Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biomed Sci ; 27(1): 2, 2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-31898491

RESUMEN

BACKGROUND: Serglycin (SRGN), previously recognized as an intracellular proteoglycan involved in the storage processes of secretory granules, has recently been shown to be upregulated in several solid tumors. We have previously shown that SRGN in non-small cell lung cancer (NSCLC) promotes malignant phenotypes in a CD44-dependent manner and increased expression of SRGN predicts poor prognosis of primary lung adenocarcinomas. However, the underlying mechanism remains to be defined. METHODS: Overexpression, knockdown and knockout approaches were performed to assess the role of SRGN in cell motility using wound healing and Boyden chamber migration assays. SRGN devoid of glycosaminoglycan (GAG) modification was produced by site-directed mutagenesis or chondroitinase treatment. Liquid chromatography/tandem mass spectrometry was applied for quantitative analysis of the disaccharide compositions and sulfation extent of SRGN GAGs. Western blot and co-immunoprecipitation analyses were performed to determine the expression and interaction of proteins of interest. Actin cytoskeleton organization was monitored by immunofluorescence staining. RESULTS: SRGN expressed by NSCLC cells is readily secreted to the extracellular matrix in a heavily glycosylated form attached with mainly chondroitin sulfate (CS)-GAG chains, and to a lesser extent with heparin sulfate (HS). The CS-GAG moiety serves as the structural motif for SRGN binding to tumor cell surface CD44 and promotes cell migration. SRGN devoid of CS-GAG modification fails to interact with CD44 and has lost the ability to promote cell migration. SRGN/CD44 interaction promotes focal adhesion turnover via Src-mediated paxillin phosphorylation and disassembly of paxillin/FAK adhesion complex, facilitating cell migration. In support, depletion of Src activity or removal of CS-GAGs efficiently blocks SRGN-mediated Src activation and cell migration. SRGN also promotes cell migration via inducing cytoskeleton reorganization mediated through RAC1 and CDC42 activation accompanied with increased lamellipodia and filopodia formation. CONCLUSIONS: Proteoglycan SRGN promotes NSCLC cell migration via the binding of its GAG motif to CD44. SRGN/CD44 interaction induces Rho-family GTPase-mediated cytoskeleton reorganization and facilitates Src-mediated focal adhesion turnover, leading to increased cell migration. These findings suggest that targeting specific glycans in tumor microenvironment that serve as ligands for oncogenic pathways may be a potential strategy for cancer therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Glicosaminoglicanos/genética , Receptores de Hialuranos/genética , Proteoglicanos/genética , Proteínas de Transporte Vesicular/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Glicosaminoglicanos/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Mutagénesis Sitio-Dirigida , Unión Proteica/genética , Proteoglicanos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rho/genética , Familia-src Quinasas/genética
2.
Mol Cell Proteomics ; 11(10): 901-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22761399

RESUMEN

Mutational activation of KRAS promotes various malignancies, including lung adenocarcinoma. Knowledge of the molecular targets mediating the downstream effects of activated KRAS is limited. Here, we provide the KRAS target proteins and N-glycoproteins using human bronchial epithelial cells with and without the expression of activated KRAS (KRAS(V12)). Using an OFFGEL peptide fractionation and hydrazide method combined with subsequent LTQ-Orbitrap analysis, we identified 5713 proteins and 608 N-glycosites on 317 proteins in human bronchial epithelial cells. Label-free quantitation of 3058 proteins (≥2 peptides; coefficient of variation (CV) ≤ 20%) and 297 N-glycoproteins (CV ≤ 20%) revealed the differential regulation of 23 proteins and 14 N-glycoproteins caused by activated KRAS, including 84% novel ones. An informatics-assisted IPA-Biomarker® filter analysis prioritized some of the differentially regulated proteins (ALDH3A1, CA2, CTSD, DST, EPHA2, and VIM) and N-glycoproteins (ALCAM, ITGA3, and TIMP-1) as cancer biomarkers. Further, integrated in silico analysis of microarray repository data of lung adenocarcinoma clinical samples and cell lines containing KRAS mutations showed positive mRNA fold changes (p < 0.05) for 61% of the KRAS-regulated proteins, including biomarker proteins, CA2 and CTSD. The most significant discovery of the integrated validation is the down-regulation of FABP5 and PDCD4. A few validated proteins, including tumor suppressor PDCD4, were further confirmed as KRAS targets by shRNA-based knockdown experiments. Finally, the studies on KRAS-regulated N-glycoproteins revealed structural alterations in the core N-glycans of SEMA4B in KRAS-activated human bronchial epithelial cells and functional role of N-glycosylation of TIMP-1 in the regulation of lung adenocarcinoma A549 cell invasion. Together, our study represents the largest proteome and N-glycoproteome data sets for HBECs, which we used to identify several novel potential targets of activated KRAS that may provide insights into KRAS-induced adenocarcinoma and have implications for both lung cancer therapy and diagnosis.


Asunto(s)
Adenocarcinoma/genética , Proteínas Reguladoras de la Apoptosis/genética , Bronquios/metabolismo , Células Epiteliales/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Unión al ARN/genética , Proteínas ras/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores de Tumor , Bronquios/patología , Línea Celular Tumoral , Células Epiteliales/patología , Proteínas de Unión a Ácidos Grasos/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilación , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteoma/genética , Proteoma/metabolismo , Proteómica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , ARN Interferente Pequeño , Proteínas de Unión al ARN/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Proteínas ras/metabolismo
3.
Mol Cell Proteomics ; 9(1): 131-44, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19752006

RESUMEN

In this study, we present a fully automated tool, called IDEAL-Q, for label-free quantitation analysis. It accepts raw data in the standard mzXML format as well as search results from major search engines, including Mascot, SEQUEST, and X!Tandem, as input data. To quantify as many identified peptides as possible, IDEAL-Q uses an efficient algorithm to predict the elution time of a peptide unidentified in a specific LC-MS/MS run but identified in other runs. Then, the predicted elution time is used to detect peak clusters of the assigned peptide. Detected peptide peaks are processed by statistical and computational methods and further validated by signal-to-noise ratio, charge state, and isotopic distribution criteria (SCI validation) to filter out noisy data. The performance of IDEAL-Q has been evaluated by several experiments. First, a serially diluted protein mixed with Escherichia coli lysate showed a high correlation with expected ratios and demonstrated good linearity (R(2) = 0.996). Second, in a biological replicate experiment on the THP-1 cell lysate, IDEAL-Q quantified 87% (1,672 peptides) of all identified peptides, surpassing the 45.7% (909 peptides) achieved by the conventional identity-based approach, which only quantifies peptides identified in all LC-MS/MS runs. Manual validation on all 11,940 peptide ions in six replicate LC-MS/MS runs revealed that 97.8% of the peptide ions were correctly aligned, and 93.3% were correctly validated by SCI. Thus, the mean of the protein ratio, 1.00 +/- 0.05, demonstrates the high accuracy of IDEAL-Q without human intervention. Finally, IDEAL-Q was applied again to the biological replicate experiment but with an additional SDS-PAGE step to show its compatibility for label-free experiments with fractionation. For flexible workflow design, IDEAL-Q supports different fractionation strategies and various normalization schemes, including multiple spiked internal standards. User-friendly interfaces are provided to facilitate convenient inspection, validation, and modification of quantitation results. In summary, IDEAL-Q is an efficient, user-friendly, and robust quantitation tool. It is available for download.


Asunto(s)
Algoritmos , Espectrometría de Masas/métodos , Péptidos/análisis , Programas Informáticos , Línea Celular Tumoral , Proteínas de Escherichia coli/análisis , Proteínas de Escherichia coli/clasificación , Humanos , Proteoma/análisis , Proteoma/clasificación , Proteómica/métodos , Reproducibilidad de los Resultados
4.
Front Oncol ; 12: 883437, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35719949

RESUMEN

Background: Addition of oxaliplatin to adjuvant 5-FU has significantly improved the disease-free survival and served as the first line adjuvant chemotherapy in advanced colorectal cancer (CRC) patients. However, a fraction of patients remains refractory to oxaliplatin-based treatment. It is urgent to establish a preclinical platform to predict the responsiveness toward oxaliplatin in CRC patients as well as to improve the efficacy in the resistant patients. Methods: A living biobank of organoid lines were established from advanced CRC patients. Oxaliplatin sensitivity was assessed in patient-derived tumor organoids (PDOs) in vitro and in PDO-xenografted tumors in mice. Based on in vitro oxaliplatin IC50 values, PDOs were classified into either oxaliplatin-resistant (OR) or oxaliplatin-sensitive (OS) PDOs. The outcomes of patients undergone oxaliplatin-based treatment was followed. RNA-sequencing and bioinformatics tools were performed for molecular profiling of OR and OS PDOs. Oxaliplatin response signatures were submitted to Connectivity Map algorithm to identify perturbagens that may antagonize oxaliplatin resistance. Results: Oxaliplatin sensitivity in PDOs was shown to correlate to oxaliplatin-mediated inhibition on PDO xenograft tumors in mice, and parallelled clinical outcomes of CRC patients who received FOLFOX treatment. Molecular profiling of transcriptomes revealed oxaliplatin-resistant and -sensitive PDOs as two separate entities, each being characterized with distinct hallmarks and gene sets. Using Leave-One-Out Cross Validation algorithm and Logistic Regression model, 18 gene signatures were identified as predictive biomarkers for oxaliplatin response. Candidate drugs identified by oxaliplatin response signature-based strategies, including inhibitors targeting c-ABL and Notch pathway, DNA/RNA synthesis inhibitors, and HDAC inhibitors, were demonstrated to potently and effectively increase oxaliplatin sensitivity in the resistant PDOs. Conclusions: PDOs are useful in informing decision-making on oxaliplatin-based chemotherapy and in designing personalized chemotherapy in CRC patients.

5.
J Surg Oncol ; 102(5): 497-502, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20872953

RESUMEN

BACKGROUND AND OBJECTIVES: Small-cell carcinomas (SCC) develop most commonly in the lung (small-cell lung carcinoma, SCLC) and only small percentages are present at extra-pulmonary sites. This study aimed to examine the distribution, treatment, and survival of SCCs. METHODS: The records for 922 SCC cases of various origins between January 1989 and December 2008 were retrieved and analyzed. RESULTS: The lung (89.2%) was the most common location, followed by the esophagus (1.8%), urinary bladder (1.6%), uterine cervix (1.5%), colorectum (1.4%), skin (1.0%), stomach (0.9%), head and neck (0.7%), prostate (0.3%), and small intestine (0.1%). Limited disease (LD) SCLC patients underwent surgery and chemotherapy had significantly higher survival rates than those who received chemotherapy alone, those who underwent combined radiotherapy and chemotherapy, and those who were administered supportive treatment. Actuarial 1-, 2-, and 3-year survival rate was 28.9%, 9.4%, and 4.8% for total SCLC cases, 41.3%, 17.5%, and 9.6% for LD-SCLC patients, and 21.9%, 4.2%, and 1.8% for extensive disease (ED)-SCLC patients (P < 0.001). The survival rates for lung and stomach SCC patients with LD were significantly better than for patients with ED; cervical SCC stages I and IIa patients had better survival rates than patients with stage IIb and above (P = 0.034). CONCLUSION: The lung was the most common location of SCCs, with 9.3% of cases being extra-pulmonary in origin. The need for combined surgery and chemotherapy in LD-SCLC patients deserves further evaluation.


Asunto(s)
Carcinoma de Células Pequeñas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Anciano , Anciano de 80 o más Años , Carcinoma de Células Pequeñas/mortalidad , Carcinoma de Células Pequeñas/patología , Carcinoma de Células Pequeñas/terapia , Femenino , Neoplasias Gastrointestinales/mortalidad , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/terapia , Humanos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Estudios Retrospectivos , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinoma Pulmonar de Células Pequeñas/terapia , Tasa de Supervivencia , Taiwán , Resultado del Tratamiento
6.
Cancer Res ; 67(5): 2089-97, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17332338

RESUMEN

Osteopontin (OPN) and splice variants of CD44 (CD44(V)) have independently been identified as markers for tumor progression. In this study, we show that both OPN and CD44(V) are frequently overexpressed in human gastric cancer and that OPN-engaged CD44(V) ligation confers cells an increased survival mediated through integrin activation. First, we show that OPN treatment confers cells an increased resistance to UV-induced apoptosis. The OPN-mediated antiapoptosis is dependent on the expression of the variant exon 6 (V6)- or V7-containing CD44 as shown by overexpression of individual CD44(V) in gastric AZ521 cells that express no or very low level of endogenous CD44 and by knockdown of the constitutively expressed V6-containing CD44 isoforms in colon HT29 cells. Although OPN also interacts with RGD integrins, OPN-RGD sequence is dispensable for OPN-mediated antiapoptosis. OPN-induced antiapoptosis is mainly attributed to the engagement of CD44(V) isoforms and the relay of an inside-out signaling via Src activity, leading to robust integrin activation. Furthermore, OPN-elicited antiapoptosis was observed when cells were plated on fibronectin but not on poly-D-lysin, and preincubation of cells with anti-integrin beta(1) antibody to block integrin-extracellular matrix (ECM) interaction or ectopic expression of the dominant-negative forms of focal adhesion kinase to block ECM-derived signal abolished OPN-induced survival, suggesting that OPN-elicited antiapoptotic function is propagated from matrix transduced by integrin. Taken together, we showed that OPN-CD44(V) interaction promotes ECM-derived survival signal mediated through integrin activation, which may play an important role in the pathogenic development and progression of gastric cancer.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Gastrointestinales/patología , Receptores de Hialuranos/metabolismo , Integrinas/metabolismo , Osteopontina/fisiología , Adenocarcinoma/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Gastrointestinales/metabolismo , Células HT29 , Humanos , Osteopontina/metabolismo , Osteopontina/farmacología , Isoformas de Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Rayos Ultravioleta/efectos adversos
7.
Int J Cancer ; 122(2): 377-86, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17918157

RESUMEN

We recently reported that low Nm23-H1 expression of primary oral squamous cell carcinoma (OSCC) was correlated with the occurrence of lymphatic metastasis. However, little is known about whether Nm23-H1 level of metastatic tumors in the cervical lymph nodes is reduced in comparison with primary oral cancers and its significance for patients' prognosis. By immunohistochemistry, we analyzed the Nm23-H1 expression in 52 pairs of OSCC specimens from primary oral cancers and their metastatic lymph nodes. Western blot analysis further confirmed the immunohistochemical interpretation. To verify the effects of Nm23-H1 on cell migration and invasion, we established several stable clones derived from a human OSCC cell line (SAS) by knockdown and overexpression. Wound-healing closure, transwell migration and invasion assays were performed to determine cell motility, migratory and invasive activities. Western blot analysis was carried out to evaluate cyclin A expression of OSCC cells with the altered Nm23-H1 levels following knockdown and overexpression. By immunohistochemistry, Nm23-H1 expression of metastatic lymph nodes was significantly lower than that of their primary oral cancers, supporting a role of Nm23-H1 in metastasis suppression. Negative Nm23-H1 interpretation of OSCC specimens, in either primary oral cancers or metastatic lymph nodes, indicated a poor survival outcome of patients. On the basis of in vitro studies of Nm23-H1 knockdown and overexpression, we demonstrated an inverse correlation between Nm23-H1 expression and the invasiveness of OSCC cells. Moreover, we observed the concomitant reduction in Nm23-H1 and cyclin A levels of metastatic tumors in both results of in vitro OSCC cells and ex vivo tumor specimens.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Regulación Neoplásica de la Expresión Génica , Ganglios Linfáticos/metabolismo , Neoplasias de la Boca/metabolismo , Nucleósido Difosfato Quinasas NM23/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/diagnóstico , Línea Celular Tumoral , Ciclina A/metabolismo , Femenino , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/diagnóstico , Metástasis de la Neoplasia
8.
Am J Clin Pathol ; 130(4): 535-42, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18794045

RESUMEN

A series of 76 cases of gastrointestinal tract endocrine tumors, including 21 poorly differentiated endocrine carcinomas (PDECs, small cell carcinomas) and 55 well-differentiated endocrine neoplasms (WDENs, carcinoids), 18 metastatic and 37 nonmetastatic rectal carcinoids, were examined by immunohistochemical analysis for p16INK4a, cyclin D1, and retinoblastoma protein (pRB) expression. Overexpression of p16INK4a was noted in 16 (76%) of the PDECs and none of the WDENs (P < .0001). Loss of pRB expression was demonstrated in 14 (67%) of the PDECs and 17 (31%) of the WDENs (P = .004). Overexpression of cyclin D1 was noted in 49 (89%) of the WDENs and 3 (14%) of the PDECs (P < .0001). Loss of pRB expression was noted in 11 (61%) of 18 metastatic WDENs and only 6 (16%) of 37 nonmetastatic WDENs (P = .001). The p16INK4a/cyclin D1/pRB pathway was altered in gastrointestinal tract endocrine tumors, and the loss of expression of pRB may be helpful in identifying patients at high risk of metastasis in rectal WDENs.


Asunto(s)
Ciclina D1/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Neoplasias Gastrointestinales/metabolismo , Tumores Neuroendocrinos/metabolismo , Proteína de Retinoblastoma/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Neoplasias Gastrointestinales/patología , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/patología
9.
Am J Clin Pathol ; 126(6): 856-65, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17074692

RESUMEN

This study was designed to determine the expression of p16, p53, and CD117 in gastrointestinal tract endocrine tumors. Immunohistochemical studies of p16, p53, and CD117 were performed in 57 gastrointestinal tract endocrine tumors, including 22 poorly differentiated endocrine carcinomas (PDECs) and 35 well-differentiated endocrine tumors (WDETs). Overexpression of p16 and p53 was observed in 16 (73%) and 10 (45%) of the PDECs, respectively, whereas only 1 WDET showed overexpression of p53 and none showed overexpression of p16. A total of 18 (82%) of the PDECs showed overexpression of p16 or p53 proteins. This is closely associated with PDEC (P < .0001). By using overexpression of p16 or p53 as the criteria for PDEC, the sensitivity and specificity are 81.8% and 97.1%, respectively, with positive and negative predictive values of 94.7% and 89.5%, respectively. CD117 was not detected in any of the 57 gastrointestinal endocrine tumors by immunohistochemical analysis.


Asunto(s)
Tumor Carcinoide/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Neoplasias Gastrointestinales/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Tumor Carcinoide/secundario , Femenino , Neoplasias Gastrointestinales/patología , Humanos , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas
10.
Sci Rep ; 6: 26050, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27198662

RESUMEN

Gliomas are aggressive brain tumors with poor prognosis. In this study, we report a novel approach combining both in vivo multi-parametric MRI and in vitro cell culture assessments to evaluate the pathogenic development of gliomas. Osteopontin (OPN), a pleiotropic factor, has been implicated in the formation and progression of various human cancers, including gliomas, through its functions in regulating cell proliferation, survival, angiogenesis, and migration. Using rat C6 glioma model, the combined approach successfully monitors the acquisition and decrease of cancer hallmarks. We show that knockdown of the expression of OPN reduces C6 cell proliferation, survival, viability and clonogenicity in vitro, and reduces tumor burden and prolongs animal survival in syngeneic rats. OPN depletion is associated with reduced tumor growth, decreased angiogenesis, and an increase of tumor-associated metabolites, as revealed by T2-weighted images, diffusion-weighted images, K(trans) maps, and 1H-MRS, respectively. These strategies allow us to define an important role of OPN in conferring cancer hallmarks, which can be further applied to assess the functional roles of other candidate genes in glioma. In particular, the non-invasive multi-parametric MRI measurement of cancer hallmarks related to proliferation, angiogenesis and altered metabolism may serve as a useful tool for diagnosis and for patient management.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Glioma/diagnóstico , Osteopontina/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular , Autorrenovación de las Células , Supervivencia Celular , Modelos Animales de Enfermedad , Humanos , Imagen por Resonancia Magnética , Trasplante de Neoplasias , Neovascularización Patológica , Osteopontina/genética , Ratas , Carga Tumoral
11.
J Thorac Oncol ; 11(9): 1469-81, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27234602

RESUMEN

INTRODUCTION: Despite recent advances in cancer therapy, the overall 5-year survival rate of patients with lung cancer remains low. The aim of our study was to search for novel markers for early diagnosis in patients with lung cancer. METHODS: Complementary DNA microarray analysis was performed in primary lung adenocarcinomas and cell lines to search for differentially expressed genes, followed by in vivo and in vitro tumorigenic assays to characterize the oncogenic potential of the candidate genes. Gene body methylation was analyzed by 450K methylation array, bisulfite sequencing, and quantitative methylation-specific polymerase chain reaction assays. In silico analysis of The Cancer Genome Atlas data set was also performed. RESULTS: Inositol-trisphosphate 3-kinase A gene (ITPKA), a kinase with limited tissue distribution, was identified as a potential oncogene. We showed that ITPKA expression is up-regulated in many forms of cancers, including lung and breast cancers, and that overexpressed ITPKA contributes to tumorigenesis. We also demonstrated that ITPKA expression is regulated by epigenetic DNA methylation of ITPKA gene body through modulation of the binding of SP1 transcription factor to the ITPKA promoter. ITPKA gene body displayed low or absent levels of methylation in most normal tissue but was significantly methylated in malignant tumors. In lung cancer, ITPKA gene body methylation first appeared at the in situ carcinoma stage and progressively increased after invasion. CONCLUSIONS: ITPKA is a potential oncogene that it is overexpressed in most tumors, and its overexpression promotes tumorigenesis. ITPKA gene body methylation regulates its expression and thus serves as a novel and potential biomarker for early cancer detection.


Asunto(s)
Metilación de ADN , Detección Precoz del Cáncer , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Biomarcadores de Tumor , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/diagnóstico
12.
Oncogene ; 23(14): 2472-83, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-14968111

RESUMEN

Oncogenic virus proteins often target to tumor suppressor p53 during virus life cycle. In the case of hepatitis C virus (HCV) core protein, it has been shown to affect p53-dependent transcription. Here, we further characterized the in vitro and in vivo interactions between HCV core protein and p53 and showed that these two proteins colocalized in subnuclear granular structures and the perinuclear area. By use of a reporter assay, we observed that while low level of HCV core protein enhanced the transactivational activity of p53, high level of HCV core protein inhibited this activity. In both cases, however, HCV core protein increased the p53 DNA-binding affinity in gel retardation analyses, likely due to the hyperacetylation of p53 Lys(373) and Lys(382) residues. Additionally, HCV core protein, depending on its expression level, had differential effects on the Ser(15) phosphorylation of p53. Moreover, HCV core protein could rescue p53-mediated suppressive effects on both RNA polymerase I and III transcriptions. Collectively, our results indicate that HCV core protein targets to p53 pathway via at least three means: physical interaction, modulation of p53 gene regulatory activity and post-translational modification. This feature of HCV core protein, may potentially contribute to the HCV-associated pathogenesis.


Asunto(s)
Regulación Viral de la Expresión Génica , Procesamiento Proteico-Postraduccional , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteínas del Núcleo Viral/metabolismo , Acetilación , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/virología , Genes Supresores de Tumor , Glutatión Transferasa/metabolismo , Células HeLa , Hepacivirus/genética , Humanos , Lisina/metabolismo , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Serina/metabolismo , Proteína p53 Supresora de Tumor/química , Proteínas del Núcleo Viral/genética
13.
Oncogene ; 23(54): 8731-42, 2004 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-15480428

RESUMEN

Rhotekin (RTKN), the gene coding for the Rho effector, RTKN, was shown to be overexpressed in human gastric cancer (GC). In this study, we further showed that RTKN is expressed at a low level in normal cells and is overexpressed in many cancer-derived cell lines. The function of RTKN as an effector protein in Rho GTPase-mediated pathways regulating apoptosis was investigated. By transfection and expression of RTKN in cells that expressed endogenous RTKN at a low basal level, we showed that RTKN overexpression conferred cell resistance to apoptosis induced by serum deprivation or treatment with sodium butyrate, and the increased resistance correlated to the level of RTKN. Conversely, reducing RTKN expression by small interfering RNAs greatly sensitized cells to apoptosis. The RTKN-mediated antiapoptotic effect was blocked by the nuclear factor-kappaB (NF-kappaB) inhibitors, curcumin or parthenolide, but not by the phosphatidylinositol 3'-OH-kinase inhibitor, LY294002, or the MAP kinase inhibitor, PD98059. Reporter gene assays and electrophoretic mobility shift assay confirmed that RTKN overexpression led to constitutive activation of NF-kappaB through the phosphorylation of IkappaB by IKKbeta. By using the RTKN truncation mutants, we showed that RTKN mediated Rho activity eliciting signaling pathway to activate NF-kappaB, with a concomitant induction of expression of the NF-kappaB antiapoptotic genes, cIAP-2, BCl-xL, A1, and A20. Consistent with these data, RTKN-expressing cells showed increased chemoresistance to 5-fluorouracil and paclitaxol, and the resistance was greatly attenuated by NF-kappaB inhibitor. In conclusion, overactivated Rho/RTKN/NF-kappaB signaling pathway through overexpression of RTKN may play a key role in gastric tumorigenesis by conferring cells resistance to apoptosis, and this signaling pathway may serve as an important target for novel therapeutic approaches to the treatment of human GC.


Asunto(s)
Apoptosis/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , FN-kappa B/metabolismo , Proteínas de Unión al GTP rho/fisiología , Secuencia de Aminoácidos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Proteínas de Unión al GTP , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Mol Biol ; 343(5): 1365-77, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15491618

RESUMEN

Hepatoma-derived growth factor (HDGF)-related proteins (HRPs) comprise a new protein family that has been implicated in nephrogenesis, tumorigenesis, vascular development, cell proliferation, and transcriptional activation. All HRPs share a conserved N-terminal homologous to the amino terminus of HDGF (HATH) domain, but vary significantly in the C-terminal region. Here, we show that in solution the N and C termini of human HDGF form two structurally independent domains. The 100 amino acid residue N-terminal HATH domain is well-structured while the 140 amino acid residue C-terminal domain is disordered. We determined the solution structure of the HATH domain by NMR. The core structure of the HATH domain is a five-stranded beta-barrel followed by two alpha-helices, similar to those of PWWP domains of known structures. Surface plasmon resonance results showed that the HATH domain is primarily responsible for heparin binding. On the basis of the chemical shift perturbation induced by binding of heparin-derived hexasaccharide, we identified a prominent, highly positively charged region as the putative heparin-binding site. Sequence comparison and structure prediction suggest that all HRPs are likely to adapt a similar modular structure.


Asunto(s)
Heparina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Espectroscopía de Resonancia Magnética , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/química , Estructura Terciaria de Proteína , Factores de Tiempo
15.
Brain Struct Funct ; 220(4): 2073-85, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24771246

RESUMEN

White matter tracts are important for the trafficking of neural progenitor cells (NPCs) in both normal and pathological conditions, but the underlying mechanism is not clear. The directionality of white matter is advantageous for molecules or cells to distribute over a long distance, but this feature is unlikely solely responsible for efficient migration. The present study hypothesizes that the efficient migration of NPCs into white matter is under the influences of neurochemical attraction­CXCL12/CXCR4 signaling, a major mechanism underlying the targeted migration of NPCs. To test this view, the present study investigated the effects of CXCL12 administration into the corpus callosum (CC) on the migratory behavior of transplanted NPCs. A living animal tracking platform based on MRI and a magnetic cell labeling technique was employed. The NPCs were magnetically labeled and then transplanted at the right end of the CC. CXCL12 was infused continuously at the left end. Migration of NPCs was monitored repeatedly over a 7-day course using 3D gradient echo T2*-weighted imaging. It was found that, CXCL12 induced NPCs to migrate up to 1,881 µm from the graft whereas the spontaneous migration was mere 200 µm. CXCL12 induced migration that was nine times as efficient in the speed. The results indicate that the CXCL12/CXCR4 signaling may be a mechanism via which NPCs efficiently migrate along the white matter tracts. The study also presents a potential strategy for facilitating the targeted migration in NPC therapy for brain disorders.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CXCL12/metabolismo , Células-Madre Neurales/fisiología , Receptores CXCR4/metabolismo , Transducción de Señal/fisiología , Sustancia Blanca/fisiología , Análisis de Varianza , Animales , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/farmacología , Embrión de Mamíferos , Femenino , Citometría de Flujo , Factor C1 de la Célula Huésped/efectos de los fármacos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Sustancia Blanca/citología
16.
Sci Rep ; 5: 9633, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25910225

RESUMEN

Identification and functional analysis of genes from genetically altered chromosomal regions would suggest new molecular targets for cancer diagnosis and treatment. Here we performed a genome-wide analysis of chromosomal copy number alterations (CNAs) in matching sets of colon mucosa-adenoma-carcinoma samples using high-throughput oligonucleotide microarray analysis. In silico analysis of NCBI GEO and TCGA datasets allowed us to uncover the significantly altered genes (p ≤ 0.001) associated with the identified CNAs. We performed quantitative PCR analysis of the genomic and complementary DNA derived from primary mucosa, adenoma, and carcinoma samples, and confirmed the recurrent loss and down-regulation of PTPRM in colon adenomas and carcinomas. Functional characterization demonstrated that PTPRM negatively regulates cell growth and colony formation, whereas loss of PTPRM promotes oncogenic cell growth. We further showed that, in accordance to Knudson's two-hit hypothesis, inactivation of PTPRM in colon cancer was mainly attributed to loss of heterozygosity and promoter hypermethylation. Taken together, this study demonstrates a putative tumor suppressive role for PTPRM and that genetic and epigenetic alterations of PTPRM may contribute to early step of colorectal tumorigenesis.


Asunto(s)
Adenoma/genética , Carcinoma/genética , Neoplasias Colorrectales/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Adenoma/patología , Carcinoma/patología , Proliferación Celular , Neoplasias Colorrectales/patología , Variaciones en el Número de Copia de ADN , Metilación de ADN , Bases de Datos Genéticas , Regulación hacia Abajo , Genotipo , Humanos , Mucosa Intestinal/metabolismo , Pérdida de Heterocigocidad , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo
17.
Methods Mol Biol ; 1120: 157-66, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24470024

RESUMEN

Identification of oncogene-mediated phosphorylation events is essential to understanding the molecular determinants responsible for cancer development and progression. Here, we identify KRAS-regulated phosphorylation events using label-free quantitation-based comparative phosphoproteomics analyses of immortalized human bronchial epithelial cells that express oncogenic KRAS as well as cells that do not. Further, we demonstrate integration of the identified phosphorylation events with the Pathway Interaction Database to infer KRAS-regulated pathways, which may have implications in KRAS-associated lung adenocarcinoma development. Taken together, our study provides an overview of the functional phosphoproteomics approach involving cell culture, preparation of whole cell lysates, trypsin digestion, phosphopeptide enrichment, mass spectrometry analyses, label-free quantitative analyses, and signaling pathway analyses to study KRAS-targeted events.


Asunto(s)
Fosfoproteínas/metabolismo , Proteómica/métodos , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Proteínas ras/metabolismo , Bronquios/citología , Transformación Celular Neoplásica , Células Cultivadas , Cromatografía Liquida , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Fosfopéptidos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas p21(ras) , Espectrometría de Masas en Tándem
18.
Neoplasia ; 16(5): 441-50, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24947187

RESUMEN

Two genes are called synthetic lethal (SL) if their simultaneous mutations lead to cell death, but each individual mutation does not. Targeting SL partners of mutated cancer genes can kill cancer cells specifically, but leave normal cells intact. We present an integrated approach to uncovering SL pairs in colorectal cancer (CRC). Screening verified SL pairs using microarray gene expression data of cancerous and normal tissues, we first identified potential functionally relevant (simultaneously differentially expressed) gene pairs. From the top-ranked pairs, ~20 genes were chosen for immunohistochemistry (IHC) staining in 171 CRC patients. To find novel SL pairs, all 169 combined pairs from the individual IHC were synergistically correlated to five clinicopathological features, e.g. overall survival. Of the 11 predicted SL pairs, MSH2-POLB and CSNK1E-MYC were consistent with literature, and we validated the top two pairs, CSNK1E-TP53 and CTNNB1-TP53 using RNAi knockdown and small molecule inhibitors of CSNK1E in isogenic HCT-116 and RKO cells. Furthermore, synthetic lethality of CSNK1E and TP53 was verified in mouse model. Importantly, multivariate analysis revealed that CSNK1E-P53, CTNNB1-P53, MSH2-RB1, and BRCA1-WNT5A were independent prognosis markers from stage, with CSNK1E-P53 applicable to early-stage and the remaining three throughout all stages. Our findings suggest that CSNK1E is a promising target for TP53-mutant CRC patients which constitute ~40% to 50% of patients, while to date safety regarding inhibition of TP53 is controversial. Thus the integrated approach is useful in finding novel SL pairs for cancer therapeutics, and it is readily accessible and applicable to other cancers.


Asunto(s)
Biomarcadores de Tumor/genética , Caseína Cinasa 1 épsilon/genética , Neoplasias Colorrectales/genética , Proteína p53 Supresora de Tumor/genética , beta Catenina/genética , Animales , Neoplasias Colorrectales/mortalidad , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Modelos de Riesgos Proporcionales , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares
19.
Magn Reson Imaging ; 30(2): 247-53, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22133285

RESUMEN

The successful translation of stem-cell therapies requires a detailed understanding of the fate of transplanted cells. Magnetic resonance imaging (MRI) has provided a noninvasive means of imaging cell dynamics in vivo by prelabeling cell with T(2) shortening iron oxide particles. However, this approach suffers from a gradual loss of sensitivity since active cell mitosis could decrease the cellular contrast agent (CA) concentration below detection level. In addition, the interpretation of images may be confounded by hypointensities induced by factors other than this CA susceptibility effect (CASE). We therefore examined the feasibility of exploiting the phase information in MRI to increase the sensitivity of cellular imaging and to differentiate the CASE from endogenous image hypointensity. Phase aliasing and the B(0) field inhomogeneity effect were removed by applying a reliable unwrapping algorithm and a high-pass filter, respectively, thus delineating phase variations originating from high spatial frequencies due to the CASE. We found that the filtered phase map detects labeled cells with high sensitivity and can readily differentiate the cell migration track from the white matter, both of which are hypointense in T(2)-weighted magnitude images. Furthermore, an approximate fivefold contrast-to-noise ratio enhancement can be achieved with an MRI phase map over conventional T(2)-weighted magnitude images.


Asunto(s)
Encéfalo/citología , Encéfalo/cirugía , Rastreo Celular/métodos , Dextranos , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita , Trasplante de Células Madre , Células Madre/citología , Animales , Células Cultivadas , Medios de Contraste , Ratas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
20.
PLoS One ; 6(5): e20199, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21637843

RESUMEN

BACKGROUND: Ras is frequently mutated in a variety of human cancers, including lung cancer, leading to constitutive activation of MAPK signaling. Despite decades of research focused on the Ras oncogene, Ras-targeted phosphorylation events and signaling pathways have not been described on a proteome-wide scale. METHODOLOGY/PRINCIPAL FINDINGS: By functional phosphoproteomics, we studied the molecular mechanics of oncogenic Ras signaling using a pathway-based approach. We identified Ras-regulated phosphorylation events (n = 77) using label-free comparative proteomics analysis of immortalized human bronchial epithelial cells with and without the expression of oncogenic Ras. Many were newly identified as potential targets of the Ras signaling pathway. A majority (∼60%) of the Ras-targeted events consisted of a [pSer/Thr]-Pro motif, indicating the involvement of proline-directed kinases. By integrating the phosphorylated signatures into the Pathway Interaction Database, we further inferred Ras-regulated pathways, including MAPK signaling and other novel cascades, in governing diverse functions such as gene expression, apoptosis, cell growth, and RNA processing. Comparisons of Ras-regulated phosphorylation events, pathways, and related kinases in lung cancer-derived cells supported a role of oncogenic Ras signaling in lung adenocarcinoma A549 and H322 cells, but not in large cell carcinoma H1299 cells. CONCLUSIONS/SIGNIFICANCE: This study reveals phosphorylation events, signaling networks, and molecular functions that are regulated by oncogenic Ras. The results observed in this study may aid to extend our knowledge on Ras signaling in lung cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Genes ras , Neoplasias Pulmonares/metabolismo , Fosfoproteínas/metabolismo , Proteómica/métodos , Transducción de Señal , Adenocarcinoma del Pulmón , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Bronquios/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Células Epiteliales/metabolismo , Humanos , Datos de Secuencia Molecular , Fosfopéptidos/metabolismo , Fosfoproteínas/química , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA