Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 164(1-2): 293-309, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26771497

RESUMEN

Large-scale genomic studies have identified multiple somatic aberrations in breast cancer, including copy number alterations and point mutations. Still, identifying causal variants and emergent vulnerabilities that arise as a consequence of genetic alterations remain major challenges. We performed whole-genome small hairpin RNA (shRNA) "dropout screens" on 77 breast cancer cell lines. Using a hierarchical linear regression algorithm to score our screen results and integrate them with accompanying detailed genetic and proteomic information, we identify vulnerabilities in breast cancer, including candidate "drivers," and reveal general functional genomic properties of cancer cells. Comparisons of gene essentiality with drug sensitivity data suggest potential resistance mechanisms, effects of existing anti-cancer drugs, and opportunities for combination therapy. Finally, we demonstrate the utility of this large dataset by identifying BRD4 as a potential target in luminal breast cancer and PIK3CA mutations as a resistance determinant for BET-inhibitors.


Asunto(s)
Algoritmos , Neoplasias de la Mama/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I , Análisis por Conglomerados , Resistencia a Antineoplásicos , Dosificación de Gen , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Modelos Lineales , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas , Factores de Transcripción/genética
2.
Adv Anat Embryol Cell Biol ; 236: 21-55, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37955770

RESUMEN

The ability to assess various cellular events consequent to perturbations, such as genetic mutations, disease states and therapies, has been recently revolutionized by technological advances in multiple "omics" fields. The resulting deluge of information has enabled and necessitated the development of tools required to both process and interpret the data. While of tremendous value to basic researchers, the amount and complexity of the data has made it extremely difficult to manually draw inference and identify factors key to the study objectives. The challenges of data reduction and interpretation are being met by the development of increasingly complex tools that integrate disparate knowledge bases and synthesize coherent models based on current biological understanding. This chapter presents an example of how genomics data can be integrated with biological network analyses to gain further insight into the developmental consequences of genetic perturbations. State of the art methods for conducting similar studies are discussed along with modern methods used to analyze and interpret the data.


Asunto(s)
Biología Computacional , Biología de Sistemas , Genómica , Músculo Esquelético , Bases del Conocimiento
3.
Haematologica ; 108(2): 382-393, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36134452

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most frequent cancer diagnosed in children. Despite the great progress achieved over the last 40 years, with cure rates now exceeding 85%, refractory or relapsed ALL still exhibit a dismal prognosis. This poor outcome reflects the lack of treatment options specifically targeting relapsed or refractory ALL. In order to address this gap, we performed whole-genome CRISPR/Cas drop-out screens on a panel of seven B-ALL cell lines. Our results demonstrate that while there was a significant overlap in gene essentiality between ALL cell lines and other cancer types survival of ALL cell lines was dependent on several unique metabolic pathways, including an exquisite sensitivity to GPX4 depletion and ferroptosis induction. Detailed molecular analysis of B-ALL cells suggest that they are primed to undergo ferroptosis as they exhibit high steady-state oxidative stress potential, a low buffering capacity, and a disabled GPX4-independent secondary lipid peroxidation detoxification pathway. Finally, we validated the sensitivity of BALL to ferroptosis induction using patient-derived B-ALL samples.


Asunto(s)
Ferroptosis , Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Humanos , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Ferroptosis/genética , Línea Celular , Peroxidación de Lípido , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico
4.
Nature ; 525(7569): 384-8, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26331541

RESUMEN

MYC (also known as c-MYC) overexpression or hyperactivation is one of the most common drivers of human cancer. Despite intensive study, the MYC oncogene remains recalcitrant to therapeutic inhibition. MYC is a transcription factor, and many of its pro-tumorigenic functions have been attributed to its ability to regulate gene expression programs. Notably, oncogenic MYC activation has also been shown to increase total RNA and protein production in many tissue and disease contexts. While such increases in RNA and protein production may endow cancer cells with pro-tumour hallmarks, this increase in synthesis may also generate new or heightened burden on MYC-driven cancer cells to process these macromolecules properly. Here we discover that the spliceosome is a new target of oncogenic stress in MYC-driven cancers. We identify BUD31 as a MYC-synthetic lethal gene in human mammary epithelial cells, and demonstrate that BUD31 is a component of the core spliceosome required for its assembly and catalytic activity. Core spliceosomal factors (such as SF3B1 and U2AF1) associated with BUD31 are also required to tolerate oncogenic MYC. Notably, MYC hyperactivation induces an increase in total precursor messenger RNA synthesis, suggesting an increased burden on the core spliceosome to process pre-mRNA. In contrast to normal cells, partial inhibition of the spliceosome in MYC-hyperactivated cells leads to global intron retention, widespread defects in pre-mRNA maturation, and deregulation of many essential cell processes. Notably, genetic or pharmacological inhibition of the spliceosome in vivo impairs survival, tumorigenicity and metastatic proclivity of MYC-dependent breast cancers. Collectively, these data suggest that oncogenic MYC confers a collateral stress on splicing, and that components of the spliceosome may be therapeutic entry points for aggressive MYC-driven cancers.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Genes myc/genética , Empalmosomas/efectos de los fármacos , Empalmosomas/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Intrones/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Precursores del ARN/biosíntesis , Precursores del ARN/genética , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Ribonucleoproteínas/metabolismo , Factor de Empalme U2AF , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Biol Chem ; 290(37): 22520-31, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26221032

RESUMEN

CUX1 and CUX2 proteins are characterized by the presence of three highly similar regions called Cut repeats 1, 2, and 3. Although CUX1 is ubiquitously expressed, CUX2 plays an important role in the specification of neuronal cells and continues to be expressed in postmitotic neurons. Cut repeats from the CUX1 protein were recently shown to stimulate 8-oxoguanine DNA glycosylase 1 (OGG1), an enzyme that removes oxidized purines from DNA and introduces a single strand break through its apurinic/apyrimidinic lyase activity to initiate base excision repair. Here, we investigated whether CUX2 plays a similar role in the repair of oxidative DNA damage. Cux2 knockdown in embryonic cortical neurons increased levels of oxidative DNA damage. In vitro, Cut repeats from CUX2 increased the binding of OGG1 to 7,8-dihydro-8-oxoguanine-containing DNA and stimulated both the glycosylase and apurinic/apyrimidinic lyase activities of OGG1. Genetic inactivation in mouse embryo fibroblasts or CUX2 knockdown in HCC38 cells delayed DNA repair and increased DNA damage. Conversely, ectopic expression of Cut repeats from CUX2 accelerated DNA repair and reduced levels of oxidative DNA damage. These results demonstrate that CUX2 functions as an accessory factor that stimulates the repair of oxidative DNA damage. Neurons produce a high level of reactive oxygen species because of their dependence on aerobic oxidation of glucose as their source of energy. Our results suggest that the persistent expression of CUX2 in postmitotic neurons contributes to the maintenance of genome integrity through its stimulation of oxidative DNA damage repair.


Asunto(s)
Corteza Cerebral/metabolismo , Daño del ADN , Reparación del ADN , Proteínas de Homeodominio/metabolismo , Neuronas/metabolismo , Animales , Línea Celular , Corteza Cerebral/citología , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Ratones , Neuronas/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Ratas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Proc Natl Acad Sci U S A ; 109(8): 2808-13, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-21628573

RESUMEN

The tyrosine kinase c-Src is activated in a large proportion of breast cancers, in which it is thought to play a key role in promoting the malignant phenotype. c-Src activity is also elevated in transgenic mouse models of breast cancer, including the widely used polyomavirus middle-T antigen (PyVmT) model, which provides an opportunity to study the importance of c-Src in mammary tumorigenesis. However, germline c-Src deletion in mammary epithelial and stromal compartments complicates the interpretation of in vivo tumorigenesis studies as a result of severe defects in mammary gland development. We have therefore engineered a mouse strain in which deletion of c-Src can be targeted to the mammary epithelium. We demonstrate that mammary epithelial disruption of c-Src impairs proliferation and tumor progression driven by PyVmT in vivo. Whereas related kinases substitute for c-Src in PyVmT signaling, c-Src ablation impairs cell cycle progression with decreased cyclin expression and elevated expression of cyclin-dependent kinase inhibitors. Our data indicate that c-Src has essential and unique functions in proliferation and tumor progression in this mouse model that may also be important in certain contexts in some human breast cancers.


Asunto(s)
Ciclo Celular , Transformación Celular Neoplásica/patología , Epitelio/enzimología , Epitelio/patología , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/patología , Familia-src Quinasas/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Adhesión Celular , Proliferación Celular , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/metabolismo , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Silenciador del Gen , Humanos , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Ratones Desnudos , Especificidad de Órganos , Fosforilación
7.
J Mammary Gland Biol Neoplasia ; 13(3): 323-35, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18651209

RESUMEN

The advent of genetically engineered mouse models (GEMs) of human breast cancer, have provided important insight into molecular basis or human breast cancer. This review will focus on two of the most extensively studied mouse models for human breast cancer involving mammary gland specific expression of the polyoma middle T (PyV MT) antigen and of the ErbB2. In addition, this review will discuss past and recent advances in understanding relative contribution of the signaling pathways in tumor induction and metastasis by these potent mammary oncogenes.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Transducción de Señal , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Humanos , Integrinas/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Metástasis de la Neoplasia , Oncogenes , Receptor ErbB-2/metabolismo
8.
NPJ Precis Oncol ; 3: 28, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31701019

RESUMEN

Germline variants such as BRCA1/2 play an important role in tumorigenesis and clinical outcomes of cancer patients. However, only a small fraction (i.e., 5-10%) of inherited variants has been associated with clinical outcomes (e.g., BRCA1/2, APC, TP53, PTEN and so on). The challenge remains in using these inherited germline variants to predict clinical outcomes of cancer patient population. In an attempt to solve this issue, we applied our recently developed algorithm, eTumorMetastasis, which constructs predictive models, on exome sequencing data to ER+ breast (n = 755) cancer patients. Gene signatures derived from the genes containing functionally germline variants significantly distinguished recurred and non-recurred patients in two ER+ breast cancer independent cohorts (n = 200 and 295, P = 1.4 × 10-3). Furthermore, we compared our results with the widely known Oncotype DX test (i.e., Oncotype DX breast cancer recurrence score) and outperformed prediction for both high- and low-risk groups. Finally, we found that recurred patients possessed a higher rate of germline variants. In addition, the inherited germline variants from these gene signatures were predominately enriched in T cell function, antigen presentation, and cytokine interactions, likely impairing the adaptive and innate immune response thus favoring a pro-tumorigenic environment. Hence, germline genomic information could be used for developing non-invasive genomic tests for predicting patients' outcomes in breast cancer.

9.
Sci Rep ; 9(1): 10989, 2019 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-31358840

RESUMEN

The efficacy of prospective cancer treatments is routinely estimated by in vitro cell-line proliferation screens. However, it is unclear whether tumor aggressiveness and patient survival are influenced more by the proliferative or the migratory properties of cancer cells. To address this question, we experimentally measured proliferation and migration phenotypes across more than 40 breast cancer cell-lines. Based on the latter, we built and validated individual predictors of breast cancer proliferation and migration levels from the cells' transcriptomics. We then apply these predictors to estimate the proliferation and migration levels of more than 1000 TCGA breast cancer tumors. Reassuringly, both estimates increase with tumor's aggressiveness, as qualified by its stage, grade, and subtype. However, predicted tumor migration levels are significantly more strongly associated with patient survival than the proliferation levels. We confirmed these findings by conducting siRNA knock-down experiments on the highly migratory MDA-MB-231 cell lines and deriving gene knock-down based proliferation and migration signatures. We show that cytoskeletal drugs might be more beneficial in patients with high predicted migration levels. Taken together, these results testify to the importance of migration levels in determining patient survival.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Transcriptoma , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica/diagnóstico , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Pronóstico , Análisis de Supervivencia
10.
Nat Cell Biol ; 18(7): 803-813, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27323329

RESUMEN

Tumours exist in a hypoxic microenvironment and must limit excessive oxygen consumption. Hypoxia-inducible factor (HIF) controls mitochondrial oxygen consumption, but how/if tumours regulate non-mitochondrial oxygen consumption (NMOC) is unknown. Protein-tyrosine phosphatase-1B (PTP1B) is required for Her2/Neu-driven breast cancer (BC) in mice, although the underlying mechanism and human relevance remain unclear. We found that PTP1B-deficient HER2(+) xenografts have increased hypoxia, necrosis and impaired growth. In vitro, PTP1B deficiency sensitizes HER2(+) BC lines to hypoxia by increasing NMOC by α-KG-dependent dioxygenases (α-KGDDs). The moyamoya disease gene product RNF213, an E3 ligase, is negatively regulated by PTP1B in HER2(+) BC cells. RNF213 knockdown reverses the effects of PTP1B deficiency on α-KGDDs, NMOC and hypoxia-induced death of HER2(+) BC cells, and partially restores tumorigenicity. We conclude that PTP1B acts via RNF213 to suppress α-KGDD activity and NMOC. This PTP1B/RNF213/α-KGDD pathway is critical for survival of HER2(+) BC, and possibly other malignancies, in the hypoxic tumour microenvironment.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Consumo de Oxígeno/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula , Femenino , Genes erbB-2/genética , Humanos , Ratones , Mitocondrias/metabolismo
11.
FASEB J ; 16(8): 899-901, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12039873

RESUMEN

Spaceflight, just like aging, causes profound changes in musculoskeletal parameters, which result in decreased bone density and muscular weakness. As these conditions decrease our ability to conduct long-term manned space missions, and increase bone frailty in the elderly, the identification of genes responsible for the apparition of these physiological changes will be of great benefit. Thus, we developed and implemented a new microarray approach to investigate the changes in normal WI38 human fibroblast gene expression that arise as a consequence of space flight. Using our microarray, we identified changes in the level of expression of 10 genes, belonging to either the tumor necrosis factor- (TNF) or interleukin- (IL) related gene families in fibroblasts when WI38 cells exposed to microgravity during the STS-93 Space Shuttle mission were compared with ground controls. The genes included two ligands from the TNF superfamily, TWEAK and TNFSF15; two TNF receptor-associated proteins, NSMAF and PTPN13; three TNF-inducible genes, ABC50, PTX3, and SCYA13; TNF-alpha converting enzyme, IL-1 receptor antagonist, and IL-15 receptor alpha chain. Most of these are involved in either the regulation of bone density, and as such the development of spaceflight osteopenia, or in the development of proinflammatory status.


Asunto(s)
Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Interleucinas/genética , Vuelo Espacial , Factor de Necrosis Tumoral alfa/genética , Línea Celular , Fibroblastos/citología , Regulación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Mech Ageing Dev ; 125(10-11): 777-83, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15541772

RESUMEN

In replicative senescence, cells undergo permanent exit from cell cycle traverse; this is traditionally thought to occur at the end of a culture's in vitro life span, after serial passaging. In general, the checkpoint for replicative senescence is found at the G(1)/S border, controlled by the modulation of a battery of proteins, typified by gaining inhibitors of cell cycle traverse, such as cyclin-dependent kinases or RB hyperphosphorylation, and losing pro-proliferation gene expressions such as c-fos, c-myc, and a cadre of proliferation-dependent kinases. Here, we present evidence that replicatively senescent fibroblasts are resistant to apoptotic death, associated with a lack of key enzyme activities, caspase-3 being the chief executioner. This observation, coupled with our earlier report that senescent fibroblasts maintain persistently high levels of pro-survival factor Bcl-2, suggests that the molecular signaling program present in fibroblasts at the end of their in vitro life span may not only cater to the state of permanent exit from cell cycle traverse, but also dictate an inability to commit cellular suicide. Future experiments will reveal whether replicatively senescent fibroblasts that can neither proliferate nor die contribute to organismic aging, and whether their accumulation over time in tissue becomes detrimental to the normal aging process.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Senescencia Celular/fisiología , Regulación hacia Abajo/fisiología , Fibroblastos/fisiología , Apoptosis/genética , Caspasa 3 , Caspasas/genética , Células Cultivadas , Senescencia Celular/genética , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo/genética , Fase G1/genética , Fase G1/fisiología , Humanos , Fase S/genética , Fase S/fisiología
13.
J Gerontol A Biol Sci Med Sci ; 57(7): B257-69, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12084796

RESUMEN

Forty years after its discovery, replicative senescence remains a rich source of information about cell-cycle regulation and the progression from a normal to a transformed phenotype. Effectors of this growth-arrested state are being discovered at a great pace. This review discusses the latest findings on the players responsible for establishing replicative senescence, as well as the associated telomere shortening.


Asunto(s)
Senescencia Celular/fisiología , Animales , Ciclo Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/fisiología , Humanos , Proteína de Retinoblastoma/fisiología , Telómero , Proteína p14ARF Supresora de Tumor/fisiología , Proteína p53 Supresora de Tumor/fisiología
14.
Cancer Cell ; 25(2): 181-95, 2014 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-24525234

RESUMEN

Cellular transformation by oncogenic RAS engages the MAPK pathway under strict regulation by the scaffold protein KSR-1. Here, we report that the guanine nucleotide exchange factor GEF-H1 plays a critical role in a positive feedback loop for the RAS/MAPK pathway independent of its RhoGEF activity. GEF-H1 acts as an adaptor protein linking the PP2A B' subunits to KSR-1, thereby mediating the dephosphorylation of KSR-1 S392 and activation of MAPK signaling. GEF-H1 is important for the growth and survival of HRAS(V12)-transformed cells and pancreatic tumor xenografts. GEF-H1 expression is induced by oncogenic RAS and is correlated with pancreatic neoplastic progression. Our results, therefore, identify GEF-H1 as an amplifier of MAPK signaling and provide mechanistic insight into the progression of RAS mutant tumors.


Asunto(s)
Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/patología , Proteínas Quinasas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas ras/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Técnicas para Inmunoenzimas , Ratones , Células 3T3 NIH , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosforilación , Regiones Promotoras Genéticas/genética , Proteínas Quinasas/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Transducción de Señal , Células Tumorales Cultivadas , Proteínas ras/genética
15.
Cancer Discov ; 2(2): 172-189, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22585861

RESUMEN

UNLABELLED: Genomic analyses are yielding a host of new information on the multiple genetic abnormalities associated with specific types of cancer. A comprehensive description of cancer-associated genetic abnormalities can improve our ability to classify tumors into clinically relevant subgroups and, on occasion, identify mutant genes that drive the cancer phenotype ("drivers"). More often, though, the functional significance of cancer-associated mutations is difficult to discern. Genome-wide pooled short hairpin RNA (shRNA) screens enable global identification of the genes essential for cancer cell survival and proliferation, providing a "functional genomic" map of human cancer to complement genomic studies. Using a lentiviral shRNA library targeting ~16,000 genes and a newly developed, dynamic scoring approach, we identified essential gene profiles in 72 breast, pancreatic, and ovarian cancer cell lines. Integrating our results with current and future genomic data should facilitate the systematic identification of drivers, unanticipated synthetic lethal relationships, and functional vulnerabilities of these tumor types. SIGNIFICANCE: This study presents a resource of genome-scale, pooled shRNA screens for 72 breast, pancreatic, and ovarian cancer cell lines that will serve as a functional complement to genomics data, facilitate construction of essential gene profiles, help uncover synthetic lethal relationships, and identify uncharacterized genetic vulnerabilities in these tumor types. SIGNIFICANCE: This study presents a resource of genome-scale, pooled shRNA screens for 72 breast, pancreatic, and ovarian cancer cell lines that will serve as a functional complement to genomics data, facilitate construction of essential gene profiles, help uncover synthetic lethal relationships, and identify uncharacterized genetic vulnerabilities in these tumor types.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Ováricas/genética , Neoplasias Pancreáticas/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Biblioteca de Genes , Humanos , Masculino , Neoplasias Ováricas/metabolismo , Neoplasias Pancreáticas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transcriptoma
16.
Mol Cell Biol ; 29(21): 5858-71, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19704002

RESUMEN

Previous studies have demonstrated that c-Src tyrosine kinase interacts specifically with ErbB2, but not with other members of the epidermal growth factor receptor (EGFR) family. To identify the site of interaction, we recently used a chimeric EGFR/ErbB2 receptor approach to show that c-Src requires the kinase region of ErbB2 for binding. Here, we demonstrate that retention of a conserved amino acid motif surrounding tyrosine 877 (referred to here as EGFR(YHAD)) is sufficient to confer binding to c-Src. Surprisingly the association of c-Src was not dependent on its SH2 or SH3 domain or on the phosphorylation or kinase activity of the receptor. We further show that the chimeric EGFRs that contain the Y877 motif are transforming in vitro and in vivo following ligand stimulation. Transformation was also partially dependent on sustained activation of Stat3. Finally, we demonstrate that EGFRs with mutations in the catalytic domain, originally identified in lung cancer and conferring increased sensitivity to gefitinib and erlotinib, two EGFR kinase inhibitors, gained the capacity to bind c-Src. Moreover, transformation by these EGFR mutants was inhibited by Src inhibitors regardless of their sensitivities to gefitinib and erlotinib. These observations have important implications for understanding the molecular basis for resistance to EGFR inhibitors and implicate c-Src as a critical signaling molecule in EGFR mutant-induced transformation.


Asunto(s)
Dominio Catalítico , Transformación Celular Neoplásica/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/química , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptor ErbB-2/metabolismo , Animales , Biotina/metabolismo , Células Cultivadas , Endocitosis , Receptores ErbB/metabolismo , Fibroblastos/enzimología , Humanos , Neoplasias Pulmonares/enzimología , Ratones , Mutación/genética , Unión Proteica , Conformación Proteica , Ratas , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
17.
Cancer Res ; 69(12): 5057-64, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19491266

RESUMEN

The phosphatidylinositol 3-kinase (PI3K)/Akt survival pathway is often dysregulated in cancer. Our previous studies have shown that coexpression of activated Akt1 with activated ErbB2 or polyoma virus middle T antigen uncoupled from the PI3K pathway (PyVmT Y315/322F) accelerates mammary tumor development but cannot rescue the metastatic phenotype associated with these models. Here, we report the generation of transgenic mice expressing activated Akt2 in the mammary epithelium. Like the mouse mammary tumor virus-Akt1 strain, mammary-specific expression of Akt2 delayed mammary gland involution. However, in contrast to Akt1, coexpression of Akt2 with activated ErbB2 or PyVmT Y315/322F in the mammary glands of transgenic mice did not affect the latency of tumor development. Strikingly, Akt2 coexpresssion markedly increased the incidence of pulmonary metastases in both tumor models, demonstrating a unique role in tumor progression. Together, these observations argue that these highly conserved kinases have distinct biological and biochemical outputs that play opposing roles in mammary tumor induction and metastasis.


Asunto(s)
Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Progresión de la Enfermedad , Inmunohistoquímica , Neoplasias Mamarias Experimentales/enzimología , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Cell Biochem ; 96(5): 1071-85, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16167342

RESUMEN

c-Myc transcriptional activity in cells is dampened by the Mad family of transcriptional repressors. The expression of one member, hMad4, is increased in growth-arrested states such as quiescence or replicative senescence; hMad4 mRNA levels in replicatively senescent fibroblasts are about twice those seen in their young contact-inhibited quiescent counterparts. Moreover, the repression of hMad4 transcription following serum stimulation observed in quiescent young fibroblasts is lost in senescent cells. This loss results in persistent expression of hMad4, which leads to an inability to switch from an hMad4/Max complex to a c-Myc/Max complex on selected c-Myc target genes following serum stimulation. We have located an initiator element (Inr), a candidate for Miz-1 binding, in the hMad4 promoter. In reporter assays, Miz-1 enhances reporter GFP expression; this enhancement is inhibited by co-expressing c-Myc. Thus hMad4, as does its murine counterpart, contains the Inr element through which Miz-1 activates its expression; but this action is inhibited in the presence of c-Myc. This inhibition may explain the down-regulation of hMad4, corresponding to the up-regulation of c-Myc, in young serum-starved quiescent fibroblasts upon serum stimulation. However, this reciprocal change does not occur in replicatively senescent fibroblasts upon serum stimulation; instead, hMad4 persists in the presence of high levels of c-Myc activation. Our results suggest that: (1) replicative senescence-specific factors may block c-Myc inhibition of Miz-1 activation of hMad4 expression; and (2) the continual presence of hMad4 protein may transcriptionally repress selected c-Myc target genes, whose functions are key to the signaling pathways leading to apoptosis inhibition and permanent exit of cell cycle traverse in normal human fibroblasts.


Asunto(s)
Fibroblastos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Northern Blotting , Línea Celular , Proliferación Celular , Separación Celular , Senescencia Celular , Inmunoprecipitación de Cromatina , Medio de Cultivo Libre de Suero/farmacología , Cartilla de ADN/química , ADN Complementario/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Factor 4E Eucariótico de Iniciación/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Timosina/análogos & derivados , Timosina/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética
19.
J Cell Biochem ; 89(3): 576-88, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12761891

RESUMEN

Mad family proteins have an antagonistic action on Myc-dependent cell proliferation and transformation. We isolated a human cDNA clone, human Mad4 (hMad4), encoding a polypeptide of 209 amino acid residues, exhibiting 90% identity with mouse Mad4. Northern blot analysis shows that hMad4 probe hybridizes to a 3.8 kb message; its expression is highest in quiescent human WI38 fibroblasts. Among tissues, hMad4 mRNA is most abundant in brain, lung, and muscle. Consistent with other members of the Mad family, hMad4 can repress the transactivation activity of Myc/Max heterodimers on an E-box chloramphenicol acteyl transferase (CAT) reporter plasmid; inhibition of both proliferation and clonogenic formation of hMad4-infected cells correlates with the in vitro reporter repression. Moreover, infection of young human fibroblasts induces a replicative senescence-like state. This phenotype was accompanied by s-beta-galactosidase and PAI-1 expression. These results suggest that hMad4 might be an important regulator of replicative senescence in human cells.


Asunto(s)
División Celular/fisiología , Senescencia Celular/fisiología , Proteínas de Unión al ADN/fisiología , Transactivadores/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , ADN Complementario , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Proteína Smad4 , Transactivadores/genética , Transactivadores/metabolismo
20.
J Biol Chem ; 277(7): 5418-25, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11724805

RESUMEN

Peptide elongation factor eEF1A-2/S1, which shares 92% homology with eEF1A-1/EF-1alpha, is exclusively expressed in brain, heart, and skeletal muscle. In these tissues, eEF1A-2/S1 is the only type 1A elongation factor expressed in adulthood because a transition from eEF1A-1/EF-1alpha to eEF1A-2/S1 occurs in early postnatal development. In this article, we report that the expression of eEF1A-2/S1 protein is activated upon myogenic differentiation. Furthermore, we show that upon serum deprivation-induced apoptosis, eEF1A-2/S1 protein disappears and is replaced by its homolog eEF1A-1/EF-1alpha in dying myotubes; cell death is characterized by the activation of caspase-3. In addition, we show that the continuous expression of eEF1A-2/S1 resulting from adenoviral gene transfer protects differentiated myotubes from apoptosis by delaying their death, thus suggesting a prosurvival function for eEF1A-2/S1 in skeletal muscle. In contrast, myotube death is accelerated by the introduction of the homologous gene, eEF1A-1/EF-1alpha, whereas cells transfected with antisense eEF1A-1/EF-1alpha are protected from apoptosis. These results demonstrate that the two sister genes, eEF1A-1/EF-1alpha and eEF1A-2/S1, regulate myotube survival with the former exerting prodeath activity and the latter a prosurvival effect.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Músculo Esquelético/citología , Factor 1 de Elongación Peptídica/biosíntesis , Factor 1 de Elongación Peptídica/química , Factores de Elongación de Péptidos/biosíntesis , Adenoviridae/genética , Animales , Western Blotting , Caspasa 3 , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , ADN/metabolismo , Fragmentación del ADN , Etiquetado Corte-Fin in Situ , Ratones , Músculo Esquelético/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Oligonucleótidos Antisentido/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factores de Tiempo , Distribución Tisular , Transfección , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA