Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Eur J Nucl Med Mol Imaging ; 49(5): 1574-1583, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34837510

RESUMEN

PURPOSE: Paediatric high-risk neuroblastoma has poor prognosis despite modern multimodality therapy. This phase I/II study aimed to determine the safety, dose-limiting toxicity (DLT), and efficacy of high-dose 131I-meta-iodobenzylguanidine (131I-mIBG) therapy combined with single high-dose chemotherapy (HDC) and haematopoietic stem cell transplantation (HSCT) in high-risk neuroblastoma in Japan. METHODS: Patients received 666 MBq/kg of 131I-mIBG and single HDC and HSCT from autologous or allogeneic stem cell sources. The primary endpoint was DLT defined as adverse events associated with 131I-mIBG treatment posing a significant obstacle to subsequent HDC. The secondary endpoints were adverse events/reactions, haematopoietic stem cell engraftment and responses according to the Response Evaluation Criteria in Solid Tumours version 1.1 (RECIST 1.1) and 123I-mIBG scintigraphy. Response was evaluated after engraftment. RESULTS: We enrolled eight patients with high-risk neuroblastoma (six females; six newly diagnosed and two relapsed high-risk neuroblastoma; median age, 4 years; range, 1-10 years). Although all patients had adverse events/reactions after high-dose 131I-mIBG therapy, we found no DLT. Adverse events and reactions were observed in 100% and 25% patients during single HDC and 100% and 12.5% patients during HSCT, respectively. No Grade 4 complications except myelosuppression occurred during single HDC and HSCT. The response rate according to RECIST 1.1 was observed in 87.5% (7/8) in stable disease and 12.5% (1/8) were not evaluated. Scintigraphic response occurred in 62.5% (5/8) and 37.5% (3/8) patients in complete response and stable disease, respectively. CONCLUSION: 131I-mIBG therapy with 666 MBq/kg followed by single HDC and autologous or allogeneic SCT is safe and efficacious in patients with high-risk neuroblastoma and has no DLT. TRIAL REGISTRATION NUMBER: jRCTs041180030. NAME OF REGISTRY: Feasibility of high-dose iodine-131-meta-iodobenzylguanidine therapy for high-risk neuroblastoma preceding myeloablative chemotherapy and haematopoietic stem cell transplantation (High-dose iodine-131-meta-iodobenzylguanidine therapy for high-risk neuroblastoma). URL OF REGISTRY: https://jrct.niph.go.jp/en-latest-detail/jRCTs041180030 . DATE OF ENROLMENT OF THE FIRST PARTICIPANT TO THE TRIAL: 12/01/2018.


Asunto(s)
3-Yodobencilguanidina , Neuroblastoma , 3-Yodobencilguanidina/administración & dosificación , 3-Yodobencilguanidina/efectos adversos , Niño , Preescolar , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Radioisótopos de Yodo , Masculino , Neuroblastoma/radioterapia , Trasplante Autólogo
2.
Semin Cancer Biol ; 27: 30-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24657939

RESUMEN

Galectin-3, a member of ß-galactoside-binding gene family is a multi-functional protein, which regulates pleiotropic biological functions such as cell growth, cell adhesion, cell-cell interactions, apoptosis, angiogenesis and mRNA processing. Its unique structure enables it to interact with a plethora of ligands in a carbohydrate dependent or independent manner. Galectin-3 is mainly a cytosolic protein, but can easily traverse the intracellular and plasma membranes to translocate into the nucleus, mitochondria or get externalized. Depending on the cell type, specific experimental conditions in vitro, cancer type and stage, galectin-3 has been reported to be exclusively cytoplasmic, predominantly nuclear or distributed between the two compartments. In this review we have summarized the dynamics of galectin-3 shuttling between the nucleus and the cytoplasm, the nuclear transport mechanisms of galectin-3, how its specific interactions with the members of ß-catenin signaling pathways affect tumor progression, and its implications as a therapeutic target.


Asunto(s)
Galectina 3/metabolismo , Neoplasias/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Galectina 3/antagonistas & inhibidores , Galectina 3/química , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Glycobiology ; 24(10): 886-91, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25138305

RESUMEN

Galectin-3 is a member of the family of ß-galactoside-binding lectins characterized by evolutionarily conserved sequences defined by structural similarities in their carbohydrate-recognition domains. Galectin-3 is a unique, chimeric protein consisting of three distinct structural motifs: (i) a short NH2 terminal domain containing a serine phosphorylation site; (ii) a repetitive proline-rich collagen-α-like sequence cleavable by matrix metalloproteases; and (iii) a globular COOH-terminal domain containing a carbohydrate-binding motif and an NWGR anti-death motif. It is ubiquitously expressed and has diverse biological functions depending on its subcellular localization. Galectin-3 is mainly found in the cytoplasm, also seen in the nucleus and can be secreted by non-classical, secretory pathways. In general, secreted galectin-3 mediates cell migration, cell adhesion and cell-cell interactions through the binding with high affinity to galactose-containing glycoproteins on the cell surface. Cytoplasmic galectin-3 exhibits anti-apoptotic activity and regulates several signal transduction pathways, whereas nuclear galectin-3 has been associated with pre-mRNA splicing and gene expression. Its unique chimeric structure enables it to interact with a plethora of ligands and modulate diverse functions such as cell growth, adhesion, migration, invasion, angiogenesis, immune function, apoptosis and endocytosis emphasizing its significance in the process of tumor progression. In this review, we have focused on the role of galectin-3 in tumor metastasis with special emphasis on angiogenesis.


Asunto(s)
Carcinogénesis/genética , Galectina 3/genética , Neoplasias/genética , Neovascularización Patológica/genética , Apoptosis/genética , Proteínas Sanguíneas , Adhesión Celular , Galectina 3/química , Galectina 3/metabolismo , Galectinas , Humanos , Metástasis de la Neoplasia , Neoplasias/patología , Precursores del ARN/genética , Empalme del ARN/genética , Transducción de Señal
4.
Biochem Biophys Res Commun ; 434(1): 155-61, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23541576

RESUMEN

Nucleoporin Nup98 is a component of the nuclear pore complex, and is important in transport across the nuclear pore. Many studies implicate nucleoporin in cancer progression, but no direct mechanistic studies of its effect in cancer have been reported. We show here that Nup98 specifically regulates nucleus-cytoplasm transport of galectin-3, which is a ß-galactoside-binding protein that affects adhesion, migration, and cancer progression, and controls cell growth through the ß-catenin signaling pathway in cancer cells. Nup98 interacted with galectin-3 on the nuclear membrane, and promoted galectin-3 cytoplasmic translocation whereas other nucleoporins did not show these functions. Inversely, silencing of Nup98 expression by siRNA technique localized galectin-3 to the nucleus and retarded cell growth, which was rescued by Nup98 transfection. In addition, Nup98 RNA interference significantly suppressed downstream mRNA expression in the ß-catenin pathway, such as cyclin D1 and FRA-1, while nuclear galectin-3 binds to ß-catenin to inhibit transcriptional activity. Reduced expression of ß-catenin target genes is consistent with the Nup98 reduction and the galectin-3-nucleus translocation rate. Overall, the results show Nup98's involvement in nuclear-cytoplasm translocation of galectin-3 and ß-catenin signaling pathway in regulating cell proliferation, and the results depicted here suggest a novel therapeutic target/modality for cancers.


Asunto(s)
Galectina 3/metabolismo , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Transporte Activo de Núcleo Celular/genética , Proliferación Celular , Progresión de la Enfermedad , Células HeLa , Humanos , Células MCF-7 , Neoplasias/metabolismo , Neoplasias/patología , Proteínas de Complejo Poro Nuclear/antagonistas & inhibidores , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/fisiología , Dominios y Motivos de Interacción de Proteínas , ARN Interferente Pequeño/genética , Transducción de Señal , beta Catenina/metabolismo
5.
Cancer Metastasis Rev ; 30(2): 239-51, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21298575

RESUMEN

One of the main reasons for cancer mortality is caused by the highly invasive behavior of cancer cells, which often due to aggressive metastasis. Metastasis is mediated by various growth factors and cytokines, operating through numerous signaling pathways. Remarkably, all these metastatic signaling pathways must enter the nucleus through a single gatekeeper, the nuclear pore complex (NPC). NPCs are the only gateway between the cytoplasm and the nucleus. NPCs are among the largest proteinaceous assemblies in the cell and are composed of multiple copies of around 30 different proteins called nucleoporins. Here, we review what is currently known about the NPC, and its role in the mechanisms of tumor progression. We will also explore potential strategies to target metastatic pathways by manipulating the karyopherins (importins/exportins) of nucleocytoplasmic traffic through NPCs.


Asunto(s)
Neoplasias/metabolismo , Poro Nuclear/metabolismo , Transducción de Señal , Transporte Activo de Núcleo Celular , Animales , Humanos , Modelos Biológicos , Metástasis de la Neoplasia , Neoplasias/patología , Proteínas de Complejo Poro Nuclear/metabolismo , Microambiente Tumoral
6.
J Biol Chem ; 285(14): 10841-9, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20133940

RESUMEN

Gain or loss of whole chromosomes is often observed in cancer cells and is thought to be due to aberrant chromosome segregation during mitosis. Proper chromosome segregation depends on a faithful interaction between spindle microtubules and kinetochores. Several components of the nuclear pore complex/nucleoporins play critical roles in orchestrating the rapid remodeling events that occur during mitosis. Our recent studies revealed that the nucleoporin, Rae1, plays critical roles in maintaining spindle bipolarity. Here, we show association of another nucleoporin, termed Tpr (translocated promoter region), with the molecular motors dynein and dynactin, which both orchestrate with the spindle checkpoints Mad1 and Mad2 during cell division. Overexpression of Tpr enhanced multinucleated cell formation. RNA interference-mediated knockdown of Tpr caused a severe lagging chromosome phenotype and disrupted spindle checkpoint proteins expression and localization. Next, we performed a series of rescue and dominant negative experiments to confirm that Tpr orchestrates proper chromosome segregation through interaction with dynein light chain. Our data indicate that Tpr functions as a spatial and temporal regulator of spindle checkpoints, ensuring the efficient recruitment of checkpoint proteins to the molecular motor dynein to promote proper anaphase formation.


Asunto(s)
Anafase/fisiología , Segregación Cromosómica , Dineínas/metabolismo , Metafase/fisiología , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Western Blotting , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Dineínas Citoplasmáticas/genética , Dineínas Citoplasmáticas/metabolismo , Dineínas/antagonistas & inhibidores , Dineínas/genética , Células HeLa , Humanos , Técnicas para Inmunoenzimas , Cinetocoros , Proteínas Mad2 , Ratones , Células 3T3 NIH , Proteínas de Complejo Poro Nuclear/antagonistas & inhibidores , Proteínas de Complejo Poro Nuclear/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Huso Acromático
7.
Cancer Res ; 67(9): 4236-43, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483335

RESUMEN

Phosphoglucose isomerase (PGI) is one of the glycolytic enzymes and is a multifunctional enzyme that functions in glucose metabolism inside the cell while acting as a cytokine outside the cell, with properties that include autocrine motility factor (AMF) regulating tumor cell motility. Although there are many studies indicating that PGI/AMF has been implicated in progression of metastasis, no direct studies of the significance of exogenous PGI/AMF on tumor progression have been reported. Here, we report on the mesenchymal-to-epithelial transition (MET), which is the reverse phenomenon of the epithelial-to-mesenchymal transition that is associated with loss of cell polarity, loss of epithelia markers, and enhancement of cell motility essential for tumor cell invasion and metastasis. Mesenchymal human fibrosarcoma HT1080 cells, which have naturally high levels of endogenous and exogenous PGI/AMF, were stably transfected with PGI/AMF small interfering RNA (siRNA). The siRNA targeting human PGI/AMF down-regulated the endogenous PGI/AMF expression and completely extinguished the secretion of PGI/AMF in a human fibrosarcoma HT1080, whereas the control siRNA showed no effects. The PGI/AMF siRNA caused cells to change shape dramatically and inhibited cell motility and invasion markedly. Suppression of PGI/AMF led to a contact-dependent inhibition of cell growth. Those PGI/AMF siRNA-transfected cells showed epithelial phenotype. Furthermore, tumor cells with PGI/AMF deficiency lost their abilities to form tumor mass. This study identifies that MET in HT1080 human lung fibrosarcoma cells was initiated by down-regulation of the housekeeping gene product/cytokine PGI/AMF, and the results depicted here suggest a novel therapeutic target/modality for mesenchymal cancers.


Asunto(s)
Fibrosarcoma/enzimología , Fibrosarcoma/patología , Glucosa-6-Fosfato Isomerasa/biosíntesis , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , ADN de Neoplasias/biosíntesis , Regulación hacia Abajo , Células Epiteliales/patología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glucosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Glucosa-6-Fosfato Isomerasa/genética , Humanos , Mesodermo/patología , Invasividad Neoplásica , ARN Interferente Pequeño/genética , Transfección
8.
Cancer Res ; 67(18): 8682-9, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875708

RESUMEN

Phosphoglucose isomerase (PGI; EC 5.3.1.9) is a ubiquitous cytosolic enzyme essential for glycolysis and gluconeogenesis. PGI is a multifunctional dimeric protein that extracellularly acts as a cytokine [autocrine motility factor (AMF)] eliciting mitogenic, motogenic, and differentiation functions through binding to its cell surface receptor gp78/AMF receptor (AMFR). AMFR contains a seven-transmembrane domain with RING-H2 and leucine zipper motifs showing ubiquitin protein ligase (E3) activity and is exposed on the endoplasmic reticulum surface. Augmented expressions of both PGI/AMF and AMFR have been implicated in tumor progression and metastasis, and an intracellular binding partner of PGI/AMF is expected to regulate in part its diverse biological functions. Thus, we screened a cDNA library using a yeast two-hybrid system to search for interacting protein(s) and report on the finding of poly(ADP-ribose) polymerase-14 (PARP-14) to be a binding partner with PGI/AMF. PARP-14-PGI/AMF interaction was confirmed by coimmunoprecipitation and immunolocalization. We also report that PGI/AMF degradation is mainly regulated by the ubiquitin-lysosome system and RNA interference experiments revealed that PARP-14 inhibits PGI/AMF ubiquitination, thus contributing to its stabilization and secretion. This newly characterized PARP-14 protein should assist in understanding the regulation of PGI/AMF intracellular function(s) and may provide a new therapeutic target for inhibition of PGI/AMF inducing tumor cell migration and invasion during metastasis.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias Óseas/enzimología , Neoplasias del Colon/enzimología , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Células Endoteliales/enzimología , Fibrosarcoma/enzimología , Células HT29 , Humanos , Osteosarcoma/enzimología , Poli(ADP-Ribosa) Polimerasas/genética
9.
FEBS Lett ; 582(13): 1877-82, 2008 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-18485900

RESUMEN

The autocrine motility factor (AMF) is a multifunctional protein that is involved in tumor progression including enhanced invasiveness via induction of matrix metalloproteinase-3 (MMP3). The increase in MMP3 was found in an AMF-high production tumor cell line, and c-Jun, c-Fos and mitogen-activated protein kinases (MAPKs) were also highly phosphorylated compared with the parent line. AMF stimulation induced the rapid phosphorylation of the cellular MAPK cascade and MMP3 secretion, which was blocked using a specific MAPK inhibitor. Results of this study suggest that AMF stimulation stimulates MMP3 expression via a MAPK signaling pathway.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Invasividad Neoplásica , Línea Celular Tumoral , Movimiento Celular , Activación Enzimática , Glucosa-6-Fosfato Isomerasa/farmacología , Humanos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Metaloproteinasa 3 de la Matriz/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores del Factor Autocrino de Motilidad , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba
10.
J Mol Biol ; 358(3): 741-53, 2006 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-16563432

RESUMEN

The autocrine motility factor (AMF) promotes cellular locomotion or invasion, and regulates tumor angiogenesis or ascites accumulation. These signals are triggered by binding between AMF and its receptor (AMFR), a glycoprotein on the cell surface. AMF has been identified as phosphohexose isomerase (PHI). Previous reports have suggested that the substrate-recognition of exo-PHI is significant for receptor binding. Crystallographic studies have shown that AMF consists of three domains, and that the substrate or inhibitor of PHI is stored between the large and small domains, corresponding to approximately residues 117-288. Here, site-directed mutagenesis was used to investigate 18 recombinant human AMF point mutants involving critical amino acid residues for substrate or enzyme inhibitor recognition or binding. Mutation of residues that interact with the phosphate group of the PHI substrate significantly reduced the cell motility-stimulating activity. Their binding capacities for AMFR were also lower than wild-type human AMF. Mutants that retained the enzymic activity showed the motility-stimulating effect and receptor binding and had sensitivity to a PHI inhibitor. Mutant AMFR lacking the N-sugar chain was expressed on the cell membrane but did not respond to AMF-stimulation, and N-glycosidase-treated AMFR did not compete with receptor binding of AMF. Furthermore, the AMF domains that contain the substrate storage domain and C-terminal region stimulate cell locomotion. These results suggest that the N-glyco side-chain of AMFR is a trigger and that interaction between the 117-C-terminal part of AMF and the extracellular core protein of AMFR is needed during AMF-AMFR interactions.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/metabolismo , Glucosa/metabolismo , Receptores de Citocinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Glucosa-6-Fosfato Isomerasa/química , Glucosa-6-Fosfato Isomerasa/genética , Humanos , Modelos Moleculares , Unión Proteica , Estructura Cuaternaria de Proteína , Receptores del Factor Autocrino de Motilidad , Receptores de Citocinas/química , Receptores de Citocinas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ubiquitina-Proteína Ligasas
11.
J Mol Biol ; 356(2): 312-24, 2006 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-16375918

RESUMEN

Autocrine motility factor (AMF), a tumor-secreted cytokine, stimulates cell migration in vitro and metastasis in vivo. AMF is identical to the extracellular cytokines neuroleukin and maturation factor and, interestingly, to the intracellular enzyme phosphoglucose isomerase. The cytokine activity of AMF is inhibited by carbohydrate phosphate compounds as they compete for AMF binding with the carbohydrate moiety of the AMF receptor (AMFR), which is a glycosylated seven transmembrane helix protein. Here, we report the first comprehensive high-resolution crystal structure analyses of the inhibitor-free form and the eight types of inhibitor (phosphate, erythrose 4-phosphate (E4P), arabinose 5-phosphate (A5P), sorbitol 6-phosphate (S6P), 6-phosphogluconic acid (6PGA), fructose 6-phosphate (F6P), glucose 6-phosphate (G6P), or mannose 6-phosphate (M6P)) complexes of mouse AMF (mAMF). We assayed the inhibitory activities of these inhibitors against the cytokine activity of mAMF. The inhibitory activities of the six-carbon sugars (G6P, F6P, M6P, and 6PGA) were found to be significantly higher than those of the four or five-carbon sugars (E4P or A5P). The inhibitory activities clearly depend on the length of the inhibitor molecules. A structural comparison revealed that a water-mediated hydrogen bond between one end of the inhibitor and a rigid portion of the protein surface in the shorter-chain inhibitor (E4P) complex is replaced by a direct hydrogen bond in the longer-chain inhibitor (6PGA) complex. Thus, to obtain a new compound with higher inhibitory activities against AMF, water molecules at the inhibitor binding site of AMF should be replaced by a functional group of inhibitors in order to introduce direct interactions with the protein surface. The present structure-activity relationship studies will be valuable not only for designing more effective AMF inhibitors but also for studying general protein-inhibitor interactions.


Asunto(s)
Carbohidratos/química , Glucosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Glucosa-6-Fosfato Isomerasa/química , Fosfatos/química , Animales , Sitios de Unión , Movimiento Celular , Cristalografía por Rayos X , Glucosa-6-Fosfato Isomerasa/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Relación Estructura-Actividad
12.
FASEB J ; 19(11): 1422-30, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16126909

RESUMEN

Phosphoglucose isomerase (PGI; EC 5.3.1.9) is a housekeeping cytosolic enzyme of the sugar metabolism pathways that plays a key role in glycolysis and gluconeogenesis. PGI is a multifunctional dimeric protein that extracellularly acts as a cytokine with properties that include autocrine motility factor (AMF) eliciting mitogenic, motogenic, differentiation functions and has been implicated in tumor progression and metastasis. Since metastasis is regulated in part by hypoxia, which induces the transcription of metastasis-associated genes and anaerobic glycolic metabolism, we questioned whether hypoxia also regulates the expression level of tumor cells' PGI/AMF. We establish here that in the human breast carcinoma BT-549 cells hypoxia enhanced expression of the transcription factor hypoxia-inducible factor (HIF)-1, which in turn led to the up-regulation of PGI/AMF expression and was specifically inhibited by inhibitors of the phosphatidylinositol 3'-kinase signaling pathway. In addition, the hypoxia induction of PGI/AMF expression was suppressed by inhibitors of vascular endothelial growth factor (VEGF) or VEGF receptors, suggesting that hypoxia-inducible VEGF regulates the PGI/AMF expression. Hypoxia also enhanced cancer cell motility, and these effects were strongly inhibited by the PGI/AMF, VEGF, or VEGF receptor inhibitors. The results presented here suggest that under hypoxic conditions the expression of PGI/AMF is regulated in part by the HIF pathway, which in turn increases the flow of the glycolytic cascade leading to an increased anaerobic energy generation; thus, inhibition of PGI/AMF expression and activities may provide a new therapeutic modality for treatment of hypoxic tumors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glucosa-6-Fosfato Isomerasa/genética , Hipoxia/metabolismo , Línea Celular Tumoral , Movimiento Celular , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Indazoles/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/biosíntesis
13.
Leuk Lymphoma ; 47(10): 2234-43, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17071500

RESUMEN

Autocrine Motility Factor (AMF)/maturation factor (MF)/neuroleukin (NLK) is a multifunctional protein, which acts as a glucose 6-phosphate isomerase (G6PI) intracellularly. Exto-G6PI stimulates invasion and metastasis of tumor cells, neurotropic growth and differentiation of leukemic cells. The cell motility and proliferation receptor is known to be gp78 (78 kilo-Dalton glycoprotein), which has seven transmembrane domains in its N-terminal region, but the maturation factor receptor remains unclear. The human acute monocytic leukemia line does not express gp78 and its motile activity is not enhanced by AMF though it is well differentiated by AMF exposure. The forced expression of gp78 in leukemic cells recovered acceptable motile stimulation, concomitant with reduced differentiation ability. Two unknown proteins were detected by crosslinking between AMF and leukemic cells. The results of this report suggest that the receptor molecule for AMF/NLK/MF in leukemic differentiation is not gp78.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/fisiología , Receptores de Citocinas/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , ADN/metabolismo , Glucosa-6-Fosfato Isomerasa/metabolismo , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Plásmidos/metabolismo , Receptores del Factor Autocrino de Motilidad , Receptores de Citocinas/metabolismo , Transfección , Células U937 , Ubiquitina-Proteína Ligasas/metabolismo
14.
J Mol Biol ; 318(4): 985-97, 2002 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-12054796

RESUMEN

Autocrine motility factor (AMF), a tumor-secreted cytokine, stimulates cell migration in vitro and metastasis in vivo. AMF is genetically identical with the extracellular cytokines neuroleukin (NLK) and maturation factor (MF) and, interestingly, the intracellular enzyme phosphohexose isomerase (PHI). The crystal structures of the inhibitor-free open form and the inhibitor (erythrose 4-phosphate, E4P, a strong inhibitor of AMF's cytokine activity)-bound closed form of human AMF have been determined at 1.9 A and 2.4 A resolution, respectively. Upon E4P binding, local conformation changes (open to closed) occur around the inhibitor-binding site. The E4P-bound structure shows that the location of the inhibitor (of cytokine activity) binding site of human AMF is very similar to those of the inhibitor (of enzymatic activity) binding sites of PHIs. The present study shows clearly that there is structural overlap of the regions responsible for the enzymatic and cytokine functions of AMF and PHI and suggests two scenarios for the inhibition mechanism of cytokine activity of AMF by the carbohydrate phosphate group. One likely scenario is that the compound could compete for AMF binding with the carbohydrate moiety of the AMF receptor (AMFR), which is a glycosylated seven-transmembrane helix protein. The other scenario is that the local conformation changes upon inhibitor binding may affect the AMF-AMFR interactions. To examine roles of the residues in the inhibitor-binding site, two mutant AMFs were prepared. Replacements of His389, which is hydrogen-bonded to the hydroxyl group of E4P by Phe, and Thr215, which is hydrogen-bonded to the phosphate group of E4P by Asp, result in mutant AMFs that are impaired in cytokine activity. These results suggest a role for these amino acids in recognition of a carbohydrate moiety of the AMFR. Since the E4P is one of the smallest compounds having AMF inhibitor activity, knowledge of the present crystal structure would provide an insight into the lead compound design of more effective AMF inhibitors.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Glucosa-6-Fosfato Isomerasa/química , Fosfatos de Azúcar/farmacología , Sitios de Unión , Western Blotting , Dicroismo Circular , Reactivos de Enlaces Cruzados , Cristalografía por Rayos X , Glucosa-6-Fosfato Isomerasa/genética , Humanos , Modelos Moleculares , Estructura Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Especificidad por Sustrato , Células Tumorales Cultivadas
15.
Sci Rep ; 2: 878, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23170199

RESUMEN

The nuclear pore complex (NPC) consists of a conserved set of ~30 different proteins, termed nucleoporins, and serves as a gateway for the exchange of materials between the cytoplasm and nucleus. Tpr (translocated promoter region) is a component of NPC that presumably localizes at intranuclear filaments. Here, we show that Tpr knockdown caused a severe reduction in the number of nuclear pores. Furthermore, our electron microscopy studies indicated a significant reduction in the number of inner nuclear filaments. In addition, Tpr siRNA treatment impaired cell growth and proliferation compared to control siRNA-treated cells. In Tpr-depleted cells, the levels of p53 and p21 proteins were enhanced. Surprisingly, Tpr depletion increased p53 nuclear accumulation and facilitated autophagy. Our study demonstrates for the first time that Tpr plays a role in autophagy through controlling HSP70 and HSF1 mRNA export, p53 trafficking with karyopherin CRM1, and potentially through direct transcriptional regulation of autophagy factors.


Asunto(s)
Autofagia/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/genética , Poro Nuclear/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Procesos de Crecimiento Celular/genética , Línea Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Factores de Transcripción del Choque Térmico , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína Exportina 1
16.
PLoS One ; 7(6): e38423, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22685567

RESUMEN

MyD88, a Toll/interleukin-1 receptor homology (TIR) domain-containing adaptor protein, mediates signals from the Toll-like receptors (TLR) or IL-1/IL-18 receptors to downstream kinases. In MyD88-dependent TLR4 signaling, the function of MyD88 is enhanced by another TIR domain-containing adaptor, Mal/TIRAP, which brings MyD88 to the plasma membrane and promotes its interaction with the cytosolic region of TLR4. Hence, Mal is recognized as the "sorting adaptor" for MyD88. In this study, a direct interaction between MyD88-TIR and another membrane-sorting adaptor, TRAM/TICAM-2, was demonstrated in vitro. Cell-based assays including RNA interference experiments and TRAM deficient mice revealed that the interplay between MyD88 and TRAM in cells is important in mediating IL-18 signal transduction. Live cell imaging further demonstrated the co-localized accumulation of MyD88 and TRAM in the membrane regions in HEK293 cells. These findings suggest that TRAM serves as the sorting adaptor for MyD88 in IL-18 signaling, which then facilitates the signal transduction. The binding sites for TRAM are located in the TIR domain of MyD88 and actually overlap with the binding sites for Mal. MyD88, the multifunctional signaling adaptor that works together with most of the TLR members and with the IL-1/IL-18 receptors, can interact with two distinct sorting adaptors, TRAM and Mal, in a conserved manner in a distinct context.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Subunidad beta del Receptor de Interleucina-18/metabolismo , Glicoproteínas de Membrana/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina-1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sitios de Unión/genética , Células HEK293 , Humanos , Inmunoprecipitación , Interferón gamma/metabolismo , Interleucina-12/farmacología , Interleucina-18/farmacología , Subunidad beta del Receptor de Interleucina-18/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Modelos Moleculares , Mutación , Factor 88 de Diferenciación Mieloide/genética , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Receptores de Interleucina-1/genética , Receptores de Interleucina-18/metabolismo , Transducción de Señal , Células TH1/efectos de los fármacos , Células TH1/metabolismo , Receptor Toll-Like 4/metabolismo
17.
Cell Cycle ; 10(3): 425-33, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21270521

RESUMEN

Many human cancers have irregular chromosome content, a condition known as aneuploidy. Several nuclear pore proteins (nucleoporins/Nups) that mediate transport of RNA or macromolecules into and out of the nucleus have been implicated in mitosis. These nucleoporins are involved in molecular networks that function in a variety of mitotic processes, including chromosome condensation, sister chromatid cohesion, kinetochore assembly and spindle formation. An alteration in the concentration of Nups inside cells often causes aneuploidy. In this review, we discuss this sprouting area and the possible functions of Nups during mitosis.


Asunto(s)
Ciclo Celular , Proteínas de Complejo Poro Nuclear/fisiología , Transporte Activo de Núcleo Celular , Humanos , Mitosis , Modelos Biológicos , Poro Nuclear/química , Poro Nuclear/metabolismo , Poro Nuclear/fisiología , Proteínas de Complejo Poro Nuclear/química
18.
Cell Cycle ; 10(9): 1456-67, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21467841

RESUMEN

Chromosomal translocations involving chimeric fusions of the nucleoporin NUP98 protein have often been described in acute myelogenous leukemia (AML). All the fusion proteins have an identical NUP98 N terminus, which contains the GLEBS motif for interaction with the mRNA export factor RAE1 and FG repeats that associate with the transcription factors HDAC1 and p300. It is virtually unknown whether these interaction partners affect leukemogenesis. We previously showed that RAE1 depletion caused aneuploidy, which enhanced tumorigenesis. We speculated that RAE1 may also be directly involved in NUP98 fusion-mediated leukemogenesis. We show here that RNA interference (RNAi)-mediated knockdown of NUP98 caused severe chromosome segregation defects and disrupted RAE1 but not HDAC1 expression and localization. Next, we performed rescue experiments to confirm that the RAE1-NUP98 complex orchestrates proper chromosome segregation. Interestingly, we found diverse behaviors of NUP98 and the leukemogenic fusion protein NUP98-HOXA9 throughout the cell cycle. Strikingly, in NUP98-HOXA9-transfected cells, RAE1 protein were reduced and mis-localized. Our cellular interpretations were further confirmed by NUP98-HOXA9 transgenic mice and the NUP98-HOXA9 AML patient. These data suggest that RAE1 orchestrates NUP98-mediated leukemogenesis and raise the possibility that targeting this negative feedback loop may provide a new strategy for the therapy of aggressive leukemias.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/fisiología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Transporte Nucleocitoplasmático/fisiología , Adulto , Animales , Segregación Cromosómica/genética , Femenino , Regulación Leucémica de la Expresión Génica/fisiología , Células HEK293 , Células HeLa , Proteínas de Homeodominio/genética , Humanos , Células K562 , Leucemia Mieloide Aguda/etiología , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
19.
Cancer Res ; 69(13): 5349-56, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19531650

RESUMEN

Phosphoglucose isomerase/autocrine motility factor (PGI/AMF) is a housekeeping gene product/cytokine that catalyzes a step in glycolysis and gluconeogenesis, and acts as a multifunctional cytokine associated with aggressive tumors. PGI/AMF has been correlated significantly with breast cancer progression and poor prognosis in breast cancer. We show here that ectopic expression of PGI/AMF induced epithelial-to-mesenchymal transition (EMT) in MCF10A normal human breast epithelial cells, and inhibition of PGI/AMF expression triggered mesenchymal-to-epithelial transition (MET) in aggressive mesenchymal-type human breast cancer MDA-MB-231 cells. EMT in MCF10A cells was shown by morphologic changes and loss of E-cadherin/beta-catenin-mediated cell-cell adhesion, which is concomitant with the induction of the E-cadherin transcriptional repressor Snail and proteosome-dependent degradation of beta-catenin protein. Molecular analysis showed that PGI/AMF suppressed epithelial marker expressions and enhanced mesenchymal marker expressions. Silencing of PGI/AMF expression by RNA interference in MDA-MB-231 cells induced the reverse processes of EMT including altered cell shape, gain of epithelial marker, and reduction of mesenchymal marker, e.g., MET. Taken together, the results show the involvement of PGI/AMF in both EMT and MET: overexpression of PGI/AMF induces EMT in normal breast epithelial cells and reduction of PGI/AMF expression led to MET in aggressive breast cancer cells. These results suggest for the first time that PGI/AMF is a key gene to both EMT in the initiating step of cancer metastasis and MET in the later stage of metastasis during breast cancer progression.


Asunto(s)
Neoplasias de la Mama/patología , Células Epiteliales/patología , Glucosa-6-Fosfato Isomerasa/fisiología , Mesodermo/patología , Secuencia de Bases , Mama/citología , Mama/patología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Cadherinas/genética , Citocinas/fisiología , Cartilla de ADN , Femenino , Gluconeogénesis , Glucosa-6-Fosfato Isomerasa/genética , Glucólisis , Humanos , Datos de Secuencia Molecular , Fenotipo , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Transfección
20.
Cancer Metastasis Rev ; 26(3-4): 725-35, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17828376

RESUMEN

Autocrine motility factor (AMF) is a tumor-secreted cytokine and is abundant at tumor sites, where it may affect the process of tumor growth and metastasis. AMF is a multifunctional protein capable of affecting cell migration, invasion, proliferation, and survival, and possesses phosphoglucose isomerase activity and can catalyze the step in glycolysis and gluconeogenesis. Here, we review the role of AMF and tumor environment on malignant processes. The outcome of metastasis depends on multiple interactions between tumor cells and homeostatic mechanisms, therefore elucidation of the tumor/host interactions in the tumor microenvironment is essential in the development of new prevention and treatment strategies. Such knowledge might provide clues to develop new future therapeutic approaches for human cancers.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/fisiología , Neoplasias/etiología , Hipoxia de la Célula , Progresión de la Enfermedad , Humanos , Neoplasias/irrigación sanguínea , Neovascularización Patológica/etiología , Receptores del Factor Autocrino de Motilidad , Receptores de Citocinas/fisiología , Transducción de Señal , Ubiquitina-Proteína Ligasas/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA