Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Genes Chromosomes Cancer ; 51(4): 313-27, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22170698

RESUMEN

Well-differentiated liposarcoma (WDLS) and dedifferentiated liposarcoma (DDLS) represent the most common biological group of liposarcoma, and there is a pressing need to develop targeted therapies for patients with advanced disease. To identify potential therapeutic targets, we sought to identify differences in the adipogenic pathways between DDLS, WDLS, and normal adipose tissue. In a microarray analysis of DDLS (n = 84), WDLS (n = 79), and normal fat (n = 23), C/EBPα, a transcription factor involved in cell cycle regulation and differentiation, was underexpressed in DDLS when compared to both WDLS and normal fat (15.2- and 27.8-fold, respectively). In normal adipose-derived stem cells, C/EBPα expression was strongly induced when cells were cultured in differentiation media, but in three DDLS cell lines, this induction was nearly absent. We restored C/EBPα expression in one of the cell lines (DDLS8817) by transfection of an inducible C/EBPα expression vector. Inducing C/EBPα expression reduced proliferation and caused cells to accumulate in G2/M. Under differentiation conditions, the cell proliferation was reduced further, and 66% of the DDLS cells containing the inducible C/EBPα expression vector underwent apoptosis as demonstrated by annexin V staining. These cells in differentiation conditions expressed early adipocyte-specific mRNAs such as LPL and FABP4, but they failed to accumulate intracellular lipid droplets, a characteristic of mature adipocytes. These results demonstrate that loss of C/EBPα is an important factor in suppressing apoptosis and maintaining the dedifferentiated state in DDLS. Restoring C/EBPα may be a useful therapeutic approach for DDLS.


Asunto(s)
Apoptosis , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Liposarcoma/metabolismo , Liposarcoma/patología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Ciclo Celular , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Lípidos/biosíntesis , Neoplasias de Tejido Adiposo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , ARN Mensajero/biosíntesis , Transducción de Señal , Células Madre/metabolismo
2.
Clin Cancer Res ; 28(1): 175-186, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34667024

RESUMEN

PURPOSE: Dedifferentiated liposarcoma (DDLS), one of the most common and aggressive sarcomas, infrequently responds to chemotherapy. DDLS survival and growth depend on underexpression of C/EBPα, a tumor suppressor and transcriptional regulator controlling adipogenesis. We sought to screen and prioritize candidate drugs that increase C/EBPα expression and may therefore serve as differentiation-based therapies for DDLS. EXPERIMENTAL DESIGN: We screened known bioactive compounds for the ability to restore C/EBPα expression and inhibit proliferation selectively in two DDLS cell lines but not in normal adipose-derived stem cells (ASC). Selected hits' activity was validated, and the mechanism of the most potent, SN-38, was investigated. The in vivo efficacy of irinotecan, the prodrug of SN-38, was evaluated in DDLS xenograft models. RESULTS: Of 3,119 compounds, screen criteria were met by 19. Validation experiments confirmed the DDLS selectivity of deguelin, emetine, and SN-38 and showed that they induce apoptosis in DDLS cells. SN-38 had the lowest IC50 (approximately 10 nmol/L), and its pro-apoptotic effects were countered by knockdown of CEBPA but not of TP53. Irinotecan significantly inhibited tumor growth at well-tolerated doses, induced nuclear expression of C/EBPα, and inhibited HIF1α expression in DDLS patient-derived and cancer cell line xenograft models. In contrast, doxorubicin, the most common treatment for nonresectable DDLS, reduced tumor growth by 30% to 50% at a dose that caused weight loss. CONCLUSIONS: This high-content screen revealed potential treatments for DDLS. These include irinotecan, which induces apoptosis of DDLS cells in a C/EBPα-dependent, p53-independent manner, and should be clinically evaluated in patients with advanced DDLS.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT , Proteínas Potenciadoras de Unión a CCAAT , Liposarcoma , Adipocitos/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/análisis , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/análisis , Genes Supresores de Tumor , Humanos , Liposarcoma/tratamiento farmacológico , Liposarcoma/genética , Liposarcoma/patología , Células Madre/metabolismo
3.
J Cell Biol ; 171(2): 361-71, 2005 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-16247032

RESUMEN

Introduction of activated p21-activated kinase (PAK) is sufficient to release primary endothelial cells from contact inhibition of growth. Confluent cells display deficient activation of PAK and translocation of Rac to the plasma membrane at matrix adhesions. Targeting Rac to the plasma membrane rescues these cells from contact inhibition. PAK's ability to release human umbilical vein endothelial cells from contact inhibition is blocked by an unphosphorylatable form of its target Merlin, suggesting that PAK promotes mitogenesis by phosphorylating, and thus inactivating, Merlin. Merlin mutants, which are presumed to exert a dominant-negative effect, enable recruitment of Rac to matrix adhesions and promote mitogenesis in confluent cells. Small interference RNA-mediated knockdown of Merlin exerts the same effects. Dominant-negative Rac blocks PAK-mediated release from contact inhibition, implying that PAK functions upstream of Rac in this signaling pathway. These results provide a framework for understanding the tumor suppressor function of Merlin and indicate that Merlin mediates contact inhibition of growth by suppressing recruitment of Rac to matrix adhesions.


Asunto(s)
Membrana Celular/metabolismo , Inhibición de Contacto/fisiología , Neurofibromina 2/fisiología , Proteínas de Unión al GTP rac/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Humanos , Neurofibromina 2/antagonistas & inhibidores , Fosforilación , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Quinasas p21 Activadas , Proteínas de Unión al GTP rac/efectos de los fármacos
4.
Cancer Res ; 80(12): 2461-2471, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32161142

RESUMEN

Myxofibrosarcoma (MFS) and undifferentiated pleomorphic sarcoma (UPS) are highly genetically complex soft tissue sarcomas. Up to 50% of patients develop distant metastases, but current systemic therapies have limited efficacy. MFS and UPS have recently been shown to commonly harbor copy number alterations or mutations in the tumor suppressor genes RB1 and TP53. As these alterations have been shown to engender dependence on the oncogenic protein Skp2 for survival of transformed cells in mouse models, we sought to examine its function and potential as a therapeutic target in MFS/UPS. Comparative genomic hybridization and next-generation sequencing confirmed that a significant fraction of MFS and UPS patient samples (n = 94) harbor chromosomal deletions and/or loss-of-function mutations in RB1 and TP53 (88% carry alterations in at least one gene; 60% carry alterations in both). Tissue microarray analysis identified a correlation between absent Rb and p53 expression and positive expression of Skp2. Downregulation of Skp2 or treatment with the Skp2-specific inhibitor C1 revealed that Skp2 drives proliferation of patient-derived MFS/UPS cell lines deficient in both Rb and p53 by degrading p21 and p27. Inhibition of Skp2 using the neddylation-activating enzyme inhibitor pevonedistat decreased growth of Rb/p53-negative patient-derived cell lines and mouse xenografts. These results demonstrate that loss of both Rb and p53 renders MFS and UPS dependent on Skp2, which can be therapeutically exploited and could provide the basis for promising novel systemic therapies for MFS and UPS. SIGNIFICANCE: Loss of both Rb and p53 renders myxofibrosarcoma and undifferentiated pleomorphic sarcoma dependent on Skp2, which could provide the basis for promising novel systemic therapies.See related commentary by Lambert and Jones, p. 2437.


Asunto(s)
Fibrosarcoma , Sarcoma , Neoplasias de los Tejidos Blandos , Adulto , Animales , Hibridación Genómica Comparativa , Fibrosarcoma/genética , Humanos , Ratones , Sarcoma/genética , Proteína p53 Supresora de Tumor/genética
5.
Mol Cancer Res ; 18(10): 1534-1544, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32561656

RESUMEN

Soft-tissue sarcomas (STS) are rare malignancies showing lineage differentiation toward diverse mesenchymal tissues. Half of all high-grade STSs develop lung metastasis with a median survival of 15 months. Here, we used a genetically engineered mouse model that mimics undifferentiated pleomorphic sarcoma (UPS) to study the molecular mechanisms driving metastasis. High-grade sarcomas were generated with Cre recombinase technology using mice with conditional mutations in Kras and Trp53 (KP) genes. After amputation of the limb bearing the primary tumor, mice were followed for the development of lung metastasis. Using RNA-sequencing of matched primary KP tumors and lung metastases, we found that the long noncoding RNA (lncRNA) Nuclear Enriched Abundant Transcript 1 (Neat1) is significantly upregulated in lung metastases. Furthermore, NEAT1 RNA ISH of human UPS showed that NEAT1 is upregulated within a subset of lung metastases compared with paired primary UPS. Remarkably, CRISPR/Cas9-mediated knockout of Neat1 suppressed the ability of KP tumor cells to colonize the lungs. To gain insight into the underlying mechanisms by which the lncRNA Neat1 promotes sarcoma metastasis, we pulled down Neat1 RNA and used mass spectrometry to identify interacting proteins. Interestingly, most Neat1 interacting proteins are involved in RNA splicing regulation. In particular, KH-Type Splicing Regulatory Protein (KHSRP) interacts with Neat1 and is associated with poor prognosis of human STS. Moreover, depletion of KHSRP suppressed the ability of KP tumor cells to colonize the lungs. Collectively, these results suggest that Neat1 and its interacting proteins, which regulate RNA splicing, are involved in mediating sarcoma metastasis. IMPLICATIONS: Understanding that lncRNA NEAT1 promotes sarcoma metastasis, at least in part, through interacting with the RNA splicing regulator KHSRP may translate into new therapeutic approaches for sarcoma.


Asunto(s)
Empalme del ARN/genética , ARN Largo no Codificante/genética , Sarcoma/genética , Humanos , Metástasis de la Neoplasia , Células PC-3 , Transfección
6.
Nat Cell Biol ; 21(4): 534, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30842593

RESUMEN

In the version of this Article originally published the same blot was inadvertently presented as both p-Rb and Cyclin A in Fig. 2a. This blot corresponds to the p-Rb panel, as can be seen in the unprocessed version of these blots in Supplementary Fig. 9. The corrected version of the panel is shown below, together with a completely uncropped image of both blots. In addition, in the 'Viral transduction' section of the Methods, the pLKO.1 plasmids encoding short hairpin RNAs targeting human Rnd1 were incorrectly listed as clones TRCN0000018338 and TRCN0000039977. The correct clone numbers are TRCN0000047434 and TRCN0000047435.

7.
Cancer Lett ; 234(2): 239-48, 2006 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-15882926

RESUMEN

Reduced expression of connexins (Cxs), gap junction proteins, is frequently reported in malignant cell lines and tumors, whereas recent studies suggested that C x 26, a subtype of Cxs, might help tumor cells acquire malignant phenotypes. To examine this suggestion in the clinical setting, 50 lung squamous cell carcinomas (SCCs) were stained with the anti-C x 26 antibody. No C x 26-specific signals were detectable in 34 tumors (group I; 68%), whereas the remaining 16 were judged positive for C x 26 (group II; 32%). In 14 tumors of group II, C x 26-specific signals were detected not in all SCC cells but in SCC cells facing the tumor stroma or capsule, in which the signals were localized on the plasma membrane. Involved lymph nodes of group-II patients often contained metastatic foci consisting of all C x 26-positive cells. The proportion of C x 26-positive to C x 26-negative SCC cells in the metastatic nodes was larger than that in the corresponding primary tumors. C x 26-positive SCC cells seemed to be more invasive and metastatic than negative ones. Consistently, the 5-year cancer-specific survival rate of group-II patients was significantly lower than that of group-I patients (12.5 vs 38.9%; P=0.0391). Multivariate analysis demonstrated that C x 26 expression (P=0.0448) as well as pathological stage (P=0.0338) and vascular invasion (P=0.0191) were independent, significant prognostic predictors. These results suggest that C x 26 may represent an essential effector for controlling the biological aggressiveness of lung SCC tumor.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Conexinas/biosíntesis , Neoplasias Pulmonares/metabolismo , Metástasis Linfática/patología , Biomarcadores de Tumor/análisis , Northern Blotting , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Conexina 26 , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Pronóstico , Análisis de Supervivencia
8.
Cancer Discov ; 6(10): 1148-1165, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27577794

RESUMEN

Myxofibrosarcoma is a common mesenchymal malignancy with complex genomics and heterogeneous clinical outcomes. Through gene-expression profiling of 64 primary high-grade myxofibrosarcomas, we defined an expression signature associated with clinical outcome. The gene most significantly associated with disease-specific death and distant metastasis was ITGA10 (integrin-α10). Functional studies revealed that myxofibrosarcoma cells strongly depended on integrin-α10, whereas normal mesenchymal cells did not. Integrin-α10 transmitted its tumor-specific signal via TRIO and RICTOR, two oncoproteins that are frequently co-overexpressed through gene amplification on chromosome 5p. TRIO and RICTOR activated RAC/PAK and AKT/mTOR to promote sarcoma cell survival. Inhibition of these proteins with EHop-016 (RAC inhibitor) and INK128 (mTOR inhibitor) had antitumor effects in tumor-derived cell lines and mouse xenografts, and combining the drugs enhanced the effects. Our results demonstrate the importance of integrin-α10/TRIO/RICTOR signaling for driving myxofibrosarcoma progression and provide the basis for promising targeted treatment strategies for patients with high-risk disease. SIGNIFICANCE: Identifying the molecular pathogenesis for myxofibrosarcoma progression has proven challenging given the highly complex genomic alterations in this tumor type. We found that integrin-α10 promotes tumor cell survival through activation of TRIO-RAC-RICTOR-mTOR signaling, and that inhibitors of RAC and mTOR have antitumor effects in vivo, thus identifying a potential treatment strategy for patients with high-risk myxofibrosarcoma. Cancer Discov; 6(10); 1148-65. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1069.


Asunto(s)
Proteínas Portadoras/genética , Fibrosarcoma/genética , Cadenas alfa de Integrinas/genética , Proteínas de Unión al GTP rac/genética , Animales , Línea Celular Tumoral , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Cadenas alfa de Integrinas/metabolismo , Ratones , Terapia Molecular Dirigida , Clasificación del Tumor , Trasplante de Neoplasias , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Regulación hacia Arriba
9.
Nat Cell Biol ; 17(1): 81-94, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25531777

RESUMEN

We identified the Rho GTPase Rnd1 as a candidate metastasis suppressor in basal-like and triple-negative breast cancer through bioinformatics analysis. Depletion of Rnd1 disrupted epithelial adhesion and polarity, induced epithelial-to-mesenchymal transition, and cooperated with deregulated expression of c-Myc or loss of p53 to cause neoplastic conversion. Mechanistic studies revealed that Rnd1 suppresses Ras signalling by activating the GAP domain of Plexin B1, which inhibits Rap1. Rap1 inhibition in turn led to derepression of p120 Ras-GAP, which was able to inhibit Ras. Inactivation of Rnd1 in mammary epithelial cells induced highly undifferentiated and invasive tumours in mice. Conversely, Rnd1 expression inhibited spontaneous and experimental lung colonization in mouse models of metastasis. Genomic studies indicated that gene deletion in combination with epigenetic silencing or, more rarely, point mutation inactivates RND1 in human breast cancer. These results reveal a previously unappreciated mechanism through which Rnd1 restrains activation of Ras-MAPK signalling and breast tumour initiation and progression.


Asunto(s)
Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/genética , Neoplasias Pulmonares/genética , Neoplasias de la Mama Triple Negativas/patología , Proteínas de Unión al GTP rho/fisiología , Animales , Adhesión Celular/genética , Línea Celular Tumoral , Polaridad Celular/genética , Proliferación Celular/genética , Senescencia Celular/genética , Femenino , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Receptores de Superficie Celular/metabolismo , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/genética , Proteína p53 Supresora de Tumor/genética , Proteína Activadora de GTPasa p120/biosíntesis , Proteínas de Unión al GTP rap1/antagonistas & inhibidores , Proteínas Activadoras de ras GTPasa/antagonistas & inhibidores , Proteínas de Unión al GTP rho/genética
10.
Oncoscience ; 4(3-4): 31-32, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28540334
11.
Mol Cell Biol ; 29(15): 4235-49, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19451229

RESUMEN

Integrin signaling promotes, through p21-activated kinase, phosphorylation and inactivation of the tumor suppressor merlin, thus removing a block to mitogenesis in normal cells. However, the biochemical function of merlin and the effector pathways critical for the pathogenesis of malignant mesothelioma and other NF2-related malignancies are not known. We report that integrin-specific signaling promotes activation of mTORC1 and cap-dependent mRNA translation. Depletion of merlin rescues mTORC1 signaling in cells deprived of anchorage to a permissive extracellular matrix, suggesting that integrin signaling controls mTORC1 through inactivation of merlin. This signaling pathway controls translation of the cyclin D1 mRNA and, thereby, cell cycle progression. In addition, it promotes cell survival. Analysis of a panel of malignant mesothelioma cell lines reveals a strong correlation between loss of merlin and activation of mTORC1. Merlin-negative lines are sensitive to the growth-inhibitory effect of rapamycin, and the expression of recombinant merlin renders them partially resistant to rapamycin. Conversely, depletion of merlin restores rapamycin sensitivity in merlin-positive lines. These results indicate that integrin-mediated adhesion promotes mTORC1 signaling through the inactivation of merlin. Furthermore, they reveal that merlin-negative mesotheliomas display unregulated mTORC1 signaling and are sensitive to rapamycin, thus providing a preclinical rationale for prospective, biomarker-driven clinical studies of mTORC1 inhibitors in these tumors.


Asunto(s)
Integrinas/metabolismo , Neurofibromina 2/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Antibióticos Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina D1/genética , Fase G1 , Humanos , Immunoblotting , Integrinas/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Mesotelioma/genética , Mesotelioma/metabolismo , Mesotelioma/patología , Complejos Multiproteicos , Neurofibromina 2/metabolismo , Biosíntesis de Proteínas , Proteínas , Caperuzas de ARN , Interferencia de ARN , ARN Mensajero/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factores de Transcripción/genética , Transfección
12.
Trends Cell Biol ; 17(5): 222-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17442573

RESUMEN

Inactivation of the tumor suppressor Merlin, encoded by the NF2 (Neurofibromatosis type 2) gene, contributes to malignant conversion in many cell types. Merlin is an Ezrin-Radixin-Moesin protein and localizes underneath the plasma membrane at cell-cell junctions and other actin-rich sites. Recent studies indicate that Merlin mediates contact inhibition of proliferation by blocking recruitment of Rac to the plasma membrane. In mitogen-stimulated cells, p21-activated kinase phosphorylates Ser518 in the C-terminus of Merlin, inactivating the growth suppressive function of the protein. Furthermore, the myosin phosphatase MYPT1-PP1delta, has been identified as a direct activator of Merlin and its inhibition has been linked to malignant transformation. Finally, studies in the fruit fly Drosophila melanogaster have revealed that Merlin functions together with the band 4.1 protein Expanded to promote [corrected] the endocytosis of many signaling receptors, limiting [corrected] their accumulation at the plasma membrane, and to activate [corrected] the Hippo signaling pathway. Here, we review these recent findings and their relevance to the tumor suppressor function of Merlin.


Asunto(s)
Genes Supresores de Tumor/fisiología , Neurofibromina 2/fisiología , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Drosophila melanogaster/fisiología , Genes de la Neurofibromatosis 2 , Humanos , Mutación , Neurofibromina 2/genética
13.
Cancer ; 98(5): 1002-7, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12942568

RESUMEN

BACKGROUND: Recently, the TSLC1 (tumor suppressor in lung cancer 1) gene has been identified as a novel tumor suppressor in human nonsmall cell lung carcinoma. To the authors' knowledge, the clinical relevance of TSLC1 gene expression has not been studied using patient data and surgical samples. The current study was designed to evaluate whether the TSLC1 gene can serve as a target for the prognostic determination of patients with pulmonary adenocarcinoma. METHODS: A total of 38 patients who were surgically treated for proven primary lung adenocarcinoma were enrolled in the current study. Surgical specimens were examined for TSLC1 protein expression immunohistochemically and by Western blot analysis. The correlation between levels of TSLC1 expression and pathologic characteristics, as well as prognosis, was investigated. RESULTS: All patients underwent a potentially curative resection of their tumor. TSLC1 antigen expression as evaluated by immunohistochemistry was confirmed by immunoblotting. The expression of TSLC1 protein was found to be inversely correlated with advanced disease stage, lymph node involvement, lymphatic permeation, and vascular invasion. The 4-year overall survival rates of patients with a tumor demonstrating high (> 70% positive cells [n = 14 patients]), intermediate (20-70% positive cells [n = 10 patients]), and low (< 20% positive cells [n = 14 patients]) expression of the TSLC1 antigen were 84%, 28%, and 7%, respectively. In addition, the disease-free survival of patients with a tumor that demonstrated a high percentage of TSLC1 protein-positive cells was reported to be significantly better than that of patients with a tumor that showed a low percentage of TSLC1 protein-positive cells. CONCLUSIONS: The loss or reduction of TSLC1 expression in resected lung adenocarcinoma cases was associated with a poor prognosis, indicating that TSLC1 represents a central effector gene for controlling the biologic aggressiveness of the tumor and that it is an essential biomarker for predicting patient prognosis. These data may help to detect those patients at high risk for recurrence who might benefit from additional therapeutic strategies such as adjuvant therapy.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Biomarcadores de Tumor , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Inmunoglobulinas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas de la Membrana , Proteínas/análisis , Adenocarcinoma/cirugía , Western Blotting , Molécula 1 de Adhesión Celular , Moléculas de Adhesión Celular , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/cirugía , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Pronóstico , Biosíntesis de Proteínas , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA