Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 1222, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38336721

RESUMEN

To survive, cells must respond to changing environmental conditions. One way that eukaryotic cells react to harsh stimuli is by forming physiological, RNA-seeded subnuclear condensates, termed amyloid bodies (A-bodies). The molecular constituents of A-bodies induced by different stressors vary significantly, suggesting this pathway can tailor the cellular response by selectively aggregating a subset of proteins under a given condition. Here, we identify critical structural elements that regulate heat shock-specific amyloid aggregation. Our data demonstrates that manipulating structural pockets in constituent proteins can either induce or restrict their A-body targeting at elevated temperatures. We propose a model where selective aggregation within A-bodies is mediated by the thermal stability of a protein, with temperature-sensitive structural regions acting as an intrinsic form of post-translational regulation. This system would provide cells with a rapid and stress-specific response mechanism, to tightly control physiological amyloid aggregation or other cellular stress response pathways.


Asunto(s)
Amiloide , Proteínas Amiloidogénicas , Amiloide/metabolismo , Respuesta al Choque Térmico , Células Eucariotas/metabolismo , Temperatura
2.
Sci Rep ; 13(1): 14471, 2023 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-37660155

RESUMEN

The formation of protein aggregates is a hallmark of many neurodegenerative diseases and systemic amyloidoses. These disorders are associated with the fibrillation of a variety of proteins/peptides, which ultimately leads to cell toxicity and tissue damage. Understanding how amyloid aggregation occurs and developing compounds that impair this process is a major challenge in the health science community. Here, we demonstrate that pathogenic proteins associated with Alzheimer's disease, diabetes, AL/AA amyloidosis, and amyotrophic lateral sclerosis can aggregate within stress-inducible physiological amyloid-based structures, termed amyloid bodies (A-bodies). Using a limited collection of small molecule inhibitors, we found that diclofenac could repress amyloid aggregation of the ß-amyloid (1-42) in a cellular setting, despite having no effect in the classic Thioflavin T (ThT) in vitro fibrillation assay. Mapping the mechanism of the diclofenac-mediated repression indicated that dysregulation of cyclooxygenases and the prostaglandin synthesis pathway was potentially responsible for this effect. Together, this work suggests that the A-body machinery may be linked to a subset of pathological amyloidosis, and highlights the utility of this model system in the identification of new small molecules that could treat these debilitating diseases.


Asunto(s)
Amiloidosis , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas , Humanos , Diclofenaco/farmacología , Proteínas Amiloidogénicas , Prostaglandina-Endoperóxido Sintasas
3.
Biochem Cell Biol ; 101(2): 160-171, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36745874

RESUMEN

Heterogeneous nuclear ribonucleoproteins (hnRNPs) are among the most abundantly expressed RNA binding proteins in the cell and play major roles in all facets of RNA metabolism. hnRNPs are increasingly appreciated as essential for mammalian B cell development by regulating the carefully ordered expression of specific genes. Due to this tight regulation of the hnRNP-RNA network, it is no surprise that a growing number of genes encoding hnRNPs have been causally associated with the onset or progression of many cancers, including B cell neoplasms. Here we discuss our current understanding of hnRNP-driven regulation in normal, perturbed, and malignant B cells, and the most recent and emerging therapeutic innovations aimed at targeting the hnRNP-RNA network in lymphoma.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas , Ribonucleoproteínas , Animales , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Mamíferos/genética , Mamíferos/metabolismo
5.
Front Mol Biosci ; 9: 998363, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36203874

RESUMEN

In recent decades, a growing number of biomolecular condensates have been identified in eukaryotic cells. These structures form through phase separation and have been linked to a diverse array of cellular processes. While a checklist of established membrane-bound organelles is present across the eukaryotic domain, less is known about the conservation of membrane-less subcellular structures. Many of these structures can be seen throughout eukaryotes, while others are only thought to be present in metazoans or a limited subset of species. In particular, the nucleus is a hub of biomolecular condensates. Some of these subnuclear domains have been found in a broad range of organisms, which is a characteristic often attributed to essential functionality. However, this does not always appear to be the case. For example, the nucleolus is critical for ribosomal biogenesis and is present throughout the eukaryotic domain, while the Cajal bodies are believed to be similarly conserved, yet these structures are dispensable for organismal survival. Likewise, depletion of the Drosophila melanogaster omega speckles reduces viability, despite the apparent absence of this domain in higher eukaryotes. By reviewing primary research that has analyzed the presence of specific condensates (nucleoli, Cajal bodies, amyloid bodies, nucleolar aggresomes, nuclear speckles, nuclear paraspeckles, nuclear stress bodies, PML bodies, omega speckles, NUN bodies, mei2 dots) in a cross-section of organisms (e.g., human, mouse, D. melanogaster, Caenorhabditis elegans, yeast), we adopt a human-centric view to explore the emergence, retention, and absence of a subset of nuclear biomolecular condensates. This overview is particularly important as numerous biomolecular condensates have been linked to human disease, and their presence in additional species could unlock new and well characterized model systems for health research.

6.
J Cell Sci ; 134(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34704593

RESUMEN

In response to environmental stress, human cells have been shown to form reversible amyloid aggregates within the nucleus, termed amyloid bodies (A-bodies). These protective physiological structures share many of the biophysical characteristics associated with the pathological amyloids found in Alzheimer's and Parkinson's disease. Here, we show that A-bodies are evolutionarily conserved across the eukaryotic domain, with their detection in Drosophila melanogaster and Saccharomyces cerevisiae marking the first examples of these functional amyloids being induced outside of a cultured cell setting. The conditions triggering amyloidogenesis varied significantly among the species tested, with results indicating that A-body formation is a severe, but sublethal, stress response pathway that is tailored to the environmental norms of an organism. RNA-sequencing analyses demonstrate that the regulatory low-complexity long non-coding RNAs that drive A-body aggregation are both conserved and essential in human, mouse and chicken cells. Thus, the identification of these natural and reversible functional amyloids in a variety of evolutionarily diverse species highlights the physiological significance of this protein conformation, and will be informative in advancing our understanding of both functional and pathological amyloid aggregation events. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Drosophila melanogaster , Animales , Biofisica , Drosophila melanogaster/genética , Ratones
7.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33568529

RESUMEN

Biomolecular condensates concentrate molecules to facilitate basic biochemical processes, including transcription and DNA replication. While liquid-like condensates have been ascribed various functions, solid-like condensates are generally thought of as amorphous sites of protein storage. Here, we show that solid-like amyloid bodies coordinate local nuclear protein synthesis (LNPS) during stress. On stimulus, translationally active ribosomes accumulate along fiber-like assemblies that characterize amyloid bodies. Mass spectrometry analysis identified regulatory ribosomal proteins and translation factors that relocalize from the cytoplasm to amyloid bodies to sustain LNPS. These amyloidogenic compartments are enriched in newly transcribed messenger RNA by Heat Shock Factor 1 (HSF1). Depletion of stress-induced ribosomal intergenic spacer noncoding RNA (rIGSRNA) that constructs amyloid bodies prevents recruitment of the nuclear protein synthesis machinery, abolishes LNPS, and impairs the nuclear HSF1 response. We propose that amyloid bodies support local nuclear translation during stress and that solid-like condensates can facilitate complex biochemical reactions as their liquid counterparts can.


Asunto(s)
Amiloide/metabolismo , Núcleo Celular/metabolismo , Respuesta al Choque Térmico , Amiloide/genética , Hipoxia de la Célula , Citoplasma/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Células MCF-7 , Biosíntesis de Proteínas , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ribosomas/metabolismo
8.
Blood ; 136(5): 572-584, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32160292

RESUMEN

Mantle cell lymphoma (MCL) is an uncommon B-cell non-Hodgkin lymphoma (NHL) that is incurable with standard therapies. The genetic drivers of this cancer have not been firmly established, and the features that contribute to differences in clinical course remain limited. To extend our understanding of the biological pathways involved in this malignancy, we performed a large-scale genomic analysis of MCL using data from 51 exomes and 34 genomes alongside previously published exome cohorts. To confirm our findings, we resequenced the genes identified in the exome cohort in 191 MCL tumors, each having clinical follow-up data. We confirmed the prognostic association of TP53 and NOTCH1 mutations. Our sequencing revealed novel recurrent noncoding mutations surrounding a single exon of the HNRNPH1gene. In RNA-seq data from 103 of these cases, MCL tumors with these mutations had a distinct imbalance of HNRNPH1 isoforms. This altered splicing of HNRNPH1 was associated with inferior outcomes in MCL and showed a significant increase in protein expression by immunohistochemistry. We describe a functional role for these recurrent noncoding mutations in disrupting an autoregulatory feedback mechanism, thereby deregulating HNRNPH1 protein expression. Taken together, these data strongly imply a role for aberrant regulation of messenger RNA processing in MCL pathobiology.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Linfoma de Células del Manto/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mutación , Secuenciación Completa del Genoma
9.
FEBS Lett ; 593(22): 3162-3172, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31512750

RESUMEN

Physiological amyloid aggregation occurs within the nuclei of stress-treated cells. These structures, termed Amyloid bodies (A-bodies), assemble through the rapid accumulation of proteins into dense membrane-less organelles, which possess the same biophysical properties as plaques observed in many amyloid-based diseases. Here, we demonstrate that A-body proteomic compositions vary significantly between stimuli, as constituent proteins can be sequestered by one or more stressors. Stimulus exposure alone was insufficient to induce aggregation, demonstrating that this pathway is not regulated solely by stress-induced conformational changes of the A-body targets. We propose that different environmental conditions induce the formation of A-body subtypes containing distinct protein residents. This selective immobilization of proteins may have evolved as a finely tuned mechanism for surviving divergent stressors.


Asunto(s)
Agregado de Proteínas , Proteínas/química , Proteínas/metabolismo , Proteómica/métodos , Células A549 , Núcleo Celular/metabolismo , Células HeLa , Calor , Humanos , Células MCF-7 , Espectrometría de Masas , Células PC-3 , Estrés Fisiológico
10.
Nat Commun ; 9(1): 4001, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30275490

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is an aggressive cancer originating from mature B-cells. Prognosis is strongly associated with molecular subgroup, although the driver mutations that distinguish the two main subgroups remain poorly defined. Through an integrative analysis of whole genomes, exomes, and transcriptomes, we have uncovered genes and non-coding loci that are commonly mutated in DLBCL. Our analysis has identified novel cis-regulatory sites, and implicates recurrent mutations in the 3' UTR of NFKBIZ as a novel mechanism of oncogene deregulation and NF-κB pathway activation in the activated B-cell (ABC) subgroup. Small amplifications associated with over-expression of FCGR2B (the Fcγ receptor protein IIB), primarily in the germinal centre B-cell (GCB) subgroup, correlate with poor patient outcomes suggestive of a novel oncogene. These results expand the list of subgroup driver mutations that may facilitate implementation of improved diagnostic assays and could offer new avenues for the development of targeted therapeutics.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes Reguladores/genética , Variación Genética , Genoma Humano/genética , Linfoma de Células B Grandes Difuso/genética , Regiones no Traducidas 3'/genética , Proteínas Adaptadoras Transductoras de Señales , Linfocitos B/metabolismo , Linfocitos B/patología , Línea Celular Tumoral , Exoma/genética , Estudio de Asociación del Genoma Completo , Centro Germinal/metabolismo , Centro Germinal/patología , Humanos , Proteínas I-kappa B/genética , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Mutación , Proteínas Nucleares/genética , Receptores de IgG/genética , Análisis de Secuencia de ADN , Transcriptoma
11.
Cell Rep ; 24(7): 1713-1721.e4, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30110628

RESUMEN

Amyloid bodies (A-bodies) are inducible membrane-less nuclear compartments composed of heterogeneous proteins that adopt an amyloid-like state. A-bodies are seeded by noncoding RNA derived from stimuli-specific loci of the rDNA intergenic spacer (rIGSRNA). This raises the question of how rIGSRNA recruits a large population of diverse proteins to confer A-body identity. Here, we show that long low-complexity dinucleotide repeats operate as the architectural determinants of rIGSRNA. On stimulus, clusters of rIGSRNA with simple cytosine/uracil (CU) or adenosine/guanine (AG) repeats spanning hundreds of nucleotides accumulate in the nucleolar area. The low-complexity sequences facilitate charge-based interactions with short cationic peptides to produce multiple nucleolar liquid-like foci. Local concentration of proteins with fibrillation propensity in these nucleolar foci induces the formation of an amyloidogenic liquid phase that seeds A-bodies. These results demonstrate the physiological importance of low-complexity RNA and repetitive regions of the genome often dismissed as "junk" DNA.


Asunto(s)
Proteínas Amiloidogénicas/química , Nucléolo Celular/genética , ADN Intergénico/química , ADN Ribosómico/química , ARN Ribosómico/química , ARN no Traducido/química , Amiloide/química , Amiloide/genética , Amiloide/metabolismo , Proteínas Amiloidogénicas/genética , Proteínas Amiloidogénicas/metabolismo , Animales , Secuencia de Bases , Hipoxia de la Célula , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestructura , ADN Intergénico/genética , ADN Intergénico/metabolismo , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Repeticiones de Dinucleótido , Expresión Génica , Respuesta al Choque Térmico , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Ratones , Transición de Fase , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Electricidad Estática , Estrés Fisiológico , Imagen de Lapso de Tiempo
12.
Trends Cell Biol ; 27(7): 465-467, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28359692

RESUMEN

Historically, amyloids were perceived as toxic/irreversible protein aggregates associated with neurodegenerative disorders including Alzheimer's and Parkinson's diseases. Recent papers are challenging this perception by uncovering widespread cellular roles for physiological amyloidogenesis. These findings suggest that the amyloid-fold should be considered, alongside the native-fold and unfolded configurations, as a physiological and reversible protein organization.


Asunto(s)
Amiloide/metabolismo , Amiloide/genética , Amiloidosis/genética , Humanos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Agregado de Proteínas , Pliegue de Proteína
13.
Eur J Cell Biol ; 95(12): 611-622, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28029379

RESUMEN

The recently identified Luman/CREB3-binding partner LRF (Luman/CREB3 recruitment factor) was shown to localize to discrete sub-nuclear foci. Luman is implicated in herpes simplex virus-1 (HSV-1) latency/reactivation and the unfolded protein response (UPR) pathway; therefore, we sought to characterize the formation of the LRF nuclear foci in the context of cellular signaling and HSV-1 replication. Here, we mapped the nuclear foci-targeting sequence to the central region containing the first leucine zipper (a.a.415-519), and found that the integrity of the whole region appears essential for LRF foci formation. LRF foci integrity was unaffected by inhibition of cellular DNA replication and translation, however, disruption of transcription resulted in altered LRF localization. When compared to other cellular and viral foci LRF co-localized with the nuclear receptor co-activator GRIP1, while the HSV-1 gene products ICP4, ICP27 and VP13/14 disrupted foci formation to varying degrees. Interestingly, cells over-expressing LRF were resistant to productive HSV-1 infection and this resistance was dependent upon protein targeting and an N-terminal transactivation domain. When LRF knockdown cells were subjected to primary infection, HSV-1 gene expression and progeny virus yield were enhanced by ∼3 fold compared to wildtype cells. Taken together, these results indicate that LRF is a key regulator that may act direct or indirectly as a repressor of essential genes required for productive viral infection.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 1/fisiología , Factores de Transcripción/metabolismo , Replicación Viral/fisiología , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Ratones , Células 3T3 NIH , Proteínas Supresoras de Tumor/metabolismo , Respuesta de Proteína Desplegada , Células Vero
14.
Dev Cell ; 39(2): 155-168, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27720612

RESUMEN

The amyloid state of protein organization is typically associated with debilitating human neuropathies and is seldom observed in physiology. Here, we uncover a systemic program that leverages the amyloidogenic propensity of proteins to regulate cell adaptation to stressors. On stimulus, cells assemble the amyloid bodies (A-bodies), nuclear foci containing heterogeneous proteins with amyloid-like biophysical properties. A discrete peptidic sequence, termed the amyloid-converting motif (ACM), is capable of targeting proteins to the A-bodies by interacting with ribosomal intergenic noncoding RNA (rIGSRNA). The pathological ß-amyloid peptide, involved in Alzheimer's disease, displays ACM-like activity and undergoes stimuli-mediated amyloidogenesis in vivo. Upon signal termination, elements of the heat-shock chaperone pathway disaggregate the A-bodies. Physiological amyloidogenesis enables cells to store large quantities of proteins and enter a dormant state in response to stressors. We suggest that cells have evolved a post-translational pathway that rapidly and reversibly converts native-fold proteins to an amyloid-like solid phase.


Asunto(s)
Adaptación Fisiológica , Amiloide/metabolismo , Estrés Fisiológico , Secuencias de Aminoácidos , Péptidos beta-Amiloides/metabolismo , Animales , Fenómenos Biofísicos , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Femenino , Respuesta al Choque Térmico , Humanos , Células MCF-7 , Ratones Desnudos , Chaperonas Moleculares/metabolismo , ARN no Traducido/genética , Ribosomas/metabolismo
15.
Cell Rep ; 14(6): 1293-1300, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26854219

RESUMEN

Protein concentrations evolve under greater evolutionary constraint than mRNA levels. Translation efficiency of mRNA represents the chief determinant of basal protein concentrations. This raises a fundamental question of how mRNA and protein levels are coordinated in dynamic systems responding to physiological stimuli. This report examines the contributions of mRNA abundance and translation efficiency to protein output in cells responding to oxygen stimulus. We show that changes in translation efficiencies, and not mRNA levels, represent the major mechanism governing cellular responses to [O2] perturbations. Two distinct cap-dependent protein synthesis machineries select mRNAs for translation: the normoxic eIF4F and the hypoxic eIF4F(H). O2-dependent remodeling of translation efficiencies enables cells to produce adaptive translatomes from preexisting mRNA pools. Differences in mRNA expression observed under different [O2] are likely neutral, given that they occur during evolution. We propose that mRNAs contain translation efficiency determinants for their triage by the translation apparatus on [O2] stimulus.


Asunto(s)
Factor 4F Eucariótico de Iniciación/genética , Oxígeno/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/genética , Hipoxia de la Célula , Línea Celular Tumoral , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Factor 4F Eucariótico de Iniciación/metabolismo , Evolución Molecular , Humanos , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , ARN Mensajero/metabolismo
16.
Biochim Biophys Acta ; 1859(1): 184-91, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26142536

RESUMEN

Genomic studies have revealed that humans possess far fewer protein-encoding genes than originally predicted. These over-estimates were drawn from the inherent developmental and stimuli-responsive complexity found in humans and other mammals, when compared to lower eukaryotic organisms. This left a conceptual void in many cellular networks, as a new class of functional molecules was necessary for "fine-tuning" the basic proteomic machinery. Transcriptomics analyses have determined that the vast majority of the genetic material is transcribed as noncoding RNA, suggesting that these molecules could provide the functional diversity initially sought from proteins. Indeed, as discussed in this review, long noncoding RNAs (lncRNAs), the largest family of noncoding transcripts, have emerged as common regulators of many cellular stressors; including heat shock, metabolic deprivation and DNA damage. These stimuli, while divergent in nature, share some common stress-responsive pathways, notably inhibition of cell proliferation. This role intrinsically makes stress-responsive lncRNA regulators potential tumor suppressor or proto-oncogenic genes. As the list of functional RNA molecules continues to rapidly expand it is becoming increasingly clear that the significance and functionality of this family may someday rival that of proteins. This article is part of a Special Issue entitled: Clues to long noncoding RNA taxonomy1, edited by Dr. Tetsuro Hirose and Dr. Shinichi Nakagawa.


Asunto(s)
Oncogenes , ARN Largo no Codificante/genética , Estrés Fisiológico/genética , Proteínas Supresoras de Tumor/genética , Animales , Proliferación Celular/genética , Humanos , Proteómica , ARN Largo no Codificante/clasificación , Proteínas Supresoras de Tumor/clasificación
17.
Proc Natl Acad Sci U S A ; 111(21): 7783-8, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24817692

RESUMEN

Epigenetic regulation of gene expression by DNA methylation plays a central role in the maintenance of cellular homeostasis. Here we present evidence implicating the DNA methylation program in the regulation of hypoxia-inducible factor (HIF) oxygen-sensing machinery and hypoxic cell metabolism. We show that DNA methyltransferase 3a (DNMT3a) methylates and silences the HIF-2α gene (EPAS1) in differentiated cells. Epigenetic silencing of EPAS1 prevents activation of the HIF-2α gene program associated with hypoxic cell growth, thereby limiting the proliferative capacity of adult cells under low oxygen tension. Naturally occurring defects in DNMT3a, observed in primary tumors and malignant cells, cause the unscheduled activation of EPAS1 in early dysplastic foci. This enables incipient cancer cells to exploit the HIF-2α pathway in the hypoxic tumor microenvironment necessary for the formation of cellular masses larger than the oxygen diffusion limit. Reintroduction of DNMT3a in DNMT3a-defective cells restores EPAS1 epigenetic silencing, prevents hypoxic cell growth, and suppresses tumorigenesis. These data support a tumor-suppressive role for DNMT3a as an epigenetic regulator of the HIF-2α oxygen-sensing pathway and the cellular response to hypoxia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinogénesis/genética , Hipoxia de la Célula/fisiología , ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , ADN Metiltransferasa 3A , Epigénesis Genética/genética , Femenino , Humanos , Ratones , Ratones Desnudos
18.
Cell Stress Chaperones ; 19(6): 813-25, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24590458

RESUMEN

Mammalian hibernators survive low body temperatures, ischemia-reperfusion, and restricted nutritional resources via global reductions in energy-expensive cellular processes and selective increases in stress pathways. Consequently, studies that analyze hibernation uncover mechanisms which balance metabolism and support survival by enhancing stress tolerance. We hypothesized processing factors that influence messenger ribonucleic acid (mRNA) maturation and translation may play significant roles in hibernation. We characterized the amino acid sequences of three RNA processing proteins (T cell intracellular antigen 1 (TIA-1), TIA1-related (TIAR), and poly(A)-binding proteins (PABP-1)) from thirteen-lined ground squirrels (Ictidomys tridecemlineatus), which all displayed a high degree of sequence identity with other mammals. Alternate Tia-1 and TiaR gene variants were found in the liver with higher expression of isoform b versus a in both cases. The localization of RNA-binding proteins to subnuclear structures was assessed by immunohistochemistry and confirmed by subcellular fractionation; TIA-1 was identified as a major component of subnuclear structures with up to a sevenfold increase in relative protein levels in the nucleus during hibernation. By contrast, there was no significant difference in the relative protein levels of TIARa/TIARb in the nucleus, and a decrease was observed for TIAR isoforms in cytoplasmic fractions of torpid animals. Finally, we used solubility tests to analyze the formation of reversible aggregates that are associated with TIA-1/R function during stress; a shift towards the soluble fraction (TIA-1a, TIA-1b) was observed during hibernation suggesting enhanced protein aggregation was not present during torpor. The present study identifies novel posttranscriptional regulatory mechanisms that may play a role in reducing translational rates and/or mRNA processing under unfavorable environmental conditions.


Asunto(s)
Hibernación , Proteína I de Unión a Poli(A)/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulación de la Expresión Génica , Variación Genética , Hibernación/genética , Proteína I de Unión a Poli(A)/genética , Biosíntesis de Proteínas , Isoformas de Proteínas , Estabilidad del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Sciuridae , Solubilidad , Transcripción Genética
19.
Mol Biol Cell ; 24(18): 2943-53, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23904269

RESUMEN

The nucleolus is a plurifunctional organelle in which structure and function are intimately linked. Its structural plasticity has long been appreciated, particularly in response to transcriptional inhibition and other cellular stresses, although the mechanism and physiological relevance of these phenomena are unclear. Using MCF-7 and other mammalian cell lines, we describe a structural and functional adaptation of the nucleolus, triggered by heat shock or physiological acidosis, that depends on the expression of ribosomal intergenic spacer long noncoding RNA (IGS lncRNA). At the heart of this process is the de novo formation of a large subnucleolar structure, termed the detention center (DC). The DC is a spatially and dynamically distinct region, characterized by an 8-anilino-1-naphthalenesulfonate-positive hydrophobic signature. Its formation is accompanied by redistribution of nucleolar factors and arrest in ribosomal biogenesis. Silencing of regulatory IGS lncRNA prevents the creation of this structure and allows the nucleolus to retain its tripartite organization and transcriptional activity. Signal termination causes a decrease in IGS transcript levels and a return to the active nucleolar conformation. We propose that the induction of IGS lncRNA by environmental signals operates as a molecular switch that regulates the structure and function of the nucleolus.


Asunto(s)
Nucléolo Celular/genética , Nucléolo Celular/metabolismo , Ambiente , ARN Largo no Codificante/metabolismo , Animales , Línea Celular Tumoral , Nucléolo Celular/ultraestructura , ADN Espaciador Ribosómico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Modelos Biológicos , Células 3T3 NIH , Transcripción Genética
20.
Nucleus ; 3(4): 315-9, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22688644

RESUMEN

The nucleolus is organized around a scaffolding of rDNA tandem repeats. These repeats, known as ribosomal cassettes, are each composed of ribosomal RNA (rRNA) genes preceding a long intergenic spacer (IGS) that has been classically perceived to be transcriptionally silent. Recent study of the IGS has contradicted the dogma that these spacers are merely inert regions of the genome, instead suggesting they are biologically significant, complex and plurifunctional transcriptional units that appear central to proper cellular functioning. Through the timely induction of various ribosomal IGS noncoding RNA (IGS RNA) transcripts, the cell is capable of both regulating rRNA synthesis and sequestering large numbers of proteins, thereby modulating essential molecular networks. Here we discuss our current understanding of the organization and function of the IGS.


Asunto(s)
ADN Espaciador Ribosómico/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Genes de ARNr/genética , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA