Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 148(1-2): 349-61, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22265420

RESUMEN

Metastasis is the leading cause of cancer-associated death but has been difficult to study because it involves a series of rare, stochastic events. To capture these events, we developed a sensitive method to tag and track pancreatic epithelial cells in a mouse model of pancreatic cancer. Tagged cells invaded and entered the bloodstream unexpectedly early, before frank malignancy could be detected by rigorous histologic analysis; this behavior was widely associated with epithelial-to-mesenchymal transition (EMT). Circulating pancreatic cells maintained a mesenchymal phenotype, exhibited stem cell properties, and seeded the liver. EMT and invasiveness were most abundant at inflammatory foci, and induction of pancreatitis increased the number of circulating pancreatic cells. Conversely, treatment with the immunosuppressive agent dexamethasone abolished dissemination. These results provide insight into the earliest events of cellular invasion in situ and suggest that inflammation enhances cancer progression in part by facilitating EMT and entry into the circulation.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Transición Epitelial-Mesenquimal , Invasividad Neoplásica , Neoplasias Pancreáticas/patología , Animales , Carcinoma Ductal Pancreático/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/inmunología , Pancreatitis/patología
2.
Genes Dev ; 33(11-12): 641-655, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31048544

RESUMEN

Pancreatic adenocarcinoma (PDA) is an aggressive disease driven by oncogenic KRAS and characterized by late diagnosis and therapeutic resistance. Here we show that deletion of the ataxia-telangiectasia group D-complementing (Atdc) gene, whose human homolog is up-regulated in the majority of pancreatic adenocarcinoma, completely prevents PDA development in the context of oncogenic KRAS. ATDC is required for KRAS-driven acinar-ductal metaplasia (ADM) and its progression to pancreatic intraepithelial neoplasia (PanIN). As a result, mice lacking ATDC are protected from developing PDA. Mechanistically, we show ATDC promotes ADM progression to PanIN through activation of ß-catenin signaling and subsequent SOX9 up-regulation. These results provide new insight into PDA initiation and reveal ATDC as a potential target for preventing early tumor-initiating events.


Asunto(s)
Carcinogénesis , Carcinoma Ductal Pancreático/fisiopatología , Neoplasias Pancreáticas/fisiopatología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción/fisiología , Células Acinares/metabolismo , Células Acinares/patología , Animales , Carcinoma in Situ/patología , Carcinoma in Situ/fisiopatología , Carcinoma Ductal Pancreático/patología , Transdiferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Humanos , Metaplasia , Ratones , Ratones Transgénicos , Conductos Pancreáticos/metabolismo , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta Catenina/metabolismo
3.
Development ; 150(2)2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36458554

RESUMEN

Adenosine deaminase acting on RNA 1 (ADAR1) is an RNA-binding protein that deaminates adenosine (A) to inosine (I). A-to-I editing alters post-transcriptional RNA processing, making ADAR1 a crucial regulator of gene expression. Consequently, Adar1 has been implicated in organogenesis. To determine the role of Adar1 in pancreatic development and homeostasis, we conditionally deleted Adar1 from the murine pancreas (Ptf1aCre/+; Adar1Fl/Fl). The resulting mice had stunted growth, likely due to malabsorption associated with exocrine pancreatic insufficiency. Analyses of pancreata revealed ductal cell expansion, heightened interferon-stimulated gene expression and an increased influx of immune cells. Concurrent deletion of Adar1 and Mavs, a signaling protein implicated in the innate immune pathway, rescued the degenerative phenotype and resulted in normal pancreatic development. Taken together, our work suggests that the primary function of Adar1 in the pancreas is to prevent aberrant activation of the Mavs-mediated innate immune pathway, thereby maintaining pancreatic homeostasis.


Asunto(s)
Páncreas Exocrino , Animales , Ratones , Páncreas Exocrino/metabolismo , Interferones/genética , Interferones/metabolismo , Fenotipo , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo
4.
Genes Dev ; 29(2): 171-83, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25593307

RESUMEN

The initiation of pancreatic ductal adenocarcinoma (PDA) is linked to activating mutations in KRAS. However, in PDA mouse models, expression of oncogenic mutant KRAS during development gives rise to tumors only after a prolonged latency or following induction of pancreatitis. Here we describe a novel mouse model expressing ataxia telangiectasia group D complementing gene (ATDC, also known as TRIM29 [tripartite motif 29]) that, in the presence of oncogenic KRAS, accelerates pancreatic intraepithelial neoplasia (PanIN) formation and the development of invasive and metastatic cancers. We found that ATDC up-regulates CD44 in mouse and human PanIN lesions via activation of ß-catenin signaling, leading to the induction of an epithelial-to-mesenchymal transition (EMT) phenotype characterized by expression of Zeb1 and Snail1. We show that ATDC is up-regulated by oncogenic Kras in a subset of PanIN cells that are capable of invading the surrounding stroma. These results delineate a novel molecular pathway for EMT in pancreatic tumorigenesis, showing that ATDC is a proximal regulator of EMT.


Asunto(s)
Carcinoma Ductal Pancreático/fisiopatología , Neoplasias Pancreáticas/fisiopatología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción/metabolismo , Animales , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Transgénicos , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/enzimología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , beta Catenina/metabolismo
5.
Genes Dev ; 27(3): 288-300, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23355395

RESUMEN

Pancreatic exocrine cell plasticity can be observed during development, pancreatitis with subsequent regeneration, and also transformation. For example, acinar-ductal metaplasia (ADM) occurs during acute pancreatitis and might be viewed as a prelude to pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDAC) development. To elucidate regulatory processes that overlap ductal development, ADM, and the progression of normal cells to PanIN lesions, we undertook a systematic approach to identify the Prrx1 paired homeodomain Prrx1 transcriptional factor as a highly regulated gene in these processes. Prrx1 annotates a subset of pancreatic ductal epithelial cells in Prrx1creER(T2)-IRES-GFP mice. Furthermore, sorted Prrx1(+) cells have the capacity to self-renew and expand during chronic pancreatitis. The two isoforms, Prrx1a and Prrx1b, regulate migration and invasion, respectively, in pancreatic cancer cells. In addition, Prrx1b is enriched in circulating pancreatic cells (Pdx1cre;LSL-Kras(G12D/+);p53(fl/+);R26YFP). Intriguingly, the Prrx1b isoform, which is also induced in ADM, binds the Sox9 promoter and positively regulates Sox9 expression. This suggests a new hierarchical scheme whereby a Prrx1-Sox9 axis may influence the emergence of acinar-ductal metaplasia and regeneration. Furthermore, our data provide a possible explanation of why pancreatic cancer is skewed toward a ductal fate.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Páncreas/patología , Páncreas/fisiología , Neoplasias Pancreáticas/fisiopatología , Regeneración/fisiología , Animales , Línea Celular Tumoral , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Páncreas/citología , Regiones Promotoras Genéticas/genética , Unión Proteica , Isoformas de Proteínas/metabolismo , Factor de Transcripción SOX9/genética
6.
Gastroenterology ; 154(5): 1509-1523.e5, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29273451

RESUMEN

BACKGROUND & AIMS: Intraductal papillary mucinous neoplasias (IPMNs) are precancerous cystic lesions that can develop into pancreatic ductal adenocarcinomas (PDACs). These large macroscopic lesions are frequently detected during medical imaging, but it is unclear how they form or progress to PDAC. We aimed to identify cells that form IPMNs and mutations that promote IPMN development and progression. METHODS: We generated mice with disruption of Pten specifically in ductal cells (Sox9CreERT2;Ptenflox/flox;R26RYFP or PtenΔDuct/ΔDuct mice) and used PtenΔDuct/+ and Pten+/+ mice as controls. We also generated KrasG12D;PtenΔDuct/ΔDuct and KrasG12D;PtenΔDuct/+ mice. Pancreata were collected when mice were 28 weeks to 14.5 months old and analyzed by histology, immunohistochemistry, and electron microscopy. We performed multiplexed droplet digital polymerase chain reaction to detect spontaneous Kras mutations in PtenΔDuct/ΔDuct mice and study the effects of Ras pathway activation on initiation and progression of IPMNs. We obtained 2 pancreatic sections from a patient with an invasive pancreatobiliary IPMN and analyzed the regions with and without the invasive IPMN (control tissue) by immunohistochemistry. RESULTS: Mice with ductal cell-specific disruption of Pten but not control mice developed sporadic, macroscopic, intraductal papillary lesions with histologic and molecular features of human IPMNs. PtenΔDuct/ΔDuct mice developed IPMNs of several subtypes. In PtenΔDuct/ΔDuct mice, 31.5% of IPMNs became invasive; invasion was associated with spontaneous mutations in Kras. KrasG12D;PtenΔDuct/ΔDuct mice all developed invasive IPMNs within 1 month. In KrasG12D;PtenΔDuct/+ mice, 70% developed IPMN, predominately of the pancreatobiliary subtype, and 63.3% developed PDAC. In all models, IPMNs and PDAC expressed the duct-specific lineage tracing marker yellow fluorescent protein. In immunohistochemical analyses, we found that the invasive human pancreatobiliary IPMN tissue had lower levels of PTEN and increased levels of phosphorylated (activated) ERK compared with healthy pancreatic tissue. CONCLUSIONS: In analyses of mice with ductal cell-specific disruption of Pten, with or without activated Kras, we found evidence for a ductal cell origin of IPMNs. We also showed that PTEN loss and activated Kras have synergistic effects in promoting development of IPMN and progression to PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/enzimología , Transformación Celular Neoplásica/metabolismo , Neoplasias Quísticas, Mucinosas y Serosas/enzimología , Fosfohidrolasa PTEN/deficiencia , Conductos Pancreáticos/enzimología , Neoplasias Pancreáticas/enzimología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Invasividad Neoplásica , Neoplasias Quísticas, Mucinosas y Serosas/genética , Neoplasias Quísticas, Mucinosas y Serosas/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Factores de Tiempo
7.
Proc Natl Acad Sci U S A ; 113(11): 3078-83, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26929329

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by an exuberant inflammatory desmoplastic response. The PDAC microenvironment is complex, containing both pro- and antitumorigenic elements, and remains to be fully characterized. Here, we show that sensory neurons, an under-studied cohort of the pancreas tumor stroma, play a significant role in the initiation and progression of the early stages of PDAC. Using a well-established autochthonous model of PDAC (PKC), we show that inflammation and neuronal damage in the peripheral and central nervous system (CNS) occurs as early as the pancreatic intraepithelial neoplasia (PanIN) 2 stage. Also at the PanIN2 stage, pancreas acinar-derived cells frequently invade along sensory neurons into the spinal cord and migrate caudally to the lower thoracic and upper lumbar regions. Sensory neuron ablation by neonatal capsaicin injection prevented perineural invasion (PNI), astrocyte activation, and neuronal damage, suggesting that sensory neurons convey inflammatory signals from Kras-induced pancreatic neoplasia to the CNS. Neuron ablation in PKC mice also significantly delayed PanIN formation and ultimately prolonged survival compared with vehicle-treated controls (median survival, 7.8 vs. 4.5 mo; P = 0.001). These data establish a reciprocal signaling loop between the pancreas and nervous system, including the CNS, that supports inflammation associated with oncogenic Kras-induced neoplasia. Thus, pancreatic sensory neurons comprise an important stromal cell population that supports the initiation and progression of PDAC and may represent a potential target for prevention in high-risk populations.


Asunto(s)
Capsaicina/uso terapéutico , Carcinoma Ductal Pancreático/prevención & control , Desnervación , Páncreas/inervación , Neoplasias Pancreáticas/prevención & control , Células Receptoras Sensoriales/fisiología , Adenocarcinoma in Situ/patología , Adenocarcinoma in Situ/fisiopatología , Vías Aferentes , Animales , Animales Recién Nacidos , Capsaicina/administración & dosificación , Capsaicina/farmacología , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/fisiopatología , Carcinoma Ductal Pancreático/terapia , Ceruletida/toxicidad , Progresión de la Enfermedad , Femenino , Ganglios Simpáticos/fisiopatología , Genes ras , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mielitis/complicaciones , Mielitis/genética , Mielitis/fisiopatología , Invasividad Neoplásica , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/fisiopatología , Neoplasias Pancreáticas/terapia , Pancreatitis/inducido químicamente , Pancreatitis/complicaciones , Pancreatitis/fisiopatología , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/complicaciones , Lesiones Precancerosas/fisiopatología , Células Receptoras Sensoriales/efectos de los fármacos , Médula Espinal/fisiopatología , Tractos Espinotalámicos/fisiopatología , Vértebras Torácicas
8.
Gastroenterology ; 152(4): 840-850.e3, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27923728

RESUMEN

BACKGROUND & AIMS: Approximately 50% of all patients with pancreatic ductal adenocarcinoma (PDA) develop diabetes mellitus before their cancer diagnosis. Screening individuals with new-onset diabetes might allow earlier diagnosis of PDA. We sought to develop and validate a PDA risk prediction model to identify high-risk individuals among those with new-onset diabetes. METHODS: We conducted a retrospective cohort study in a population representative database from the United Kingdom. Individuals with incident diabetes after the age of 35 years and 3 or more years of follow-up after diagnosis of diabetes were eligible for inclusion. Candidate predictors consisted of epidemiologic and clinical characteristics available at the time of diabetes diagnosis. Variables with P values <.25 in the univariable analyses were evaluated using backward stepwise approach. Model discrimination was assessed using receiver operating characteristic curve analysis. Calibration was evaluated using the Hosmer-Lemeshow test. Results were internally validated using a bootstrapping procedure. RESULTS: We analyzed data from 109,385 patients with new-onset diabetes. Among them, 390 (0.4%) were diagnosed with PDA within 3 years. The final model (area under the curve, 0.82; 95% confidence interval, 0.75-0.89) included age, body mass index, change in body mass index, smoking, use of proton pump inhibitors, and anti-diabetic medications, as well as levels of hemoglobin A1C, cholesterol, hemoglobin, creatinine, and alkaline phosphatase. Bootstrapping validation showed negligible optimism. If the predicted risk threshold for definitive PDA screening was set at 1% over 3 years, only 6.19% of the new-onset diabetes population would undergo definitive screening, which would identify patients with PDA with 44.7% sensitivity, 94.0% specificity, and a positive predictive value of 2.6%. CONCLUSIONS: We developed a risk model based on widely available clinical parameters to help identify patients with new-onset diabetes who might benefit from PDA screening.


Asunto(s)
Carcinoma Ductal Pancreático/epidemiología , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/epidemiología , Modelos Teóricos , Neoplasias Pancreáticas/epidemiología , Factores de Edad , Anciano , Fosfatasa Alcalina/sangre , Área Bajo la Curva , Índice de Masa Corporal , Carcinoma Ductal Pancreático/diagnóstico , Colesterol/sangre , Creatinina/sangre , Diabetes Mellitus/sangre , Detección Precoz del Cáncer , Femenino , Hemoglobina Glucada/metabolismo , Hemoglobinas/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/diagnóstico , Valor Predictivo de las Pruebas , Inhibidores de la Bomba de Protones/uso terapéutico , Curva ROC , Estudios Retrospectivos , Medición de Riesgo , Fumar/epidemiología , Reino Unido/epidemiología
10.
Psychooncology ; 27(4): 1221-1228, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29388275

RESUMEN

OBJECTIVE: To examine the dose-response effects of aerobic exercise on health-related quality of life (HRQoL) among colon cancer survivors. METHODS: Thirty-nine stage I to III colon cancer survivors were randomized to 1 of 3 groups: usual-care control, 150 min·wk-1 of aerobic exercise (low-dose) and 300 min·wk-1 of aerobic exercise (high-dose) for 6 months. HRQoL outcomes included the Short Form (SF)-36 physical and mental component summary, Functional Assessment of Cancer Therapy-Colorectal, Pittsburgh Sleep Quality Index, Fear of Cancer Recurrence Inventory, Fatigue Symptom Inventory, and North Central Cancer Treatment Group bowel function questionnaire, assessed at baseline and post intervention. The primary hypothesis was that exercise would improve HRQoL outcomes in a dose-response fashion, such that high-dose aerobic exercise would yield the largest improvements in HRQoL outcomes. RESULTS: Over 6 months, the low-dose group completed 141 ± 10 min·wk-1 of aerobic exercise, and the high-dose group completed 247 ± 11 min·wk-1 of aerobic exercise. Over 6 months, exercise improved the physical component summary score of the SF-36 (Ptrend  = 0.002), the Functional Assessment of Cancer Therapy-Colorectal (Ptrend  = 0.025), the Pittsburgh Sleep Quality Index (Ptrend  = 0.049), and the Fatigue Symptom Inventory (Ptrend  = 0.045) in a dose-response fashion. Between-group standardized mean difference effects sizes for the above-described findings were small to moderate in magnitude (0.35-0.75). No dose-response effects were observed for the mental component summary score of the SF-36, the Fear of Cancer Recurrence Inventory, or bowel function. CONCLUSION: Higher doses of aerobic exercise, up to 300 min·wk-1 , improve multiple HRQoL outcomes among stage I to III colon cancer survivors. These findings provide evidence that aerobic exercise may provide multiple health benefits for colon cancer survivors.


Asunto(s)
Supervivientes de Cáncer/psicología , Neoplasias del Colon/psicología , Terapia por Ejercicio/métodos , Ejercicio Físico/psicología , Calidad de Vida , Neoplasias del Colon/fisiopatología , Neoplasias del Colon/rehabilitación , Ejercicio Físico/fisiología , Fatiga/fisiopatología , Fatiga/psicología , Femenino , Estado de Salud , Humanos , Masculino , Persona de Mediana Edad , Medición de Resultados Informados por el Paciente
11.
Gut ; 66(1): 124-136, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27402485

RESUMEN

BACKGROUND: Pancreatic cancer is characterised by the accumulation of a fibro-inflammatory stroma. Within this stromal reaction, myeloid cells are a predominant population. Distinct myeloid subsets have been correlated with tumour promotion and unmasking of anti-tumour immunity. OBJECTIVE: The goal of this study was to determine the effect of myeloid cell depletion on the onset and progression of pancreatic cancer and to understand the relationship between myeloid cells and T cell-mediated immunity within the pancreatic cancer microenvironment. METHODS: Primary mouse pancreatic cancer cells were transplanted into CD11b-diphtheria toxin receptor (DTR) mice. Alternatively, the iKras* mouse model of pancreatic cancer was crossed into CD11b-DTR mice. CD11b+ cells (mostly myeloid cell population) were depleted by diphtheria toxin treatment during tumour initiation or in established tumours. RESULTS: Depletion of myeloid cells prevented KrasG12D-driven pancreatic cancer initiation. In pre-established tumours, myeloid cell depletion arrested tumour growth and in some cases, induced tumour regressions that were dependent on CD8+ T cells. We found that myeloid cells inhibited CD8+ T-cell anti-tumour activity by inducing the expression of programmed cell death-ligand 1 (PD-L1) in tumour cells in an epidermal growth factor receptor (EGFR)/mitogen-activated protein kinases (MAPK)-dependent manner. CONCLUSION: Our results show that myeloid cells support immune evasion in pancreatic cancer through EGFR/MAPK-dependent regulation of PD-L1 expression on tumour cells. Derailing this crosstalk between myeloid cells and tumour cells is sufficient to restore anti-tumour immunity mediated by CD8+ T cells, a finding with implications for the design of immune therapies for pancreatic cancer.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/inmunología , Células Mieloides/inmunología , Neoplasias Pancreáticas/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral/inmunología , Animales , Antígeno CD11b/análisis , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Receptores ErbB/metabolismo , Humanos , Tolerancia Inmunológica , Inmunidad Celular , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor , Sistema de Señalización de MAP Quinasas , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células Mieloides/química , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Escape del Tumor
12.
Br J Cancer ; 117(11): 1614-1620, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-28934762

RESUMEN

BACKGROUND: Physical activity is associated with a lower risk of disease recurrence among colon cancer survivors. Excess visceral adipose tissue is associated with a higher risk of disease recurrence among colon cancer survivors. The pathways through which physical activity may alter disease outcomes are unknown, but may be mediated by changes in visceral adipose tissue. METHODS: Thirty-nine stage I-III colon cancer survivors were randomised to one of three groups: usual-care control, 150 min wk-1 of aerobic exercise (low dose) and 300 min wk-1 of aerobic exercise (high dose) for 6 months. The prespecified key body composition outcome was visceral adipose tissue quantified using dual energy X-ray absorptiometry. RESULTS: Exercise reduced visceral adipose tissue in dose-response fashion (Ptrend=0.008). Compared with the control group, the low- and high-dose exercise groups lost 9.5 cm2 (95% CI: -22.4, 3.5) and 13.6 cm2 (95% CI: -27.0, -0.1) in visceral adipose tissue, respectively. Each 60 min wk-1 increase in exercise predicted a 2.7 cm2 (95% CI: -5.4, -0.1) reduction in visceral adipose tissue. CONCLUSIONS: Aerobic exercise reduces visceral adipose tissue in dose-response fashion among patients with stage I-III colon cancer. Visceral adipose tissue may be a mechanism through which exercise reduces the risk of disease recurrence among colon cancer survivors.


Asunto(s)
Composición Corporal , Supervivientes de Cáncer , Neoplasias del Colon/metabolismo , Neoplasias del Colon/mortalidad , Ejercicio Físico , Adulto , Anciano , Femenino , Humanos , Grasa Intraabdominal/metabolismo , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/prevención & control
13.
Gastroenterology ; 146(3): 647-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24333829

RESUMEN

Hematogenous dissemination is thought to be a late event in cancer progression. We recently showed in a genetic model of pancreatic ductal adenocarcinoma that pancreas cells can be detected in the bloodstream before tumor formation. To confirm these findings in humans, we used microfluidic geometrically enhanced differential immunocapture to detect circulating pancreas epithelial cells in patient blood samples. We captured more than 3 circulating pancreas epithelial cells/mL in 7 of 21 (33%) patients with cystic lesions and no clinical diagnosis of cancer (Sendai criteria negative), 8 of 11 (73%) with pancreatic ductal adenocarcinoma, and in 0 of 19 patients without cysts or cancer (controls). These findings indicate that cancer cells are present in the circulation of patients before tumors are detected, which might be used in risk assessment.


Asunto(s)
Células Epiteliales/patología , Células Neoplásicas Circulantes/patología , Páncreas/patología , Quiste Pancreático/diagnóstico , Quiste Pancreático/patología , Adenocarcinoma/diagnóstico , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/patología , Estudios de Casos y Controles , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Técnicas Analíticas Microfluídicas , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Medición de Riesgo
14.
Curr Opin Gastroenterol ; 30(5): 506-10, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25023382

RESUMEN

PURPOSE OF REVIEW: Recent advances in sequencing technology have led to a deeper and more comprehensive understanding of the molecular biology of pancreatic ductal adenocarcinoma. This timely review seeks to summarize these recent advances which will provide a foundation for future studies in the field. RECENT FINDINGS: Stereotypical genetic alterations have been identified and confirmed. However, additional alterations have highlighted the importance and complexity of a number of intracellular signaling pathways that present unique opportunities for therapeutic targeting. SUMMARY: A genetic signature of pancreatic ductal adenocarcinoma has been identified. This recent and important work is currently in the process of being applied in many clinical applications from early diagnostics to customized therapeutic regimens for this disease. A fundamental understanding of these findings will thus be of utmost importance for future research in the field and in the clinical care of patients with this lethal disease.


Asunto(s)
Carcinoma Ductal Pancreático , Técnicas Genéticas , Biología Molecular/métodos , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Pancreáticas/metabolismo , Transducción de Señal
15.
Carcinogenesis ; 34(11): 2647-54, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23764754

RESUMEN

Igf2 mRNA binding protein 1 (IMP1, CRD-BP, ZBP-1) is a messenger RNA binding protein that we have shown previously to regulate colorectal cancer (CRC) cell growth in vitro. Furthermore, increased IMP1 expression correlates with enhanced metastasis and poor prognosis in CRC patients. In the current study, we sought to elucidate IMP1-mediated functions in CRC pathogenesis in vivo. Using CRC cell xenografts, we demonstrate that IMP1 overexpression promotes xenograft tumor growth and dissemination into the blood. Furthermore, intestine-specific knockdown of Imp1 dramatically reduces tumor number in the Apc (Min/+) mouse model of intestinal tumorigenesis. In addition, IMP1 knockdown xenografts exhibit a reduced number of tumor cells entering the circulation, suggesting that IMP1 may directly modulate this early metastatic event. We further demonstrate that IMP1 overexpression decreases E-cadherin expression, promotes survival of single tumor cell-derived colonospheres and promotes enrichment and maintenance of a population of CD24+CD44+ cells, signifying that IMP1 overexpressing cells display evidence of loss of epithelial identity and enhancement of a tumor-initiating cell phenotype. Taken together, these findings implicate IMP1 as a modulator of tumor growth and provide evidence for a novel role of IMP1 in early events in CRC metastasis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/patología , Intestinos/patología , Células Madre Neoplásicas/patología , Proteínas de Unión al ARN/fisiología , Animales , Apoptosis , Western Blotting , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/metabolismo , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Transición Epitelial-Mesenquimal , Fibroblastos/metabolismo , Fibroblastos/patología , Xenoinjertos , Humanos , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Células Madre Neoplásicas/metabolismo , Fenotipo , Células Tumorales Cultivadas
16.
Pancreatology ; 13(2): 114-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23561968

RESUMEN

An epithelial-to-mesenchymal transition (EMT) is thought to be an important process in the acquisition of capabilities required for metastasis. Until recently, studies of EMT involved mostly in vitro assays and transplantation experiments of cancer cells that overexpressed known EMT drivers. While valuable, these studies do not allow us to conclude if an EMT sustained under "physiologic conditions" within the tumor microenvironment leads to the myriad changes in phenotype observed in vitro. Here we review our recently published work using a lineage labeled genetically engineered mouse model of pancreatic ductal adenocarcinoma to characterize cells that have sustained an EMT in vivo.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Neoplasias Pancreáticas/patología , Animales , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Páncreas/citología , Páncreas/patología
18.
Cancer Res ; 82(15): 2761-2776, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35666804

RESUMEN

Conventional genetically engineered mouse models (GEMM) are time-consuming, laborious, and offer limited spatiotemporal control. Here, we describe the development of a streamlined platform for in vivo gene activation using CRISPR activation (CRISPRa) technology. Unlike conventional GEMMs, this model system allows for flexible, sustained, and timed activation of one or more target genes using single or pooled lentiviral guides. Myc and Yap1 were used as model oncogenes to demonstrate gene activation in primary pancreatic organoid cultures in vitro and enhanced tumorigenic potential in Myc-activated organoids when transplanted orthotopically in vivo. Implementation of this model as an autochthonous lung cancer model showed that transduction-mediated activation of Myc led to accelerated tumor progression and significantly reduced overall survival relative to nontargeted tumor controls. Furthermore, Myc activation led to the acquisition of an immune suppressive, "cold" tumor microenvironment. Cross-species validation of these results using publicly available RNA/DNA-seq datasets linked MYC to a previously described immunosuppressive molecular subtype in patient tumors, thus identifying a patient cohort that may benefit from combined MYC- and immune-targeted therapies. Overall, this work demonstrates how CRISPRa can be used for rapid functional validation of putative oncogenes and may allow for the identification and evaluation of potential metastatic and oncogenic drivers through competitive screening. SIGNIFICANCE: A streamlined platform for programmable CRISPR gene activation enables rapid evaluation and functional validation of putative oncogenes in vivo.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas c-myc , Adenocarcinoma del Pulmón/genética , Animales , Carcinogénesis/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Oncogenes/genética , Proteínas Proto-Oncogénicas c-myc/genética , Microambiente Tumoral/genética
19.
Eur J Gastroenterol Hepatol ; 34(1): 33-38, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33470698

RESUMEN

BACKGROUND: Early detection of pancreatic ductal adenocarcinoma (PDA) may improve survival. We previously developed a clinical prediction model among patients with new-onset diabetes to help identify PDAs 6 months prior to the clinical diagnosis of the cancer. We developed and internally validated a new model to predict PDA risk among those newly diagnosed with impaired fasting glucose (IFG). METHODS: We conducted a retrospective cohort study in The Health Improvement Network (THIN) (1995-2013) from the UK. Eligible study patients had newly diagnosed IFG during follow-up in THIN. The outcome was incident PDA diagnosed within 3 years of IFG diagnosis. Candidate predictors were factors associated with PDA, glucose metabolism or both. RESULTS: Among the 138 232 eligible patients with initial IFG diagnosis, 245 (0.2%) were diagnosed with PDA within 3 years. The median time from IFG diagnosis to clinical PDA diagnosis was 326 days (IQR 120-588). The final prediction model included age, BMI, proton pump inhibitor use, total cholesterol, low-density lipoprotein, alanine aminotransferase and alkaline phosphatase. The model achieved good discrimination [area under the curve 0.71 (95% CI, 0.67-0.75)] and calibration (Hosmer and Lemeshow goodness-of-fit test P > 0.05 in 17 of the 20 imputed data sets) with optimism of 0.0012662 (95% CI, -0.00932 to 0.0108771). CONCLUSIONS: We developed and internally validated a sequential PDA prediction model based on clinical information routinely available at the initial appearance of IFG. If externally validated, this model could significantly extend our ability to detect PDAs at an earlier stage.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Estado Prediabético , Glucemia , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/epidemiología , Humanos , Modelos Estadísticos , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/epidemiología , Estado Prediabético/diagnóstico , Estado Prediabético/epidemiología , Pronóstico , Estudios Retrospectivos , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA