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.
Curr Biol ; 34(9): 1853-1865.e6, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38604167

RESUMEN

Different signaling mechanisms concur to ensure robust tissue patterning and cell fate instruction during animal development. Most of these mechanisms rely on signaling proteins that are produced, transported, and detected. The spatiotemporal dynamics of signaling molecules are largely unknown, yet they determine signal activity's spatial range and time frame. Here, we use the Caenorhabditis elegans embryo to study how Wnt ligands, an evolutionarily conserved family of signaling proteins, dynamically organize to establish cell polarity in a developing tissue. We identify how Wnt ligands, produced in the posterior half of the embryos, spread extracellularly to transmit information to distant target cells in the anterior half. With quantitative live imaging and fluorescence correlation spectroscopy, we show that Wnt ligands diffuse through the embryo over a timescale shorter than the cell cycle, in the intercellular space, and outside the tissue below the eggshell. We extracted diffusion coefficients of Wnt ligands and their receptor Frizzled and characterized their co-localization. Integrating our different measurements and observations in a simple computational framework, we show how fast diffusion in the embryo can polarize individual cells through a time integration of the arrival of the ligands at the target cells. The polarity established at the tissue level by a posterior Wnt source can be transferred to the cellular level. Our results support a diffusion-based long-range Wnt signaling, which is consistent with the dynamics of developing processes.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Polaridad Celular , Embrión no Mamífero , Proteínas Wnt , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Embrión no Mamífero/metabolismo , Embrión no Mamífero/embriología , Ligandos , Vía de Señalización Wnt , Difusión
2.
Nature ; 627(8005): 873-879, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38418882

RESUMEN

Cyclic GMP-AMP synthase (cGAS) senses aberrant DNA during infection, cancer and inflammatory disease, and initiates potent innate immune responses through the synthesis of 2'3'-cyclic GMP-AMP (cGAMP)1-7. The indiscriminate activity of cGAS towards DNA demands tight regulatory mechanisms that are necessary to maintain cell and tissue homeostasis under normal conditions. Inside the cell nucleus, anchoring to nucleosomes and competition with chromatin architectural proteins jointly prohibit cGAS activation by genomic DNA8-15. However, the fate of nuclear cGAS and its role in cell physiology remains unclear. Here we show that the ubiquitin proteasomal system (UPS) degrades nuclear cGAS in cycling cells. We identify SPSB3 as the cGAS-targeting substrate receptor that associates with the cullin-RING ubiquitin ligase 5 (CRL5) complex to ligate ubiquitin onto nuclear cGAS. A cryo-electron microscopy structure of nucleosome-bound cGAS in a complex with SPSB3 reveals a highly conserved Asn-Asn (NN) minimal degron motif at the C terminus of cGAS that directs SPSB3 recruitment, ubiquitylation and cGAS protein stability. Interference with SPSB3-regulated nuclear cGAS degradation primes cells for type I interferon signalling, conferring heightened protection against infection by DNA viruses. Our research defines protein degradation as a determinant of cGAS regulation in the nucleus and provides structural insights into an element of cGAS that is amenable to therapeutic exploitation.


Asunto(s)
Proteínas Nucleares , Nucleosomas , Nucleotidiltransferasas , Proteolisis , Ubiquitina-Proteína Ligasas , Animales , Humanos , Ratones , Núcleo Celular/metabolismo , Microscopía por Crioelectrón , Degrones , Infecciones por Virus ADN/inmunología , Virus ADN/inmunología , Virus ADN/metabolismo , ADN Viral/inmunología , ADN Viral/metabolismo , Inmunidad Innata , Reconocimiento de Inmunidad Innata , Interferón Tipo I/inmunología , Proteínas Nucleares/metabolismo , Nucleosomas/química , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/ultraestructura , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Especificidad por Sustrato , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/ultraestructura , Ubiquitinación
3.
Nat Cancer ; 4(9): 1326-1344, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37640930

RESUMEN

The lysyl oxidase family represents a promising target in stromal targeting of solid tumors due to the importance of this family in crosslinking and stabilizing fibrillar collagens and its known role in tumor desmoplasia. Using small-molecule drug-design approaches, we generated and validated PXS-5505, a first-in-class highly selective and potent pan-lysyl oxidase inhibitor. We demonstrate in vitro and in vivo that pan-lysyl oxidase inhibition decreases chemotherapy-induced pancreatic tumor desmoplasia and stiffness, reduces cancer cell invasion and metastasis, improves tumor perfusion and enhances the efficacy of chemotherapy in the autochthonous genetically engineered KPC model, while also demonstrating antifibrotic effects in human patient-derived xenograft models of pancreatic cancer. PXS-5505 is orally bioavailable, safe and effective at inhibiting lysyl oxidase activity in tissues. Our findings present the rationale for progression of a pan-lysyl oxidase inhibitor aimed at eliciting a reduction in stromal matrix to potentiate chemotherapy in pancreatic ductal adenocarcinoma.


Asunto(s)
Enfermedades Pancreáticas , Neoplasias Pancreáticas , Humanos , Gemcitabina , Proteína-Lisina 6-Oxidasa , Neoplasias Pancreáticas/tratamiento farmacológico
4.
Sci Adv ; 9(17): eadf9063, 2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37126544

RESUMEN

Aberrant AKT activation occurs in a number of cancers, metabolic syndrome, and immune disorders, making it an important target for the treatment of many diseases. To monitor spatial and temporal AKT activity in a live setting, we generated an Akt-FRET biosensor mouse that allows longitudinal assessment of AKT activity using intravital imaging in conjunction with image stabilization and optical window technology. We demonstrate the sensitivity of the Akt-FRET biosensor mouse using various cancer models and verify its suitability to monitor response to drug targeting in spheroid and organotypic models. We also show that the dynamics of AKT activation can be monitored in real time in diverse tissues, including in individual islets of the pancreas, in the brown and white adipose tissue, and in the skeletal muscle. Thus, the Akt-FRET biosensor mouse provides an important tool to study AKT dynamics in live tissue contexts and has broad preclinical applications.


Asunto(s)
Técnicas Biosensibles , Proteínas Proto-Oncogénicas c-akt , Ratones , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Técnicas Biosensibles/métodos
5.
Cell Rep ; 36(11): 109689, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525350

RESUMEN

Assessing drug response within live native tissue provides increased fidelity with regards to optimizing efficacy while minimizing off-target effects. Here, using longitudinal intravital imaging of a Rac1-Förster resonance energy transfer (FRET) biosensor mouse coupled with in vivo photoswitching to track intratumoral movement, we help guide treatment scheduling in a live breast cancer setting to impair metastatic progression. We uncover altered Rac1 activity at the center versus invasive border of tumors and demonstrate enhanced Rac1 activity of cells in close proximity to live tumor vasculature using optical window imaging. We further reveal that Rac1 inhibition can enhance tumor cell vulnerability to fluid-flow-induced shear stress and therefore improves overall anti-metastatic response to therapy during transit to secondary sites such as the lung. Collectively, this study demonstrates the utility of single-cell intravital imaging in vivo to demonstrate that Rac1 inhibition can reduce tumor progression and metastases in an autochthonous setting to improve overall survival.


Asunto(s)
Técnicas Biosensibles/métodos , Neoplasias de la Mama/patología , Proteína de Unión al GTP rac1/metabolismo , Aminoquinolinas/farmacología , Animales , Neoplasias de la Mama/diagnóstico por imagen , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Transferencia Resonante de Energía de Fluorescencia , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Pirimidinas/farmacología , Resistencia al Corte , Transducción de Señal , Proteína de Unión al GTP rac1/antagonistas & inhibidores
6.
Nature ; 587(7835): 668-672, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32911482

RESUMEN

The DNA sensor cyclic GMP-AMP synthase (cGAS) initiates innate immune responses following microbial infection, cellular stress and cancer1. Upon activation by double-stranded DNA, cytosolic cGAS produces 2'3' cGMP-AMP, which triggers the induction of inflammatory cytokines and type I interferons 2-7. cGAS is also present inside the cell nucleus, which is replete with genomic DNA8, where chromatin has been implicated in restricting its enzymatic activity9. However, the structural basis for inhibition of cGAS by chromatin remains unknown. Here we present the cryo-electron microscopy structure of human cGAS bound to nucleosomes. cGAS makes extensive contacts with both the acidic patch of the histone H2A-H2B heterodimer and nucleosomal DNA. The structural and complementary biochemical analysis also find cGAS engaged to a second nucleosome in trans. Mechanistically, binding of the nucleosome locks cGAS into a monomeric state, in which steric hindrance suppresses spurious activation by genomic DNA. We find that mutations to the cGAS-acidic patch interface are sufficient to abolish the inhibitory effect of nucleosomes in vitro and to unleash the activity of cGAS on genomic DNA in living cells. Our work uncovers the structural basis of the interaction between cGAS and chromatin and details a mechanism that permits self-non-self discrimination of genomic DNA by cGAS.


Asunto(s)
Microscopía por Crioelectrón , Nucleosomas/metabolismo , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/química , Células HeLa , Histonas/metabolismo , Humanos , Modelos Moleculares , Mutación , Nucleosomas/química , Nucleosomas/ultraestructura , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/ultraestructura
7.
Development ; 147(7)2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32156756

RESUMEN

Wnt/ß-catenin signalling has been implicated in the terminal asymmetric divisions of neuronal progenitors in vertebrates and invertebrates. However, the role of Wnt ligands in this process remains poorly characterized. Here, we used the terminal divisions of the embryonic neuronal progenitors in C. elegans to characterize the role of Wnt ligands during this process, focusing on a lineage that produces the cholinergic interneuron AIY. We observed that, during interphase, the neuronal progenitor is elongated along the anteroposterior axis, then divides along its major axis, generating an anterior and a posterior daughter with different fates. Using time-controlled perturbations, we show that three Wnt ligands, which are transcribed at higher levels at the posterior of the embryo, regulate the orientation of the neuronal progenitor and its asymmetric division. We also identify a role for a Wnt receptor (MOM-5) and a cortical transducer APC (APR-1), which are, respectively, enriched at the posterior and anterior poles of the neuronal progenitor. Our study establishes a role for Wnt ligands in the regulation of the shape and terminal asymmetric divisions of neuronal progenitors, and identifies downstream components.


Asunto(s)
División Celular Asimétrica/genética , Caenorhabditis elegans/embriología , Células-Madre Neurales/citología , Proteínas Wnt/fisiología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , División Celular/genética , Polaridad Celular , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/fisiología , Ligandos , Células-Madre Neurales/fisiología , Neuronas/citología , Neuronas/fisiología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
8.
Nat Commun ; 10(1): 3637, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31406163

RESUMEN

Heterogeneous subtypes of cancer-associated fibroblasts (CAFs) coexist within pancreatic cancer tissues and can both promote and restrain disease progression. Here, we interrogate how cancer cells harboring distinct alterations in p53 manipulate CAFs. We reveal the existence of a p53-driven hierarchy, where cancer cells with a gain-of-function (GOF) mutant p53 educate a dominant population of CAFs that establish a pro-metastatic environment for GOF and null p53 cancer cells alike. We also demonstrate that CAFs educated by null p53 cancer cells may be reprogrammed by either GOF mutant p53 cells or their CAFs. We identify perlecan as a key component of this pro-metastatic environment. Using intravital imaging, we observe that these dominant CAFs delay cancer cell response to chemotherapy. Lastly, we reveal that depleting perlecan in the stroma combined with chemotherapy prolongs mouse survival, supporting it as a potential target for anti-stromal therapies in pancreatic cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Resistencia a Antineoplásicos/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Neoplasias Pancreáticas/patología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/genética , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica/patología , Páncreas/patología , Neoplasias Pancreáticas/genética , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/genética
9.
Elife ; 72018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-29985127

RESUMEN

Intravital microscopy can provide unique insights into the function of biological processes in a native context. However, physiological motion caused by peristalsis, respiration and the heartbeat can present a significant challenge, particularly for functional readouts such as fluorescence lifetime imaging (FLIM), which require longer acquisition times to obtain a quantitative readout. Here, we present and benchmark Galene, a versatile multi-platform software tool for image-based correction of sample motion blurring in both time resolved and conventional laser scanning fluorescence microscopy data in two and three dimensions. We show that Galene is able to resolve intravital FLIM-FRET images of intra-abdominal organs in murine models and NADH autofluorescence of human dermal tissue imaging subject to a wide range of physiological motions. Thus, Galene can enable FLIM imaging in situations where a stable imaging platform is not always possible and rescue previously discarded quantitative imaging data.


Asunto(s)
Imagenología Tridimensional , Microscopía Intravital , Movimiento (Física) , Algoritmos , Animales , Técnicas Biosensibles , Adhesión Celular , Simulación por Computador , Transferencia Resonante de Energía de Fluorescencia , Guanosina Trifosfato/metabolismo , Humanos , Intestinos/fisiología , Ratones , Microscopía Fluorescente , Modelos Biológicos , Metástasis de la Neoplasia , Neuropéptidos/metabolismo , Neoplasias Pancreáticas/patología , Piel/anatomía & histología , Programas Informáticos , Proteína de Unión al GTP rac1/metabolismo , Familia-src Quinasas/metabolismo
10.
Cell Rep ; 23(11): 3312-3326, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29898401

RESUMEN

Application of advanced intravital imaging facilitates dynamic monitoring of pathway activity upon therapeutic inhibition. Here, we assess resistance to therapeutic inhibition of the PI3K pathway within the hypoxic microenvironment of pancreatic ductal adenocarcinoma (PDAC) and identify a phenomenon whereby pronounced hypoxia-induced resistance is observed for three clinically relevant inhibitors. To address this clinical problem, we have mapped tumor hypoxia by both immunofluorescence and phosphorescence lifetime imaging of oxygen-sensitive nanoparticles and demonstrate that these hypoxic regions move transiently around the tumor. To overlay this microenvironmental information with drug response, we applied a FRET biosensor for Akt activity, which is a key effector of the PI3K pathway. Performing dual intravital imaging of drug response in different tumor compartments, we demonstrate an improved drug response to a combination therapy using the dual mTORC1/2 inhibitor AZD2014 with the hypoxia-activated pro-drug TH-302.


Asunto(s)
Resistencia a Antineoplásicos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Animales , Benzamidas , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Quimioterapia Combinada , Femenino , Transferencia Resonante de Energía de Fluorescencia , Humanos , Hipoxia , Microscopía Intravital/métodos , Ratones , Ratones Endogámicos BALB C , Morfolinas/farmacología , Morfolinas/uso terapéutico , Nanopartículas/química , Nitroimidazoles/farmacología , Nitroimidazoles/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Mostazas de Fosforamida/farmacología , Mostazas de Fosforamida/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas , Transducción de Señal/efectos de los fármacos , Trasplante Heterólogo , Microambiente Tumoral
11.
Sci Transl Med ; 9(384)2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28381539

RESUMEN

The emerging standard of care for patients with inoperable pancreatic cancer is a combination of cytotoxic drugs gemcitabine and Abraxane, but patient response remains moderate. Pancreatic cancer development and metastasis occur in complex settings, with reciprocal feedback from microenvironmental cues influencing both disease progression and drug response. Little is known about how sequential dual targeting of tumor tissue tension and vasculature before chemotherapy can affect tumor response. We used intravital imaging to assess how transient manipulation of the tumor tissue, or "priming," using the pharmaceutical Rho kinase inhibitor Fasudil affects response to chemotherapy. Intravital Förster resonance energy transfer imaging of a cyclin-dependent kinase 1 biosensor to monitor the efficacy of cytotoxic drugs revealed that priming improves pancreatic cancer response to gemcitabine/Abraxane at both primary and secondary sites. Transient priming also sensitized cells to shear stress and impaired colonization efficiency and fibrotic niche remodeling within the liver, three important features of cancer spread. Last, we demonstrate a graded response to priming in stratified patient-derived tumors, indicating that fine-tuned tissue manipulation before chemotherapy may offer opportunities in both primary and metastatic targeting of pancreatic cancer.


Asunto(s)
Progresión de la Enfermedad , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/uso terapéutico , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Paclitaxel Unido a Albúmina/farmacología , Paclitaxel Unido a Albúmina/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Técnicas Biosensibles , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Matriz Extracelular/metabolismo , Humanos , Hígado/patología , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Quinasas Asociadas a rho/metabolismo , Familia-src Quinasas/metabolismo , Gemcitabina
12.
Dev Cell ; 33(6): 737-45, 2015 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-26073017

RESUMEN

Transcription factors of the TCF family are key mediators of the Wnt/ß-catenin pathway. TCF usually activates transcription on cis-regulatory elements containing TCF binding sites when the pathway is active and represses transcription when the pathway is inactive. However, some direct targets display an opposite regulation (activated by TCF in the absence of Wnt), but the mechanism behind this atypical regulation remains poorly characterized. Here, we use the cis-regulatory region of an opposite target gene, ttx-3, to dissect the mechanism of this atypical regulation. Using a combination of genetic, molecular, and biochemical experiments, we establish that, in the absence of Wnt pathway activation, TCF activates ttx-3 expression via a Zic binding site by forming a complex with a Zic transcription factor. This mechanism is later reinforced by specific bHLH factors. This study reveals an atypical mode of action for TCF that may apply to other binary decisions mediated by Wnt signaling.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción TCF/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Modelos Neurológicos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Factores de Transcripción TCF/genética , Factores de Transcripción/genética , Activación Transcripcional , Vía de Señalización Wnt
13.
J Vis Exp ; (87)2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24836407

RESUMEN

Fast and low phototoxic imaging techniques are pre-requisite to study the development of organisms in toto. Light sheet based microscopy reduces photo-bleaching and phototoxic effects compared to confocal microscopy, while providing 3D images with subcellular resolution. Here we present the setup of a light sheet based microscope, which is composed of an upright microscope and a small set of opto-mechanical elements for the generation of the light sheet. The protocol describes how to build, align the microscope and characterize the light sheet. In addition, it details how to implement the method for in toto imaging of C. elegans embryos using a simple observation chamber. The method allows the capture of 3D two-colors time-lapse movies over few hours of development. This should ease the tracking of cell shape, cell divisions and tagged proteins over long periods of time.


Asunto(s)
Caenorhabditis elegans/embriología , Microscopía/instrumentación , Microscopía/métodos , Animales , Embrión no Mamífero , Imagenología Tridimensional/instrumentación , Imagenología Tridimensional/métodos , Imagen de Lapso de Tiempo/instrumentación , Imagen de Lapso de Tiempo/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA