Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 629
Filtrar
1.
Cell Metab ; 34(4): 564-580.e8, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385705

RESUMEN

Hepatokines, secretory proteins from the liver, mediate inter-organ communication to maintain a metabolic balance between food intake and energy expenditure. However, molecular mechanisms by which hepatokine levels are rapidly adjusted following stimuli are largely unknown. Here, we unravel how CNOT6L deadenylase switches off hepatokine expression after responding to stimuli (e.g., exercise and food) to orchestrate energy intake and expenditure. Mechanistically, CNOT6L inhibition stabilizes hepatic Gdf15 and Fgf21 mRNAs, increasing corresponding serum protein levels. The resulting upregulation of GDF15 stimulates the hindbrain to suppress appetite, while increased FGF21 affects the liver and adipose tissues to induce energy expenditure and lipid consumption. Despite the potential of hepatokines to treat metabolic disorders, their administration therapies have been challenging. Using small-molecule screening, we identified a CNOT6L inhibitor enhancing GDF15 and FGF21 hepatokine levels, which dramatically improves diet-induced metabolic syndrome. Our discovery, therefore, lays the foundation for an unprecedented strategy to treat metabolic syndrome.


Asunto(s)
Síndrome Metabólico , Estabilidad del ARN , Animales , Ingestión de Alimentos , Metabolismo Energético/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Hígado/metabolismo , Síndrome Metabólico/metabolismo , Ratones , Estabilidad del ARN/genética , Estabilidad del ARN/fisiología , Ribonucleasas/metabolismo
2.
Elife ; 112022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35103592

RESUMEN

Accurate control of the cell cycle is critical for development and tissue homeostasis, and requires precisely timed expression of many genes. Cell cycle gene expression is regulated through transcriptional and translational control, as well as through regulated protein degradation. Here, we show that widespread and temporally controlled mRNA decay acts as an additional mechanism for gene expression regulation during the cell cycle in human cells. We find that two waves of mRNA decay occur sequentially during the mitosis-to-G1 phase transition, and we identify the deadenylase CNOT1 as a factor that contributes to mRNA decay during this cell cycle transition. Collectively, our data show that, akin to protein degradation, scheduled mRNA decay helps to reshape cell cycle gene expression as cells move from mitosis into G1 phase.


Asunto(s)
Ciclo Celular/genética , Ciclo Celular/fisiología , Estabilidad del ARN/fisiología , Línea Celular , Regulación de la Expresión Génica , Células HEK293 , Humanos , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35177478

RESUMEN

The role of N6-methyladenosine (m6A) modifications has increasingly been associated with a diverse set of roles in modulating viruses and influencing the outcomes of viral infection. Here, we report that the landscape of m6A deposition is drastically shifted during Kaposi's sarcoma-associated herpesvirus (KSHV) lytic infection for both viral and host transcripts. In line with previous reports, we also saw an overall decrease in host methylation in favor of viral messenger RNA (mRNA), along with 5' hypomethylation and 3' hypermethylation. During KSHV lytic infection, a major shift in overall mRNA abundance is driven by the viral endoribonuclease SOX, which induces the decay of greater than 70% of transcripts. Here, we reveal that interlukin-6 (IL-6) mRNA, a well-characterized, SOX-resistant transcript, is m6A modified during lytic infection. Furthermore, we show that this modification falls within the IL-6 SOX resistance element, an RNA element in the IL-6 3' untranslated region (UTR) that was previously shown to be sufficient for protection from SOX cleavage. We show that the presence of this m6A modification is essential to confer SOX resistance to the IL-6 mRNA. We next show that this modification recruits the m6A reader YTHDC2 and found that YTHDC2 is necessary for the escape of the IL-6 transcript. These results shed light on how the host cell has evolved to use RNA modifications to circumvent viral manipulation of RNA fate during KSHV infection.


Asunto(s)
Endorribonucleasas/metabolismo , ARN Helicasas/metabolismo , Estabilidad del ARN/fisiología , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/metabolismo , Línea Celular Tumoral , Endorribonucleasas/genética , Expresión Génica/genética , Regulación Viral de la Expresión Génica/genética , Células HEK293 , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Metilación , ARN Helicasas/genética , Estabilidad del ARN/genética , ARN Mensajero/metabolismo , ARN Viral/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
4.
Chem Biol Interact ; 351: 109705, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34656559

RESUMEN

Non-small cell lung cancer (NSCLC) is one of the prevalent and deadly cancers worldwide. Cisplatin (CDDP) has been used as a standard adjuvant therapy for advanced NSCLC patients, while chemoresistance is one of the most challenging problems to limit its clinical application. Our data showed that the expression of visfatin was significantly increased in CDDP resistant NSCLC cells as compared with that in their parental cells, while knockdown of visfatin or its neutralization antibody can restore the CDDP sensitivity of resistant NSCLC cells. The upregulation of visfatin in CDDP resistant NSCLC cells was due to the increased mRNA stability and promoter activity. Further, we found that signal transducer and activator of transcription 3 (STAT3), which was increased in chemoresistant cells, can increase the transcription of visfatin. While tristetraprolin (TTP), which can decease mRNA stability of visfatin, was decreased in chemoresistant cells. Inhibition of STAT3 or over expression of TTP can restore CDDP sensitivity of resistant NSCLC cells. Collectively, our data showed that STAT3 and TTP-regulated expression of visfatin was involved in CDDP resistance of NSCLC cells. It indicated that targeted inhibition of visfatin should be a potential approach to overcome CDDP resistance of NSCLC treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Citocinas/metabolismo , Resistencia a Antineoplásicos/fisiología , Neoplasias Pulmonares/fisiopatología , Nicotinamida Fosforribosiltransferasa/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Estabilidad del ARN/fisiología , Factor de Transcripción STAT3/metabolismo , Tristetraprolina/metabolismo , Regulación hacia Arriba/fisiología
5.
Crit Rev Biochem Mol Biol ; 57(1): 48-72, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34547957

RESUMEN

Detailed studies of the Gram-negative model bacterium, Escherichia coli, have demonstrated that post-transcriptional events exert important and possibly greater control over gene regulation than transcription initiation or effective translation. Thus, over the past 30 years, considerable effort has been invested in understanding the pathways of mRNA turnover in E. coli. Although it is assumed that most of the ribonucleases and accessory proteins involved in mRNA decay have been identified, our understanding of the regulation of mRNA decay is still incomplete. Furthermore, the vast majority of the studies on mRNA decay have been conducted on exponentially growing cells. Thus, the mechanism of mRNA decay as currently outlined may not accurately reflect what happens when cells find themselves under a variety of stress conditions, such as, nutrient starvation, changes in pH and temperature, as well as a host of others. While the cellular machinery for degradation is relatively constant over a wide range of conditions, intracellular levels of specific ribonucleases can vary depending on the growth conditions. Substrate competition will also modulate ribonucleolytic activity. Post-transcriptional modifications of transcripts by polyadenylating enzymes may favor a specific ribonuclease activity. Interactions with small regulatory RNAs and RNA binding proteins add additional complexities to mRNA functionality and stability. Since many of the ribonucleases are found at the inner membrane, the physical location of a transcript may help determine its half-life. Here we discuss the properties and role of the enzymes involved in mRNA decay as well as the multiple factors that may affect mRNA decay under various in vivo conditions.


Asunto(s)
Escherichia coli , ARN Bacteriano , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Estabilidad del ARN/fisiología , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleasas/metabolismo
6.
Int J Mol Sci ; 22(24)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34948199

RESUMEN

The genome is pervasively transcribed across various species, yielding numerous non-coding RNAs. As a counterbalance for pervasive transcription, various organisms have a nuclear RNA exosome complex, whose structure is well conserved between yeast and mammalian cells. The RNA exosome not only regulates the processing of stable RNA species, such as rRNAs, tRNAs, small nucleolar RNAs, and small nuclear RNAs, but also plays a central role in RNA surveillance by degrading many unstable RNAs and misprocessed pre-mRNAs. In addition, associated cofactors of RNA exosome direct the exosome to distinct classes of RNA substrates, suggesting divergent and/or multi-layer control of RNA quality in the cell. While the RNA exosome is essential for cell viability and influences various cellular processes, mutations and alterations in the RNA exosome components are linked to the collection of rare diseases and various diseases including cancer, respectively. The present review summarizes the relationships between pervasive transcription and RNA exosome, including evolutionary crosstalk, mechanisms of RNA exosome-mediated RNA surveillance, and physiopathological effects of perturbation of RNA exosome.


Asunto(s)
Complejo Multienzimático de Ribonucleasas del Exosoma/fisiología , Estabilidad del ARN/fisiología , Transcripción Genética/genética , Animales , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Genoma/genética , Humanos , ARN/genética , ARN/metabolismo , Estabilidad del ARN/genética , ARN Nuclear/genética , ARN Nuclear/metabolismo
7.
Nat Commun ; 12(1): 7006, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34853315

RESUMEN

Nicotine addiction and the occurrence of lymph node spread are two major significant factors associated with esophageal cancer's poor prognosis; however, nicotine's role in inducing lymphatic metastasis of esophageal cancer remains unclear. Here we show that OTU domain-containing protein 3 (OTUD3) is downregulated by nicotine and correlates with poor prognosis in heavy-smoking esophageal cancer patients. OTUD3 directly interacts with ZFP36 ring finger protein (ZFP36) and stabilizes it by inhibiting FBXW7-mediated K48-linked polyubiquitination. ZFP36 binds with the VEGF-C 3-'UTR and recruits the RNA degrading complex to induce its rapid mRNA decay. Downregulation of OTUD3 and ZFP36 is essential for nicotine-induced VEGF-C production and lymphatic metastasis in esophageal cancer. This study establishes that the OTUD3/ZFP36/VEGF-C axis plays a vital role in nicotine addiction-induced lymphatic metastasis, suggesting that OTUD3 may serve as a prognostic marker, and induction of the VEGF-C mRNA decay might be a potential therapeutic strategy against human esophageal cancer.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Neoplasias Esofágicas/metabolismo , Metástasis Linfática , Nicotina/farmacología , Estabilidad del ARN/fisiología , Proteasas Ubiquitina-Específicas/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Inductores de la Angiogénesis/farmacología , Animales , Línea Celular Tumoral , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/metabolismo , Tristetraprolina/metabolismo , Proteasas Ubiquitina-Específicas/genética , Factor C de Crecimiento Endotelial Vascular/genética
8.
J Virol ; 95(20): e0103421, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34346764

RESUMEN

One of the many challenges faced by RNA viruses is the maintenance of their genomes during infections of host cells. Members of the family Tombusviridae are plus-strand RNA viruses with unmodified triphosphorylated genomic 5' termini. The tombusvirus Carnation Italian ringspot virus was used to investigate how it protects its RNA genome from attack by 5'-end-targeting degradation enzymes. In vivo and in vitro assays were employed to determine the role of genomic RNA structure in conferring protection from the 5'-to-3' exoribonuclease Xrn. The results revealed that (i) the CIRV RNA genome is more resistant to Xrn than its sg mRNAs, (ii) the genomic 5'-untranslated region (UTR) folds into a compact RNA structure that effectively and independently prevents Xrn access, (iii) the RNA structure limiting 5' access is formed by secondary and tertiary interactions that function cooperatively, (iv) the structure is also able to block access of RNA pyrophosphohydrolase to the genomic 5' terminus, and (v) the RNA structure does not stall an actively digesting Xrn. Based on its proficiency at impeding Xrn 5' access, we have termed this 5'-terminal structure an Xrn-evading RNA, or xeRNA. These and other findings demonstrate that the 5'UTR of the CIRV RNA genome folds into a complex structural conformation that helps to protect its unmodified 5' terminus from enzymatic decay during infections. IMPORTANCE The plus-strand RNA genomes of plant viruses in the large family Tombusviridae are not 5' capped. Here, we explored how a species in the type genus Tombusvirus protects its genomic 5' end from cellular nuclease attack. Our results revealed that the 5'-terminal sequence of the CIRV genome folds into a complex RNA structure that limits access of the 5'-to-3' exoribonuclease Xrn, thereby protecting it from processive degradation. The RNA conformation also impeded access of RNA pyrophosphohydrolase, which converts 5'-triphosphorylated RNA termini into 5'-monophosphorylated forms, the preferred substrate for Xrn. This study represents the first report of a higher-order RNA structure in an RNA plant virus genome independently conferring resistance to 5'-end-attacking cellular enzymes.


Asunto(s)
Regiones no Traducidas 5'/genética , Estabilidad del ARN/genética , Tombusvirus/genética , Regiones no Traducidas 3'/genética , Secuencia de Bases/genética , Exorribonucleasas , Genoma Viral/genética , Conformación de Ácido Nucleico , Biosíntesis de Proteínas/genética , Estabilidad del ARN/fisiología , Virus ARN/genética , ARN Mensajero/metabolismo , ARN Viral/genética , Ribonucleasas/metabolismo , Relación Estructura-Actividad , Tombusvirus/metabolismo , Proteínas Virales/metabolismo
9.
Neurochem Res ; 46(11): 3003-3011, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34297301

RESUMEN

This study aims to investigate the biological role of 6-methyladenine (m6A) methylation in inducing the carcinogenesis of glioma and its proliferation. Relative levels of ALKBH5 and glucose-6-phosphate dehydrogenase (G6PD) in glioma tissues and cell lines were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. Gain-of-function and loss-of-function approaches were used to investigate the role of ALKBH5 in mediating proliferation and energy metabolism of glioma cells. The regulatory effect of ALKBH5 on G6PD was analyzed using m6A-qRT-PCR. Our results showed that ALKBH5 was upregulated in glioma, which stimulated glioma cells to proliferate. Serving as a m6A eraser, ALKBH5 demethylated the target transcript G6PD and enhanced its mRNA stability, thereby promoting G6PD translation and activating the pentose phosphate pathway (PPP). Collectively, ALKBH5 stimulates glioma cells to proliferate through erasing the m6A methylation of G6PD, which can be utilized as a potential therapeutic target for glioma.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB/biosíntesis , Neoplasias Encefálicas/metabolismo , Proliferación Celular/fisiología , Glioma/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Estabilidad del ARN/fisiología , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Glioma/genética , Glucosafosfato Deshidrogenasa/genética , Humanos
10.
Nat Commun ; 12(1): 4071, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34210974

RESUMEN

Molecular left-right (L-R) asymmetry is established at the node of the mouse embryo as a result of the sensing of a leftward fluid flow by immotile cilia of perinodal crown cells and the consequent degradation of Dand5 mRNA on the left side. We here examined how the fluid flow induces Dand5 mRNA decay. We found that the first 200 nucleotides in the 3' untranslated region (3'-UTR) of Dand5 mRNA are necessary and sufficient for the left-sided decay and to mediate the response of a 3'-UTR reporter transgene to Ca2+, the cation channel Pkd2, the RNA-binding protein Bicc1 and their regulation by the flow direction. We show that Bicc1 preferentially recognizes GACR and YGAC sequences, which can explain the specific binding to a conserved GACGUGAC motif located in the proximal Dand5 3'-UTR. The Cnot3 component of the Ccr4-Not deadenylase complex interacts with Bicc1 and is also required for Dand5 mRNA decay at the node. These results suggest that Ca2+ currents induced by leftward fluid flow stimulate Bicc1 and Ccr4-Not to mediate Dand5 mRNA degradation specifically on the left side of the node.


Asunto(s)
Embrión de Mamíferos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Estabilidad del ARN/fisiología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores CCR4/metabolismo , Regiones no Traducidas 3' , Animales , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/genética , Receptores CCR4/genética , Canales Catiónicos TRPP/metabolismo , Factores de Transcripción
11.
Cells ; 10(6)2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-34202997

RESUMEN

The current SARS-CoV-2 pandemic underscores the importance of understanding the evolution of RNA genomes. While RNA is subject to the formation of similar lesions as DNA, the evolutionary and physiological impacts RNA lesions have on viral genomes are yet to be characterized. Lesions that may drive the evolution of RNA genomes can induce breaks that are repaired by recombination or can cause base substitution mutagenesis, also known as base editing. Over the past decade or so, base editing mutagenesis of DNA genomes has been subject to many studies, revealing that exposure of ssDNA is subject to hypermutation that is involved in the etiology of cancer. However, base editing of RNA genomes has not been studied to the same extent. Recently hypermutation of single-stranded RNA viral genomes have also been documented though its role in evolution and population dynamics. Here, we will summarize the current knowledge of key mechanisms and causes of RNA genome instability covering areas from the RNA world theory to the SARS-CoV-2 pandemic of today. We will also highlight the key questions that remain as it pertains to RNA genome instability, mutations accumulation, and experimental strategies for addressing these questions.


Asunto(s)
Evolución Molecular , ARN Viral/genética , SARS-CoV-2/genética , COVID-19/epidemiología , COVID-19/virología , Genoma Viral/genética , Humanos , Mutación , Pandemias , Edición de ARN/fisiología , Estabilidad del ARN/fisiología
12.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203408

RESUMEN

TENT4A (PAPD7) is a non-canonical poly(A) polymerase, of which little is known. Here, we show that TENT4A regulates multiple biological pathways and focuses on its multilayer regulation of translesion DNA synthesis (TLS), in which error-prone DNA polymerases bypass unrepaired DNA lesions. We show that TENT4A regulates mRNA stability and/or translation of DNA polymerase η and RAD18 E3 ligase, which guides the polymerase to replication stalling sites and monoubiquitinates PCNA, thereby enabling recruitment of error-prone DNA polymerases to damaged DNA sites. Remarkably, in addition to the effect on RAD18 mRNA stability via controlling its poly(A) tail, TENT4A indirectly regulates RAD18 via the tumor suppressor CYLD and via the long non-coding antisense RNA PAXIP1-AS2, which had no known function. Knocking down the expression of TENT4A or CYLD, or overexpression of PAXIP1-AS2 led each to reduced amounts of the RAD18 protein and DNA polymerase η, leading to reduced TLS, highlighting PAXIP1-AS2 as a new TLS regulator. Bioinformatics analysis revealed that TLS error-prone DNA polymerase genes and their TENT4A-related regulators are frequently mutated in endometrial cancer genomes, suggesting that TLS is dysregulated in this cancer.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , Neoplasias Endometriales/metabolismo , Mutación/genética , Polinucleotido Adenililtransferasa/metabolismo , ARN Mensajero/metabolismo , Western Blotting , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/genética , Biología Computacional , Daño del ADN/genética , Daño del ADN/fisiología , Reparación del ADN/genética , Replicación del ADN/genética , Replicación del ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Neoplasias Endometriales/genética , Femenino , Células HEK293 , Humanos , Inmunoprecipitación , Células MCF-7 , Reacción en Cadena de la Polimerasa , Polinucleotido Adenililtransferasa/genética , Estabilidad del ARN/genética , Estabilidad del ARN/fisiología , ARN Mensajero/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/genética , Ubiquitinación/fisiología
13.
PLoS Comput Biol ; 17(7): e1009188, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34297727

RESUMEN

Cellular RNA levels typically fluctuate and are influenced by different transcription rates and RNA degradation rates. However, the understanding of the fundamental relationships between RNA abundance, environmental stimuli, RNA activities, and RNA age distributions is incomplete. Furthermore, the rates of RNA degradation and transcription are difficult to measure in transcriptomic experiments in living organisms, especially in studies involving humans. A model based on activity demands and RNA age was developed to explore the mechanisms of RNA level fluctuations. Using single-cell time-series gene expression experimental data, we assessed the transcription rates, RNA degradation rates, RNA life spans, RNA demand, accumulated transcription levels, and accumulated RNA degradation levels. This model could also predict RNA levels under simulation backgrounds, such as stimuli that induce regular oscillations in RNA abundance, stable RNA levels over time that result from long-term shortage of total RNA activity or from uncontrollable transcription, and relationships between RNA/protein levels and metabolic rates. This information contributes to existing knowledge.


Asunto(s)
Modelos Biológicos , Estabilidad del ARN , ARN Mensajero/metabolismo , Biología Computacional , Simulación por Computador , Estabilidad del ARN/genética , Estabilidad del ARN/fisiología , ARN de Hongos/genética , ARN de Hongos/metabolismo , ARN Mensajero/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Análisis de la Célula Individual , Transcripción Genética , Transcriptoma
14.
J Immunother Cancer ; 9(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34117117

RESUMEN

SARS-CoV-2 infection and the resulting COVID-19 have afflicted millions of people in an ongoing worldwide pandemic. Safe and effective vaccination is needed urgently to protect not only the general population but also vulnerable subjects such as patients with cancer. Currently approved mRNA-based SARS-CoV-2 vaccines seem suitable for patients with cancer based on their mode of action, efficacy, and favorable safety profile reported in the general population. Here, we provide an overview of mRNA-based vaccines including their safety and efficacy. Extrapolating from insights gained from a different preventable viral infection, we review existing data on immunity against influenza A and B vaccines in patients with cancer. Finally, we discuss COVID-19 vaccination in light of the challenges specific to patients with cancer, such as factors that may hinder protective SARS-CoV-2 immune responses in the context of compromised immunity and the use of immune-suppressive or immune-modulating drugs.


Asunto(s)
Vacunas contra la COVID-19 , Neoplasias/terapia , ARN Mensajero , SARS-CoV-2/inmunología , Vacunas Virales , COVID-19/epidemiología , COVID-19/prevención & control , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/uso terapéutico , Estabilidad de Medicamentos , Humanos , Gripe Humana/epidemiología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Neoplasias/epidemiología , Neoplasias/inmunología , Pandemias , Estabilidad del ARN/fisiología , ARN Mensajero/administración & dosificación , ARN Mensajero/efectos adversos , ARN Mensajero/química , ARN Mensajero/genética , SARS-CoV-2/genética , Vacunación/métodos , Vacunas Virales/efectos adversos , Vacunas Virales/química , Vacunas Virales/genética
15.
J Neurochem ; 158(3): 766-778, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34107054

RESUMEN

Trans-active response DNA-binding protein of 43 kDa (TDP-43) promotes tau mRNA instability and tau exon 10 inclusion. Aggregation of phosphorylated TDP-43 is associated with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration. Casein kinase 1ε (CK1ε) phosphorylates TDP-43 at multiple sites, enhances its cytoplasmic aggregation, and modulates its function in tau mRNA processing. To determine roles of TDP-43 site-specific phosphorylation in its localization, aggregation, and function in tau mRNA processing, TDP-43 was mutated to alanine or aspartic acid at Ser379, Ser403/404, or Ser409/410 to block or mimic phosphorylation. Site-specific phosphorylation of TDP-43 and its mutants by CK1ε was studied in vitro and in cultured cells. Cytoplasmic and nuclear TDP-43 and phospho-TDP-43 were analyzed by western blots. Aggregation of TDP-43 was assessed by immunostaining and level of radioimmunoprecipitation assay buffer-insoluble TDP-43. Green florescent protein tailed with tau 3'-untranslated region and mini-tau gene pCI/SI9-LI10 were used to study tau mRNA stability and alternative splicing of tau exon 10. We found that phospho-blocking mutations of TDP-43 at Ser379, Ser403/404, or Ser409/410 were not effectively phosphorylated by CK1ε. Compared with TDP-43, higher level of phosphorylated TDP-43 in the cytoplasm was observed. Phospho-mimicking mutations at these sites enhanced cytoplasmic aggregation of TDP-43. Green florescent protein expression was not inhibited by phospho-blocking mutants of TDP-43, but tau exon 10 inclusion was further enhanced by phospho-blocking mutations at Ser379 and Ser403/404. Phosphorylation of TDP-43 at Ser379, Ser403/404, or Ser409/410 primes its phosphorylation by CK1ε, promotes TDP-43 cytoplasmic aggregation, and modulates its function in tau mRNA processing in site-specific manner.


Asunto(s)
Empalme Alternativo/fisiología , Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Exones/fisiología , Estabilidad del ARN/fisiología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Agregación Celular/fisiología , Proteínas de Unión al ADN/genética , Femenino , Lóbulo Frontal/metabolismo , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Fosforilación/fisiología , Proteínas tau/genética
16.
PLoS One ; 16(5): e0251290, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33983964

RESUMEN

Extracellular vesicles (EVs) have emerged as promising candidates in biomarker discovery and diagnostics. Protected by the lipid bilayer, the molecular content of EVs in diverse biofluids are protected from RNases and proteases in the surrounding environment that may rapidly degrade targets of interests. Nonetheless, cryopreservation of EV-containing samples to -80°C may expose the lipid bilayer to physical and biological stressors which may result in cryoinjury and contribute to changes in EV yield, function, or molecular cargo. In the present work, we systematically evaluate the effect of cryopreservation at -80°C for a relatively short duration of storage (up to 12 days) on plasma- and media-derived EV particle count and/or RNA yield/quality, as compared to paired fresh controls. On average, we found that the plasma-derived EV concentration of stored samples decreased to 23% of fresh samples. Further, this significant decrease in EV particle count was matched with a corresponding significant decrease in RNA yield whereby plasma-derived stored samples contained only 47-52% of the total RNA from fresh samples, depending on the extraction method used. Similarly, media-derived EVs showed a statistically significant decrease in RNA yield whereby stored samples were 58% of the total RNA from fresh samples. In contrast, we did not obtain clear evidence of decreased RNA quality through analysis of RNA traces. These results suggest that samples stored for up to 12 days can indeed produce high-quality RNA; however, we note that when directly comparing fresh versus cryopreserved samples without cryoprotective agents there are significant losses in total RNA. Finally, we demonstrate that the addition of the commonly used cryoprotectant agent, DMSO, alongside greater control of the rate of cooling/warming, can rescue EVs from damaging ice formation and improve RNA yield.


Asunto(s)
Vesículas Extracelulares/metabolismo , ARN/aislamiento & purificación , Manejo de Especímenes/métodos , Criopreservación/métodos , Medios de Cultivo/química , Voluntarios Sanos , Humanos , Plasma/química , ARN/metabolismo , Estabilidad del ARN/efectos de los fármacos , Estabilidad del ARN/fisiología
17.
Mol Cell Biol ; 41(8): e0010421, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-33972392

RESUMEN

N6-Methyladenosine (m6A) is the most prevalent internal RNA modification and has a widespread impact on mRNA stability and translation. Methyltransferase-like 3 (Mettl3) is a methyltransferase responsible for RNA m6A modification, and it is essential for early embryogenesis before or during gastrulation in mice and zebrafish. However, due to the early embryonic lethality, loss-of-function phenotypes of Mettl3 beyond gastrulation, especially during neurulation stages when spatial neural patterning takes place, remain elusive. Here, we address multiple roles of Mettl3 during Xenopus neurulation in anteroposterior neural patterning, neural crest specification, and neuronal cell differentiation. Knockdown of Mettl3 causes anteriorization of neurulae and tailbud embryos along with the loss of neural crest and neuronal cells. Knockdown of the m6A reader Ythdf1 and mRNA degradation factors, such as 3' to 5' exonuclease complex component Lsm1 or mRNA uridylation enzyme Tut7, also show similar neural patterning defects, suggesting that m6A-dependent mRNA destabilization regulates spatial neural patterning in Xenopus. We also address that canonical WNT signaling is inhibited in Mettl3 morphants, which may underlie the neural patterning defects of the morphants. Altogether, this study reveals functions of Mettl3 during spatial neural patterning in Xenopus.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Metiltransferasas/metabolismo , Nucleotidiltransferasas/metabolismo , ARN/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Línea Celular Tumoral , Metiltransferasas/genética , Nucleotidiltransferasas/genética , Estabilidad del ARN/fisiología , Vía de Señalización Wnt/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Pez Cebra/metabolismo
18.
Neuroreport ; 32(9): 824-832, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-33994531

RESUMEN

Amyloid precursor protein (APP) is a transmembrane protein that plays a crucial role in the production of amyloid-ß peptides. Any disruption in APP protein production, its mRNA decay rate or processing may result in abnormal production of amyloid-ß peptides and subsequent development of protein aggregation diseases. Therefore, the equilibrium is crucial for neuronal function. An association study of heterogeneous nuclear ribonucleoprotein (hnRNP)-F and hnRNP H1 with APP was carried out in Neuro-2a (N2a) cells. In the present study, we found that hnRNP F and hnRNP H1 were significantly upregulated in the hippocampus of APP/PS1 mice. The changes in APP expression were positively associated with hnRNP F and hnRNP H1 when hnRNP F and hnRNP H1 were depleted or increased in N2a cells. Importantly, cross-linked RNA immunoprecipitation demonstrated binding affinities of hnRNP F and hnRNP H1 for App mRNA. Mechanistically, mRNA stability assay revealed that overexpression of hnRNP F or hnRNP H1 increases the APP level by stabilizing App mRNA half-life, implying that levels of hnRNP F and hnRNP H1 can change the production of APP. Further understanding of the regulatory mechanism of APP expression in association with hnRNP F and hnRNP H1 would provide insights into the mechanism underlying the maintenance of brain health and cognition. This study provides a theoretical basis for the development of hnRNP-stabilizing compounds to regulate APP.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Hipocampo/metabolismo , Estabilidad del ARN/fisiología , ARN Mensajero/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/genética , Hipocampo/patología , Ratones , Ratones Transgénicos
19.
J Virol ; 95(13): e0033621, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33853967

RESUMEN

To replicate efficiently and evade the antiviral immune response of the host, some viruses degrade host mRNA to induce host gene shutoff via encoding shutoff factors. In this study, we found that feline calicivirus (FCV) infection promotes the degradation of endogenous and exogenous mRNAs and induces host gene shutoff, which results in global inhibition of host protein synthesis. Screening assays revealed that proteinase-polymerase (PP) is a most effective factor in reducing mRNA expression. Moreover, PP from differently virulent strains of FCV could induce mRNA degradation. Further, we found that the key sites of the PP protein required for its proteinase activity are also essential for its shutoff activity but also required for viral replication. The mechanism analysis showed that PP mainly targets Pol II-transcribed RNA in a ribosome-, 5' cap-, and 3' poly(A) tail-independent manner. Moreover, purified glutathione S-transferase (GST)-PP fusion protein exhibits RNase activity in vitro in assays using green fluorescent protein (GFP) RNA transcribed in vitro as a substrate in the absence of other viral or cellular proteins. Finally, PP-induced shutoff requires host Xrn1 to complete further RNA degradation. This study provides a newly discovered strategy in which FCV PP protein induces host gene shutoff by promoting the degradation of host mRNAs. IMPORTANCE Virus infection-induced shutoff is the result of targeted or global manipulation of cellular gene expression and leads to efficient viral replication and immune evasion. FCV is a highly contagious pathogen that persistently infects cats. It is unknown how FCV blocks the host immune response and persistently exists in cats. In this study, we found that FCV infection promotes the degradation of host mRNAs and induces host gene shutoff via a common strategy. Further, PP protein for different FCV strains is a key factor that enhances mRNA degradation. An in vitro assay showed that the GST-PP fusion protein possesses RNase activity in the absence of other viral or cellular proteins. This study demonstrates that FCV induces host gene shutoff by promoting the degradation of host mRNAs, thereby introducing a potential mechanism by which FCV infection inhibits the immune response.


Asunto(s)
Calicivirus Felino/crecimiento & desarrollo , Evasión Inmune/inmunología , Péptido Hidrolasas/metabolismo , Estabilidad del ARN/fisiología , ARN Mensajero/metabolismo , Ribonucleasas/metabolismo , Animales , Infecciones por Caliciviridae/patología , Calicivirus Felino/genética , Calicivirus Felino/metabolismo , Gatos , Línea Celular , Células HEK293 , Humanos , Evasión Inmune/genética , Péptido Hidrolasas/genética , Biosíntesis de Proteínas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Ribonucleasas/genética , Replicación Viral
20.
Nat Commun ; 12(1): 1518, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33750796

RESUMEN

Growing evidences suggest that cancer stem cells exhibit many molecular characteristics and phenotypes similar to their ancestral progenitor cells. In the present study, human embryonic stem cells are induced to differentiate into hepatocytes along hepatic lineages to mimic liver development in vitro. A liver progenitor specific gene, RALY RNA binding protein like (RALYL), is identified. RALYL expression is associated with poor prognosis, poor differentiation, and metastasis in clinical HCC patients. Functional studies reveal that RALYL could promote HCC tumorigenicity, self-renewal, chemoresistance, and metastasis. Moreover, molecular mechanism studies show that RALYL could upregulate TGF-ß2 mRNA stability by decreasing N6-methyladenosine (m6A) modification. TGF-ß signaling and the subsequent PI3K/AKT and STAT3 pathways, upregulated by RALYL, contribute to the enhancement of HCC stemness. Collectively, RALYL is a liver progenitor specific gene and regulates HCC stemness by sustaining TGF-ß2 mRNA stability. These findings may inspire precise therapeutic strategies for HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo C/metabolismo , Neoplasias Hepáticas/metabolismo , Estabilidad del ARN/fisiología , Factor de Crecimiento Transformador beta2/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Células Madre Embrionarias , Femenino , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo C/genética , Humanos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...