Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Gan To Kagaku Ryoho ; 48(3): 331-335, 2021 Mar.
Artículo en Japonés | MEDLINE | ID: mdl-33790151

RESUMEN

BACKGROUND: Though irinotecan is commonly used for treating advanced gastric cancer, there is no predictive biomarker to date. We have studied the resistant mechanism for irinotecan and found that phosphorylation of serine 10 residue of topoisomerase Ⅰ(topo Ⅰ)is an important step for irinotecan resistance. We have developed an immunohistochemical staining-based biomarker; topo Ⅰ-pS10, for predicting irinotecan efficacy. PURPOSE: The purpose of this study is to test the accuracy of topo Ⅰ-pS10 immunohistochemical staining in gastric cancer clinical samples. METHODS: In this study we performed 2 sets of tests. In the training set, we stained 79 gastric cancer clinical samples which efficacy of irinotecan was measured by succinate dehydrogenase inhibition(SDI)test. In the validation set, we used 27 gastric cancer clinical samples which irinotecan was used and the efficacy was known. RESULTS: Training set: From the ROC curve the cut-off point was set at 35% positive nuclei. Sixty three cases were positive with topo Ⅰ-pS10 in the nuclei. With the result of irinotecan SDI, the sensitivity was 76.6% and the positive predictive value was 92.5%. This result showed that topo Ⅰ-pS10 positive case does not respond to irinotecan. Validation set: In this set, the sensitivity was 82.4% and the positive predictive value was 82.4%. CONCLUSION: topo Ⅰ-pS10 staining can be used as a predictive biomarker for irinotecan for gastric cancer patients.


Asunto(s)
ADN-Topoisomerasas de Tipo I , Neoplasias Gástricas , Biomarcadores , Camptotecina , ADN-Topoisomerasas de Tipo I/metabolismo , Humanos , Irinotecán , Neoplasias Gástricas/tratamiento farmacológico
2.
Clin Colorectal Cancer ; 20(2): e129-e138, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33731288

RESUMEN

PURPOSE: The camptothecin (CPT) analogs topotecan and irinotecan specifically target topoisomerase I (topoI) and are used to treat colorectal, gastric, and pancreatic cancer. Response rate for this class of drug varies from 10% to 30%, and there is no predictive biomarker for patient stratification by response. On the basis of our understanding of CPT drug resistance mechanisms, we developed an immunohistochemistry-based predictive test, P-topoI-Dx, to stratify the patient population into those who did and did not experience a response. PATIENTS AND METHODS: The retrospective validation studies included a training set (n = 79) and a validation cohort (n = 27) of gastric cancer (GC) patients, and 8 cohorts of colorectal cancer (CRC) patient tissue (n = 176). Progression-free survival for 6 months was considered a positive response to CPT-based therapy. Formalin-fixed, paraffin-embedded slides were immunohistochemically stained with anti-phospho-specific topoI-Serine10 (topoI-pS10), quantitated, and analyzed statistically. RESULTS: We determined a threshold of 35% positive staining to offer optimal test characteristics in GC. The GC (n = 79) training set demonstrated 76.6% (95% confidence interval, 64-86) sensitivity; 68.8% (41-88) specificity; positive predictive value (PPV) 92.5% (81-98); and negative predictive value (NPV) 42.3% (24-62). The GC validation set (n = 27) demonstrated 82.4% (56-95) sensitivity and 70.0% (35-92) specificity. Estimated PPV and NPV were 82.4% (56-95) and 70.0% (35-92) respectively. In the CRC validation set (n = 176), the 40% threshold demonstrated 87.5% (78-94) sensitivity; 70.0% (59-79) specificity; PPV 70.7% (61-79); and NPV 87.0 % (77-93). CONCLUSION: The analysis of retrospective data from patients (n = 282) provides clinical validity to our P-topoI-Dx immunohistochemical test to identify patients with disease that is most likely to respond to topoI inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Estudios Retrospectivos , Medición de Riesgo/métodos
3.
PLoS One ; 15(8): e0228002, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32764831

RESUMEN

Irinotecan specifically targets topoisomerase I (topoI), and is used to treat various solid tumors, but only 13-32% of patients respond to the therapy. Now, it is understood that the rapid rate of topoI degradation in response to irinotecan causes irinotecan resistance. We have published that the deregulated DNA-PKcs kinase cascade ensures rapid degradation of topoI and is at the core of the drug resistance mechanism of topoI inhibitors, including irinotecan. We also identified CTD small phosphatase 1 (CTDSP1) (a nuclear phosphatase) as a primary upstream regulator of DNA-PKcs in response to topoI inhibitors. Previous reports showed that rabeprazole, a proton pump inhibitor (PPI) inhibits CTDSP1 activity. The purpose of this study was to confirm the effects of rabeprazole on CTDSP1 activity and its impact on irinotecan-based therapy in colon cancer. Using differentially expressing CTDSP1 cells, we demonstrated that CTDSP1 contributes to the irinotecan sensitivity by preventing topoI degradation. Retrospective analysis of patients receiving irinotecan with or without rabeprazole has shown the effects of CTDSP1 on irinotecan response. These results indicate that CTDSP1 promotes sensitivity to irinotecan and rabeprazole prevents this effect, resulting in drug resistance. To ensure the best chance at effective treatment, rabeprazole may not be a suitable PPI for cancer patients treated with irinotecan.


Asunto(s)
Neoplasias Colorrectales/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , Rabeprazol/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias Colorrectales/fisiopatología , ADN , ADN-Topoisomerasas de Tipo I/fisiología , Proteína Quinasa Activada por ADN/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Humanos , Irinotecán/metabolismo , Irinotecán/farmacología , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Inhibidores de la Bomba de Protones/farmacología , Rabeprazol/farmacología , Estudios Retrospectivos , Inhibidores de Topoisomerasa I/farmacología
4.
JCI Insight ; 3(12)2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29925694

RESUMEN

Mucin 1 (MUC1) is a heterodimeric protein that is aberrantly overexpressed on the surface of diverse human carcinomas and is an attractive target for the development of mAb-based therapeutics. However, attempts at targeting the shed MUC1 N-terminal subunit have been unsuccessful. We report here the generation of mAb 3D1 against the nonshed oncogenic MUC1 C-terminal (MUC1-C) subunit. We show that mAb 3D1 binds with low nM affinity to the MUC1-C extracellular domain at the restricted α3 helix. mAb 3D1 reactivity is selective for MUC1-C-expressing human cancer cell lines and primary cancer cells. Internalization of mAb 3D1 into cancer cells further supported the conjugation of mAb 3D1 to monomethyl auristatin E (MMAE). The mAb 3D1-MMAE antibody-drug conjugate (ADC) (a) kills MUC1-C-positive cells in vitro, (b) is nontoxic in MUC1-transgenic (MUC1.Tg) mice, and (c) is active against human HCC827 lung tumor xenografts. Humanized mAb (humAb) 3D1 conjugated to MMAE also exhibited antitumor activity in (a) MUC1.Tg mice harboring syngeneic MC-38/MUC1 tumors, (b) nude mice bearing human ZR-75-1 breast tumors, and (c) NCG mice engrafted with a patient-derived triple-negative breast cancer. These findings and the absence of associated toxicities support clinical development of humAb 3D1-MMAE ADCs as a therapeutic for the many cancers with MUC1-C overexpression.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoconjugados/farmacología , Mucina-1/inmunología , Proteínas Oncogénicas/inmunología , Animales , Anticuerpos Monoclonales/química , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Desnudos , Modelos Moleculares , Mucina-1/química , Oligopéptidos , Proteínas Oncogénicas/química , Conformación Proteica en Hélice alfa , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Histochem Cytochem ; 66(8): 549-561, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29587004

RESUMEN

Phosphorylation is the most extensively studied posttranslational modification of proteins. There are approximately 500 kinases known in the human genome. The kinase-activated pathways regulate almost every aspect of cell function and a deregulated kinase cascade leads to impaired cellular function. Impaired regulation of several kinase cascades, including the epidermal growth factor receptor (EGFR) pathway, leading to tumor pathogenesis, is well documented. Thus, a phosphospecific test with prognostic or predictive value was expected in oncology. However, no phosphospecific IHC test is used in oncology clinics. Human topoisomerase I (topoI) inhibitors, camptothecin and its analogues (CPT), are used extensively to treat various solid tumors. Depending on tumor type, the response rate is only 13-32%. We have demonstrated that the deregulated kinase cascade is at the core of CPT resistance. DNA-PKcs, a kinase central to the DNA-double-strand break (DSB) response pathway, phosphorylates topoI at serine 10 (topoI-pS10), and cells with higher basal levels of topoI-pS10 degrade topoI rapidly and are resistant to this class of drug. The higher basal level of topoI phosphorylation is due to continual activation of DNA-PKcs, and one potential mechanism of this pathway activation is failure of upstream effector phosphatases such as phosphatase and tensin homolog (PTEN). Based on this understanding, we have developed an IHC-based test (P-topoIDx) that can stratify the responder and non-responder patient population.


Asunto(s)
Camptotecina/farmacología , ADN-Topoisomerasas de Tipo I/análisis , Inmunohistoquímica/métodos , Inhibidores de Topoisomerasa I/farmacología , Animales , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo I/inmunología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosforilación
6.
Oncotarget ; 8(27): 43733-43751, 2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28415827

RESUMEN

Proteasomal degradation of topoisomerase I (topoI) is one of the most remarkable cellular phenomena observed in response to camptothecin (CPT). Importantly, the rate of topoI degradation is linked to CPT resistance. Formation of the topoI-DNA-CPT cleavable complex inhibits DNA re-ligation resulting in DNA-double strand break (DSB). The degradation of topoI marks the first step in the ubiquitin proteasome pathway (UPP) dependent DNA damage response (DDR). Here, we show that the Ku70/Ku80 heterodimer binds with topoI, and that the DNA-dependent protein kinase (DNA-PKcs) phosphorylates topoI on serine 10 (topoI-pS10), which is subsequently ubiquitinated by BRCA1. A higher basal level of topoI-pS10 ensures rapid topoI degradation leading to CPT resistance. Importantly, PTEN regulates DNA-PKcs kinase activity in this pathway and PTEN deletion ensures DNA-PKcs dependent higher topoI-pS10, rapid topoI degradation and CPT resistance.


Asunto(s)
Camptotecina/farmacología , ADN-Topoisomerasas de Tipo I/metabolismo , Resistencia a Antineoplásicos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Topoisomerasa I/farmacología , Ubiquitina/metabolismo , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Edición Génica , Humanos , Autoantígeno Ku/metabolismo , Complejos Multiproteicos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Unión Proteica , Proteína Quinasa C/metabolismo , Proteolisis , Interferencia de ARN
7.
PLoS One ; 10(8): e0135156, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26267657

RESUMEN

Mucin 1 (MUC1) is a heterodimeric protein that is aberrantly expressed in diverse human carcinomas and certain hematologic malignancies. The oncogenic MUC1 transmembrane C-terminal subunit (MUC1-C) functions in part by transducing growth and survival signals from cell surface receptors. However, little is known about the structure of the MUC1-C cytoplasmic domain as a potential drug target. Using methods for structural predictions, our results indicate that a highly conserved CQCRRK sequence, which is adjacent to the cell membrane, forms a small pocket that exposes the two cysteine residues for forming disulfide bonds. By contrast, the remainder of the MUC1-C cytoplasmic domain has no apparent structure, consistent with an intrinsically disordered protein. Our studies thus focused on targeting the MUC1 CQCRRK region. The results show that L- and D-amino acid CQCRRK-containing peptides bind directly to the CQC motif. We further show that the D-amino acid peptide, designated GO-203, blocks homodimerization of the MUC1-C cytoplasmic domain in vitro and in transfected cells. Moreover, GO-203 binds directly to endogenous MUC1-C in breast and lung cancer cells. Colocalization studies further demonstrate that GO-203 predominantly binds to MUC1-C at the cell membrane. These findings support the further development of agents that target the MUC1-C cytoplasmic domain CQC motif and thereby MUC1-C function in cancer cells.


Asunto(s)
Mucina-1/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular Tumoral , Células HEK293 , Humanos , Datos de Secuencia Molecular , Mucina-1/metabolismo , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas
8.
Mol Cancer Ther ; 13(3): 576-84, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24327519

RESUMEN

Small cell lung cancer (SCLC) is a devastating disease, and current therapies have not greatly improved the 5-year survival rates. Topoisomerase (Top) inhibition is a treatment modality for SCLC; however, the response is short lived. Consequently, our research has focused on improving SCLC therapeutics through the identification of novel targets. Previously, we identified MNNG HOS transforming gene (MET) to be overexpressed and functional in SCLC. Herein, we investigated the therapeutic potential of combinatorial targeting of MET using SU11274 and Top1 using 7-ethyl-10-hydroxycamptothecin (SN-38). MET and TOP1 gene copy numbers and protein expression were determined in 29 patients with limited (n = 11) and extensive (n = 18) disease. MET gene copy number was significantly increased (>6 copies) in extensive disease compared with limited disease (P = 0.015). Similar TOP1 gene copy numbers were detected in limited and extensive disease. Immunohistochemical staining revealed a significantly higher Top1 nuclear expression in extensive (0.93) versus limited (0.15) disease (P = 0.04). Interestingly, a significant positive correlation was detected between MET gene copy number and Top1 nuclear expression (r = 0.5). In vitro stimulation of H82 cells revealed hepatocyte growth factor (HGF)-induced nuclear colocalization of p-MET and Top1. Furthermore, activation of the HGF/MET axis enhanced Top1 activity, which was abrogated by SU11274. Combination of SN-38 with SU11274 dramatically decreased SCLC growth as compared with either drug alone. Collectively, these findings suggest that the combinatorial inhibition of MET and Top1 is a potentially efficacious treatment strategy for SCLC.


Asunto(s)
Carcinoma de Células Pequeñas/tratamiento farmacológico , ADN-Topoisomerasas de Tipo I/genética , Proteínas Proto-Oncogénicas c-met/genética , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dosificación de Gen/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/administración & dosificación , Irinotecán , Persona de Mediana Edad , Piperazinas/administración & dosificación , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Sulfonamidas/administración & dosificación , Inhibidores de Topoisomerasa I/administración & dosificación
9.
Biochem Biophys Res Commun ; 418(1): 116-21, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22244893

RESUMEN

The intracellular domain of ErbB4 receptor tyrosine kinase is known to translocate to the nucleus of cells where it can regulate p53 transcriptional activity. The purpose of this study was to examine whether ErbB4 can localize to the nucleus of adult rat ventricular myocytes (ARVM), and regulate p53 in these cells. We demonstrate that ErbB4 does locate to the nucleus of cardiac myocytes as a full-length protein, although nuclear location occurs as a full-length protein that does not require Protein Kinase C or γ-secretase activity. Consistent with this we found that only the non-cleavable JM-b isoform of ErbB4 is expressed in ARVM. Doxorubicin was used to examine ErbB4 role in regulation of a DNA damage response in ARVM. Doxorubicin induced p53 and p21 was suppressed by treatment with AG1478, an EGFR and ErbB4 kinase inhibitor, or suppression of ErbB4 expression with small interfering RNA. Thus ErbB4 localizes to the nucleus as a full-length protein, and plays a role in the DNA damage response induced by doxorubicin in cardiac myocytes.


Asunto(s)
Núcleo Celular/enzimología , Daño del ADN , Receptores ErbB/metabolismo , Miocitos Cardíacos/enzimología , Animales , Núcleo Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Doxorrubicina/farmacología , Receptores ErbB/antagonistas & inhibidores , Ventrículos Cardíacos/citología , Ratones , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Ratas , Receptor ErbB-4 , Proteína p53 Supresora de Tumor/metabolismo , Tirfostinos/farmacología
10.
Mol Microbiol ; 81(5): 1390-401, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21797942

RESUMEN

Anthrax toxin is an A/B bacterial protein toxin which is composed of the enzymatically active Lethal Factor (LF) and/or Oedema Factor (EF) bound to Protective Antigen 63 (PA63) which functions as both the receptor binding and transmembrane domains. Once the toxin binds to its cell surface receptors it is internalized into the cell and traffics through Rab5- and Rab7-associated endosomal vesicles. Following acidification of the vesicle lumen, PA63 undergoes a dynamic change forming a beta-barrel that inserts into and forms a pore through the endosomal membrane. It is widely recognized that LF, and the related fusion protein LFnDTA, must be completely denatured in order to transit through the PA63 formed pore and enter the eukaryotic cell cytosol. We demonstrate by protease protection assays that the molecular chaperone GRP78 mediates the unfolding of LFnDTA and LF at neutral pH and thereby converts these proteins from a trypsin resistant to sensitive conformation. We have used immunoelectron microscopy and gold-labelled antibodies to demonstrate that both GRP78 and GRP94 chaperones are present in the lumen of endosomal vesicles. Finally, we have used siRNA to demonstrate that knock-down of GRP78 results in the emergence of resistance to anthrax lethal toxin and oedema toxin action.


Asunto(s)
Antígenos Bacterianos/química , Antígenos Bacterianos/metabolismo , Bacillus anthracis/química , Bacillus anthracis/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Proteínas de Choque Térmico/metabolismo , Transporte de Proteínas/fisiología , Animales , Carbunco/metabolismo , Carbunco/microbiología , Proteínas Bacterianas/metabolismo , Línea Celular , Citosol/enzimología , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/genética , Glicoproteínas de Membrana/biosíntesis , Ratones , Desplegamiento Proteico , Interferencia de ARN , ARN Interferente Pequeño , Tripsina/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Proteínas de Unión a GTP rab7
11.
PLoS One ; 5(11): e13830, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-21085490

RESUMEN

BACKGROUND: Many inducible transcription factors are regulated through batteries of posttranslational modifications that couple their activity to inducing stimuli. We have studied such regulation of Heat Shock Factor 1 (HSF1), a key protein in control of the heat shock response, and a participant in carcinogenisis, neurological health and aging. As the mechanisms involved in the intracellular regulation of HSF1 in good health and its dysregulation in disease are still incomplete we are investigating the role of posttranslational modifications in such regulation. METHODOLOGY/PRINCIPAL FINDINGS: In a proteomic study of HSF1 binding partners, we have discovered its association with the pleiotropic protein kinase A (PKA). HSF1 binds avidly to the catalytic subunit of PKA, (PKAcα) and becomes phosphorylated on a novel serine phosphorylation site within its central regulatory domain (serine 320 or S320), both in vitro and in vivo. Intracellular PKAcα levels and phosphorylation of HSF1 at S320 were both required for HSF1 to be localized to the nucleus, bind to response elements in the promoter of an HSF1 target gene (hsp70.1) and activate hsp70.1 after stress. Reduction in PKAcα levels by small hairpin RNA led to HSF1 exclusion from the nucleus, its exodus from the hsp70.1 promoter and decreased hsp70.1 transcription. Likewise, null mutation of HSF1 at S320 by alanine substitution for serine led to an HSF1 species excluded from the nucleus and deficient in hsp70.1 activation. CONCLUSIONS: These findings of PKA regulation of HSF1 through S320 phosphorylation add to our knowledge of the signaling networks converging on this factor and may contribute to elucidating its complex roles in the stress response and understanding HSF1 dysregulation in disease.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Sustitución de Aminoácidos , Sitios de Unión/genética , Dominio Catalítico/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas de Unión al ADN/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Células HeLa , Factores de Transcripción del Choque Térmico , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Confocal , Mutación , Fosforilación , Unión Proteica , Interferencia de ARN , Elementos de Respuesta/genética , Serina/genética , Serina/metabolismo , Factores de Transcripción/genética
12.
Curr Proteomics ; 7(1): 49-65, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20526421

RESUMEN

Lung cancer is the leading cause of cancer death for both men and women in the United States, and similar trends are seen world wide. The lack of early diagnosis is one of the primary reasons for the high mortality rate. A number of biomarkers have been evaluated in lung cancer patients, however, their specificity and early stage diagnostic values are limited. Using traditional protein chemistry and proteomics tool we have demonstrated higher serum haptoglobin levels in small cell lung cancer (SCLC). Similar findings have been reported for other cancers including ovarian cancer and glioblastoma. Haptoglobin is an acute phase protein with at least six possible phenotypes. The six phenotypes, in combination with two post translational modifications, glycosylation and deamidation, lead to large numbers of possible haptoglobin isoforms. Recent studies indicate a possible correlation between specific haptoglobin glycosylation and particular disease conditions. In our current study, we have fractionated control and SCLC patient serum by 2-D gel electrophoresis to identify differentially expressed haptoglobin isoforms in SCLC serum samples.

13.
Nat Cell Biol ; 10(10): 1208-16, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18806787

RESUMEN

The von Hippel-Lindau protein pVHL suppresses renal tumorigenesis in part by promoting the degradation of hypoxia-inducible HIF-alpha transcription factors; additional mechanisms have been proposed. pVHL also stabilizes the plant homeodomain protein Jade-1, which is a candidate renal tumour suppressor that may correlate with renal cancer risk. Here we show that Jade-1 binds the oncoprotein beta-catenin in Wnt-responsive fashion. Moreover, Jade-1 destabilizes wild-type beta-catenin but not a cancer-causing form of beta-catenin. Whereas the well-established beta-catenin E3 ubiquitin ligase component beta-TrCP ubiquitylates only phosphorylated beta-catenin, Jade-1 ubiquitylates both phosphorylated and non-phosphorylated beta-catenin and therefore regulates canonical Wnt signalling in both Wnt-off and Wnt-on phases. Thus, the different characteristics of beta-TrCP and Jade-1 may ensure optimal Wnt pathway regulation. Furthermore, pVHL downregulates beta-catenin in a Jade-1-dependent manner and inhibits Wnt signalling, supporting a role for Jade-1 and Wnt signalling in renal tumorigenesis. The pVHL tumour suppressor and the Wnt tumorigenesis pathway are therefore directly linked through Jade-1.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular , Humanos , Unión Proteica , Proteínas Wnt/antagonistas & inhibidores , Xenopus
14.
Proc Natl Acad Sci U S A ; 105(13): 5254-9, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18356299

RESUMEN

The delivery of the diphtheria toxin catalytic domain (DTA) from acidified endosomes into the cytoplasm of eukaryotic cells requires protein-protein interactions between the toxin and a cytosolic translocation factor (CTF) complex. A conserved peptide motif, T1, within the DT transmembrane helix 1 mediates these interactions. Because the T1 motif is also present in the N-terminal segments of lethal factor (LF) and edema factor (EF) in anthrax toxin, we asked whether LF entry into the cell might also be facilitated by target cell cytosolic proteins. In this study, we have used LFnDTA and its associated ADP-ribosyltransferase activity (DTA) to determine the requirements for LF translocation from the lumen of endosomal vesicles to the external medium in vitro. Although low-level release of LFnDTA from enriched endosomal vesicles occurs in the absence of added factors, translocation was enhanced by the addition of cytosolic proteins and ATP to the reaction mixture. We show by GST-LFn pull-down assays that LFn specifically interacts with at least zeta-COP and beta-COP of the COPI coatomer complex. Immunodepletion of COPI coatomer complex and associated proteins from cytosolic extracts blocks in vitro LFnDTA translocation. Translocation may be reconstituted by the addition of partially purified bovine COPI to the translocation assay mixture. Taken together, these data suggest that the delivery of LF to the cytosol requires either COPI coatomer complex or a COPI subcomplex for translocation from the endosomal lumen. This facilitated delivery appears to use a mechanism that is analogous to that of DT entry.


Asunto(s)
Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Proteína Coat de Complejo I/metabolismo , Membranas Intracelulares/metabolismo , Bacillus anthracis/metabolismo , Línea Celular Tumoral , Proteína Coat de Complejo I/genética , Citosol/metabolismo , Humanos , Microscopía Electrónica , Unión Proteica , Subunidades de Proteína/metabolismo , Transporte de Proteínas , Sensibilidad y Especificidad
15.
Mass Spectrom Rev ; 26(3): 451-66, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17407130

RESUMEN

Lung cancer is a devastating illness with an overall poor prognosis. To effectively address this disease, early detection and novel therapeutics are required. Early detection of lung cancer is challenging, in part because of the lack of adequate tumor biomarkers. The goal of this review is to summarize the knowledge of current biomarkers in lung cancer, with a focus on important serum biomarkers. The current knowledge on the known serum cytokines and tumor biomarkers of lung cancer will be presented. Emerging trends and new findings in the search for novel diagnostic and therapeutic tumor biomarkers using proteomics technologies and platforms are emphasized, including recent advances in mass spectrometry to facilitate tumor biomarker discovery program in lung cancer. It is our hope that validation of these new research platforms and technologies will result in improved early detection, prognostication, and finally the treatment of lung cancer with potential novel molecularly targeted therapeutics.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Espectrometría de Masas/tendencias , Proteínas de Neoplasias/análisis , Proteoma/análisis , Proteómica/tendencias , Diagnóstico por Computador/tendencias , Predicción
16.
J Immunol ; 177(9): 5946-55, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17056519

RESUMEN

We have developed a molecular chaperone-based tumor vaccine that reverses the immune tolerance of cancer cells. Heat shock protein (HSP) 70 extracted from fusions of dendritic (DC) and tumor cells (HSP70.PC-F) possess superior properties such as stimulation of DC maturation and T cell proliferation over its counterpart from tumor cells. More importantly, immunization of mice with HSP70.PC-F resulted in a T cell-mediated immune response including significant increase of CD8 T cells and induction of the effector and memory T cells that was able to break T cell unresponsiveness to a nonmutated tumor Ag and provide protection of mice against challenge with tumor cells. By contrast, the immune response to vaccination with HSP70-PC derived from tumor cells is muted against such nonmutated tumor Ag. HSP70.PC-F complexes differed from those derived from tumor cells in a number of key manners, most notably, enhanced association with immunologic peptides. In addition, the molecular chaperone HSP90 was found to be associated with HSP70.PC-F as indicated by coimmunoprecipitation, suggesting ability to carry an increased repertoire of antigenic peptides by the two chaperones. Significantly, activation of DC by HSP70.PC-F was dependent on the presence of an intact MyD88 gene, suggesting a role for TLR signaling in DC activation and T cell stimulation. These experiments indicate that HSP70-peptide complexes (PC) derived from DC-tumor fusion cells have increased their immunogenicity and therefore constitute an improved formulation of chaperone protein-based tumor vaccine.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Proteínas HSP70 de Choque Térmico/inmunología , Complejos Multiproteicos/inmunología , Neoplasias/prevención & control , Animales , Fusión Celular , Proteínas HSP70 de Choque Térmico/aislamiento & purificación , Inmunidad Activa/genética , Inmunización , Ratones , Ratones Noqueados , Mucina-1/análisis , Complejos Multiproteicos/química , Complejos Multiproteicos/aislamiento & purificación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Fragmentos de Péptidos/análisis , Péptidos/inmunología , Linfocitos T/inmunología , Receptores Toll-Like/metabolismo
17.
Tumour Biol ; 27(1): 17-26, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16340246

RESUMEN

The 25-kDa heat shock protein (Hsp25) is associated with various malignancies and is expressed at high levels in biopsies as well as circulating in the serum of breast cancer patients. In this study, we used RNA interference technology to silence the hsp25 gene in 4T1 breast adenocarcinoma cells, known as a poorly immunogenic, highly metastatic cell line. We demonstrate that transfection of 4T1 cells with short interference RNA-Hsp25 dramatically inhibits proliferation as compared with control transfected cells. In addition, we show that 4T1 cells transfected with short interference RNA-Hsp25 abrogates tumor migration potential by a mechanism that is in part due to the repression of matrix metalloproteinase 9 expression and a concomitant upregulation of its antagonist, tissue inhibitor metalloproteinase 1. Taken together, these findings provide a model system for the study of metastatic potential of tumors and are suggestive of an earlier unrecognized role for Hsp25 in tumor migration.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Choque Térmico/biosíntesis , Proteínas de Choque Térmico/genética , Metástasis de la Neoplasia/fisiopatología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Interferencia de ARN , Secuencia de Bases , Movimiento Celular , Proliferación Celular , Femenino , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/fisiología , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Chaperonas Moleculares , Datos de Secuencia Molecular , Proteínas de Neoplasias/fisiología , Transfección , Células Tumorales Cultivadas
18.
Proc Natl Acad Sci U S A ; 102(43): 15635-40, 2005 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-16230620

RESUMEN

A 10-aa motif in transmembrane helix 1 of diphtheria toxin that is conserved in anthrax edema factor, anthrax lethal factor, and botulinum neurotoxin serotypes A, C, and D was identified by blast, clustal w, and meme computational analysis. Using the diphtheria toxin-related fusion protein toxin DAB(389)IL-2, we demonstrate that introduction of the L221E mutation into a highly conserved residue within this motif results in a nontoxic catalytic domain translocation deficient phenotype. To further probe the function of this motif in the process by which the catalytic domain is delivered from the lumen of early endosomes to the cytosol, we constructed a gene encoding a portion of diphtheria toxin transmembrane helix 1, T1, which carries the motif and is expressed from a CMV promoter. We then isolated stable transfectants of Hut102/6TG cells that express the T1 peptide, Hut102/6TG-T1. In contrast to the parental cell line, Hut102/6TG-T1 cells are ca. 10(4)-fold more resistant to the fusion protein toxin. This resistance is completely reversed by coexpression of small interfering RNA directed against the gene encoding the T1 peptide in Hut102/6TG-T1 cells. We further demonstrate by GST-DT140-271 pull-down experiments in the presence and absence of synthetic T1 peptides the specific binding of coatomer protein complex subunit beta to this region of the diphtheria toxin transmembrane domain.


Asunto(s)
Citosol/metabolismo , Toxina Diftérica/química , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Secuencia Conservada , Toxina Diftérica/metabolismo , Datos de Secuencia Molecular , Transporte de Proteínas , Conejos
19.
Oncogene ; 24(43): 6564-73, 2005 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-16007186

RESUMEN

Elevation of heat shock protein (HSP) levels is widespread in cancer and predicts a poor prognosis and resistance to therapy. We show that HSP elevation in tumor cells can be induced by the highly malignant factor heregulin beta1 (HRGbeta1), which induces HSP expression through heat shock transcription factor 1 (HSF1). Inactivation of the hsf1 gene prevents HSP induction by HRGbeta1. HSP expression is induced through a cascade response initiated by HRGbeta1 binding to c-erbB receptors on the cell surface and which leads to the inhibition of intracellular HSF1 antagonist glycogen synthase kinase 3. HSF1 activated by this pathway plays a key role in the protection of cells from apoptosis and the mediation of anchorage independent growth by HRGbeta1, indicating a role for HSF1 in this tumorigenic pathway.


Asunto(s)
Apoptosis/fisiología , Proteínas de Choque Térmico/metabolismo , Neurregulina-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/genética , Humanos , Ratones , Neurregulina-1/farmacología , Proteínas Oncogénicas v-erbB/genética , Proteínas Oncogénicas v-erbB/metabolismo , Transducción de Señal , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
20.
J Biol Chem ; 280(13): 13148-52, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15671027

RESUMEN

Heat shock protein (Hsp) 90 is a ubiquitously expressed chaperone that stabilizes expression of multiple signaling kinases involved in growth regulation, including ErbB2, Raf-1, and Akt. The chaperone activity of Hsp90 requires ATP, which binds with approximately 10-fold lower affinity than ADP. This suggests that Hsp90 may be a physiological ATP sensor, regulating the stability of growth signaling cascades in relation to cellular energy charge. Here we show that lowering ATP concentration by inhibiting glycolysis or mitochondrial respiration in isolated myocytes triggers rapid dissociation of Hsp90 from ErbB2 and degradation of ErbB2 along with other client proteins. The effect of disrupting Hsp90 chaperone activity by ATP depletion was similar to the effect of the pharmacological Hsp90 inhibitor geldanamycin. ATP depletion-induced disruption of Hsp90 chaperone activity was associated with cellular resistance to growth factor activation of intracellular signaling. ErbB2 degradation was also induced by the physiological stress of beta-adrenergic receptor stimulation in electrically stimulated cells. These results support a role for Hsp90 as an ATP sensor that modulates tissue growth factor responsiveness under metabolically stressed conditions and provide a novel mechanism by which cellular responsiveness to growth factor stimulation is modulated by cellular energy charge.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Células Musculares/metabolismo , Miocitos Cardíacos/citología , Receptor ErbB-2/metabolismo , Adenosina Difosfato/química , Adenosina Trifosfato/química , Animales , Benzoquinonas , Proliferación Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Glucólisis , Proteínas HSP90 de Choque Térmico/química , Inmunoprecipitación , Lactamas Macrocíclicas , Ligandos , Masculino , Miocitos Cardíacos/metabolismo , Consumo de Oxígeno , Fosforilación , Unión Proteica , Quinonas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA