Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Appetite ; 146: 104509, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31698015

RESUMEN

BACKGROUND: Maternal feeding styles have been associated with children's eating behaviors and obesity risk. Few works have identified maternal feeding styles using a multi-method person-centered approach. OBJECTIVES: (1) To identify maternal feeding styles using a person-centered multi-method approach, and (2) to examine the association of child weight status with maternal feeding styles. METHODS: Participants were low-income mother-child dyads (N = 255) (mean child age 5.9 years) from the United States. Mothers completed questionnaires and participated in a semi-structured interview. Interview transcripts were reliably coded for constructs of child feeding including beliefs, goals, and concerns. Family mealtime video recordings were reliably coded for feeding behaviors. Child anthropometrics were measured. Latent class analysis (LCA) was used to determine empirically-driven typologies of maternal feeding styles. Chi-square analyses tested the association of maternal feeding styles with child overweight or obese (vs. not) weight status. RESULTS: Two maternal feeding styles were identified by LCA which we term "High Coercive Control" (27% child overweight/obese) and "Low Coercive Control" (55% child overweight/obese). High Coercive Control mothers were more likely to believe their child was too thin, self-reported being more demanding in feeding and pressuring the child to eat, worried more about their child not eating enough and were observed to use more bribery. Low Coercive Control mothers were concerned about their child eating too much, and were less likely to self-report engaging in pressuring or restricting feeding behaviors. CONCLUSIONS: The findings suggest that although there is a "feeding style" characterized by substantial control, this style was most common among mothers of thinner children. The mothers of children with overweight/obesity were primarily characterized by engaging in the "recommended" feeding behaviors and being appropriately concerned about their child's risk for excess weight.


Asunto(s)
Conducta Infantil/psicología , Conducta Alimentaria/psicología , Comidas/psicología , Responsabilidad Parental/psicología , Pobreza/psicología , Adulto , Antropometría , Peso Corporal , Preescolar , Coerción , Femenino , Humanos , Análisis de Clases Latentes , Masculino , Relaciones Madre-Hijo , Madres , Obesidad Infantil/psicología , Proyectos de Investigación , Factores de Riesgo , Encuestas y Cuestionarios , Estados Unidos
2.
Int J Radiat Oncol Biol Phys ; 118(5): 1315-1327, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38104870

RESUMEN

PURPOSE: Despite aggressive multimodal treatment that typically includes definitive or adjuvant radiation therapy (RT), locoregional recurrence rates approach 50% for patients with locally advanced human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC). Thus, more effective therapeutics are needed to improve patient outcomes. We evaluated the radiosensitizing effects of ataxia telangiectasia and RAD3-related (ATR) inhibitor (ATRi) BAY 1895344 in preclinical models of HNSCC. METHODS AND MATERIALS: Murine and human HPV-negative HNSCC cells (MOC2, MOC1, JHU-012) were treated with vehicle or ATRi with or without 4 Gy. Checkpoint kinase 1 phosphorylation and DNA damage (γH2AX) were evaluated by Western blot, and ATRi half-maximal inhibitory concentration was determined by MTT assay for HNSCC cells and immortalized murine oral keratinocytes. In vitro radiosensitization was tested by clonogenic assay. Cell cycle distribution and mitotic catastrophe were evaluated by flow cytometry. Mitotic aberrations were quantified by fluorescent microscopy. Tumor growth delay and survival were assessed in mice bearing MOC2 or JHU-012 transplant tumors treated with vehicle, ATRi, RT (10 Gy × 1 or 8 Gy × 3), or combined ATRi + RT. RESULTS: ATRi caused dose-dependent reduction in checkpoint kinase 1 phosphorylation at 1 hour post-RT (4 Gy) and dose-dependent increase in γH2AX at 18 hours post-RT. Addition of RT to ATRi led to decreased BAY 1895344 half-maximal inhibitory concentration in HNSCC cell lines but not in normal tissue surrogate immortalized murine oral keratinocytes. Clonogenic assays demonstrated radiosensitization in the HNSCC cell lines. ATRi abrogated the RT-induced G2/M checkpoint, leading to mitosis with unrepaired DNA damage and increased mitotic aberrations (multinucleated cells, micronuclei, nuclear buds, nucleoplasmic bridges). ATRi and RT significantly delayed tumor growth in MOC2 and JHU-012 in vivo models, with improved overall survival in the MOC2 model. CONCLUSIONS: These findings demonstrated that BAY 1895344 increased in vitro and in vivo radiosensitivity in HPV-negative HNSCC preclinical models, suggesting therapeutic potential warranting evaluation in clinical trials for patients with locally advanced or recurrent HPV-negative HNSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Morfolinas , Infecciones por Papillomavirus , Pirazoles , Fármacos Sensibilizantes a Radiaciones , Humanos , Animales , Ratones , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Recurrencia Local de Neoplasia/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/farmacología , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/radioterapia , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo
3.
Nat Commun ; 15(1): 3018, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38589357

RESUMEN

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced acute GI syndrome. Through single-cell RNA-sequencing of the irradiated mouse small intestine, we find that p53 target genes are specifically enriched in regenerating epithelial cells that undergo fetal-like reversion, including revival stem cells (revSCs) that promote animal survival after severe damage of the GI tract. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce fetal-like revSCs. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells and is controlled by an Mdm2-mediated negative feedback loop. Together, our findings reveal that p53 suppresses severe radiation-induced GI injury by promoting fetal-like reprogramming of irradiated intestinal epithelial cells.


Asunto(s)
Traumatismos por Radiación , Proteína p53 Supresora de Tumor , Ratones , Animales , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Intestinos , Tracto Gastrointestinal/metabolismo , Traumatismos por Radiación/genética , Traumatismos por Radiación/metabolismo , Células Madre/metabolismo , Apoptosis/genética
4.
Methods Cell Biol ; 180: 147-175, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37890927

RESUMEN

Given the potential risk of radiological terrorism and disasters, it is essential to develop plans to prepare for such events. In these hazardous scenarios, radiation-induced gastrointestinal (GI) syndrome is one of the many manifestations that may happen after the organism is exposed to a lethal dose of ionizing radiation. Therefore, it is critical to better understand how the intestinal tissues initiate and orchestrate regeneration following severe radiation injury. In this chapter, we aimed to provide several key considerations for researchers who utilize histological assessment to study radiation-induced intestinal injury. Rigor and reproducibility are critical in experimental design and can be achieved by maintaining proper radiation administration, maintaining consistency in sample collection, and selecting and using appropriate controls. We also provided technical details of histological preparation of the intestines with tips on dissecting, cleaning, fixing, and preserving. Step-by-step descriptions of both bundling and Swiss rolling are provided with discussion on how to choose between the two approaches. In the following section, we detailed several histological assessment methods and then provided suggestions on how to use histological assessment to study cellular dynamics in the small intestines. Finally, we touched on some non-histological assessments. We hope that the information provided in this chapter will contribute to the research society of radiation-induced intestinal injury with an ultimate goal of promoting the development of radiation countermeasures against the GI acute radiation syndrome.


Asunto(s)
Intestino Delgado , Intestinos , Reproducibilidad de los Resultados , Intestinos/patología , Radiación Ionizante
5.
bioRxiv ; 2023 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-37066384

RESUMEN

The adaptive immune system plays an essential anti-tumor role through immunosurveillance and response to immunotherapies. Characterizing phenotypic features and mechanisms of dysfunction of tumor-specific T cell populations may uncover novel immunotherapeutic targets and biomarkers of response. To study tumor-specific T cell responses in vivo, a tumor model must express a known neoantigen. While transplant models with known neoantigen expression are widely available, autochthonous tumor models in which the tumor coevolves with the immune system are limited. In this study, we combined CRISPR/Cas9 and sleeping beauty transposase technology to develop an autochthonous orthotopic murine sarcoma model with oncogenic KrasG12D, functionally impaired p53, and expression of known MHCI and MHCII sarcoma neoantigens. Using MHC tetramer flow cytometry, we identified a tumor-specific immune response in the peripheral blood as early as 10 days after tumor induction leading to tumor clearance. Tumors developed at high penetrance after co-depletion of CD8 and CD4 T cells, but depletion of either CD8 or CD4 T cells alone was insufficient to permit tumor growth. These results suggest that CD8 and CD4 T cells can independently contribute to immunosurveillance leading to clearance of sarcomas expressing MHCI and MHCII neoantigens.

6.
bioRxiv ; 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37162959

RESUMEN

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced GI injury. Through single-cell RNA-sequencing of the irradiated mouse intestine, we find that p53 target genes are specifically enriched in stem cells of the regenerating epithelium, including revival stem cells that promote animal survival after GI damage. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce revival stem cells. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells that is controlled by an Mdm2-mediated negative feedback loop. These results suggest that p53 suppresses severe radiation-indued GI injury by promoting intestinal epithelial cell reprogramming. One-Sentence Summary: After severe radiation injury to the intestine, transient p53 activity induces revival stem cells to promote regeneration.

7.
Cancer Res Commun ; 3(12): 2455-2467, 2023 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-37982576

RESUMEN

Approximately half of patients with cancer receive radiotherapy and, as cancer survivorship increases, the low rate of radiation-associated sarcomas is rising. Pharmacologic inhibition of p53 has been proposed as an approach to ameliorate acute injury of normal tissues from genotoxic therapies, but how this might impact the risk of therapy-induced cancer and normal tissue injuries remains unclear. We utilized mice that express a doxycycline (dox)-inducible p53 short hairpin RNA to reduce Trp53 expression temporarily during irradiation. Mice were placed on a dox diet 10 days prior to receiving 30 or 40 Gy hind limb irradiation in a single fraction and then returned to normal chow. Mice were examined weekly for sarcoma development and scored for radiation-induced normal tissue injuries. Radiation-induced sarcomas were subjected to RNA sequencing. Following single high-dose irradiation, 21% of animals with temporary p53 knockdown during irradiation developed a sarcoma in the radiation field compared with 2% of control animals. Following high-dose irradiation, p53 knockdown preserves muscle stem cells, and increases sarcoma development. Mice with severe acute radiation-induced injuries exhibit an increased risk of developing late persistent wounds, which were associated with sarcomagenesis. RNA sequencing revealed radiation-induced sarcomas upregulate genes related to translation, epithelial-mesenchymal transition (EMT), inflammation, and the cell cycle. Comparison of the transcriptomes of human and mouse sarcomas that arose in irradiated tissues revealed regulation of common gene programs, including elevated EMT pathway gene expression. These results suggest that blocking p53 during radiotherapy could minimize acute toxicity while exacerbating late effects including second cancers. SIGNIFICANCE: Strategies to prevent or mitigate acute radiation toxicities include pharmacologic inhibition of p53 and other cell death pathways. Our data show that temporarily reducing p53 during irradiation increases late effects including sarcomagenesis.


Asunto(s)
Traumatismos por Radiación , Sarcoma , Humanos , Ratones , Animales , Proteína p53 Supresora de Tumor/genética , Sarcoma/genética , Ciclo Celular , Daño del ADN
8.
Breast Cancer Res ; 14(3): R95, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22697792

RESUMEN

INTRODUCTION: Progesterone receptors (PR) are emerging as important breast cancer drivers. Phosphorylation events common to breast cancer cells impact PR transcriptional activity, in part by direct phosphorylation. PR-B but not PR-A isoforms are phosphorylated on Ser294 by mitogen activated protein kinase (MAPK) and cyclin dependent kinase 2 (CDK2). Phospho-Ser294 PRs are resistant to ligand-dependent Lys388 SUMOylation (that is, a repressive modification). Antagonism of PR small ubiquitin-like modifier (SUMO)ylation by mitogenic protein kinases suggests a mechanism for derepression (that is, transcriptional activation) of target genes. As a broad range of PR protein expression is observed clinically, a PR gene signature would provide a valuable marker of PR contribution to early breast cancer progression. METHODS: Global gene expression patterns were measured in T47D and MCF-7 breast cancer cells expressing either wild-type (SUMOylation-capable) or K388R (SUMOylation-deficient) PRs and subjected to pathway analysis. Gene sets were validated by RT-qPCR. Recruitment of coregulators and histone methylation levels were determined by chromatin immunoprecipitation. Changes in cell proliferation and survival were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and western blotting. Finally, human breast tumor cohort datasets were probed to identify PR-associated gene signatures; metagene analysis was employed to define survival rates in patients whose tumors express a PR gene signature. RESULTS: 'SUMO-sensitive' PR target genes primarily include genes required for proliferative and pro-survival signaling. DeSUMOylated K388R receptors are preferentially recruited to enhancer regions of derepressed genes (that is, MSX2, RGS2, MAP1A, and PDK4) with the steroid receptor coactivator, CREB-(cAMP-response element-binding protein)-binding protein (CBP), and mixed lineage leukemia 2 (MLL2), a histone methyltransferase mediator of nucleosome remodeling. PR SUMOylation blocks these events, suggesting that SUMO modification of PR prevents interactions with mediators of early chromatin remodeling at 'closed' enhancer regions. SUMO-deficient (phospho-Ser294) PR gene signatures are significantly associated with human epidermal growth factor 2 (ERBB2)-positive luminal breast tumors and predictive of early metastasis and shortened survival. Treatment with antiprogestin or MEK inhibitor abrogated expression of SUMO-sensitive PR target-genes and inhibited proliferation in BT-474 (estrogen receptor (ER)+/PR+/ERBB2+) breast cancer cells. CONCLUSIONS: We conclude that reversible PR SUMOylation/deSUMOylation profoundly alters target gene selection in breast cancer cells. Phosphorylation-induced PR deSUMOylation favors a permissive chromatin environment via recruitment of CBP and MLL2. Patients whose ER+/PR+ tumors are driven by hyperactive (that is, derepressed) phospho-PRs may benefit from endocrine (antiestrogen) therapies that contain an antiprogestin.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Células MCF-7 , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/genética , Fosforilación , Regiones Promotoras Genéticas , Receptor ErbB-2/metabolismo , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/antagonistas & inhibidores , Sumoilación , Activación Transcripcional , Transcriptoma
9.
Proc Natl Acad Sci U S A ; 106(34): 14287-92, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19706513

RESUMEN

In advanced breast tumors, protein kinases are upregulated and steroid hormone receptors often function independently of ligand. Herein, we explored mechanisms of ligand-independent progesterone receptor (PR) activity. We showed previously that growth factor-induced phosphorylation of PR Ser-294 blocks PR Lys-388 sumoylation. SUMO-deficient mutant PR-B (K388R) thus provides a model receptor for the study of PR function in the context of high kinase activities. T47D cells stably expressing K388R PR-B exhibited increased ligand-independent proliferation and growth in soft agar relative to cells expressing wt PR-B or phospho-mutant (sumoylated) S294A PR-B. Expression of selected PR target genes (HB-EGF, IRS-1, and STC1) was significantly elevated in cells containing desumoylated (K388R) PR-B. Basal PR transcriptional activity occurred independently of progestins, was increased by activated CDK2, and attenuated by RU486. Notably, ChIP assays demonstrated that K388R PR-B and SRC1 were constitutively recruited to the STC1 promoter in the absence of progestin; PR Lys-388 sumoylation was required for HDAC3 recruitment. Knock-down of STC1 inhibited proliferation of cells expressing K388R PR-B. These data suggest a mechanism whereby phosphorylated, and thus desumoylated, PRs mediate increased expression of growth promoting genes. Our data explain why breast cancer models often remain insensitive to progestins, but are growth-inhibited by antiprogestins, and underscore the need to target PR-B and associated kinase activities as part of breast cancer therapy.


Asunto(s)
Mutación , Proteínas Quinasas/metabolismo , Receptores de Progesterona/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células HeLa , Humanos , Immunoblotting , Factor I del Crecimiento Similar a la Insulina/farmacología , Ligandos , Fosforilación , Progestinas/farmacología , Regiones Promotoras Genéticas/genética , Receptores de Progesterona/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/efectos de los fármacos
10.
Radiat Res ; 196(6): 686-689, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34644390

RESUMEN

Exposure to high dose radiation causes life-threatening acute and delayed effects. Defining the mechanisms of lethal radiation-induced acute toxicity of gastrointestinal and hematopoietic tissues are critical steps to identify drug targets to mitigate and protect against the acute radiation syndrome (ARS). For example, one rational approach would be to design pharmaceuticals that block cell death pathways to preserve tissue integrity in radiation-sensitive organ systems including the gastrointestinal tract and hematopoietic compartment. A previous study reported that the inflammasome pathway, which mediates inflammatory cell death through pyroptosis, promotes ARS. However, we show that mice lacking the inflammatory executioner caspases, caspase-1 and caspase-11, are not protected from ARS when compared directly to littermates expressing caspase-1 and caspase-11. These results suggest that alternative pathways will need to be targeted by drugs that successfully mitigate and protect against the ARS.


Asunto(s)
Síndrome de Radiación Aguda/enzimología , Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Inflamasomas/metabolismo , Animales , Sistema Hematopoyético/efectos de la radiación , Inflamasomas/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Piroptosis/efectos de la radiación
11.
Radiother Oncol ; 157: 155-162, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33545252

RESUMEN

BACKGROUND AND PURPOSE: Late cardiac toxicity is a major side effect of radiation therapy (RT) for breast cancer. We developed and characterized a mouse model of radiation-induced heart disease that mimics the dose, fractionation, and beam arrangement of left breast and chest wall RT. MATERIAL AND METHODS: Female wild-type (C57BL6/J) and atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice (on a C57BL/6J background) on regular chow were treated with 2 Gy × 25 fractions of partial-heart irradiation via opposed tangential beams to the left chest wall. The changes in myocardial perfusion and cardiac function of C57BL/6J mice were examined by single-photon emission computed tomography (SPECT) and echocardiography, respectively. In addition to SPECT and echocardiography, the formation of calcified plaques and changes in cardiac function of ApoE-/- mice were examined by dual-energy microCT (DE-CT) and pressure-volume (PV) loop analysis, respectively. The development of myocardial fibrosis was examined by histopathology. RESULTS: Compared to unirradiated controls, irradiated C57BL/6J mice showed no significant changes by SPECT or echocardiography up to 18 months after 2 Gy × 25 partial-heart irradiation even though irradiated mice exhibited a modest increase in myocardial fibrosis. For ApoE-/- mice, 2 Gy × 25 partial-heart irradiation did not cause significant changes by SPECT, DE-CT, or echocardiography. However, PV loop analysis revealed a significant decrease in load-dependent systolic and diastolic function measures including cardiac output, dV/dtmax and dV/dt min 12 months after RT. CONCLUSIONS: Following clinically relevant doses of partial-heart irradiation in C57BL/6J and ApoE-/- mice, assessment with noninvasive imaging modalities such as echocardiography, SPECT, and DE-CT yielded no evidence of decreased myocardial perfusion and cardiac dysfunction related to RT. However, invasive hemodynamic assessment with PV loop analysis indicated subtle, but significant, changes in cardiac function of irradiated ApoE-/- mice. PV loop analysis may be useful for future preclinical studies of radiation-induced heart disease, especially if subtle changes in cardiac function are expected.


Asunto(s)
Corazón , Tomografía Computarizada de Emisión de Fotón Único , Animales , Fraccionamiento de la Dosis de Radiación , Ecocardiografía , Femenino , Corazón/diagnóstico por imagen , Ratones , Ratones Endogámicos C57BL
12.
Cancer Res ; 81(19): 4939-4948, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34385184

RESUMEN

Chromosomal translocations generate oncogenic fusion proteins in approximately one-third of sarcomas, but how these proteins promote tumorigenesis is not well understood. Interestingly, some translocation-driven cancers exhibit dramatic clinical responses to therapy, such as radiotherapy, although the precise mechanism has not been elucidated. Here we reveal a molecular mechanism by which the fusion oncoprotein FUS-CHOP promotes tumor maintenance that also explains the remarkable sensitivity of myxoid liposarcomas to radiation therapy. FUS-CHOP interacted with chromatin remodeling complexes to regulate sarcoma cell proliferation. One of these chromatin remodelers, SNF2H, colocalized with FUS-CHOP genome-wide at active enhancers. Following ionizing radiation, DNA damage response kinases phosphorylated the prion-like domain of FUS-CHOP to impede these protein-protein interactions, which are required for transformation. Therefore, the DNA damage response after irradiation disrupted oncogenic targeting of chromatin remodelers required for FUS-CHOP-driven sarcomagenesis. This mechanism of disruption links phosphorylation of the prion-like domain of an oncogenic fusion protein to DNA damage after ionizing radiation and reveals that a dependence on oncogenic chromatin remodeling underlies sensitivity to radiation therapy in myxoid liposarcoma. SIGNIFICANCE: Prion-like domains, which are frequently translocated in cancers as oncogenic fusion proteins that drive global epigenetic changes, confer sensitivity to radiation via disruption of oncogenic interactions.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Radiación Ionizante , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Sitios de Unión , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de la radiación , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Fusión Oncogénica/química , Fosforilación/efectos de la radiación , Unión Proteica , Proteína FUS de Unión a ARN/química , Sarcoma/etiología , Sarcoma/metabolismo , Sarcoma/patología , Factor de Transcripción CHOP/química , Translocación Genética
13.
Cancer Res ; 81(14): 3777-3790, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34035082

RESUMEN

Mouse models of radiation-induced thymic lymphoma are widely used to study the development of radiation-induced blood cancers and to gain insights into the biology of human T-cell lymphoblastic leukemia/lymphoma. Here we aimed to identify key oncogenic drivers for the development of radiation-induced thymic lymphoma by performing whole-exome sequencing using tumors and paired normal tissues from mice with and without irradiation. Thymic lymphomas from irradiated wild-type (WT), p53+/-, and KrasLA1 mice were not observed to harbor significantly higher numbers of nonsynonymous somatic mutations compared with thymic lymphomas from unirradiated p53-/- mice. However, distinct patterns of recurrent mutations arose in genes that control the Notch1 signaling pathway based on the mutational status of p53. Preferential activation of Notch1 signaling in p53 WT lymphomas was also observed at the RNA and protein level. Reporter mice for activation of Notch1 signaling revealed that total-body irradiation (TBI) enriched Notch1hi CD44+ thymocytes that could propagate in vivo after thymocyte transplantation. Mechanistically, genetic inhibition of Notch1 signaling in immature thymocytes prevented formation of radiation-induced thymic lymphoma in p53 WT mice. Taken together, these results demonstrate a critical role of activated Notch1 signaling in driving multistep carcinogenesis of thymic lymphoma following TBI in p53 WT mice. SIGNIFICANCE: These findings reveal the mutational landscape and key drivers in murine radiation-induced thymic lymphoma, a classic animal model that has been used to study radiation carcinogenesis for over 70 years.


Asunto(s)
Secuenciación del Exoma/métodos , Linfoma/inducido químicamente , Receptor Notch1/metabolismo , Neoplasias del Timo/inducido químicamente , Proteína p53 Supresora de Tumor/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones
14.
Radiat Res ; 197(3): 0, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34724704

RESUMEN

Delayed radiation myelopathy is a rare, but significant late side effect from radiation therapy that can lead to paralysis. The cellular and molecular mechanisms leading to delayed radiation myelopathy are not completely understood but may be a consequence of damage to oligodendrocyte progenitor cells and vascular endothelial cells. Here, we aimed to determine the contribution of endothelial cell damage to the development of radiation-induced spinal cord injury using a genetically defined mouse model in which endothelial cells are sensitized to radiation due to loss of the tumor suppressor p53. Tie2Cre; p53FL/+ and Tie2Cre; p53FL/- mice, which lack one and both alleles of p53 in endothelial cells, respectively, were treated with focal irradiation that specifically targeted the lumbosacral region of the spinal cord. The development of hindlimb paralysis was followed for up to 18 weeks after either a 26.7 Gy or 28.4 Gy dose of radiation. During 18 weeks of follow-up, 83% and 100% of Tie2Cre; p53FL/- mice developed hindlimb paralysis after 26.7 and 28.4 Gy, respectively. In contrast, during this period only 8% of Tie2Cre; p53FL/+ mice exhibited paralysis after 28.4 Gy. In addition, 8 weeks after 28.4 Gy the irradiated spinal cord from Tie2Cre; p53FL/- mice showed a significantly higher fractional area positive for the neurological injury marker glial fibrillary acidic protein (GFAP) compared with the irradiated spinal cord from Tie2Cre; p53FL/+ mice. Together, our findings show that deletion of p53 in endothelial cells sensitizes mice to the development of delayed radiation myelopathy indicating that endothelial cells are a critical cellular target of radiation that regulates myelopathy.


Asunto(s)
Traumatismos de la Médula Espinal/radioterapia , Animales , Relación Dosis-Respuesta en la Radiación , Células Endoteliales , Femenino , Proteína Ácida Fibrilar de la Glía/efectos de la radiación , Humanos , Masculino , Ratones , Traumatismos Experimentales por Radiación , Radiación Ionizante , Médula Espinal/efectos de los fármacos , Factores de Tiempo , Proteína p53 Supresora de Tumor/efectos de la radiación
15.
Health Phys ; 119(3): 315-321, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32175929

RESUMEN

The Radiation and Nuclear Countermeasures Program at the National Institute of Allergy and Infectious Diseases (NIAID) mandated that medical countermeasures for treating Acute Radiation Syndrome (ARS) must have efficacy when administered at least 24 h after radiation exposure. At this time point, many cells within key target tissues, such as the hematopoietic system and the gastrointestinal (GI) tract, will already be dead. Therefore, drugs that promote the regeneration of surviving cells may improve outcomes. The serine/threonine kinase glycogen synthase kinase-3 (GSK-3) regulates stem and progenitor cell self-renewal and regeneration in the hematopoietic and GI compartments. We tested inhibition of GSK-3ß by SB216763 24 h after total body irradiation (TBI) and sub-total body irradiation (SBI). Here, we show that subcutaneous administration of SB216763 promotes the regeneration of surviving hematopoietic stem/progenitor cells (HSPCs), including myeloid progenitor cells, and improves survival of C57Bl/6 male mice when administered 24 h after TBI. However, these results were not recapitulated in female C57Bl/6 animals, suggesting a sex difference in GSK-3ß signaling in HSPCs. Subcutaneous administration of SB216763 in male mice stimulated activation of Sox2 transcription but failed to induce Sox2 transcription in female C57Bl/6 mice. Using TCF/lef-GFP reporter mice, we examined Wnt signaling in HSPCs of irradiated male and female mice treated with SB216763. GSK-3 inhibition elevated Wnt reporter activity in HSPCs isolated from male but not female mice. SB216763 did not mitigate hematopoietic ARS in males or females of a second strain of wild-type mice, C3H. In addition, administration of SB216763 did not mitigate hematopoietic ARS beyond the currently available standard approved therapy of ciprofloxacin and granulocyte-colony stimulating factor (G-CSF) in male C57Bl/6 mice. Further, SB216763 did not mitigate GI-ARS after SBI in C57Bl/6 male mice. The lack of efficacy in both sexes and multiple strains of mice indicate that SB216763 is not suitable for further drug development as a mitigator of ARS. Our studies demonstrate that activation of Wnt signaling in HSPCs promotes hematopoietic regeneration following radiation exposure, and targeting this pathway downstream of GSK-3ß may mitigate ARS in a sex- and strain-independent manner.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Hematopoyesis/efectos de la radiación , Indoles/uso terapéutico , Maleimidas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Protectores contra Radiación/uso terapéutico , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/enzimología , Médula Ósea/efectos de la radiación , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Hematopoyesis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores Sexuales , Especificidad de la Especie
16.
Science ; 368(6487): 181-186, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32273467

RESUMEN

Embryonic development is a complex process that is unamenable to direct observation. In this study, we implanted a window to the mouse uterus to visualize the developing embryo from embryonic day 9.5 to birth. This removable intravital window allowed manipulation and high-resolution imaging. In live mouse embryos, we observed transient neurotransmission and early vascularization of neural crest cell (NCC)-derived perivascular cells in the brain, autophagy in the retina, viral gene delivery, and chemical diffusion through the placenta. We combined the imaging window with in utero electroporation to label and track cell division and movement within embryos and observed that clusters of mouse NCC-derived cells expanded in interspecies chimeras, whereas adjacent human donor NCC-derived cells shrank. This technique can be combined with various tissue manipulation and microscopy methods to study the processes of development at unprecedented spatiotemporal resolution.


Asunto(s)
Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Desarrollo Embrionario , Microscopía Intravital/métodos , Cresta Neural , Animales , Encéfalo/embriología , Encéfalo/fisiología , División Celular , Movimiento Celular , Quimera/embriología , Quimera/fisiología , Electroporación , Femenino , Técnicas de Transferencia de Gen , Ratones , Ratones Transgénicos , Neovascularización Fisiológica , Cresta Neural/irrigación sanguínea , Cresta Neural/citología , Cresta Neural/embriología , Placenta/fisiología , Embarazo , Retina/embriología , Retina/fisiología , Transmisión Sináptica , Útero
17.
Endocr Relat Cancer ; 16(2): 351-61, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19357196

RESUMEN

Progesterone receptors (PR), members of the nuclear receptor superfamily, function as ligand-activated transcription factors and initiators of c-Src kinase and mitogen-activated protein kinase signaling. Bidirectional cross-talk between PR and mitogenic protein kinases results in changes in PR post-translational modification, leading to alterations in PR transcriptional activity and promoter selectivity. PR-induced rapid activation of cytoplasmic protein kinases insures precise regulatory input to downstream cellular processes that are dependent upon nuclear PR, such as cell-cycle progression, and pro-survival signaling. Here, we review interactions between PR and mitogenic protein kinases and discuss the consequences of specific post-translational modifications on PR action in breast cancer cell-line models.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Quinasas/metabolismo , Receptores de Progesterona/fisiología , Neoplasias de la Mama/patología , Femenino , Humanos , Procesamiento Proteico-Postraduccional , Transducción de Señal
18.
Mol Endocrinol ; 22(4): 823-37, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18202149

RESUMEN

Human progesterone receptors (PR) rapidly activate cytosolic signaling pathways, in addition to their classical function as ligand-activated transcription factors. Using ER+/PR-B+ T47D breast cancer cells, we probed the role of progestin-stimulated rapid PR signaling in the transcriptional regulation of target genes involved in breast cancer cell proliferation. Epidermal growth factor receptor (EGFR) was rapidly activated after a 10-min treatment with R5020. Progestin induced EGFR-, c-Src-, and MAPK-dependent phosphorylation of PR-B on the MAPK consensus site, Ser345. Ser345-phosphorylated PR-B receptors strongly associated with specificity protein 1 (Sp1) transcription factors to regulate PR cell cycle (p21) and growth-promoting (EGFR) target genes whose promoters lack canonical progesterone response element sequences. Inhibitors of EGFR, c-Src, or MAPK activities blocked PR tethering to Sp1 and progestin-stimulated S-phase entry. Mutant PR-B receptors defective for c-Src binding (mPro) were not phosphorylated on Ser345 in response to progestin and failed to interact with Sp1. Hormone-induced complexes containing Sp1 and wild-type PR-B, but not S345A or mPro PR-B, were recruited to Sp1 sites within the endogenous p21 promoter. Progestin-induced S-phase entry was attenuated in T47D cells containing wild-type PR-B and treated with EGFR, c-Src, or MAPK kinase inhibitors or in T47D cells stably expressing mPro or mutant DNA-binding domain PR-B. In sum, rapid progestin-activated PR signaling leads to PR Ser345 phosphorylation and tethering to Sp1. These events are critical for progestin-stimulated regulation of Sp1 target genes and breast cancer cell proliferation. Our data demonstrate the therapeutic potential for PR-targeted breast cancer treatment by exploiting multiple nodes along the PR signaling pathway, including PR-B, EGFR, c-Src, MAPK, or Sp1.


Asunto(s)
Receptores de Progesterona/metabolismo , Serina/metabolismo , Transducción de Señal/fisiología , Factor de Transcripción Sp1/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Citometría de Flujo , Humanos , Immunoblotting , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación/efectos de los fármacos , Progestinas/farmacología , Promegestona/farmacología , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptores de Progesterona/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Transcripción Sp1/genética , Transcripción Genética
19.
Cancer Res ; 67(9): 4199-209, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483331

RESUMEN

Total tyrosine kinase activity is often elevated in both cytosolic and membrane fractions of malignant breast tissue and correlates with a decrease in disease-free survival. Breast tumor kinase (Brk; protein tyrosine kinase 6) is a soluble tyrosine kinase that was cloned from a metastatic breast tumor and found to be overexpressed in a majority of breast tumors. Herein, we show that Brk is overexpressed in 86% of invasive ductal breast tumors and coexpressed with ErbB family members in breast cancer cell lines. Additionally, the ErbB ligand, heregulin, activates Brk kinase activity. Knockdown of Brk by stable expression of short hairpin RNA (shRNA) in T47D breast cancer cells decreases proliferation and blocks epidermal growth factor (EGF)- and heregulin-induced activation of Rac GTPase, extracellular signal-regulated kinase (ERK) 5, and p38 mitogen-activated protein kinase (MAPK) but not Akt, ERK1/2, or c-Jun NH(2)-terminal kinase. Furthermore, EGF- and heregulin-induced cyclin D1 expression is dependent on p38 signaling and inhibited by Brk shRNA knockdown. The myocyte enhancer factor 2 transcription factor target of p38 MAPK and ERK5 signaling is also sensitive to altered Brk expression. Finally, heregulin-induced migration of T47D cells requires p38 MAPK activity and is blocked by Brk knockdown. These results place Brk in a novel signaling pathway downstream of ErbB receptors and upstream of Rac, p38 MAPK, and ERK5 and establish the ErbB-Brk-Rac-p38 MAPK pathway as a critical mediator of breast cancer cell migration.


Asunto(s)
Neoplasias de la Mama/enzimología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Neurregulina-1/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Procesos de Crecimiento Celular/fisiología , Movimiento Celular/fisiología , Ciclina D1/biosíntesis , Activación Enzimática , Factor de Crecimiento Epidérmico , Receptores ErbB/metabolismo , Humanos , Proteínas de Dominio MADS/metabolismo , Factores de Transcripción MEF2 , Factores Reguladores Miogénicos/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neurregulina-1/antagonistas & inhibidores , Neurregulina-1/metabolismo , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-4 , Transducción de Señal
20.
Radiat Res ; 192(3): 258-266, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31265788

RESUMEN

Exposure of the gastrointestinal (GI) tract to ionizing radiation can cause acute and delayed injury. However, critical cellular targets that regulate the development of radiation-induced GI injury remain incompletely understood. Here, we investigated the role of vascular endothelial cells in controlling acute and delayed GI injury after total-abdominal irradiation (TAI). To address this, we used genetically engineered mice in which endothelial cells are sensitized to radiation due to the deletion of the tumor suppressor p53. Remarkably, we found that VE-cadherin-Cre; p53FL/FL mice, in which both alleles of p53 are deleted in endothelial cells, were not sensitized to the acute GI radiation syndrome, but these mice were highly susceptible to delayed radiation enteropathy. Histological examination indicated that VE-cadherin-Cre; p53FL/FL mice that developed delayed radiation enteropathy had severe vascular injury in the small intestine, which was manifested by hemorrhage, loss of microvessels and tissue hypoxia. In addition, using dual-energy CT imaging, we showed that VE-cadherin-Cre; p53FL/FL mice had a significant increase in vascular permeability of the small intestine in vivo 28 days after TAI. Together, these findings demonstrate that while sensitization of endothelial cells to radiation does not exacerbate the acute GI radiation syndrome, it is sufficient to promote the development of late radiation enteropathy.


Asunto(s)
Células Endoteliales/patología , Células Endoteliales/efectos de la radiación , Intestinos/patología , Intestinos/efectos de la radiación , Tolerancia a Radiación , Animales , Permeabilidad Capilar/efectos de la radiación , Hipoxia de la Célula/efectos de la radiación , Células Endoteliales/metabolismo , Eliminación de Gen , Ratones , Factores de Tiempo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA