Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
RNA ; 26(11): 1603-1620, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32675111

RESUMEN

Cellular quiescence and cell cycle reentry regulate vital biological processes such as cellular development and tissue homeostasis and are controlled by precise regulation of gene expression. The roles of long noncoding RNAs (lncRNAs) during these processes remain to be elucidated. By performing genome-wide transcriptome analyses, we identify differential expression of several hundreds of lncRNAs, including a significant number of the less-characterized class of microRNA-host-gene (MIRHG) lncRNAs or lnc-MIRHGs, during cellular quiescence and cell cycle reentry in human diploid fibroblasts. We observe that MIR222HG lncRNA displays serum-stimulated RNA processing due to enhanced splicing of the host nascent pri-MIR222HG transcript. The pre-mRNA splicing factor SRSF1 negatively regulates the microprocessor-catalyzed cleavage of pri-miR-222, thereby increasing the cellular pool of the mature MIR222HG Association of SRSF1 to pri-MIR222HG, including to a mini-exon, which partially overlaps with the primary miR-222 precursor, promotes serum-stimulated splicing over microRNA processing of MIR222HG Further, we observe that the increased levels of spliced MIR222HG in serum-stimulated cells promote the cell cycle reentry post quiescence in a microRNA-independent manner. MIR222HG interacts with DNM3OS, another lncRNA whose expression is elevated upon serum-stimulation, and promotes cell cycle reentry. The double-stranded RNA binding protein ILF3/2 complex facilitates MIR222HG:DNM3OS RNP complex assembly, thereby promoting DNM3OS RNA stability. Our study identifies a novel mechanism whereby competition between the splicing and microprocessor machinery modulates the serum-induced RNA processing of MIR222HG, which dictates cell cycle reentry.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Pulmón/citología , ARN Largo no Codificante/genética , Factores de Empalme Serina-Arginina/metabolismo , Suero/química , Ciclo Celular , Línea Celular , Fibroblastos/química , Fibroblastos/citología , Células HEK293 , Humanos , Pulmón/química , Proteína del Factor Nuclear 45/metabolismo , Proteínas del Factor Nuclear 90/metabolismo , Procesamiento Postranscripcional del ARN , Empalme del ARN , Análisis de Secuencia de ARN , Imagen Individual de Molécula , Regulación hacia Arriba , Secuenciación del Exoma
2.
Methods ; 155: 131-139, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30500367

RESUMEN

The regulation of gene expression occurs through complex relationships between transcription, processing, turnover, and translation, which are only beginning to be elucidated. We know that at least for certain messenger (m) RNAs, processing, modifications, and sequence elements can greatly influence their translational output through recognition by translation and turn-over machinery. Recently, we and others have combined high-throughput sequencing technologies with traditional biochemical methods of studying translation to extend our understanding of these relationships. Additionally, there is growing importance given to how these processes may be regulated across varied cell types as a means to achieve tissue-specific expression of proteins. Here, we provide an in-depth methodology for polysome profiling to dissect the composition of mRNAs and proteins that make up the translatome from both whole tissues and a specific cell type isolated from mammalian tissue. Also, we provide a detailed computational workflow for the analysis of the next-generation sequencing data generated from these experiments.


Asunto(s)
Biología Computacional/métodos , Polirribosomas/genética , Biosíntesis de Proteínas , ARN Mensajero/genética , Análisis de Secuencia de ARN/estadística & datos numéricos , Animales , Encéfalo/citología , Encéfalo/metabolismo , Fraccionamiento Celular/métodos , Centrifugación por Gradiente de Densidad/métodos , Ontología de Genes , Redes Reguladoras de Genes , Hepatocitos/citología , Hepatocitos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Hígado/citología , Hígado/metabolismo , Ratones , Anotación de Secuencia Molecular , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos , Polirribosomas/clasificación , Polirribosomas/metabolismo , ARN Mensajero/metabolismo
3.
Curr Probl Cardiol ; 49(6): 102527, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38492618

RESUMEN

This comprehensive study delves into the epidemiological landscape of Pulmonary Heart Disease (PHD) mortality in the United States from 1999 to 2020, leveraging the extensive CDC WONDER database. PHD encompasses conditions affecting the right side of the heart due to lung disorders or elevated pressure in the pulmonary arteries, including pulmonary hypertension, pulmonary embolism, and chronic thromboembolic pulmonary hypertension (CTEPH). Analyzing data from death certificates, demographic characteristics, and geographical segmentation, significant trends emerge. The age-adjusted mortality rates (AAMRs) for PHD-related deaths show a fluctuating pattern, initially decreasing from 1999 to 2006, followed by a steady increase until 2020. Male patients consistently exhibit higher AAMRs than females, with notable disparities observed among racial/ethnic groups and geographic regions. Non-hispanic (NH) Black or African American individuals, residents of specific states like Colorado and the District of Columbia, and those in the Midwest region demonstrate elevated AAMRs. Furthermore, nonmetropolitan areas consistently manifest higher AAMRs than metropolitan areas. These findings underscore the urgent need for intensified prevention and treatment strategies to address the rising mortality associated with PHD, particularly among vulnerable populations. Insights from this study offer valuable guidance for public health initiatives aimed at reducing PHD-related mortality and improving outcomes nationwide.


Asunto(s)
Disparidades en el Estado de Salud , Enfermedad Cardiopulmonar , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Centers for Disease Control and Prevention, U.S. , Etnicidad/estadística & datos numéricos , Estudios Longitudinales , Enfermedad Cardiopulmonar/epidemiología , Enfermedad Cardiopulmonar/mortalidad , Grupos Raciales/estadística & datos numéricos , Distribución por Sexo , Factores Sexuales , Tasa de Supervivencia/tendencias , Estados Unidos/epidemiología , Negro o Afroamericano , Hispánicos o Latinos
4.
Nat Commun ; 14(1): 551, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759613

RESUMEN

Regulation of RNA processing contributes profoundly to tissue development and physiology. Here, we report that serine-arginine-rich splicing factor 1 (SRSF1) is essential for hepatocyte function and survival. Although SRSF1 is mainly known for its many roles in mRNA metabolism, it is also crucial for maintaining genome stability. We show that acute liver damage in the setting of targeted SRSF1 deletion in mice is associated with the excessive formation of deleterious RNA-DNA hybrids (R-loops), which induce DNA damage. Combining hepatocyte-specific transcriptome, proteome, and RNA binding analyses, we demonstrate that widespread genotoxic stress following SRSF1 depletion results in global inhibition of mRNA transcription and protein synthesis, leading to impaired metabolism and trafficking of lipids. Lipid accumulation in SRSF1-deficient hepatocytes is followed by necroptotic cell death, inflammation, and fibrosis, resulting in NASH-like liver pathology. Importantly, SRSF1-depleted human liver cancer cells recapitulate this pathogenesis, illustrating a conserved and fundamental role for SRSF1 in preserving genome integrity and tissue homeostasis. Thus, our study uncovers how the accumulation of detrimental R-loops impedes hepatocellular gene expression, triggering metabolic derangements and liver damage.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Animales , Ratones , Factores de Empalme de ARN/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , ARN/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , ARN Mensajero/metabolismo , Empalme Alternativo
5.
Biochemistry ; 50(25): 5806-11, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21591687

RESUMEN

Binding of the thiazolidinedione antidiabetic drug pioglitazone led to the discovery of a novel outer mitochondrial membrane protein of unknown function called mitoNEET. The protein is homodimeric and contains a uniquely ligated two iron-two sulfur cluster in each of its two cytosolic domains. Electrospray ionization mass spectrometry was employed to characterize solutions of the soluble cytosolic domain (amino acids 32--108) of the protein. Ions characteristic of dimers containing the cofactors were readily detected under native conditions. mitoNEET responded to exposure to solutions at low pH by dissociation to give monomers that retained the cofactor, followed by dissociation of the cofactor in a concerted fashion. mitoNEET formed complexes with resveratrol-3-sulfate, one of the primary metabolites of the natural product resveratrol. Resveratrol itself showed no tendency to interact with mitoNEET. The formation of complexes was evident in both electrospray ionization mass spectrometry and isothermal titration calorimetry measurements. Up to eight molecules of the compound associated with the dimeric form of the protein in a sequential fashion. Dissociation constants determined by micorcalorimetry were in the range 5-16 µM for the various binding sites. The only other known naturally occurring binding partner for mitoNEET at present is NADPH. It is very interesting that the iron-sulfur cluster containing protein interacts with two potentially redox active substances at the surface of mitochondria. These findings provide a new direction for research into two poorly understood, yet biomedically relevant, species.


Asunto(s)
Membranas Mitocondriales/química , Proteínas Mitocondriales/química , Estilbenos/química , Calorimetría , Dimerización , Humanos , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , Oxidación-Reducción , Unión Proteica , Resveratrol , Espectrometría de Masa por Ionización de Electrospray , Estilbenos/metabolismo , Termodinámica
6.
Sci Adv ; 7(17)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33883138

RESUMEN

Nuclear receptors farnesoid X receptor (FXR) and small heterodimer partner (SHP) are key regulators of metabolism. Here, we report a previously unknown function for the hepatic FXR-SHP axis in controlling protein N-linked glycosylation. Transcriptome analysis in liver-specific Fxr-Shp double knockout (LDKO) livers revealed induction of genes encoding enzymes in the N-glycosylation pathway, including Mgat5, Fut8, St3gal6, and St6gal1 FXR activation suppressed Mgat5, while Shp deletion induced St3gal6 and St6gal1 Increased percentages of core-fucosylated and triantennary glycan moieties were seen in LDKO livers, and proteins with the "hyperglycoforms" preferentially localized to exosomes and lysosomes. This up-regulation of N-glycosylation machinery was specific to the Golgi apparatus and not the endoplasmic reticulum. The increased glycan complexity in the LDKO correlated well with dilated unstacked Golgi ribbons and alterations in the secretion of albumin, cholesterol, and triglycerides. Our findings demonstrate a role for the FXR-SHP axis in maintaining glycoprotein diversity in the liver.


Asunto(s)
Hígado , Receptores Citoplasmáticos y Nucleares , Colesterol/metabolismo , Hígado/metabolismo , Polisacáridos/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Activación Transcripcional
7.
Toxicol Lett ; 329: 38-46, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32320774

RESUMEN

Perfluorooctanoic acid (PFOA) is a persistent organic pollutant prevalent in the environment and implicated in damage to the liver leading to a fatty liver phenotype called hepatocellular steatosis. Our goal is to provide a basis for PFOA-induced hepatocellular steatosis in relation to epigenetic alterations and mRNA splicing. Young adult female mice exposed to different concentrations of PFOA showed an increase in liver weight with decreased global DNA methylation (5-mC). At higher concentrations, the expression of DNA methyltransferase 3A (Dnmt3a) was significantly reduced and the expression of tet methycytosine dioxygenase 1 (Tet1) was significantly increased. There was no significant change in the other Dnmts and Tets. PFOA exposure significantly increased the expression of cell cycle regulators and anti-apoptotic genes. The expression of multiple genes involved in mTOR (mammalian target of rapamycin) signaling pathway were altered significantly with reduction in Pten (phosphatase and tensin homolog, primary inhibitor of mTOR pathway) expression. Multiple splicing factors whose protein but not mRNA levels affected by PFOA exposure were identified. The changes in protein abundance of the splicing factors was also reflected in altered splicing pattern of their target genes, which provided new insights on the previously unexplored mechanisms of PFOA-mediated hepatotoxicity and pathogenesis.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Caprilatos/farmacología , Metilación de ADN/efectos de los fármacos , Fluorocarburos/farmacología , Hígado/efectos de los fármacos , Animales , Apoptosis , Proliferación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas , ADN Metiltransferasa 3A , Hígado Graso/inducido químicamente , Femenino , Ratones , Isoformas de Proteínas , Proteínas de Unión al ARN , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
8.
JHEP Rep ; 2(5): 100140, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32875282

RESUMEN

BACKGROUND & AIMS: Liver diseases are caused by many factors, such as genetics, nutrition, and viruses. Therefore, it is important to delineate transcriptomic changes that occur in various liver diseases. METHODS: We performed high-throughput sequencing of mouse livers with diverse types of injuries, including cholestasis, diet-induced steatosis, and partial hepatectomy. Comparative analysis of liver transcriptome from mice and human samples of viral infections (HBV and HCV), alcoholic hepatitis (AH), non-alcoholic steatohepatitis (NASH), and biliary atresia revealed distinct and overlapping gene profiles associated with liver diseases. We hypothesised that discrete molecular signatures could be utilised to assess therapeutic outcomes. We focused on cholestasis to test and validate the hypothesis using pharmacological approaches. RESULTS: Here, we report significant overlap in the expression of inflammatory and proliferation-related genes across liver diseases. However, cholestatic livers were unique and displayed robust induction of genes involved in drug metabolism. Consistently, we found that constitutive androstane receptor (CAR) activation is crucial for the induction of the drug metabolic gene programme in cholestasis. When challenged, cholestatic mice were protected against zoxazolamine-induced paralysis and acetaminophen-induced hepatotoxicity. These protective effects were diminished upon inhibition of CAR activity. Further, drug metabolic genes were also induced in the livers from a subset of biliary atresia patients, but not in HBV and HCV infections, AH, or NASH. We also found a higher expression of CYP2B6, a CAR target, in the livers of biliary atresia patients, underscoring the clinical importance of our findings. CONCLUSIONS: Comparative transcriptome analysis of different liver disorders revealed specific induction of phase I and II metabolic genes in cholestasis. Our results demonstrate that CAR activation may lead to variations in drug metabolism and clinical outcomes in biliary atresia. LAY SUMMARY: Transcriptomic analysis of diverse liver diseases revealed alterations in common and distinct pathways. Specifically, in cholestasis, we found that detoxification genes and their activity are increased. Thus, cholestatic patients may have an unintended consequence on drug metabolism and not only have a beneficial effect against liver toxicity, but also may require adjustments to their therapeutic dosage.

9.
Nat Struct Mol Biol ; 25(10): 928-939, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30250226

RESUMEN

During liver regeneration, most new hepatocytes arise via self-duplication; yet, the underlying mechanisms that drive hepatocyte proliferation following injury remain poorly defined. By combining high-resolution transcriptome and polysome profiling of hepatocytes purified from quiescent and toxin-injured mouse livers, we uncover pervasive alterations in messenger RNA translation of metabolic and RNA-processing factors, which modulate the protein levels of a set of splicing regulators. Specifically, downregulation of the splicing regulator ESRP2 activates a neonatal alternative splicing program that rewires the Hippo signaling pathway in regenerating hepatocytes. We show that production of neonatal splice isoforms attenuates Hippo signaling, enables greater transcriptional activation of downstream target genes, and facilitates liver regeneration. We further demonstrate that ESRP2 deletion in mice causes excessive hepatocyte proliferation upon injury, whereas forced expression of ESRP2 inhibits proliferation by suppressing the expression of neonatal Hippo pathway isoforms. Thus, our findings reveal an alternative splicing axis that supports regeneration following chronic liver injury.


Asunto(s)
Empalme Alternativo , Regeneración Hepática/genética , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Proliferación Celular/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hepatocitos/citología , Hepatocitos/metabolismo , Hepatocitos/fisiología , Vía de Señalización Hippo , Ratones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Transducción de Señal/genética
10.
Biochim Biophys Acta Gene Regul Mech ; 1860(3): 349-362, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28088440

RESUMEN

Intermediary metabolism studies have typically concentrated on four major regulatory mechanisms-substrate availability, allosteric enzyme regulation, post-translational enzyme modification, and regulated enzyme synthesis. Although transcriptional control has been a big focus, it is becoming increasingly evident that many post-transcriptional events are deeply embedded within the core regulatory circuits of enzyme synthesis/breakdown that maintain metabolic homeostasis. The prominent post-transcriptional mechanisms affecting intermediary metabolism include alternative pre-mRNA processing, mRNA stability and translation control, and the more recently discovered regulation by noncoding RNAs. In this review, we discuss the latest advances in our understanding of these diverse mechanisms at the cell-, tissue- and organismal-level. We also highlight the dynamics, complexity and non-linear nature of their regulatory roles in metabolic decision making, and deliberate some of the outstanding questions and challenges in this rapidly expanding field.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , Animales , Humanos
11.
Sci Rep ; 5: 9512, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25824884

RESUMEN

In the present study, we perform an association analysis focusing on the expression changes of 1344 RNA Binding proteins (RBPs) as a function of age and gender in human liver. We identify 88 and 45 RBPs to be significantly associated with age and gender respectively. Experimental verification of several of the predicted associations in mice confirmed our findings. Our results suggest that a small fraction of the gender-associated RBPs (~40%) are expressed higher in males than females. Altogether, these observations show that several of these RBPs are important and conserved regulators in maintaining liver function. Further analysis of the protein interaction network of RBPs associated with age and gender based on the centrality measures like degree, betweenness and closeness revealed that several of these RBPs might be prominent players in aging liver and impart gender specific alterations in gene expression via the formation of protein complexes. Indeed, both age and gender-associated RBPs in liver were found to show significantly higher clustering coefficients and network centrality measures compared to non-associated RBPs. The compendium of RBPs and this study will help us gain insight into the role of post-transcriptional regulatory molecules in aging and gender specific expression of genes.


Asunto(s)
Hígado/crecimiento & desarrollo , Hígado/metabolismo , Proteínas de Unión al ARN/genética , Factores de Edad , Animales , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Ratas , Reproducibilidad de los Resultados , Factores Sexuales , Transcriptoma
12.
Nat Commun ; 6: 8768, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26531099

RESUMEN

Although major genetic networks controlling early liver specification and morphogenesis are known, the mechanisms responsible for postnatal hepatic maturation are poorly understood. Here we employ global analyses of the mouse liver transcriptome to demonstrate that postnatal remodelling of the liver is accompanied by large-scale transcriptional and post-transcriptional transitions that are cell-type-specific and temporally coordinated. Combining detailed expression analyses with gain- and loss-of-function studies, we identify epithelial splicing regulatory protein 2 (ESRP2) as a conserved regulatory factor that controls the neonatal-to-adult switch of ∼20% of splice isoforms in mouse and human hepatocytes. The normal shift in splicing coincides tightly with dramatic postnatal induction of ESRP2 in hepatocytes. We further demonstrate that forced expression of ESRP2 in immature mouse and human hepatocytes is sufficient to drive a reciprocal shift in splicing and causes various physiological abnormalities. These findings define a direct role for ESRP2 in the generation of conserved repertoires of adult splice isoforms that facilitate terminal differentiation and maturation of hepatocytes.


Asunto(s)
Empalme Alternativo/genética , Regulación del Desarrollo de la Expresión Génica , Hepatocitos/metabolismo , Hígado/metabolismo , Procesamiento Postranscripcional del ARN/genética , Proteínas de Unión al ARN/genética , Animales , Western Blotting , Diferenciación Celular , Humanos , Inmunohistoquímica , Hibridación in Situ , Hígado/crecimiento & desarrollo , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA