Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Biol Chem ; 298(12): 102622, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36272642

RESUMEN

Fibrosis, stiffening and scarring of an organ/tissue due to genetic abnormalities, environmental factors, infection, and/or injury, is responsible for > 40% of all deaths in the industrialized world, and to date, there is no cure for it despite extensive research and numerous clinical trials. Several biomarkers have been identified, but no effective therapeutic targets are available. Human galectin-3 is a chimeric gene product formed by the fusion of the internal domain of the collagen alpha gene [N-terminal domain (ND)] at the 5'-end of galectin-1 [C-terminal domain (CRD)] that appeared during evolution together with vertebrates. Due to the overlapping structural similarities between collagen and galectin-3 and their shared susceptibility to cleavage by matrix metalloproteases to generate circulating collagen-like peptides, this review will discuss present knowledge on the role of collagen and galectin-3 as biomarkers of fibrosis. We will also highlight the need for transformative approaches targeting both the ND and CRD domains of galectin-3, since glycoconjugate binding by the CRD is triggered by ND-mediated oligomerization and the therapies targeted only at the CRD have so far achieved limited success.


Asunto(s)
Colágeno , Fibrosis , Galectina 3 , Animales , Humanos , Biomarcadores , Colágeno/genética , Colágeno/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Vertebrados , Glicoconjugados , Metaloproteinasas de la Matriz
2.
Glycobiology ; 28(4): 172-181, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29315388

RESUMEN

Over the last few decades galectin-3, a carbohydrate binding protein, with affinity for N-acetyllactosamine residues, has been unique due to the regulatory roles it performs in processes associated with tumor progression and metastasis such as cell proliferation, homotypic/heterotypic aggregation, dynamic cellular transformation, migration and invasion, survival and apoptosis. Structure-function association of galectin-3 reveals that it consists of a short amino terminal motif, which regulates its nuclear-cytoplasmic shuttling; a collagen α-like domain, susceptible to cleavage by matrix metalloproteases and prostate specific antigen; accountable for its oligomerization and lattice formation, and a carbohydrate-recognition/binding domain containing the anti-death motif of the Bcl2 protein family. This structural complexity permits galectin-3 to associate with numerous molecules utilizing protein-protein and/or protein-carbohydrate interactions in the extra-cellular as well as intracellular milieu and regulate diverse signaling pathways, a number of which appear directed towards epithelial-mesenchymal transition and cancer stemness. Self-renewal, differentiation, long-term culturing and drug-resistance potential characterize cancer stem cells (CSCs), a small cell subpopulation within the tumor that is thought to be accountable for heterogeneity, recurrence and metastasis of tumors. Despite the fact that association of galectin-3 to the tumor stemness phenomenon is still in its infancy, there is sufficient direct evidence of its regulatory roles in CSC-associated phenotypes and signaling pathways. In this review, we have highlighted the available data on galectin-3 regulated functions pertinent to cancer stemness and explored the opportunities of its exploitation as a CSC marker and a therapeutic target.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Galectina 3/metabolismo , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Proteínas Sanguíneas , Galectina 3/antagonistas & inhibidores , Galectinas , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos
3.
Cancer Metastasis Rev ; 35(2): 333-46, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27067726

RESUMEN

The skeleton is frequently a secondary growth site of disseminated cancers, often leading to painful and devastating clinical outcomes. Metastatic cancer distorts bone marrow homeostasis through tumor-derived factors, which shapes different bone tumor microenvironments depending on the tumor cells' origin. Here, we propose a novel insight on tumor-secreted Galectin-3 (Gal-3) that controls the induction of an inflammatory cascade, differentiation of osteoblasts, osteoclasts, and bone marrow cells, resulting in bone destruction and therapeutic failure. In the approaching era of personalized medicine, the current treatment modalities targeting bone metastatic environments are provided to the patient with limited consideration of the cancer cells' origin. Our new outlook suggests delivering individual tumor microenvironment treatments based on the expression level/activity/functionality of tumor-derived factors, rather than utilizing a commonly shared therapeutic umbrella. The notion of "Gal-3-associated bone remodeling" could be the first step toward a specific personalized therapy for each cancer type generating a different bone niche in patients afflicted with non-curable bone metastasis.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Galectina 3/metabolismo , Microambiente Tumoral , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Remodelación Ósea , Proteínas Portadoras/metabolismo , Comunicación Celular , Diferenciación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Citocinas/metabolismo , Femenino , Galectina 3/genética , Humanos , Masculino , Células Mieloides/citología , Células Mieloides/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Unión Proteica , Transducción de Señal , Microambiente Tumoral/genética
4.
Oncotarget ; 13: 534-550, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309869

RESUMEN

The accepted notion of dNTP transport following cytoplasmic biosynthesis is 'facilitated diffusion'; however, whether this alone is sufficient for moving dNTPs for DNA synthesis remains an open question. The data presented here show that the MYH9 gene encoded heavy chain of non-muscle myosin IIA binds dNTPs potentially serving as a 'reservoir'. Pull-down assays showed that MYH9 present in the cytoplasmic, mitochondrial and nuclear compartments bind to DNA and this interaction is inhibited by dNTPs and 2-deoxyribose-5-phosphate (dRP) suggesting that MYH9-DNA binding is mediated via pentose sugar recognition. Direct dNTP-MYH9 binding was demonstrated by ELISA and a novel PCR-based method, which showed that all dNTPs bind to MYH9 with varying efficiencies. Cellular thermal shift assays showed that MYH9 thermal stability is enhanced by dNTPs. MYH9 siRNA transfection or treatment with myosin II selective inhibitors ML7 or blebbistatin decreased cell proliferation compared to controls. EdU labeling and cell cycle analysis by flow cytometry confirmed MYH9 siRNA and myosin II inhibitors decreased progression to S-phase with accumulation of cells in G0/G1 phase. Taken together, our data suggest a novel role for MYH9 in dNTP binding and DNA synthesis.


Asunto(s)
Cadenas Pesadas de Miosina , Miosina Tipo IIA no Muscular , Proteínas del Citoesqueleto , ADN/genética , Desoxirribosa , Humanos , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo II , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIA no Muscular/metabolismo , Pentosas , Fosfatos , ARN Interferente Pequeño , Azúcares
5.
Int J Cancer ; 127(11): 2530-41, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20162566

RESUMEN

Galectin-3 cleavage is related to progression of human breast and prostate cancer and is partly responsible for tumor growth, angiogenesis and apoptosis resistance in mouse models. A functional polymorphism in galectin-3 gene, determining its susceptibility to cleavage by matrix metalloproteinases (MMPs)-2/-9 is related to racial disparity in breast cancer incidence in Asian and Caucasian women. The purpose of our study is to evaluate (i) if cleavage of galectin-3 could be related to angiogenesis during the progression of human breast cancer, (ii) the role of cleaved galectin-3 in induction of angiogenesis and (iii) determination of the galectin-3 domain responsible for induction of angiogenic response. Galectin-3 null breast cancer cells BT-459 were transfected with either cleavable full-length galectin-3 or its fragmented peptides. Chemotaxis, chemoinvasion, heterotypic aggregation, epithelial-endothelial cell interactions and angiogenesis were compared to noncleavable galectin-3. BT-549-H(64) cells harboring cleavable galectin-3 exhibited increased chemotaxis, invasion and interactions with endothelial cells resulting in angiogenesis and 3D morphogenesis compared to BT-549-P(64) cells harboring noncleavable galectin-3. BT-549-H(64) cells induced increased migration and phosphorylation of focal adhesion kinase in migrating endothelial cells. Endothelial cells cocultured with BT-549 cells transfected with galectin-3 peptides indicate that amino acids 1-62 and 33-250 stimulate migration and morphogenesis of endothelial cells. Immunohistochemical analysis of blood vessel density and galectin-3 cleavage in a breast cancer progression tissue array support the in vitro findings. We conclude that the cleavage of the N terminus of galectin-3 followed by its release in the tumor microenvironment in part leads to breast cancer angiogenesis and progression.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Galectina 3/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Sustitución de Aminoácidos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Quimiotaxis/fisiología , Técnicas de Cocultivo , Células Endoteliales/patología , Femenino , Galectina 3/genética , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Polimorfismo de Nucleótido Simple , Transfección
6.
Am J Pathol ; 174(4): 1515-23, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19286570

RESUMEN

Galectin-3, a beta-galactoside-binding protein, has been implicated in a variety of biological functions including cell proliferation, apoptosis, angiogenesis, tumor progression, and metastasis. The present study was undertaken to understand the role of galectin-3 in the progression of prostate cancer. Immunohistochemical analysis of galectin-3 expression revealed that galectin-3 was cleaved during the progression of prostate cancer. Galectin-3 knockdown by small interfering RNA (siRNA) was associated with reduced cell migration, invasion, cell proliferation, anchorage-independent colony formation, and tumor growth in the prostates of nude mice. Galectin-3 knockdown in human prostate cancer PC3 cells led to cell-cycle arrest at G(1) phase, up-regulation of nuclear p21, and hypophosphorylation of the retinoblastoma tumor suppressor protein (pRb), with no effect on cyclin D1, cyclin E, cyclin-dependent kinases (CDK2 and CDK4), and p27 protein expression levels. The data obtained here implicate galectin-3 in prostate cancer progression and suggest that galectin-3 may serve as both a diagnostic marker and therapeutic target for future disease treatments.


Asunto(s)
Biomarcadores de Tumor/análisis , Galectina 3/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Técnica del Anticuerpo Fluorescente , Galectina 3/genética , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
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.
Carbohydr Polym ; 211: 141-151, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824074

RESUMEN

While chemically and thermally modified citrus pectin (MCP) has already been studied for health benefits, it is unknown how size-fractionated oligo- and polysaccharides differentially affect cancer cell behavior. We produced thermally MCP and fractionated it by molecular size to evaluate the effect these polymers have on cancer cells. MCP30/10 (between 30 and 10 kDa) had more esterified homogalacturonans (HG) and fewer rhamnogalacturonans (RG-I) than MCP and MCP30 (higher than 30 kDa), while MCP10/3 (between 10 and 3 kDa) showed higher amounts of type I arabinogalactans (AGI) and lower amounts of RG-I. MCP3 (smaller than 3 kDa) presented less esterified HG and the lowest amount of AGI and RG-I. Our data indicate that the enrichment of de-esterified HG oligomers and the AGI and RG-I depletions in MCP3, or the increase of AGI and loss of RGI in MCP30/10, enhance the anticancer behaviors by inhibiting migration, aggregation, and proliferation of cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Pectinas/farmacología , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Peso Molecular , Neoplasias/tratamiento farmacológico , Pectinas/química
9.
Cancer Res ; 66(20): 9995-10006, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17047062

RESUMEN

Galectin-3 (Gal-3), a pleiotropic beta-galactoside-binding protein, was shown to be involved in several nuclear-dependent functions, including up-regulation of transcriptional factors, RNA processing, and cell cycle regulation. Gal-3 compartmentalization in the nucleus versus the cytoplasm affects, in part, the malignant phenotype of various cancers. However, to date, the mechanism by which Gal-3 translocates into the nucleus remains debatable. Thus, we have constructed and expressed a variety of fusion proteins containing deletion mutants of Gal-3 fused with monomers, dimers, and trimers of enhanced green fluorescent protein and searched for the Gal-3 sequence motifs essential for its nuclear localization in vivo. In addition, a digitonin-permeabilized, cell-free transport in vitro assay was used to directly examine the mechanism of Gal-3 nuclear import. Partial deletions of the COOH-terminal region (114-250) of the human Gal-3 significantly decreases its nuclear translocation, whereas a peptide (1-115) was transported to the nuclei. The in vitro nuclear import assay revealed that there are at least two independent nuclear pathways for shuttling Gal-3 into the nucleus: a passive diffusion and an active transport. This is the first article providing direct evidence for the nuclear import mechanisms of Gal-3 and suggests that Gal-3 nuclear translocation is governed by dual pathways, whereas the cytoplasmic/nuclear distribution may be regulated by multiple processes, including cytoplasmic anchorage, nuclear retention, and or nuclear export. These results may lead to the development of a therapeutic modality aiming at abrogating Gal-3 translocation into the nucleus and thus hampering its activity during cancer progression and metastasis.


Asunto(s)
Núcleo Celular/metabolismo , Galectina 3/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Antígenos Transformadores de Poliomavirus/genética , Transporte Biológico Activo , Neoplasias de la Mama/metabolismo , Células COS , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Chlorocebus aethiops , Difusión , Digitonina/farmacología , Galectina 3/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Mutación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Fracciones Subcelulares/metabolismo , Transfección
10.
Cancer Res ; 65(17): 7546-53, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16140916

RESUMEN

The antiapoptotic molecule galectin-3 was previously shown to regulate CD95, a member of the tumor necrosis factor (TNF) family of proteins in the apoptotic signaling pathway. Here, we question the generality of the phenomenon by studying a different member of this family of proteins [e.g., TNF-related apoptosis-inducing ligand (TRAIL), which induces apoptosis in a wide variety of cancer cells]. Overexpression of galectin-3 in J82 human bladder carcinoma cells rendered them resistant to TRAIL-induced apoptosis, whereas phosphatidylinositol 3-kinase (PI3K) inhibitors (wortmannin and LY-294002) blocked the galectin-3 protecting effect. Because Akt is a major downstream PI3K target reported to play a role in TRAIL-induced apoptosis, we questioned the possible relationship between galectin-3 and Akt. Parental J82 and the control vector-transfected J82 cells (barely detectable galectin-3) exhibit low level of constitutively active Akt, resulting in sensitivity to TRAIL. On the other hand, J82 cells overexpressing galectin-3 cells expressed a high level of constitutively active Akt and were resistant to TRAIL. Moreover, the blockage of TRAIL-induced apoptosis in J82 cells seemed to be mediated by Akt through the inhibition of BID cleavage. These results suggest that galectin-3 involves Akt as a modulator molecule in protecting bladder carcinoma cells from TRAIL-induced apoptosis.


Asunto(s)
Apoptosis/fisiología , Galectina 3/fisiología , Glicoproteínas de Membrana/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Neoplasias de la Vejiga Urinaria/enzimología , Neoplasias de la Vejiga Urinaria/patología , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Activación Enzimática , Galectina 3/biosíntesis , Galectina 3/genética , Humanos , Membranas Intracelulares/fisiología , Potenciales de la Membrana/fisiología , Mitocondrias/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF , Transfección , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo
11.
Cancer Res ; 65(9): 3535-7, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867344

RESUMEN

Galectin-3 (gal-3), a member of the beta-galactoside-binding proteins family, was identified as a binding partner of beta-catenin. Analysis of the human gal-3 sequence reveled a structural similarity to beta-catenin as it also contains the consensus sequence (S92XXXS96) for glycogen synthase kinase-3beta (GSK-3beta) phosphorylation and can serve as its substrate. In addition, Axin, a regulator protein of Wnt that complexes with beta-catenin, also binds gal-3 using the same sequence motif identified here by a deletion mutant analysis. The data presented here give credence to the suggestion that gal-3 is a key regulator in the Wnt/beta-catenin signaling pathway and highlight the functional similarities between gal-3 and beta-catenin.


Asunto(s)
Galectina 3/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Animales , Proteína Axina , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Galectina 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Fosforilación , Unión Proteica , Ratas , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Transactivadores/metabolismo , Transfección , Proteínas Wnt , beta Catenina
12.
Cancer Res ; 77(20): 5441-5444, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28838888

RESUMEN

The hurdles in realizing successful cancer immunotherapy stem from the fact that cancer patients are either refractory to immune response and/or develop resistance. Here, we propose that these phenomena are due, in part, to the deployment/secretion of a "decoy flare," for example, anomalous cancer-associated antigens by the tumor cells. The cancer secretome, which resembles the parent cell make-up, is composed of soluble macromolecules (proteins, glycans, lipids, DNAs, RNAs, etc.) and insoluble vesicles (exosomes), thus hindering cancer detection/recognition by immunotherapeutic agents, resulting in a "cancer-stealth" effect. Immunotherapy, or any treatment that relies on antigens' expression/function, could be improved by the understanding of the properties of the cancer secretome, as its clinical evaluation may change the therapeutic landscape. Cancer Res; 77(20); 5441-4. ©2017 AACR.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Animales , Humanos
13.
Oncotarget ; 8(11): 17643-17650, 2017 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-27741522

RESUMEN

The U.S. Preventive Services Task Force (USPSTF) has recommended against PSA-based screening for prostate cancer due to potential possibilities of false-results. Since no alternative test is available to replace it, we have initiated a trial with the purpose of establishing whether Galectin-3 (Gal-3) serum level and/or the patients' immune response to PSA and Gal-3 antigens could complement the PSA test as diagnostic tools for prostate cancer patients. A blind, prospective, single institution, pilot study was conducted. A total of 95 men were recruited and classified into 5 different groups: healthy controls (Group1), newly diagnosed patients (Group2), no recurrence after local therapy (Group3), rising PSA after local therapy (Group4), and metastatic patients (Group5). The primary endpoints were the levels of serum PSA, PSA autoantibodies (AAPSA), Gal-3, and Gal-3 autoantibodies (AAGal-3). Data were analyzed by Spearman's rank correlation (rho) and least squares linear regression modeling. The expression levels of PSA, AAPSA, Gal-3, and AAGal-3 were determined in both healthy controls and prostate cancer patients. Negative correlations were observed between PSA and AAPSA levels among all 95 men combined (rho = -0.321, P = 0.0021; fitted slope -0.288, P = 0.0048), and in metastatic patients (rho = -0.472, P = 0.0413; fitted slope -1.145, P = 0.0061). We suggest an association between PSA and AAPSA, whereby the AAPSA may alter PSA levels. It provides a novel outlook for prostate cancer diagnosis, and should serve as a basis for an all-inclusive diagnostic trial centering on patients with metastasis.


Asunto(s)
Autoanticuerpos/sangre , Biomarcadores de Tumor/sangre , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/sangre , Anciano , Autoantígenos/inmunología , Biomarcadores de Tumor/inmunología , Ensayo de Inmunoadsorción Enzimática , Galectina 3/inmunología , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/inmunología
14.
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
15.
J Natl Cancer Inst ; 94(24): 1854-62, 2002 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-12488479

RESUMEN

BACKGROUND: The role of dietary components in cancer progression and metastasis is an emerging field of clinical importance. Many stages of cancer progression involve carbohydrate-mediated recognition processes. We therefore studied the effects of high pH- and temperature-modified citrus pectin (MCP), a nondigestible, water-soluble polysaccharide fiber derived from citrus fruit that specifically inhibits the carbohydrate-binding protein galectin-3, on tumor growth and metastasis in vivo and on galectin-3-mediated functions in vitro. METHODS: In vivo tumor growth, angiogenesis, and metastasis were studied in athymic mice that had been fed with MCP in their drinking water and then injected orthotopically with human breast carcinoma cells (MDA-MB-435) into the mammary fat pad region or with human colon carcinoma cells (LSLiM6) into the cecum. Galectin-3-mediated functions during tumor angiogenesis in vitro were studied by assessing the effect of MCP on capillary tube formation by human umbilical vein endothelial cells (HUVECs) in Matrigel. The effects of MCP on galectin-3-induced HUVEC chemotaxis and on HUVEC binding to MDA-MB-435 cells in vitro were studied using Boyden chamber and labeling assays, respectively. The data were analyzed by two-sided Student's t test or Fisher's protected least-significant-difference test. RESULTS: Tumor growth, angiogenesis, and spontaneous metastasis in vivo were statistically significantly reduced in mice fed MCP. In vitro, MCP inhibited HUVEC morphogenesis (capillary tube formation) in a dose-dependent manner. In vitro, MCP inhibited the binding of galectin-3 to HUVECs: At concentrations of 0.1% and 0.25%, MCP inhibited the binding of galectin-3 (10 micro g/mL) to HUVECs by 72.1% (P =.038) and 95.8% (P =.025), respectively, and at a concentration of 0.25% it inhibited the binding of galectin-3 (1 micro g/mL) to HUVECs by 100% (P =.032). MCP blocked chemotaxis of HUVECs toward galectin-3 in a dose-dependent manner, reducing it by 68% at 0.005% (P<.001) and inhibiting it completely at 0.1% (P<.001). Finally, MCP also inhibited adhesion of MDA-MB-435 cells, which express galectin-3, to HUVECs in a dose-dependent manner. CONCLUSIONS: MCP, given orally, inhibits carbohydrate-mediated tumor growth, angiogenesis, and metastasis in vivo, presumably via its effects on galectin-3 function. These data stress the importance of dietary carbohydrate compounds as agents for the prevention and/or treatment of cancer.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Citrus , Galectina 3/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Pectinas/administración & dosificación , Adenocarcinoma/tratamiento farmacológico , Administración Oral , Animales , Western Blotting , Neoplasias de la Mama/tratamiento farmacológico , Quimiotaxis , Neoplasias del Colon/tratamiento farmacológico , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Endotelio Vascular , Técnica del Anticuerpo Fluorescente Indirecta , Inmunohistoquímica , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/prevención & control , Trasplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/prevención & control , Neovascularización Patológica/tratamiento farmacológico , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas , Venas Umbilicales
16.
Cancer Res ; 63(1): 242-9, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12517804

RESUMEN

Autocrine motility factor (AMF)/phosphoglucose isomerase (PGI) is a ubiquitous cytosolic enzyme and is produced as a leaderless secretory protein, released from cells via a nonclassical pathway. AMF/PGI acts extracellularly as a potent mitogen/cytokine (CXXC, chemokine). Increased expression of AMF/PGI and its receptor/CXXC-R has been found in a wide spectrum of malignancies, and is associated with cancer progression and metastasis. To directly elucidate the functional role of AMF/PGI on cell motility and neoplastic transformation, we stably transfected AMF/PGI cDNA into NIH-3T3 cells. Ectopic overexpression of AMF/PGI results in its secretion and activation via a constitutive autocrine activation loop that renders the cells highly motile, acquiring a transformed phenotype in vitro and tumorigenicity in vivo. The transformed phenotype of AMF/PGI-transfected cells leads in part resistance to induction of apoptosis induced by serum starvation, through the activation of phosphatidylinositol 3'-kinase/Akt signaling pathway and down-regulation of caveolin-1 expression. Overexpression of this housekeeping gene induces resistance to apoptosis and neoplastic transformation, and, thus, AMF/PGI represents a novel class of oncogenic protein.


Asunto(s)
Supervivencia Celular/fisiología , Transformación Celular Neoplásica , Fibrosarcoma/patología , Glucosa-6-Fosfato Isomerasa/genética , Glucosa-6-Fosfato Isomerasa/metabolismo , Células 3T3 , Animales , División Celular , Medio de Cultivo Libre de Suero , Fibroblastos/citología , Fibrosarcoma/genética , Humanos , Cinética , Ratones , Ratones Desnudos , Invasividad Neoplásica , Fenotipo , Proteínas Recombinantes/metabolismo , Transfección
17.
Cancer Res ; 64(18): 6363-7, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15374939

RESUMEN

Galectin-3 (gal-3), a pleiotrophic protein, is an important regulator of tumor metastasis, which like beta-catenin shuttles between the nucleus and the cytosol in a phosphorylation-dependent manner. We report herein that beta-catenin stimulation of cyclin D1 and c-myc expression is gal-3 dependent. Gal-3 binds to beta-catenin/Tcf complex, colocalizes with beta-catenin in the nucleus, and induces the transcriptional activity of Tcf-4 as determined by the TOP/FOPFLASH reporter system. We have identified the beta-catenin-gal-3-binding sequences, which are in the NH2 and COOH termini of the proteins encompassing amino acid residues 1 to 131 and 143 to 250, respectively. These data indicate that gal-3 is a novel binding partner for beta-catenin involved in the regulation of Wnt/beta-catenin signaling pathway.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Galectina 3/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Galectina 3/genética , Regulación Neoplásica de la Expresión Génica , Genes bcl-1 , Células HCT116 , Células HT29 , Humanos , Plásmidos/genética , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Factores de Transcripción TCF , Transactivadores/genética , Proteína 2 Similar al Factor de Transcripción 7 , Factores de Transcripción/genética , Activación Transcripcional , Transfección , Regulación hacia Arriba , beta Catenina
18.
Cancer Res ; 62(15): 4484-90, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12154059

RESUMEN

The hallmark of tumor metastasis is the dissemination of cells from the primary growth site to distant organs. Autocrine motility factor (AMF), a tumor-associated C-X-X-C cytokine, the ligand for a unique 78 kDa seven transmembrane receptor, is a potent simulator of cell motility, a process that is a prerequisite for tumor progression and metastasis. Because little is known about AMF-dependent signaling, we sought to study whether AMF signaling involves family members of the Rho-like GTPases. AMF stimulation of human melanoma cells resulted in stress-fiber formation, concomitant with up-regulation and activation of both RhoA and Rac1 expression with no apparent changes in the expression level or activation state of Cdc42. Treatment of the cells with C3 exoenzyme before AMF stimulation inhibited both the formation of stress-fiber-like structures and the activation of RhoA. In addition, both c-Jun NH(2)-terminal kinase 1 and c-Jun NH(2)-terminal kinase 2 were simultaneously activated by AMF, supporting the notion that they are involved in the signaling pathway of RhoA. We thus conclude that AMF signaling shares a similar pathway to previously established paracrine factors signaling involving cytoskeletal rearrangement and morphological alterations mediated by the small RhoA-like GTPases.


Asunto(s)
Toxinas Botulínicas , Glucosa-6-Fosfato Isomerasa/fisiología , Proteínas de Unión al GTP rho/fisiología , ADP Ribosa Transferasas/farmacología , Movimiento Celular/fisiología , Citoesqueleto/enzimología , Citoesqueleto/patología , Activación Enzimática , Fibrosarcoma/enzimología , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Humanos , Melanoma/enzimología , Melanoma/metabolismo , Melanoma/patología , Receptores del Factor Autocrino de Motilidad , Receptores de Citocinas/fisiología , Transducción de Señal/fisiología , Fibras de Estrés/enzimología , Fibras de Estrés/metabolismo , Fibras de Estrés/patología , Ubiquitina-Proteína Ligasas , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/biosíntesis , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
19.
Cancer Res ; 63(23): 8302-11, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678989

RESUMEN

Galectin (Gal)-3, a M(r) 31000 member of the beta-galactoside-binding protein family, is a multifunctional protein implicated in a variety of biological functions, including tumor cell adhesion, proliferation, differentiation, angiogenesis, apoptosis, cancer progression, and metastasis. Here, we report that secreted extracellular Gal-3 can signal apoptosis of human T leukemia cell lines, human peripheral blood mononuclear cells, and activated mouse T cells after binding to cell surface glycoconjugate receptors through carbohydrate-dependent interactions because the apoptotic effect was found to be inhibited by lactose, specific sugar inhibitor, and to be dose dependent. However, the apoptosis sensitivity to Gal-3 varied among the different cell lines tested. We report that Gal-3-null Jurkat, CEM, and MOLT-4 cells were significantly more sensitive to exogenous Gal-3 than SKW6.4 and H9 cells, which express Gal-3, suggesting a cross-talk between the antiapoptotic activity of intracellular Gal-3 and proapoptotic activity of extracellular Gal-3. Furthermore, Gal-3-transfected CEM cells were found to be more resistant to C(2)-ceramide-induced apoptosis than the control CEM cells. Identification of Gal-3 cell surface receptors revealed that Gal-3 binding to CD7 and CD29 (beta(1) integrin) induced apoptosis. Gal-3 binding to its cell surface receptors results in activation of mitochondrial apoptosis events including cytochrome c release and caspase-3 activation, but not caspase-8 activation. Taken together, these results suggest that the induction of T-cell apoptosis by secreted Gal-3 may play a role in the immune escape mechanism during tumor progression through the induction of apoptosis to cancer-infiltrating T cells. The induction of T-cell apoptosis by secreted Gal-3 is dependent in part on the presence or absence of cytoplasmic Gal-3, providing a new insight for the immune escape mechanism of cancer cells.


Asunto(s)
Antígenos CD7/fisiología , Apoptosis/efectos de los fármacos , Galectina 3/farmacología , Integrina beta1/fisiología , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD7/metabolismo , Apoptosis/inmunología , Inhibidores de Caspasas , Caspasas/metabolismo , Activación Enzimática , Galectina 3/genética , Galectina 3/metabolismo , Humanos , Integrina beta1/metabolismo , Células Jurkat , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Linfocitos T/citología , Linfocitos T/inmunología , Transfección
20.
Cancer Res ; 64(10): 3376-9, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15150087

RESUMEN

Studies of CD95 (APO-1/Fas), a member of the death receptor family, have revealed that it is involved in two primary CD95 apoptotic signaling pathways, one regulated by the large amount of active caspase-8 (type I) formed at the death-inducing signaling complex and the other by the apoptogenic activity of mitochondria (type II). To date, it is still unclear which pathway will be activated in response to an apoptotic insult. Here, we demonstrate that the antiapoptotic molecule galectin-3, which contains the four amino acid-anti-death-motif (NWGR) conserved in the BH1 domain of the Bcl-2 member proteins, is expressed only in type I cells. Transfection of galectin-3 cDNA into galectin-3 null cells (type II) resulted converting them to type I apoptotic phenotype. In addition, we show that galectin-3 is complexed with CD95 in vivo identifying galectin-3 as a novel CD95-binding partner that determines which of the CD95 apoptotic signaling pathways the cell will select.


Asunto(s)
Apoptosis/fisiología , Galectina 3/fisiología , Receptor fas/fisiología , Linfocitos B/fisiología , Caspasa 8 , Caspasas/metabolismo , Línea Celular , Galectina 3/genética , Galectina 3/metabolismo , Humanos , Células Jurkat , Transducción de Señal/fisiología , Linfocitos T/fisiología , Transfección , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda