Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 60(7): 692-696, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36539277

RESUMEN

Pathogenic germline variants in the protection of telomeres 1 gene (POT1) have been associated with predisposition to a range of tumour types, including melanoma, glioma, leukaemia and cardiac angiosarcoma. We sequenced all coding exons of the POT1 gene in 2928 European-descent melanoma cases and 3298 controls, identifying 43 protein-changing genetic variants. We performed POT1-telomere binding assays for all missense and stop-gained variants, finding nine variants that impair or disrupt protein-telomere complex formation, and we further define the role of variants in the regulation of telomere length and complex formation through molecular dynamics simulations. We determine that POT1 coding variants are a minor contributor to melanoma burden in the general population, with only about 0.5% of melanoma cases carrying germline pathogenic variants in this gene, but should be screened in individuals with a strong family history of melanoma and/or multiple malignancies.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanoma/genética , Neoplasias Cutáneas/genética , Complejo Shelterina , Proteínas de Unión a Telómeros/genética , Telómero/metabolismo , Estudios de Casos y Controles , Melanoma Cutáneo Maligno
2.
Am J Hum Genet ; 104(5): 985-989, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31006513

RESUMEN

We report a recurrent CNOT1 de novo missense mutation, GenBank: NM_016284.4; c.1603C>T (p.Arg535Cys), resulting in a syndrome of pancreatic agenesis and abnormal forebrain development in three individuals and a similar phenotype in mice. CNOT1 is a transcriptional repressor that has been suggested as being critical for maintaining embryonic stem cells in a pluripotent state. These findings suggest that CNOT1 plays a critical role in pancreatic and neurological development and describe a novel genetic syndrome of pancreatic agenesis and holoprosencephaly.


Asunto(s)
Discapacidades del Desarrollo/etiología , Holoprosencefalia/etiología , Enfermedades del Recién Nacido/etiología , Mutación , Enfermedades del Sistema Nervioso/etiología , Páncreas/anomalías , Enfermedades Pancreáticas/congénito , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Discapacidades del Desarrollo/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Holoprosencefalia/patología , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/patología , Masculino , Ratones , Ratones Noqueados , Enfermedades del Sistema Nervioso/patología , Páncreas/patología , Enfermedades Pancreáticas/etiología , Enfermedades Pancreáticas/patología , Linaje , Fenotipo , Homología de Secuencia , Síndrome
3.
Nat Genet ; 39(4): 486-95, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17353896

RESUMEN

The autosomal recessive disorder Shwachman-Diamond syndrome, characterized by bone marrow failure and leukemia predisposition, is caused by deficiency of the highly conserved Shwachman-Bodian-Diamond syndrome (SBDS) protein. Here, we identify the function of the yeast SBDS ortholog Sdo1, showing that it is critical for the release and recycling of the nucleolar shuttling factor Tif6 from pre-60S ribosomes, a key step in 60S maturation and translational activation of ribosomes. Using genome-wide synthetic genetic array mapping, we identified multiple TIF6 gain-of-function alleles that suppressed the pre-60S nuclear export defects and cytoplasmic mislocalization of Tif6 observed in sdo1Delta cells. Sdo1 appears to function within a pathway containing elongation factor-like 1, and together they control translational activation of ribosomes. Thus, our data link defective late 60S ribosomal subunit maturation to an inherited bone marrow failure syndrome associated with leukemia predisposition.


Asunto(s)
Biosíntesis de Proteínas/genética , Ribosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/genética , Proteínas Portadoras/genética , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/fisiología , Eliminación de Gen , Proteínas de Filamentos Intermediarios/genética , Modelos Biológicos , Modelos Moleculares , Mutación , Organismos Modificados Genéticamente , Factores de Elongación de Péptidos/genética , Factores de Elongación de Péptidos/fisiología , Fosfoproteínas/genética , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Ribosómicas , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética
4.
Pharm Res ; 32(5): 1663-75, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25392229

RESUMEN

PURPOSE: The purpose of the study was to evaluate the metabolism, pharmacokinetics and efficacy of phospho-NSAIDs in Ces1c-knockout mice. METHODS: Hydrolysis of phospho-NSAIDs by Ces1c was investigated using Ces1c-overexpressing cells. The rate of phospho-NSAID hydrolysis was compared between wild-type, Ces1c+/- and Ces1c-/- mouse plasma in vitro, and the effect of plasma Ces1c on the cytotoxicity of phospho-NSAIDs was evaluated. Pharmacokinetics of phospho-sulindac was examined in wild-type and Ces1c-/- mice. The impact of Ces1c on the efficacy of phospho-sulindac was investigated using lung and pancreatic cancer models in vivo. RESULTS: Phospho-NSAIDs were extensively hydrolyzed in Ces1c-overexpressing cells. Phospho-NSAID hydrolysis in wild-type mouse plasma was 6-530-fold higher than that in the plasma of Ces1c-/- mice. Ces1c-expressing wild-type mouse serum attenuated the in vitro cytotoxicity of phospho-NSAIDs towards cancer cells. Pharmacokinetic studies of phospho-sulindac using wild-type and Ces1c-/- mice demonstrated 2-fold less inactivation of phospho-sulindac in the latter. Phospho-sulindac was 2-fold more efficacious in inhibiting the growth of lung and pancreatic carcinoma in Ces1c -/- mice, as compared to wild-type mice. CONCLUSIONS: Our results indicate that intact phospho-NSAIDs are the pharmacologically active entities and phospho-NSAIDs are expected to be more efficacious in humans than in rodents due to their differential expression of carboxylesterases.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Aspirina/análogos & derivados , Hidrolasas de Éster Carboxílico/genética , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Ibuprofeno/análogos & derivados , Organofosfatos/uso terapéutico , Compuestos Organofosforados/uso terapéutico , Sulindac/análogos & derivados , Animales , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacocinética , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Aspirina/metabolismo , Aspirina/farmacocinética , Aspirina/uso terapéutico , Hidrolasas de Éster Carboxílico/sangre , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Ibuprofeno/metabolismo , Ibuprofeno/farmacocinética , Ibuprofeno/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organofosfatos/metabolismo , Organofosfatos/farmacocinética , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/farmacocinética , Sulindac/metabolismo , Sulindac/farmacocinética , Sulindac/uso terapéutico
5.
J Pharmacol Exp Ther ; 351(1): 61-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25047517

RESUMEN

Esterase hydrolysis of drugs can accelerate their elimination, thereby limiting their efficacy. Polyethylene glycol (PEG) covalently attached to drugs (pegylation) is known to improve the efficiency of many drugs. Using as a test agent the novel phospho-ibuprofen (PI), we examined whether pegylation of PI could abrogate its hydrolytic degradation by esterases; PI, known to inhibit colon cancer growth, has a carboxylic ester hydrolyzable by carboxylesterases (CES). We covalently attached mPEG-2000 to PI (PI-PEG) and studied its stability by exposing it to cells overexpressing CES and by administering it to mice. We also evaluated PI-PEG's anticancer efficacy in human colon cancer xenografts and in Apc(min/+) mice. PI-PEG was stable in the presence of cells overexpressing CES1 or CES2, whereas PI was extensively hydrolyzed (90.2 ± 0.7%, 14.3 ± 1.1%, mean ± S.E.M.). In mice, PI was nearly completely hydrolyzed. Intravenous administration of PI-PEG resulted in significant levels in blood and in colon cancer xenografts (xenograft values in parentheses): area under the curve for 0-24 hours = 2351 (2621) (nmol/g) × h; Cmax = 1965 (886) nmol/g; Tmax = 0.08 (2) hour. The blood levels of ibuprofen, its main hydrolytic product, were minimal. Compared with controls, PI-PEG inhibited the growth of the xenografts by 74.8% (P < 0.01) and reduced intestinal tumor multiplicity in Apc(min/+) mice by 73.1% (P < 0.01), prolonging their survival (100% versus 55.1% of controls; P = 0.013). Pegylation protects PI from esterase hydrolysis and improves its pharmacokinetics. In preclinical models of colon cancer, PI-PEG is a safe and efficacious agent that merits further evaluation.


Asunto(s)
Antineoplásicos/farmacocinética , Ibuprofeno/análogos & derivados , Organofosfatos/farmacocinética , Polietilenglicoles/química , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Células HEK293 , Humanos , Hidrólisis , Ibuprofeno/efectos adversos , Ibuprofeno/síntesis química , Ibuprofeno/farmacocinética , Ibuprofeno/uso terapéutico , Ratones , Organofosfatos/efectos adversos , Organofosfatos/síntesis química , Organofosfatos/uso terapéutico , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
6.
BMC Cancer ; 14: 141, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24575839

RESUMEN

BACKGROUND: The anticancer properties of aspirin are restricted by its gastrointestinal toxicity and its limited efficacy. Therefore, we synthesized phospho-aspirin (PA-2; MDC-22), a novel derivative of aspirin, and evaluated its chemotherapeutic and chemopreventive efficacy in preclinical models of triple negative breast cancer (TNBC). METHODS: Efficacy of PA-2 was evaluated in human breast cancer cells in vitro, and in orthotopic and subcutaneous TNBC xenografts in nude mice. Mechanistic studies were also carried out to elucidate the mechanism of action of PA-2. RESULTS: PA-2 inhibited the growth of TNBC cells in vitro more potently than aspirin. Treatment of established subcutaneous TNBC xenografts (MDA-MB-231 and BT-20) with PA-2 induced a strong growth inhibitory effect, resulting in tumor stasis (79% and 90% inhibition, respectively). PA-2, but not aspirin, significantly prevented the development of orthotopic MDA-MB-231 xenografts (62% inhibition). Mechanistically, PA-2: 1) inhibited the activation of epidermal growth factor receptor (EGFR) and suppressed its downstream signaling cascades, including PI3K/AKT/mTOR and STAT3; 2) induced acetylation of p53 at multiple lysine residues and enhanced its DNA binding activity, leading to cell cycle arrest; and 3) induced oxidative stress by suppressing the thioredoxin system, consequently inhibiting the activation of the redox sensitive transcription factor NF-κB. These molecular alterations were observed in vitro and in vivo, demonstrating their relevance to the anticancer effect of PA-2. CONCLUSIONS: Our findings demonstrate that PA-2 possesses potent chemotherapeutic efficacy against TNBC, and is also effective in its chemoprevention, warranting further evaluation as an anticancer agent.


Asunto(s)
Aspirina/análogos & derivados , Receptores ErbB/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/prevención & control , Organofosfatos/uso terapéutico , Estrés Oxidativo/fisiología , Proteína p53 Supresora de Tumor/administración & dosificación , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Animales , Aspirina/administración & dosificación , Aspirina/uso terapéutico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/prevención & control , Línea Celular Tumoral , Receptores ErbB/fisiología , Femenino , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Organofosfatos/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
Carcinogenesis ; 34(4): 943-51, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23338686

RESUMEN

We have synthesized a novel derivative of indomethacin, phospho-tyrosol-indomethacin (PTI; MPI-621), and evaluated its anticancer efficacy in vitro and in vivo. PTI inhibited the growth of human colon, breast and lung cancer cell lines 6-30-fold more potently than indomethacin. In vivo, in contrast to indomethacin that was unable to inhibit colon cancer xenograft growth, PTI inhibited the growth of colon (69% at 10mg/kg/day, P < 0.01) and lung (91% at 15mg/kg/day, P < 0.01) subcutaneous cancer xenografts in immunodeficient mice, suppressing cell proliferation by 33% and inducing apoptosis by 75% (P < 0.05, for both). Regarding its pharmacokinetics in mice, after a single intraperitoneal injection of PTI, its plasma levels reached the maximum concentration (Cmax = 46 µM) at 2h (Tmax) and became undetectable at 4h. Indomethacin is the major metabolite of PTI, with plasma Cmax = 378 µM and Tmax = 2.5h; it became undetectable 24h postadministration. The cellular uptake of PTI (50-200 µM) at 6h was about 200-fold greater than that of indomethacin. Regarding its safety, PTI had no significant genotoxicity, showed less gastrointestinal toxicity than indomethacin and presented no cardiac toxicity. Mechanistically, PTI suppressed prostaglandin E2 production in A549 human lung cancer cells and strongly inhibited nuclear factor-κB activation in A549 xenografts. These findings indicate that PTI merits further evaluation as an anticancer agent.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Indometacina/análogos & derivados , Indometacina/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Organofosfatos/farmacología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dinoprostona/biosíntesis , Femenino , Humanos , Indometacina/sangre , Ratones , Ratones Desnudos , Ratones SCID , FN-kappa B/antagonistas & inhibidores , FN-kappa B/efectos de los fármacos , Trasplante de Neoplasias , Organofosfatos/sangre , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Blood ; 118(16): 4305-12, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-21803848

RESUMEN

Shwachman-Diamond syndrome (SDS), a recessive leukemia predisposition disorder characterized by bone marrow failure, exocrine pancreatic insufficiency, skeletal abnormalities and poor growth, is caused by mutations in the highly conserved SBDS gene. Here, we test the hypothesis that defective ribosome biogenesis underlies the pathogenesis of SDS. We create conditional mutants in the essential SBDS ortholog of the ancient eukaryote Dictyostelium discoideum using temperature-sensitive, self-splicing inteins, showing that mutant cells fail to grow at the restrictive temperature because ribosomal subunit joining is markedly impaired. Remarkably, wild type human SBDS complements the growth and ribosome assembly defects in mutant Dictyostelium cells, but disease-associated human SBDS variants are defective. SBDS directly interacts with the GTPase elongation factor-like 1 (EFL1) on nascent 60S subunits in vivo and together they catalyze eviction of the ribosome antiassociation factor eukaryotic initiation factor 6 (eIF6), a prerequisite for the translational activation of ribosomes. Importantly, lymphoblasts from SDS patients harbor a striking defect in ribosomal subunit joining whose magnitude is inversely proportional to the level of SBDS protein. These findings in Dictyostelium and SDS patient cells provide compelling support for the hypothesis that SDS is a ribosomopathy caused by corruption of an essential cytoplasmic step in 60S subunit maturation.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/patología , Insuficiencia Pancreática Exocrina/genética , Insuficiencia Pancreática Exocrina/patología , Lipomatosis/genética , Lipomatosis/patología , Proteínas/genética , Ribosomas/patología , Enfermedades de la Médula Ósea/metabolismo , Línea Celular , Dictyostelium/genética , Dictyostelium/metabolismo , Insuficiencia Pancreática Exocrina/metabolismo , GTP Fosfohidrolasas/metabolismo , Humanos , Lipomatosis/metabolismo , Mutación , Factores de Iniciación de Péptidos/metabolismo , Proteínas/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Síndrome de Shwachman-Diamond
9.
J Pharmacol Exp Ther ; 340(2): 422-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22085648

RESUMEN

Phospho-nonsteroidal anti-inflammatory drugs (phospho-NSAIDs) are novel NSAID derivatives with improved anticancer activity and reduced side effects in preclinical models. Here, we studied the metabolism of phospho-NSAIDs by carboxylesterases and assessed the impact of carboxylesterases on the anticancer activity of phospho-NSAIDs in vitro and in vivo. The expression of human liver carboxylesterase (CES1) and intestinal carboxylesterase (CES2) in human embryonic kidney 293 cells resulted in the rapid intracellular hydrolysis of phospho-NSAIDs. Kinetic analysis revealed that CES1 is more active in the hydrolysis of phospho-sulindac, phospho-ibuprofen, phospho-naproxen, phospho-indomethacin, and phospho-tyrosol-indomethacin that possessed a bulky acyl moiety, whereas the phospho-aspirins are preferentially hydrolyzed by CES2. Carboxylesterase expression leads to a significant attenuation of the in vitro cytotoxicity of phospho-NSAIDs, suggesting that the integrity of the drug is critical for anticancer activity. Benzil and bis-p-nitrophenyl phosphate (BNPP), two carboxylesterase inhibitors, abrogated the effect of carboxylesterases and resensitized carboxylesterase-expressing cells to the potent cytotoxic effects of phospho-NSAIDs. In mice, coadministration of phospho-sulindac and BNPP partially protected the former from esterase-mediated hydrolysis, and this combination more effectively inhibited the growth of AGS human gastric xenografts in nude mice (57%) compared with phospho-sulindac alone (28%) (p = 0.037). Our results show that carboxylesterase mediates that metabolic inactivation of phospho-NSAIDs, and the inhibition of carboxylesterases improves the efficacy of phospho-NSAIDs in vitro and in vivo.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Carboxilesterasa/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Organofosfatos/metabolismo , Organofosfatos/farmacología , Animales , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/uso terapéutico , Aspirina/análogos & derivados , Aspirina/metabolismo , Aspirina/farmacología , Carboxilesterasa/antagonistas & inhibidores , Carboxilesterasa/genética , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Hidrolasas de Éster Carboxílico/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Interacciones Farmacológicas/fisiología , Quimioterapia Combinada/métodos , Inhibidores Enzimáticos/farmacología , Femenino , Células HEK293 , Humanos , Hidrólisis , Ibuprofeno/análogos & derivados , Ibuprofeno/metabolismo , Ibuprofeno/farmacología , Indometacina/análogos & derivados , Indometacina/metabolismo , Indometacina/farmacología , Concentración 50 Inhibidora , Cinética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Naproxeno/análogos & derivados , Naproxeno/metabolismo , Naproxeno/farmacología , Nitrofenoles/farmacología , Nitrofenoles/uso terapéutico , Organofosfatos/sangre , Organofosfatos/uso terapéutico , Compuestos Organofosforados/sangre , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/farmacología , Compuestos Organofosforados/uso terapéutico , Fenilglioxal/análogos & derivados , Fenilglioxal/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Sulindac/análogos & derivados , Sulindac/sangre , Sulindac/metabolismo , Sulindac/farmacología , Sulindac/uso terapéutico
10.
Pharm Res ; 29(12): 3292-301, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22782648

RESUMEN

PURPOSE: To investigate the metabolism of phospho-aspirin (PA, MDC-22), a novel anti-cancer and anti-inflammatory agent. METHODS: The metabolism of PA was studied in the liver and intestinal microsomes from mouse, rat and human. RESULTS: PA is rapidly deacetylated to phospho-salicylic acid (PSA), which undergoes regioselective oxidation to generate 3-OH-PSA and 5-OH-PSA. PSA also can be hydrolyzed to give salicylic acid (SA), which can be further glucuronidated. PA is far more stable in human liver or intestinal microsomes compared to those from mouse or rat due to its slowest deacetylation in human microsomes. Of the five major human cytochrome P450 (CYP) isoforms, CYP2C19 and 2D6 are the most active towards PSA. In contrast to PSA, conventional SA is not appreciably oxidized by the CYPs and liver microsomes, indicating that PSA is a preferred substrate of CYPs. Similarly, PA, in contrast to PSA, cannot be directly oxidized by CYPs and liver microsomes, indicating that the acetyl group of PA abrogates its oxidation by CYPs. CONCLUSIONS: Our findings establish the metabolism of PA, reveal significant inter-species differences in its metabolic transformations, and provide an insight into the role of CYPs in these processes.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Aspirina/análogos & derivados , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Microsomas/metabolismo , Organofosfatos/metabolismo , Animales , Aspirina/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Ratones , Oxidación-Reducción , Ratas
11.
Nat Commun ; 13(1): 1562, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35322020

RESUMEN

Protein synthesis is a cyclical process consisting of translation initiation, elongation, termination and ribosome recycling. The release factors SBDS and EFL1-both mutated in the leukemia predisposition disorder Shwachman-Diamond syndrome - license entry of nascent 60S ribosomal subunits into active translation by evicting the anti-association factor eIF6 from the 60S intersubunit face. We find that in mammalian cells, eIF6 holds all free cytoplasmic 60S subunits in a translationally inactive state and that SBDS and EFL1 are the minimal components required to recycle these 60S subunits back into additional rounds of translation by evicting eIF6. Increasing the dose of eIF6 in mice in vivo impairs terminal erythropoiesis by sequestering post-termination 60S subunits in the cytoplasm, disrupting subunit joining and attenuating global protein synthesis. These data reveal that ribosome maturation and recycling are dynamically coupled by a mechanism that is disrupted in an inherited leukemia predisposition disorder.


Asunto(s)
Leucemia , Proteínas , Animales , Leucemia/metabolismo , Mamíferos/metabolismo , Ratones , Proteínas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/genética , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Síndrome de Shwachman-Diamond
12.
Nat Commun ; 12(1): 2482, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33931647

RESUMEN

While oncogenes promote tumorigenesis, they also induce deleterious cellular stresses, such as apoptosis, that cancer cells must combat by coopting adaptive responses. Whether tumor suppressor gene haploinsufficiency leads to such phenomena and their mechanistic basis is unclear. Here, we demonstrate that elevated levels of the anti-apoptotic factor, CASP8 and FADD-like apoptosis regulator (CFLAR), promotes apoptosis evasion in acute myeloid leukemia (AML) cells haploinsufficient for the cut-like homeobox 1 (CUX1) transcription factor, whose loss is associated with dismal clinical prognosis. Genome-wide CRISPR/Cas9 screening identifies CFLAR as a selective, acquired vulnerability in CUX1-deficient AML, which can be mimicked therapeutically using inhibitor of apoptosis (IAP) antagonists in murine and human AML cells. Mechanistically, CUX1 deficiency directly alleviates CUX1 repression of the CFLAR promoter to drive CFLAR expression and leukemia survival. These data establish how haploinsufficiency of a tumor suppressor is sufficient to induce advantageous anti-apoptosis cell survival pathways and concurrently nominate CFLAR as potential therapeutic target in these poor-prognosis leukemias.


Asunto(s)
Apoptosis/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Haploinsuficiencia , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/genética , Inmunoprecipitación de Cromatina , Dipéptidos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ontología de Genes , Genes Supresores de Tumor , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Indoles/farmacología , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Leucemia Mielomonocítica Crónica/genética , Leucemia Mielomonocítica Crónica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Análisis por Matrices de Proteínas , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
14.
JAMA Dermatol ; 155(5): 604-609, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30586141

RESUMEN

Importance: The protection of telomeres 1 protein (POT1) is a critical component of the shelterin complex, a multiple-protein machine that regulates telomere length and protects telomere ends. Germline variants in POT1 have been linked to familial melanoma, and somatic mutations are associated with a range of cancers including cutaneous T-cell lymphoma (CTCL). Objective: To characterize pathogenic variation in POT1 in families with melanoma to inform clinical management. Design, Setting, and Participants: In this case study and pedigree evaluation, analysis of the pedigree of 1 patient with melanoma revealed a novel germline POT1 variant (p.I78T, c.233T>C, chromosome 7, g.124870933A>G, GRCh38) that was subsequently found in 2 other pedigrees obtained from the GenoMEL Consortium. Main Outcomes and Measures: (1) Identification of the POT1 p.I78T variant; (2) evaluation of the clinical features and characteristics of patients with this variant; (3) analysis of 3 pedigrees; (4) genomewide single-nucleotide polymorphism genotyping of germline DNA; and (5) a somatic genetic analysis of available nevi and 1 melanoma lesion. Results: The POT1 p.I78T variant was found in 3 melanoma pedigrees, all of persons who self-reported as being of Jewish descent, and was shown to disrupt POT1-telomere binding. A UV mutation signature was associated with nevus and melanoma formation in POT1 variant carriers, and somatic mutations in driver genes such as BRAF, NRAS, and KIT were associated with lesion development in these patients. Conclusions and Relevance: POT1 p.I78T is a newly identified, likely pathogenic, variant meriting screening for in families with melanoma after more common predisposition genes such as CDKN2A have been excluded. It could also be included as part of gene panel testing.


Asunto(s)
Mutación de Línea Germinal , Melanoma/genética , Mutación Missense , Neoplasias Cutáneas/genética , Proteínas de Unión a Telómeros/genética , Adulto , Anciano , Humanos , Judíos , Masculino , Melanoma/etnología , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Complejo Shelterina , Neoplasias Cutáneas/etnología , Melanoma Cutáneo Maligno
15.
Genome Biol ; 16: 229, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26481584

RESUMEN

The use of transposons as insertional mutagens to identify cancer genes in mice has generated a wealth of information over the past decade. Here, we discuss recent major advances in transposon-mediated insertional mutagenesis screens and compare this technology with other screening strategies.


Asunto(s)
Elementos Transponibles de ADN , Genes Relacionados con las Neoplasias , Mutagénesis Insercional/métodos , Neoplasias/genética , Animales , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Genoma , Humanos , Ratones , Metástasis de la Neoplasia
16.
Nat Struct Mol Biol ; 22(11): 914-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26479198

RESUMEN

SBDS protein (deficient in the inherited leukemia-predisposition disorder Shwachman-Diamond syndrome) and the GTPase EFL1 (an EF-G homolog) activate nascent 60S ribosomal subunits for translation by catalyzing eviction of the antiassociation factor eIF6 from nascent 60S ribosomal subunits. However, the mechanism is completely unknown. Here, we present cryo-EM structures of human SBDS and SBDS-EFL1 bound to Dictyostelium discoideum 60S ribosomal subunits with and without endogenous eIF6. SBDS assesses the integrity of the peptidyl (P) site, bridging uL16 (mutated in T-cell acute lymphoblastic leukemia) with uL11 at the P-stalk base and the sarcin-ricin loop. Upon EFL1 binding, SBDS is repositioned around helix 69, thus facilitating a conformational switch in EFL1 that displaces eIF6 by competing for an overlapping binding site on the 60S ribosomal subunit. Our data reveal the conserved mechanism of eIF6 release, which is corrupted in both inherited and sporadic leukemias.


Asunto(s)
Factores Eucarióticos de Iniciación/metabolismo , GTP Fosfohidrolasas/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Proteínas/metabolismo , Proteínas Protozoarias/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Microscopía por Crioelectrón , Dictyostelium/genética , GTP Fosfohidrolasas/química , Humanos , Modelos Biológicos , Modelos Moleculares , Conformación Molecular , Factores de Elongación de Péptidos , Proteínas/química , Ribonucleoproteína Nuclear Pequeña U5 , Subunidades Ribosómicas Grandes de Eucariotas/química
17.
PLoS One ; 9(11): e111720, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25369051

RESUMEN

Phospho-aspirin (PA-2) is a novel aspirin derivative that exhibits promising anticancer properties and is considerably safer than conventional aspirin. In this study, we investigated the chemotherapeutic efficacy of PA-2 in preclinical models of estrogen receptor positive (ER+) breast cancer and elucidated its mechanism of action. PA-2 inhibited the growth of ER+ cells more potently than aspirin in vitro, and exerted a triple cytokinetic effect that includes induction of apoptosis and cell cycle arrest as well as the inhibition of cell proliferation. PA-2 is highly efficacious in vivo, as treatment of established MCF7 xenografts with PA-2 induced tumor stasis (98.2% inhibition, p<0.01). PA-2 triggered the activation of p53-dependent apoptosis via two distinct mechanisms: 1) acetylation of p53 (at K373), which disrupts its interaction with its transcription repressor MDM2, and 2) translocation of p53 to the mitochondria leading to the dissipation of mitochondrial transmembrane potential (ΔΨ(m)). Consistent with these observations, both the RNAi-mediated knockdown of p53 and forced deactylation via HDAC1 over-expression attenuated the anticancer effect of PA-2 in MCF7 cells. An upstream mediator of the signaling effects of PA-2 is RONS. PA-2 induced oxidative stress in vitro and in mice bearing MCF7 xenografts; its induction effect appears to be tumor-specific. Crucially, administration of N-acetylcysteine, a ROS scavenger, abrogated the effect of PA-2 on p53 acetylation and mitochondria translocation, thus identifying RONS as proximal molecules mediating the anticancer effect of PA-2. In summary, our findings demonstrate that PA-2 is a promising antineoplastic compound against ER+ breast cancer, warranting further evaluation as an anticancer agent.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Aspirina/análogos & derivados , Aspirina/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Mama/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Ratones , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Receptores de Estrógenos/análisis , Proteína p53 Supresora de Tumor/metabolismo
18.
Int J Oncol ; 44(2): 521-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24284479

RESUMEN

Phospho-sulindac (P-S), a promising anticancer agent, is efficacious in pre-clinical models of human cancer and is apparently safe. Here, we studied the effect of P-S on pancreatic cancer growth. We found that P-S strongly inhibits the growth of human pancreatic cancer cells in vitro, is efficacious in inhibiting the growth of pancreatic xenografts in nude mice, and has an excellent safety profile. Microarray analysis revealed that P-S induced the expression of nuclear factor of activated T-cells, isoform c1 (NFATc1) gene. NFATc1, a calcineurin-responsive transcription factor associated with aggressive pancreatic cancer. The role of increased NFATc1 expression on the growth inhibitory effect of P-S on cancer growth was evaluated by silencing or by overexpressing it both in vitro and in vivo. We found that when the expression of NFATc1 was abrogated by RNAi, pancreatic cancer cells were more responsive to treatment with P-S. Conversely, overexpressing the NFATc1 gene made the pancreatic cancer cells less responsive to treatment with P-S. NFATc1 likely mediates drug resistance to P-S and is an unfavorable prognostic factor that predicts poor tumor response. We also demonstrated that NFATc1-mediated resistance can be overcome by cyclosporin A (CsA), an NFAT inhibitor, and that the combination of P-S and CsA synergistically inhibited pancreatic cancer cell growth. In conclusion, our preclinical data establish P-S as an efficacious drug for pancreatic cancer in preclinical models, which merits further evaluation.


Asunto(s)
Resistencia a Antineoplásicos , Factores de Transcripción NFATC/metabolismo , Compuestos Organofosforados/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Sulindac/análogos & derivados , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Neoplasias Pancreáticas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulindac/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Pharm ; 477(1-2): 236-43, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25311177

RESUMEN

Phospho-non-steroidal anti-inflammatory drugs (phospho-NSAIDs) are a novel class of NSAID derivatives with potent antitumor activity. However, phospho-NSAIDs have limited stability in vivo due to their rapid hydrolysis by carboxylesterases at their carboxylic ester link. Here, we synthesized phospho-ibuprofen amide (PIA), a metabolically stable analog of phospho-ibuprofen, formulated it in nanocarriers, and evaluated its pharmacokinetics and anticancer efficacy in pre-clinical models of human lung cancer. PIA was 10-fold more potent than ibuprofen in suppressing the growth of human non-small-cell lung cancer (NSCLC) cell lines, an effect mediated by favorably altering cytokinetics and inducing oxidative stress. Pharmacokinetic studies in rats revealed that liposome-encapsulated PIA exhibited remarkable resistance to hydrolysis by carboxylesterases, remaining largely intact in the systemic circulation, and demonstrated selective distribution to the lungs. The antitumor activity of liposomal PIA was evaluated in a metastatic model of human NSCLC in mice. Liposomal PIA strongly inhibited lung tumorigenesis (>95%) and was significantly (p<0.05) more efficacious than ibuprofen. We observed a significant induction of urinary 8-iso-prostaglandin F2αin vivo, which indicates that ROS stress probably plays an important role in mediating the antitumor efficacy of PIA. Our findings suggest that liposomal PIA is a potent agent in the treatment of lung cancer and merits further evaluation.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Portadores de Fármacos/química , Ibuprofeno/análogos & derivados , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/química , Organofosfatos/síntesis química , Organofosfatos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Estabilidad de Medicamentos , Humanos , Ibuprofeno/síntesis química , Ibuprofeno/química , Ibuprofeno/farmacocinética , Ibuprofeno/farmacología , Liposomas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones Desnudos , Estructura Molecular , Organofosfatos/química , Organofosfatos/farmacocinética , Estrés Oxidativo/efectos de los fármacos , Ratas , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Nat Genet ; 46(1): 33-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24316979

RESUMEN

A major challenge in cancer genetics is to determine which low-frequency somatic mutations are drivers of tumorigenesis. Here we interrogate the genomes of 7,651 diverse human cancers and find inactivating mutations in the homeodomain transcription factor gene CUX1 (cut-like homeobox 1) in ~1-5% of various tumors. Meta-analysis of CUX1 mutational status in 2,519 cases of myeloid malignancies reveals disruptive mutations associated with poor survival, highlighting the clinical significance of CUX1 loss. In parallel, we validate CUX1 as a bona fide tumor suppressor using mouse transposon-mediated insertional mutagenesis and Drosophila cancer models. We demonstrate that CUX1 deficiency activates phosphoinositide 3-kinase (PI3K) signaling through direct transcriptional downregulation of the PI3K inhibitor PIK3IP1 (phosphoinositide-3-kinase interacting protein 1), leading to increased tumor growth and susceptibility to PI3K-AKT inhibition. Thus, our complementary approaches identify CUX1 as a pan-driver of tumorigenesis and uncover a potential strategy for treating CUX1-mutant tumors.


Asunto(s)
Genes Supresores de Tumor , Proteínas de Homeodominio/genética , Mutación , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Animales , Elementos Transponibles de ADN , Drosophila/genética , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis Insercional , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Factores de Transcripción , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA