Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
PLoS One ; 18(9): e0292423, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37769019

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0050281.].

2.
J Pathol ; 260(3): 289-303, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37186300

RESUMEN

Breast cancer invasion and metastasis result from a complex interplay between tumor cells and the tumor microenvironment (TME). Key oncogenic changes in the TME include aberrant synthesis, processing, and signaling of hyaluronan (HA). Hyaluronan-mediated motility receptor (RHAMM, CD168; HMMR) is an HA receptor enabling tumor cells to sense and respond to this aberrant TME during breast cancer progression. Previous studies have associated RHAMM expression with breast tumor progression; however, cause and effect mechanisms are incompletely established. Focused gene expression analysis of an internal breast cancer patient cohort confirmed that increased RHAMM expression correlates with aggressive clinicopathological features. To probe mechanisms, we developed a novel 27-gene RHAMM-related signature (RRS) by intersecting differentially expressed genes in lymph node (LN)-positive patient cases with the transcriptome of a RHAMM-dependent model of cell transformation, which we validated in an independent cohort. We demonstrate that the RRS predicts for poor survival and is enriched for cell cycle and TME-interaction pathways. Further analyses using CRISPR/Cas9-generated RHAMM-/- breast cancer cells provided direct evidence that RHAMM promotes invasion in vitro and in vivo. Immunohistochemistry studies highlighted heterogeneous RHAMM protein expression, and spatial transcriptomics associated the RRS with RHAMM-high microanatomic foci. We conclude that RHAMM upregulation leads to the formation of 'invasive niches', which are enriched in RRS-related pathways that drive invasion and could be targeted to limit invasive progression and improve patient outcomes. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/patología , Ácido Hialurónico/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Receptores de Hialuranos/metabolismo , Microambiente Tumoral
3.
JCO Precis Oncol ; 6: e2100463, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35476550

RESUMEN

PURPOSE: The 80-gene molecular subtyping signature (80-GS) reclassifies a proportion of immunohistochemistry (IHC)-defined luminal breast cancers (estrogen receptor-positive [ER+], human epidermal growth factor receptor 2-negative [HER2-]) as Basal-Type. We report the association of 80-GS reclassification with neoadjuvant treatment response and 5-year outcome in patients with breast cancer. METHODS: Neoadjuvant Breast Registry Symphony Trial (NBRST; NCT01479101) is an observational, prospective study that included 1,069 patients with early-stage breast cancer age 18-90 years who received neoadjuvant therapy. Pathologic complete response (pCR) and 5-year distant metastasis-free survival (DMFS) and overall survival (OS) were assessed in 477 patients with IHC-defined ER+, HER2- tumors and in a reference group of 229 patients with IHC-defined triple-negative breast cancer (TNBC). RESULTS: 80-GS reclassified 15% of ER+, HER2- tumors (n = 73) as Basal-Type (ER+/Basal), which had similar pCR compared with TNBC/Basal tumors (34% v 38%; P = .52), and significantly higher pCR than ER+/Luminal A (2%; P < .001) and ER+/Luminal B (6%; P < .001) tumors. The 5-year DMFS (%, [95% CI]) was significantly lower for patients with ER+/Basal tumors (66% [52.6 to 77.3]), compared with those with ER+/Luminal A tumors (92.3% [85.2 to 96.1]) and ER+/Luminal B tumors (73.5% [44.5 to 79.3]). Importantly, patients with ER+/Basal or TNBC/Basal tumors that had a pCR exhibited significantly improved DMFS and OS compared with those with residual disease. By contrast, patients with ER+/Luminal B tumors had comparable 5-year DMFS and OS whether or not they achieved pCR. CONCLUSION: Significant differences in chemosensitivity and 5-year outcome suggest patients with ER+/Basal molecular subtype may benefit from neoadjuvant regimens optimized for patients with TNBC/Basal tumors compared with patients with ER+/Luminal subtype. These data highlight the importance of identifying this subset of patients to improve treatment planning and long-term survival.


Asunto(s)
Terapia Neoadyuvante , Neoplasias de la Mama Triple Negativas , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Humanos , Persona de Mediana Edad , Estudios Prospectivos , Receptor ErbB-2 , Receptores de Estrógenos/genética , Receptores de Progesterona/análisis , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adulto Joven
4.
Oncotarget ; 11(3): 304, 2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-32076492

RESUMEN

[This corrects the article DOI: 10.18632/oncotarget.2500.].

6.
Support Care Cancer ; 27(4): 1365-1373, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30112722

RESUMEN

SIGNIFICANCE: Poor sleep quality is a common and persistent problem reported by women with breast cancer (BC). Empirical evidence identifies many risk factors for self-reported sleep deficiency, but inconsistencies limit translation to practice. PURPOSE: To increase understanding of risk factors predicting self-reported poor sleep quality in women with BC who completed the Breast Cancer Collaborative Registry (BCCR) questionnaire. METHODS: This cross-sectional study recruited women with a first diagnosis of BC (n = 1302) at five sites in Nebraska and South Dakota. Women completed the BCCR that includes numerous variables as well as the Pittsburgh Sleep Quality Index (PSQI) and SF36v2 (n = 1260). Descriptive statistics and non-parametric correlations were used to determine associations and create predictive models of sleep quality with BCCR variables and SF36v2 subscales. RESULTS: Most women were white (93.7%) and married (71.5%); mean age was 60.1 (21-90) years. Poor sleep was self-reported by 53% of women. Seven variables were highly associated with sleep quality (p ≤ 0.001). The first model found younger age, lower physical activity, and higher fatigue were the strongest combined and independent variables predicting poor sleep quality (F = 23.0 (p < .001), R2 = 0.103). Participants self-reported lower health status on most SF36v2 subscales [Z = 44.9 (11.6) to 49.1 (10.1)]. A second model found that all subscales were predictors of poor sleep; vitality, mental health, bodily pain, and general health were the strongest predictors (F = 101.3 (p < .001), R2 = 0.26). CONCLUSIONS: Results confirm previously identified risk factors and reveal inconsistencies in other variables. Clinicians need to routinely screen for the identified risk factors of self-reported poor sleep quality.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Dolor en Cáncer/complicaciones , Dolor en Cáncer/fisiopatología , Sistema de Registros , Trastornos del Sueño-Vigilia/etiología , Trastornos del Sueño-Vigilia/fisiopatología , Sueño/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/psicología , Dolor en Cáncer/psicología , Estudios Transversales , Fatiga/etiología , Fatiga/psicología , Femenino , Humanos , Persona de Mediana Edad , Calidad de Vida , Autoinforme , Trastornos del Sueño-Vigilia/psicología , Encuestas y Cuestionarios , Adulto Joven
8.
J Natl Compr Canc Netw ; 13(7): 880-915, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26150582

RESUMEN

Breast cancer is the most frequently diagnosed malignancy in women in the United States and is second only to lung cancer as a cause of cancer death. To assist women who are at increased risk of developing breast cancer and their physicians in the application of individualized strategies to reduce breast cancer risk, NCCN has developed these guidelines for breast cancer risk reduction.


Asunto(s)
Neoplasias de la Mama/prevención & control , Conducta de Reducción del Riesgo , Femenino , Humanos , Factores de Riesgo
9.
Breast Cancer Res Treat ; 151(1): 219-24, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25833210

RESUMEN

Partner and localizer of BRCA2 (PALB2), plays an important functional role in DNA damage repair. Recent studies indicate that germline mutations in PALB2 predispose individuals to a high risk of developing familial breast cancer. Therefore, comprehensive identification of PALB2 germline mutations is potentially important for understanding their roles in tumorigenesis and for testing their potential utility as clinical targets. Most of the previous studies of PALB2 have focused on familial breast cancer cases with normal/wild-type BRCA1 and BRCA2 (BRCAx). We hypothesize that PALB2 genetic mutations also exist in individuals with BRCA mutations (BRCA+). To test this hypothesis, PALB2 germline mutations were screened in 107 exome data sets collected from familial breast cancer families who were either BRCA1+ or BRCAx. Two novel heterozygous mutations predicted to alter the function of PALB2 were identified (c.2014G>C, p.E672Q and c.2993G>A, p.G998E). Notably, both of these mutations co-existed in BRCA1+ and BRCA1x families. These studies show that mutations in PALB2 can occur independent of the status of BRCA1 mutations, and they highlight the importance to include BRCA1+ families in PALB2 mutation screens.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Masculino , Linaje
10.
PLoS One ; 10(3): e0119383, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25803781

RESUMEN

Breast cancers exhibit highly heterogeneous molecular profiles. Although gene expression profiles have been used to predict the risks and prognostic outcomes of breast cancers, the high variability of gene expression limits its clinical application. In contrast, genetic mutation profiles would be more advantageous than gene expression profiles because genetic mutations can be stably detected and the mutational heterogeneity widely exists in breast cancer genomes. We analyzed 98 breast cancer whole exome samples that were sorted into three subtypes, two grades and two stages. The sum deleterious effect of all mutations in each gene was scored to identify differentially mutated genes (DMGs) for this case-control study. DMGs were corroborated using extensive published knowledge. Functional consequences of deleterious SNVs on protein structure and function were also investigated. Genes such as ERBB2, ESP8, PPP2R4, KIAA0922, SP4, CENPJ, PRCP and SELP that have been experimentally or clinically verified to be tightly associated with breast cancer prognosis are among the DMGs identified in this study. We also identified some genes such as ARL6IP5, RAET1E, and ANO7 that could be crucial for breast cancer development and prognosis. Further, SNVs such as rs1058808, rs2480452, rs61751507, rs79167802, rs11540666, and rs2229437 that potentially influence protein functions are observed at significantly different frequencies in different comparison groups. Protein structure modeling revealed that many non-synonymous SNVs have a deleterious effect on protein stability, structure and function. Mutational profiling at gene- and SNV-level revealed differential patterns within each breast cancer comparison group, and the gene signatures correlate with expected prognostic characteristics of breast cancer classes. Some of the genes and SNVs identified in this study show high promise and are worthy of further investigation by experimental studies.


Asunto(s)
Neoplasias de la Mama/genética , Exoma , Mutación , Adulto , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Femenino , Estudios de Asociación Genética , Humanos , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Análisis de Secuencia de ADN , Transcriptoma
11.
Oncotarget ; 5(21): 10251-70, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25344910

RESUMEN

Radiation therapy is a staple treatment for pancreatic cancer. However, owing to the intrinsic radioresistance of pancreatic cancer cells, radiation therapy often fails to increase survival of pancreatic cancer patients. Radiation impedes cancer cells by inducing DNA damage, which can activate cell cycle checkpoints. Normal cells possess both a G1 and G2 checkpoint. However, cancer cells are often defective in G1 checkpoint due to mutations/alterations in key regulators of this checkpoint. Accordingly, our results show that normal pancreatic ductal cells respond to ionizing radiation (IR) with activation of both checkpoints whereas pancreatic cancer cells respond to IR with G2/M arrest only. Overexpression/hyperactivation of Rac1 GTPase is detected in the majority of pancreatic cancers. Rac1 plays important roles in survival and Ras-mediated transformation. Here, we show that Rac1 also plays a critical role in the response of pancreatic cancer cells to IR. Inhibition of Rac1 using specific inhibitor and dominant negative Rac1 mutant not only abrogates IR-induced G2 checkpoint activation, but also increases radiosensitivity of pancreatic cancer cells through induction of apoptosis. These results implicate Rac1 signaling in the survival of pancreatic cancer cells following IR, raising the possibility that this pathway contributes to the intrinsic radioresistance of pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/radioterapia , Rayos gamma/uso terapéutico , Neoplasias Pancreáticas/radioterapia , Proteína de Unión al GTP rac1/metabolismo , Aminoquinolinas/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína Quinasa CDC2 , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Mutación/genética , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Pirimidinas/farmacología , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
12.
BMC Cancer ; 14: 470, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24969172

RESUMEN

BACKGROUND: Genetic predisposition is the primary risk factor for familial breast cancer. For the majority of familial breast cancer, however, the genetic predispositions remain unknown. All newly identified predispositions occur rarely in disease population, and the unknown genetic predispositions are estimated to reach up to total thousands. Family unit is the basic structure of genetics. Because it is an autosomal dominant disease, individuals with a history of familial breast cancer must carry the same genetic predisposition across generations. Therefore, focusing on the cases in lineages of familial breast cancer, rather than pooled cases in disease population, is expected to provide high probability to identify the genetic predisposition for each family. METHODS: In this study, we tested genetic predispositions by analyzing the family-specific variants in familial breast cancer. Using exome sequencing, we analyzed three families and 22 probands with BRCAx (BRCA-negative) familial breast cancer. RESULTS: We observed the presence of family-specific, novel, deleterious germline variants in each family. Of the germline variants identified, many were shared between the disease-affected family members of the same family but not found in different families, which have their own specific variants. Certain variants are putative deleterious genetic predispositions damaging functionally important genes involved in DNA replication and damaging repair, tumor suppression, signal transduction, and phosphorylation. CONCLUSIONS: Our study demonstrates that the predispositions for many BRCAx familial breast cancer families can lie in each disease family. The application of a family-focused approach has the potential to detect many new predispositions.


Asunto(s)
Genes BRCA1 , Genes BRCA2 , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Exoma , Femenino , Humanos , Modelos Biológicos , Linaje
13.
BMC Cancer ; 14: 342, 2014 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-24884718

RESUMEN

BACKGROUND: BRCA1 plays an essential role in maintaining genome stability. Inherited BRCA1 germline mutation (BRCA1+) is a determined genetic predisposition leading to high risk of breast cancer. While BRCA1+ induces breast cancer by causing genome instability, most of the knowledge is known about somatic genome instability in breast cancer cells but not germline genome instability. METHODS: Using the exome-sequencing method, we analyzed the genomes of blood cells in a typical BRCA1+ breast cancer family with an exon 13-duplicated founder mutation, including six breast cancer-affected and two breast cancer unaffected members. RESULTS: We identified 23 deleterious mutations in the breast cancer-affected family members, which are absent in the unaffected members. Multiple mutations damaged functionally important and breast cancer-related genes, including transcriptional factor BPTF and FOXP1, ubiquitin ligase CUL4B, phosphorylase kinase PHKG2, and nuclear receptor activator SRA1. Analysis of the mutations between the mothers and daughters shows that most mutations were germline mutation inherited from the ancestor(s) while only a few were somatic mutation generated de novo. CONCLUSION: Our study indicates that BRCA1+ can cause genome instability with both germline and somatic mutations in non-breast cells.


Asunto(s)
Proteína BRCA1/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Inestabilidad Genómica , Adulto , Proteína BRCA1/sangre , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/sangre , Análisis Mutacional de ADN , Exones , Femenino , Efecto Fundador , Predisposición Genética a la Enfermedad , Herencia , Humanos , Masculino , Persona de Mediana Edad , Madres , Mutación , Núcleo Familiar , Linaje , Fenotipo
14.
Breast J ; 19(5): 520-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23800003

RESUMEN

Genetic predisposition plays a key role in the development of familial breast cancer. In spite of strong familial clustering of the disease and extensive efforts made during the past decade; however, progress has been slow in identifying genetic predisposition for the majority of familial breast cancer families. The question arises therefore as to whether current approaches are adequate in identifying the unknown genetic predisposition. We analyzed eight members of a BRCA1-, BRCA2-, p53-, and PTEN-negative breast cancer family, of which five had breast cancer, one is an obligate gene carrier, and two were unaffected. We sequenced the entire coding region of the genome for each member using exome sequencing to identify nonsynonymous variants. We identified 55 nonsynonymous germline variants affecting 49 genes in multiple members of the family, of which 22 are predicted to have damaging effects. We validated 20 of the 22 selected variants in the family by Sanger sequencing. Two variants in KAT6B, an acetal transferase gene, were identified in six family members of which five were affected with breast cancer and one is the unaffected obligate carrier. We further examined the presence of the identified variants in a cohort of 40 additional breast cancer cases from 22 familial breast cancer families, but none of the 22 variants was detected in these cases. Sequencing the entire coding exons in KAT6B detects no variants in these cases. Our results show that genetic predisposition for familial breast cancer can be rich in an affected family, but the predisposition can be family-specific. As such, it will be difficult to detect them by applying population-based approach. Our study supports the concept that focusing on each affected family will be required to determine the genetic predisposition for many familial breast cancer families whose genetic dispositions remain unknown.


Asunto(s)
Neoplasias de la Mama/congénito , Predisposición Genética a la Enfermedad , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/genética , Femenino , Genes BRCA1 , Genes BRCA2 , Histona Acetiltransferasas/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación
15.
PLoS One ; 7(11): e50281, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166842

RESUMEN

Topo II poisons, which target topoisomerase II (topo II) to generate enzyme mediated DNA damage, have been commonly used for anti-cancer treatment. While clinical evidence demonstrate a capability of topo II poisons in inducing apoptosis in cancer cells, accumulating evidence also show that topo II poison treatment frequently results in cell cycle arrest in cancer cells, which was associated with subsequent resistance to these treatments. Results in this report indicate that treatment of MCF-7 and T47D breast cancer cells with topo II poisons resulted in an increased phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2) and an subsequent induction of G2/M cell cycle arrest. Furthermore, inhibition of ERK1/2 activation using specific inhibitors markedly attenuated the topo II poison-induced G2/M arrest and diminished the topo II poison-induced activation of ATR and Chk1 kinases. Moreover, decreased expression of ATR by specific shRNA diminished topo II poison-induced G2/M arrest but had no effect on topo II poison-induced ERK1/2 activation. In contrast, inhibition of ERK1/2 signaling had little, if any, effect on topo II poison-induced ATM activation. In addition, ATM inhibition by either incubation of cells with ATM specific inhibitor or transfection of cells with ATM specific siRNA did not block topo II poison-induced G2/M arrest. Ultimately, inhibition of ERK1/2 signaling greatly enhanced topo II poison-induced apoptosis. These results implicate a critical role for ERK1/2 signaling in the activation of G2/M checkpoint response following topo II poison treatment, which protects cells from topo II poison-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Inhibidores de Topoisomerasa II/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Doxorrubicina/farmacología , Etopósido/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Vectores Genéticos/genética , Humanos , Immunoblotting , Inmunoprecipitación , Indoles , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células MCF-7 , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Retroviridae , Transfección
16.
Breast Cancer Res ; 14(2): R60, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22494620

RESUMEN

INTRODUCTION: In response to gamma-irradiation (IR)-induced double-strand DNA breaks, cells undergo cell-cycle arrest, allowing time for DNA repair before reentering the cell cycle. G2/M checkpoint activation involves activation of ataxia telangiectasia mutated (ATM)/ATM- and rad3-related (ATR) kinases and inhibition of Cdc25 phosphatases, resulting in inhibition of Cdc2 kinase and subsequent G2/M cell-cycle arrest. Previous studies from our laboratory showed that the G2/M checkpoint activation after IR exposure of MCF-7 breast cancer cells is dependent on the activation of extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) signaling. In the present studies, we investigated the role of Ras-related C3 botulinum toxin substrate 1 (Rac1) guanosine triphosphatase (GTPase) in IR-induced G2/M checkpoint response and ERK1/2 activation, as well as in cell survival after IR. METHODS: With Rac1-specific inhibitor, dominant negative mutant Rac1 (N17Rac1) and specific small interfering RNA, the effect of Rac1 on IR-induced G2/M checkpoint response and ERK1/2 activation was examined in human breast cancer cells. In addition, the effect of Rac1 on cell survival after irradiation was assessed by using Rac1-specific inhibitor. RESULTS: IR exposure of MCF-7 breast cancer cells was associated with a marked activation of Rac1 GTPase. Furthermore, inhibition of Rac1 by using specific inhibitor, dominant-negative Rac1 mutant, or specific siRNA resulted in attenuation of IR-induced G2/M arrest and concomitant diminution of IR-induced activation of ATM, ATR, Chk1, and Chk2 kinases, as well as phosphorylation of Cdc2-Tyr15. Moreover, Rac1 inhibition or decreased Rac1 expression also abrogated IR-induced phosphorylation of mitogen-activated protein kinase kinase 1 and 2 (MEK1/2) and ERK1/2. Ultimately, inhibition of Rac1 markedly increased cellular sensitivity to IR exposure, which involves induction of apoptosis. CONCLUSION: Studies in this report suggest that Rac1 GTPase plays an essential role in the activation of IR-induced ERK1/2 signaling and subsequent G2/M checkpoint response. Furthermore, results also support a role for Rac1 in promoting cell survival after irradiation treatment.


Asunto(s)
Neoplasias de la Mama/radioterapia , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Puntos de Control de la Fase M del Ciclo Celular/efectos de la radiación , Proteína de Unión al GTP rac1/metabolismo , Aminoquinolinas/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína Quinasa CDC2 , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Femenino , Rayos gamma , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/farmacología , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Tirosina/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
18.
J Biol Chem ; 284(11): 6705-15, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19147494

RESUMEN

KSR1 (kinase suppressor of Ras 1) is a molecular scaffold and positive regulator of the Raf/MEK/ERK phosphorylation cascade. KSR1 is required for maximal ERK activation induced by growth factors and by some cytotoxic agents. We show here that KSR1 is also required for maximal ERK activation induced by UV light, ionizing radiation, or the DNA interstrand cross-linking agent mitomycin C (MMC). We further demonstrate a role for KSR1 in the reinitiation of the cell cycle and proliferation following cell cycle arrest induced by MMC. Cells lacking KSR1 underwent but did not recover from MMC-induced G(2)/M arrest. Expression of KSR1 allowed KSR1(-/-) cells to re-enter the cell cycle following MMC treatment. However, cells expressing a mutated form of KSR1 unable to bind ERK did not recover from MMC-induced cell cycle arrest, demonstrating the requirement for the KSR1-ERK interaction. In addition, constitutive activation of ERK was not sufficient to promote cell cycle reinitiation in MMC-treated KSR1(-/-) cells. Only cells expressing KSR1 recovered from MMC-induced cell cycle arrest. Importantly, MMC-induced DNA damage was repaired in KSR1(-/-) cells, as determined by resolution of gamma-H2AX-containing foci. These data indicate that cell cycle reinitiation is not actively signaled in the absence of KSR1, even when DNA damage has been resolved. These data reveal a specific role for the molecular scaffold KSR1 and KSR1-mediated ERK signaling in the cellular response to DNA interstrand cross-links.


Asunto(s)
Ciclo Celular/fisiología , Daño del ADN/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Noqueados , Mitomicina/farmacología , Mutación , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Quinasas/genética
19.
Cancer Chemother Pharmacol ; 63(5): 807-18, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18661133

RESUMEN

Previous studies have indicated that the ERK1/2 MAP kinase signaling pathway plays an important role not only in cell growth, cell cycle regulation, and differentiation, but also in determining the sensitivity of cells to anticancer agents as well. Furthermore, expression of kinase suppressor of Ras-1 (KSR1), a molecular scaffold that modulates signaling through the ERK1/2 MAP kinase pathway, has been shown to influence the cellular sensitivity to the anticancer agent cisplatin. To further define the role of KSR1 expression on drug sensitivity, the expression of KSR1 was examined in the NCI60 anticancer drug screen, a panel of cancer cell lines representing nine tissue types, established by the Developmental Therapeutics Program (DTP) at the National Cancer Institute (NCI). The expression of thousands of molecular targets has been examined in the NCI60 panel as well as the cellular toxicity for greater than 400,000 compounds. KSR1 expression varied almost 30-fold difference between the highest and lowest expressing cell lines in the NCI60. Using the COMPARE analysis algorithm, KSR1 expression was correlated with sensitivity of the compounds screened by DTP and several novel agents were identified whose sensitivity correlated with KSR1 expression in the NCI60 panel. Cytotoxicity of two agents, cytochalasin H and tunicamycin, identified through the COMPARE analysis of KSR1 expression and drug sensitivity, was also examined in wild type (KSR(+/+)) mouse embryo fibroblasts (MEFs) and MEFs deficient in KSR1 expression (KSR1(-/-)). These studies demonstrated enhanced sensitivity, as well as increased ERK activation, in KSR(-/-) MEFs following exposure to tunicamycin or cytochalasin H compared to KSR(+/+) MEFs. Furthermore, restoration of KSR1 expression in KSR(-/-) MEFs following stable transduction of cells with a KSR1 expression vector, enhanced sensitivity of cells to tunicamycin and cytochalasin H and decreased ERK1/2 activation following exposure to these drugs. In addition, the sensitivity to cytochalasin H and tunicamycin of breast cancer cell lines with low KSR1 expression, (HS578T and MDA-MB-231/ATCC), was increased relative to the sensitivity of breast cancer cells with higher levels of KSR1 (MCF7). These studies indicate that KSR1 may play an important role in the determination of cellular sensitivity to anticancer agents.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Quinasas/fisiología , Animales , Antivirales/farmacología , Western Blotting , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Células Cultivadas , Citocalasinas/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/enzimología , Activación Enzimática/efectos de los fármacos , Femenino , Fibroblastos/citología , Fibroblastos/enzimología , Humanos , Ratones , Ratones Noqueados , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Tunicamicina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...