Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(6): 114243, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38805398

RESUMEN

Xeroderma pigmentosum (XP) is caused by defective nucleotide excision repair of DNA damage. This results in hypersensitivity to ultraviolet light and increased skin cancer risk, as sunlight-induced photoproducts remain unrepaired. However, many XP patients also display early-onset neurodegeneration, which leads to premature death. The mechanism of neurodegeneration is unknown. Here, we investigate XP neurodegeneration using pluripotent stem cells derived from XP patients and healthy relatives, performing functional multi-omics on samples during neuronal differentiation. We show substantially increased levels of 5',8-cyclopurine and 8-oxopurine in XP neuronal DNA secondary to marked oxidative stress. Furthermore, we find that the endoplasmic reticulum stress response is upregulated and reversal of the mutant genotype is associated with phenotypic rescue. Critically, XP neurons exhibit inappropriate downregulation of the protein clearance ubiquitin-proteasome system (UPS). Chemical enhancement of UPS activity in XP neuronal models improves phenotypes, albeit inadequately. Although more work is required, this study presents insights with intervention potential.

2.
Cell Rep ; : 114016, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38636520

RESUMEN

How cancer cells determine their shape in response to three-dimensional (3D) geometric and mechanical cues is unclear. We develop an approach to quantify the 3D cell shape of over 60,000 melanoma cells in collagen hydrogels using high-throughput stage-scanning oblique plane microscopy (ssOPM). We identify stereotypic and environmentally dependent changes in shape and protrusivity depending on whether a cell is proximal to a flat and rigid surface or is embedded in a soft environment. Environmental sensitivity metrics calculated for small molecules and gene knockdowns identify interactions between the environment and cellular factors that are important for morphogenesis. We show that the Rho guanine nucleotide exchange factor (RhoGEF) TIAM2 contributes to shape determination in environmentally independent ways but that non-muscle myosin II, microtubules, and the RhoGEF FARP1 regulate shape in ways dependent on the microenvironment. Thus, changes in cancer cell shape in response to 3D geometric and mechanical cues are modulated in both an environmentally dependent and independent fashion.

3.
Nucleic Acids Res ; 51(12): 6337-6354, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37224534

RESUMEN

Accurate genome replication is essential for all life and a key mechanism of disease prevention, underpinned by the ability of cells to respond to replicative stress (RS) and protect replication forks. These responses rely on the formation of Replication Protein A (RPA)-single stranded (ss) DNA complexes, yet this process remains largely uncharacterized. Here, we establish that actin nucleation-promoting factors (NPFs) associate with replication forks, promote efficient DNA replication and facilitate association of RPA with ssDNA at sites of RS. Accordingly, their loss leads to deprotection of ssDNA at perturbed forks, impaired ATR activation, global replication defects and fork collapse. Supplying an excess of RPA restores RPA foci formation and fork protection, suggesting a chaperoning role for actin nucleators (ANs) (i.e. Arp2/3, DIAPH1) and NPFs (i.e, WASp, N-WASp) in regulating RPA availability upon RS. We also discover that ß-actin interacts with RPA directly in vitro, and in vivo a hyper-depolymerizing ß-actin mutant displays a heightened association with RPA and the same dysfunctional replication phenotypes as loss of ANs/NPFs, which contrasts with the phenotype of a hyper-polymerizing ß-actin mutant. Thus, we identify components of actin polymerization pathways that are essential for preventing ectopic nucleolytic degradation of perturbed forks by modulating RPA activity.


Asunto(s)
Actinas , Replicación del ADN , Actinas/genética , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , ADN de Cadena Simple/genética , Chaperonas Moleculares/genética
4.
bioRxiv ; 2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36711944

RESUMEN

Accurate genome replication is essential for all life and a key mechanism of disease prevention, underpinned by the ability of cells to respond to replicative stress (RS) and protect replication forks. These responses rely on the formation of Replication Protein A (RPA)-single stranded (ss) DNA complexes, yet this process remains largely uncharacterized. Here we establish that actin nucleation-promoting factors (NPFs) associate with replication forks, promote efficient DNA replication and facilitate association of RPA with ssDNA at sites of RS. Accordingly, their loss leads to deprotection of ssDNA at perturbed forks, impaired ATR activation, global replication defects and fork collapse. Supplying an excess of RPA restores RPA foci formation and fork protection, suggesting a chaperoning role for actin nucleators (ANs) (i.e., Arp2/3, DIAPH1) and NPFs (i.e, WASp, N-WASp) in regulating RPA availability upon RS. We also discover that ß-actin interacts with RPA directly in vitro , and in vivo a hyper-depolymerizing ß-actin mutant displays a heightened association with RPA and the same dysfunctional replication phenotypes as loss of ANs/NPFs, which contrasts with the phenotype of a hyper-polymerizing ß-actin mutant. Thus, we identify components of actin polymerization pathways that are essential for preventing ectopic nucleolytic degradation of perturbed forks by modulating RPA activity.

5.
STAR Protoc ; 4(1): 101942, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36525347

RESUMEN

Small interfering RNA (siRNA) screening approaches used with quantitative single-cell analysis can uncover the roles of genes in cell morphogenesis. Here, we present a high-throughput automated phenotypic screening technique to quantify a single cell shape in cancer cells cultured on top of soft 3D hydrogels. We describe reverse transfection of cells with siRNAs and seeding of these cells on top of collagen, followed by image analysis to quantify morphology of a single cell and population levels in low-elasticity matrices. For complete details on the use and execution of this protocol, please refer to Bousgouni et al. (2022).1.


Asunto(s)
Hidrogeles , Neoplasias , Ensayos Analíticos de Alto Rendimiento/métodos , ARN Interferente Pequeño/genética , Diagnóstico por Imagen , Fenotipo , Neoplasias/genética
6.
iScience ; 25(8): 104795, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-36039362

RESUMEN

Rho GTP Exchange Factors (RhoGEFs) and Rho GTPase Activating Proteins (RhoGAPs) are large families of molecules that regulate shape determination in all eukaryotes. In pathologies such as melanoma, RhoGEF and RhoGAP activity underpins the ability of cells to invade tissues of varying elasticity. To identify RhoGEFs and RhoGAPs that regulate melanoma cell shape on soft and/or stiff materials, we performed genetic screens, in tandem with single-cell quantitative morphological analysis. We show that ARHGEF9/Collybistin (Cb) is essential for cell shape determination on both soft and stiff materials, and in cells embedded in 3D soft hydrogel. ARHGEF9 is required for melanoma cells to invade 3D matrices. Depletion of ARHGEF9 results in loss of tension at focal adhesions decreased cell-wide contractility, and the inability to stabilize protrusions. Taken together we show that ARHGEF9 promotes the formation of actin-rich filopodia, which serves to establish and stabilize adhesions and determine melanoma cell shape.

7.
J Microsc ; 288(2): 130-141, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-34089183

RESUMEN

We presenta robust, long-range optical autofocus system for microscopy utilizing machine learning. This can be useful for experiments with long image data acquisition times that may be impacted by defocusing resulting from drift of components, for example due to changes in temperature or mechanical drift. It is also useful for automated slide scanning or multiwell plate imaging where the sample(s) to be imaged may not be in the same horizontal plane throughout the image data acquisition. To address the impact of (thermal or mechanical) fluctuations over time in the optical autofocus system itself, we utilize a convolutional neural network (CNN) that is trained over multiple days to account for such fluctuations. To address the trade-off between axial precision and range of the autofocus, we implement orthogonal optical readouts with separate CNN training data, thereby achieving an accuracy well within the 600 nm depth of field of our 1.3 numerical aperture objective lens over a defocus range of up to approximately +/-100 µm. We characterize the performance of this autofocus system and demonstrate its application to automated multiwell plate single molecule localization microscopy.


Many microscopy experiments involve extended imaging of samples over timescales from minutes to days, during which the microscope can 'drift' out of focus. When imaging at high magnification, the depth of field is of the order of one micron and so the imaging system should keep the sample in the focal plane of the microscope objective lens to this precision. Unfortunately, temperature changes in the laboratory can cause thermal expansion of microscope components that can move the focal plane by more than a micron and such changes can occur on a timescale of minutes. This is a particular issue for super-resolved microscopy experiments using single molecule localization microscopy (SMLM) techniques, for which 1000s of images are acquired, and for automated imaging of multiple samples in multiwell plates. It is possible to maintain the sample in the focal plane focus position by either automatically moving the sample or adjusting the imaging system, for example by moving the objective lens. This is called 'autofocus' and is frequently achieved by reflecting a light beam from the microscope coverslip and measuring its position of beam profile as a function of defocus of the microscope. The correcting adjustment is then usually calculated analytically but there is recent interest in using machine learning techniques to determine the required focussing adjustment. Here, we present a system that uses a neural network to determine the required defocus correcting adjustment from camera images of a laser beam that is reflected from the coverslip. Unfortunately, this approach will only work when the microscope is in the same condition as it was when the neural network was trained - and this can be compromised by the same drift of the optical system that causes the defocus needing to be corrected. We show, however, that by training a neural network over an extended period, for example 10 days, this approach can 'learn' about the optical system drifts and provide the required autofocus function. We also show that an optical system utilizing a rectangular slit can make two measurements of the defocus simultaneously, with one measurement being optimized for high accuracy over a limited range (±10 µm) near focus and the other providing lower accuracy but over a much longer range (±100 µm). This robust autofocus system is suitable for automated super-resolved microscopy of arrays of samples in a multiwell plate using SMLM, for which an experiment routinely lasts more than 5 h.


Asunto(s)
Aprendizaje Profundo , Microscopía , Microscopía/métodos , Imagen Individual de Molécula , Aprendizaje Automático
8.
Nat Commun ; 11(1): 1851, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32296040

RESUMEN

Genome stability relies on proper coordination of mitosis and cytokinesis, where dynamic microtubules capture and faithfully segregate chromosomes into daughter cells. With a high-content RNAi imaging screen targeting more than 2,000 human lncRNAs, we identify numerous lncRNAs involved in key steps of cell division such as chromosome segregation, mitotic duration and cytokinesis. Here, we provide evidence that the chromatin-associated lncRNA, linc00899, leads to robust mitotic delay upon its depletion in multiple cell types. We perform transcriptome analysis of linc00899-depleted cells and identify the neuronal microtubule-binding protein, TPPP/p25, as a target of linc00899. We further show that linc00899 binds TPPP/p25 and suppresses its transcription. In cells depleted of linc00899, upregulation of TPPP/p25 alters microtubule dynamics and delays mitosis. Overall, our comprehensive screen uncovers several lncRNAs involved in genome stability and reveals a lncRNA that controls microtubule behaviour with functional implications beyond cell division.


Asunto(s)
División Celular/genética , División Celular/fisiología , ARN Largo no Codificante/genética , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Mitosis/genética , Mitosis/fisiología , Proteínas/genética , Interferencia de ARN/fisiología
9.
Stem Cell Reports ; 12(4): 743-756, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30880078

RESUMEN

Differentiated cells are epigenetically stable, but can be reprogrammed to pluripotency by expression of the OSKM transcription factors. Despite significant effort, relatively little is known about the cellular requirements for reprogramming and how they affect the properties of induced pluripotent stem cells. We have performed high-content screening with small interfering RNAs targeting 300 chromatin-associated factors and extracted colony-level quantitative features. This revealed five morphological phenotypes in early reprogramming, including one displaying large round colonies exhibiting an early block of reprogramming. Using RNA sequencing, we identified transcriptional changes associated with these phenotypes. Furthermore, double knockdown epistasis experiments revealed that BRCA1, BARD1, and WDR5 functionally interact and are required for the DNA damage response. In addition, the mesenchymal-to-epithelial transition is affected in Brca1, Bard1, and Wdr5 knockdowns. Our data provide a resource of chromatin-associated factors in early reprogramming and underline colony morphology as an important high-dimensional readout for reprogramming quality.


Asunto(s)
Proteína BRCA1/genética , Reprogramación Celular/genética , Daño del ADN , Transición Epitelial-Mesenquimal/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Proteína BRCA1/metabolismo , Cromatina/genética , Cromatina/metabolismo , Reparación del ADN , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Fenotipo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
10.
Sci Data ; 4: 170018, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28248929

RESUMEN

In order to metastasise, triple negative breast cancer (TNBC) must make dynamic changes in cell shape. The shape of all eukaryotic cells is regulated by Rho Guanine Nucleotide Exchange Factors (RhoGEFs), which activate Rho-family GTPases in response to mechanical and informational cues. In contrast, Rho GTPase-activating proteins (RhoGAPs) inhibit Rho GTPases. However, which RhoGEFs and RhoGAPS couple TNBC cell shape to changes in their environment is very poorly understood. Moreover, whether the activity of particular RhoGEFs and RhoGAPs become dysregulated as cells evolve the ability to metastasise is not clear. Towards the ultimate goal of identifying RhoGEFs and RhoGAPs that are essential for TNBC metastasis, we performed an RNAi screen to isolate RhoGEFs and RhoGAPs that contribute to the morphogenesis of the highly metastatic TNBC cell line LM2, and its less-metastatic parental cell line MDA-MB-231. For ~6 million cells from each cell line, we measured 127 different features following the depletion of 142 genes. Using a linear classifier scheme we also describe the morphological heterogeneity of each gene-depleted population.


Asunto(s)
Proteínas Activadoras de GTPasa , Neoplasias de la Mama Triple Negativas , Femenino , Humanos , Interferencia de ARN , Neoplasias de la Mama Triple Negativas/enzimología , Neoplasias de la Mama Triple Negativas/patología
11.
Mol Biol Cell ; 26(22): 4163-70, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26310441

RESUMEN

Melanoma cells can adopt two functionally distinct forms, amoeboid and mesenchymal, which facilitates their ability to invade and colonize diverse environments during the metastatic process. Using quantitative imaging of single living tumor cells invading three-dimensional collagen matrices, in tandem with unsupervised computational analysis, we found that melanoma cells can switch between amoeboid and mesenchymal forms via two different routes in shape space--an apolar and polar route. We show that whereas particular Rho-family GTPases are required for the morphogenesis of amoeboid and mesenchymal forms, others are required for transitions via the apolar or polar route and not amoeboid or mesenchymal morphogenesis per se. Altering the transition rates between particular routes by depleting Rho-family GTPases can change the morphological heterogeneity of cell populations. The apolar and polar routes may have evolved in order to facilitate conversion between amoeboid and mesenchymal forms, as cells are either searching for, or attracted to, particular migratory cues, respectively.


Asunto(s)
Melanoma/patología , Animales , Bovinos , Línea Celular Tumoral , Movimiento Celular/fisiología , Forma de la Célula/fisiología , Colágeno/metabolismo , Humanos , Mesodermo/metabolismo , Invasividad Neoplásica , Cuerpos Polares/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Proteínas de Unión al GTP rho/metabolismo
12.
PLoS One ; 9(7): e101164, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25007267

RESUMEN

The function and capacity of the endoplasmic reticulum (ER) is determined by multiple processes ranging from the local regulation of peptide translation, translocation, and folding, to global changes in lipid composition. ER homeostasis thus requires complex interactions amongst numerous cellular components. However, describing the networks that maintain ER function during changes in cell behavior and environmental fluctuations has, to date, proven difficult. Here we perform a systems-level analysis of ER homeostasis, and find that although signaling networks that regulate ER function have a largely modular architecture, the TORC1-SREBP signaling axis is a central node that integrates signals emanating from different sub-networks. TORC1-SREBP promotes ER homeostasis by regulating phospholipid biosynthesis and driving changes in ER morphology. In particular, our network model shows TORC1-SREBP serves to integrate signals promoting growth and G1-S progression in order to maintain ER function during cell proliferation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Retículo Endoplásmico/fisiología , Homeostasis , Transducción de Señal , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Proliferación Celular , Drosophila melanogaster , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Ácidos Grasos Insaturados/fisiología , Puntos de Control de la Fase G1 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Metabolismo de los Lípidos , Interferencia de ARN , Respuesta de Proteína Desplegada
13.
Open Biol ; 4: 130132, 2014 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-24451547

RESUMEN

One goal of cell biology is to understand how cells adopt different shapes in response to varying environmental and cellular conditions. Achieving a comprehensive understanding of the relationship between cell shape and environment requires a systems-level understanding of the signalling networks that respond to external cues and regulate the cytoskeleton. Classical biochemical and genetic approaches have identified thousands of individual components that contribute to cell shape, but it remains difficult to predict how cell shape is generated by the activity of these components using bottom-up approaches because of the complex nature of their interactions in space and time. Here, we describe the regulation of cellular shape by signalling systems using a top-down approach. We first exploit the shape diversity generated by systematic RNAi screening and comprehensively define the shape space a migratory cell explores. We suggest a simple Boolean model involving the activation of Rac and Rho GTPases in two compartments to explain the basis for all cell shapes in the dataset. Critically, we also generate a probabilistic graphical model to show how cells explore this space in a deterministic, rather than a stochastic, fashion. We validate the predictions made by our model using live-cell imaging. Our work explains how cross-talk between Rho and Rac can generate different cell shapes, and thus morphological heterogeneity, in genetically identical populations.


Asunto(s)
Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Teorema de Bayes , Línea Celular , Forma de la Célula , Análisis por Conglomerados , Citoesqueleto/metabolismo , Drosophila/enzimología , Drosophila/metabolismo , Análisis de Componente Principal , Interferencia de ARN , Transducción de Señal , Programas Informáticos , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rho/genética
14.
Mol Biosyst ; 8(10): 2605-13, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22790786

RESUMEN

Reactive Oxygen Species (ROS) are a natural by-product of cellular growth and proliferation, and are required for fundamental processes such as protein-folding and signal transduction. However, ROS accumulation, and the onset of oxidative stress, can negatively impact cellular and genomic integrity. Signalling networks have evolved to respond to oxidative stress by engaging diverse enzymatic and non-enzymatic antioxidant mechanisms to restore redox homeostasis. The architecture of oxidative stress response networks during periods of normal growth, and how increased ROS levels dynamically reconfigure these networks are largely unknown. In order to gain insight into the structure of signalling networks that promote redox homeostasis we first performed genome-scale RNAi screens to identify novel suppressors of superoxide accumulation. We then infer relationships between redox regulators by hierarchical clustering of phenotypic signatures describing how gene inhibition affects superoxide levels, cellular viability, and morphology across different genetic backgrounds. Genes that cluster together are likely to act in the same signalling pathway/complex and thus make "functional interactions". Moreover we also calculate differential phenotypic signatures describing the difference in cellular phenotypes following RNAi between untreated cells and cells submitted to oxidative stress. Using both phenotypic signatures and differential signatures we construct a network model of functional interactions that occur between components of the redox homeostasis network, and how such interactions become rewired in the presence of oxidative stress. This network model predicts a functional interaction between the transcription factor Jun and the IRE1 kinase, which we validate in an orthogonal assay. We thus demonstrate the ability of systems-biology approaches to identify novel signalling events.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Endorribonucleasas/metabolismo , Redes Reguladoras de Genes , Paraquat/farmacología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Antioxidantes/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Endorribonucleasas/genética , Expresión Génica/efectos de los fármacos , Familia de Multigenes , Oxidación-Reducción , Estrés Oxidativo , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-jun/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Superóxidos/metabolismo
15.
J Alzheimers Dis ; 19(2): 631-46, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20110608

RESUMEN

In a recent clinical study, the thiazolidinedione (TZD) pioglitazone (Actos was reported to preserve cognitive function in patients with mild to moderate Alzheimer's disease and type II diabetes mellitus. TZDs are agonists of the nuclear hormone receptor peroxisome proliferator-activated receptor-gamma (PPARgamma), are peripheral insulin sensitizers, and have recently been reported to increase mitochondrial biogenesis in the central nervous system and dendritic spine density. We report a transcriptional profile of the TZD pioglitazone and the non-TZD PPARgamma agonist GW347845 in primary cortical culture. We observed that pioglitazone, but not GW347845, increased cholesterol biosynthetic and lipogenic gene expression after 6 h, and the expression of the cholesterol efflux transporters Abca1 and Abcg1 after 24 h. Co-treatment of pioglitazone with the PPARgamma antagonist GW9662 did not significantly reduce these effects, suggesting a PPARgamma-independent mechanism. These findings suggest a novel effect of TZDs in neurons that may be of relevance as a novel approach against Alzheimer's disease.


Asunto(s)
Hipoglucemiantes/farmacología , Neuronas/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Tiazolidinedionas/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Células Cultivadas , Corteza Cerebral/citología , Colesterol/biosíntesis , Colesterol/genética , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , PPAR gamma/antagonistas & inhibidores , Pioglitazona , Ratas , Ratas Sprague-Dawley , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA