we demonstrated that pre-rRNA processing defects are conserved in different cell types (including primary cells), defects are not due to activation of a p53-dependent nucleolar tumor surveillance pathway, and they precede cell-cycle arrest and apoptosis. We also investigated the exosome's role in processing internal transcribed spacers (ITSs) and report that 3' end maturation of 18S rRNA involves EXOSC10/Rrp6, a yeast ITS2 processing factor. We conclude that human cells adopt unique strategies and recruit distinct trans-acting factors to carry out essential processing steps, posing fundamental implications for understanding ribosomopathies at the molecular level and developing effective therapeutic agents.


Asunto(s)
Nucléolo Celular/genética , Proteínas Nucleares/metabolismo , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN , ARN Ribosómico/genética , Ribosomas/metabolismo , Transactivadores/metabolismo , Apoptosis , Northern Blotting , Puntos de Control del Ciclo Celular , Nucléolo Celular/metabolismo , Células Cultivadas , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Células HCT116 , Células HeLa , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteínas Nucleares/genética , Precursores del ARN/metabolismo , ARN Ribosómico/metabolismo , Transactivadores/genética
12.
Mol Cell Biol ; 32(12): 2254-67, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22493060

RESUMEN

Posttranscriptional and posttranslational modification of macromolecules is known to fine-tune their functions. Trm112 is unique, acting as an activator of both tRNA and protein methyltransferases. Here we report that in Saccharomyces cerevisiae, Trm112 is required for efficient ribosome synthesis and progression through mitosis. Trm112 copurifies with pre-rRNAs and with multiple ribosome synthesis trans-acting factors, including the 18S rRNA methyltransferase Bud23. Consistent with the known mechanisms of activation of methyltransferases by Trm112, we found that Trm112 interacts directly with Bud23 in vitro and that it is required for its stability in vivo. Consequently, trm112Δ cells are deficient for Bud23-mediated 18S rRNA methylation at position G1575 and for small ribosome subunit formation. Bud23 failure to bind nascent preribosomes activates a nucleolar surveillance pathway involving the TRAMP complexes, leading to preribosome degradation. Trm112 is thus active in rRNA, tRNA, and translation factor modification, ideally placing it at the interface between ribosome synthesis and function.


Asunto(s)
Metiltransferasas/metabolismo , ARN Ribosómico 18S/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , ARNt Metiltransferasas/metabolismo , Guanina , Metilación , Proteína Metiltransferasas/metabolismo , Procesamiento Postranscripcional del ARN , ARN de Transferencia/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA