Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 136(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37232246

RESUMEN

Endocytic recycling controls the return of internalised cargoes to the plasma membrane to coordinate their positioning, availability and downstream signalling. The Rab4 and Rab11 small GTPase families regulate distinct recycling routes, broadly classified as fast recycling from early endosomes (Rab4) and slow recycling from perinuclear recycling endosomes (Rab11), and both routes handle a broad range of overlapping cargoes to regulate cell behaviour. We adopted a proximity labelling approach, BioID, to identify and compare the protein complexes recruited by Rab4a, Rab11a and Rab25 (a Rab11 family member implicated in cancer aggressiveness), revealing statistically robust protein-protein interaction networks of both new and well-characterised cargoes and trafficking machinery in migratory cancer cells. Gene ontological analysis of these interconnected networks revealed that these endocytic recycling pathways are intrinsically connected to cell motility and cell adhesion. Using a knock-sideways relocalisation approach, we were further able to confirm novel links between Rab11, Rab25 and the ESCPE-1 and retromer multiprotein sorting complexes, and identify new endocytic recycling machinery associated with Rab4, Rab11 and Rab25 that regulates cancer cell migration in the 3D matrix.


Asunto(s)
Proteínas de Unión al GTP rab , Proteínas de Unión al GTP rab4 , Humanos , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab4/metabolismo , Transporte Biológico , Transporte de Proteínas/fisiología , Endosomas/metabolismo
2.
Cell ; 139(7): 1327-41, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20064378

RESUMEN

p53 is a tumor suppressor protein whose function is frequently lost in cancers through missense mutations within the Tp53 gene. This results in the expression of point-mutated p53 proteins that have both lost wild-type tumor suppressor activity and show gain of functions that contribute to transformation and metastasis. Here, we show that mutant p53 expression can promote invasion, loss of directionality of migration, and metastatic behavior. These activities of p53 reflect enhanced integrin and epidermal growth factor receptor (EGFR) trafficking, which depends on Rab-coupling protein (RCP) and results in constitutive activation of EGFR/integrin signaling. We provide evidence that mutant p53 promotes cell invasion via the inhibition of TAp63, and simultaneous loss of p53 and TAp63 recapitulates the phenotype of mutant p53 in cells. These findings open the possibility that blocking alpha5/beta1-integrin and/or the EGF receptor will have therapeutic benefit in mutant p53-expressing cancers.


Asunto(s)
Movimiento Celular , Integrina alfa5beta1/metabolismo , Metástasis de la Neoplasia , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Mutación , Seudópodos/metabolismo , Proteína p53 Supresora de Tumor/genética
3.
Am J Physiol Gastrointest Liver Physiol ; 323(3): G239-G254, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35819177

RESUMEN

The small GTPase, Rab11a, regulates vesicle trafficking and cell polarity in epithelial cells through interaction with Rab11 family-interacting proteins (Rab11-FIPs). We hypothesized that deficiency of Rab11-FIP1 would affect mucosal integrity in the intestine. Global Rab11FIP1 knockout (KO) mice were generated by deletion of the second exon. Pathology of intestinal tissues was analyzed by immunostaining of colonic sections and RNA-sequencing of isolated colonic epithelial cells. A low concentration of dextran sodium sulfate (DSS, 2%) was added to drinking water for 5 days, and injury score was compared between Rab11FIP1 KO, Rab11FIP2 KO, and heterozygous littermates. Rab11FIP1 KO mice showed normal fertility and body weight gain. More frequent lymphoid patches and infiltration of macrophages and neutrophils were identified in Rab11FIP1 KO mice before the development of rectal prolapse compared with control mice. The population of trefoil factor 3 (TFF3)-positive goblet cells was significantly lower, and the ratio of proliferative to nonproliferative cells was higher in Rab11FIP1 KO colons. Transcription signatures indicated that Rab11FIP1 deletion downregulated genes that mediate stress tolerance response, whereas genes mediating the response to infection were significantly upregulated, consistent with the inflammatory responses in the steady state. Lack of Rab11FIP1 also resulted in abnormal accumulation of subapical vesicles in colonocytes and the internalization of transmembrane mucin, MUC13, with Rab14. After DSS treatment, Rab11FIP1 KO mice showed greater body weight loss and more severe mucosal damage than those in heterozygous littermates. These findings suggest that Rab11FIP1 is important for cytoprotection mechanisms and for the maintenance of colonic mucosal integrity.NEW & NOTEWORTHY Although Rab11FIP1 is important in membrane trafficking in epithelial cells, the gastrointestinal phenotype of Rab11FIP1 knockout (KO) mice had never been reported. This study demonstrated that Rab11FIP1 loss induces mistrafficking of Rab14 and MUC13 and decreases in colonic goblet cells, resulting in impaired mucosal integrity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Colitis , Proteínas de la Membrana , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Colitis/metabolismo , Colon/metabolismo , Sulfato de Dextran , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/genética , Ratones Noqueados
4.
Nat Rev Mol Cell Biol ; 10(12): 843-53, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19904298

RESUMEN

Since it has become clear that adhesion receptors are trafficked through the endosomal pathway and that this can influence their function, much effort has been invested in obtaining detailed descriptions of the molecular machinery responsible for internalizing and recycling integrins. New findings indicate that integrin trafficking dictates the nature of Rho GTPase signalling during cytokinesis and cell migration. Furthermore, integrins can exert control over the trafficking of other receptors in a way that drives cancer cell invasion and tumour angiogenesis.


Asunto(s)
Endocitosis , Integrinas/metabolismo , Animales , Matriz Extracelular/metabolismo , Humanos , Transporte de Proteínas , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
5.
Nucleic Acids Res ; 47(9): 4375-4392, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30927008

RESUMEN

Antisense oligonucleotides (ASOs) modulate cellular target gene expression through direct binding to complementary RNA. Advances in ASO chemistry have led to the development of phosphorothioate (PS) ASOs with constrained-ethyl modifications (cEt). These next-generation cEt-ASOs can enter cells without transfection reagents. Factors involved in intracellular uptake and trafficking of cEt-ASOs leading to successful target knockdown are highly complex and not yet fully understood. AZD4785 is a potent and selective therapeutic KRAS cEt-ASO currently under clinical development for the treatment of cancer. Therefore, we used this to investigate mechanisms of cEt-ASO trafficking across a panel of cancer cells. We found that the extent of ASO-mediated KRAS mRNA knockdown varied significantly between cells and that this did not correlate with bulk levels of intracellular accumulation. We showed that in cells with good productive uptake, distribution of ASO was perinuclear and in those with poor productive uptake distribution was peripheral. Furthermore, ASO rapidly trafficked to the late endosome/lysosome in poor productive uptake cells compared to those with more robust knockdown. An siRNA screen identified several factors mechanistically involved in productive ASO uptake, including the endosomal GTPase Rab5C. This work provides novel insights into the trafficking of cEt-ASOs and mechanisms that may determine their cellular fate.


Asunto(s)
Neoplasias/genética , Oligonucleótidos Antisentido/genética , Oligonucleótidos Fosforotioatos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al GTP rab5/genética , Endosomas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Neoplasias/patología , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Fosforotioatos/química , Oligonucleótidos Fosforotioatos/farmacología , ARN Mensajero/genética , ARN Interferente Pequeño/genética
6.
J Cell Sci ; 130(4): 697-711, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28062852

RESUMEN

We have previously shown that Rab17, a small GTPase associated with epithelial polarity, is specifically suppressed by ERK2 (also known as MAPK1) signalling to promote an invasive phenotype. However, the mechanisms through which Rab17 loss permits invasiveness, and the endosomal cargoes that are responsible for mediating this, are unknown. Using quantitative mass spectrometry-based proteomics, we have found that knockdown of Rab17 leads to a highly selective reduction in the cellular levels of a v-SNARE (Vamp8). Moreover, proteomics and immunofluorescence indicate that Vamp8 is associated with Rab17 at late endosomes. Reduced levels of Vamp8 promote transition between ductal carcinoma in situ (DCIS) and a more invasive phenotype. We developed an unbiased proteomic approach to elucidate the complement of receptors that redistributes between endosomes and the plasma membrane, and have pin-pointed neuropilin-2 (NRP2) as a key pro-invasive cargo of Rab17- and Vamp8-regulated trafficking. Indeed, reduced Rab17 or Vamp8 levels lead to increased mobilisation of NRP2-containing late endosomes and upregulated cell surface expression of NRP2. Finally, we show that NRP2 is required for the basement membrane disruption that accompanies the transition between DCIS and a more invasive phenotype.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Endosomas/metabolismo , Proteómica/métodos , Aminoácidos/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Membranas Intracelulares/metabolismo , Marcaje Isotópico , Modelos Biológicos , Clasificación del Tumor , Invasividad Neoplásica , Neuropilina-2/metabolismo , Unión Proteica , Transporte de Proteínas , Proteínas R-SNARE/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/metabolismo , Proteínas SNARE/metabolismo , Análisis de Supervivencia , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
7.
PLoS Genet ; 10(3): e1004262, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24676055

RESUMEN

Receptor Tyrosine Kinases (RTKs) and Focal Adhesion Kinase (FAK) regulate multiple signalling pathways, including mitogen-activated protein (MAP) kinase pathway. FAK interacts with several RTKs but little is known about how FAK regulates their downstream signalling. Here we investigated how FAK regulates signalling resulting from the overexpression of the RTKs RET and EGFR. FAK suppressed RTKs signalling in Drosophila melanogaster epithelia by impairing MAPK pathway. This regulation was also observed in MDA-MB-231 human breast cancer cells, suggesting it is a conserved phenomenon in humans. Mechanistically, FAK reduced receptor recycling into the plasma membrane, which resulted in lower MAPK activation. Conversely, increasing the membrane pool of the receptor increased MAPK pathway signalling. FAK is widely considered as a therapeutic target in cancer biology; however, it also has tumour suppressor properties in some contexts. Therefore, the FAK-mediated negative regulation of RTK/MAPK signalling described here may have potential implications in the designing of therapy strategies for RTK-driven tumours.


Asunto(s)
Neoplasias de la Mama/genética , Quinasa 1 de Adhesión Focal/genética , Sistema de Señalización de MAP Quinasas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Drosophila melanogaster/genética , Células Epiteliales/metabolismo , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Fosforilación , Proteínas Tirosina Quinasas Receptoras/metabolismo
8.
Traffic ; 15(6): 665-83, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24589086

RESUMEN

Fibroblast growth factor receptor 4 (FGFR4) plays important roles during development and in the adult to maintain tissue homeostasis. Moreover, overexpression of FGFR4 or activating mutations in FGFR4 has been identified as tumour-promoting events in several forms of cancer. Endocytosis is important for regulation of signalling receptors and we have previously shown that FGFR4 is mainly localized to transferrin-positive structures after ligand-induced endocytosis. Here, using a cell line with a defined pericentriolar endocytic recycling compartment, we show that FGFR4 accumulates in this compartment after endocytosis. Furthermore, using classical recycling assays and a new, photoactivatable FGFR4-PA-GFP fusion protein combined with live-cell imaging, we demonstrate that recycling of FGFR4 is dependent on Rab11. Upon Rab11b depletion, FGFR4 is trapped in the pericentriolar recycling compartment and the total levels of FGFR4 in cells are increased. Moreover, fibroblast growth factor 1 (FGF1)-induced autophosphorylation of FGFR4 as well as phosphorylation of phospholipase C (PLC)-γ is prolonged in cells depleted of Rab11. Interestingly, the activation of mitogen-activated protein kinase and AKT pathways were not prolonged but rather reduced in Rab11-depleted cells, indicating that recycling of FGFR4 is important for the nature of its signalling output. Thus, Rab11-dependent recycling of FGFR4 maintains proper levels of FGFR4 in cells and regulates FGF1-induced FGFR4 signalling.


Asunto(s)
Endocitosis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Línea Celular Tumoral , Endosomas/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Humanos , Fosfolipasa C gamma/metabolismo , Transporte de Proteínas , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal , Proteínas de Unión al GTP rab/genética
9.
J Cell Sci ; 127(Pt 18): 3893-901, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015290

RESUMEN

Chloride intracellular channel 3 (CLIC3) drives invasiveness of pancreatic and ovarian cancer by acting in concert with Rab25 to regulate the recycling of α5ß1 integrin from late endosomes to the plasma membrane. Here, we show that in two estrogen receptor (ER)-negative breast cancer cell lines, CLIC3 has little influence on integrin recycling, but controls trafficking of the pro-invasive matrix metalloproteinase MT1-MMP (also known as MMP14). In MDA-MB-231 cells, MT1-MMP and CLIC3 are localized primarily to late endosomal/lysosomal compartments located above the plane of adhesion and near the nucleus. MT1-MMP is transferred from these late endosomes to sites of cell-matrix adhesion in a CLIC3-dependent fashion. Correspondingly, CLIC3-knockdown opposes MT1-MMP-dependent invasive processes. These include the disruption of the basement membrane as acini formed from MCF10DCIS.com cells acquire invasive characteristics in 3D culture, and the invasion of MDA-MB-231 cells into Matrigel or organotypic plugs of type I collagen. Consistent with this, expression of CLIC3 predicts poor prognosis in ER-negative breast cancer. The identification of MT1-MMP as a cargo of a CLIC3-regulated pathway that drives invasion highlights the importance of late endosomal sorting and trafficking in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Canales de Cloruro/metabolismo , Endosomas/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Movimiento Celular , Canales de Cloruro/genética , Femenino , Humanos , Metaloproteinasa 14 de la Matriz/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Transporte de Proteínas
10.
J Biol Chem ; 289(1): 122-32, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24220032

RESUMEN

The control and processing of microRNAs (miRs) is critical in the regulation of all cellular responses. Previous studies have suggested that a reduction in the expression of certain miRs, or an overall decrease in miR processing through the partial depletion of Dicer, can promote enhanced metastatic potential. We show here that Dicer depletion can promote the invasive behavior of cells that is reflected in enhanced recycling and activation of the growth factor receptors Met and EGF receptor. These responses are also seen in response to the expression of tumor-derived mutant p53s, and we show that mutant p53 can down-regulate Dicer expression through both direct inhibition of the TAp63-mediated transcriptional activation of Dicer and a TAp63-independent control of Dicer protein expression. Our results delineate a clear relationship between mutant p53, TAp63, and Dicer that might contribute to the metastatic function of mutant p53 but, interestingly, also reveal TAp63-independent functions of mutant p53 in controlling Dicer activity.


Asunto(s)
ARN Helicasas DEAD-box/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias/metabolismo , Ribonucleasa III/biosíntesis , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular Tumoral , ARN Helicasas DEAD-box/genética , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Ribonucleasa III/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética
11.
Nat Cell Biol ; 10(9): 1017-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18758489

RESUMEN

Talin can activate integrins to bind the extracellular matrix and also connect matrix-engaged integrins to the actin cytoskeleton. New work shows that cell spreading can be dissected into three distinct phases according to their differential requirements for talin function.


Asunto(s)
Movimiento Celular , Tamaño de la Célula , Talina/deficiencia , Animales , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibronectinas/farmacología , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/enzimología , Humanos , Integrina beta1/metabolismo , Ratones
12.
Bioessays ; 35(6): 523-32, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23605698

RESUMEN

Recently it has become clear that trafficking of integrins to late endosomes is key to the regulation of integrin expression and function during cell migration. Here we discuss the molecular machinery that dictates whether integrins are sorted to recycling endosomes or are targeted to late endosomes and lysosomes. Integrins and other receptors that are sorted to late endosomes are not necessarily degraded and, under certain circumstances, can be spared destruction and returned to the cell surface to drive cell migration and invasion. We will discuss how the exchange of adhesion receptors and other key regulators of cell migration between late endosomes/lysosomes and the plasma membrane can promote dynamic turnover of adhesions during cell migration.


Asunto(s)
Movimiento Celular , Endosomas/metabolismo , Integrinas/fisiología , Lisosomas/metabolismo , Neoplasias/patología , Animales , Humanos , Invasividad Neoplásica , Transporte de Proteínas
13.
J Cell Sci ; 125(Pt 6): 1465-77, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22328529

RESUMEN

Upregulation of the extracellular signal-regulated kinase (ERK) pathway has been shown to contribute to tumour invasion and progression. Because the two predominant ERK isoforms (ERK1 and ERK2, also known as MAPK3 and MAPK1, respectively) are highly homologous and have indistinguishable kinase activities in vitro, both enzymes were believed to be redundant and interchangeable. To challenge this view, we show that ERK2 silencing inhibits invasive migration of MDA-MB-231 cells, and re-expression of ERK2 but not ERK1 restores the normal invasive phenotype. A detailed quantitative analysis of cell movement on 3D matrices indicates that ERK2 knockdown impairs cellular motility by decreasing the migration velocity as well as increasing the time that cells spend not moving. Using gene expression arrays we found that the expression of the genes for Rab17 and liprin-ß2 was increased by knockdown of ERK2 and restored to normal levels following re-expression of ERK2, but not ERK1. Both play inhibitory roles in the invasive behaviour of three independent cancer cell lines. Importantly, knockdown of either Rab17 or liprin-ß2 restores invasiveness of ERK2-depleted cells, indicating that ERK2 drives invasion of MDA-MB-231 cells by suppressing expression of these genes.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Portadoras/antagonistas & inhibidores , Movimiento Celular/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Microambiente Tumoral/fisiología , Proteínas de Unión al GTP rab/antagonistas & inhibidores , Neoplasias de la Mama/genética , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas de Unión al GTP rab/biosíntesis , Proteínas de Unión al GTP rab/genética
14.
Cancer Res Commun ; 4(2): 588-606, 2024 02 29.
Artículo en Inglés | MEDLINE | ID: mdl-38358352

RESUMEN

Neutrophils are a highly heterogeneous cellular population. However, a thorough examination of the different transcriptional neutrophil states between health and malignancy has not been performed. We utilized single-cell RNA sequencing of human and murine datasets, both publicly available and independently generated, to identify neutrophil transcriptomic subtypes and developmental lineages in health and malignancy. Datasets of lung, breast, and colorectal cancer were integrated to establish and validate neutrophil gene signatures. Pseudotime analysis was used to identify genes driving neutrophil development from health to cancer. Finally, ligand-receptor interactions and signaling pathways between neutrophils and other immune cell populations in primary colorectal cancer and metastatic colorectal cancer were investigated. We define two main neutrophil subtypes in primary tumors: an activated subtype sharing the transcriptomic signatures of healthy neutrophils; and a tumor-specific subtype. This signature is conserved in murine and human cancer, across different tumor types. In colorectal cancer metastases, neutrophils are more heterogeneous, exhibiting additional transcriptomic subtypes. Pseudotime analysis implicates IL1ß/CXCL8/CXCR2 axis in the progression of neutrophils from health to cancer and metastasis, with effects on T-cell effector function. Functional analysis of neutrophil-tumoroid cocultures and T-cell proliferation assays using orthotopic metastatic mouse models lacking Cxcr2 in neutrophils support our transcriptional analysis. We propose that the emergence of metastatic-specific neutrophil subtypes is driven by the IL1ß/CXCL8/CXCR2 axis, with the evolution of different transcriptomic signals that impair T-cell function at the metastatic site. Thus, a better understanding of neutrophil transcriptomic programming could optimize immunotherapeutic interventions into early and late interventions, targeting different neutrophil states. SIGNIFICANCE: We identify two recurring neutrophil populations and demonstrate their staged evolution from health to malignancy through the IL1ß/CXCL8/CXCR2 axis, allowing for immunotherapeutic neutrophil-targeting approaches to counteract immunosuppressive subtypes that emerge in metastasis.


Asunto(s)
Neoplasias Colorrectales , Neutrófilos , Animales , Ratones , Humanos , Recurrencia Local de Neoplasia/metabolismo , Transducción de Señal/genética , Neoplasias Colorrectales/genética , Análisis de la Célula Individual
15.
Neuro Oncol ; 26(4): 625-639, 2024 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37936324

RESUMEN

BACKGROUND: Glioblastomas have highly infiltrative growth patterns that contribute to recurrence and poor survival. Despite infiltration being a critical therapeutic target, no clinically useful therapies exist that counter glioblastoma invasion. Here, we report that inhibition of ataxia telangiectasia and Rad 3 related kinase (ATR) reduces invasion of glioblastoma cells through dysregulation of cytoskeletal networks and subsequent integrin trafficking. METHODS: Glioblastoma motility and invasion were assessed in vitro and in vivo in response to ATR inhibition (ATRi) and ATR overexpression using time-lapse microscopy, two orthotopic glioblastoma models, and intravital imaging. Disruption to cytoskeleton networks and endocytic processing were investigated via high-throughput, super-resolution and intravital imaging. RESULTS: High ATR expression was associated with significantly poorer survival in clinical datasets while histological, protein expression, and spatial transcriptomics using glioblastoma tumor specimens revealed higher ATR expression at infiltrative margins. Pharmacological inhibition with two different compounds and RNAi targeting of ATR opposed the invasion of glioblastoma, whereas overexpression of ATR drove migration. Subsequent investigation revealed that cytoskeletal dysregulation reduced macropinocytotic internalization of integrins at growth-cone-like structures, resulting in a tumor microtube retraction defect. The biological relevance and translational potential of these findings were confirmed using two orthotopic in vivo models of glioblastoma and intravital imaging. CONCLUSIONS: We demonstrate a novel role for ATR in determining invasion in glioblastoma cells and propose that pharmacological targeting of ATR could have far-reaching clinical benefits beyond radiosensitization.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/patología , Integrinas/metabolismo , Línea Celular Tumoral , Citoesqueleto/metabolismo , Citoesqueleto/patología , Invasividad Neoplásica , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo
16.
J Neurosci ; 32(30): 10352-64, 2012 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-22836268

RESUMEN

Integrins are involved in axon growth and regeneration. Manipulation of integrins is a route to promoting axon regeneration and understanding regeneration failure in the CNS. Expression of α9 integrin promotes axon regeneration, so we have investigated α9ß1 trafficking and transport in axons and at the growth cone. We have previously found that α9 and ß1 integrins traffic via Rab11-positive recycling endosomes in peripheral axons and growth cones. However, transport via Rab11 is slow, while rapid transport occurs in vesicles lacking Rab11. We have further studied α9 and ß1 integrin transport and traffic in adult rat dorsal root ganglion axons and PC12 cells. Integrins are in ARF6 vesicles during rapid axonal transport and during trafficking in the growth cone. We report that rapid axonal transport of these integrins and their trafficking at the cell surface is regulated by ARF6. ARF6 inactivation by expression of ACAP1 leads to increased recycling of ß1 integrins to the neuronal surface and to increased anterograde axonal transport. ARF6 activation by expression of the neuronal guanine nucleotide exchange factors, ARNO or EFA6, increases retrograde integrin transport in axons and increases integrin internalization. ARF6 inactivation increases integrin-mediated outgrowth, while activation decreases it. The coordinated changes in integrin transport and recycling resulting from ARF6 activation or inactivation are the probable mechanism behind this regulation of axon growth. Our data suggest a novel mechanism of integrin traffic and transport in peripheral axons, regulated by the activation state of ARF6, and suggest that ARF6 might be targeted to enhance integrin-dependent axon regeneration after injury.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Transporte Axonal/fisiología , Ganglios Espinales/metabolismo , Integrinas/metabolismo , Neuronas/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Animales , Axones/metabolismo , Células Cultivadas , Endocitosis/fisiología , Ganglios Espinales/citología , Conos de Crecimiento/metabolismo , Masculino , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo
17.
J Cell Sci ; 124(Pt 22): 3753-9, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22114305

RESUMEN

The actin cytoskeleton provides scaffolding and physical force to effect fundamental processes such as motility, cytokinesis and vesicle trafficking. The Arp2/3 complex nucleates actin structures and contributes to endocytic vesicle invagination and trafficking away from the plasma membrane. Internalisation and directed recycling of integrins are major driving forces for invasive cell motility and potentially for cancer metastasis. Here, we describe a direct requirement for WASH and Arp2/3-mediated actin polymerisation on the endosomal membrane system for α5ß1 integrin recycling. WASH regulates the trafficking of endosomal α5ß1 integrin to the plasma membrane and is fundamental for integrin-driven cell morphology changes and integrin-mediated cancer cell invasion. Thus, we implicate WASH and Arp2/3-driven actin nucleation in receptor recycling leading to invasive motility.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Movimiento Celular , Integrina alfa5beta1/metabolismo , Neoplasias/fisiopatología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/genética , Actinas/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Endosomas/genética , Endosomas/metabolismo , Humanos , Integrina alfa5beta1/genética , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Transporte de Proteínas , Proteína del Síndrome de Wiskott-Aldrich/genética
18.
Curr Opin Cell Biol ; 18(5): 549-57, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16904305

RESUMEN

The past five years have seen a steady accumulation of data reinforcing the view that Rab-regulated recycling pathways contribute to cell migration. In particular, detailed descriptions have emerged of the mechanisms that recruit integrins and growth factor receptors to Rab4- and Rab11-driven pathways. Recent work provides new insight into the importance of particular recycling events in cell migration within a variety of physiological contexts.


Asunto(s)
Membrana Celular/metabolismo , Movimiento Celular/fisiología , Endocitosis/fisiología , Transducción de Señal/fisiología , Factores de Ribosilacion-ADP/metabolismo , Animales , Exocitosis/fisiología , Integrinas/metabolismo , Receptores de Quimiocina/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Proteínas de Unión al GTP rab/metabolismo
19.
J Am Soc Nephrol ; 23(9): 1506-17, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22859853

RESUMEN

The aquaporin 2 (AQP2) water channel, expressed in kidney collecting ducts, contributes critically to water homeostasis in mammals. Animals lacking or having significantly reduced levels of AQP2, however, have not only urinary concentrating abnormalities but also renal tubular defects that lead to neonatal mortality from renal failure. Here, we show that AQP2 is not only a water channel but also an integrin-binding membrane protein that promotes cell migration and epithelial morphogenesis. AQP2 expression modulates the trafficking and internalization of integrin ß1, facilitating its turnover at focal adhesions. In vitro, disturbing the interaction between AQP2 and integrin ß1 by mutating the RGD motif led to reduced endocytosis, retention of integrin ß1 at the cell surface, and defective cell migration and tubulogenesis. Similarly, in vivo, AQP2-null mice exhibited significant retention of integrin ß1 at the basolateral membrane and had tubular abnormalities. In summary, these data suggest that the water channel AQP2 interacts with integrins to promote renal epithelial cell migration, contributing to the structural and functional integrity of the mammalian kidney.


Asunto(s)
Acuaporina 2/fisiología , Movimiento Celular/fisiología , Células Epiteliales/citología , Riñón/citología , Morfogénesis/fisiología , Animales , Acuaporina 2/deficiencia , Acuaporina 2/genética , Línea Celular , Permeabilidad de la Membrana Celular/fisiología , Perros , Endocitosis/fisiología , Células Epiteliales/fisiología , Técnicas In Vitro , Integrina beta1/fisiología , Riñón/crecimiento & desarrollo , Riñón/fisiología , Ratones , Ratones Noqueados , Modelos Animales , Mutación/genética , Oligopéptidos/genética , Oligopéptidos/fisiología , Porcinos , Transfección
20.
Neurooncol Adv ; 5(1): vdad067, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37334166

RESUMEN

Background: Infiltration of glioblastoma (GBM) throughout the brain leads to its inevitable recurrence following standard-of-care treatments, such as surgical resection, chemo-, and radiotherapy. A deeper understanding of the mechanisms invoked by GBM to infiltrate the brain is needed to develop approaches to contain the disease and reduce recurrence. The aim of this study was to discover mechanisms through which extracellular vesicles (EVs) released by GBM influence the brain microenvironment to facilitate infiltration, and to determine how altered extracellular matrix (ECM) deposition by glial cells might contribute to this. Methods: CRISPR was used to delete genes, previously established to drive carcinoma invasiveness and EV production, from patient-derived primary and GBM cell lines. We purified and characterized EVs released by these cells, assessed their capacity to foster pro-migratory microenvironments in mouse brain slices, and evaluated the contribution made by astrocyte-derived ECM to this. Finally, we determined how CRISPR-mediated deletion of genes, which we had found to control EV-mediated communication between GBM cells and astrocytes, influenced GBM infiltration when orthotopically injected into CD1-nude mice. Results: GBM cells expressing a p53 mutant (p53R273H) with established pro-invasive gain-of-function release EVs containing a sialomucin, podocalyxin (PODXL), which encourages astrocytes to deposit ECM with increased levels of hyaluronic acid (HA). This HA-rich ECM, in turn, promotes migration of GBM cells. Consistently, CRISPR-mediated deletion of PODXL opposes infiltration of GBM in vivo. Conclusions: This work describes several key components of an EV-mediated mechanism though which GBM cells educate astrocytes to support infiltration of the surrounding healthy brain tissue.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA