Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell Immunol ; 397-398: 104814, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38422979

RESUMEN

The aim of this study was to assess the potential use of a selective small molecule MALT1 inhibitor in solid tumor treatment as an immunotherapy targeting regulatory T-cells (Tregs). In vitro, MALT1 inhibition suppressed the proteolytic cleavage of the MALT1-substrate HOIL1 and blocked IL-2 secretion in Jurkat cells. It selectively suppressed the proliferation of PBMC-derived Tregs, with no effect on conventional CD4+T-cells. In vivo, however, no evident anti-tumor effect was achieved by MALT1 inhibition monotherapy or in combination with anti-CTLA4 in the MB49 cancer model. Despite decreased Treg-frequencies in lymph nodes of tumor-bearing animals, intratumoral Treg depletion was not observed. We also showed that MALT1-inhibition caused a reduction of antigen-specific CD8+T-cells in an adoptive T-cell transfer model. Thus, selective targeting of Tregs would be required to improve the immunotherapeutic effect of MALT1-inhibition. Also, various dosing schedules and combination therapy strategies should be carefully designed and evaluated further.


Asunto(s)
Leucocitos Mononucleares , Neoplasias , Animales , Linfocitos T Reguladores , Linfocitos T CD8-positivos , Proteolisis
2.
J Hepatol ; 62(5): 1156-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25445398

RESUMEN

BACKGROUND & AIMS: Low-grade chronic inflammation is a cardinal feature of the metabolic syndrome, yet its pathogenesis is not well defined. The purpose of this study was to examine the role of TRAIL receptor (TR) signaling in the pathogenesis of obesity-associated inflammation using mice with the genetic deletion of TR. METHODS: TR knockout (TR(-/-)) mice and their littermate wild-type (WT) mice were fed a diet high in saturated fat, cholesterol and fructose (FFC) or chow. Metabolic phenotyping, liver injury, and liver and adipose tissue inflammation were assessed. Chemotaxis and activation of mouse bone marrow-derived macrophages (BMDMϕ) was measured. RESULTS: Genetic deletion of TR completely repressed weight gain, adiposity and insulin resistance in FFC-fed mice. Moreover, TR(-/-) mice suppressed steatohepatitis, with essentially normal serum ALT, hepatocyte apoptosis and liver triglyceride accumulation. Gene array data implicated inhibition of macrophage-associated hepatic inflammation in the absence of the TR. In keeping with this, there was diminished accumulation and activation of inflammatory macrophages in liver and adipose tissue. TR(-/-) BMDMϕ manifest reduced chemotaxis and diminished activation of nuclear factor-κ B signaling upon activation by palmitate and lipopolysaccharide. CONCLUSIONS: These data advance the concept that macrophage-associated hepatic and adipose tissue inflammation of nutrient excess requires TR signaling.


Asunto(s)
Tejido Adiposo , Inflamación , Hígado , Macrófagos , Obesidad , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Quimiotaxis , Dieta Alta en Grasa/métodos , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Activación de Macrófagos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal
3.
J Biol Chem ; 286(45): 39336-48, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21941003

RESUMEN

Nonalcoholic steatohepatitis is characterized by hepatic steatosis, elevated levels of circulating free fatty acids (FFA), endoplasmic reticulum (ER) stress, and hepatocyte lipoapoptosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor 5 (DR5) is significantly elevated in patients with nonalcoholic steatohepatitis, and steatotic hepatocytes demonstrate increased sensitivity to TRAIL-mediated cell death. Nonetheless, a role for TRAIL and/or DR5 in mediating lipoapoptotic pathways is unexplored. Here, we examined the contribution of DR5 death signaling to lipoapoptosis by free fatty acids. The toxic saturated free fatty acid palmitate induces an increase in DR5 mRNA and protein expression in Huh-7 human hepatoma cells leading to DR5 localization into lipid rafts, cell surface receptor clustering with subsequent recruitment of the initiator caspase-8, and ultimately cellular demise. Lipoapoptosis by palmitate was not inhibited by a soluble human recombinant DR5-Fc chimera protein suggesting that DR5 cytotoxic signaling is ligand-independent. Hepatocytes from murine TRAIL receptor knock-out mice (DR(-/-)) displayed reduced palmitate-mediated lipotoxicity. Likewise, knockdown of DR5 or caspase-8 expression by shRNA technology attenuated palmitate-induced Bax activation and apoptosis in Huh-7 cells, without altering induction of ER stress markers. Similar observations were verified in other cell models. Finally, knockdown of CHOP, an ER stress-mediated transcription factor, reduced DR5 up-regulation and DR5-mediated caspase-8 activation upon palmitate treatment. Collectively, these results suggest that ER stress-induced CHOP activation by palmitate transcriptionally up-regulates DR5, likely resulting in ligand-independent cytotoxic signaling by this death receptor.


Asunto(s)
Apoptosis , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Caspasa 8/genética , Caspasa 8/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Hígado Graso/genética , Hígado Graso/patología , Técnicas de Silenciamiento del Gen , Hepatocitos/patología , Humanos , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Microdominios de Membrana/patología , Ratones , Ratones Noqueados , Ácido Palmítico/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
4.
Am J Pathol ; 179(5): 2169-76, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21975022

RESUMEN

Bone marrow-derived cells (BMDCs) participate in the growth and spread of tumors of the breast, brain, lung, and stomach. To date, there are limited reports of bone marrow involvement in colon cancer pathogenesis, but such findings would have the potential to generate novel treatments for colon cancer patients. We have established a mouse model for imaging BMDCs from whole tumor to single-cell resolution, whereby the bone marrow of lethally irradiated host animals is reconstituted with EGFP-expressing bone marrow cells from matched TgActb(EGFP) donors. The BM transplants yield mice with fluorescently labeled bone marrow, and so BMDCs can subsequently be monitored within a tumor through optical imaging. Successful BM reconstitution was confirmed at 8 weeks after transplantation, when surviving BALB/c mice were injected with CT26 mouse colon cancer cells. We find that up to 45% of cells dissociated from the tumors are GFP(+) and approximately 50% of Lin(+), CD11b(+), and CD3(+) cells express high levels of GFP. Notably, tumor growth is reduced in BM transplanted animals, compared with untransplanted host mice or EGFP-expressing BM donor mice. A needed next step is to separate the molecular and cellular (eg, T cells, NK cells, macrophages) bases of the antitumor effect of the BMDCs from any protumorigenic effect that could be subverted for therapeutic gain.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea/métodos , Neoplasias del Colon/terapia , Proteínas Fluorescentes Verdes/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Neoplasias del Colon/patología , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Trasplante de Neoplasias/métodos , Trasplante Isogénico/métodos , Microambiente Tumoral , Irradiación Corporal Total
5.
Am J Cancer Res ; 12(5): 2118-2131, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35693092

RESUMEN

Colorectal cancer (CRC) incidence is rising globally. Hence, preventing this disease is a high priority. With this aim, we determined the CRC prevention potential of the TRAIL-inducing small molecule ONC201/TIC10 using a preclinical model representing high-risk familial adenomatous polyposis (FAP) patients, Apc min/+ mice. Prior to the efficacy study, optimal and non-toxic doses of ONC201 were determined by testing five different doses of ONC201 (0-100 mg/kg body weight (BW); twice weekly by oral gavage) in C57BL/6J mice (n=6/group) for 6 weeks. BW gain, organ weights and histopathology, blood profiling, and the plasma liver enzyme profile suggested no toxicities of ONC201 at doses up to 100 mg/kg BW. For efficacy determination, beginning at six weeks of age, groups of Apc min/+ male and female mice (n≥20) treated with colon carcinogen azoxymethane (AOM) (AOM-Apc min/+) were administered ONC201 (0, 25, and 50 mg/kg BW) as above up to 20 weeks of age. At termination, efficacy was determined by comparing the incidence and multiplicity of intestinal tumors between vehicle- and drug-treated groups. ONC201 showed a strong suppressive effect against the development of both large and small intestinal tumors in male and female mice. Apc min/+ mice treated with ONC201 (50 mg/kg BW) showed >50% less colonic tumor incidence (P<0.0002) than controls. Colonic tumor multiplicity was also significantly reduced by 68% in male mice (0.44 ± 0.11 in treated vs. 1.4 ± 0.14 in controls; P<0.0001) and by 75% in female mice (0.30 ± 0.10 in treated vs. 1.19 ± 0.19 in controls; P<0.0003) with ONC201 treatment (50 mg/kg BW). Small intestinal polyps were reduced by 68% in male mice (11.40 ± 1.19 in treated vs. 36.08 ± 2.62 in controls; P<0.0001) and female mice (9.65 ± 1.15 in treated vs. 29.24 ± 2.51 in controls; P<0.0001). Molecular analysis of the tumors suggested an increase in TRAIL, DR5, cleaved caspases 3/7/8, Fas-associated death domain protein (FADD), and p21 (WAF1) in response to drug treatment. Serum analysis indicated a decrease in pro-inflammatory serum biomarkers, such as IL1ß, IL6, TNFα, G-CSF, and GM-CSF, in the ONC201-treated mice compared with controls. Our data demonstrated excellent chemopreventive potential of orally administered ONC201 against intestinal tumorigenesis in the AOM-Apc min/+ mouse model.

6.
J Clin Invest ; 118(1): 111-23, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18079962

RESUMEN

Preclinical data support the potential of the death-signaling receptors for TRAIL as targets for cancer therapy. However, it is unclear whether these death-signaling receptors suppress the emergence and growth of malignant tumors in vivo. Herein we show that TNF-related apoptosis-inducing ligand receptor (TRAIL-R), the only proapoptotic death-signaling receptor for TRAIL in the mouse, suppresses inflammation and tumorigenesis. Loss of a single TRAIL-R allele on the lymphoma-prone Emu-myc genetic background significantly reduced median lymphoma-free survival. TRAIL-R-deficient lymphomas developed with equal frequency irrespective of mono- or biallelic loss of TRAIL-R, had increased metastatic potential, and showed apoptotic defects relative to WT littermates. In addition, TRAIL-R-/- mice showed decreased long-term survival following a sublethal dose of ionizing radiation. Histological evaluation of moribund irradiated TRAIL-R-/- animals showed hallmarks of bronchopneumonia as well as tumor formation with increased NF-kappaB p65 expression. TRAIL-R also suppressed diethylnitrosamine-induced (DEN-induced) hepatocarcinogenesis, as an increased number of large tumors with apoptotic defects developed in the livers of DEN-treated TRAIL-R-/- mice. Thus TRAIL-R may function as an inflammation and tumor suppressor in multiple tissues in vivo.


Asunto(s)
Bronquitis/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Hepáticas/metabolismo , Linfoma/metabolismo , Neumonía/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Alquilantes/toxicidad , Animales , Bronquitis/inducido químicamente , Bronquitis/genética , Bronquitis/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/efectos de la radiación , Enfermedad Crónica , Dietilnitrosamina/toxicidad , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Linfoma/inducido químicamente , Linfoma/genética , Linfoma/patología , Ratones , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/patología , Radiación Ionizante , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Proteínas Supresoras de Tumor/genética , Irradiación Corporal Total/efectos adversos
7.
Radiat Res ; 172(2): 175-86, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19630522

RESUMEN

Abstract Dietary antioxidants have radioprotective effects after gamma-radiation exposure that limit hematopoietic cell depletion and improve animal survival. The purpose of this study was to determine whether a dietary supplement consisting of l-selenomethionine, vitamin C, vitamin E succinate, alpha-lipoic acid and N-acetyl cysteine could improve survival of mice after proton total-body irradiation (TBI). Antioxidants significantly increased 30-day survival of mice only when given after irradiation at a dose less than the calculated LD(50/30); for these data, the dose-modifying factor (DMF) was 1.6. Pretreatment of animals with antioxidants resulted in significantly higher serum total white blood cell, polymorphonuclear cell and lymphocyte cell counts at 4 h after 1 Gy but not 7.2 Gy proton TBI. Antioxidants significantly modulated plasma levels of the hematopoietic cytokines Flt-3L and TGFbeta1 and increased bone marrow cell counts and spleen mass after TBI. Maintenance of the antioxidant diet resulted in improved recovery of peripheral leukocytes and platelets after sublethal and potentially lethal TBI. Taken together, oral supplementation with antioxidants appears to be an effective approach for radioprotection of hematopoietic cells and improvement of animal survival after proton TBI.


Asunto(s)
Antioxidantes/administración & dosificación , Supervivencia Celular/efectos de la radiación , Suplementos Dietéticos , Células Madre Hematopoyéticas/efectos de la radiación , Traumatismos por Radiación/mortalidad , Irradiación Corporal Total/efectos adversos , Administración Oral , Animales , Células Madre Hematopoyéticas/patología , Masculino , Ratones , Ratones Endogámicos ICR , Protones/efectos adversos , Traumatismos por Radiación/dietoterapia , Traumatismos por Radiación/prevención & control , Traumatismos por Radiación/veterinaria , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/efectos de la radiación , Protectores contra Radiación/administración & dosificación , Análisis de Supervivencia , Tasa de Supervivencia
8.
Cancer Biol Ther ; 20(11): 1389-1397, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31423878

RESUMEN

Hydroxyurea (HU) has been widely used in sickle cell disease. Its potential long-term risk for carcinogenesis or leukemogenic risk remains undefined. Here, we report a 26 y old African-American female with Sickle Cell Disease (SCD) who developed refractory/relapsed acute myeloid leukemia (AML) 6 months after 26 months of HU use. That patient's cytogenetics and molecular genetics analyses demonstrated a complex mutation profile with 5q deletion, trisomy 8, and P53 deletion (deletion of 17p13.1). P53 gene sequence studies revealed a multitude of somatic mutations that most suggest a treatment-related etiology. The above-mentioned data indicates that the patient may have developed acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) as a direct result of HU exposure.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Carcinogénesis/efectos de los fármacos , Hidroxiurea/efectos adversos , Leucemia Mieloide Aguda/genética , Adulto , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/epidemiología , Anemia de Células Falciformes/patología , Carcinogénesis/genética , Deleción Cromosómica , Cromosomas Humanos Par 17/efectos de los fármacos , Cromosomas Humanos Par 5/efectos de los fármacos , Cromosomas Humanos Par 8/efectos de los fármacos , Femenino , Humanos , Hidroxiurea/uso terapéutico , Leucemia Mieloide Aguda/inducido químicamente , Leucemia Mieloide Aguda/epidemiología , Leucemia Mieloide Aguda/patología , Mutación/efectos de los fármacos , Factores de Riesgo
9.
Mol Cell Biol ; 25(5): 2000-13, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15713653

RESUMEN

DR5 (also called TRAIL receptor 2 and KILLER) is an apoptosis-inducing membrane receptor for tumor necrosis factor-related apoptosis-inducing ligand (also called TRAIL and Apo2 ligand). DR5 is a transcriptional target of p53, and its overexpression induces cell death in vitro. However, the in vivo biology of DR5 has remained largely unexplored. To better understand the role of DR5 in development and in adult tissues, we have created a knockout mouse lacking DR5. This mouse is viable and develops normally with the exception of having an enlarged thymus. We show that DR5 is not expressed in developing embryos but is present in the decidua and chorion early in development. DR5-null mouse embryo fibroblasts expressing E1A are resistant to treatment with TRAIL, suggesting that DR5 may be the primary proapoptotic receptor for TRAIL in the mouse. When exposed to ionizing radiation, DR5-null tissues exhibit reduced amounts of apoptosis compared to wild-type thymus, spleen, Peyer's patches, and the white matter of the brain. In the ileum, colon, and stomach, DR5 deficiency was associated with a subtle phenotype of radiation-induced cell death. These results indicate that DR5 has a limited role during embryogenesis and early stages of development but plays an organ-specific role in the response to DNA-damaging stimuli.


Asunto(s)
Apoptosis , Radiación Ionizante , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/fisiología , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis , Caspasa 3 , Caspasas/análisis , Caspasas/metabolismo , Corion/metabolismo , Daño del ADN , Decidua/metabolismo , Doxorrubicina/farmacología , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Expresión Génica , Marcación de Gen , Masculino , Glicoproteínas de Membrana/farmacología , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Eliminación de Secuencia/genética , Bazo/química , Bazo/citología , Bazo/efectos de la radiación , Ligando Inductor de Apoptosis Relacionado con TNF , Timo/química , Timo/citología , Timo/efectos de la radiación , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/fisiología
10.
Sci Rep ; 7(1): 9711, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851987

RESUMEN

Nucleotide metabolism in cancer cells can influence malignant behavior and intrinsic resistance to therapy. Here we describe p53-dependent control of the rate-limiting enzyme in the pyrimidine catabolic pathway, dihydropyrimidine dehydrogenase (DPYD) and its effect on pharmacokinetics of and response to 5-fluorouracil (5-FU). Using in silico/chromatin-immunoprecipitation (ChIP) analysis we identify a conserved p53 DNA-binding site (p53BS) downstream of the DPYD gene with increased p53 occupancy following 5-FU treatment of cells. Consequently, decrease in Histone H3K9AC and increase in H3K27me3 marks at the DPYD promoter are observed concomitantly with reduced expression of DPYD mRNA and protein in a p53-dependent manner. Mechanistic studies reveal inhibition of DPYD expression by p53 is augmented following thymidylate synthase (TS) inhibition and DPYD repression by p53 is dependent on DNA-dependent protein kinase (DNA-PK) and Ataxia telangiectasia mutated (ATM) signaling. In-vivo, liver specific Tp53 loss increases the conversion of 5-FU to 5-FUH2 in plasma and elicits a diminished 5-FU therapeutic response in a syngeneic colorectal tumor model consistent with increased DPYD-activity. Our data suggest that p53 plays an important role in controlling pyrimidine catabolism through repression of DPYD expression, following metabolic stress imposed by nucleotide imbalance. These findings have implications for the toxicity and efficacy of the cancer therapeutic 5-FU.


Asunto(s)
Dihidrouracilo Deshidrogenasa (NADP)/genética , Regulación Enzimológica de la Expresión Génica , Pirimidinas/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromatografía Liquida , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Proteínas Nucleares/metabolismo , Polimorfismo Genético , Unión Proteica , Transducción de Señal/efectos de los fármacos , Espectrometría de Masas en Tándem , Proteína p53 Supresora de Tumor/genética
11.
Cell Cycle ; 16(12): 1171-1174, 2017 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-28486014

RESUMEN

The release of inflammatory cytokines has been implicated in the toxicity of conventional radiotherapy (CRT). Transforming growth factor ß (TGF-ß) has been suggested to be a risk marker for pulmonary toxicity following radiotherapy. Pulsed low-dose rate radiotherapy (PLDR) is a technique that involves spreading out a conventional radiotherapy dose into short pulses of dose with breaks in between to reduce toxicities. We hypothesized that the more tolerable toxicity profile of PLDR compared with CRT may be related to differential expression of inflammatory cytokines such as TGF-ß in normal tissues. To address this, we analyzed tissues from mice that had been subjected to lethal doses of CRT and PLDR by histology and immunohistochemistry (IHC). Equivalent physical doses of CRT triggered more cellular atrophy in the bone marrow, intestine, and pancreas when compared with PLDR as indicated by hematoxylin and eosin staining. IHC data indicates that TGF-ß expression is increased in the bone marrow, intestine, and lungs of mice subjected to CRT as compared with tissues from mice subjected to PLDR. Our in vivo data suggest that differential expression of inflammatory cytokines such as TGF-ß may play a role in the more favorable normal tissue late response following treatment with PLDR.


Asunto(s)
Traumatismos Experimentales por Radiación/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Intestino Delgado/metabolismo , Intestino Delgado/patología , Intestino Delgado/efectos de la radiación , Pulmón/metabolismo , Pulmón/patología , Pulmón/efectos de la radiación , Masculino , Ratones Endogámicos BALB C , Especificidad de Órganos , Traumatismos Experimentales por Radiación/patología , Radioterapia
12.
Oncotarget ; 8(40): 66747-66757, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28977993

RESUMEN

We have developed 3D-tumoroids and tumor slice in vitro culture systems from surgical tumor specimens derived from patients with colorectal cancer (CRC) or lung cancer to evaluate immune cell populations infiltrating cultured tissues. The system incorporates patient's peripherally and tumor-derived immune cells into tumoroid in vitro cultures to evaluate the ability of the culture to mimic an immunosuppressive tumor microenvironment (ITM). This system enables analysis of tumor response to standard therapy within weeks of surgical resection. Here we show that tumoroid cultures from a CRC patient are highly sensitive to the thymidylate synthase inhibitor 5-fluorouracil (adrucil) but less sensitive to the combination of nucleoside analog trifluridine and thymidine phosphorylase inhibitor tipiracil (Lonsurf). Moreover, re-introduction of isolated immune cells derived from surrounding and infiltrating tumor tissue as well as CD45+ tumor infiltrating hematopoietic cells displayed prolonged (>10 days) survival in co-culture. Established tumor slice cultures were found to contain both an outer epithelial and inner stromal cell compartment mimicking tumor structure in vivo. Collectively, these data suggest that, 3D-tumoroid and slice culture assays may provide a feasible in vitro approach to assess efficacy of novel therapeutics in the context of heterogeneous tumor-associated cell types including immune and non-transformed stromal cells. In addition, delineating the impact of therapeutics on immune cells, and cell types involved in therapeutic resistance mechanisms may be possible in general or for patient-specific responses.

13.
Oncotarget ; 8(18): 29887-29905, 2017 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-28187452

RESUMEN

Previous studies have linked increased frequency of glycosylphosphatidylinositol-anchor protein (GPI-AP) deficiency with genomic instability and the risk of carcinogenesis. However, the underlying mechanism is still not clear. A randomForest analysis of the gene expression array data from 55 MDS patients (GSE4619) demonstrated a significant (p = 0.0007) correlation (Pearson r =-0.4068) between GPI-anchor biosynthesis gene expression and genomic instability, in which PIGN, a gene participating in GPI-AP biosynthesis, was ranked as the third most important in predicting risk of MDS progression. Furthermore, we observed that PIGN gene expression aberrations (increased transcriptional activity but diminished to no protein production) were associated with increased frequency of GPI-AP deficiency in leukemic cells during leukemic transformation/progression. PIGN gene expression aberrations were attributed to partial intron retentions between exons 14 and 15 resulting in frameshifts and premature termination which were confirmed by examining the RNA-seq data from a group of AML patients (phs001027.v1.p1). PIGN gene expression aberration correlated with the elevation of genomic instability marker expression that was independent of the TP53 regulatory pathway. Suppression/elimination of PIGN protein expression caused a similar pattern of genomic instability that was rescued by PIGN restoration. Finally, we found that PIGN bound to the spindle assembly checkpoint protein, MAD1, and regulated its expression during the cell cycle. In conclusion, PIGN gene is crucial in regulating mitotic integrity to maintain chromosomal stability and prevents leukemic transformation/progression.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Inestabilidad Genómica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Fosfotransferasas/genética , Médula Ósea/patología , Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica/genética , Biología Computacional/métodos , Progresión de la Enfermedad , Exones , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes p53 , Humanos , Intrones , Leucemia Mieloide Aguda/metabolismo , Masculino , Modelos Biológicos , Mutación , Proteínas Nucleares/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal , Huso Acromático/metabolismo
14.
Oncotarget ; 7(20): 29520-30, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27121056

RESUMEN

The DNA damage response (DDR) gene cell cycle checkpoint kinase 2 (Chk2) triggers programmed cell death and lethal radiation-induced toxicity in mice in vivo. However, it is not well established to what extent targeting of Chk2 may protect from dose-limiting toxicities (DLT) inflicted by mainstay cancer chemotherapy. We screened different classes of chemotherapy in wild type and Chk2-deficient cells. Here we show that loss of Chk2 protect from cell death in vitro and lethal toxicity in vivo following treatment with topoisomerase II (TOP2)-inhibitors whereas no such protection was observed following treatment with topoisomerase I (TOP1) inhibitors. Furthermore, through combined in silico and functional screens of the Diversity Set II (NCI/NTP) chemical library we identified the carbanilide-derivative NSC105171, also known as ptu-23, as a novel Chk2 inhibitor (Chk2i). Indeed, NSC105171 can be administered safely to mice to countermeasure etoposide-induced toxicity. Incorporation of Chk2i into chemotherapy protocols employing TOP2-inhibitors may be an effective strategy to prevent DLT's without interfering with treatment.


Asunto(s)
Quinasa de Punto de Control 2/antagonistas & inhibidores , Feniltiourea/análogos & derivados , Inhibidores de Topoisomerasa II/toxicidad , Animales , Masculino , Dosis Máxima Tolerada , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Feniltiourea/farmacología
15.
Cancer Res ; 76(3): 700-12, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26609054

RESUMEN

The combination of TRAIL death receptor agonists and radiochemotherapy to treat advanced cancers continues to be investigated in clinical trials. We previously showed that normal cells with a functional DNA damage response (DDR) upregulate the expression of death-inducing receptor DR5/TRAILR2/TNFRSF10B in a p53-dependent manner that sensitizes them to treatment with DR5 agonists. However, it is unclear if targeting DR5 selectively sensitizes cancer cells to agonist treatment following exposure to DNA-damaging chemotherapy, and to what extent normal tissues are targeted. Here, we show that the combined administration of the DR5 agonistic monoclonal antibody (mAb) and chemotherapy to wild-type mice triggered synergistic gastrointestinal toxicities (GIT) that were associated with the death of Lgr5(+) crypt base columnar stem cells in a p53- and DR5-dependent manner. Furthermore, we confirmed that normal human epithelial cells treated with the human DR5-agonistic mAb and chemotherapeutic agents were also greatly sensitized to cell death. Interestingly, our data also indicated that genetic or pharmacologic targeting of Chk2 may counteract GIT without negatively affecting the antitumor responses of combined DR5 agonist/chemotherapy treatment, further linking the DDR to TRAIL death receptor signaling in normal cells. In conclusion, the combination of DR5-targeting agonistic mAbs with DNA damaging chemotherapy may pose a risk of developing toxicity-induced conditions, and the effects of mAb-based strategies on the dose-limiting toxicity of chemotherapy must be considered when establishing new combination therapies.


Asunto(s)
Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Células Madre/efectos de los fármacos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Femenino , Enfermedades Gastrointestinales/inducido químicamente , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Células Madre/metabolismo
16.
Clin Cancer Res ; 22(24): 6192-6203, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27307592

RESUMEN

PURPOSE AND EXPERIMENTAL DESIGN: Anaplastic thyroid cancer (ATC) comprises approximately 2% of all thyroid cancers, and its median survival rate remains poor. It is responsible for more than one third of thyroid cancer-related deaths. ATC is frequently resistant to conventional therapy, and NFκB signaling has been proposed to be a feature of the disease. We aimed to assess the activity of the antimalaria drug quinacrine known to target NFκB signaling in combination with the clinically relevant kinase inhibitor sorafenib in ATC cells. The presence of NFκB-p65/RELA and its target MCL1 was demonstrated in ATC by meta-data gene set enrichment analysis and IHC. We assessed the responses of a panel of human ATC cell lines to quinacrine and sorafenib in vitro and in vivo RESULTS: We detected increased expression of NFκB-p65/RELA and MCL1 in the nucleus of a subset of ATC compared with non-neoplastic thyroid. ATC cells were found to respond with additive/synergistic tumor cell killing to the combination of sorafenib plus quinacrine in vitro, and the drug combination improves survival of immunodeficient mice injected orthotopically with ATC cells as compared with mice administered either compound alone or doxorubicin. We also demonstrate that the combination of sorafenib and quinacrine is well tolerated in mice. At the molecular level, quinacrine and sorafenib inhibited expression of prosurvival MCL1, pSTAT3, and dampened NFκB signaling. CONCLUSIONS: The combination of quinacrine and sorafenib targets emerging molecular hallmarks of ATC and shows promising results in clinically relevant models for the disease. Further testing of sorafenib plus quinacrine can be conducted in ATC patients. Clin Cancer Res; 22(24); 6192-203. ©2016 AACR.


Asunto(s)
Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Quinacrina/farmacología , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Carcinoma Anaplásico de Tiroides/metabolismo , Animales , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Doxorrubicina/farmacología , Sinergismo Farmacológico , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , FN-kappa B/metabolismo , Niacinamida/farmacología , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Sorafenib , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/metabolismo , Factor de Transcripción ReIA/metabolismo
17.
Cancer Biol Ther ; 4(9): 937-42, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16251801

RESUMEN

Apoptosis is instrumental in several physiological/pathophysiological processes and is a frequently used end-point in the development of anti-neoplastic compounds. Despite ample data on several colon cancer cell lines, little is known about the susceptibility of human colon to apoptosis following treatment with established chemotherapeutics. By treating fresh human colonic explants with 5-Fluorouracil (200 microg/ml), CPT-11 (100 microg/ml) and/or TRAIL (100 ng/ml) we readily detected a signal in situ using FITC-VAD-FMK at different time points, whereas labeling of colonic explants with EGFP-conjugated Annexin V proved less specific. Although TRAIL treatment alone appeared to cause little apoptosis in human colonic epithelia versus the control, we observed a greater number of cells undergoing apoptosis when a combination of CPT-11 and TRAIL was used as compared to either agent alone. This is the initial demonstration of TRAIL-induced apoptosis with or without a chemotherapeutic agent in fresh primary human colon epithelia explants. Thus, human colonic explants may provide a valuable reference point when candidate therapeutic compounds triggering apoptosis in colon cancer cell lines, xenografts or mouse models are developed. The results support the feasibility of developing non-invasive optical imaging strategies to detect apoptosis through direct visualization of injury to human colonic epithelia in vivo.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/farmacología , Apoptosis/efectos de los fármacos , Camptotecina/análogos & derivados , Colon/efectos de los fármacos , Fluorouracilo/farmacología , Glicoproteínas de Membrana/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Adolescente , Camptotecina/farmacología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colon/citología , Colon/cirugía , Combinación de Medicamentos , Femenino , Fluorescencia , Humanos , Procesamiento de Imagen Asistido por Computador , Irinotecán , Técnicas de Cultivo de Órganos , Ligando Inductor de Apoptosis Relacionado con TNF
18.
Curr Clin Pharmacol ; 10(4): 279-98, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26548905

RESUMEN

The survival rate of patients with colorectal cancer (CRC) is steadily increasing over the past decade. However, CRC continue to be one of the leading causes of cancer-related fatality in the United States. Current targeted strategies offer limited clinical benefits and the overall survival rate for CRC remains low. Improved understanding of the molecular changes associated with CRC that control growth factor signaling and evasion of cell death allow for the development of improved targeted therapy. This review aims to discuss some of the emerging therapies aimed to target CRC.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Terapia Molecular Dirigida , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Diseño de Fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia
19.
Expert Opin Ther Targets ; 19(9): 1171-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26004811

RESUMEN

INTRODUCTION: While apoptosis is critical for maintaining homeostasis in normal cells, defective apoptosis contributes to the survival of cancer cells. TNF-related apoptosis-inducing ligand (TRAIL)-targeted therapy has attracted significant effort for treating cancer, but the clinical results have revealed limitations. The authors review the current status of development of TRAIL-targeted therapy with an outlook towards the future. AREAS COVERED: Recombinant human proteins, small molecules and agonistic monoclonal antibodies targeting death receptors that trigger TRAIL-mediated apoptosis are covered in this article. The authors review both intrinsic and extrinsic apoptotic pathways, highlighting how the apoptosis serves as a promising therapeutic target. They also review different categories of TRAIL pathway targeting agents and provide a brief overview of clinical trials using these agents. The authors discuss the limitations of conventional approaches for targeting the TRAIL pathway as well as future directions. EXPERT OPINION: The development of better combination partners for pro-apoptotic TRAIL pathway modulators including novel agents inhibiting anti-apoptotic molecules or targeting alternative resistance pathways may improve the chances for anti-tumor responses in the clinic. Developing predictive biomarkers via circulating tumor cells/DNA, apoptosis signal products, and genetic signatures/protein biomarkers from tumor tissue are also suggested as future directions.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Diseño de Fármacos , Humanos , Neoplasias/patología , Proteínas Recombinantes/farmacología
20.
Cancer Biol Ther ; 3(7): 614-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15254408

RESUMEN

Tumor cells frequently recruit the PI3K/Akt pathway in order to evade cell death, terminal differentiation and replicative inhibition. A wealth of targets for the PI3K/Akt pathway involved in these processes has been described. Among others, targets for the Akt-kinase include certain members of the Forkhead Box Class O (FOXO) transcription factors, involved in DNA damage repair, apoptosis, cell cycle progression and arrest. Akt regulates the sub-cellular localization of FOXO3a by phosphorylation thereby preventing the protein to translocate to the nucleus and regulate transcription. Constitutive Akt-activation is frequently correlated with cytoplasmatic FOXO3a in breast tumors and this is associated with decreased patient survival. In a recent paper (Hu MC, et al. Cell 2004; 117:225-237) FOXO3a was found in the cytoplasm in the absence of activated Akt. Instead, IKKbeta was shown to interact with and phosphorylate FOXO3a. The recent findings suggest that the IKKs might serve as a potential drug target in anti-cancer therapy since multiple signal transduction pathways inhibiting proliferation and facilitating cell death could be activated.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , FN-kappa B/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Humanos , Quinasa I-kappa B , Fosfatidilinositol 3-Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA