Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 25(11): 2182-2193, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27053713

RESUMEN

The most common congenital disorder of glycosylation (CDG), phosphomannomutase 2 (PMM2)-CDG, is caused by mutations in PMM2 that limit availability of mannose precursors required for protein N-glycosylation. The disorder has no therapy and there are no models to test new treatments. We generated compound heterozygous mice with the R137H and F115L mutations in Pmm2 that correspond to the most prevalent alleles found in patients with PMM2-CDG. Many Pmm2R137H/F115L mice died prenatally, while survivors had significantly stunted growth. These animals and cells derived from them showed protein glycosylation deficiencies similar to those found in patients with PMM2-CDG. Growth-related glycoproteins insulin-like growth factor (IGF) 1, IGF binding protein-3 and acid-labile subunit, along with antithrombin III, were all deficient in Pmm2R137H/F115L mice, but their levels in heterozygous mice were comparable to wild-type (WT) littermates. These imbalances, resulting from defective glycosylation, are likely the cause of the stunted growth seen both in our model and in PMM2-CDG patients. Both Pmm2R137H/F115L mouse and PMM2-CDG patient-derived fibroblasts displayed reductions in PMM activity, guanosine diphosphate mannose, lipid-linked oligosaccharide precursor and total cellular protein glycosylation, along with hypoglycosylation of a new endogenous biomarker, glycoprotein 130 (gp130). Over-expression of WT-PMM2 in patient-derived fibroblasts rescued all these defects, showing that restoration of mutant PMM2 activity is a viable therapeutic strategy. This functional mouse model of PMM2-CDG, in vitro assays and identification of the novel gp130 biomarker all shed light on the human disease, and moreover, provide the essential tools to test potential therapeutics for this untreatable disease.


Asunto(s)
Biomarcadores , Trastornos Congénitos de Glicosilación/genética , Receptor gp130 de Citocinas/genética , Fosfotransferasas (Fosfomutasas)/genética , Animales , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/patología , Receptor gp130 de Citocinas/biosíntesis , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Genotipo , Glicosilación , Humanos , Manosa/genética , Manosa/metabolismo , Ratones , Mutación
2.
Proc Natl Acad Sci U S A ; 112(29): 9088-93, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26150517

RESUMEN

Mitochondrial aldehyde dehydrogenase 2 (ALDH2) in the liver removes toxic aldehydes including acetaldehyde, an intermediate of ethanol metabolism. Nearly 40% of East Asians inherit an inactive ALDH2*2 variant, which has a lysine-for-glutamate substitution at position 487 (E487K), and show a characteristic alcohol flush reaction after drinking and a higher risk for gastrointestinal cancers. Here we report the characterization of knockin mice in which the ALDH2(E487K) mutation is inserted into the endogenous murine Aldh2 locus. These mutants recapitulate essentially all human phenotypes including impaired clearance of acetaldehyde, increased sensitivity to acute or chronic alcohol-induced toxicity, and reduced ALDH2 expression due to a dominant-negative effect of the mutation. When treated with a chemical carcinogen, these mutants exhibit increased DNA damage response in hepatocytes, pronounced liver injury, and accelerated development of hepatocellular carcinoma (HCC). Importantly, ALDH2 protein levels are also significantly lower in patient HCC than in peritumor or normal liver tissues. Our results reveal that ALDH2 functions as a tumor suppressor by maintaining genomic stability in the liver, and the common human ALDH2 variant would present a significant risk factor for hepatocarcinogenesis. Our study suggests that the ALDH2*2 allele-alcohol interaction may be an even greater human public health hazard than previously appreciated.


Asunto(s)
Aldehído Deshidrogenasa/genética , Carcinoma Hepatocelular/enzimología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Mutación/genética , Intoxicación Alcohólica/enzimología , Intoxicación Alcohólica/patología , Aldehído Deshidrogenasa Mitocondrial , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Carcinogénesis/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Etanol/efectos adversos , Técnicas de Sustitución del Gen , Técnicas de Genotipaje , Hepatocitos/enzimología , Hepatocitos/patología , Humanos , Hiperpigmentación/patología , Inmunohistoquímica , Hígado/enzimología , Hígado/patología , Neoplasias Hepáticas/patología , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , Polimorfismo Genético , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Piel/patología , Análisis de Supervivencia
3.
Blood ; 125(2): 296-303, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25398940

RESUMEN

Mutations of IDH1 and IDH2, which produce the oncometabolite 2-hydroxyglutarate (2HG), have been identified in several tumors, including acute myeloid leukemia. Recent studies have shown that expression of the IDH mutant enzymes results in high levels of 2HG and a block in cellular differentiation that can be reversed with IDH mutant-specific small-molecule inhibitors. To further understand the role of IDH mutations in cancer, we conducted mechanistic studies in the TF-1 IDH2 R140Q erythroleukemia model system and found that IDH2 mutant expression caused both histone and genomic DNA methylation changes that can be reversed when IDH2 mutant activity is inhibited. Specifically, histone hypermethylation is rapidly reversed within days, whereas reversal of DNA hypermethylation proceeds in a progressive manner over the course of weeks. We identified several gene signatures implicated in tumorigenesis of leukemia and lymphoma, indicating a selective modulation of relevant cancer genes by IDH mutations. As methylation of DNA and histones is closely linked to mRNA expression and differentiation, these results indicate that IDH2 mutant inhibition may function as a cancer therapy via histone and DNA demethylation at genes involved in differentiation and tumorigenesis.


Asunto(s)
Metilación de ADN/genética , Inhibidores Enzimáticos/farmacología , Histonas/genética , Isocitrato Deshidrogenasa/genética , Mutación , Transcriptoma/efectos de los fármacos , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cromatografía Liquida , Histonas/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/genética , Compuestos de Fenilurea/farmacología , Análisis de Componente Principal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología , Espectrometría de Masas en Tándem
4.
Blood ; 124(11): 1777-89, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25006129

RESUMEN

Postchemotherapy relapse presents a major unmet medical need in acute myeloid leukemia (AML), where treatment options are limited. CD25 is a leukemic stem cell marker and a conspicuous prognostic marker for overall/relapse-free survival in AML. Rare occurrence of genetic alterations among PIM family members imposes a substantial hurdle in formulating a compelling patient stratification strategy for the clinical development of selective PIM inhibitors in cancer. Here we show that CD25, a bona fide STAT5 regulated gene, is a mechanistically relevant predictive biomarker for sensitivity to PIM kinase inhibitors. Alone or in combination with tyrosine kinase inhibitors, PIM inhibitors can suppress STAT5 activation and significantly shorten the half-life of MYC to achieve substantial growth inhibition of high CD25-expressing AML cells. Our results highlight the importance of STAT5 and MYC in rendering cancer cells sensitive to PIM inhibitors. Because the presence of a CD25-positive subpopulation in leukemic blasts correlates with poor overall or relapse-free survival, our data suggest that a combination of PIM inhibitors with chemotherapy and tyrosine kinase inhibitors could improve long-term therapeutic outcomes in CD25-positive AML.


Asunto(s)
Antineoplásicos/farmacología , Crisis Blástica , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Factor de Transcripción STAT5/metabolismo , Antineoplásicos/química , Crisis Blástica/tratamiento farmacológico , Crisis Blástica/genética , Crisis Blástica/metabolismo , Crisis Blástica/patología , Femenino , Células HL-60 , Humanos , Subunidad alfa del Receptor de Interleucina-2/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-pim-1/genética , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Factor de Transcripción STAT5/genética
5.
J Inherit Metab Dis ; 39(6): 807-820, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27469509

RESUMEN

D-2-hydroxyglutaric aciduria (D2HGA) type II is a rare neurometabolic disorder caused by germline gain-of-function mutations in isocitrate dehydrogenase 2 (IDH2), resulting in accumulation of D-2-hydroxyglutarate (D2HG). Patients exhibit a wide spectrum of symptoms including cardiomyopathy, epilepsy, developmental delay and limited life span. Currently, there are no effective therapeutic interventions. We generated a D2HGA type II mouse model by introducing the Idh2R140Q mutation at the native chromosomal locus. Idh2R140Q mice displayed significantly elevated 2HG levels and recapitulated multiple defects seen in patients. AGI-026, a potent, selective inhibitor of the human IDH2R140Q-mutant enzyme, suppressed 2HG production, rescued cardiomyopathy, and provided a survival benefit in Idh2R140Q mice; treatment withdrawal resulted in deterioration of cardiac function. We observed differential expression of multiple genes and metabolites that are associated with cardiomyopathy, which were largely reversed by AGI-026. These findings demonstrate the potential therapeutic benefit of an IDH2R140Q inhibitor in patients with D2HGA type II.


Asunto(s)
Encefalopatías Metabólicas Innatas/tratamiento farmacológico , Cardiomiopatías/tratamiento farmacológico , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Mutación/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Encefalopatías Metabólicas Innatas/genética , Modelos Animales de Enfermedad , Isocitrato Deshidrogenasa/genética , Ratones , Mutación/genética
6.
J Biol Chem ; 288(42): 30125-30138, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24003220

RESUMEN

PRP4 kinase is known for its roles in regulating pre-mRNA splicing and beyond. Therefore, a wider spectrum of PRP4 kinase substrates could be expected. The role of PRP4 kinase in cancer is also yet to be fully elucidated. Attaining specific and potent PRP4 inhibitors would greatly facilitate the study of PRP4 biological function and its validation as a credible cancer target. In this report, we verified the requirement of enzymatic activity of PRP4 in regulating cancer cell growth and identified an array of potential novel substrates through orthogonal proteomics approaches. The ensuing effort in structural biology unveiled for the first time unique features of PRP4 kinase domain and its potential mode of interaction with a low molecular weight inhibitor. These results provide new and important information for further exploration of PRP4 kinase function in cancer.


Asunto(s)
Proteínas de Neoplasias , Neoplasias , Inhibidores de Proteínas Quinasas , Ribonucleoproteína Nuclear Pequeña U4-U6 , Línea Celular Tumoral , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteómica/métodos , Ribonucleoproteína Nuclear Pequeña U4-U6/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U4-U6/química , Ribonucleoproteína Nuclear Pequeña U4-U6/genética , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo
7.
Sci Transl Med ; 16(749): eabp8334, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38809966

RESUMEN

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease driven by gain-of-function variants in activin receptor-like kinase 2 (ALK2), the most common variant being ALK2R206H. In FOP, ALK2 variants display increased and dysregulated signaling through the bone morphogenetic protein (BMP) pathway resulting in progressive and permanent replacement of skeletal muscle and connective tissues with heterotopic bone, ultimately leading to severe debilitation and premature death. Here, we describe the discovery of BLU-782 (IPN60130), a small-molecule ALK2R206H inhibitor developed for the treatment of FOP. A small-molecule library was screened in a biochemical ALK2 binding assay to identify potent ALK2 binding compounds. Iterative rounds of structure-guided drug design were used to optimize compounds for ALK2R206H binding, ALK2 selectivity, and other desirable pharmacokinetic properties. BLU-782 preferentially bound to ALK2R206H with high affinity, inhibiting signaling from ALK2R206H and other rare FOP variants in cells in vitro without affecting signaling of closely related homologs ALK1, ALK3, and ALK6. In vivo efficacy of BLU-782 was demonstrated using a conditional knock-in ALK2R206H mouse model, where prophylactic oral dosing reduced edema and prevented cartilage and heterotopic ossification (HO) in both muscle and bone injury models. BLU-782 treatment preserved the normal muscle-healing response in ALK2R206H mice. Delayed dosing revealed a short 2-day window after injury when BLU-782 treatment prevented HO in ALK2R206H mice, but dosing delays of 4 days or longer abrogated HO prevention. Together, these data suggest that BLU-782 may be a candidate for prevention of HO in FOP.


Asunto(s)
Modelos Animales de Enfermedad , Miositis Osificante , Osificación Heterotópica , Animales , Miositis Osificante/tratamiento farmacológico , Miositis Osificante/metabolismo , Osificación Heterotópica/tratamiento farmacológico , Osificación Heterotópica/metabolismo , Osificación Heterotópica/prevención & control , Ratones , Humanos , Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo I/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
8.
Gastro Hep Adv ; 2(3): 307-321, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-39132655

RESUMEN

Background and Aims: Fibrolamellar carcinoma (FLC) is a rare, difficult-to-treat liver cancer primarily affecting pediatric and adolescent patients, and for which precision medicine approaches have historically not been possible. The DNAJB1-PRKACA gene fusion was identified as a driver of FLC pathogenesis. We aimed to assess whether FLC tumors maintain dependency on this gene fusion and determine if PRKACA is a viable therapeutic target. Methods: FLC patient-derived xenograft (PDX) shRNA cell lines were implanted subcutaneously into female NOD-SCID mice and tumors were allowed to develop prior to randomization to doxycycline (to induce knockdown) or control groups. Tumor development was assessed every 2 days. To assess the effect of treatment with novel selective PRKACA small molecule kinase inhibitors, BLU0588 and BLU2864, FLC PDX tumor cells were implanted subcutaneously into NOD-SCID mice and tumors allowed to develop. Mice were randomized to treatment (BLU0588 and BLU2864, orally, once daily) or control groups and tumor size determined as previously. Results: Knockdown of DNAJB1-PRKACA reversed a FLC-specific gene signature and reduced PDX tumor growth in mice compared to the control group. Furthermore, FLC PDX tumor growth was significantly reduced with BLU0588 and BLU2864 treatment vs control (P = .003 and P = .0005, respectively). Conclusion: We demonstrated, using an inducible knockdown and small molecule approaches, that FLC PDX tumors were dependent upon DNAJB1-PRKACA fusion activity. In addition, this study serves as a proof-of-concept that PRKACA is a viable therapeutic target for FLC and warrants further investigation.

9.
Nature ; 441(7092): 451-6, 2006 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-16724057

RESUMEN

A cancer drug target is only truly validated by demonstrating that a given therapeutic agent is clinically effective and acts through the target against which it was designed. Nevertheless, it is desirable to declare an early-stage drug target as 'validated' before investing in a full-scale drug discovery programme dedicated to it. Although the outcome of validation studies can guide cancer research programmes, strictly defined universal validation criteria have not been established.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Células/efectos de los fármacos , Células/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/normas , Humanos , Neoplasias/genética , Neoplasias/patología , Reproducibilidad de los Resultados , Especificidad por Sustrato
10.
Stem Cells ; 26(3): 621-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18055448

RESUMEN

Although Notch ligands are considered to activate signaling through direct cell-cell contact, the existence of soluble forms has been demonstrated. However, their roles remain controversial: soluble forms have been reported to mimic the biological activity of membrane-bound form, whereas other studies rather suggested an antagonistic activity toward their full-length counterparts. We previously observed that membrane-bound Delta4-expressing S17 stroma (mbD4/S17) reduced human CD34+CD38(low) cell proliferation and favored self-renewal. Here, we assessed the effects of a soluble form of Delta4 (solD4) by exposing CD34+CD38(low) cells to S17 feeders engineered to express solD4 (solD4/S17). In contrast to mbD4/S17, (a) solD4/S17 increased 10-fold cell production after 2 weeks, through enhanced cell proliferation, and (b) it did not preserve colony-forming cell and long-term culture-initiating cell potential of output CD34+ cells. mbD4 and solD4 appeared to also differ in their signaling. Indeed, mbD4, but not solD4, strongly activated both CSL (the nuclear mediator of Notch signaling) in Hela cells overexpressing Notch1 and transcription of some classic Notch target genes in CD34+CD38(low) cells. Furthermore, both biological effects and CSL activation elicited by mbD4 were strictly dependent upon the gamma-secretase complex, whereas solD4 enhanced cell expansion in a partially gamma-secretase-independent manner. Altogether, these results suggest that part of solD4 activity did not rely upon canonical Notch pathway.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD34/metabolismo , Diferenciación Celular , Membrana Celular/metabolismo , Sangre Fetal/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores Notch/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Núcleo Celular/metabolismo , Proliferación Celular , Técnicas de Cocultivo , Regulación de la Expresión Génica , Células HeLa , Humanos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Solubilidad
11.
Cancer Discov ; 9(12): 1686-1695, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31575540

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide with no clinically confirmed oncogenic driver. Although preclinical studies implicate the FGF19 receptor FGFR4 in hepatocarcinogenesis, the dependence of human cancer on FGFR4 has not been demonstrated. Fisogatinib (BLU-554) is a potent and selective inhibitor of FGFR4 and demonstrates clinical benefit and tumor regression in patients with HCC with aberrant FGF19 expression. Mutations were identified in the gatekeeper and hinge-1 residues in the kinase domain of FGFR4 upon disease progression in 2 patients treated with fisogatinib, which were confirmed to mediate resistance in vitro and in vivo. A gatekeeper-agnostic, pan-FGFR inhibitor decreased HCC xenograft growth in the presence of these mutations, demonstrating continued FGF19-FGFR4 pathway dependence. These results validate FGFR4 as an oncogenic driver and warrant further therapeutic targeting of this kinase in the clinic. SIGNIFICANCE: Our study is the first to demonstrate on-target FGFR4 kinase domain mutations as a mechanism of acquired clinical resistance to targeted therapy. This further establishes FGF19-FGFR4 pathway activation as an oncogenic driver. These findings support further investigation of fisogatinib in HCC and inform the profile of potential next-generation inhibitors.See related commentary by Subbiah and Pal, p. 1646.This article is highlighted in the In This Issue feature, p. 1631.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico por imagen , Resistencia a Antineoplásicos , Neoplasias Hepáticas/diagnóstico por imagen , Piranos/farmacología , Quinazolinas/farmacología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Anciano de 80 o más Años , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Modelos Moleculares , Mutación , Trasplante de Neoplasias , Dominios Proteicos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo
12.
Nat Commun ; 10(1): 96, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626880

RESUMEN

Aberrant metabolism of cancer cells is well appreciated, but the identification of cancer subsets with specific metabolic vulnerabilities remains challenging. We conducted a chemical biology screen and identified a subset of neuroendocrine tumors displaying a striking pattern of sensitivity to inhibition of the cholesterol biosynthetic pathway enzyme squalene epoxidase (SQLE). Using a variety of orthogonal approaches, we demonstrate that sensitivity to SQLE inhibition results not from cholesterol biosynthesis pathway inhibition, but rather surprisingly from the specific and toxic accumulation of the SQLE substrate, squalene. These findings highlight SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly small cell lung cancers.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Escualeno-Monooxigenasa/antagonistas & inhibidores , Escualeno-Monooxigenasa/metabolismo , Antineoplásicos/química , Línea Celular Tumoral , Colesterol/biosíntesis , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos
13.
Cell Signal ; 18(12): 2223-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16824733

RESUMEN

Receptor-interacting protein 2 (RIP2) is a serine-threonine kinase that mediates signaling for many receptors of the innate and adaptive immune systems. Toll like receptors (TLR) are an important component of the innate immune response. Stimulation of RIP2-deficient cells with ligands for TLR 2, 3 and 4 results in impaired cytokine production and decreased activation of NF-kB and MAP kinases compared to wild-type cells. Stimulation of TLR 4 with its ligand lipopolysaccaride (LPS) leads to the activation of RIP2 kinase activity and its autophosphorylation. Here we identify serine residue 176 as a site of autophosphorylation using a combination of mass spectrometry and mutational analysis. Mutation of S176 to alanine not only abolishes autophosphorylation of RIP2 but also significantly decreases its catalytic activity. A phospho-specific anti-S176 antibody detects wild-type RIP2 but not kinase-dead RIP2 or the RIP2 S176A mutant. Endogenous RIP2 in THP-1 cells and mouse bone marrow derived macrophages can be detected by the phospho-RIP2 (S176) antibody only after stimulation with LPS suggesting that the antibody recognizes activated RIP2. In summary, our results indicate that S176 is a regulatory autophosphorylation site for RIP2 and that S176 phosphorylation can be used to monitor the activation state of RIP2.


Asunto(s)
Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/química , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos/inmunología , Sitios de Unión , Western Blotting , Línea Celular , Cromatografía Liquida , Humanos , Espectrometría de Masas/métodos , Ratones , Datos de Secuencia Molecular , Mutación/genética , Fosforilación , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Análisis de Secuencia de Proteína/métodos , Spodoptera
14.
Cancer Discov ; 7(5): 478-493, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28193778

RESUMEN

Somatic gain-of-function mutations in isocitrate dehydrogenases (IDH) 1 and 2 are found in multiple hematologic and solid tumors, leading to accumulation of the oncometabolite (R)-2-hydroxyglutarate (2HG). 2HG competitively inhibits α-ketoglutarate-dependent dioxygenases, including histone demethylases and methylcytosine dioxygenases of the TET family, causing epigenetic dysregulation and a block in cellular differentiation. In vitro studies have provided proof of concept for mutant IDH inhibition as a therapeutic approach. We report the discovery and characterization of AG-221, an orally available, selective, potent inhibitor of the mutant IDH2 enzyme. AG-221 suppressed 2HG production and induced cellular differentiation in primary human IDH2 mutation-positive acute myeloid leukemia (AML) cells ex vivo and in xenograft mouse models. AG-221 also provided a statistically significant survival benefit in an aggressive IDH2R140Q-mutant AML xenograft mouse model. These findings supported initiation of the ongoing clinical trials of AG-221 in patients with IDH2 mutation-positive advanced hematologic malignancies.Significance: Mutations in IDH1/2 are identified in approximately 20% of patients with AML and contribute to leukemia via a block in hematopoietic cell differentiation. We have shown that the targeted inhibitor AG-221 suppresses the mutant IDH2 enzyme in multiple preclinical models and induces differentiation of malignant blasts, supporting its clinical development. Cancer Discov; 7(5); 478-93. ©2017 AACR.See related commentary by Thomas and Majeti, p. 459See related article by Shih et al., p. 494This article is highlighted in the In This Issue feature, p. 443.


Asunto(s)
Aminopiridinas/farmacología , Antineoplásicos/farmacología , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Leucemia Mieloide Aguda/genética , Triazinas/farmacología , Animales , Línea Celular Tumoral , Humanos , Isocitrato Deshidrogenasa/genética , Ratones , Mutación , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Autoimmunity ; 39(6): 469-78, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17060026

RESUMEN

In the present study we have characterized T cell-driven immune function in mice that are genetically deficient in PKC theta. In response to simple immunologic stimulation invoked by in vivo T cell receptor (TCR) cross-linking, these mice showed significantly depressed plasma cytokine levels for IL-2, IL-4, IFNgamma, and TNFalpha compared to wild-type (WT) mice. In parallel, spleen mRNA levels for these cytokines were reduced, and NF-kappaB activation was also reduced in PKC theta knockouts (KO). Injection of allogeneic cells into the footpad of PKC theta deficient mice provoked a significantly diminished local T cell response compared to WT mice similarly challenged. Unlike comparable cells from wild type mice, CD45RBhi T cells harvested from PKC theta deficient mice failed to induce colitis in the SCID-CD45RB cell transfer model of IBD. In another T cell-dependent model of inflammatory disease, PKC theta deficient animals developed far less severe neurologic signs and reduced spinal cord inflammatory cell infiltrate compared to WT controls in the MOG-induced EAE model. A fundamental role for PKC theta in T cell activation and in the development of T cell-mediated inflammatory diseases is indicated by these results.


Asunto(s)
Inflamación/inmunología , Isoenzimas/genética , Proteína Quinasa C/genética , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Antígenos CD4/inmunología , Proliferación Celular , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Activación Enzimática , Femenino , Inflamación/patología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Isoenzimas/inmunología , Antígenos Comunes de Leucocito/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Activación de Linfocitos , Ratones , Ratones Noqueados , Ratones SCID , FN-kappa B/metabolismo , Proteína Quinasa C/inmunología , Proteína Quinasa C-theta , ARN Mensajero/sangre , ARN Mensajero/metabolismo , Médula Espinal/inmunología , Médula Espinal/patología , Bazo/metabolismo
16.
J Leukoc Biol ; 78(2): 426-34, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15908459

RESUMEN

Macrophages exist as sentinels in innate immune response and react by expressing proinflammatory cytokines and up-regulating antigen-presenting and costimulatory molecules. We report a novel function for prokineticin-1 (PK1)/endocrine gland-derived vascular endothelial growth factor. Screening of murine tissue sections and cells for specific binding site leads to the identification of macrophages as an in vivo cellular target for PK1. We demonstrate PK1 induces differentiation of murine and human bone marrow cells into the monocyte/macrophage lineage. Human peripheral blood monocytes respond to PK1 by morphological changes and down-regulation of B7-1, CD14, CC chemokine receptor 5, and CXC chemokine receptor 4. Monocytes treated with PK1 have elevated interleukin (IL)-12 and tumor necrosis factor alpha and down-regulated IL-10 production in response to lipopolysaccharide. PK1 induces a distinct monocyte-derived cell population, which is primed for release of proinflammatory cytokines that favor a T helper cell type 1 response.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Macrófagos/fisiología , Células Progenitoras Mieloides/fisiología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/farmacología , Animales , Antígeno B7-1/biosíntesis , Células CHO , Diferenciación Celular/fisiología , Cricetinae , Cricetulus , Citocinas/biosíntesis , Humanos , Receptores de Lipopolisacáridos/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos/citología , Ratones , Receptores CCR5/biosíntesis , Receptores CXCR4/biosíntesis , Células TH1/fisiología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo
17.
Cell Rep ; 15(3): 574-587, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27068473

RESUMEN

Homozygous deletions of p16/CDKN2A are prevalent in cancer, and these mutations commonly involve co-deletion of adjacent genes, including methylthioadenosine phosphorylase (MTAP). Here, we used shRNA screening and identified the metabolic enzyme, methionine adenosyltransferase II alpha (MAT2A), and the arginine methyltransferase, PRMT5, as vulnerable enzymes in cells with MTAP deletion. Metabolomic and biochemical studies revealed a mechanistic basis for this synthetic lethality. The MTAP substrate methylthioadenosine (MTA) accumulates upon MTAP loss. Biochemical profiling of a methyltransferase enzyme panel revealed that MTA is a potent and selective inhibitor of PRMT5. MTAP-deleted cells have reduced PRMT5 methylation activity and increased sensitivity to PRMT5 depletion. MAT2A produces the PRMT5 substrate S-adenosylmethionine (SAM), and MAT2A depletion reduces growth and PRMT5 methylation activity selectively in MTAP-deleted cells. Furthermore, this vulnerability extends to PRMT5 co-complex proteins such as RIOK1. Thus, the unique biochemical features of PRMT5 create an axis of targets vulnerable in CDKN2A/MTAP-deleted cancers.


Asunto(s)
Adenosina/análogos & derivados , Antígenos de Neoplasias/metabolismo , Eliminación de Gen , Metionina Adenosiltransferasa/metabolismo , Neoplasias/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Purina-Nucleósido Fosforilasa/metabolismo , Transducción de Señal , Tionucleósidos/metabolismo , Adenosina/metabolismo , Genómica , Células HCT116 , Humanos , Complejos Multiproteicos/metabolismo , Neoplasias/metabolismo , Purina-Nucleósido Fosforilasa/deficiencia , ARN Interferente Pequeño/metabolismo
18.
Cell Rep ; 17(3): 876-890, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27732861

RESUMEN

Although aberrant metabolism in tumors has been well described, the identification of cancer subsets with particular metabolic vulnerabilities has remained challenging. Here, we conducted an siRNA screen focusing on enzymes involved in the tricarboxylic acid (TCA) cycle and uncovered a striking range of cancer cell dependencies on OGDH, the E1 subunit of the alpha-ketoglutarate dehydrogenase complex. Using an integrative metabolomics approach, we identified differential aspartate utilization, via the malate-aspartate shuttle, as a predictor of whether OGDH is required for proliferation in 3D culture assays and for the growth of xenograft tumors. These findings highlight an anaplerotic role of aspartate and, more broadly, suggest that differential nutrient utilization patterns can identify subsets of cancers with distinct metabolic dependencies for potential pharmacological intervention.


Asunto(s)
Ácido Aspártico/metabolismo , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Ciclo del Ácido Cítrico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Humanos , ARN Interferente Pequeño/metabolismo
19.
PLoS One ; 9(12): e115144, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25502225

RESUMEN

Recent work has highlighted glutaminase (GLS) as a key player in cancer cell metabolism, providing glutamine-derived carbon and nitrogen to pathways that support proliferation. There is significant interest in targeting GLS for cancer therapy, although the gene is not known to be mutated or amplified in tumors. As a result, identification of tractable markers that predict GLS dependence is needed for translation of GLS inhibitors to the clinic. Herein we validate a small molecule inhibitor of GLS and show that non-small cell lung cancer cells marked by low E-cadherin and high vimentin expression, hallmarks of a mesenchymal phenotype, are particularly sensitive to inhibition of the enzyme. Furthermore, lung cancer cells induced to undergo epithelial to mesenchymal transition (EMT) acquire sensitivity to the GLS inhibitor. Metabolic studies suggest that the mesenchymal cells have a reduced capacity for oxidative phosphorylation and increased susceptibility to oxidative stress, rendering them unable to cope with the perturbations induced by GLS inhibition. These findings elucidate selective metabolic dependencies of mesenchymal lung cancer cells and suggest novel pathways as potential targets in this aggressive cancer type.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Glutaminasa/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Estrés Oxidativo/efectos de los fármacos , Sulfuros/farmacología , Tiadiazoles/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Transición Epitelial-Mesenquimal , Estudios de Asociación Genética , Glutaminasa/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Terapia Molecular Dirigida
20.
Nat Med ; 19(11): 1410-22, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24202394

RESUMEN

Major progress has been made in recent years in the development of Hedgehog (Hh) pathway inhibitors for the treatment of patients with cancer. Promising clinical trial results have been obtained in cancers that harbor activating mutations of the Hh pathway, such as basal cell carcinoma and medulloblastoma. However, for many cancers, in which Hh ligand overexpression is thought to drive tumor growth, results have been disappointing. Here we review the preclinical data that continue to shape our understanding of the Hh pathway in tumorigenesis and the emerging clinical experience with smoothened inhibitors.


Asunto(s)
Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Descubrimiento de Drogas , Resistencia a Antineoplásicos/genética , Proteínas Hedgehog/genética , Humanos , Ligandos , Modelos Biológicos , Terapia Molecular Dirigida , Mutación , Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Receptor Smoothened , Factores de Transcripción/metabolismo , Investigación Biomédica Traslacional , Proteína con Dedos de Zinc GLI1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA