Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 41(13): 111893, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36577377

RESUMEN

Within the scope of the FANTOM6 consortium, we perform a large-scale knockdown of 200 long non-coding RNAs (lncRNAs) in human induced pluripotent stem cells (iPSCs) and systematically characterize their roles in self-renewal and pluripotency. We find 36 lncRNAs (18%) exhibiting cell growth inhibition. From the knockdown of 123 lncRNAs with transcriptome profiling, 36 lncRNAs (29.3%) show molecular phenotypes. Integrating the molecular phenotypes with chromatin-interaction assays further reveals cis- and trans-interacting partners as potential primary targets. Additionally, cell-type enrichment analysis identifies lncRNAs associated with pluripotency, while the knockdown of LINC02595, CATG00000090305.1, and RP11-148B6.2 modulates colony formation of iPSCs. We compare our results with previously published fibroblasts phenotyping data and find that 2.9% of the lncRNAs exhibit a consistent cell growth phenotype, whereas we observe 58.3% agreement in molecular phenotypes. This highlights that molecular phenotyping is more comprehensive in revealing affected pathways.


Asunto(s)
Células Madre Pluripotentes Inducidas , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Oligonucleótidos Antisentido , Perfilación de la Expresión Génica/métodos , Células Madre Embrionarias/metabolismo
2.
Nat Genet ; 54(11): 1675-1689, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36333502

RESUMEN

The value of genome-wide over targeted driver analyses for predicting clinical outcomes of cancer patients is debated. Here, we report the whole-genome sequencing of 485 chronic lymphocytic leukemia patients enrolled in clinical trials as part of the United Kingdom's 100,000 Genomes Project. We identify an extended catalog of recurrent coding and noncoding genetic mutations that represents a source for future studies and provide the most complete high-resolution map of structural variants, copy number changes and global genome features including telomere length, mutational signatures and genomic complexity. We demonstrate the relationship of these features with clinical outcome and show that integration of 186 distinct recurrent genomic alterations defines five genomic subgroups that associate with response to therapy, refining conventional outcome prediction. While requiring independent validation, our findings highlight the potential of whole-genome sequencing to inform future risk stratification in chronic lymphocytic leukemia.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Secuenciación Completa del Genoma , Mutación , Genómica , Pronóstico
3.
BMC Biol ; 19(1): 218, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34592985

RESUMEN

BACKGROUND: Niemann-Pick disease, type C (NPC) is a childhood-onset, lethal, neurodegenerative disorder caused by autosomal recessive mutations in the genes NPC1 or NPC2 and characterized by impaired cholesterol homeostasis, a lipid essential for cellular function. Cellular cholesterol levels are tightly regulated, and mutations in either NPC1 or NPC2 lead to deficient transport and accumulation of unesterified cholesterol in the late endosome/lysosome compartment, and progressive neurodegeneration in affected individuals. Previous cell-based studies to understand the NPC cellular pathophysiology and screen for therapeutic agents have mainly used patient fibroblasts. However, these do not allow modeling the neurodegenerative aspect of NPC disease, highlighting the need for an in vitro system that permits understanding the cellular mechanisms underlying neuronal loss and identifying appropriate therapies. This study reports the development of a novel human iPSC-derived, inducible neuronal model of Niemann-Pick disease, type C1 (NPC1). RESULTS: We generated a null i3Neuron (inducible × integrated × isogenic) (NPC1-/- i3Neuron) iPSC-derived neuron model of NPC1. The NPC1-/- and the corresponding isogenic NPC1+/+ i3Neuron cell lines were used to efficiently generate homogenous, synchronized neurons that can be used in high-throughput screens. NPC1-/- i3Neurons recapitulate cardinal cellular NPC1 pathological features including perinuclear endolysosomal storage of unesterified cholesterol, accumulation of GM2 and GM3 gangliosides, mitochondrial dysfunction, and impaired axonal lysosomal transport. Cholesterol storage, mitochondrial dysfunction, and axonal trafficking defects can be ameliorated by treatment with 2-hydroxypropyl-ß-cyclodextrin, a drug that has shown efficacy in NPC1 preclinical models and in a phase 1/2a trial. CONCLUSION: Our data demonstrate the utility of this new cell line in high-throughput drug/chemical screens to identify potential therapeutic agents. The NPC1-/- i3Neuron line will also be a valuable tool for the NPC1 research community to explore the pathological mechanisms contributing to neuronal degeneration.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Niemann-Pick Tipo C , Colesterol , Humanos , Neuronas , Enfermedad de Niemann-Pick Tipo C/genética , Preparaciones Farmacéuticas
4.
Essays Biochem ; 65(4): 761-773, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33835127

RESUMEN

Recent efforts on the characterization of long non-coding RNAs (lncRNAs) revealed their functional roles in modulating diverse cellular processes. These include pluripotency maintenance, lineage commitment, carcinogenesis, and pathogenesis of various diseases. By interacting with DNA, RNA and protein, lncRNAs mediate multifaceted mechanisms to regulate transcription, RNA processing, RNA interference and translation. Of more than 173000 discovered lncRNAs, the majority remain functionally unknown. The cell type-specific expression and localization of the lncRNA also suggest potential distinct functions of lncRNAs across different cell types. This highlights the niche of identifying functional lncRNAs in different biological processes and diseases through high-throughput (HTP) screening. This review summarizes the current work performed and perspectives on HTP screening of functional lncRNAs where different technologies, platforms, cellular responses and the downstream analyses are discussed. We hope to provide a better picture in applying different technologies to facilitate functional annotation of lncRNA efficiently.


Asunto(s)
ARN Largo no Codificante , Ensayos Analíticos de Alto Rendimiento , ARN Largo no Codificante/genética
5.
J Biol Chem ; 295(9): 2850-2865, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31911440

RESUMEN

Cholesterol synthesis is a tightly regulated process, both transcriptionally and post-translationally. Transcriptional control of cholesterol synthesis is relatively well-understood. However, of the ∼20 enzymes in cholesterol biosynthesis, post-translational regulation has only been examined for a small number. Three of the four sterol reductases in cholesterol production, 7-dehydrocholesterol reductase (DHCR7), 14-dehydrocholesterol reductase (DHCR14), and lamin-B receptor (LBR), share evolutionary ties with a high level of sequence homology and predicted structural homology. DHCR14 and LBR uniquely share the same Δ-14 reductase activity in cholesterol biosynthesis, yet little is known about their post-translational regulation. We have previously identified specific modes of post-translational control of DHCR7, but it is unknown whether these regulatory mechanisms are shared by DHCR14 and LBR. Using CHO-7 cells stably expressing epitope-tagged DHCR14 or LBR, we investigated the post-translational regulation of these enzymes. We found that DHCR14 and LBR undergo differential post-translational regulation, with DHCR14 being rapidly turned over, triggered by cholesterol and other sterol intermediates, whereas LBR remained stable. DHCR14 is degraded via the ubiquitin-proteasome system, and we identified several DHCR14 and DHCR7 putative interaction partners, including a number of E3 ligases that modulate DHCR14 levels. Interestingly, we found that gene expression across an array of human tissues showed a negative relationship between the C14-sterol reductases; one enzyme or the other tends to be predominantly expressed in each tissue. Overall, our findings indicate that whereas LBR tends to be the constitutively active C14-sterol reductase, DHCR14 levels are tunable, responding to the local cellular demands for cholesterol.


Asunto(s)
Colesterol/biosíntesis , Regulación de la Expresión Génica , Oxidorreductasas/genética , Procesamiento Proteico-Postraduccional , Receptores Citoplasmáticos y Nucleares/genética , Animales , Células CHO , Cricetulus , Humanos , Especificidad de Órganos , Oxidorreductasas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Estabilidad Proteica , Ubiquitina-Proteína Ligasas/metabolismo , Receptor de Lamina B
6.
Neuron ; 104(2): 239-255.e12, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31422865

RESUMEN

CRISPR/Cas9-based functional genomics have transformed our ability to elucidate mammalian cell biology. However, most previous CRISPR-based screens were conducted in cancer cell lines rather than healthy, differentiated cells. Here, we describe a CRISPR interference (CRISPRi)-based platform for genetic screens in human neurons derived from induced pluripotent stem cells (iPSCs). We demonstrate robust and durable knockdown of endogenous genes in such neurons and present results from three complementary genetic screens. First, a survival-based screen revealed neuron-specific essential genes and genes that improved neuronal survival upon knockdown. Second, a screen with a single-cell transcriptomic readout uncovered several examples of genes whose knockdown had strikingly cell-type-specific consequences. Third, a longitudinal imaging screen detected distinct consequences of gene knockdown on neuronal morphology. Our results highlight the power of unbiased genetic screens in iPSC-derived differentiated cell types and provide a platform for systematic interrogation of normal and disease states of neurons. VIDEO ABSTRACT.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Silenciamiento del Gen/métodos , Células Madre Pluripotentes Inducidas , Neuronas/metabolismo , Supervivencia Celular , Humanos , Microscopía Confocal , Neuronas/citología , RNA-Seq , Análisis de la Célula Individual
7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(7): 647-657, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28342963

RESUMEN

BACKGROUND: The two control points of cholesterol synthesis, 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) and squalene monooxygenase (SQLE) are known targets of the transcription factor sterol-regulatory element binding protein-2 (SREBP-2). Yet the location of the sterol-regulatory elements (SREs) and cofactor binding sites, nuclear factor-Y (NF-Y) and specificity protein 1 (Sp1), have not been satisfactorily mapped in the human SQLE promoter, or at all in the human HMGCR promoter. METHODS: We used luciferase reporter assays to screen the sterol-responsiveness of a library of predicted SRE, Sp1 and NF-Y site mutants and hence identify bone fide binding sites. We confirmed SREs via an electrophoretic mobility shift assay (EMSA) and ChIP-PCR. RESULTS: We identified two SREs in close proximity in both the human HMGCR and SQLE promoters, as well as one NF-Y site in HMGCR and two in SQLE. In addition, we found that HMGCR expression is highly activated only when SREBP-2 levels are very high, in contrast to the low density lipoprotein receptor (LDLR), a result reflected in mouse models used in other studies. CONCLUSIONS: Both HMGCR and SQLE promoters have two SREs that may act as a homing region to attract a single SREBP-2 homodimer, with HMGCR being activated only when there is absolute need for cholesterol synthesis. This ensures preferential uptake of exogenous cholesterol via LDLR, thereby conserving energy. GENERAL SIGNIFICANCE: We provide the first comprehensive investigation of SREs and NF-Ys in the human HMGCR and SQLE promoters, increasing our fundamental understanding of the transcriptional regulation of cholesterol synthesis.


Asunto(s)
Colesterol/metabolismo , Hidroximetilglutaril-CoA Reductasas/genética , NADH NADPH Oxidorreductasas/genética , Regiones Promotoras Genéticas/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Colesterol/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/genética , Genes Reporteros/genética , Células HeLa , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Luciferasas/metabolismo , Ratones , NADH NADPH Oxidorreductasas/metabolismo , Proteínas Nucleares/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Alineación de Secuencia , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
8.
Methods Mol Biol ; 1583: 211-219, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28205177

RESUMEN

The development of gas chromatography/mass spectrometry (GC/MS) technology has improved the ease and efficiency with which sterols in biological samples can be analyzed. Its advantages include that it needs only a small amount of sample, a short analysis time, and has enhanced specificity over traditional methods. Furthermore, a major benefit is its nonselective properties, which means that a complete scan of the sample will display the relative abundance of every sterol in the sample. This property has made it possible to define the abnormal, but distinctive, sterol profiles in a number of inborn errors of cholesterol synthesis. Here, we describe a semiquantitative method to determine relative activity of cholesterol synthesis enzymes. As an example, we measure the relative abundance of the substrate and product sterols of a cholesterol synthetic enzyme, 24-dehydrocholesterol reductase (DHCR24), which is defective in the hereditary developmental disease desmosterolosis.


Asunto(s)
Colesterol , Cromatografía de Gases y Espectrometría de Masas/métodos , Proteínas del Tejido Nervioso , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Anomalías Múltiples/enzimología , Animales , Colesterol/biosíntesis , Colesterol/química , Humanos , Errores Innatos del Metabolismo Lipídico/enzimología , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/análisis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/química , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo
9.
J Steroid Biochem Mol Biol ; 165(Pt B): 363-368, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27520299

RESUMEN

Cholesterol is essential for survival, but too much or too little can cause disease. Thus, cholesterol levels must be kept within close margins. 7-dehydrocholesterol reductase (DHCR7) is a terminal enzyme of cholesterol synthesis, and is essential for embryonic development. Largely, DHCR7 research is associated with the developmental disease Smith-Lemli-Opitz syndrome, which is caused by mutations in the DHCR7 gene. However, little is known about what regulates DHCR7 activity. Here we provide evidence that phosphorylation plays a role in controlling DHCR7 activity, which may provide a means to divert flux from cholesterol synthesis to vitamin D production. DHCR7 activity was significantly decreased when we used pharmacological inhibitors against two important kinases, AMP-activated protein kinase and protein kinase A. Moreover, mutating a known phosphorylated residue, S14, also decreased DHCR7 activity. Thus, we demonstrate that phosphorylation modulates DHCR7 activity in cells, and contributes to the overall synthesis of cholesterol, and probably vitamin D.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Colesterol/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Enzimológica de la Expresión Génica , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Cromatografía de Gases y Espectrometría de Masas , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , ARN Interferente Pequeño/metabolismo , Síndrome de Smith-Lemli-Opitz/metabolismo , Vitamina D/metabolismo
10.
Prog Lipid Res ; 64: 138-151, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27697512

RESUMEN

The conversion of 7-dehydrocholesterol to cholesterol, the final step of cholesterol synthesis in the Kandutsch-Russell pathway, is catalyzed by the enzyme 7-dehydrocholesterol reductase (DHCR7). Homozygous or compound heterozygous mutations in DHCR7 lead to the developmental disease Smith-Lemli-Opitz syndrome, which can also result in fetal mortality, highlighting the importance of this enzyme in human development and survival. Besides serving as a substrate for DHCR7, 7-dehydrocholesterol is also a precursor of vitamin D via the action of ultraviolet light on the skin. Thus, DHCR7 exerts complex biological effects, involved in both cholesterol and vitamin D production. Indeed, we argue that DHCR7 can act as a switch between cholesterol and vitamin D synthesis. This review summarizes current knowledge about the critical enzyme DHCR7, highlighting recent findings regarding its structure, transcriptional and post-transcriptional regulation, and its links to vitamin D synthesis. Greater understanding about DHCR7 function, regulation and its place within cellular metabolism will provide important insights into its biological roles.


Asunto(s)
Colesterol/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Vitamina D/metabolismo , Animales , Deshidrocolesteroles/metabolismo , Embrión no Mamífero/metabolismo , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/química , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Dominios Proteicos , Síndrome de Smith-Lemli-Opitz/metabolismo , Síndrome de Smith-Lemli-Opitz/patología , Xenopus/crecimiento & desarrollo , Xenopus/metabolismo
11.
J Biol Chem ; 291(16): 8363-73, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26887953

RESUMEN

Cholesterol is detrimental to human health in excess but is also essential for normal embryogenesis. Hence, enzymes involved in its synthesis possess many layers of regulation to achieve balanced cholesterol levels. 7-Dehydrocholesterol reductase (DHCR7) is the terminal enzyme of cholesterol synthesis in the Kandutsch-Russell pathway, converting 7-dehydrocholesterol (7DHC) to cholesterol. In the absence of functional DHCR7, accumulation of 7DHC and a lack of cholesterol production leads to the devastating developmental disorder, Smith-Lemli-Opitz syndrome. This study identifies that statin treatment can ameliorate the low DHCR7 expression seen with common Smith-Lemli-Opitz syndrome mutations. Furthermore, we show that wild-type DHCR7 is also relatively labile. In an example of end-product inhibition, cholesterol accelerates the proteasomal degradation of DHCR7, resulting in decreased protein levels and activity. The loss of enzymatic activity results in the accumulation of the substrate 7DHC, which leads to an increased production of vitamin D. Thus, these findings highlight DHCR7 as an important regulatory switch between cholesterol and vitamin D synthesis.


Asunto(s)
Deshidrocolesteroles/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Vitamina D/biosíntesis , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Complejo de la Endopetidasa Proteasomal/genética , Síndrome de Smith-Lemli-Opitz/enzimología , Síndrome de Smith-Lemli-Opitz/genética , Síndrome de Smith-Lemli-Opitz/patología , Vitamina D/genética
12.
J Nutr Sci Vitaminol (Tokyo) ; 61 Suppl: S154-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26598836

RESUMEN

Cholesterol is vital for human life, but its levels must be tightly regulated. Too little cholesterol leads to developmental disorders, but too much is widely appreciated as contributing to heart disease. Levels are regulated through the coordinated control of cholesterol synthesis, uptake and efflux. Here, we focus on cholesterol synthesis. The cholesterol synthesis pathway involves more than twenty enzymes, but most research so far has focused on a very early enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR), a well characterised control point. This is largely because HMGCR is the target of the successful cholesterol-lowering drugs, the statins. Our recent work has examined several other enzymes in the pathway and revealed complex regulatory mechanisms that also contribute to the control of cholesterol synthesis. In this review, we discuss the transcriptional regulation of the two terminal enzymes, 7- and 24-dehydrocholesterol reductase (DHCR7 and DHCR24), where we have found that a cooperative transcriptional program exists. We also discuss the post-translational regulation of another critical enzyme, squalene monooxygenase (SM), which has its protein levels controlled by cholesterol, and DHCR24, which has its activity affected by sterols and related compounds, as well as via phosphorylation/signalling. There is an unforeseen complexity in the regulation of cholesterol synthesis which requires further investigation.


Asunto(s)
Vías Biosintéticas , Colesterol/biosíntesis , Regulación de la Expresión Génica , Hidroximetilglutaril-CoA Reductasas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Humanos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Fosforilación , Escualeno-Monooxigenasa/metabolismo
13.
Biochim Biophys Acta ; 1842(10): 1431-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25048193

RESUMEN

The enzyme 7-dehydrocholesterol reductase (DHCR7) catalyzes the final step of cholesterol synthesis via the Kandutsch-Russell pathway, and is crucial in maintaining cellular cholesterol levels. Its absence leads to the devastating fetal developmental disorder Smith-Lemli-Opitz Syndrome (SLOS). How this enzyme is regulated has implications in controlling not only cholesterol synthesis, but also the synthesis of Vitamin D - another product of 7-dehydrocholesterol. In this study, we look specifically at how DHCR7 is regulated by the sterol regulatory element-binding protein-2 (SREBP-2) transcription factor. Sterol regulation has previously been studied in the rat DHCR7 promoter, but we have found that its regulatory elements are not all conserved in humans. Rather, the human promoter contains two binding sites for SREBP-2 (at -155 and -55) and a binding site for the nuclear factor-Y (NF-Y) cofactor (at -136). The -155 site is a particularly responsive sterol regulatory element (SRE) which is well conserved in mammals, and was possibly overlooked in the rat promoter study. The exact location of the weaker -55 site (close to the known rat SRE) may have shifted during evolution. Furthermore, we established that the two SREs that bind SREBP-2 work in cooperation to synergistically activate DHCR7. We have previously characterized the SREs in DHCR24, the final enzyme in the alternate Bloch pathway of cholesterol synthesis. Here, comparison of the sterol regulation of these terminal enzymes demonstrates the unique cooperative system that helps to control cholesterol synthesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA