Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(15): 4016-4031.e22, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34081922

RESUMEN

Cross-presentation of antigens from dead tumor cells by type 1 conventional dendritic cells (cDC1s) is thought to underlie priming of anti-cancer CD8+ T cells. cDC1 express high levels of DNGR-1 (a.k.a. CLEC9A), a receptor that binds to F-actin exposed by dead cell debris and promotes cross-presentation of associated antigens. Here, we show that secreted gelsolin (sGSN), an extracellular protein, decreases DNGR-1 binding to F-actin and cross-presentation of dead cell-associated antigens by cDC1s. Mice deficient in sGsn display increased DNGR-1-dependent resistance to transplantable tumors, especially ones expressing neoantigens associated with the actin cytoskeleton, and exhibit greater responsiveness to cancer immunotherapy. In human cancers, lower levels of intratumoral sGSN transcripts, as well as presence of mutations in proteins associated with the actin cytoskeleton, are associated with signatures of anti-cancer immunity and increased patient survival. Our results reveal a natural barrier to cross-presentation of cancer antigens that dampens anti-tumor CD8+ T cell responses.


Asunto(s)
Reactividad Cruzada/inmunología , Gelsolina/metabolismo , Inmunidad , Lectinas Tipo C/metabolismo , Neoplasias/inmunología , Receptores Inmunológicos/metabolismo , Receptores Mitogénicos/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reactividad Cruzada/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Gelsolina/química , Gelsolina/deficiencia , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad/efectos de los fármacos , Ratones Endogámicos C57BL , Mutación/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Unión Proteica/efectos de los fármacos , Análisis de Supervivencia
2.
BMC Biol ; 22(1): 26, 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38302906

RESUMEN

BACKGROUND: The ability of recombinant adeno-associated virus to transduce preimplantation mouse embryos has led to the use of this delivery method for the production of genetically altered knock-in mice via CRISPR-Cas9. The potential exists for this method to simplify the production and extend the types of alleles that can be generated directly in the zygote, obviating the need for manipulations of the mouse genome via the embryonic stem cell route. RESULTS: We present the production data from a total of 13 genetically altered knock-in mouse models generated using CRISPR-Cas9 electroporation of zygotes and delivery of donor repair templates via transduction with recombinant adeno-associated virus. We explore the efficiency of gene targeting at a total of 12 independent genetic loci and explore the effects of allele complexity and introduce strategies for efficient identification of founder animals. In addition, we investigate the reliability of germline transmission of the engineered allele from founder mice generated using this methodology. By comparing our production data against genetically altered knock-in mice generated via gene targeting in embryonic stem cells and their microinjection into blastocysts, we assess the animal cost of the two methods. CONCLUSIONS: Our results confirm that recombinant adeno-associated virus transduction of zygotes provides a robust and effective delivery route for donor templates for the production of knock-in mice, across a range of insertion sizes (0.9-4.7 kb). We find that the animal cost of this method is considerably less than generating knock-in models via embryonic stem cells and thus constitutes a considerable 3Rs reduction.


Asunto(s)
Sistemas CRISPR-Cas , Dependovirus , Ratones , Animales , Dependovirus/genética , Reproducibilidad de los Resultados , Cigoto , Marcación de Gen , Técnicas de Sustitución del Gen/métodos
3.
Nature ; 514(7523): 498-502, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25341788

RESUMEN

After immunogenic challenge, infiltrating and dividing lymphocytes markedly increase lymph node cellularity, leading to organ expansion. Here we report that the physical elasticity of lymph nodes is maintained in part by podoplanin (PDPN) signalling in stromal fibroblastic reticular cells (FRCs) and its modulation by CLEC-2 expressed on dendritic cells. We show in mouse cells that PDPN induces actomyosin contractility in FRCs via activation of RhoA/C and downstream Rho-associated protein kinase (ROCK). Engagement by CLEC-2 causes PDPN clustering and rapidly uncouples PDPN from RhoA/C activation, relaxing the actomyosin cytoskeleton and permitting FRC stretching. Notably, administration of CLEC-2 protein to immunized mice augments lymph node expansion. In contrast, lymph node expansion is significantly constrained in mice selectively lacking CLEC-2 expression in dendritic cells. Thus, the same dendritic cells that initiate immunity by presenting antigens to T lymphocytes also initiate remodelling of lymph nodes by delivering CLEC-2 to FRCs. CLEC-2 modulation of PDPN signalling permits FRC network stretching and allows for the rapid lymph node expansion--driven by lymphocyte influx and proliferation--that is the critical hallmark of adaptive immunity.


Asunto(s)
Células Dendríticas/fisiología , Fibroblastos/citología , Ganglios Linfáticos/citología , Células del Estroma/citología , Actomiosina/metabolismo , Animales , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Células Dendríticas/inmunología , Femenino , Fibroblastos/fisiología , Inflamación/inmunología , Lectinas Tipo C/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Células del Estroma/fisiología , Proteínas ras/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA , Proteína rhoC de Unión a GTP
4.
EMBO J ; 33(14): 1582-98, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24920579

RESUMEN

We have identified a new function for the dynein adaptor Bicaudal D homolog 1 (BICD1) by screening a siRNA library for genes affecting the dynamics of neurotrophin receptor-containing endosomes in motor neurons (MNs). Depleting BICD1 increased the intracellular accumulation of brain-derived neurotrophic factor (BDNF)-activated TrkB and p75 neurotrophin receptor (p75(NTR)) by disrupting the endosomal sorting, reducing lysosomal degradation and increasing the co-localisation of these neurotrophin receptors with retromer-associated sorting nexin 1. The resulting re-routing of active receptors increased their recycling to the plasma membrane and altered the repertoire of signalling-competent TrkB isoforms and p75(NTR) available for ligand binding on the neuronal surface. This resulted in attenuated, but more sustained, AKT activation in response to BDNF stimulation. These data, together with our observation that Bicd1 expression is restricted to the developing nervous system when neurotrophin receptor expression peaks, indicate that BICD1 regulates neurotrophin signalling by modulating the endosomal sorting of internalised ligand-activated receptors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Endosomas/metabolismo , Modelos Biológicos , Neuronas Motoras/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Proteínas del Citoesqueleto/genética , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Proteínas Luminiscentes , Ratones , Microscopía Electrónica de Transmisión , Transporte de Proteínas/fisiología , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Proteína Fluorescente Roja
5.
Eur J Immunol ; 45(12): 3386-403, 2015 12.
Artículo en Inglés | MEDLINE | ID: mdl-26457795

RESUMEN

IFN-α/ß allow cells to fight virus infection by inducing the expression of many genes that encode effectors of antiviral defense. One of these, the Ski2-like DExH-box helicase DDX60, was recently implicated in resistance of human cells to hepatitis C virus, as well as in induction of IFN-α/ß by retinoic acid inducible gene 1-like receptors (RLRs) that detect the presence of RNA viruses in a cell-intrinsic manner. Here, we sought to investigate the role of DDX60 in IFN-α/ß induction and in resistance to virus infection. Analysis of fibroblasts and myeloid cells from Ddx60-deficient mice revealed no impairment in IFN-α/ß production in response to RLR agonists, RNA viruses, or other stimuli. Moreover, overexpression of DDX60 did not potentiate IFN induction and DDX60 did not interact with RLRs or capture RLR agonists from virally infected cells. We also failed to identify any impairment in Ddx60-deficient murine cells or mice in resistance to infection with influenza A virus, encephalomyocarditis virus, Sindbis virus, vaccinia virus, or herpes simplex virus-1. These results put in question the reported role of DDX60 as a broad-acting positive regulator of RLR responses and hint at the possibility that it may function as a restriction factor highly specific for a particular virus or class of viruses.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Interferón Tipo I/biosíntesis , Virosis/inmunología , Animales , Línea Celular , Citocinas/biosíntesis , Humanos , Ratones , Receptores Toll-Like/fisiología
6.
Development ; 140(11): 2321-33, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23674601

RESUMEN

Efficient angiogenic sprouting is essential for embryonic, postnatal and tumor development. Serum response factor (SRF) is known to be important for embryonic vascular development. Here, we studied the effect of inducible endothelial-specific deletion of Srf in postnatal and adult mice. We find that endothelial SRF activity is vital for postnatal growth and survival, and is equally required for developmental and pathological angiogenesis, including during tumor growth. Our results demonstrate that SRF is selectively required for endothelial filopodia formation and cell contractility during sprouting angiogenesis, but seems dispensable for vascular remodeling. At the molecular level, we observe that vascular endothelial growth factor A induces nuclear accumulation of myocardin-related transcription factors (MRTFs) and regulates MRTF/SRF-dependent target genes including Myl9, which is important for endothelial cell migration in vitro. We conclude that SRF has a unique function in regulating migratory tip cell behavior during sprouting angiogenesis. We hypothesize that targeting the SRF pathway could provide an opportunity to selectively target tip cell filopodia-driven angiogenesis to restrict tumor growth.


Asunto(s)
Vasos Sanguíneos/embriología , Regulación del Desarrollo de la Expresión Génica , Neovascularización Patológica , Vasos Retinianos/embriología , Factor de Respuesta Sérica/fisiología , Actinas/metabolismo , Animales , Eliminación de Gen , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Miosinas/metabolismo , Trasplante de Neoplasias , Seudópodos/metabolismo , ARN Interferente Pequeño/metabolismo , Vasos Retinianos/patología , Factor de Respuesta Sérica/metabolismo
7.
Cancer Cell ; 11(4): 311-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17418408

RESUMEN

Germline mutations in the fumarate hydratase (FH) tumor suppressor gene predispose to leiomyomatosis, renal cysts, and renal cell cancer (HLRCC). HLRCC tumors overexpress HIF1alpha and hypoxia pathway genes. We conditionally inactivated mouse Fh1 in the kidney. Fh1 mutants developed multiple clonal renal cysts that overexpressed Hif1alpha and Hif2alpha. Hif targets, such as Glut1 and Vegf, were upregulated. We found that Fh1-deficient murine embryonic stem cells and renal carcinomas from HLRCC showed similar overexpression of HIF and hypoxia pathway components to the mouse cysts. Our data have shown in vivo that pseudohypoxic drive, resulting from HIF1alpha (and HIF2alpha) overexpression, is a direct consequence of Fh1 inactivation. Our mouse may be useful for testing therapeutic interventions that target angiogenesis and HIF-prolyl hydroxylation.


Asunto(s)
Carcinoma de Células Renales/etiología , Fumarato Hidratasa/genética , Silenciador del Gen/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Enfermedades Renales Quísticas/etiología , Neoplasias Renales/etiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Hipoxia de la Célula , Proliferación Celular , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Nature ; 458(7240): 899-903, 2009 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-19219027

RESUMEN

Injury or impaired clearance of apoptotic cells leads to the pathological accumulation of necrotic corpses, which induce an inflammatory response that initiates tissue repair. In addition, antigens present in necrotic cells can sometimes provoke a specific immune response and it has been argued that necrosis could explain adaptive immunity in seemingly infection-free situations, such as after allograft transplantation or in spontaneous and therapy-induced tumour rejection. In the mouse, the CD8alpha+ subset of dendritic cells phagocytoses dead cell remnants and cross-primes CD8+ T cells against cell-associated antigens. Here we show that CD8alpha+ dendritic cells use CLEC9A (also known as DNGR-1), a recently-characterized C-type lectin, to recognize a preformed signal that is exposed on necrotic cells. Loss or blockade of CLEC9A does not impair the uptake of necrotic cell material by CD8+ dendritic cells, but specifically reduces cross-presentation of dead-cell-associated antigens in vitro and decreases the immunogenicity of necrotic cells in vivo. The function of CLEC9A requires a key tyrosine residue in its intracellular tail that allows the recruitment and activation of the tyrosine kinase SYK, which is also essential for cross-presentation of dead-cell-associated antigens. Thus, CLEC9A functions as a SYK-coupled C-type lectin receptor to mediate sensing of necrosis by the principal dendritic-cell subset involved in regulating cross-priming to cell-associated antigens.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Necrosis/inmunología , Necrosis/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Mitogénicos/metabolismo , Animales , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Reactividad Cruzada/inmunología , Humanos , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Ligandos , Ratones , Fagocitosis , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Receptores Mitogénicos/genética , Transducción de Señal
9.
J Immunol ; 188(3): 1514-22, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22210910

RESUMEN

Autoimmune alopecia is characterized by an extensive epidermal T cell infiltrate that mediates hair follicle destruction. We have investigated the role of cell adhesion molecule 1 (Cadm1; Necl2) in this disease. Cadm1 is expressed by epidermal cells and mediates heterotypic adhesion to lymphocytes expressing class 1-restricted T cell-associated molecule (CRTAM). Using a murine autoimmune alopecia model, we observed an increase in early-activated cytotoxic (CD8-restricted, CRTAM-expressing) T cells, which preferentially associated with hair follicle keratinocytes expressing Cadm1. Coculture with Cadm1-transduced MHC-matched APCs stimulated alopecic lymph node cells to release IL-2 and IFN-γ. Overexpression of Cadm1 in cultured human keratinocytes did not promote cytokine secretion, but led to increased adhesion of alopecic cytotoxic T cells and enhanced T cell cytotoxicity in an MHC-independent manner. Epidermal overexpression of Cadm1 in transgenic mice led to increased autoimmune alopecia susceptibility relative to nontransgenic littermate controls. Our findings reveal that Cadm1 expression in the hair follicle plays a role in autoimmune alopecia.


Asunto(s)
Alopecia/inmunología , Moléculas de Adhesión Celular/fisiología , Citotoxicidad Inmunológica , Epidermis/inmunología , Inmunoglobulinas/fisiología , Linfocitos T/patología , Alopecia/etiología , Alopecia/patología , Animales , Enfermedades Autoinmunes , Adhesión Celular , Molécula 1 de Adhesión Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Epidermis/química , Folículo Piloso/inmunología , Folículo Piloso/patología , Humanos , Queratinocitos , Ratones , Ratones Transgénicos , Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología
10.
EMBO Rep ; 12(11): 1135-43, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21979816

RESUMEN

How individual components of the vascular basement membrane influence endothelial cell behaviour remains unclear. Here we show that laminin α4 (Lama4) regulates tip cell numbers and vascular density by inducing endothelial Dll4/Notch signalling in vivo. Lama4 deficiency leads to reduced Dll4 expression, excessive filopodia and tip cell formation in the mouse retina, phenocopying the effects of Dll4/Notch inhibition. Lama4-mediated Dll4 expression requires a combination of integrins in vitro and integrin ß1 in vivo. We conclude that appropriate laminin/integrin-induced signalling is necessary to induce physiologically functional levels of Dll4 expression and regulate branching frequency during sprouting angiogenesis in vivo.


Asunto(s)
Membrana Basal/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Membrana Basal/ultraestructura , Proteínas de Unión al Calcio , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Integrinas/metabolismo , Laminina/deficiencia , Laminina/metabolismo , Ratones , Neovascularización Fisiológica , Receptores Notch/antagonistas & inhibidores
11.
Proc Natl Acad Sci U S A ; 107(46): 19903-8, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21041641

RESUMEN

In mammalian epidermis, integrin expression is normally confined to the basal proliferative layer that contains stem cells. However, in epidermal hyperproliferative disorders and tumors, integrins are also expressed by suprabasal cells, with concomitant up-regulation of Erk mitogen-activated protein kinase (MAPK) signaling. In transgenic mice, expression of activated MAPK kinase 1 (MEK1) in the suprabasal, nondividing, differentiated cell layers (InvEE transgenics) results in epidermal hyperproliferation and skin inflammation. We now demonstrate that wounding induces benign tumors (papillomas and keratoacanthomas) in InvEE mice. By generating chimeras between InvEE mice and mice that lack the MEK1 transgene, we demonstrate that differentiating, nondividing cells that express MEK1 stimulate adjacent transgene-negative cells to divide and become incorporated into the tumor mass. Dexamethasone treatment inhibits tumor formation, suggesting that inflammation is involved. InvEE skin and tumors express high levels of IL1α; treatment with an IL1 receptor antagonist delays tumor onset and reduces incidence. Depletion of γδ T cells and macrophages also reduces tumor incidence. Because a hallmark of cancer is uncontrolled proliferation, it is widely assumed that tumors arise only from dividing cells. In contrast, our studies show that differentiated epidermal cells can initiate tumor formation without reacquiring the ability to divide and that they do so by triggering an inflammatory infiltrate.


Asunto(s)
División Celular , Epidermis/patología , Inflamación/patología , Neoplasias/patología , Lesiones Precancerosas/patología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Epidermis/efectos de los fármacos , Interleucina-1alfa/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Papiloma/metabolismo , Papiloma/patología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Transgenes , Cicatrización de Heridas/efectos de los fármacos
12.
Proc Natl Acad Sci U S A ; 106(23): 9286-91, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19478060

RESUMEN

Local tissue stem cells have been described in airways of the lung but their contribution to normal epithelial maintenance is currently unknown. We therefore developed aggregation chimera mice and a whole-lung imaging method to determine the relative contributions of progenitor (Clara) and bronchiolar stem cells to epithelial maintenance and repair. In normal and moderately injured airways chimeric patches were small in size and not associated with previously described stem cell niches. This finding suggested that single, randomly distributed progenitor cells maintain normal epithelial homeostasis. In contrast we found that repair following severe lung injury resulted in the generation of rare, large clonal cell patches that were associated with stem cell niches. This study provides evidence that epithelial stem cells are dispensable for normal airway homeostasis. We also demonstrate that stem cell activation and robust clonal cellular expansion occur only during repair from severe lung injury.


Asunto(s)
Bronquiolos/citología , Pulmón/citología , Células Madre/citología , Animales , Células Epiteliales/citología , Femenino , Homeostasis , Pulmón/fisiología , Masculino , Ratones , Células Madre/fisiología
13.
Cell Rep ; 38(4): 110227, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35081338

RESUMEN

In pancreatic ductal adenocarcinoma (PDAC), differentiation of pancreatic stellate cells (PSCs) into myofibroblast-like cancer-associated fibroblasts (CAFs) can both promote and suppress tumor progression. Here, we show that the Rho effector protein kinase N2 (PKN2) is critical for PSC myofibroblast differentiation. Loss of PKN2 is associated with reduced PSC proliferation, contractility, and alpha-smooth muscle actin (α-SMA) stress fibers. In spheroid co-cultures with PDAC cells, loss of PKN2 prevents PSC invasion but, counter-intuitively, promotes invasive cancer cell outgrowth. PKN2 deletion induces a myofibroblast to inflammatory CAF switch in the PSC matrisome signature both in vitro and in vivo. Further, deletion of PKN2 in the pancreatic stroma induces more locally invasive, orthotopic pancreatic tumors. Finally, we demonstrate that a PKN2KO matrisome signature predicts poor outcome in pancreatic and other solid human cancers. Our data indicate that suppressing PSC myofibroblast function can limit important stromal tumor-suppressive mechanisms, while promoting a switch to a cancer-supporting CAF phenotype.


Asunto(s)
Invasividad Neoplásica/patología , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Animales , Humanos , Ratones , Células Estrelladas Pancreáticas/metabolismo , Fenotipo , Proteína Quinasa C/metabolismo , Microambiente Tumoral/fisiología
14.
Elife ; 102021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34636321

RESUMEN

Lung squamous cell carcinoma (LSCC) is a considerable global health burden, with an incidence of over 600,000 cases per year. Treatment options are limited, and patient's 5-year survival rate is less than 5%. The ubiquitin-specific protease 28 (USP28) has been implicated in tumourigenesis through its stabilization of the oncoproteins c-MYC, c-JUN, and Δp63. Here, we show that genetic inactivation of Usp28-induced regression of established murine LSCC lung tumours. We developed a small molecule that inhibits USP28 activity in the low nanomole range. While displaying cross-reactivity against the closest homologue USP25, this inhibitor showed a high degree of selectivity over other deubiquitinases. USP28 inhibitor treatment resulted in a dramatic decrease in c-MYC, c-JUN, and Δp63 proteins levels and consequently induced substantial regression of autochthonous murine LSCC tumours and human LSCC xenografts, thereby phenocopying the effect observed by genetic deletion. Thus, USP28 may represent a promising therapeutic target for the treatment of squamous cell lung carcinoma.


Asunto(s)
Proteínas de Unión al ADN/genética , Eliminación de Gen , Neoplasias Pulmonares/genética , Neoplasias de Células Escamosas/genética , Factores de Transcripción/genética , Ubiquitina Tiolesterasa/genética , Animales , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Factores de Transcripción/metabolismo , Ubiquitina Tiolesterasa/metabolismo
15.
Cancer Cell ; 36(1): 68-83.e9, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31257073

RESUMEN

RAC1 P29 is the third most commonly mutated codon in human cutaneous melanoma, after BRAF V600 and NRAS Q61. Here, we study the role of RAC1P29S in melanoma development and reveal that RAC1P29S activates PAK, AKT, and a gene expression program initiated by the SRF/MRTF transcriptional pathway, which results in a melanocytic to mesenchymal phenotypic switch. Mice with ubiquitous expression of RAC1P29S from the endogenous locus develop lymphoma. When expressed only in melanocytes, RAC1P29S cooperates with oncogenic BRAF or with NF1-loss to promote tumorigenesis. RAC1P29S also drives resistance to BRAF inhibitors, which is reversed by SRF/MRTF inhibitors. These findings establish RAC1P29S as a promoter of melanoma initiation and mediator of therapy resistance, while identifying SRF/MRTF as a potential therapeutic target.


Asunto(s)
Transformación Celular Neoplásica/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Melanoma/etiología , Melanoma/patología , Mutación , Proteína de Unión al GTP rac1/genética , Alelos , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Masculino , Melanocitos/metabolismo , Melanoma/mortalidad , Melanoma/terapia , Ratones , Ratones Transgénicos , Modelos Biológicos , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Factor de Respuesta Sérica , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Mol Cell Biol ; 25(18): 8323-33, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16135819

RESUMEN

To elucidate the physiological role(s) of DUSP9 (dual-specificity phosphatase 9), also known as MKP-4 (mitogen-activated protein kinase [MAPK] phosphatase 4), the gene was deleted in mice. Crossing male chimeras with wild-type females resulted in heterozygous (DUSP9(+/-)) females. However, when these animals were crossed with wild-type (DUSP9(+/y)) males none of the progeny carried the targeted DUSP9 allele, indicating that both female heterozygous and male null (DUSP9(-/y)) animals die in utero. The DUSP9 gene is on the X chromosome, and this pattern of embryonic lethality is consistent with the selective inactivation of the paternal X chromosome in the extraembryonic tissues of the mouse, suggesting that DUSP9/MKP4 performs an essential function during placental development. Examination of embryos between 8 and 10.5 days postcoitum confirmed that lethality was due to a failure of labyrinth development, and this correlates exactly with the normal expression pattern of DUSP9/MKP-4 in the trophoblast giant cells and labyrinth of the placenta. Finally, when the placental defect was rescued, male null (DUSP9(-/y)) embryos developed to term, appeared normal, and were fertile. Our results indicate that DUSP9/MKP-4 is essential for placental organogenesis but is otherwise dispensable for mammalian embryonic development and highlights the critical role of dual-specificity MAPK phosphatases in the regulation of developmental outcomes in vertebrates.


Asunto(s)
Desarrollo Embrionario , Placenta/enzimología , Proteínas Tirosina Fosfatasas/fisiología , Alelos , Animales , Cromosomas , Fosfatasas de Especificidad Dual , Embrión de Mamíferos/citología , Femenino , Eliminación de Gen , Genes Letales , Masculino , Ratones , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Organogénesis/genética , Fosforilación , Placenta/embriología , Proteínas Tirosina Fosfatasas/genética
17.
Mol Cell Biol ; 24(15): 6719-27, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254239

RESUMEN

TREX1, originally designated DNase III, was isolated as a major nuclear DNA-specific 3'-->5' exonuclease that is widely distributed in both proliferating and nonproliferating mammalian tissues. The cognate cDNA shows homology to the editing subunit of the Escherichia coli replicative DNA polymerase III holoenzyme and encodes an exonuclease which was able to serve a DNA-editing function in vitro, promoting rejoining of a 3' mismatched residue in a reconstituted DNA base excision repair system. Here we report the generation of gene-targeted Trex1(-/-) mice. The null mice are viable and do not show the increase in spontaneous mutation frequency or cancer incidence that would be predicted if Trex1 served an obligatory role of editing mismatched 3' termini generated during DNA repair or DNA replication in vivo. Unexpectedly, Trex1(-/-) mice exhibit a dramatically reduced survival and develop inflammatory myocarditis leading to progressive, often dilated, cardiomyopathy and circulatory failure.


Asunto(s)
Exodesoxirribonucleasas/fisiología , Marcación de Gen , Miocarditis/genética , Fosfoproteínas/fisiología , Animales , Southern Blotting , División Celular , Supervivencia Celular , ADN/metabolismo , Reparación del ADN , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Exodesoxirribonucleasas/genética , Genotipo , Inflamación , Ratones , Ratones Transgénicos , Modelos Genéticos , Mutagénesis , Mutación , Miocardio/patología , Fosfoproteínas/genética , Estructura Terciaria de Proteína , Timo/patología , Factores de Tiempo
18.
Cell Rep ; 21(4): 966-978, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29069604

RESUMEN

The cell of origin of pancreatic ductal adenocarcinoma (PDAC) has been controversial. Here, we show that identical oncogenic drivers trigger PDAC originating from both ductal and acinar cells with similar histology but with distinct pathophysiology and marker expression dependent on cell of origin. Whereas acinar-derived tumors exhibited low AGR2 expression and were preceded by pancreatic intraepithelial neoplasias (PanINs), duct-derived tumors displayed high AGR2 and developed independently of a PanIN stage via non-mucinous lesions. Using orthotopic transplantation and chimera experiments, we demonstrate that PanIN-like lesions can be induced by PDAC as bystanders in adjacent healthy tissues, explaining the co-existence of mucinous and non-mucinous lesions and highlighting the need to distinguish between true precursor PanINs and PanIN-like bystander lesions. Our results suggest AGR2 as a tool to stratify PDAC according to cell of origin, highlight that not all PanIN-like lesions are precursors of PDAC, and add an alternative progression route to the current model of PDAC development.


Asunto(s)
Células Acinares/patología , Carcinogénesis , Carcinoma Ductal Pancreático/patología , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/patología , Células Acinares/metabolismo , Animales , Carcinoma Ductal Pancreático/metabolismo , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Mucoproteínas/genética , Mucoproteínas/metabolismo , Proteínas Oncogénicas , Conductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo
19.
Cell Rep ; 14(3): 440-448, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26774483

RESUMEN

In animals, the protein kinase C (PKC) family has expanded into diversely regulated subgroups, including the Rho family-responsive PKN kinases. Here, we describe knockouts of all three mouse PKN isoforms and reveal that PKN2 loss results in lethality at embryonic day 10 (E10), with associated cardiovascular and morphogenetic defects. The cardiovascular phenotype was not recapitulated by conditional deletion of PKN2 in endothelial cells or the developing heart. In contrast, inducible systemic deletion of PKN2 after E7 provoked collapse of the embryonic mesoderm. Furthermore, mouse embryonic fibroblasts, which arise from the embryonic mesoderm, depend on PKN2 for proliferation and motility. These cellular defects are reflected in vivo as dependence on PKN2 for mesoderm proliferation and neural crest migration. We conclude that failure of the mesoderm to expand in the absence of PKN2 compromises cardiovascular integrity and development, resulting in lethality.


Asunto(s)
Mesodermo/metabolismo , Proteína Quinasa C/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Genes Reporteros , Corazón/crecimiento & desarrollo , Mesodermo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Rastreo , Miocardio/metabolismo , Miocardio/patología , Proteína Quinasa C/deficiencia , Proteína Quinasa C/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología
20.
Mol Endocrinol ; 17(12): 2418-35, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14500757

RESUMEN

Recent work indicates that thyroid hormone receptor-associated protein 220 (TRAP220), a subunit of the multiprotein TRAP coactivator complex, is essential for embryonic survival. We have generated TRAP220 conditional null mice that are hypomorphic and express the gene at reduced levels. In contrast to TRAP220 null mice, which die at embryonic d 11.5 (E11.5), hypomorphic mice survive until E13.5. The reduced expression in hypomorphs results in hepatic necrosis, defects in hematopoiesis, and hypoplasia of the ventricular myocardium, similar to that observed in TRAP220 null embryos at an earlier stage. The embryonic lethality of null embryos at E11.5 is due to placental insufficiency. Tetraploid aggregation assays partially rescues embryonic development until E13.5, when embryonic loss occurs due to hepatic necrosis coupled with poor myocardial development as observed in hypomorphs. These findings demonstrate that, for normal placental function, there is an absolute requirement for TRAP220 in extraembryonic tissues at E11.5, with an additional requirement in embryonic tissues for hepatic and cardiovascular development thereafter.


Asunto(s)
Corazón/embriología , Hígado/embriología , Placenta/fisiología , Factores de Transcripción/fisiología , Animales , Femenino , Muerte Fetal , Corazón/fisiología , Heterocigoto , Homocigoto , Hibridación in Situ , Hígado/patología , Subunidad 1 del Complejo Mediador , Ratones , Ratones Noqueados , Miocardio/patología , Poliploidía , Embarazo , Receptores de Hormona Tiroidea/deficiencia , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA