Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Hum Cell ; 37(5): 1593-1601, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39103560

RESUMEN

Neuroendocrine tumors (NETs) of the pancreas are rare neoplasms that present complex challenges to diagnosis and treatment due to their indolent course. The incidence of pancreatic neuroendocrine tumors has increased significantly over the past two decades. A limited number of pancreatic neuroendocrine cell lines are currently available for the research. Here, we present 3D-iNET ORION, a novel 3-dimensional (spheroid) cell line, isolated from human pancreatic neuroendocrine tumor liver metastasis. Three-dimensionally grown (3D) cancer cell lines have gained interest over the past years as 3D cancer cell lines better recapitulate the in vivo structure of tumors, and are more suitable for in vitro and in vivo experiments. 3D-iNET ORION cancer cell line showed high potential to form tumorspheres when embedded in Matrigel matrix and expresses synaptophysin and EpCAM. Electron microscopy analysis of cancer cell line proved the presence of dense neurosecretory granules. When xenografted into athymic mice, 3D-iNET ORION cells produce slow-growing tumors, positive for chromogranin and synaptophysin. Human Core Exome Panel Analysis has shown that 3DiNET ORION cell line retains the genetic aberration profile detected in the original tumor. In conclusion, our newly developed neuroendocrine cancer cell line can be considered as a new research tool for in vitro and in vivo experiments.


Asunto(s)
Ratones Desnudos , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/patología , Tumores Neuroendocrinos/patología , Animales , Línea Celular Tumoral , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Esferoides Celulares/patología , Técnicas de Cultivo Tridimensional de Células/métodos , Modelos Biológicos , Ratones , Técnicas de Cultivo de Célula/métodos
2.
Nat Biomed Eng ; 6(7): 882-897, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34931077

RESUMEN

Targeting the delivery of therapeutics specifically to diseased tissue enhances their efficacy and decreases their side effects. Here we show that mesenchymal stromal cells with their nuclei removed by density-gradient centrifugation following the genetic modification of the cells for their display of chemoattractant receptors and endothelial-cell-binding molecules are effective vehicles for the targeted delivery of therapeutics. The enucleated cells neither proliferate nor permanently engraft in the host, yet retain the organelles for energy and protein production, undergo integrin-regulated adhesion to inflamed endothelial cells, and actively home to chemokine gradients established by diseased tissues. In mouse models of acute inflammation and of pancreatitis, systemically administered enucleated cells expressing two types of chemokine receptor and an endothelial adhesion molecule enhanced the delivery of an anti-inflammatory cytokine to diseased tissue (with respect to unmodified stromal cells and to exosomes derived from bone-marrow-derived stromal cells), attenuating inflammation and ameliorating disease pathology. Enucleated cells retain most of the cells' functionality, yet acquire the cargo-carrying characteristics of cell-free delivery systems, and hence represent a versatile delivery vehicle and therapeutic system.


Asunto(s)
Sistemas de Liberación de Medicamentos , Células Madre Mesenquimatosas , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Humanos , Inflamación/metabolismo , Ratones
3.
Front Oncol ; 11: 697626, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422650

RESUMEN

Angiogenesis is the formation of new vessels from pre-existing vasculature. The heparan sulfate chains from endothelial cell proteoglycans interact with the major angiogenic factors, regulating blood vessels´ formation. Since the FDA´s first approval, anti-angiogenic therapy has shown tumor progression inhibition and increased patient survival. Previous work in our group has selected an HS-binding peptide using a phage display system. Therefore, we investigated the effect of the selected peptide in angiogenesis and tumor progression. The HS-binding peptide showed a higher affinity for heparin N-sulfated. The HS-binding peptide was able to inhibit the proliferation of human endothelial umbilical cord cells (HUVEC) by modulation of FGF-2. It was verified a significant decrease in the tube formation of human endothelial cells and capillary formation of mice aorta treated with HS-binding peptide. HS-binding peptide also inhibited the formation of sub-intestinal blood vessels in zebrafish embryos. Additionally, in zebrafish embryos, the tumor size decreased after treatment with HS-binding peptide.

4.
Am J Pathol ; 191(12): 2203-2218, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34428425

RESUMEN

Bladder cancer invasion depends on mammalian target of rapamycin complex 2 (mTORC2) activity, although the downstream mTORC2 effectors that mediate this effect have not been fully defined. One potential downstream effector is the arginine derivative nitric oxide (NO). This study identified a stage-associated increase in the expression of the NO-generating enzymes endothelial NO synthase (eNOS) and inducible NOS (iNOS) in human bladder cancer. Reduction of NOS activity by pharmacologic inhibition or silencing of NOS enzymes reduced cancer cell invasion, with similar effects observed using the NO scavenger cobinamide. By contrast, enhanced invasion was seen with the NO donor Deta-NONOate and an analog of the downstream NO second messenger cGMP. Next, NOS expression was evaluated in invadopodia, which are cellular protrusions that form the invasive tips of cancer cells. Invadopodia were enriched in both iNOS protein and mTORC2 activity, and invadopodia formation was increased by Deta-NONOate and decreased by cobinamide and ablation of mTORC2 activity. Additionally, mTORC2 increased expression of iNOS. Using a zebrafish model, injection of iNOS- or rictor-silenced cells reduced the frequency of bladder cancer cell metastasis in zebrafish. These results indicate that mTORC2 can mediate bladder cancer cell invasion through increased iNOS expression, resulting in increased NO and cGMP production in invadopodia and further propagation of invadopodia formation.


Asunto(s)
Diana Mecanicista del Complejo 2 de la Rapamicina/fisiología , Óxido Nítrico/metabolismo , Podosomas/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Animales , Carcinoma de Células Transicionales/genética , Carcinoma de Células Transicionales/metabolismo , Carcinoma de Células Transicionales/patología , Embrión no Mamífero , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Podosomas/genética , Podosomas/patología , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Pez Cebra/embriología
5.
Cell Discov ; 4: 26, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29872538

RESUMEN

PEAK1 is a newly described tyrosine kinase and scaffold protein that transmits integrin-mediated extracellular matrix (ECM) signals to facilitate cell movement and growth. While aberrant expression of PEAK1 has been linked to cancer progression, its normal physiological role in vertebrate biology is not known. Here we provide evidence that PEAK1 plays a central role in orchestrating new vessel formation in vertebrates. Deletion of the PEAK1 gene in zebrafish, mice, and human endothelial cells (ECs) induced severe defects in new blood vessel formation due to deficiencies in EC proliferation, survival, and migration. Gene transcriptional and proteomic analyses of PEAK1-deficient ECs revealed a significant loss of vascular endothelial growth factor receptor 2 (VEGFR2) mRNA and protein expression, as well as downstream signaling to its effectors, ERK, Akt, and Src kinase. PEAK1 regulates VEGFR2 expression by binding to and increasing the protein stability of the transcription factor GATA-binding protein 2 (GATA2), which controls VEGFR2 transcription. Importantly, PEAK1-GATA2-dependent VEGFR2 expression is mediated by EC adhesion to the ECM and is required for breast cancer-induced new vessel formation in mice. Also, elevated expression of PEAK1 and VEGFR2 mRNA are highly correlated in many human cancers including breast cancer. Together, our findings reveal a novel PEAK1-GATA2-VEGFR2 signaling axis that integrates cell adhesion and growth factor cues from the extracellular environment necessary for new vessel formation during vertebrate development and cancer.

6.
Nat Commun ; 9(1): 1310, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29615667

RESUMEN

Lipoprotein lipase (LPL) mediates hydrolysis of triglycerides (TGs) to supply free fatty acids (FFAs) to tissues. Here, we show that LPL activity is also required for hematopoietic stem progenitor cell (HSPC) maintenance. Knockout of Lpl or its obligatory cofactor Apoc2 results in significantly reduced HSPC expansion during definitive hematopoiesis in zebrafish. A human APOC2 mimetic peptide or the human very low-density lipoprotein, which carries APOC2, rescues the phenotype in apoc2 but not in lpl mutant zebrafish. Creating parabiotic apoc2 and lpl mutant zebrafish rescues the hematopoietic defect in both. Docosahexaenoic acid (DHA) is identified as an important factor in HSPC expansion. FFA-DHA, but not TG-DHA, rescues the HSPC defects in apoc2 and lpl mutant zebrafish. Reduced blood cell counts are also observed in Apoc2 mutant mice at the time of weaning. These results indicate that LPL-mediated release of the essential fatty acid DHA regulates HSPC expansion and definitive hematopoiesis.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Lipoproteína Lipasa/metabolismo , Células Madre/citología , Animales , Apoptosis , Compuestos Azo/química , Separación Celular , Femenino , Citometría de Flujo , Cromatografía de Gases y Espectrometría de Masas , Hematopoyesis , Humanos , Hidrólisis , Hibridación in Situ , Lipoproteína Lipasa/genética , Lipoproteínas VLDL/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Péptidos/química , Triglicéridos/química , Pez Cebra
7.
Sci Rep ; 8(1): 2982, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445239

RESUMEN

PLAUR encodes the urokinase receptor (uPAR), which promotes cell survival, migration, and resistance to targeted cancer therapeutics in glioblastoma cells in culture and in mouse model systems. Herein, we show that patient survival correlates inversely with PLAUR mRNA expression in gliomas of all grades, in glioblastomas, and in the subset of glioblastomas that demonstrate the mesenchymal gene expression signature. PLAUR clusters with genes that define the more aggressive mesenchymal subtype in transcriptome profiles of glioblastoma tissue and glioblastoma cells in neurospheres, which are enriched for multipotent cells with stem cell-like qualities. When PLAUR was over-expressed or silenced in glioblastoma cells, neurosphere growth and expression of mesenchymal subtype biomarkers correlated with uPAR abundance. uPAR also promoted glioblastoma cell survival in neurospheres. Constitutively-active EGF Receptor (EGFRvIII) promoted neurosphere growth; however, unlike uPAR, EGFRvIII did not induce the mesenchymal gene expression signature. Immunohistochemical analysis of human glioblastomas showed that uPAR is typically expressed by a small sub-population of the cancer cells; it is thus reasonable to conclude that this subpopulation of cells is responsible for the effects of PLAUR on patient survival. We propose that uPAR-expressing glioblastoma cells demonstrate a mesenchymal gene signature, an increased capacity for cell survival, and stem cell-like properties.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Células Madre Mesenquimatosas/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Animales , Neoplasias Encefálicas/mortalidad , Movimiento Celular , Proliferación Celular , Supervivencia Celular/genética , Estudios de Cohortes , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Glioblastoma/mortalidad , Humanos , Ratones , ARN Interferente Pequeño/genética , Análisis de Supervivencia , Análisis de Matrices Tisulares , Transcriptoma , Células Tumorales Cultivadas
8.
Cancer Res ; 78(6): 1444-1456, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29321164

RESUMEN

There remains intense interest in tractable approaches to target or silence the KRAS oncoprotein as a rational therapeutic strategy to attack pancreatic ductal adenocarcinoma (PDAC) and other cancers that overexpress it. Here we provide evidence that accumulation of the KRAS oncoprotein is controlled by a self-regulating feed-forward regulatory loop that utilizes a unique hypusinated isoform of the translation elongation factor eIF5A and the tyrosine kinase PEAK1. Oncogenic activation of KRAS increased eIF5A-PEAK1 translational signaling, which in turn facilitated increased KRAS protein synthesis. Mechanistic investigations show that this feed-forward positive regulatory pathway was controlled by oncogenic KRAS-driven metabolic demands, operated independently of canonical mTOR signaling, and did not involve new KRAS gene transcription. Perturbing eIF5A-PEAK1 signaling, by genetic or pharmacologic strategies or by blocking glutamine synthesis, was sufficient to inhibit expression of KRAS, eIF5A, and PEAK1, to attenuate cancer cell growth and migration, and to block tumor formation in established preclinical mouse models of PDAC. Levels of KRAS, eIF5A, and PEAK1 protein increased during cancer progression with the highest levels of expression observed in metastatic cell populations. Combinatorial targeting of eIF5A hypusination and the RAS-ERK signaling pathway cooperated to attenuate KRAS expression and its downstream signaling along with cell growth in vitro and tumor formation in vivo Collectively, our findings highlight a new mechanistic strategy to attenuate KRAS expression as a therapeutic strategy to target PDAC and other human cancers driven by KRAS activation.Significance: These findings highlight a new mechanistic strategy to attenuate KRAS expression as a therapeutic strategy to target human cancers driven by KRAS activation. Cancer Res; 78(6); 1444-56. ©2018 AACR.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Retroalimentación Fisiológica , Femenino , GTP Fosfohidrolasas/metabolismo , Glutamina/metabolismo , Humanos , Lisina/análogos & derivados , Lisina/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Factores de Iniciación de Péptidos/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al ARN/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Factor 5A Eucariótico de Iniciación de Traducción
9.
Exp Cell Res ; 363(2): 271-282, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29355494

RESUMEN

Extracellular matrix (ECM) serves as a reservoir for biologically active factors, such as growth factors and proteases that influence the tumor cell behavior. ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin motifs) is a secreted protease that has the ability to modify the ECM during physiological and pathological processes. Here, we analyzed the role played by ADAMTS-1 regulating HGF and TGF-ß1 activities in the high-grade fibrosarcoma cell line (HT1080). We generated HT1080 and HEK293T cells overexpressing ADAMTS-1. HT1080 cells overexpressing ADAMTS-1 (HT1080-MPA) exhibited a significant decrease in cell proliferation and migration velocity, both in presence of HGF. We obtained similar results with ADAMTS-1-enriched conditioned medium from other cell type. However, ADAMTS-1 overexpression failed to affect TGF-ß1 activity associated with HT1080 cell proliferation and migration velocity. Immunoblotting showed that ADAMTS-1 overexpression disturbs c-Met activation upon HGF stimulation. Downstream ERK1/2 and FAK signaling pathways are also influenced by this protease. Additionally, ADAMTS-1 decreased the size of the fibrosarcospheres, both under normal conditions and in the presence of HGF. Likewise, in presence of HGF, ADAMTS-1 overexpression in HT1080 disrupted microtumors formation in vivo. These microtumors, including individual cells, presented characteristics of non-invasive lesions (rounded morphology). Our results suggest that ADAMTS-1 is involved in regulating HGF-related functions on fibrosarcoma cells. This protease may then represent an endogenous mechanism in controlling the bioavailability of different growth factors that have a direct influence on tumor cell behavior.


Asunto(s)
Proteína ADAMTS1/metabolismo , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Células HEK293 , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo
10.
Cancer Res ; 77(8): 1997-2007, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28381547

RESUMEN

In pancreatic ductal adenocarcinoma (PDAC), mutant KRAS stimulates the translation initiation factor eIF5A and upregulates the focal adhesion kinase PEAK1, which transmits integrin and growth factor signals mediated by the tumor microenvironment. Although eIF5A-PEAK1 signaling contributes to multiple aggressive cancer cell phenotypes, the downstream signaling processes that mediate these responses are uncharacterized. Through proteomics and informatic analyses of PEAK1-depleted PDAC cells, we defined protein translation, cytoskeleton organization, and cell-cycle regulatory pathways as major pathways controlled by PEAK1. Biochemical and functional studies revealed that the transcription factors YAP1 and TAZ are key targets of eIF5A-PEAK1 signaling. YAP1/TAZ coimmunoprecipitated with PEAK1. Interfering with eIF5A-PEAK1 signaling in PDAC cells inhibited YAP/TAZ protein expression, decreasing expression of stem cell-associated transcription factors (STF) including Oct4, Nanog, c-Myc, and TEAD, thereby decreasing three-dimensional (3D) tumor sphere growth. Conversely, amplified eIF5A-PEAK1 signaling increased YAP1/TAZ expression, increasing expression of STF and enhancing 3D tumor sphere growth. Informatic interrogation of mRNA sequence databases revealed upregulation of the eIF5A-PEAK1-YAP1-TEAD signaling module in PDAC patients. Taken together, our findings indicate that eIF5A-PEAK1-YAP signaling contributes to PDAC development by regulating an STF program associated with increased tumorigenicity. Cancer Res; 77(8); 1997-2007. ©2017 AACR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pancreáticas/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Ciclo Celular/fisiología , Línea Celular Tumoral , Citoesqueleto/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Factores de Iniciación de Péptidos/biosíntesis , Factores de Iniciación de Péptidos/genética , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Transducción de Señal , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP , Factor 5A Eucariótico de Iniciación de Traducción
11.
PLoS One ; 10(8): e0135748, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26267863

RESUMEN

Transforming Growth Factor ß (TGFß) has dual functions as both a tumor suppressor and a promoter of cancer progression within the tumor microenvironment, but the molecular mechanisms by which TGFß signaling switches between these outcomes and the contexts in which this switch occurs remain to be fully elucidated. We previously identified PEAK1 as a new non-receptor tyrosine kinase that associates with the cytoskeleton, and facilitates signaling of HER2/Src complexes. We also showed PEAK1 functions downstream of KRas to promote tumor growth, metastasis and therapy resistance using preclinical in vivo models of human tumor progression. In the current study, we analyzed PEAK1 expression in human breast cancer samples and found PEAK1 levels correlate with mesenchymal gene expression, poor cellular differentiation and disease relapse. At the cellular level, we also observed that PEAK1 expression was highest in mesenchymal breast cancer cells, correlated with migration potential and increased in response to TGFß-induced epithelial-mesenchymal transition (EMT). Thus, we sought to evaluate the role of PEAK1 in the switching of TGFß from a tumor suppressing to tumor promoting factor. Notably, we discovered that high PEAK1 expression causes TGFß to lose its anti-proliferative effects, and potentiates TGFß-induced proliferation, EMT, cell migration and tumor metastasis in a fibronectin-dependent fashion. In the presence of fibronectin, PEAK1 caused a switching of TGFß signaling from its canonical Smad2/3 pathway to non-canonical Src and MAPK signaling. This report is the first to provide evidence that PEAK1 mediates signaling cross talk between TGFß receptors and integrin/Src/MAPK pathways and that PEAK1 is an important molecular regulator of TGFß-induced tumor progression and metastasis in breast cancer. Finally, PEAK1 overexpression/upregulation cooperates with TGFß to reduce breast cancer sensitivity to Src kinase inhibition. These findings provide a rational basis to develop therapeutic agents to target PEAK1 expression/function or upstream/downstream pathways to abrogate breast cancer progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Células MCF-7 , Proteínas Tirosina Quinasas/genética , Transducción de Señal/efectos de los fármacos
12.
Cancer Res ; 74(22): 6671-81, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25261239

RESUMEN

Deregulation of protein synthesis is a hallmark of cancer cell proliferation, survival, and metastatic progression. eIF5A1 and its highly related isoform eIF5A2 are translation initiation factors that have been implicated in a range of human malignancies, but how they control cancer development and disease progression is still poorly understood. Here, we investigated how eIF5A proteins regulate pancreatic ductal adenocarcinoma (PDAC) pathogenesis. eIF5A proteins are the only known proteins regulated by a distinct posttranslational modification termed hypusination, which is catalyzed by two enzymes, deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH). The highly selective nature of the hypusine modification and its amenability to pharmacologic inhibition make eIF5A proteins attractive therapeutic targets. We found that the expression and hypusination of eIF5A proteins are upregulated in human PDAC tissues and in premalignant pancreatic intraepithelial neoplasia tissues isolated from Pdx-1-Cre: LSL-KRAS(G12D) mice. Knockdown of eIF5A proteins in PDAC cells inhibited their growth in vitro and orthotopic tumor growth in vivo, whereas amplification of eIF5A proteins increased PDAC cell growth and tumor formation in mice. Small-molecule inhibitors of DHPS and DOHH both suppressed eIF5A hypusination, preventing PDAC cell growth. Interestingly, we found that eIF5A proteins regulate PDAC cell growth by modulating the expression of PEAK1, a nonreceptor tyrosine kinase essential for PDAC cell growth and therapy resistance. Our findings suggest that eIF5A proteins utilize PEAK1 as a downstream effector to drive PDAC pathogenesis and that pharmacologic inhibition of the eIF5A-hypusine-PEAK1 axis may provide a novel therapeutic strategy to combat this deadly disease.


Asunto(s)
Carcinoma Ductal Pancreático/etiología , Lisina/análogos & derivados , Neoplasias Pancreáticas/etiología , Factores de Iniciación de Péptidos/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas de Unión al ARN/fisiología , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Ciclopirox , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Femenino , Humanos , Lisina/fisiología , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas p21(ras) , Piridonas/farmacología , Proteínas ras/fisiología , Gemcitabina , Factor 5A Eucariótico de Iniciación de Traducción
13.
Methods Mol Biol ; 1046: 203-18, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23868590

RESUMEN

The cytoskeleton is fundamental to many cellular functions including cell proliferation, differentiation, adhesion, and migration. It is composed of actin, microtubules, intermediate filaments, and integrin cell surface receptors, which form focal adhesions with the extracellular matrix. These elements are highly integrated in the cell providing a rigid network of interconnected cables and protein scaffolds, which generate force and mechanical support to maintain cell shape and movement. However, the cytoskeleton is not just a simple compilation of static filaments that dictate cell adhesion and morphology-it is highly plastic with the inherent ability to assemble and disassemble in response to diverse and complex cellular cues. Thus, biochemical and proteomic methods are needed to better understand the cytoskeleton network and its dynamic signal transduction functions in health and disease. This chapter describes methods for the biochemical enrichment and mass spectrometry-based proteomic analyses of the cytoskeletome. We also detail how these methods can be used to investigate the cytoskeletome of migrating cells and their purified pseudopodia membrane projections.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Espectrometría de Masas/métodos , Proteómica , Actinas/genética , Adhesión Celular/genética , Citoesqueleto/genética , Eptifibatida , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Humanos , Filamentos Intermedios/metabolismo , Microtúbulos/metabolismo , Péptidos/metabolismo , Receptores de Superficie Celular , Transducción de Señal
14.
J Cell Sci ; 126(Pt 4): 904-13, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23321642

RESUMEN

Breast cancer and melanoma cells commonly metastasize to the brain using homing mechanisms that are poorly understood. Cancer patients with brain metastases display poor prognosis and survival due to the lack of effective therapeutics and treatment strategies. Recent work using intravital microscopy and preclinical animal models indicates that metastatic cells colonize the brain, specifically in close contact with the existing brain vasculature. However, it is not known how contact with the vascular niche promotes microtumor formation. Here, we investigate the role of connexins in mediating early events in brain colonization using transparent zebrafish and chicken embryo models of brain metastasis. We provide evidence that breast cancer and melanoma cells utilize connexin gap junction proteins (Cx43, Cx26) to initiate brain metastatic lesion formation in association with the vasculature. RNAi depletion of connexins or pharmacological blocking of connexin-mediated cell-cell communication with carbenoxolone inhibited brain colonization by blocking tumor cell extravasation and blood vessel co-option. Activation of the metastatic gene twist in breast cancer cells increased Cx43 protein expression and gap junction communication, leading to increased extravasation, blood vessel co-option and brain colonization. Conversely, inhibiting twist activity reduced Cx43-mediated gap junction coupling and brain colonization. Database analyses of patient histories revealed increased expression of Cx26 and Cx43 in primary melanoma and breast cancer tumors, respectively, which correlated with increased cancer recurrence and metastasis. Together, our data indicate that Cx43 and Cx26 mediate cancer cell metastasis to the brain and suggest that connexins might be exploited therapeutically to benefit cancer patients with metastatic disease.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Conexinas/metabolismo , Melanoma/complicaciones , Melanoma/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias de la Mama/genética , Embrión de Pollo , Conexina 26 , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Femenino , Humanos , Melanoma/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/genética , Interferencia de ARN
15.
J Biol Chem ; 288(1): 123-31, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23105102

RESUMEN

Pseudopodium-enriched atypical kinase 1 (PEAK1) is a recently described tyrosine kinase that associates with the actin cytoskeleton and focal adhesion (FA) in migrating cells. PEAK1 is known to promote cell migration, but the responsible mechanisms remain unclear. Here, we show that PEAK1 controls FA assembly and disassembly in a dynamic pathway controlled by PEAK1 phosphorylation at Tyr-665. Knockdown of endogenous PEAK1 inhibits random cell migration. In PEAK1-deficient cells, FA lifetimes are decreased, FA assembly times are shortened, and FA disassembly times are extended. Phosphorylation of Tyr-665 in PEAK1 is essential for normal PEAK1 localization and its function in the regulation of FAs; however, constitutive phosphorylation of PEAK1 Tyr-665 is also disruptive of its function, indicating a requirement for precise spatiotemporal regulation of PEAK1. Src family kinases are required for normal PEAK1 localization and function. Finally, we provide evidence that PEAK1 promotes cancer cell invasion through Matrigel by a mechanism that requires dynamic regulation of Tyr-665 phosphorylation.


Asunto(s)
Adhesiones Focales/química , Regulación de la Expresión Génica , Proteínas Tirosina Quinasas/química , Tirosina/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Colágeno/química , Combinación de Medicamentos , Humanos , Laminina/química , Paxillin/metabolismo , Fosforilación , Proteoglicanos/química , Factores de Tiempo , Familia-src Quinasas/metabolismo
16.
Curr Opin Cell Biol ; 24(5): 662-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22980730

RESUMEN

Metastatic cancer cells produce invasive membrane protrusions called invadopodia and pseudopodia, which play a central role in driving cancer cell dissemination in the body. Malignant cells use these structures to attach to and degrade extracellular matrix proteins, generate force for cell locomotion, and to penetrate the vasculature. Recent work using unique subcellular fractionation methodologies combined with spatial genomic, proteomic, and phosphoproteomic profiling has provided insight into the invadopodiome and pseudopodiome signaling networks that control the protrusion of invasive membranes. Here I highlight how these powerful spatial 'omics' approaches reveal important signatures of metastatic cancer cells and possible new therapeutic targets aimed at treating metastatic disease.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Animales , Movimiento Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Invasividad Neoplásica/patología , Proteómica , Seudópodos/genética , Seudópodos/metabolismo , Seudópodos/patología , ARN Neoplásico/análisis , ARN Neoplásico/genética
17.
Biomaterials ; 33(29): 7064-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22809641

RESUMEN

Our current understanding of 3-dimensional (3D) cell migration is primarily based on results from fibrous scaffolds with randomly organized internal architecture. Manipulations that change the stiffness of these 3D scaffolds often alter other matrix parameters that can modulate cell motility independently or synergistically, making observations less predictive of how cells behave when migrating in 3D. In order to decouple microstructural influences and stiffness effects, we have designed and fabricated 3D polyethylene glycol (PEG) scaffolds that permit orthogonal tuning of both elastic moduli and microstructure. Scaffolds with log-pile architectures were used to compare the 3D migration properties of normal breast epithelial cells (HMLE) and Twist-transformed cells (HMLET). Our results indicate that the nature of cell migration is significantly impacted by the ability of cells to migrate in the third dimension. 2D ECM-coated PEG substrates revealed no statistically significant difference in cell migration between HMLE and HMLET cells among substrates of different stiffness. However, when cells were allowed to move along the third dimension, substantial differences were observed for cell displacement, velocity and path straightness parameters. Furthermore, these differences were sensitive to both substrate stiffness and the presence of the Twist oncogene. Importantly, these 3D modes of migration provide insight into the potential for oncogene-transformed cells to migrate within and colonize tissues of varying stiffness.


Asunto(s)
Neoplasias/metabolismo , Polietilenglicoles/química , Andamios del Tejido/química , Biofisica/métodos , Mama/citología , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Reactivos de Enlaces Cruzados/farmacología , Elasticidad , Células Epiteliales/citología , Diseño de Equipo , Femenino , Humanos , Microscopía Electrónica de Rastreo/métodos , Metástasis de la Neoplasia , Polímeros/química , Estrés Mecánico
18.
Cancer Res ; 72(10): 2554-64, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22589274

RESUMEN

Early biomarkers and effective therapeutic strategies are desperately needed to treat pancreatic ductal adenocarcinoma (PDAC), which has a dismal 5-year patient survival rate. Here, we report that the novel tyrosine kinase PEAK1 is upregulated in human malignancies, including human PDACs and pancreatic intraepithelial neoplasia (PanIN). Oncogenic KRas induced a PEAK1-dependent kinase amplification loop between Src, PEAK1, and ErbB2 to drive PDAC tumor growth and metastasis in vivo. Surprisingly, blockade of ErbB2 expression increased Src-dependent PEAK1 expression, PEAK1-dependent Src activation, and tumor growth in vivo, suggesting a mechanism for the observed resistance of patients with PDACs to therapeutic intervention. Importantly, PEAK1 inactivation sensitized PDAC cells to trastuzumab and gemcitabine therapy. Our findings, therefore, suggest that PEAK1 is a novel biomarker, critical signaling hub, and new therapeutic target in PDACs.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Genes erbB-2 , Proteína Oncogénica pp60(v-src)/genética , Neoplasias Pancreáticas/genética , Proteínas Tirosina Quinasas/genética , Animales , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Resistencia a Antineoplásicos , Genes ras , Humanos , Ratones , Ratones Transgénicos , Modelos Moleculares , Metástasis de la Neoplasia/genética , Trasplante de Neoplasias , Transducción de Señal/genética , Activación Transcripcional , Regulación hacia Arriba
19.
Methods Mol Biol ; 757: 349-65, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21909922

RESUMEN

Cell migration requires actin/myosin-mediated membrane protrusion of a pseudopodium (or lamellipodium) and its attachment to the substratum. This process guides the direction of cell movement through cytoskeletal remodeling and is regulated by complex signaling networks that act spatially downstream of integrin adhesion receptors. Understanding how these regulatory networks are organized in migratory cells is important for many physiological and pathological processes, including wound healing, immune function, and cancer metastasis. Here, we describe methods for the immunoaffinity purification of phosphotyrosine proteins (pY) from pseudopodia that have been isolated from migratory cells. These methods are compatible with current mass spectrometry-based protein identification technologies and can be utilized for the large-scale identification of the pseudopodium pY proteome in various migratory cell lines, including primary and cancer cells.


Asunto(s)
Fosfotirosina/metabolismo , Proteínas/metabolismo , Proteómica , Seudópodos/metabolismo , Animales , Células COS , Movimiento Celular/fisiología , Chlorocebus aethiops , Ratones , Células 3T3 NIH , Proteínas/aislamiento & purificación
20.
Proteomics ; 11(10): 2019-26, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21500348

RESUMEN

In this study, we evaluated a concatenated low pH (pH 3) and high pH (pH 10) reversed-phase liquid chromatography strategy as a first dimension for two-dimensional liquid chromatography tandem mass spectrometry ("shotgun") proteomic analysis of trypsin-digested human MCF10A cell sample. Compared with the more traditional strong cation exchange method, the use of concatenated high pH reversed-phase liquid chromatography as a first-dimension fractionation strategy resulted in 1.8- and 1.6-fold increases in the number of peptide and protein identifications (with two or more unique peptides), respectively. In addition to broader identifications, advantages of the concatenated high pH fractionation approach include improved protein sequence coverage, simplified sample processing, and reduced sample losses. The results demonstrate that the concatenated high pH reversed-phased strategy is an attractive alternative to strong cation exchange for two-dimensional shotgun proteomic analysis.


Asunto(s)
Mama/química , Cromatografía de Fase Inversa/métodos , Fragmentos de Péptidos/aislamiento & purificación , Mapeo Peptídico/métodos , Proteoma/química , Acetonitrilos/química , Mama/citología , Mama/metabolismo , Línea Celular , Análisis por Conglomerados , Células Epiteliales/química , Células Epiteliales/citología , Células Epiteliales/metabolismo , Formiatos/química , Humanos , Concentración de Iones de Hidrógeno , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Tripsina/metabolismo , Urea/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA