Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
Cell Death Differ ; 29(10): 2089-2104, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35473984

RESUMEN

Glioblastoma (GBM) is the most prevalent malignant primary brain tumour in adults. GBM typically has a poor prognosis, mainly due to a lack of effective treatment options leading to tumour persistence or recurrence. We investigated the therapeutic potential of targeting anti-apoptotic BCL-2 proteins in GBM. Levels of anti-apoptotic BCL-xL and MCL-1 were consistently increased in GBM compared with non-malignant cells and tissue. Moreover, we found that relative to their differentiated counterparts, patient-derived GBM stem-like cells also displayed higher expression of anti-apoptotic BCL-2 family members. High anti-apoptotic BCL-xL and MCL-1 expression correlated with heightened susceptibility of GBM to BCL-2 family protein-targeting BH3-mimetics. This is indicative of increased apoptotic priming. Indeed, GBM displayed an obligate requirement for MCL-1 expression in both tumour development and maintenance. Investigating this apoptotic sensitivity, we found that sequential inhibition of BCL-xL and MCL-1 led to robust anti-tumour responses in vivo, in the absence of overt toxicity. These data demonstrate that BCL-xL and MCL-1 pro-survival function is a fundamental prerequisite for GBM survival that can be therapeutically exploited by BH3-mimetics.


Asunto(s)
Glioblastoma , Adulto , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X
4.
RSC Med Chem ; 13(2): 150-155, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35308027

RESUMEN

Rab27A is a small GTPase, which mediates transport and docking of secretory vesicles at the plasma membrane via protein-protein interactions (PPIs) with effector proteins. Rab27A promotes the growth and invasion of multiple cancer types such as breast, lung and pancreatic, by enhancing secretion of chemokines, metalloproteases and exosomes. The significant role of Rab27A in multiple cancer types and the minor role in adults suggest that Rab27A may be a suitable target to disrupt cancer metastasis. Similar to many GTPases, the flat topology of the Rab27A-effector PPI interface and the high affinity for GTP make it a challenging target for inhibition by small molecules. Reported co-crystal structures show that several effectors of Rab27A interact with the Rab27A SF4 pocket ('WF-binding pocket') via a conserved tryptophan-phenylalanine (WF) dipeptide motif. To obtain structural insight into the ligandability of this pocket, a novel construct was designed fusing Rab27A to part of an effector protein (fRab27A), allowing crystallisation of Rab27A in high throughput. The paradigm of KRas covalent inhibitor development highlights the challenge presented by GTPase proteins as targets. However, taking advantage of two cysteine residues, C123 and C188, that flank the WF pocket and are unique to Rab27A and Rab27B among the >60 Rab family proteins, we used the quantitative Irreversible Tethering (qIT) assay to identify the first covalent ligands for native Rab27A. The binding modes of two hits were elucidated by co-crystallisation with fRab27A, exemplifying a platform for identifying suitable lead fragments for future development of competitive inhibitors of the Rab27A-effector interaction interface, corroborating the use of covalent libraries to tackle challenging targets.

5.
Nat Commun ; 12(1): 6829, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819513

RESUMEN

Signals are relayed from receptor tyrosine kinases (RTKs) at the cell surface to effector systems in the cytoplasm and nucleus, and coordination of this process is important for the execution of migratory phenotypes, such as cell scattering and invasion. The endosomal system influences how RTK signalling is coded, but the ways in which it transmits these signals to the nucleus to influence gene expression are not yet clear. Here we show that hepatocyte growth factor, an activator of MET (an RTK), promotes Rab17- and clathrin-dependent endocytosis of EphA2, another RTK, followed by centripetal transport of EphA2-positive endosomes. EphA2 then mediates physical capture of endosomes on the outer surface of the nucleus; a process involving interaction between the nuclear import machinery and a nuclear localisation sequence in EphA2's cytodomain. Nuclear capture of EphA2 promotes RhoG-dependent phosphorylation of the actin-binding protein, cofilin to oppose nuclear import of G-actin. The resulting depletion of nuclear G-actin drives transcription of Myocardin-related transcription factor (MRTF)/serum-response factor (SRF)-target genes to implement cell scattering and the invasive behaviour of cancer cells.


Asunto(s)
Núcleo Celular/metabolismo , Endosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias/patología , Factores Complejos Ternarios/metabolismo , Actinas/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Línea Celular Tumoral , Citoplasma/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo
6.
J Cell Biol ; 220(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34623384

RESUMEN

The cystine-glutamate antiporter, xCT, supports a glutathione synthesis program enabling cancer cells to cope with metabolically stressful microenvironments. Up-regulated xCT, in combination with glutaminolysis, leads to increased extracellular glutamate, which promotes invasive behavior by activating metabotropic glutamate receptor 3 (mGluR3). Here we show that activation of mGluR3 in breast cancer cells activates Rab27-dependent release of extracellular vesicles (EVs), which can transfer invasive characteristics to "recipient" tumor cells. These EVs contain mitochondrial DNA (mtDNA), which is packaged via a PINK1-dependent mechanism. We highlight mtDNA as a key EV cargo necessary and sufficient for intercellular transfer of invasive behavior by activating Toll-like receptor 9 in recipient cells, and this involves increased endosomal trafficking of pro-invasive receptors. We propose that an EV-mediated mechanism, through which altered cellular metabolism in one cell influences endosomal trafficking in other cells, is key to generation and dissemination of pro-invasive microenvironments during mammary carcinoma progression.


Asunto(s)
ADN Mitocondrial/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas Quinasas/metabolismo , Línea Celular Tumoral , Cisplatino/farmacología , Empaquetamiento del ADN/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/ultraestructura , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Invasividad Neoplásica , Receptores de Glutamato Metabotrópico/metabolismo , Tetraspanina 30/metabolismo , Receptor Toll-Like 9/metabolismo , Proteínas rab27 de Unión a GTP/metabolismo
7.
Elife ; 102021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34096503

RESUMEN

RAS-like (RAL) GTPases function in Wnt signalling-dependent intestinal stem cell proliferation and regeneration. Whether RAL proteins work as canonical RAS effectors in the intestine and the mechanisms of how they contribute to tumourigenesis remain unclear. Here, we show that RAL GTPases are necessary and sufficient to activate EGFR/MAPK signalling in the intestine, via induction of EGFR internalisation. Knocking down Drosophila RalA from intestinal stem and progenitor cells leads to increased levels of plasma membrane-associated EGFR and decreased MAPK pathway activation. Importantly, in addition to influencing stem cell proliferation during damage-induced intestinal regeneration, this role of RAL GTPases impacts on EGFR-dependent tumourigenic growth in the intestine and in human mammary epithelium. However, the effect of oncogenic RAS in the intestine is independent from RAL function. Altogether, our results reveal previously unrecognised cellular and molecular contexts where RAL GTPases become essential mediators of adult tissue homeostasis and malignant transformation.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Receptores ErbB/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Receptores de Péptidos de Invertebrados/metabolismo , Células Madre/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Animales , Animales Modificados Genéticamente , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Endocitosis , Receptores ErbB/genética , Femenino , Humanos , Hiperplasia , Mucosa Intestinal/patología , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Glándulas Mamarias Humanas/enzimología , Glándulas Mamarias Humanas/patología , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Receptores de Péptidos de Invertebrados/genética , Transducción de Señal , Células Madre/patología , Proteínas de Unión al GTP ral/genética
8.
Cell Death Dis ; 12(2): 207, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627632

RESUMEN

TP53 is the most frequently mutated gene in cancers. Mutations lead to loss of p53 expression or expression of a mutant protein. Mutant p53 proteins commonly lose wild-type function, but can also acquire novel functions in promoting metastasis and chemoresistance. Previously, we uncovered a role for Rab-coupling protein (RCP) in mutant p53-dependent invasion. RCP promotes endosomal recycling and signalling of integrins and receptor tyrosine kinases. In a screen to identify novel RCP-interacting proteins, we discovered P-glycoprotein (P-gp). Thus, we hypothesised that mutant p53 could promote chemoresistance through RCP-dependent recycling of P-gp. The interaction between RCP and P-gp was verified endogenously and loss of RCP or mutant p53 rendered cells more sensitive to cisplatin and etoposide. In mutant p53 cells we detected an RCP-dependent delivery of P-gp to the plasma membrane upon drug treatment and decreased retention of P-gp substrates. A co-localisation of P-gp and RCP was seen in mutant p53 cells, but not in p53-null cells upon chemotherapeutic exposure. In conclusion, mutant p53 expression enhanced co-localisation of P-gp and RCP to allow for rapid delivery of P-gp to the plasma membrane and increased resistance to chemotherapeutics.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/farmacología , Membrana Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Proteínas de la Membrana/metabolismo , Mutación , Neoplasias/tratamiento farmacológico , Proteína p53 Supresora de Tumor/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antineoplásicos/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/patología , Cisplatino/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Etopósido/farmacología , Células HCT116 , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Dev Cell ; 56(1): 3-4, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33434524

RESUMEN

Carcinoma-associated fibroblast (CAF) infiltration confers poor clinical outcomes in colorectal cancer (CRC) through mechanisms that are still unclear. In this issue of Developmental Cell, Kasashima et al. report that loss of PKCζ engenders a SOX2/SFRP2-positive CAF subpopulation that increases CRC aggressiveness by creating an immunosuppressed environment.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Colorrectales , Carcinogénesis , Transformación Celular Neoplásica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana , Factores de Transcripción SOXB1/genética , Regulación hacia Arriba
10.
Sci Rep ; 10(1): 15361, 2020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32958777

RESUMEN

Despite extensive research, little progress has been made in glioblastoma therapy, owing in part to a lack of adequate preclinical in vivo models to study this disease. To mitigate this, primary patient-derived cell lines, which maintain their specific stem-like phenotypes, have replaced established glioblastoma cell lines. However, due to heterogenous tumour growth inherent in glioblastoma, the use of primary cells for orthotopic in vivo studies often requires large experimental group sizes. Therefore, when using intracranial patient-derived xenograft (PDX) approaches, it is advantageous to deploy imaging techniques to monitor tumour growth and allow stratification of mice. Here we show that stable expression of near-infrared fluorescent protein (iRFP) in patient-derived glioblastoma cells enables rapid, direct non-invasive monitoring of tumour development without compromising tumour stemness or tumorigenicity. Moreover, as this approach does not depend on the use of agents like luciferin, which can cause variability due to changing bioavailability, it can be used for quantitative longitudinal monitoring of tumour growth. Notably, we show that this technique also allows quantitative assessment of tumour burden in highly invasive models spreading throughout the brain. Thus, iRFP transduction of primary patient-derived glioblastoma cells is a reliable, cost- and time-effective way to monitor heterogenous orthotopic PDX growth.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Xenoinjertos/patología , Animales , Encéfalo/patología , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Mediciones Luminiscentes/métodos , Ratones Desnudos , Pacientes , Carga Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
11.
J Cell Biol ; 219(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32311005

RESUMEN

Microtubule-associated serine/threonine-protein kinase-like (MASTL) is a mitosis-accelerating kinase with emerging roles in cancer progression. However, possible cell cycle-independent mechanisms behind its oncogenicity remain ambiguous. Here, we identify MASTL as an activator of cell contractility and MRTF-A/SRF (myocardin-related transcription factor A/serum response factor) signaling. Depletion of MASTL increased cell spreading while reducing contractile actin stress fibers in normal and breast cancer cells and strongly impairing breast cancer cell motility and invasion. Transcriptome and proteome profiling revealed MASTL-regulated genes implicated in cell movement and actomyosin contraction, including Rho guanine nucleotide exchange factor 2 (GEF-H1, ARHGEF2) and MRTF-A target genes tropomyosin 4.2 (TPM4), vinculin (VCL), and nonmuscle myosin IIB (NM-2B, MYH10). Mechanistically, MASTL associated with MRTF-A and increased its nuclear retention and transcriptional activity. Importantly, MASTL kinase activity was not required for regulation of cell spreading or MRTF-A/SRF transcriptional activity. Taken together, we present a previously unknown kinase-independent role for MASTL as a regulator of cell adhesion, contractility, and MRTF-A/SRF activity.


Asunto(s)
Citoesqueleto de Actina/enzimología , Adhesión Celular/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal/genética , Transactivadores/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Núcleo Celular/metabolismo , Perfilación de la Expresión Génica , Humanos , Integrinas/genética , Integrinas/metabolismo , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Miosina Tipo IIB no Muscular/genética , Miosina Tipo IIB no Muscular/metabolismo , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteoma/metabolismo , ARN Interferente Pequeño , Factores de Intercambio de Guanina Nucleótido Rho/genética , Fibras de Estrés/genética , Fibras de Estrés/metabolismo , Transactivadores/genética , Transcriptoma/genética , Tropomiosina/genética , Tropomiosina/metabolismo , Vinculina/genética , Vinculina/metabolismo
12.
Curr Biol ; 30(11): 2146-2155.e5, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32302585

RESUMEN

Rho guanosine triphosphatases (GTPases) are master regulators of cell shape and cell movement [1]. The archetypal family members RhoA, Rac1, and Cdc42 arose early in eukaryotic evolution and coordinate a diverse range of cell morphologies and migrations. Evolution of the vertebrates was paralleled by expansion of this family through gene duplication. Emergence of an adaptive immune system and more complex neural systems presented new roles for Rho GTPases, filled by new family members. Cdc42 underwent gene duplication to produce two related proteins-RhoQ and RhoJ [2]. RhoQ is active in neural dynamics; however, RhoJ is highly expressed in endothelial cells under control of the endothelial-specific promoter ERG [3, 4]. RhoJ is required for angiogenesis [5, 6] and has multiple roles in this process [7, 8]. We recently demonstrated that RhoJ regulates the endosomal trafficking of podocalyxin during angiogenesis to control lumen formation [9]. Here, we use vesicle purification and proteomic analysis to identify the endothelial targets of RhoJ-mediated trafficking. We identify α5ß1 integrin as a major RhoJ cargo and show that RhoJ regulates the intracellular trafficking of active α5ß1 integrin in endothelial cells to repress fibronectin fibrillogenesis. Accordingly, mice lacking RhoJ show deregulated deposition of fibronectin around vessels during developmental angiogenesis. Intriguingly, we show that RhoJ acts in opposition to Cdc42 in this process through competition for a shared partner, PAK3. These studies identify a critical role for RhoJ in matrix remodeling during blood vessel formation and demonstrate a functional interrelationship between RhoJ and its evolutionary parent.


Asunto(s)
Integrina alfa5beta1/genética , Neovascularización Fisiológica/fisiología , Proteínas de Unión al GTP rho/genética , Animales , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa5beta1/metabolismo , Masculino , Ratones , Neovascularización Fisiológica/genética , Proteínas de Unión al GTP rho/metabolismo
13.
Dev Cell ; 51(4): 431-445.e7, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31668663

RESUMEN

Pancreatic ductal adenocarcinoma is one of the most invasive and metastatic cancers and has a dismal 5-year survival rate. We show that N-WASP drives pancreatic cancer metastasis, with roles in both chemotaxis and matrix remodeling. lysophosphatidic acid, a signaling lipid abundant in blood and ascites fluid, is both a mitogen and chemoattractant for cancer cells. Pancreatic cancer cells break lysophosphatidic acid down as they respond to it, setting up a self-generated gradient driving tumor egress. N-WASP-depleted cells do not recognize lysophosphatidic acid gradients, leading to altered RhoA activation, decreased contractility and traction forces, and reduced metastasis. We describe a signaling loop whereby N-WASP and the endocytic adapter SNX18 promote lysophosphatidic acid-induced RhoA-mediated contractility and force generation by controlling lysophosphatidic acid receptor recycling and preventing degradation. This chemotactic loop drives collagen remodeling, tumor invasion, and metastasis and could be an important target against pancreatic cancer spread.


Asunto(s)
Lisofosfolípidos/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptores del Ácido Lisofosfatídico/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Quimiotaxis , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transporte de Proteínas , Ratas , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/aislamiento & purificación , Transducción de Señal , Nexinas de Clasificación/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética , Proteína de Unión al GTP rhoA/metabolismo
14.
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
15.
Cancer Discov ; 9(5): 617-627, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837243

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) develops a pronounced stromal response reflecting an aberrant wound-healing process. This stromal reaction features transdifferentiation of tissue-resident pancreatic stellate cells (PSC) into activated cancer-associated fibroblasts, a process induced by PDAC cells but of unclear significance for PDAC progression. Here, we show that PSCs undergo a dramatic lipid metabolic shift during differentiation in the context of pancreatic tumorigenesis, including remodeling of the intracellular lipidome and secretion of abundant lipids in the activated, fibroblastic state. Specifically, stroma-derived lysophosphatidylcholines support PDAC cell synthesis of phosphatidylcholines, key components of cell membranes, and also facilitate production of the potent wound-healing mediator lysophosphatidic acid (LPA) by the extracellular enzyme autotaxin, which is overexpressed in PDAC. The autotaxin-LPA axis promotes PDAC cell proliferation, migration, and AKT activation, and genetic or pharmacologic autotaxin inhibition suppresses PDAC growth in vivo. Our work demonstrates how PDAC cells exploit the local production of wound-healing mediators to stimulate their own growth and migration. SIGNIFICANCE: Our work highlights an unanticipated role for PSCs in producing the oncogenic LPA signaling lipid and demonstrates how PDAC tumor cells co-opt the release of wound-healing mediators by neighboring PSCs to promote their own proliferation and migration.See related commentary by Biffi and Tuveson, p. 578.This article is highlighted in the In This Issue feature, p. 565.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Lisofosfatidilcolinas/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Células del Estroma/metabolismo , Animales , Carcinoma Ductal Pancreático/patología , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Transducción de Señal , Células del Estroma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nat Commun ; 9(1): 5069, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30498210

RESUMEN

Mutant p53s (mutp53) increase cancer invasiveness by upregulating Rab-coupling protein (RCP) and diacylglycerol kinase-α (DGKα)-dependent endosomal recycling. Here we report that mutp53-expressing tumour cells produce exosomes that mediate intercellular transfer of mutp53's invasive/migratory gain-of-function by increasing RCP-dependent integrin recycling in other tumour cells. This process depends on mutp53's ability to control production of the sialomucin, podocalyxin, and activity of the Rab35 GTPase which interacts with podocalyxin to influence its sorting to exosomes. Exosomes from mutp53-expressing tumour cells also influence integrin trafficking in normal fibroblasts to promote deposition of a highly pro-invasive extracellular matrix (ECM), and quantitative second harmonic generation microscopy indicates that this ECM displays a characteristic orthogonal morphology. The lung ECM of mice possessing mutp53-driven pancreatic adenocarcinomas also displays increased orthogonal characteristics which precedes metastasis, indicating that mutp53 can influence the microenvironment in distant organs in a way that can support invasive growth.


Asunto(s)
Exosomas/metabolismo , Sialoglicoproteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Exosomas/genética , Femenino , Humanos , Ratones , Ratones Desnudos , Microscopía de Fuerza Atómica , Mutación/genética , Sialoglicoproteínas/genética , Sialomucinas/genética , Sialomucinas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
18.
Nat Commun ; 8(1): 2255, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29269878

RESUMEN

The role of glutaminolysis in providing metabolites to support tumour growth is well-established, but the involvement of glutamine metabolism in invasive processes is yet to be elucidated. Here we show that normal mammary epithelial cells consume glutamine, but do not secrete glutamate. Indeed, low levels of extracellular glutamate are necessary to maintain epithelial homoeostasis, and provision of glutamate drives disruption of epithelial morphology and promotes key characteristics of the invasive phenotype such as lumen-filling and basement membrane disruption. By contrast, primary cultures of invasive breast cancer cells convert glutamine to glutamate which is released from the cell through the system Xc- antiporter to activate a metabotropic glutamate receptor. This contributes to the intrinsic aggressiveness of these cells by upregulating Rab27-dependent recycling of the transmembrane matrix metalloprotease, MT1-MMP to promote invasive behaviour leading to basement membrane disruption. These data indicate that acquisition of the ability to release glutamate is a key watershed in disease aggressiveness.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Glándulas Mamarias Humanas/metabolismo , Neoplasias Mamarias Animales/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Espacio Extracelular/metabolismo , Femenino , Homeostasis , Humanos , Neoplasias Mamarias Animales/patología , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Invasividad Neoplásica , Receptores de Glutamato Metabotrópico/metabolismo , Regulación hacia Arriba , Proteínas rab27 de Unión a GTP/metabolismo
19.
J Cell Biol ; 216(4): 867-869, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28325807

RESUMEN

The regulation of integrin function is key to fundamental cellular processes, including cell migration and extracellular matrix (ECM) assembly. In this issue, Georgiadou et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201609066) report that the metabolic sensor adenosine monophosphate-activated protein kinase influences tensin production to regulate α5ß1-integrin and fibrillar adhesion assembly and thus reveal an important connection between energy metabolism and ECM assembly.


Asunto(s)
Metabolismo Energético/fisiología , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Tensinas/metabolismo , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Humanos , Integrina alfa5beta1/metabolismo
20.
Nat Commun ; 8: 14646, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28294115

RESUMEN

The Rab GTPase effector, Rab-coupling protein (RCP) is known to promote invasive behaviour in vitro by controlling integrin and receptor tyrosine kinase (RTK) trafficking, but how RCP influences metastasis in vivo is unclear. Here we identify an RTK of the Eph family, EphA2, to be a cargo of an RCP-regulated endocytic pathway which controls cell:cell repulsion and metastasis in vivo. Phosphorylation of RCP at Ser435 by Lemur tyrosine kinase-3 (LMTK3) and of EphA2 at Ser897 by Akt are both necessary to promote Rab14-dependent (and Rab11-independent) trafficking of EphA2 which generates cell:cell repulsion events that drive tumour cells apart. Genetic disruption of RCP or EphA2 opposes cell:cell repulsion and metastasis in an autochthonous mouse model of pancreatic adenocarcinoma-whereas conditional knockout of another RCP cargo, α5 integrin, does not suppress pancreatic cancer metastasis-indicating a role for RCP-dependent trafficking of an Eph receptor to drive tumour dissemination in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor EphA2/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Adenocarcinoma/patología , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...