Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Oncol ; 11: 769385, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34900719

RESUMEN

Radiation therapy for abdominal tumors is challenging because the small intestine is exquisitely radiosensitive. Unfortunately, there are no FDA-approved therapies to prevent or mitigate GI radiotoxicity. The EGLN protein family are oxygen sensors that regulate cell survival and metabolism through the degradation of hypoxia-inducible factors (HIFs). Our group has previously shown that stabilization of HIF2 through genetic deletion or pharmacologic inhibition of the EGLNs mitigates and protects against GI radiotoxicity in mice by improving intestinal crypt stem cell survival. Here we aimed to elucidate the molecular mechanisms by which HIF2 confers GI radioprotection. We developed duodenal organoids from mice, transiently overexpressed non-degradable HIF2, and performed bulk RNA sequencing. Interestingly, HIF2 upregulated known radiation modulators and genes involved in GI homeostasis, including Wnt5a. Non-canonical Wnt5a signaling has been shown by other groups to improve intestinal crypt regeneration in response to injury. Here we show that HIF2 drives Wnt5a expression in multiple duodenal organoid models. Luciferase reporter assays performed in human cells showed that HIF2 directly activates the WNT5A promoter via a hypoxia response element. We then evaluated crypt regeneration using spheroid formation assays. Duodenal organoids that were pre-treated with recombinant Wnt5a had a higher cryptogenic capacity after irradiation, compared to vehicle-treated organoids. Conversely, we found that Wnt5a knockout decreased the cryptogenic potential of intestinal stem cells following irradiation. Treatment with recombinant Wnt5a prior to irradiation rescued the cryptogenic capacity of Wnt5a knockout organoids, indicating that Wnt5a is necessary and sufficient for duodenal radioprotection. Taken together, our results suggest that HIF2 radioprotects the GI tract by inducing Wnt5a expression.

2.
Cell Rep ; 37(8): 110044, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34818540

RESUMEN

ß-hydroxybutyrate (ß-OHB) is an essential metabolic energy source during fasting and functions as a chromatin regulator by lysine ß-hydroxybutyrylation (Kbhb) modification of the core histones H3 and H4. We report that Kbhb on histone H3 (H3K9bhb) is enriched at proximal promoters of critical gene subsets associated with lipolytic and ketogenic metabolic pathways in small intestine (SI) crypts during fasting. Similar Kbhb enrichment is observed in Lgr5+ stem cell-enriched epithelial spheroids treated with ß-OHB in vitro. Combinatorial chromatin state analysis reveals that H3K9bhb is associated with active chromatin states and that fasting enriches for an H3K9bhb-H3K27ac signature at active metabolic gene promoters and distal enhancer elements. Intestinal knockout of Hmgcs2 results in marked loss of H3K9bhb-associated loci, suggesting that local production of ß-OHB is responsible for chromatin reprogramming within the SI crypt. We conclude that modulation of H3K9bhb in SI crypts is a key gene regulatory event in response to fasting.


Asunto(s)
Ácido 3-Hidroxibutírico/metabolismo , Ayuno/metabolismo , Histonas/metabolismo , Acetilación , Animales , Cromatina/metabolismo , Ayuno/fisiología , Femenino , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Intestino Delgado/metabolismo , Cuerpos Cetónicos/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética
3.
Int J Radiat Oncol Biol Phys ; 105(3): 537-547, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31271824

RESUMEN

PURPOSE: Chemotherapy combined with radiation therapy is the most commonly used approach for treating locally advanced pancreatic cancer. The use of curative doses of radiation in this disease setting is constrained because of the close proximity of the head of the pancreas to the duodenum. The purpose of this study was to determine whether fasting protects the duodenum from high-dose radiation, thereby enabling dose escalation for efficient killing of pancreatic tumor cells. METHODS AND MATERIALS: C57BL/6J mice were either fed or fasted for 24 hours and then exposed to total abdominal radiation at 11.5 Gy. Food intake, body weight, overall health, and survival were monitored. Small intestines were harvested at various time points after radiation, and villi length, crypt depth, and number of crypts per millimeter of intestine were determined. Immunohistochemistry was performed to assess apoptosis and double-strand DNA breaks, and microcolony assays were performed to determine intestinal stem cell regeneration capacity. A syngeneic KPC model of pancreatic cancer was used to determine the effects of fasting on the radiation responses of both pancreatic cancer and host intestinal tissues. RESULTS: We demonstrated that a 24-hour fast in mice improved intestinal stem cell regeneration, as revealed by microcolony assay, and improved host survival of lethal doses of total abdominal irradiation compared with fed controls. Fasting also improved survival of mice with orthotopic pancreatic tumors subjected to lethal abdominal radiation compared with controls with free access to food. Furthermore, fasting did not affect tumor cell killing by radiation therapy and enhanced γ-H2AX staining after radiation therapy, suggesting an additional mild radiosensitizing effect. CONCLUSIONS: These results establish proof of concept for fasting as a dose-escalation strategy, enabling ablative radiation in the treatment of unresectable pancreatic cancer.


Asunto(s)
Duodeno/efectos de la radiación , Ayuno , Tratamientos Conservadores del Órgano , Neoplasias Pancreáticas/radioterapia , Tolerancia a Radiación , Células Madre/efectos de la radiación , Abdomen/efectos de la radiación , Animales , Apoptosis , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Femenino , Histonas/metabolismo , Intestino Delgado/citología , Intestino Delgado/efectos de la radiación , Masculino , Dosis Máxima Tolerada , Ratones , Ratones Endogámicos C57BL , Órganos en Riesgo/efectos de la radiación , Neoplasias Pancreáticas/mortalidad , Prueba de Estudio Conceptual , Traumatismos por Radiación/mortalidad , Traumatismos por Radiación/prevención & control , Dosificación Radioterapéutica , Distribución Aleatoria , Regeneración , Células Madre/fisiología , Factores de Tiempo , Ensayo de Tumor de Célula Madre/métodos
4.
Cancer Res ; 79(9): 2327-2338, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31043430

RESUMEN

When pancreatic cancer cannot be removed surgically, patients frequently experience morbidity and death from progression of their primary tumor. Radiation therapy (RT) cannot yet substitute for an operation because radiation causes fatal bleeding and ulceration of the nearby stomach and intestines before achieving tumor control. There are no FDA-approved medications that prevent or reduce radiation-induced gastrointestinal injury. Here, we overcome this fundamental problem of anatomy and biology with the use of the oral EGLN inhibitor FG-4592, which selectively protects the intestinal tract from radiation toxicity without protecting tumors. A total of 70 KPC mice with autochthonous pancreatic tumors received oral FG-4592 or vehicle control ± ablative RT to a cumulative 75 Gy administered in 15 daily fractions to a limited tumor field. Although ablative RT reduced complications from local tumor progression, fatal gastrointestinal bleeding was observed in 56% of mice that received high-dose RT with vehicle control. However, radiation-induced bleeding was completely ameliorated in mice that received high-dose RT with FG-4592 (0% bleeding, P < 0.0001 compared with vehicle). Furthermore, FG-4592 reduced epithelial apoptosis by half (P = 0.002) and increased intestinal microvessel density by 80% compared with vehicle controls. EGLN inhibition did not stimulate cancer growth, as treatment with FG-4592 alone, or overexpression of HIF2 within KPC tumors independently improved survival. Thus, we provide a proof of concept for the selective protection of the intestinal tract by the EGLN inhibition to enable ablative doses of cytotoxic therapy in unresectable pancreatic cancer by reducing untoward morbidity and death from radiation-induced gastrointestinal bleeding. SIGNIFICANCE: Selective protection of the intestinal tract by EGLN inhibition enables potentially definitive doses of radiation therapy. This might allow radiation to be a surgical surrogate for unresectable pancreatic cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/9/2327/F1.large.jpg.


Asunto(s)
Glicina/análogos & derivados , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Isoquinolinas/farmacología , Neoplasias Pancreáticas/mortalidad , Traumatismos por Radiación/prevención & control , Protectores contra Radiación/farmacología , Radioterapia/mortalidad , Animales , Apoptosis , Femenino , Glicina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/radioterapia , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Traumatismos por Radiación/etiología , Traumatismos por Radiación/mortalidad , Radioterapia/efectos adversos , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/fisiología
5.
Sci Rep ; 9(1): 1949, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30760738

RESUMEN

Unresectable pancreatic cancer is almost universally lethal because chemotherapy and radiation cannot completely stop the growth of the cancer. The major problem with using radiation to approximate surgery in unresectable disease is that the radiation dose required to ablate pancreatic cancer exceeds the tolerance of the nearby duodenum. WR-2721, also known as amifostine, is a well-known radioprotector, but has significant clinical toxicities when given systemically. WR-2721 is a prodrug and is converted to its active metabolite, WR-1065, by alkaline phosphatases in normal tissues. The small intestine is highly enriched in these activating enzymes, and thus we reasoned that oral administration of WR-2721 just before radiation would result in localized production of the radioprotective WR-1065 in the small intestine, providing protective benefits without the significant systemic side effects. Here, we show that oral WR-2721 is as effective as intraperitoneal WR-2721 in promoting survival of intestinal crypt clonogens after morbid irradiation. Furthermore, oral WR-2721 confers full radioprotection and survival after lethal upper abdominal irradiation of 12.5 Gy × 5 fractions (total of 62.5 Gy, EQD2 = 140.6 Gy). This radioprotection enables ablative radiation therapy in a mouse model of pancreatic cancer and nearly triples the median survival compared to controls. We find that the efficacy of oral WR-2721 stems from its selective accumulation in the intestine, but not in tumors or other normal tissues, as determined by in vivo mass spectrometry analysis. Thus, we demonstrate that oral WR-2721 is a well-tolerated, and quantitatively selective, radioprotector of the intestinal tract that is capable of enabling clinically relevant ablative doses of radiation to the upper abdomen without unacceptable gastrointestinal toxicity.


Asunto(s)
Amifostina/farmacología , Mercaptoetilaminas/farmacología , Protectores contra Radiación/uso terapéutico , Administración Oral , Amifostina/metabolismo , Animales , Femenino , Intestino Delgado/efectos de los fármacos , Masculino , Mercaptoetilaminas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/tratamiento farmacológico , Dosis de Radiación , Protección Radiológica/métodos , Neoplasias Pancreáticas
6.
Sci Rep ; 8(1): 15410, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337664

RESUMEN

Radiation therapy is one of the main treatment options for many cancer patients. Although high doses of radiation may maximize tumor cell killing, dose escalation is limited by toxicity to neighboring normal tissues. This limitation applies particularly to the small intestine, the second most radiosensitive organ in the body. Identifying small intestinal (SI) radioprotectors could enable dose escalation in the treatment of abdominopelvic malignancies. However, the only assay currently available to identify effects of radiomodulating drugs on the regenerating capacity of SI stem cells is the Withers-Elkind microcolony assay, which requires large numbers of mice, making it a costly and low throughput method. Here, we describe a novel spheroid formation assay (SFA) that utilizes SI stem cell-enriched three-dimensional epithelial spheroid cultures to identify gastrointestinal radiomodulators ex vivo. The SFA is scalable for high throughput screening and can be used to identify both radioprotectors and radiosensitizers.


Asunto(s)
Ensayo de Unidades Formadoras de Colonias/métodos , Intestino Delgado/citología , Fármacos Sensibilizantes a Radiaciones/aislamiento & purificación , Esferoides Celulares/citología , Células Madre/citología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Técnicas de Cocultivo , Femenino , Intestino Delgado/efectos de los fármacos , Intestino Delgado/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Neoplasias/radioterapia , Traumatismos por Radiación/prevención & control , Protección Radiológica/métodos , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/efectos de la radiación , Células Madre/efectos de los fármacos , Células Madre/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA