Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(7): 114405, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38923463

RESUMEN

The RNA cap methyltransferase CMTR1 methylates the first transcribed nucleotide of RNA polymerase II transcripts, impacting gene expression mechanisms, including during innate immune responses. Using mass spectrometry, we identify a multiply phosphorylated region of CMTR1 (phospho-patch [P-Patch]), which is a substrate for the kinase CK2 (casein kinase II). CMTR1 phosphorylation alters intramolecular interactions, increases recruitment to RNA polymerase II, and promotes RNA cap methylation. P-Patch phosphorylation occurs during the G1 phase of the cell cycle, recruiting CMTR1 to RNA polymerase II during a period of rapid transcription and RNA cap formation. CMTR1 phosphorylation is required for the expression of specific RNAs, including ribosomal protein gene transcripts, and promotes cell proliferation. CMTR1 phosphorylation is also required for interferon-stimulated gene expression. The cap-snatching virus, influenza A, utilizes host CMTR1 phosphorylation to produce the caps required for virus production and infection. We present an RNA cap methylation control mechanism whereby CK2 controls CMTR1, enhancing co-transcriptional capping.

2.
Discov Immunol ; 3(1): kyad021, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38572449

RESUMEN

On T cell activation, upregulation of gene expression produces the protein required for the differentiation and proliferation of effector cell populations. RAM (RNMT-Activating Mini protein/RAMAC/Fam103a1), the cofactor of the RNA cap methyltransferase RNMT (RNA guanosine N-7 cap methyltransferase), is upregulated following activation. Formation of the RNA cap protects RNA during synthesis and guides RNA processing and translation. Using conditional gene deletion, we found that Ram expression stabilizes RNMT protein in T cells and is required for its upregulation on activation. When the Ram gene is deleted in naïve T cells, there are major impacts on activation-induced RNA cap formation and gene expression. Activated T cell proliferation is dependent on increased ribosome production; in Ram knockout T cells, activation-induced expression of ribosomal protein genes and snoRNAs is most severely reduced. Consistent with these changes, Ram deletion resulted in reduced protein synthesis, and reduced growth and proliferation of CD4 T cells. Deletion of Ram results in a similar but milder phenotype to Rnmt deletion, supporting the role of RAM as a RNMT cofactor.

3.
Biochem Soc Trans ; 51(3): 1131-1141, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37145036

RESUMEN

Regulation of RNA cap formation has potent impacts on gene regulation, controlling which transcripts are expressed, processed and translated into protein. Recently, the RNA cap methyltransferases RNA guanine-7 methyltransferase (RNMT) and cap-specific mRNA (nucleoside-2'-O-)-methyltransferase 1 (CMTR1) have been found to be independently regulated during embryonic stem (ES) cell differentiation controlling the expression of overlapping and distinct protein families. During neural differentiation, RNMT is repressed and CMTR1 is up-regulated. RNMT promotes expression of the pluripotency-associated gene products; repression of the RNMT complex (RNMT-RAM) is required for repression of these RNAs and proteins during differentiation. The predominant RNA targets of CMTR1 encode the histones and ribosomal proteins (RPs). CMTR1 up-regulation is required to maintain the expression of histones and RPs during differentiation and to maintain DNA replication, RNA translation and cell proliferation. Thus the co-ordinate regulation of RNMT and CMTR1 is required for different aspects of ES cell differentiation. In this review, we discuss the mechanisms by which RNMT and CMTR1 are independently regulated during ES cell differentiation and explore how this influences the co-ordinated gene regulation required of emerging cell lineages.


Asunto(s)
Metiltransferasas , Caperuzas de ARN , Diferenciación Celular , Histonas/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , Transcripción Genética , Humanos , Animales
4.
Nucleic Acids Res ; 50(5): 2905-2922, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35212377

RESUMEN

CMTR1 (cap methyltransferase 1) catalyses methylation of the first transcribed nucleotide of RNAPII transcripts (N1 2'-O-Me), creating part of the mammalian RNA cap structure. In addition to marking RNA as self, N1 2'-O-Me has ill-defined roles in RNA expression and translation. Here, we investigated the gene specificity of CMTR1 and its impact on RNA expression in embryonic stem cells. Using chromatin immunoprecipitation, CMTR1 was found to bind to transcription start sites (TSS) correlating with RNAPII levels, predominantly binding at histone genes and ribosomal protein (RP) genes. Repression of CMTR1 expression resulted in repression of RNAPII binding at the TSS and repression of RNA expression, particularly of histone and RP genes. In correlation with regulation of histones and RP genes, CMTR1 repression resulted in repression of translation and induction of DNA replication stress and damage. Indicating a direct role for CMTR1 in transcription, addition of recombinant CMTR1 to purified nuclei increased transcription of the histone and RP genes. CMTR1 was found to be upregulated during neural differentiation and there was an enhanced requirement for CMTR1 for gene expression and proliferation during this process. We highlight the distinct roles of the cap methyltransferases RNMT and CMTR1 in target gene expression and differentiation.


Asunto(s)
Células Madre Embrionarias , Histonas , Metiltransferasas , Proteínas Ribosómicas , Animales , Células Madre Embrionarias/metabolismo , Expresión Génica , Histonas/genética , Histonas/metabolismo , Mamíferos/genética , Caperuzas de ARN/genética , ARN Polimerasa II/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética
5.
J Mol Biol ; 434(5): 167451, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35026230

RESUMEN

The control of RNA metabolism is an important aspect of molecular biology with wide-ranging impacts on cells. Central to processing of coding RNAs is the addition of the methyl-7 guanosine (m7G) "cap" on their 5' end. The eukaryotic translation initiation factor eIF4E directly binds the m7G cap and through this interaction plays key roles in many steps of RNA metabolism including nuclear RNA export and translation. eIF4E also stimulates capping of many transcripts through its ability to drive the production of the enzyme RNMT which methylates the G-cap to form the mature m7G cap. Here, we found that eIF4E also physically associated with RNMT in human cells. Moreover, eIF4E directly interacted with RNMT in vitro. eIF4E is only the second protein reported to directly bind the methyltransferase domain of RNMT, the first being its co-factor RAM. We combined high-resolution NMR methods with biochemical studies to define the binding interfaces for the RNMT-eIF4E complex. Further, we found that eIF4E competes for RAM binding to RNMT and conversely, RNMT competes for binding of well-established eIF4E-binding partners such as the 4E-BPs. RNMT uses novel structural means to engage eIF4E. Finally, we observed that m7G cap-eIF4E-RNMT trimeric complexes form, and thus RNMT-eIF4E complexes may be employed so that eIF4E captures newly capped RNA. In all, we show for the first time that the cap-binding protein eIF4E directly binds to the cap-maturation enzyme RNMT.


Asunto(s)
Factor 4E Eucariótico de Iniciación , Caperuzas de ARN , Factor 4E Eucariótico de Iniciación/genética , Guanosina/metabolismo , Humanos , Metiltransferasas/metabolismo , Unión Proteica , Proteínas de Unión a Caperuzas de ARN/genética , Proteínas de Unión a Caperuzas de ARN/metabolismo , Caperuzas de ARN/química , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo
6.
Biochem J ; 478(13): 2481-2497, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34198328

RESUMEN

The COVID-19 pandemic has presented itself as one of the most critical public health challenges of the century, with SARS-CoV-2 being the third member of the Coronaviridae family to cause a fatal disease in humans. There is currently only one antiviral compound, remdesivir, that can be used for the treatment of COVID-19. To identify additional potential therapeutics, we investigated the enzymatic proteins encoded in the SARS-CoV-2 genome. In this study, we focussed on the viral RNA cap methyltransferases, which play key roles in enabling viral protein translation and facilitating viral escape from the immune system. We expressed and purified both the guanine-N7 methyltransferase nsp14, and the nsp16 2'-O-methyltransferase with its activating cofactor, nsp10. We performed an in vitro high-throughput screen for inhibitors of nsp14 using a custom compound library of over 5000 pharmaceutical compounds that have previously been characterised in either clinical or basic research. We identified four compounds as potential inhibitors of nsp14, all of which also showed antiviral capacity in a cell-based model of SARS-CoV-2 infection. Three of the four compounds also exhibited synergistic effects on viral replication with remdesivir.


Asunto(s)
Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Exorribonucleasas/antagonistas & inhibidores , Metiltransferasas/antagonistas & inhibidores , Caperuzas de ARN/metabolismo , SARS-CoV-2/enzimología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Animales , Antivirales/química , Clorobencenos/farmacología , Chlorocebus aethiops , Pruebas de Enzimas , Exorribonucleasas/genética , Exorribonucleasas/aislamiento & purificación , Exorribonucleasas/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Indazoles/farmacología , Indenos/farmacología , Indoles/farmacología , Metiltransferasas/genética , Metiltransferasas/aislamiento & purificación , Metiltransferasas/metabolismo , Nitrilos/farmacología , Fenotiazinas/farmacología , Purinas/farmacología , Reproducibilidad de los Resultados , SARS-CoV-2/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Especificidad por Sustrato , Trifluperidol/farmacología , Células Vero , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/aislamiento & purificación , Proteínas no Estructurales Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/aislamiento & purificación , Proteínas Reguladoras y Accesorias Virales/metabolismo
7.
Nucleic Acids Res ; 49(12): 6722-6738, 2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34125914

RESUMEN

The m7G cap is ubiquitous on RNAPII-transcribed RNA and has fundamental roles in eukaryotic gene expression, however its in vivo role in mammals has remained unknown. Here, we identified the m7G cap methyltransferase, RNMT, as a key mediator of T cell activation, which specifically regulates ribosome production. During T cell activation, induction of mRNA expression and ribosome biogenesis drives metabolic reprogramming, rapid proliferation and differentiation generating effector populations. We report that RNMT is induced by T cell receptor (TCR) stimulation and co-ordinates the mRNA, snoRNA and rRNA production required for ribosome biogenesis. Using transcriptomic and proteomic analyses, we demonstrate that RNMT selectively regulates the expression of terminal polypyrimidine tract (TOP) mRNAs, targets of the m7G-cap binding protein LARP1. The expression of LARP1 targets and snoRNAs involved in ribosome biogenesis is selectively compromised in Rnmt cKO CD4 T cells resulting in decreased ribosome synthesis, reduced translation rates and proliferation failure. By enhancing ribosome abundance, upregulation of RNMT co-ordinates mRNA capping and processing with increased translational capacity during T cell activation.


Asunto(s)
Activación de Linfocitos , Metiltransferasas/fisiología , Biosíntesis de Proteínas , Ribosomas/metabolismo , Linfocitos T/enzimología , Animales , Técnicas de Inactivación de Genes , Guanosina/metabolismo , Activación de Linfocitos/genética , Metiltransferasas/biosíntesis , Metiltransferasas/genética , Ratones , Caperuzas de ARN/química , Caperuzas de ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/química , ARN Mensajero/metabolismo , ARN Pequeño no Traducido/metabolismo , Proteínas de Unión al ARN/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Regulación hacia Arriba
8.
Nucleic Acids Res ; 49(6): 3109-3126, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33684220

RESUMEN

Recruitment of the mRNA capping enzyme (CE/RNGTT) to the site of transcription is essential for the formation of the 5' mRNA cap, which in turn ensures efficient transcription, splicing, polyadenylation, nuclear export and translation of mRNA in eukaryotic cells. The CE GTase is recruited and activated by the Serine-5 phosphorylated carboxyl-terminal domain (CTD) of RNA polymerase II. Through the use of molecular dynamics simulations and enhanced sampling techniques, we provide a systematic and detailed characterization of the human CE-CTD interface, describing the effect of the CTD phosphorylation state, length and orientation on this interaction. Our computational analyses identify novel CTD interaction sites on the human CE GTase surface and quantify their relative contributions to CTD binding. We also identify, for the first time, allosteric connections between the CE GTase active site and the CTD binding sites, allowing us to propose a mechanism for allosteric activation. Through binding and activity assays we validate the novel CTD binding sites and show that the CDS2 site is essential for CE GTase activity stimulation. Comparison of the novel sites with cocrystal structures of the CE-CTD complex in different eukaryotic taxa reveals that this interface is considerably more conserved than previous structures have indicated.


Asunto(s)
Nucleotidiltransferasas/química , ARN Polimerasa II/química , Regulación Alostérica , Animales , Sitios de Unión , Dominio Catalítico , Activación Enzimática , Humanos , Ratones , Simulación de Dinámica Molecular , Nucleotidiltransferasas/metabolismo , Fosforilación , Fosfoserina/química , Fosfoserina/metabolismo , Phycodnaviridae/enzimología , Unión Proteica , Conformación Proteica , Dominios Proteicos , ARN Polimerasa II/metabolismo
9.
SLAS Discov ; 26(6): 749-756, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33724070

RESUMEN

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) represents a significant threat to human health. Despite its similarity to related coronaviruses, there are currently no specific treatments for COVID-19 infection, and therefore there is an urgent need to develop therapies for this and future coronavirus outbreaks. Formation of the cap at the 5' end of viral RNA has been shown to help coronaviruses evade host defenses. Nonstructural protein 14 (nsp14) is responsible for N7-methylation of the cap guanosine in coronaviruses. This enzyme is highly conserved among coronaviruses and is a bifunctional protein with both N7-methyltransferase and 3'-5' exonuclease activities that distinguish nsp14 from its human equivalent. Mutational analysis of SARS-CoV nsp14 highlighted its role in viral replication and translation efficiency of the viral genome. In this paper, we describe the characterization and development of a high-throughput assay for nsp14 utilizing RapidFire technology. The assay has been used to screen a library of 1771 Food and Drug Administration (FDA)-approved drugs. From this, we have validated nitazoxanide as a selective inhibitor of the methyltransferase activity of nsp14. Although modestly active, this compound could serve as a starting point for further optimization.


Asunto(s)
Antivirales/farmacología , Exorribonucleasas/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Nitrocompuestos/farmacología , Caperuzas de ARN/antagonistas & inhibidores , ARN Viral/antagonistas & inhibidores , SARS-CoV-2/efectos de los fármacos , Tiazoles/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antiparasitarios/química , Antiparasitarios/farmacología , Antivirales/química , COVID-19/virología , Clonación Molecular , Reposicionamiento de Medicamentos , Pruebas de Enzimas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Escherichia coli/genética , Escherichia coli/metabolismo , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Cinética , Espectrometría de Masas/métodos , Metilación , Nitrocompuestos/química , Medicamentos bajo Prescripción/química , Medicamentos bajo Prescripción/farmacología , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , SARS-CoV-2/enzimología , SARS-CoV-2/genética , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Tiazoles/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
10.
Proc Natl Acad Sci U S A ; 117(43): 26773-26783, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33055213

RESUMEN

Methyl-7-guanosine (m7G) "capping" of coding and some noncoding RNAs is critical for their maturation and subsequent activity. Here, we discovered that eukaryotic translation initiation factor 4E (eIF4E), itself a cap-binding protein, drives the expression of the capping machinery and increased capping efficiency of ∼100 coding and noncoding RNAs. To quantify this, we developed enzymatic (cap quantification; CapQ) and quantitative cap immunoprecipitation (CapIP) methods. The CapQ method has the further advantage that it captures information about capping status independent of the type of 5' cap, i.e., it is not restricted to informing on m7G caps. These methodological advances led to unanticipated revelations: 1) Many RNA populations are inefficiently capped at steady state (∼30 to 50%), and eIF4E overexpression increased this to ∼60 to 100%, depending on the RNA; 2) eIF4E physically associates with noncoding RNAs in the nucleus; and 3) approximately half of eIF4E-capping targets identified are noncoding RNAs. eIF4E's association with noncoding RNAs strongly positions it to act beyond translation. Coding and noncoding capping targets have activities that influence survival, cell morphology, and cell-to-cell interaction. Given that RNA export and translation machineries typically utilize capped RNA substrates, capping regulation provides means to titrate the protein-coding capacity of the transcriptome and, for noncoding RNAs, to regulate their activities. We also discovered a cap sensitivity element (CapSE) which conferred eIF4E-dependent capping sensitivity. Finally, we observed elevated capping for specific RNAs in high-eIF4E leukemia specimens, supporting a role for cap dysregulation in malignancy. In all, levels of capping RNAs can be regulated by eIF4E.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Guanosina/análogos & derivados , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Línea Celular Tumoral , Factor 4E Eucariótico de Iniciación/química , Factor 4E Eucariótico de Iniciación/genética , Guanosina/química , Guanosina/genética , Guanosina/metabolismo , Humanos , Polirribosomas/metabolismo , Caperuzas de ARN/química , Caperuzas de ARN/genética , ARN Mensajero/química , ARN Mensajero/genética , Transcriptoma/genética
11.
Emerg Med J ; 37(9): 567-570, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32669319

RESUMEN

For many of us in emergency medicine, rising to the challenge of the COVID-19 crisis will be the single most exciting and challenging episode of our careers. Lessons have been learnt on how to make quick and effective changes without being hindered by the normal restraints of bureaucracy. Changes that would normally have taken months to years to implement have been successfully introduced over a period of several weeks. Although we have managed these changes largely by command and control, compassionate leadership has identified leaders within our team and paved the way for the future. This article covers the preparation and changes made in response to COVID-19 in a London teaching hospital.


Asunto(s)
Defensa Civil , Infecciones por Coronavirus , Servicio de Urgencia en Hospital , Innovación Organizacional , Pandemias , Neumonía Viral , Planificación Estratégica , Capacidad de Reacción , Betacoronavirus , COVID-19 , Gestión del Cambio , Defensa Civil/métodos , Defensa Civil/organización & administración , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/prevención & control , Eficiencia Organizacional , Servicio de Urgencia en Hospital/organización & administración , Servicio de Urgencia en Hospital/tendencias , Humanos , Liderazgo , Londres , Pandemias/prevención & control , Neumonía Viral/epidemiología , Neumonía Viral/prevención & control , SARS-CoV-2
12.
Chemistry ; 26(49): 11266-11275, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32259329

RESUMEN

In eukaryotes, mature mRNA is formed through modifications of precursor mRNA, one of which is 5' cap biosynthesis, involving RNA cap guanine-N7 methyltransferase (N7-MTase). N7-MTases are also encoded by some eukaryotic viruses and facilitate their replication. N7-MTase inhibitors have therapeutic potential, but their discovery is difficult because long RNA substrates are usually required for activity. Herein, we report a universal N7-MTase activity assay based on small-molecule fluorescent probes. We synthesized 12 fluorescent substrate analogues (GpppA and GpppG derivatives) varying in the dye type, dye attachment site, and linker length. GpppA labeled with pyrene at the 3'-O position of adenosine acted as an artificial substrate with the properties of a turn-off probe for all three tested N7-MTases (human, parasite, and viral). Using this compound, a N7-MTase inhibitor assay adaptable to high-throughput screening was developed and used to screen synthetic substrate analogues and a commercial library. Several inhibitors with nanomolar activities were identified.


Asunto(s)
Evaluación Preclínica de Medicamentos , Pruebas de Enzimas , Inhibidores Enzimáticos/aislamiento & purificación , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Metiltransferasas/metabolismo , Caperuzas de ARN/metabolismo , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Metiltransferasas/antagonistas & inhibidores , Caperuzas de ARN/química
13.
Open Biol ; 10(2): 190306, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32097574

RESUMEN

Eukaryotic messenger RNA (mRNA) is modified by the addition of an inverted guanosine cap to the 5' triphosphate. The cap guanosine and initial transcribed nucleotides are further methylated by a series of cap methyltransferases to generate the mature cap structures which protect RNA from degradation and recruit proteins involved in RNA processing and translation. Research demonstrating that the cap methyltransferases are regulated has generated interest in determining the methylation status of the mRNA cap structures present in cells. Here, we present CAP-MAP: cap analysis protocol with minimal analyte processing, a rapid and sensitive method for detecting cap structures present in mRNA isolated from tissues or cultured cells.


Asunto(s)
Hígado/citología , Metiltransferasas/metabolismo , Caperuzas de ARN/análisis , Animales , Células Cultivadas , Cromatografía Liquida , Guanosina/metabolismo , Hígado/química , Espectrometría de Masas , Ratones , Estructura Molecular , Caperuzas de ARN/química
14.
Nucleic Acids Res ; 47(16): 8675-8692, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31329932

RESUMEN

The RNA guanine-N7 methyltransferase (RNMT) in complex with RNMT-activating miniprotein (RAM) catalyses the formation of a N7-methylated guanosine cap structure on the 5' end of nascent RNA polymerase II transcripts. The mRNA cap protects the primary transcript from exonucleases and recruits cap-binding complexes that mediate RNA processing, export and translation. By using microsecond standard and accelerated molecular dynamics simulations, we provide for the first time a detailed molecular mechanism of allosteric regulation of RNMT by RAM. We show that RAM selects the RNMT active site conformations that are optimal for binding of substrates (AdoMet and the cap), thus enhancing their affinity. Furthermore, our results strongly suggest the likely scenario in which the cap binding promotes the subsequent AdoMet binding, consistent with the previously suggested cooperative binding model. By employing the network community analyses, we revealed the underlying long-range allosteric networks and paths that are crucial for allosteric regulation by RAM. Our findings complement and explain previous experimental data on RNMT activity. Moreover, this study provides the most complete description of the cap and AdoMet binding poses and interactions within the enzyme's active site. This information is critical for the drug discovery efforts that consider RNMT as a promising anti-cancer target.


Asunto(s)
Metiltransferasas/química , Caperuzas de ARN/química , Proteínas de Unión al ARN/química , S-Adenosilhomocisteína/química , S-Adenosilmetionina/química , Regulación Alostérica , Secuencia de Aminoácidos , Sitios de Unión , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Cinética , Metiltransferasas/genética , Metiltransferasas/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Termodinámica , Transcripción Genética
15.
Open Biol ; 9(4): 190052, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30991934

RESUMEN

Basic mechanisms in gene expression are currently being investigated as targets in cancer therapeutics. One such fundamental process is the addition of the cap to pre-mRNA, which recruits mediators of mRNA processing and translation initiation. Maturation of the cap involves mRNA cap guanosine N-7 methylation, catalysed by RNMT (RNA guanine-7 methyltransferase). In a panel of breast cancer cell lines, we investigated whether all are equivalently dependent on RNMT for proliferation. When cellular RNMT activity was experimentally reduced by 50%, the proliferation rate of non-transformed mammary epithelial cells was unchanged, whereas a subset of breast cancer cell lines exhibited reduced proliferation and increased apoptosis. Most of the cell lines which exhibited enhanced dependency on RNMT harboured oncogenic mutations in PIK3CA, which encodes the p110α subunit of PI3Kα. Conversely, all cell lines insensitive to RNMT depletion expressed wild-type PIK3CA. Expression of oncogenic PIK3CA mutants, which increase PI3K p110α activity, was sufficient to increase dependency on RNMT. Conversely, inhibition of PI3Kα reversed dependency on RNMT, suggesting that PI3Kα signalling is required. Collectively, these findings provide evidence to support RNMT as a therapeutic target in breast cancer and suggest that therapies targeting RNMT would be most valuable in a PIK3CA mutant background.


Asunto(s)
Neoplasias de la Mama/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Metiltransferasas/genética , Mutación , Caperuzas de ARN , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Humanos , Células MCF-7 , Metiltransferasas/metabolismo , ARN Mensajero/genética , Transducción de Señal/genética , Transcripción Genética
16.
Trends Biochem Sci ; 44(3): 183-185, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30679132

RESUMEN

The mRNA cap is a structure that protects mRNA from degradation and recruits processing and translation factors. A new mRNA capping enzyme has been identified, PCIF1/CAPAM, which methylates adenosine when it is the first transcribed nucleotide. This discovery is crucial for understanding the function of cap adenosine methylation.


Asunto(s)
Metiltransferasas/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Adenosina/metabolismo , Animales , Humanos , Metilación
17.
Biochim Biophys Acta Gene Regul Mech ; 1862(3): 270-279, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30312682

RESUMEN

In this review we explore the regulation of mRNA cap formation and its impact on mammalian cells. The mRNA cap is a highly methylated modification of the 5' end of RNA pol II-transcribed RNA. It protects RNA from degradation, recruits complexes involved in RNA processing, export and translation initiation, and marks cellular mRNA as "self" to avoid recognition by the innate immune system. The mRNA cap can be viewed as a unique mark which selects RNA pol II transcripts for specific processing and translation. Over recent years, examples of regulation of mRNA cap formation have emerged, induced by oncogenes, developmental pathways and during the cell cycle. These signalling pathways regulate the rate and extent of mRNA cap formation, resulting in changes in gene expression, cell physiology and cell function.


Asunto(s)
Caperuzas de ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Animales , Diferenciación Celular , Humanos , Nucleotidiltransferasas/metabolismo , ARN Mensajero/genética , Transducción de Señal
18.
Life Sci Alliance ; 1(3): e201800092, 2018 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-30079402

RESUMEN

CMTR1 contributes to mRNA cap formation by methylating the first transcribed nucleotide ribose at the O-2 position. mRNA cap O-2 methylation has roles in mRNA stabilisation and translation, and self-RNA tolerance in innate immunity. We report that CMTR1 is recruited to serine-5-phosphorylated RNA Pol II C-terminal domain, early in transcription. We isolated CMTR1 in a complex with DHX15, an RNA helicase functioning in splicing and ribosome biogenesis, and characterised it as a regulator of CMTR1. When DHX15 is bound, CMTR1 activity is repressed and the methyl-transferase does not bind to RNA pol II. Conversely, CMTR1 activates DHX15 helicase activity, which is likely to impact several nuclear functions. In HCC1806 breast carcinoma cell line, the DHX15-CMTR1 interaction controls ribosome loading of a subset of mRNAs and regulates cell proliferation. The impact of the CMTR1-DHX15 interaction is complex and will depend on the relative expression of these enzymes and their interactors, and the cellular dependency on different RNA processing pathways.

19.
PLoS One ; 13(7): e0201263, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30040830

RESUMEN

Messenger RNA is modified by the addition of a 5' methylated cap structure, which protects the transcript and recruits protein complexes that mediate RNA processing and/or the initiation of translation. Two genes encoding mRNA cap methyltransferases have been identified in T. brucei: TbCMT1 and TbCGM1. Here we analysed the impact of TbCMT1 gene deletion on bloodstream form T. brucei cells. TbCMT1 was dispensable for parasite proliferation in in vitro culture. However, significantly decreased parasitemia was observed in mice inoculated with TbCMT1 null and conditional null cell lines. Using RNA-Seq, we observed that several cysteine peptidase mRNAs were downregulated in TbCMT1 null cells lines. The cysteine peptidase Cathepsin-L was also shown to be reduced at the protein level in TbCMT1 null cell lines. Our data suggest that TbCMT1 is not essential to bloodstream form T. brucei growth in vitro or in vivo but that it contributes significantly to parasite virulence in vivo.


Asunto(s)
Metiltransferasas/genética , Proteínas Protozoarias/genética , Caperuzas de ARN/genética , ARN Protozoario/genética , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/patogenicidad , Tripanosomiasis Africana/parasitología , Animales , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Ratones Endogámicos BALB C , Trypanosoma brucei brucei/crecimiento & desarrollo , Tripanosomiasis Africana/patología , Tripanosomiasis Africana/veterinaria , Virulencia , Factores de Virulencia/genética
20.
Cell Rep ; 23(5): 1530-1542, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29719263

RESUMEN

mRNA cap addition occurs early during RNA Pol II-dependent transcription, facilitating pre-mRNA processing and translation. We report that the mammalian mRNA cap methyltransferase, RNMT-RAM, promotes RNA Pol II transcription independent of mRNA capping and translation. In cells, sublethal suppression of RNMT-RAM reduces RNA Pol II occupancy, net mRNA synthesis, and pre-mRNA levels. Conversely, expression of RNMT-RAM increases transcription independent of cap methyltransferase activity. In isolated nuclei, recombinant RNMT-RAM stimulates transcriptional output; this requires the RAM RNA binding domain. RNMT-RAM interacts with nascent transcripts along their entire length and with transcription-associated factors including the RNA Pol II subunits SPT4, SPT6, and PAFc. Suppression of RNMT-RAM inhibits transcriptional markers including histone H2BK120 ubiquitination, H3K4 and H3K36 methylation, RNA Pol II CTD S5 and S2 phosphorylation, and PAFc recruitment. These findings suggest that multiple interactions among RNMT-RAM, RNA Pol II factors, and RNA along the transcription unit stimulate transcription.


Asunto(s)
Metiltransferasas/metabolismo , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética/fisiología , Células HEK293 , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Metiltransferasas/genética , ARN Polimerasa II/genética , Proteínas de Unión al ARN/genética , Ubiquitinación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...