Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell Biol ; 39(11)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31061093

RESUMEN

GP78 is an autocrine motility factor (AMF) receptor (AMFR) with E3 ubiquitin ligase activity that plays a significant role in tumor cell proliferation, motility, and metastasis. Aberrant extracellular signal-regulated kinase (ERK) activation via receptor tyrosine kinases promotes tumor proliferation and invasion. The activation of GP78 leads to ERK activation, but its underlying mechanism is not fully understood. Here, we show that GP78 is required for epidermal growth factor receptor (EGFR)-mediated ERK activation. On one hand, GP78 interacts with and promotes the ubiquitination and subsequent degradation of dual-specificity phosphatase 1 (DUSP1), an endogenous negative regulator of mitogen-activated protein kinases (MAPKs), resulting in ERK activation. On the other hand, GP78 maintains the activation status of EGFR, as evidenced by the fact that EGF fails to induce EGFR phosphorylation in GP78-deficient cells. By the regulation of both EGFR and ERK activation, GP78 promotes cell proliferation, motility, and invasion. Therefore, this study identifies a previously unknown signaling pathway by which GP78 stimulates ERK activation via DUSP1 degradation to mediate EGFR-dependent cancer cell proliferation and invasion.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Fosfatasa 1 de Especificidad Dual/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores del Factor Autocrino de Motilidad/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Fosfatasa 1 de Especificidad Dual/química , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Invasividad Neoplásica , Fosforilación , Proteolisis , Ubiquitinación
2.
Biochem Biophys Res Commun ; 509(2): 348-353, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30585151

RESUMEN

The E3 ubiquitin (Ub) ligase gp78 plays an important role in endoplasmic reticulum (ER)-associated degradation (ERAD) and regulation of lipid biogenesis. Although a variety of substrates of gp78 have been described, the regulation of the degradation of gp78 itself remains poorly understood. To address this problem, we used co-immunoprecipitation-coupled liquid chromatography-tandem mass spectrometry (Co-IP/LC-MS/MS) to identify novel proteins interacting with gp78. One of the proteins identified in this study is the deubiquitylating (DUB) enzyme USP34 (Ub-specific protease 34). We demonstrate that knockdown of USP34 facilitates proteasomal degradation of gp78 and consequently impairs the function of gp78 in regulating lipid droplet formation. This study unveils a previously unknown function of USP34 in regulating the metabolic stability of gp78 and adds to our understanding of the relevance of partnering of DUBs and E3s in regulation of protein ubiquitylation.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Hepatocitos/metabolismo , Receptores del Factor Autocrino de Motilidad/genética , Proteasas Ubiquitina-Específicas/genética , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Células HEK293 , Hepatocitos/citología , Humanos , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores del Factor Autocrino de Motilidad/metabolismo , Transducción de Señal , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación
3.
Cancer Res ; 76(6): 1391-402, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26837763

RESUMEN

Management of bone metastasis remains clinically challenging and requires the identification of new molecular target(s) that can be therapeutically exploited to improve patient outcome. Galectin-3 (Gal-3) has been implicated as a secreted factor that alters the bone microenvironment. Proteolytic cleavage of Gal-3 may also contribute to malignant cellular behaviors, but has not been addressed in cancer metastasis. Here, we report that Gal-3 modulates the osteolytic bone tumor microenvironment in the presence of RANKL. Gal-3 was localized on the osteoclast cell surface, and its suppression by RNAi or a specific antagonist markedly inhibited osteoclast differentiation markers, including tartrate-resistant acid phosphatase, and reduced the number of mature osteoclasts. Structurally, the 158-175 amino acid sequence in the carbohydrate recognition domain (CRD) of Gal-3 was responsible for augmented osteoclastogenesis. During osteoclast maturation, Gal-3 interacted and colocalized with myosin-2A along the surface of cell-cell fusion. Pathologically, bone metastatic cancers expressed and released an intact form of Gal-3, mainly detected in breast cancer bone metastases, as well as a cleaved form, more abundant in prostate cancer bone metastases. Secreted intact Gal-3 interacted with myosin-2A, leading to osteoclastogenesis, whereas a shift to cleaved Gal-3 attenuated the enhancement in osteoclast differentiation. Thus, our studies demonstrate that Gal-3 shapes the bone tumor microenvironment through distinct roles contingent on its cleavage status, and highlight Gal-3 targeting through the CRD as a potential therapeutic strategy for mitigating osteolytic bone remodeling in the metastatic niche.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Remodelación Ósea/fisiología , Neoplasias de la Mama/patología , Galectina 3/metabolismo , Metástasis de la Neoplasia/patología , Neoplasias de la Próstata/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Neoplasias Óseas/secundario , Huesos/metabolismo , Huesos/patología , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Femenino , Humanos , Isoenzimas/metabolismo , Masculino , Ratones , Miosinas/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patología , Neoplasias de la Próstata/patología , Ligando RANK/metabolismo , Fosfatasa Ácida Tartratorresistente
4.
Clin Cancer Res ; 22(5): 1284-94, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26527747

RESUMEN

PURPOSE AND EXPERIMENTAL DESIGN: The molecular events in the malignant progression of colon adenoma after loss of adenomatous polyposis coli (APC) are not fully understood. KITENIN (KAI1 C-terminal interacting tetraspanin) increases the invasiveness of colorectal cancer cells, and we identified a novel EGFR-independent oncogenic signal of EGF that works under coexpressed KITENIN and ErbB4. Here we tested whether elevated KITENIN and ErbB4 contribute to further progression of intestinal adenoma following APC loss. RESULTS: The intestinal tissues of villin-KITENIN transgenic mice in which villin-driven KITENIN expression induces increased c-Jun expression exhibit mild epithelial cell proliferation but no epithelial lineage changes compared with those of nontransgenic mice. Among the four ErbB4 isoforms, JM-a/CYT-2 and JM-b/CYT-2 exhibited the highest AP-1 activity when cells coexpressing KITENIN and each isoform were stimulated by EGF. Interestingly, predominant overexpression of the ErB4-CYT-2 mRNA as well as increased EGFR expression were observed in intestinal adenoma of APC(min/+) mice, which makes the microenvironment of activated EGF signaling. When we crossed villin-KITENIN mice with APC(min/+) mice, intestinal tumor tissues in the crossed mice showed the characteristics of early-stage invading adenocarcinoma. In patients with colorectal cancer, ErbB4-CYT-2 mRNA expression was significantly greater in tumor tissues than in normal adjacent tissues, but no significant differences in tumor tissue expression were found between different colorectal cancer stages. Furthermore, the mRNA expression of KITENIN and that of ErbB4-CYT-2 were positively correlated in human colorectal cancer tissue. CONCLUSIONS: Elevated coexpression of KITENIN and ErbB4-CYT-2 promotes the transition of colon adenoma to adenocarcinoma within an APC loss-associated tumor microenvironment.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/biosíntesis , Biomarcadores de Tumor/biosíntesis , Proteínas Portadoras/biosíntesis , Neoplasias Colorrectales/genética , Proteínas de la Membrana/biosíntesis , Receptor ErbB-4/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenoma/genética , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Biomarcadores de Tumor/genética , Proteínas Portadoras/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Receptor ErbB-4/genética , Microambiente Tumoral/genética
5.
Nat Commun ; 6: 8698, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26507712

RESUMEN

Induction of T-cell clonal anergy involves serial activation of transcription factors, including NFAT and Egr2/3. However, downstream effector mechanisms of these transcription factors are not fully understood yet. Here we identify Ndrg1 as an anergy factor induced by Egr2. Ndrg1 is upregulated by anergic signalling and maintained at high levels in resting anergic T cells. Overexpression of Ndrg1 mimics the anergic state and knockout of the gene prevents anergy induction. Interestingly, Ndrg1 is phosphorylated and degraded by CD28 signalling in a proteasome-dependent manner, explaining the costimulation dependence of anergy prevention. Similarly, IL-2 treatment of anergic T cells, under conditions that lead to the reversal of anergy, also induces Ndrg1 phosphorylation and degradation. Finally, older Ndrg1-deficient mice show T-cell hyperresponsiveness and Ndrg1-deficient T cells aggravate inducible autoimmune inflammation. Thus, Ndrg1 contributes to the maintenance of clonal anergy and inhibition of T-cell-mediated inflammation.


Asunto(s)
Antígenos CD28/inmunología , Proteínas de Ciclo Celular/genética , Anergia Clonal , Regulación hacia Abajo , Interleucina-2/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Linfocitos T/inmunología , Animales , Antígenos CD28/genética , Proteínas de Ciclo Celular/inmunología , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/inmunología , Interleucina-2/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Oncotarget ; 6(23): 19592-604, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26158764

RESUMEN

Galectin-3 (Gal-3, LGALS3) is a pleotropic versatile, 29-35 kDa chimeric gene product, and involved in diverse physiological and pathological processes, including cell growth, homeostasis, apoptosis, pre-mRNA splicing, cell-cell and cell-matrix adhesion, cellular polarity, motility, adhesion, activation, differentiation, transformation, signaling, regulation of innate/adaptive immunity, and angiogenesis. In multiple diseases, it was found that the level of circulating Gal-3 is markedly elevated, suggesting that Gal-3-dependent function is mediated by specific interaction with yet an unknown ubiquitous cell-surface protein. Recently, we showed that Gal-3 attenuated drug-induced apoptosis, which is one of the mechanisms underlying multidrug resistance (MDR). Here, we document that MDR could be mediated by Gal-3 interaction with the house-keeping gene product e.g., Na+/K+-ATPase, and P-glycoprotein (P-gp). Gal-3 interacts with Na+/K+-ATPase and induces the phosphorylation of P-gp. We also find that Gal-3 binds P-gp and enhances its ATPase activity. Furthermore Gal-3 antagonist suppresses this interaction and results in a decrease of the phosphorylation and the ATPase activity of P-gp, leading to an increased sensitivity to doxorubicin-mediated cell death. Taken together, these findings may explain the reported roles of Gal-3 in diverse diseases and suggest that a combined therapy of inhibitors of Na+/K+-ATPase and Gal-3, and a disease specific drug(s) might be superior to a single therapeutic modality.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Galectina 3/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Proteínas Sanguíneas , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Activación Enzimática , Femenino , Galectina 3/farmacología , Galectinas , Células HeLa , Humanos , Masculino , Neoplasias/genética , Neoplasias/patología , Fenotipo , Fosforilación , Unión Proteica , Proteómica/métodos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/genética , Transfección , Familia-src Quinasas/metabolismo
7.
PLoS One ; 10(3): e0118448, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25789613

RESUMEN

Nonalcoholic steatohepatitis (NASH) is related to metabolic dysregulation and the perturbation of endoplasmic reticulum (ER) homeostasis that frequently develops into hepatocellular carcinoma (HCC). Gp78 is E3 ligase, which regulates endoplasmic reticulum-associated degradation (ERAD) by ubiquitinylation of misfolded ER proteins. Here, we report that upon ageing (12 months), gp78-/- mice developed obesity, recapitulating age-related human NASH. Liver histology of gp78-/- mice revealed typical steatosis, hepatic inflammation and fibrosis, followed by progression to hepatocellular tumors. Acute ER stress revealed that loss of gp78 results in up regulation of unfolded protein response (UPR) pathways and SREBP-1 regulating de novo lipogenesis, responsible for fatty liver. Tissue array of human hepatocellular carcinoma (HCC) demonstrated that the expression of gp78 was inversely correlated with clinical grades of cancer. Here, we have described the generation of the first preclinical experimental model system which spontaneously develops age-related NASH and HCC, linking ERAD to hepatosteatosis, cirrhosis, and cancer. It suggests that gp78 is a regulator of normal liver homeostasis and a tumor suppressor in human liver.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/fisiología , Neoplasias Hepáticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Receptores del Factor Autocrino de Motilidad/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Western Blotting , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/patología , Estrés del Retículo Endoplásmico/genética , Inmunohistoquímica , Inmunoprecipitación , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Receptores del Factor Autocrino de Motilidad/genética , Ubiquitinación
8.
Oncotarget ; 5(20): 9992-10001, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25393982

RESUMEN

Cancer cells survive escaping normal apoptosis and the blocks in apoptosis that keep cancer cells alive are promising candidates for targeted therapy. Galectin-3 (Gal-3) is, a member of the lectin family, which is involved in cell growth, adhesion, proliferation and apoptosis. It remains elusive to understand the role of Gal-3 on apoptosis in thyroid carcinoma cells. Here, we report that Gal-3 heterodimerizes Bax, mediated by the carbohydrate recognition domain (CRD) of Gal-3, leading to anti-apoptotic characteristic. Gal-3/Bax interaction was suppressed by an antagonist of Gal-3, in which in turn cells became sensitive to apoptosis. The data presented here highlight that Gal-3 is involved in the anti-apoptosis of thyroid carcinoma cells. Thus, it suggests that targeting Gal-3 may lead to an improved therapeutic modality for thyroid cancer.


Asunto(s)
Galectina 3/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Proteína X Asociada a bcl-2/metabolismo , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Humanos , Multimerización de Proteína , Neoplasias de la Tiroides/genética
9.
Neoplasia ; 16(11): 939-49, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25425968

RESUMEN

Patients with bone cancer metastasis suffer from unbearable pain and bone fractures due to bone remodeling. This is caused by tumor cells that disturb the bone microenvironment. Here, we have investigated the role of tumor-secreted sugar-binding protein, i.e., galectin-3, on osteoblast differentiation and report that it downregulates the expression of osteoblast differentiation markers, e.g., RUNX2, SP7, ALPL, COL1A1, IBSP, and BGLAP, of treated human fetal osteoblast (hFOB) cells. Co-culturing of hFOB cells with human breast cancer BT-549 and prostate cancer LNCaP cells harboring galectin-3 has resulted in inhibition of osteoblast differentiation by the secreted galectin-3 into culture medium. The inhibitory effect of galectin-3 was found to be through its binding to Notch1 in a sugar-dependent manner that has led to accelerated Notch1 cleavage and activation of Notch signaling. Taken together, our findings show that soluble galectin-3 in the bone microenvironment niche regulates bone remodeling through Notch signaling, suggesting a novel bone metastasis therapeutic target.


Asunto(s)
Diferenciación Celular , Galectina 3/metabolismo , Osteoblastos/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Galectina 3/genética , Expresión Génica , Células HEK293 , Humanos , Sialoproteína de Unión a Integrina/genética , Microscopía Confocal , Osteoblastos/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Oncotarget ; 5(8): 2044-51, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24810856

RESUMEN

gp78 is a ubiquitin ligase that plays a vital role in endoplasmic reticulum (ER)-associated degradation (ERAD). Here we report that autocrine motility factor (AMF), also known as phosphoglucose isomerase (PGI), is a novel substrate of gp78. We show that polyubiquitylation of AMF requires cooperative interaction between gp78 and the ubiquitin ligase TRIM25 (tripartite motif-containing protein 25). While TRIM25 mediates the initial round of ubiquitylation, gp78 catalyzes polyubiquitylation of AMF. The E4-like activity of gp78 was illustrated by an in vitro polyubiquitylation assay using Ub-DHFR as a model substrate. We further demonstrate that TRIM25 ubiquitylates gp78 and that overexpression of TRIM25 accelerates the degradation of gp78. Our data suggest that TRIM25 not only cooperates with gp78 in polyubiquitylation of AMF but also gauges the steady-state level of gp78. This study uncovers a previously unknown functional link between gp78 and TRIM25 and provides mechanistic insight into gp78-mediated protein ubiquitylation.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/metabolismo , Receptores del Factor Autocrino de Motilidad/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Proteínas de Motivos Tripartitos , Ubiquitinación
11.
Cancer Res ; 74(8): 2229-37, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24576828

RESUMEN

Autocrine motility factor (AMF) enhances invasion by breast cancer cells, but how its secretion and effector signaling are controlled in the tumor microenvironment is not fully understood. In this study, we investigated these issues with a chimeric AMF that is secreted at high levels through a canonical endoplasmic reticulum (ER)/Golgi pathway. Using this tool, we found that AMF enhances tumor cell motility by activating AKT/ERK, altering actin organization, and stimulating ß-catenin/TCF and activating protein 1 transcription. EGF enhanced secretion of AMF through its casein kinase II-mediated phosphorylation. RNA interference-mediated attenuation of AMF expression inhibited EGF-induced invasion by suppressing extracellular signal-regulated kinase signaling. Conversely, exogenous AMF overcame the inhibitory effect of EGF receptor inhibitor gefitinib on invasive motility by activating HER2 signaling. Taken together, our findings show how AMF modulates EGF-induced invasion while affecting acquired resistance to cytotoxic drugs in the tumor microenvironment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Glucosa-6-Fosfato Isomerasa/metabolismo , Línea Celular Tumoral , Sistema Enzimático del Citocromo P-450/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucosa-6-Fosfato Isomerasa/genética , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Sistema de Señalización de MAP Quinasas , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción AP-1/metabolismo , Transfección , Microambiente Tumoral , beta Catenina/metabolismo
12.
Cell Cycle ; 13(23): 3750-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25551360

RESUMEN

Protein phosphatase 2A (PP2A) is the major serine-threonine phosphatase that regulates a number of cell signaling pathways. PP2A activity is controlled partially through protein degradation; however, the underlying mechanism is not fully understood. Here we show that PP2A/C, a catalytic subunit of PP2A, is degraded by the Cullin3 (Cul3) ligase-mediated ubiquitin-proteasome pathway. In response to death receptor signaling by tumor-necrosis factor-related apoptosis-inducing ligand (TRAIL), PP2A/C, caspase-8 and Cul3, a subunit of the cullin family of E3 ligases, are recruited into the death-inducing signaling complex (DISC) where the Cul3 ligase targets PP2A/C for ubiquitination and subsequent degradation. Functionally, knockdown of PP2A/C expression by siRNA or pharmacological inhibition of PP2A activity increases TRAIL-induced apoptosis. In cancer cells that have developed acquired TRAIL resistance, PP2A phosphatase activity is increased, and PP2A/C protein is resistant to TRAIL-induced degradation. Thus, this work identifies a new mechanism by which PP2A/C is regulated by Cul3 ligase-mediated degradation in response to death receptor signaling and suggests that inhibition of PP2A/C degradation may contribute to resistance of cancer cells to death receptor-induced apoptosis.


Asunto(s)
Dominio Catalítico/fisiología , Proteínas Cullin/fisiología , Proteína Fosfatasa 2/metabolismo , Transducción de Señal/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ubiquitinación/fisiología , Línea Celular Tumoral , Células HEK293 , Humanos
13.
Cancer Res ; 73(4): 1411-9, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23248119

RESUMEN

Trastuzumab (Herceptin) is an effective targeted therapy in HER2-overexpressing human breast carcinoma. However, many HER2-positive patients initially or eventually become resistant to this treatment, so elucidating mechanisms of trastuzumab resistance that emerge in breast carcinoma cells is clinically important. Here, we show that autocrine motility factor (AMF) binds to HER2 and induces cleavage to the ectodomain-deleted and constitutively active form p95HER2. Mechanistic investigations indicated that interaction of AMF with HER2 triggers HER2 phosphorylation and metalloprotease-mediated ectodomain shedding, activating phosphoinositide-3-kinase (PI3K) and mitogen-activated protein kinase signaling and ablating the ability of trastuzumab to inhibit breast carcinoma cell growth. Furthermore, we found that HER2 expression and AMF secretion were inversely related in breast carcinoma cells. On the basis of this evidence that AMF may contribute to HER2-mediated breast cancer progression, our findings suggest that AMF-HER2 interaction might be a novel target for therapeutic management of patients with breast cancer, whose disease is resistant to trastuzumab.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Femenino , Glucosa-6-Fosfato Isomerasa/genética , Glucosa-6-Fosfato Isomerasa/farmacología , Células HEK293 , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Interferencia de ARN , Receptor ErbB-2/genética , Receptores del Factor Autocrino de Motilidad/genética , Receptores del Factor Autocrino de Motilidad/metabolismo , Trastuzumab
14.
J Biol Chem ; 287(8): 5192-8, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22232548

RESUMEN

Galectin-3 is a chimeric carbohydrate-binding protein, which interacts with cell surface carbohydrate-containing molecules and extracellular matrix glycoproteins and has been implicated in various biological processes such as cell growth, angiogenesis, motility, and metastasis. It is expressed in a wide range of tumor cells and is associated with tumor progression. The functions of galectin-3 are dependent on its localization and post-translational modifications such as cleavage and phosphorylation. Recently, we showed that galectin-3 Tyr-107 is phosphorylated by c-Abl; concomitantly, it was also shown that galectin-3 can be cleaved at this site by prostate-specific antigen (PSA), a chymotrypsin-like serine protease, after Tyr-107, resulting in loss of galectin-3 multivalency while preserving its carbohydrate binding activity. Galectin-3 is largely a monomer in solution but may form a homodimer by self-association through its carbohydrate recognition domain, whereas, in the presence of a ligand, galectin-3 polymerizes up to pentamers utilizing its N-terminal domain. Oligomerization is a unique feature of secreted galectin-3, which allows its function by forming ordered galectin-glycan structures, i.e. lattices, on the cell surface or through direct engagement of specific cell surface glycoconjugates by traditional ligand-receptor binding. We questioned whether Tyr-107 phosphorylation by c-Abl affects galectin-3 cleavage by PSA. The data suggest a role for galectin-3 in prostate cells associated with increased activity of c-Abl kinase and loss of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) activity. In addition, the ratio of phosphorylated/dephosphorylated galectin-3 might be used as a complementary value to that of PSA for prognosis of prostate cancer and a novel therapeutic target for the treatment of prostate cancer.


Asunto(s)
Galectina 3/química , Galectina 3/metabolismo , Antígeno Prostático Específico/metabolismo , Proteolisis , Tirosina/metabolismo , Línea Celular Tumoral , Quimiotaxis , Espacio Extracelular/metabolismo , Humanos , Modelos Moleculares , Neovascularización Patológica/metabolismo , Fosfohidrolasa PTEN/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Conformación Proteica , Proteínas Proto-Oncogénicas c-abl/metabolismo
15.
Gut ; 59(7): 907-17, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20581239

RESUMEN

BACKGROUND AND AIMS: 90K, a tumour-associated glycoprotein, interacts with galectins and has roles in host defence by augmenting the immune response, but the serum 90K level was suggested to indicate poor prognosis in several cancers. The cellular mechanisms of 90K action on colorectal cancer (CRC) cell motility and its effect on CRC progression were investigated. METHODS: The impact of 90K was analysed by combining cell cultures, in vitro assays, and immunohistochemistry. RESULTS: Secreted 90K suppresses CRC cell invasion, but this action of 90K is masked through binding with extracellular galectins. A novel pathway is identified comprising a secretory 90K and a CD9/CD82 tetraspanin web; in this pathway, 90K interacts with CD9/CD82, suppresses the Wnt/beta-catenin signal via a novel proteasomal-ubiquitination mechanism of beta-catenin that is dependent on ISG15 (interferon-stimulated gene-15) modification (ISGylation) but not on glycogen synthase kinase 3beta (GSK-3beta) and Siah/Adenomatous polyposis coli (APC). In a syngeneic mouse colon tumour model, tumour growth and lung metastasis were increased with 90K knockdown. In colon tissues from stage IV human CRC and invading cancer cells of corresponding metastatic liver tissues, in which beta-catenin and galectin expression was higher, immunostained 90K and CD9/CD82 were lower than in adjacent hepatic tissues or colon tissues from stage I. CONCLUSIONS: 90K itself has antitumour activity in CRC cells via suppression of Wnt signalling with a novel mechanism of ISGylation-dependent ubiquitination of beta-catenin when it interacts with CD9/CD82, but is downregulated in advanced CRC tissues. The data suggest a strategy of strengthening this novel pathway with concomitant knockdown of galectins as a potential therapeutic approach to CRC progression.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias Colorrectales/metabolismo , Glicoproteínas/metabolismo , Proteína Kangai-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , beta Catenina/metabolismo , Animales , Antígenos de Neoplasias , Biomarcadores de Tumor , Proteínas Portadoras , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Medios de Cultivo Condicionados/farmacología , Citocinas/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Galectinas/metabolismo , Glicoproteínas/fisiología , Humanos , Lactosa/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Proteínas de Neoplasias/fisiología , Transducción de Señal/fisiología , Tetraspanina 29 , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Ubiquitinas/fisiología , Proteínas Wnt/metabolismo
16.
FEBS Lett ; 580(2): 669-76, 2006 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-16412436

RESUMEN

Previously, we reported that decreased brain-specific angiogenesis inhibitor 2 (BAI2) induced increased VEGF expression. The regulatory mechanisms for this process are not understood. Here we show that GA-binding protein gamma (GABPgamma) associates with the cytoplasmic domain of BAI2, and GABPalpha/gamma or GABPalpha/beta works as a transcriptional repressor of VEGF in SHSY5Y cells. Transcriptional activity of wild-type VEGF promoter was significantly increased in anti-sense BAI2-transfected cells, but not that of VEGF promoter harboring mutated GABP sites. In in vivo focal cerebral ischemia model, the decrease in BAI2 accompanied by decreased GABPalpha and GABPgamma elicited increased VEGF expression before the onset of HIF-1alpha. Our results point out that BAI2 controls VEGF transcription through GABP under normal conditions and cerebral ischemia.


Asunto(s)
Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Línea Celular , Factor de Transcripción de la Proteína de Unión a GA/genética , Humanos , Proteínas de la Membrana , Ratones , Proteínas del Tejido Nervioso/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Técnicas del Sistema de Dos Híbridos , Factor A de Crecimiento Endotelial Vascular/genética
17.
Cancer Res ; 65(19): 8993-9003, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16204073

RESUMEN

KITENIN promotes invasion of mouse colon adenocarcinoma (CT-26) cells in vivo. Here, we studied the effects of in vivo KITENIN ablation on established tumors by using pSUPER vectors (pSUPER-KITENIN) producing short interfering RNA (siRNA). When pSUPER-KITENIN was given weekly or semiweekly for 1 month into tail vein of syngeneic mice that have established colon tumors, tumor size regressed markedly and metastases were inhibited. In mice injected with pSUPER-KITENIN, serum interleukin-2 (IL-2) and IFN-gamma increased and CD4+ and CD8+ T cells infiltrated in the regressed tumor tissues. These effects, observed beginning 2 days after i.v. injection, imply that immune response is involved in the antitumor action of pSUPER-KITENIN. Using a yeast two-hybrid assay, we identified two KITENIN-interacting proteins for the possible mediators of these actions: 90K protein, a known immune modulatory glycoprotein, and protein kinase C inhibitor (PKCI). 90K was increased in the culture medium from CT-26/antisense KITENIN/90K cells. Double culture of accessory cells with CT-26/antisense KITENIN/90K cells revealed increased secretion of IL-1 and IL-6. Overexpression of 90K in CT-26/antisense KITENIN cells further delayed tumor growth compared with that of CT-26/antisense KITENIN cells. Actin arrangement was distorted in CT-26/antisense KITENIN and CT-26/antisense PKCI cells, whereas overexpression of PKCI resulted in increased invasiveness to fibronectin. Thus, antitumor effects of KITENIN siRNA derives from both the generation of a tumor-specific immune response in vivo through increased 90K secretion from tumor cells and the suppression of tumor invasion in which PKCI is related to increased invasiveness. Moreover, siRNA targeting of KITENIN can function as a chemotherapeutic strategy against colon cancer.


Asunto(s)
Adenocarcinoma/terapia , Proteínas Portadoras/antagonistas & inhibidores , Neoplasias del Colon/terapia , Proteínas de la Membrana/antagonistas & inhibidores , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Antígenos de Neoplasias , Biomarcadores de Tumor , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , ADN sin Sentido/genética , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Inyecciones Intravenosas , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa C/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/inmunología , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Bacteriol ; 187(17): 5984-95, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16109940

RESUMEN

The manganese-containing superoxide dismutase (MnSOD) of Vibrio vulnificus, normally detected after the onset of the stationary phase, is expressed during the lag that immediately follows the transfer of cells grown exponentially to a fresh medium acidified to pH 5.0, whereas Fe-containing SOD is constitutively expressed. The signal triggering the growth lag and MnSOD induction therein is not low pH but intracellular superoxide accumulated under these conditions, since addition of a superoxide scavenger not only shortened the lag but also abrogated the MnSOD induction. If the lysine decarboxylase reaction proceeds in the presence of sufficient lysine, the broth is rapidly neutralized to abolish the generation of oxidative stress. Accordingly, the acid tolerance response was examined without the addition of lysine. SoxR regulates MnSOD induction. Lack of MnSOD caused by mutations in soxR or sodA resulted in low tolerance to low pH. The fur mutant derepressing MnSOD showed better tolerance than the wild type. Thus, an increase in total cytosolic SOD activity through MnSOD induction is essential for the cell to withstand the acid challenge. The contribution of cuprozinc-containing SOD to acid tolerance is not significant compared with those of cytosolic SODs.


Asunto(s)
Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Vibrio vulnificus/enzimología , Secuencia de Bases , Carboxiliasas/genética , Carboxiliasas/metabolismo , Conjugación Genética , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Concentración de Iones de Hidrógeno , Cinética , Mutagénesis , Plásmidos , Mapeo Restrictivo , Transcripción Genética , Vibrio vulnificus/genética , Vibrio vulnificus/crecimiento & desarrollo
19.
FEMS Microbiol Lett ; 234(2): 261-7, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15135531

RESUMEN

Rhodobacter sphaeroides has a cuprozinc-containing superoxide dismutase (CuZnSOD) in its periplasm in addition to the cytoplasmic SOD that appears to contain iron (FeSOD). The FeSOD is constitutively expressed under the growth conditions examined, whereas the CuZnSOD is detected only when the light-harvesting complexes are found. The CuZnSOD expression is regulated post-transcriptionally, and the enzyme appears to protect the photoheterotrophic cells from periplasmic superoxide that may be generated by exposure of the cells to low O2 under the illuminated conditions at 10 W/m2.


Asunto(s)
Rhodobacter sphaeroides/enzimología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Citosol/enzimología , Isoenzimas/genética , Isoenzimas/metabolismo , Mutagénesis , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/crecimiento & desarrollo , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...