Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cancer Sci ; 115(4): 1333-1345, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38320747

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies worldwide. However, drug discovery for PDAC treatment has proven complicated, leading to stagnant therapeutic outcomes. Here, we identify Glycogen synthase kinase 3 (GSK3) as a therapeutic target through a whole-body genetic screening utilizing a '4-hit' Drosophila model mimicking the PDAC genotype. Reducing the gene dosage of GSK3 in a whole-body manner or knocking down GSK3 specifically in transformed cells suppressed 4-hit fly lethality, similar to Mitogen-activated protein kinase kinase (MEK), the therapeutic target in PDAC we have recently reported. Consistently, a combination of the GSK3 inhibitor CHIR99021 and the MEK inhibitor trametinib suppressed the phosphorylation of Polo-like kinase 1 (PLK1) as well as the growth of orthotopic human PDAC xenografts in mice. Additionally, reducing PLK1 genetically in 4-hit flies rescued their lethality. Our results reveal a therapeutic vulnerability in PDAC that offers a treatment opportunity for patients by inhibiting multiple targets.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Quinasas de Proteína Quinasa Activadas por Mitógenos , Glucógeno Sintasa Quinasa 3/metabolismo , Transducción de Señal , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo
2.
Cancer Sci ; 112(2): 505-514, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33275812

RESUMEN

Cancer burden has been increasing worldwide, making cancer the second leading cause of death in the world. Over the past decades, various experimental models have provided important insights into the nature of cancer. Among them, the fruit fly Drosophila as a whole-animal toolkit has made a decisive contribution to our understanding of fundamental mechanisms of cancer development including loss of cell polarity. In recent years, scalable Drosophila platforms have proven useful also in developing anti-cancer regimens that are effective not only in mammalian models but also in patients. Here, we review studies using Drosophila as a tool to advance cancer study by complementing other traditional research systems.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila , Neoplasias , Animales , Humanos
3.
Nat Chem Biol ; 14(3): 291-298, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29355849

RESUMEN

Synthetic tailoring of approved drugs for new indications is often difficult, as the most appropriate targets may not be readily apparent, and therefore few roadmaps exist to guide chemistry. Here, we report a multidisciplinary approach for accessing novel target and chemical space starting from an FDA-approved kinase inhibitor. By combining chemical and genetic modifier screening with computational modeling, we identify distinct kinases that strongly enhance ('pro-targets') or limit ('anti-targets') whole-animal activity of the clinical kinase inhibitor sorafenib in a Drosophila medullary thyroid carcinoma (MTC) model. We demonstrate that RAF-the original intended sorafenib target-and MKNK kinases function as pharmacological liabilities because of inhibitor-induced transactivation and negative feedback, respectively. Through progressive synthetic refinement, we report a new class of 'tumor calibrated inhibitors' with unique polypharmacology and strongly improved therapeutic index in fly and human MTC xenograft models. This platform provides a rational approach to creating new high-efficacy and low-toxicity drugs.


Asunto(s)
Carcinoma Neuroendocrino/metabolismo , Carcinoma/metabolismo , Drosophila/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Tiroides/metabolismo , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Diseño de Fármacos , Femenino , Células HCT116 , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Simulación del Acoplamiento Molecular , Trasplante de Neoplasias , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal , Sorafenib/farmacología
4.
PLoS Comput Biol ; 15(4): e1006878, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31026276

RESUMEN

Drosophila provides an inexpensive and quantitative platform for measuring whole animal drug response. A complementary approach is virtual screening, where chemical libraries can be efficiently screened against protein target(s). Here, we present a unique discovery platform integrating structure-based modeling with Drosophila biology and organic synthesis. We demonstrate this platform by developing chemicals targeting a Drosophila model of Medullary Thyroid Cancer (MTC) characterized by a transformation network activated by oncogenic dRetM955T. Structural models for kinases relevant to MTC were generated for virtual screening to identify unique preliminary hits that suppressed dRetM955T-induced transformation. We then combined features from our hits with those of known inhibitors to create a 'hybrid' molecule with improved suppression of dRetM955T transformation. Our platform provides a framework to efficiently explore novel kinase inhibitors outside of explored inhibitor chemical space that are effective in inhibiting cancer networks while minimizing whole body toxicity.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Neuroendocrino , Evaluación Preclínica de Medicamentos/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas , Neoplasias de la Tiroides , Animales , Carcinoma Neuroendocrino/enzimología , Carcinoma Neuroendocrino/metabolismo , Biología Computacional/métodos , Drosophila , Modelos Biológicos , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/metabolismo , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/metabolismo
5.
Genes Cells ; 23(7): 580-589, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29749672

RESUMEN

Jellyfish green fluorescent protein (GFP) and firefly luciferase can serve as versatile tracking markers for identification and quantification of transplanted cancer cells in vivo. However, immune reactions against these markers can hamper the formation of syngraft tumors and metastasis that follows. Here, we report two transgenic (Tg) mouse lines that express nonfunctional mutant marker proteins, namely modified firefly luciferase (Luc2) or enhanced GFP (EGFP). These mice, named as Tg-mLuc2 and Tg-mEGFP, turned out to be immunologically tolerant to the respective tracking markers and thus efficiently accepted syngeneic cancer cells expressing the active forms of the markers. We then injected intrarectally the F1 hybrid Tg mice (BALB/c × C57BL/6J) with Colon-26 (C26) colon cancer cells that originated from a BALB/c mouse. Even when C26 cells expressed active Luc2 or EGFP, they formed primary tumors in the Tg mice with only 104 cells per mouse compared with more than 106 cells required in the nontransgenic BALB/c hosts. Furthermore, we detected metastatic foci of C26 cells in the liver and lungs of the Tg mice by tracking the specific reporter activities. These results show the usefulness of the Tg mouse lines as recipients for transplantation experiments with the non-self tracking marker-expressing cells.


Asunto(s)
Isoinjertos/metabolismo , Trasplante de Neoplasias/métodos , Animales , Proteínas Fluorescentes Verdes , Luciferasas , Proteínas Luminiscentes , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos/fisiología , Neoplasias
6.
Cancer Sci ; 108(4): 744-752, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28178391

RESUMEN

A major cause of cancer death is its metastasis to the vital organs. Few effective therapies are available for metastatic castration-resistant prostate cancer (PCa), and progressive metastatic lesions such as lymph nodes and bones cause mortality. We recently identified AES as a metastasis suppressor for colon cancer. Here, we have studied the roles of AES in PCa progression. We analyzed the relationship between AES expression and PCa stages of progression by immunohistochemistry of human needle biopsy samples. We then performed overexpression and knockdown of AES in human PCa cell lines LNCaP, DU145 and PC3, and determined the effects on proliferation, invasion and metastasis in culture and in a xenograft model. We also compared the PCa phenotypes of Aes/Pten compound knockout mice with those of Pten simple knockout mice. Expression levels of AES were inversely correlated with clinical stages of human PCa. Exogenous expression of AES suppressed the growth of LNCaP cells, whereas the AES knockdown promoted it. We also found that AES suppressed transcriptional activities of androgen receptor and Notch signaling. Notably, AES overexpression in AR-defective DU145 and PC3 cells reduced invasion and metastasis to lymph nodes and bones without affecting proliferation in culture. Consistently, prostate epithelium-specific inactivation of Aes in Ptenflox/flox mice increased expression of Snail and MMP9, and accelerated growth, invasion and lymph node metastasis of the mouse prostate tumor. These results suggest that AES plays an important role in controlling tumor growth and metastasis of PCa by regulating both AR and Notch signaling pathways.


Asunto(s)
Movimiento Celular/genética , Neoplasias de la Próstata/genética , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética , Anciano , Animales , Western Blotting , Línea Celular Tumoral , Proteínas Co-Represoras , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Ratones Transgénicos , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Trasplante Heterólogo , Proteínas Supresoras de Tumor/metabolismo
7.
Cancer Sci ; 107(11): 1622-1631, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27561171

RESUMEN

We recently found that the product of the AES gene functions as a metastasis suppressor of colorectal cancer (CRC) in both humans and mice. Expression of amino-terminal enhancer of split (AES) protein is significantly decreased in liver metastatic lesions compared with primary colon tumors. To investigate its downregulation mechanism in metastases, we searched for transcriptional regulators of AES in human CRC and found that its expression is reduced mainly by transcriptional dysregulation and, in some cases, by additional haploidization of its coding gene. The AES promoter-enhancer is in a typical CpG island, and contains a Yin-Yang transcription factor recognition sequence (YY element). In human epithelial cells of normal colon and primary tumors, transcription factor YY2, a member of the YY family, binds directly to the YY element, and stimulates expression of AES. In a transplantation mouse model of liver metastases, however, expression of Yy2 (and therefore of Aes) is downregulated. In human CRC metastases to the liver, the levels of AES protein are correlated with those of YY2. In addition, we noticed copy-number reduction for the AES coding gene in chromosome 19p13.3 in 12% (5/42) of human CRC cell lines. We excluded other mechanisms such as point or indel mutations in the coding or regulatory regions of the AES gene, CpG methylation in the AES promoter enhancer, expression of microRNAs, and chromatin histone modifications. These results indicate that Aes may belong to a novel family of metastasis suppressors with a CpG-island promoter enhancer, and it is regulated transcriptionally.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Islas de CpG/genética , Metástasis de la Neoplasia/genética , Regiones Promotoras Genéticas/genética , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proteínas Co-Represoras , Metilación de ADN/genética , Regulación hacia Abajo , Genes Supresores de Tumor , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Ratones , Mutación/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Elementos de Respuesta/genética , Factores de Transcripción/genética , Transcripción Genética/genética , Factor de Transcripción YY1/metabolismo
8.
Cancer Res ; 83(16): 2704-2715, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37378549

RESUMEN

Significant progress has been made in understanding the pathogenesis of pancreatic ductal adenocarcinoma (PDAC) by generating and using murine models. To accelerate drug discovery by identifying novel therapeutic targets on a systemic level, here we generated a Drosophila model mimicking the genetic signature in PDAC (KRAS, TP53, CDKN2A, and SMAD4 alterations), which is associated with the worst prognosis in patients. The '4-hit' flies displayed epithelial transformation and decreased survival. Comprehensive genetic screening of their entire kinome revealed kinases including MEK and AURKB as therapeutic targets. Consistently, a combination of the MEK inhibitor trametinib and the AURKB inhibitor BI-831266 suppressed the growth of human PDAC xenografts in mice. In patients with PDAC, the activity of AURKB was associated with poor prognosis. This fly-based platform provides an efficient whole-body approach that complements current methods for identifying therapeutic targets in PDAC. SIGNIFICANCE: Development of a Drosophila model mimicking genetic alterations in human pancreatic ductal adenocarcinoma provides a tool for genetic screening that identifies MEK and AURKB inhibition as a potential treatment strategy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Drosophila , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Aurora Quinasa B , Neoplasias Pancreáticas
9.
Lab Chip ; 23(6): 1561-1575, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36648503

RESUMEN

Circulating tumor cells (CTCs) are precursors to cancer metastasis. In blood circulation, they take various forms such as single CTCs, CTC clusters, and CTC-leukocyte clusters, all of which have unique characteristics in terms of physiological function and have been a subject of extensive research in the last several years. Unfortunately, conventional methods are limited in accurately analysing the highly heterogeneous nature of CTCs. Here we present an effective strategy for simultaneously analysing all forms of CTCs in blood by virtual-freezing fluorescence imaging (VIFFI) flow cytometry with 5-aminolevulinic acid (5-ALA) stimulation and antibody labeling. VIFFI is an optomechanical imaging method that virtually freezes the motion of fast-flowing cells on an image sensor to enable high-throughput yet sensitive imaging of every single event. 5-ALA stimulates cancer cells to induce the accumulation of protoporphyrin (PpIX), a red fluorescent substance, making it possible to detect all cancer cells even if they show no expression of the epithelial cell adhesion molecule, a typical CTC biomarker. Although PpIX signals are generally weak, VIFFI flow cytometry can detect them by virtue of its high sensitivity. As a proof-of-principle demonstration of the strategy, we applied cancer cells spiked in blood to the strategy to demonstrate image-based detection and accurate classification of single cancer cells, clusters of cancer cells, and clusters of a cancer cell(s) and a leukocyte(s). To show the clinical utility of our method, we used it to evaluate blood samples of four breast cancer patients and four healthy donors and identified EpCAM-positive PpIX-positive cells in one of the patient samples. Our work paves the way toward the determination of cancer prognosis, the guidance and monitoring of treatment, and the design of antitumor strategies for cancer patients.


Asunto(s)
Neoplasias de la Mama , Células Neoplásicas Circulantes , Humanos , Femenino , Células Neoplásicas Circulantes/patología , Citometría de Flujo , Ácido Aminolevulínico/farmacología , Congelación , Línea Celular Tumoral , Molécula de Adhesión Celular Epitelial , Neoplasias de la Mama/patología , Anticuerpos , Imagen Óptica , Biomarcadores de Tumor/metabolismo
10.
Front Oncol ; 12: 982751, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091180

RESUMEN

Cancer is one of the most severe health problems worldwide accounting for the second leading cause of death. Studies have indicated that cancers utilize different metabolic systems as compared with normal cells to produce extra energy and substances required for their survival, which contributes to tumor formation and progression. Recently, the fruit fly Drosophila has been attracting significant attention as a whole-body model for elucidating the cancer mechanisms including metabolism. This tiny organism offers a valuable toolkit with various advantages such as high genetic conservation and similar drug response to mammals. In this review, we introduce flies modeling for cancer patient genotypes which have pinpointed novel therapeutic targets and drug candidates in the salivary gland, thyroid, colon, lung, and brain. Furthermore, we introduce fly models for metabolic diseases such as diabetes mellitus, obesity, and cachexia. Diabetes mellitus and obesity are widely acknowledged risk factors for cancer, while cachexia is a cancer-related metabolic condition. In addition, we specifically focus on two cancer metabolic alterations: the Warburg effect and redox metabolism. Indeed, flies proved useful to reveal the relationship between these metabolic changes and cancer. Such accumulating achievements indicate that Drosophila offers an efficient platform to clarify the mechanisms of cancer as a systemic disease.

11.
Int J Clin Oncol ; 16(5): 464-72, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21847533

RESUMEN

Colorectal cancer is the second most common cancer, and is the third leading cause of cancer-related death in Japan. The majority of these deaths is attributable to liver metastasis. Recent studies have provided increasing evidence that the chemokine-chemokine receptor system is a potential mechanism of tumor metastasis via multiple complementary actions: (a) by promoting cancer cell migration, invasion, survival and angiogenesis; and (b) by recruiting distal stromal cells (i.e., myeloid bone marrow-derived cells) to indirectly facilitate tumor invasion and metastasis. Here, we discuss recent preclinical and clinical data supporting the view that chemokine pathways are potential therapeutic targets for liver metastasis of colorectal cancer.


Asunto(s)
Quimiocinas/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Receptores de Quimiocina/metabolismo , Movimiento Celular/genética , Quimiocinas/antagonistas & inhibidores , Quimiocinas/genética , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Terapia Molecular Dirigida , Células Mieloides/metabolismo , Células Mieloides/patología , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/genética , Microambiente Tumoral
12.
Cancer Res ; 62(23): 6846-9, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12460897

RESUMEN

Cyclooxygenase-2 (COX-2), the inducible COX isozyme, plays a key role in intestinal tumorigenesis. We have demonstrated recently that COX-2 protein is induced in the polyp stroma near the intestinal luminal surface in the Apc(Delta716) mouse, a model for human familial adenomatous polyposis, and stimulate tumor angiogenesis. However, the precise cell types that express COX-2 are still to be determined. By immunohistochemical analysis, here we show that the majority of COX-2-expressing cells in the intestinal polyps of Apc(Delta716) mice are fibroblasts and endothelial cells. Furthermore, the COX-2-expressing cells in human familial adenomatous polyposis polyps are also fibroblasts and endothelial cells. In contrast, bone marrow-derived cells such as macrophages and leukocytes express little COX-2 protein in the intestinal polyps. These results clearly indicate that fibroblasts and endothelial cells play important roles in polyp expansion by expressing COX-2, resulting in tumor angiogenesis.


Asunto(s)
Pólipos Intestinales/enzimología , Isoenzimas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Actinas/biosíntesis , Poliposis Adenomatosa del Colon/enzimología , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Animales , Antígenos CD34/biosíntesis , Antígenos de Diferenciación/biosíntesis , Ciclooxigenasa 2 , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Fibroblastos/enzimología , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Pólipos Intestinales/patología , Macrófagos/enzimología , Macrófagos/metabolismo , Proteínas de la Membrana , Ratones , Ratones Noqueados , Células del Estroma/enzimología , Células del Estroma/metabolismo , Vimentina/biosíntesis
13.
Cancer Res ; 63(16): 4872-7, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12941808

RESUMEN

Membrane arachidonic acid is converted by cyclooxygenase (COX) into prostaglandin (PG) G(2) and then to PGH(2) which is subsequently metabolized to PGE(2) by PGE synthase (PGES). Both COX-1 and COX-2 play critical roles in intestinal polyp formation, whereas COX-2 is also expressed in cancers of a variety of organs. Likewise, inducible microsomal PGES (mPGES-1) is expressed in several types of cancer, although its role in benign polyp formation has not been investigated. We demonstrated recently that most COX-2-expressing cells in the polyps are stromal fibroblasts. Here we show colocalization of COX-1, COX-2 and mPGES in the intestinal polyp stromal fibroblasts of Apc(Delta 716) mice, a model for familial adenomatous polyposis. Contrary to COX-2 that was induced only in polyps >1 mm in diameter, COX-1 was found in polyps of any size. In polyps >1 mm, not only COX-2 but also mPGES was induced in the stromal fibroblasts where COX-1 had already been expressed. Although polyp number and size were markedly reduced in COX-1 (-/-) or COX-2 (-/-) compound mutant Apc mice, both COX-2 and mPGES were induced in the COX-1 (-/-) polyps, whereas COX-1 was expressed in the COX-2 (-/-) polyps. We found also in human familial adenomatous polyposis polyps that COX-2 and mPGES were induced in the COX-1-expressing fibroblasts. On the basis of these results, we propose that COX-1 expression in the stromal cells secures the basal level of PGE(2) that can support polyp growth to approximately 1 mm, and that simultaneous inductions of COX-2 and mPGES support the polyp expansion beyond approximately 1 mm by boosting the stromal PGE(2) production.


Asunto(s)
Pólipos Intestinales/enzimología , Isoenzimas/fisiología , Prostaglandina-Endoperóxido Sintasas/fisiología , Animales , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inducción Enzimática , Oxidorreductasas Intramoleculares/biosíntesis , Proteínas de la Membrana , Ratones , Prostaglandina-E Sintasas , Células del Estroma/enzimología
14.
Cancer Res ; 64(11): 4010-7, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15173015

RESUMEN

Chemokines and their receptors play key roles in leukocyte trafficking and are also implicated in cancer metastasis to specific organs. Here we show that mouse B16F10 melanoma cells constitutively express chemokine receptor CXCR3, and that its ligands CXCL9/Mig, CXCL10/IP-10, and CXCL11/I-TAC induce cellular responses in vitro, such as actin polymerization, migration, invasion, and cell survival. To determine whether CXCR3 could play a role in metastasis to lymph nodes (LNs), we constructed B16F10 cells with reduced CXCR3 expression by antisense RNA and investigated their metastatic activities after s.c. inoculations to syngeneic hosts, C57BL/6 mice. The metastatic frequency of these cells to LNs was markedly reduced to approximately 15% (P < 0.05) compared with the parental or empty vector-transduced cells. On the other hand, pretreatment of mice with complete Freund's adjuvant increased the levels of CXCL9 and CXCL10 in the draining LNs, which caused 2.5-3.0-fold increase (P < 0.05) in the metastatic frequency of B16F10 cells to the nodes with much larger foci. Importantly, such a stimulation of metastasis was largely suppressed when CXCR3 expression in B16F10 cells was reduced by antisense RNA or when mice were treated with specific antibodies against CXCL9 and CXCL10. We also demonstrate that CXCR3 is expressed on several human melanoma cell lines as well as primary human melanoma tissues (5 of 9 samples tested). These results suggest that CXCR3 inhibitors may be promising therapeutic agents for treatment of LN metastasis, including that of melanoma.


Asunto(s)
Ganglios Linfáticos/patología , Melanoma Experimental/patología , Receptores de Quimiocina/fisiología , Actinas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/genética , Citoesqueleto/metabolismo , Adhesiones Focales/fisiología , Adyuvante de Freund/farmacología , Humanos , Metástasis Linfática , Melanoma/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Invasividad Neoplásica , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores CXCR3 , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/genética , Transfección
15.
J Biochem ; 159(1): 133-40, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26229111

RESUMEN

Amino-terminal enhancer of split (Aes) is a member of Groucho/Transducin-like enhancer (TLE) family. Aes is a recently found metastasis suppressor of colorectal cancer (CRC) that inhibits Notch signalling, and forms nuclear foci together with TLE1. Although some Notch-associated proteins are known to form subnuclear bodies, little is known regarding the dynamics or functions of these structures. Here, we show that Aes nuclear foci in CRC observed under an electron microscope are in a rather amorphous structure, lacking surrounding membrane. Investigation of their behaviour during the cell cycle by time-lapse cinematography showed that Aes nuclear foci dissolve during mitosis and reassemble after completion of cytokinesis. We have also found that heat shock cognate 70 (HSC70) is an essential component of Aes foci. Pharmacological inhibition of the HSC70 ATPase activity with VER155008 reduces Aes focus formation. These results provide insight into the understanding of Aes-mediated inhibition of Notch signalling.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Núcleo Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas Represoras/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Animales , Núcleo Celular/ultraestructura , Proteínas Co-Represoras , Citocinesis , Células HCT116 , Células HEK293 , Proteínas del Choque Térmico HSC70/antagonistas & inhibidores , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Mitosis , Nucleósidos de Purina/farmacología , Receptores Notch/metabolismo , Proteínas Represoras/genética , Transducción de Señal , Imagen de Lapso de Tiempo
16.
Biochim Biophys Acta ; 1585(2-3): 72-6, 2002 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-12531539

RESUMEN

Phospolipase A(2) (PLA(2)) is the esterase activity that cleaves the sn-2 ester bond in glycerophospholipids, releasing free fatty acids and lysophospholipids. The PLA(2) activity is found in a variety of enzymes which can be divided in several types based on their Ca(2+) dependence for their activity; Ca(2+)-dependent secretory phosholipases (sPLA(2)s) and cytosolic phospholipases (cPLA(2)s), and Ca(2+)-independent phospholipase A(2)s (iPLA(2)s). These enzymes also show diverse size and substrate specificity (i.e., in the fatty acid chain length and extent of saturation). Among the fatty acids released by PLA(2), arachidonic acid (AA) is of particular biological importance, because it is subsequently converted to prostanoids and leukotrienes by cyclooxygenases (COX) and lipoxygenases (LOX), respectively. Free AA may also stimulate apoptosis through activation of sphingomyelinase. Alternatively, it is suggested that oxidized metabolites generated from AA by LOX induce apoptosis. Although the precise mechanisms remain to be elucidated, changes are observed in glycerolipid metabolism during apoptotic processes. In some cells induced to undergo apoptosis, AA is released concomitant with loss of cell viability, caspase activation and DNA fragmentation. Such AA releases appear to be mediated by activation of cPLA(2) and/or iPLA(2). For example, tumor necrosis factor-alpha (TNF-alpha)-induced cell death is mediated by cPLA(2), whereas Fas-induced apoptosis appears to be mediated by iPLA(2). Some discrepancies among early experimental results were probably caused by differences in the experimental conditions such as the serum concentration, inhibitors used that are not necessarily specific to a single-type enzyme, or differential expression of each PLA(2) in cells employed in the experiments. Recent studies eliminated such problems, by carefully defining the experimental conditions, and using multiple inhibitors that show different specificities. Accordingly, more convincing data are available that demonstrate involvement of some PLA(2)s in the apoptotic processes. In addition to cPLA(2) and iPLA(2), sPLA(2)s were recently found to play roles in apoptosis. Moreover, new proteins that appear to control PLA(2)s are being discovered. Here, the roles of PLA(2)s in apoptosis are discussed by reviewing recent reports.


Asunto(s)
Apoptosis/fisiología , Fosfolipasas A/fisiología , Animales , Línea Celular , Citosol/enzimología , Humanos , Isoenzimas/fisiología , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A/clasificación , Receptores de Superficie Celular/metabolismo , Factor de Necrosis Tumoral alfa
17.
Cancer Discov ; 5(2): 198-211, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25432929

RESUMEN

UNLABELLED: We have recently identified a metastasis suppressor gene for colorectal cancer: AES/Aes, which encodes an endogenous inhibitor of NOTCH signaling. When Aes is knocked out in the adenomatous epithelium of intestinal polyposis mice, their tumors become malignant, showing marked submucosal invasion and intravasation. Here, we show that one of the genes induced by NOTCH signaling in colorectal cancer is DAB1/Dab1. Genetic depletion of DAB1 suppresses cancer invasion and metastasis in the NOTCH signaling-activated mice. DAB1 is phosphorylated by ABL tyrosine kinase, which activates ABL reciprocally. Consistently, inhibition of ABL suppresses cancer invasion in mice. Furthermore, we show that one of the targets of ABL is the RAC/RHOGEF protein TRIO, and that phosphorylation at its Tyr residue 2681 (pY2681) causes RHO activation in colorectal cancer cells. Its unphosphorylatable mutation TRIO Y2681F reduces RHOGEF activity and inhibits invasion of colorectal cancer cells. Importantly, TRIO pY2681 correlates with significantly poorer prognosis of patients with colorectal cancer after surgery. SIGNIFICANCE: These results indicate that TRIO pY2681 is one of the downstream effectors of NOTCH signaling activation in colorectal cancer, and can be a prognostic marker, helping to determine the therapeutic modality of patients with colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Notch/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias Colorrectales/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Ratones , Metástasis de la Neoplasia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Receptores Notch/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Transducción de Señal
18.
Cancer Cell ; 19(1): 125-37, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21251616

RESUMEN

Metastasis is responsible for most cancer deaths. Here, we show that Aes (or Grg5) gene functions as an endogenous metastasis suppressor. Expression of Aes was decreased in liver metastases compared with primary colon tumors in both mice and humans. Aes inhibited Notch signaling by converting active Rbpj transcription complexes into repression complexes on insoluble nuclear matrix. In tumor cells, Notch signaling was triggered by ligands on adjoining blood vessels, and stimulated transendothelial migration. Genetic depletion of Aes in Apc(Δ716) intestinal polyposis mice caused marked tumor invasion and intravasation that were suppressed by Notch signaling inhibition. These results suggest that inhibition of Notch signaling can be a promising strategy for prevention and treatment of colon cancer metastasis.


Asunto(s)
Neoplasias del Colon/patología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Benzodiazepinonas/farmacología , Benzodiazepinonas/uso terapéutico , Línea Celular Tumoral , Proteínas Co-Represoras , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Regulación hacia Abajo/genética , Expresión Génica/genética , Silenciador del Gen/fisiología , Células HCT116 , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Poliposis Intestinal/tratamiento farmacológico , Poliposis Intestinal/metabolismo , Poliposis Intestinal/patología , Ligandos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Desnudos , Ratones Transgénicos , Modelos Biológicos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Matriz Nuclear/metabolismo , Receptor Notch1/metabolismo , Receptores Notch/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma/metabolismo , Factores de Transcripción/genética , Migración Transendotelial y Transepitelial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA