Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Proteome Res ; 14(3): 1420-31, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25643065

RESUMEN

Although significant progress has been made in the diagnosis and treatment of colorectal cancer (CRC), it remains a leading cause of cancer death worldwide. Early identification and removal of polyps that may progress to overt CRC is the cornerstone of CRC prevention. Expression of the High Mobility Group A1 (HMGA1) gene is significantly elevated in CRCs as compared with adjacent, nonmalignant tissues. We investigated metabolic aberrations induced by HMGA1 overexpression in small intestinal and colonic epithelium using traveling wave ion mobility mass spectrometry (TWIMMS) in a transgenic model in which murine Hmga1 was misexpressed in colonic epithelium. To determine if these Hmga1-induced metabolic alterations in mice were relevant to human colorectal carcinogenesis, we also investigated tumors from patients with CRC and matched, adjacent, nonmalignant tissues. Multivariate statistical methods and manual comparisons were used to identify metabolites specific to Hmga1 and CRC. Statistical modeling of data revealed distinct metabolic patterns in Hmga1 transgenics and human CRC samples as compared with the control tissues. We discovered that 13 metabolites were specific for Hmga1 in murine intestinal epithelium and also found in human CRC. Several of these metabolites function in fatty acid metabolism and membrane composition. Although further validation is needed, our results suggest that high levels of HMGA1 protein drive metabolic alterations that contribute to CRC pathogenesis through fatty acid synthesis. These metabolites could serve as potential biomarkers or therapeutic targets.


Asunto(s)
Poliposis Adenomatosa del Colon/fisiopatología , Proliferación Celular/fisiología , Neoplasias Colorrectales/patología , Proteína HMGA1a/fisiología , Mucosa Intestinal/patología , Neoplasias Colorrectales/metabolismo , Proteína HMGA1a/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Espectrometría de Masas en Tándem
2.
Pancreatology ; 12(4): 372-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22898640

RESUMEN

CONTEXT: Although pancreatic cancer is a common, highly lethal malignancy, the molecular events that enable precursor lesions to become invasive carcinoma remain unclear. We previously reported that the high-mobility group A1 (HMGA1) protein is overexpressed in >90% of primary pancreatic cancers, with absent or low levels in early precursor lesions. METHODS: Here, we investigate the role of HMGA1 in reprogramming pancreatic epithelium into invasive cancer cells. We assessed oncogenic properties induced by HMGA1 in non-transformed pancreatic epithelial cells expressing activated K-RAS. We also explored the HMGA1-cyclooxygenase (COX-2) pathway in human pancreatic cancer cells and the therapeutic effects of COX-2 inhibitors in xenograft tumorigenesis. RESULTS: HMGA1 cooperates with activated K-RAS to induce migration, invasion, and anchorage-independent cell growth in a cell line derived from normal human pancreatic epithelium. Moreover, HMGA1 and COX-2 expression are positively correlated in pancreatic cancer cell lines (r(2) = 0.93; p < 0.001). HMGA1 binds directly to the COX-2 promoter at an AT-rich region in vivo in three pancreatic cancer cell lines. In addition, HMGA1 induces COX-2 expression in pancreatic epithelial cells, while knock-down of HMGA1 results in repression of COX-2 in pancreatic cancer cells. Strikingly, we also discovered that Sulindac (a COX-1/COX-2 inhibitor) or Celecoxib (a more specific COX-2 inhibitor) block xenograft tumorigenesis from pancreatic cancer cells expressing high levels of HMGA1. CONCLUSIONS: Our studies identify for the first time an important role for the HMGA1-COX-2 pathway in pancreatic cancer and suggest that targeting this pathway could be effective to treat, or even prevent, pancreatic cancer.


Asunto(s)
Adenocarcinoma/genética , Ciclooxigenasa 2/genética , Proteína HMGA1a/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/prevención & control , Animales , Celecoxib , División Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Ciclooxigenasa 2/fisiología , Inhibidores de la Ciclooxigenasa/administración & dosificación , Expresión Génica , Proteína HMGA1a/fisiología , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Trasplante de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/prevención & control , Pirazoles/administración & dosificación , Sulfonamidas/administración & dosificación , Sulindac/administración & dosificación , Trasplante Heterólogo , Proteínas ras/fisiología
3.
BMC Genomics ; 12: 549, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-22053823

RESUMEN

BACKGROUND: Although the high mobility group A1 (HMGA1) gene is widely overexpressed in diverse cancers and portends a poor prognosis in some tumors, the molecular mechanisms that mediate its role in transformation have remained elusive. HMGA1 functions as a potent oncogene in cultured cells and induces aggressive lymphoid tumors in transgenic mice. Because HMGA1 chromatin remodeling proteins regulate transcription, HMGA1 is thought to drive malignant transformation by modulating expression of specific genes. Genome-wide studies to define HMGA1 transcriptional networks during tumorigenesis, however, are lacking. To define the HMGA1 transcriptome, we analyzed gene expression profiles in lymphoid cells from HMGA1a transgenic mice at different stages in tumorigenesis. RESULTS: RNA from lymphoid samples at 2 months (before tumors develop) and 12 months (after tumors are well-established) was screened for differential expression of > 20,000 unique genes by microarray analysis (Affymetrix) using a parametric and nonparametric approach. Differential expression was confirmed by quantitative RT-PCR in a subset of genes. Differentially expressed genes were analyzed for cellular pathways and functions using Ingenuity Pathway Analysis. Early in tumorigenesis, HMGA1 induced inflammatory pathways with NFkappaB identified as a major node. In established tumors, HMGA1 induced pathways involved in cell cycle progression, cell-mediated immune response, and cancer. At both stages in tumorigenesis, HMGA1 induced pathways involved in cellular development, hematopoiesis, and hematologic development. Gene set enrichment analysis showed that stem cell and immature T cell genes are enriched in the established tumors. To determine if these results are relevant to human tumors, we knocked-down HMGA1 in human T-cell leukemia cells and identified a subset of genes dysregulated in both the transgenic and human lymphoid tumors. CONCLUSIONS: We found that HMGA1 induces inflammatory pathways early in lymphoid tumorigenesis and pathways involved in stem cells, cell cycle progression, and cancer in established tumors. HMGA1 also dyregulates genes and pathways involved in stem cells, cellular development and hematopoiesis at both early and late stages of tumorigenesis. These results provide insight into HMGA1 function during tumor development and point to cellular pathways that could serve as therapeutic targets in lymphoid and other human cancers with aberrant HMGA1 expression.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes cdc , Proteína HMGA1a/metabolismo , Inflamación/genética , Tejido Linfoide/patología , Células Madre/metabolismo , Transcriptoma , Animales , Regulación Neoplásica de la Expresión Génica , Proteína HMGA1a/genética , Humanos , Leucemia de Células T/genética , Tejido Linfoide/metabolismo , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Neoplásico/genética
4.
Mod Pathol ; 23(1): 98-104, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19820691

RESUMEN

Although pancreatic ductal adenocarcinoma is a common and almost uniformly fatal cancer, little is known about the molecular events that lead to tumor progression. The high-mobility group A1 (HMGA1) protein is an architectural transcription factor that has been implicated in the pathogenesis and progression of diverse human cancers, including pancreatic ductal adenocarcinoma. In this study, we investigated HMGA1 expression in pancreatic ductal adenocarcinoma cell lines and surgically resected tumors to determine whether it could be a marker for more advanced disease. By real-time quantitative RT-PCR, we measured HMGA1a mRNA in cultured pancreatic ductal adenocarcinoma cell lines and found increased levels in all cancer cells compared with normal pancreatic tissue. To investigate HMGA1 in primary human tumors, we performed immunohistochemical analysis of 125 cases of pancreatic adenocarcinoma and 99 precursor lesions (PanIN 1-3). We found nuclear staining for HMGA1 in 98% of cases of pancreatic adenocarcinoma, but only 43% of cases of PanIN precursor lesions. Moreover, HMGA1 immunoreactivity correlates positively with decreased survival and advanced tumor and PanIN grade. These results suggest that HMGA1 promotes tumor progression in pancreatic ductal adenocarcinoma and could be a useful biomarker and rational therapeutic target in advanced disease.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma Ductal Pancreático/patología , Proteína HMGA1a/biosíntesis , Neoplasias Pancreáticas/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Pronóstico , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares
5.
Mech Ageing Dev ; 128(11-12): 706-16, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18078670

RESUMEN

Manganese superoxide dismutase (MnSOD or SOD2) is a key mitochondrial enzymatic antioxidant. Arguably the most striking phenotype associated with complete loss of SOD2 in flies and mice is shortened life span. To further explore the role of SOD2 in protecting animals from aging and age-associated pathology, we generated a unique collection of Drosophila mutants that progressively reduce SOD2 expression and function. Mitochondrial aconitase activity was substantially reduced in the Sod2 mutants, suggesting that SOD2 normally ensures the functional capacity of mitochondria. Flies with severe reductions in SOD2 expression exhibited accelerated senescence of olfactory behavior as well as precocious neurodegeneration and DNA strand breakage in neurons. Furthermore, life span was progressively shortened and age-dependent mortality was increased in conjunction with reduced SOD2 expression, while initial mortality and developmental viability were unaffected. Interestingly, life span and age-dependent mortality varied exponentially with SOD2 activity, indicating that there might normally be a surplus of this enzyme for protecting animals from premature death. Our data support a model in which disruption of the protective effects of SOD2 on mitochondria manifests as profound changes in behavioral and demographic aging as well as exacerbated age-related pathology in the nervous system.


Asunto(s)
Envejecimiento/metabolismo , Antioxidantes/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Longevidad , Mitocondrias/enzimología , Superóxido Dismutasa/metabolismo , Aconitato Hidratasa/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Conducta Animal , Encéfalo/enzimología , Encéfalo/patología , Roturas del ADN , Regulación hacia Abajo , Drosophila/genética , Proteínas de Drosophila/genética , Longevidad/genética , Mutación , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/patología , Estrés Oxidativo , Olfato/genética , Superóxido Dismutasa/genética
6.
Nat Commun ; 8: 15008, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28452345

RESUMEN

High-mobility group A1 (Hmga1) chromatin remodelling proteins are enriched in intestinal stem cells (ISCs), although their function in this setting was unknown. Prior studies showed that Hmga1 drives hyperproliferation, aberrant crypt formation and polyposis in transgenic mice. Here we demonstrate that Hmga1 amplifies Wnt/ß-catenin signalling to enhance self-renewal and expand the ISC compartment. Hmga1 upregulates genes encoding both Wnt agonist receptors and downstream Wnt effectors. Hmga1 also helps to 'build' an ISC niche by expanding the Paneth cell compartment and directly inducing Sox9, which is required for Paneth cell differentiation. In human intestine, HMGA1 and SOX9 are positively correlated, and both become upregulated in colorectal cancer. Our results define a unique role for Hmga1 in intestinal homeostasis by maintaining the stem cell pool and fostering terminal differentiation to establish an epithelial stem cell niche. This work also suggests that deregulated Hmga1 perturbs this equilibrium during intestinal carcinogenesis.


Asunto(s)
Proteína HMGA1a/metabolismo , Mucosa Intestinal/metabolismo , Células de Paneth/metabolismo , Células Madre/metabolismo , Vía de Señalización Wnt , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Proteína HMGA1a/genética , Humanos , Mucosa Intestinal/citología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Células de Paneth/citología , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Nicho de Células Madre , Células Madre/citología , Imagen de Lapso de Tiempo
7.
Genetics ; 165(4): 2295-9, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14704205

RESUMEN

A null mutation for the Sod2 gene, Sod2n283, was obtained in Drosophila melanogaster. Homozygous Sod2 null (Sodn283/Sodn283) adult flies survive up to 24 hr following eclosion, a phenotype reminiscent of mice, where Sod2-/- progeny suffer neonatal lethality. Sodn283/+ heterozygotes are sensitive to oxidative stress induced by paraquat treatment.


Asunto(s)
Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Genes Letales , Mutación , Superóxido Dismutasa/fisiología , Animales , Animales Modificados Genéticamente , Drosophila melanogaster/efectos de los fármacos , Femenino , Herbicidas/toxicidad , Homocigoto , Esperanza de Vida , Masculino , Estrés Oxidativo , Paraquat/toxicidad , Fenotipo , Tasa de Supervivencia
8.
PLoS One ; 8(5): e63419, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23658826

RESUMEN

Emerging evidence suggests that tumor cells metastasize by co-opting stem cell transcriptional networks, although the molecular underpinnings of this process are poorly understood. Here, we show for the first time that the high mobility group A1 (HMGA1) gene drives metastatic progression in triple negative breast cancer cells (MDA-MB-231, Hs578T) by reprogramming cancer cells to a stem-like state. Silencing HMGA1 expression in invasive, aggressive breast cancer cells dramatically halts cell growth and results in striking morphologic changes from mesenchymal-like, spindle-shaped cells to cuboidal, epithelial-like cells. Mesenchymal genes (Vimentin, Snail) are repressed, while E-cadherin is induced in the knock-down cells. Silencing HMGA1 also blocks oncogenic properties, including proliferation, migration, invasion, and orthotopic tumorigenesis. Metastatic progression following mammary implantation is almost completely abrogated in the HMGA1 knock-down cells. Moreover, silencing HMGA1 inhibits the stem cell property of three-dimensional mammosphere formation, including primary, secondary, and tertiary spheres. In addition, knock-down of HMGA1 depletes cancer initiator/cancer stem cells and prevents tumorigenesis at limiting dilutions. We also discovered an HMGA1 signature in triple negative breast cancer cells that is highly enriched in embryonic stem cells. Together, these findings indicate that HMGA1 is a master regulator of tumor progression in breast cancer by reprogramming cancer cells through stem cell transcriptional networks. Future studies are needed to determine how to target HMGA1 in therapy.


Asunto(s)
Progresión de la Enfermedad , Proteína HMGA1a/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Proteína HMGA1a/deficiencia , Proteína HMGA1a/genética , Humanos , Mesodermo/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Neoplasias de la Mama Triple Negativas/metabolismo
9.
PLoS One ; 7(1): e30034, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22276142

RESUMEN

BACKGROUND: Although metastatic colon cancer is a leading cause of cancer death worldwide, the molecular mechanisms that enable colon cancer cells to metastasize remain unclear. Emerging evidence suggests that metastatic cells develop by usurping transcriptional networks from embryonic stem (ES) cells to facilitate an epithelial-mesenchymal transition (EMT), invasion, and metastatic progression. Previous studies identified HMGA1 as a key transcription factor enriched in ES cells, colon cancer, and other aggressive tumors, although its role in these settings is poorly understood. METHODS/PRINCIPAL FINDINGS: To determine how HMGA1 functions in metastatic colon cancer, we manipulated HMGA1 expression in transgenic mice and colon cancer cells. We discovered that HMGA1 drives proliferative changes, aberrant crypt formation, and intestinal polyposis in transgenic mice. In colon cancer cell lines from poorly differentiated, metastatic tumors, knock-down of HMGA1 blocks anchorage-independent cell growth, migration, invasion, xenograft tumorigenesis and three-dimensional colonosphere formation. Inhibiting HMGA1 expression blocks tumorigenesis at limiting dilutions, consistent with depletion of tumor-initiator cells in the knock-down cells. Knock-down of HMGA1 also inhibits metastatic progression to the liver in vivo. In metastatic colon cancer cells, HMGA1 induces expression of Twist1, a gene involved in embryogenesis, EMT, and tumor progression, while HMGA1 represses E-cadherin, a gene that is down-regulated during EMT and metastatic progression. In addition, HMGA1 is among the most enriched genes in colon cancer compared to normal mucosa. CONCLUSIONS: Our findings demonstrate for the first time that HMGA1 drives proliferative changes and polyp formation in the intestines of transgenic mice and induces metastatic progression and stem-like properties in colon cancer cells. These findings indicate that HMGA1 is a key regulator, both in metastatic progression and in the maintenance of a stem-like state. Our results also suggest that HMGA1 or downstream pathways could be rational therapeutic targets in metastatic, poorly differentiated colon cancer.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Proteínas HMGA/metabolismo , Poliposis Intestinal/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Neoplasias del Colon/genética , Células HCT116 , Proteínas HMGA/genética , Humanos , Inmunohistoquímica , Poliposis Intestinal/genética , Ratones , Ratones Desnudos , Ratones Transgénicos , Invasividad Neoplásica/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
10.
PLoS One ; 7(11): e48533, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166588

RESUMEN

BACKGROUND: Although recent studies have identified genes expressed in human embryonic stem cells (hESCs) that induce pluripotency, the molecular underpinnings of normal stem cell function remain poorly understood. The high mobility group A1 (HMGA1) gene is highly expressed in hESCs and poorly differentiated, stem-like cancers; however, its role in these settings has been unclear. METHODS/PRINCIPAL FINDINGS: We show that HMGA1 is highly expressed in fully reprogrammed iPSCs and hESCs, with intermediate levels in ECCs and low levels in fibroblasts. When hESCs are induced to differentiate, HMGA1 decreases and parallels that of other pluripotency factors. Conversely, forced expression of HMGA1 blocks differentiation of hESCs. We also discovered that HMGA1 enhances cellular reprogramming of somatic cells to iPSCs together with the Yamanaka factors (OCT4, SOX2, KLF4, cMYC - OSKM). HMGA1 increases the number and size of iPSC colonies compared to OSKM controls. Surprisingly, there was normal differentiation in vitro and benign teratoma formation in vivo of the HMGA1-derived iPSCs. During the reprogramming process, HMGA1 induces the expression of pluripotency genes, including SOX2, LIN28, and cMYC, while knockdown of HMGA1 in hESCs results in the repression of these genes. Chromatin immunoprecipitation shows that HMGA1 binds to the promoters of these pluripotency genes in vivo. In addition, interfering with HMGA1 function using a short hairpin RNA or a dominant-negative construct blocks cellular reprogramming to a pluripotent state. CONCLUSIONS: Our findings demonstrate for the first time that HMGA1 enhances cellular reprogramming from a somatic cell to a fully pluripotent stem cell. These findings identify a novel role for HMGA1 as a key regulator of the stem cell state by inducing transcriptional networks that drive pluripotency. Although further studies are needed, these HMGA1 pathways could be exploited in regenerative medicine or as novel therapeutic targets for poorly differentiated, stem-like cancers.


Asunto(s)
Desdiferenciación Celular/fisiología , Células Madre Embrionarias/citología , Regulación de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Proteína HMGA1a/metabolismo , Células Madre Pluripotentes/citología , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Redes Reguladoras de Genes/genética , Proteína HMGA1a/genética , Humanos , Factor 4 Similar a Kruppel , Regiones Promotoras Genéticas/genética , Interferencia de ARN , Retroviridae , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sales de Tetrazolio , Tiazoles , Factores de Transcripción/metabolismo , Transducción Genética
11.
Fly (Austin) ; 5(1): 39-46, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21212740

RESUMEN

Definitive evidence on the impact of MnSOD/SOD2-deficiency and the consequent effects of high flux of mitochondrial reactive oxygen species (ROS) on pre-natal/pre-adult development has yet to be reported for either Drosophila or mice. Here we report that oocytes lacking maternal SOD2 protein develop into adults just like normal SOD2-containing oocytes suggesting that maternal SOD2-mediated protection against mitochondrial ROS is not essential for oocyte viability. However, the capacity of SOD2-null larvae to undergo successful metamorphosis into adults is negatively influenced in the absence of SOD2. We therefore determined the impact of a high superoxide environment on cell size, progression through the cell cycle, cell differentiation, and cell death and found no difference between SOD2-null and SOD2+ larva and pupa. Thus loss of SOD2 activity clearly has no effect on pre-adult imaginal tissues. Instead, we found that the high mitochondrial superoxide environment arising from the absence of SOD2 leads to the induction of autophagy. Such autophagic response may underpin the resistance of pre-adult tissues to unscavenged ROS. Finally, while our data establish that SOD2 activity is less essential for normal development, the mortality of Sod2-/- neonates of both Drosophila and mice suggests that SOD2 activity is indeed essential for the viability of adults. We therefore asked if the early mortality of SOD2-null young adults could be rescued by activation of SOD2 expression. The results support the conclusion that the early mortality of SOD2-null adults is largely attributable to the absence of SOD2 activity in the adult per se. This finding somewhat contradicts the widely held notion that failure to scavenge the high volume of superoxide emanating from the oxidative demands of development would be highly detrimental to developing tissues.


Asunto(s)
Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Animales , Autofagia/genética , Drosophila melanogaster/enzimología , Mitocondrias/enzimología , Mutación , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/deficiencia , Superóxidos/metabolismo
12.
Leuk Lymphoma ; 57(11): 2681-4, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26952843
14.
Mol Cancer Res ; 7(11): 1803-12, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19903768

RESUMEN

Although lung cancer is the leading cause of cancer death worldwide, the precise molecular mechanisms that give rise to lung cancer are incompletely understood. Here, we show that HMGA1 is an important oncogene that drives transformation in undifferentiated, large-cell carcinoma. First, we show that the HMGA1 gene is overexpressed in lung cancer cell lines and primary human lung tumors. Forced overexpression of HMGA1 induces a transformed phenotype with anchorage-independent cell growth in cultured lung cells derived from normal tissue. Conversely, inhibiting HMGA1 expression blocks anchorage-independent cell growth in the H1299 metastatic, undifferentiated, large-cell human lung carcinoma cells. We also show that the matrix metalloproteinase-2 (MMP-2) gene is a downstream target upregulated by HMGA1 in large-cell carcinoma cells. In chromatin immunoprecipitation experiments, HMGA1 binds directly to the MMP-2 promoter in vivo in large-cell lung cancer cells, but not in squamous cell carcinoma cells. In large-cell carcinoma cell lines, there is a significant, positive correlation between HMGA1 and MMP-2 mRNA. Moreover, interfering with MMP-2 expression blocks anchorage-independent cell growth in H1299 large-cell carcinoma cells, indicating that the HMGA1-MMP-2 pathway is required for this transformation phenotype in these cells. Blocking MMP-2 expression also inhibits migration and invasion in the H1299 large-cell carcinoma cells. Our findings suggest an important role for MMP-2 in transformation mediated by HMGA1 in large-cell, undifferentiated lung carcinoma and support the development of strategies to target this pathway in selected tumors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Transformación Celular Neoplásica/metabolismo , Proteína HMGA1a/metabolismo , Neoplasias Pulmonares/metabolismo , Metaloproteinasa 2 de la Matriz/biosíntesis , Carcinoma de Células Grandes/enzimología , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/metabolismo , Carcinoma de Células Grandes/patología , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteína HMGA1a/antagonistas & inhibidores , Proteína HMGA1a/biosíntesis , Proteína HMGA1a/genética , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transfección , Regulación hacia Arriba
16.
Cancer Res ; 68(24): 10121-7, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19074878

RESUMEN

Although HMGA1 (high-mobility group A1; formerly HMG-I/Y) is an oncogene that is widely overexpressed in aggressive cancers, the molecular mechanisms underlying transformation by HMGA1 are only beginning to emerge. HMGA1 encodes the HMGA1a and HMGA1b protein isoforms, which function in regulating gene expression. To determine how HMGA1 leads to neoplastic transformation, we looked for genes regulated by HMGA1 using gene expression profile analysis. Here, we show that the STAT3 gene, which encodes the signaling molecule signal transducer and activator of transcription 3 (STAT3), is a critical downstream target of HMGA1a. STAT3 mRNA and protein are up-regulated in fibroblasts overexpressing HMGA1a and activated STAT3 recapitulates the transforming activity of HMGA1a in fibroblasts. HMGA1a also binds directly to a conserved region of the STAT3 promoter in vivo in human leukemia cells by chromatin immunoprecipitation and activates transcription of the STAT3 promoter in transfection experiments. To determine if this pathway contributes to HMGA1-mediated transformation, we investigated STAT3 expression in our HMGA1a transgenic mice, all of which developed aggressive lymphoid malignancy. STAT3 expression was increased in the leukemia cells from our transgenics but not in control cells. Blocking STAT3 function induced apoptosis in the transgenic leukemia cells but not in controls. In primary human leukemia samples, there was a positive correlation between HMGA1a and STAT3 mRNA. Moreover, blocking STAT3 function in human leukemia or lymphoma cells led to decreased cellular motility and foci formation. Our results show that the HMGA1a-STAT3 axis is a potential Achilles heel that could be exploited therapeutically in hematopoietic and other malignancies overexpressing HMGA1a.


Asunto(s)
Proteína HMGA1a/genética , Neoplasias Hematológicas/genética , Factor de Transcripción STAT3/genética , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Proteína HMGA1a/biosíntesis , Proteína HMGA1a/metabolismo , Neoplasias Hematológicas/metabolismo , Humanos , Ratones , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/metabolismo , Transfección , Regulación hacia Arriba
17.
Genome ; 49(7): 746-51, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16936783

RESUMEN

Two deletions, Df(2R)Sod2-11 and Df(2R)Sod2-332, are recovered that encompass the manganese superoxide dismutase (MnSOD) gene or a null mutant referred to as SOD2n283 in Drosophila. Molecular analysis has revealed that the Df(2R)Sod2-332 deletion completely uncovered both MnSOD and its adjacent gene, Arp53D, whereas Df(2R)Sod2-11 was missing the promoter region of MnSOD gene. As a consequence of reduced MnSOD expression, these deletion heterozygotes are now sensitive to oxidative stress. Complementation analysis with some recently recovered deletions in the 53C/D region has established that other essential loci exist in this interval, and second, that Arp53D function is not essential for the survival of the organism. These deletions will be instrumental in the recovery of missense substitutions in the MnSOD peptide and their influence on oxidative stress resistance.


Asunto(s)
Drosophila melanogaster/enzimología , Genoma de los Insectos , Superóxido Dismutasa/genética , Actinas/genética , Animales , Secuencia de Bases , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Eliminación de Gen , Homocigoto , Datos de Secuencia Molecular , Estrés Oxidativo , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA