Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Microsurgery ; 39(3): 228-233, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30666705

RESUMEN

BACKGROUND: Thoracoacromial vein (TAv) is seldomly considered as a secondary outflow recipient option when venous congestion of deep inferior epigastric artery perforator (DIEP) flap is encountered. The purpose of this study was to present a computed tomography (CT)-based anatomy and a method of approaching TAv in performing superdrainage using superficial inferior epigastric vein (SIEV) in DIEP flap breast reconstruction. METHODS: For CT-based anatomical study, 42 thoracoacromial vessels (TAV) of 21 patients who underwent DIEP flap breast reconstruction were analyzed. From November 2016 to May 2018, pectoralis major (PM) muscle splitting approach to TAv in the first intercostal space was applied to 7 patients who required superdrainage via SIEV. RESULTS: TAVs at mid-first intercostal space (ICS) were located 83.5 ± 9.8 mm lateral to the sternal border (H), 41.5 ± 12.9 mm below the clavicle (V), and 11.7 ± 3.2 mm deep to the outer surface of PM muscle (D). Mean oblique distances from TAV to internal mammary vessels in the 2nd and 3rd ICS were 75.7 ± 9.7 mm and 98.2 ± 10.9 mm, respectively. Seven DIEP flaps presenting intraoperative venous congestion were successfully salvaged intraoperatively with superdrainge procedure. TAvs were harvested without cutting the PM muscle in any patient. Their mean size at anastomosis was 1.61 ± 3.2 mm (range, 0.9-2.5 mm). All flaps survived without perfusion-related complications including fat necrosis. CONCLUSIONS: Harvest of TAv by muscle-splitting approach is an alternative option when additional venous anastomosis using SIEV is mandated for managing venous congestion of DIEP flap.


Asunto(s)
Anastomosis Quirúrgica/métodos , Vena Axilar/cirugía , Arterias Epigástricas/cirugía , Mamoplastia/métodos , Microcirugia/métodos , Colgajo Perforante/efectos adversos , Colgajo Perforante/irrigación sanguínea , Procedimientos de Cirugía Plástica/métodos , Adulto , Vena Axilar/diagnóstico por imagen , Femenino , Humanos , Hiperemia/etiología , Persona de Mediana Edad , Colgajo Perforante/trasplante , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Tomógrafos Computarizados por Rayos X , Resultado del Tratamiento
2.
Nat Cell Biol ; 9(11): 1303-10, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17952061

RESUMEN

Mitochondrial proteins function as essential regulators in apoptosis. Here, we show that mitochondrial adenylate kinase 2 (AK2) mediates mitochondrial apoptosis through the formation of an AK2-FADD-caspase-10 (AFAC10) complex. Downregulation of AK2 attenuates etoposide- or staurosporine-induced apoptosis in human cells, but not that induced by tumour-necrosis-factor-related apoptosis-inducing ligand (TRAIL) or Fas ligand (FasL). During intrinsic apoptosis, AK2 translocates to the cytoplasm, whereas this event is diminished in Apaf-1 knockdown cells and prevented by Bcl-2 or Bcl-X(L). Addition of purified AK2 protein to cell extracts first induces activation of caspase-10 via FADD and subsequently caspase-3 activation, but does not affect caspase-8. AFAC10 complexes are detected in cells undergoing intrinsic cell death and AK2 promotes the association of caspase-10 with FADD. In contrast, AFAC10 complexes are not detected in several etoposide-resistant human tumour cell lines. Taken together, these results suggest that, acting in concert with FADD and caspase-10, AK2 mediates a novel intrinsic apoptotic pathway that may be involved in tumorigenesis.


Asunto(s)
Adenilato Quinasa/fisiología , Apoptosis/fisiología , Caspasa 10/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Isoenzimas/fisiología , Adenilato Quinasa/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Células HeLa , Humanos , Isoenzimas/farmacología , Complejos Multienzimáticos/metabolismo , Fracciones Subcelulares/metabolismo
3.
Nat Cancer ; 4(6): 812-828, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37277530

RESUMEN

The Hippo pathway is a key growth control pathway that is conserved across species. The downstream effectors of the Hippo pathway, YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif), are frequently activated in cancers to drive proliferation and survival. Based on the premise that sustained interactions between YAP/TAZ and TEADs (transcriptional enhanced associate domain) are central to their transcriptional activities, we discovered a potent small-molecule inhibitor (SMI), GNE-7883, that allosterically blocks the interactions between YAP/TAZ and all human TEAD paralogs through binding to the TEAD lipid pocket. GNE-7883 effectively reduces chromatin accessibility specifically at TEAD motifs, suppresses cell proliferation in a variety of cell line models and achieves strong antitumor efficacy in vivo. Furthermore, we uncovered that GNE-7883 effectively overcomes both intrinsic and acquired resistance to KRAS (Kirsten rat sarcoma viral oncogene homolog) G12C inhibitors in diverse preclinical models through the inhibition of YAP/TAZ activation. Taken together, this work demonstrates the activities of TEAD SMIs in YAP/TAZ-dependent cancers and highlights their potential broad applications in precision oncology and therapy resistance.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Proteínas Proto-Oncogénicas p21(ras)/genética , Medicina de Precisión , Factores de Transcripción/metabolismo , Transducción de Señal
4.
Proc Natl Acad Sci U S A ; 106(36): 15326-31, 2009 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-19706414

RESUMEN

Osteopontin (OPN) is highly expressed in cancer patients and plays important roles in many stages of tumor progression, such as anti-apoptosis, proliferation, and metastasis. From functional screening of human cDNA library, we isolated OPN as a caspase-8 substrate that regulates cell death during hypoxia/reoxygenation (Hyp/RO). In vitro cleavage assays demonstrate that OPN is cleaved at Asp-135 and Asp-157 by caspase-8. Cellular cleavage of OPN is observed in apoptotic cells exposed to Hyp/RO among various apoptotic stimuli and its cleavage is blocked by zVAD or IETD caspase inhibitor. Further, over-expression of OPN, the form with secretion signal, inhibits Hyp/RO-induced cell death. Caspase cleavage-defective OPN mutant (OPN D135A/D157A) is more efficient to suppress Hyp/RO-induced cell death than wild-type OPN. OPN D135A/D157A sustains AKT activity to increase cell viability through inhibition of caspase-9 during Hyp/RO. In addition, OPN is highly induced in some tumor cells during Hyp/RO, such as HeLa and Huh-7 cells, which is associated with their resistance to Hyp/RO by sustaining AKT activity. Notably, OPN C-terminal cleavage fragment produced by caspase-8 is detected in the nucleus. Plasmid-encoded expression of OPN C-terminal cleavage fragment increases p53 protein level and induces apoptosis of wild-type mouse embryonic fibroblast cells, but not p53(-/-) mouse embryonic fibroblast cells. These observations suggest that the protective function of OPN during Hyp/RO is inactivated via the proteolytic cleavage by caspase-8 and its cleavage product subsequently induces cell death via p53, postulating caspase-8 as a negative regulator of tumorigenic activity of OPN.


Asunto(s)
Apoptosis/fisiología , Caspasa 8/metabolismo , Hipoxia de la Célula/fisiología , Osteopontina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , Densitometría , Células HeLa , Humanos , Osteopontina/genética
5.
Cell Death Dis ; 13(5): 469, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35585049

RESUMEN

The RAS-BRAF signaling is a major pathway of cell proliferation and their mutations are frequently found in human cancers. Adenylate kinase 2 (AK2), which modulates balance of adenine nucleotide pool, has been implicated in cell death and cell proliferation independently of its enzyme activity. Recently, the role of AK2 in tumorigenesis was in part elucidated in some cancer types including lung adenocarcinoma and breast cancer, but the underlying mechanism is not clear. Here, we show that AK2 is a BRAF-suppressor. In in vitro assays and cell model, AK2 interacted with BRAF and inhibited BRAF activity and downstream ERK phosphorylation. Energy-deprived conditions in cell model and the addition of AMP to cell lysates strengthened the AK2-BRAF interaction, suggesting that AK2 is involved in the regulation of BRAF activity in response to cell metabolic state. AMP facilitated the AK2-BRAF complex formation through binding to AK2. In a panel of HCC cell lines, AK2 expression was inversely correlated with ERK/MAPK activation, and AK2-knockdown or -knockout increased BRAF activity and promoted cell proliferation. Tumors from HCC patients showed low-AK2 protein expression and increased ERK activation compared to non-tumor tissues and the downregulation of AK2 was also verified by two microarray datasets (TCGA-LIHC and GSE14520). Moreover, AK2/BRAF interaction was abrogated by RAS activation in in vitro assay and cell model and in a mouse model of HRASG12V-driven HCC, and AK2 ablation promoted tumor growth and BRAF activity. AK2 also bound to BRAF inhibitor-insensitive BRAF mutants and attenuated their activities. These findings indicate that AK2 monitoring cellular AMP levels is indeed a negative regulator of BRAF, linking the metabolic status to tumor growth.


Asunto(s)
Adenosina Monofosfato , Adenilato Quinasa , Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas Proto-Oncogénicas B-raf , Adenosina Monofosfato/metabolismo , Adenilato Quinasa/metabolismo , Animales , Carcinogénesis/genética , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo
6.
Cancer Discov ; 11(3): 778-793, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33208393

RESUMEN

Hippo pathway dysregulation occurs in multiple cancers through genetic and nongenetic alterations, resulting in translocation of YAP to the nucleus and activation of the TEAD family of transcription factors. Unlike other oncogenic pathways such as RAS, defining tumors that are Hippo pathway-dependent is far more complex due to the lack of hotspot genetic alterations. Here, we developed a machine-learning framework to identify a robust, cancer type-agnostic gene expression signature to quantitate Hippo pathway activity and cross-talk as well as predict YAP/TEAD dependency across cancers. Further, through chemical genetic interaction screens and multiomics analyses, we discover a direct interaction between MAPK signaling and TEAD stability such that knockdown of YAP combined with MEK inhibition results in robust inhibition of tumor cell growth in Hippo dysregulated tumors. This multifaceted approach underscores how computational models combined with experimental studies can inform precision medicine approaches including predictive diagnostics and combination strategies. SIGNIFICANCE: An integrated chemicogenomics strategy was developed to identify a lineage-independent signature for the Hippo pathway in cancers. Evaluating transcriptional profiles using a machine-learning method led to identification of a relationship between YAP/TAZ dependency and MAPK pathway activity. The results help to nominate potential combination therapies with Hippo pathway inhibition.This article is highlighted in the In This Issue feature, p. 521.


Asunto(s)
Quimioinformática/métodos , Biología Computacional/métodos , Genómica/métodos , Vía de Señalización Hippo , Sistema de Señalización de MAP Quinasas , Aprendizaje Automático , Transducción de Señal , Humanos
7.
Cancer Res ; 80(8): 1656-1668, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31988076

RESUMEN

The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk for mesothelioma and melanocytic tumors. Here, we show that pancreatic intraepithelial neoplasia driven by oncogenic mutant KrasG12D progressed to pancreatic adenocarcinoma in the absence of BAP1. The Hippo pathway was deregulated in BAP1-deficient pancreatic tumors, with the tumor suppressor LATS exhibiting enhanced ubiquitin-dependent proteasomal degradation. Therefore, BAP1 may limit tumor progression by stabilizing LATS and thereby promoting activity of the Hippo tumor suppressor pathway. SIGNIFICANCE: BAP1 is mutated in a broad spectrum of tumors. Pancreatic Bap1 deficiency causes acinar atrophy but combines with oncogenic Ras to produce pancreatic tumors. BAP1-deficient tumors exhibit deregulation of the Hippo pathway.See related commentary by Brekken, p. 1624.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Vía de Señalización Hippo , Humanos , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Proteínas Supresoras de Tumor , Ubiquitina Tiolesterasa
8.
Cancer Res ; 79(11): 2839-2852, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30987996

RESUMEN

TNF-related apoptosis-inducing ligand (TRAIL) resistance, including nongenetically acquired tolerance in cancer persister cells, is a major obstacle to translating TRAIL therapy into patients with cancer. However, the underlying mechanisms remain to be elucidated. Here, we show that DR4/TRAIL-R1 is O-GlcNAcylated at Ser424 in its death domain to mediate both apoptosis and necrosis upon TRAIL ligation. We found that DR4-Ser424 mutations, identified from our cell-based functional screen using a cancer patient-derived cDNA expression library and from The Cancer Genome Atlas, caused TRAIL resistance in various human cancer cell lines. Using O-GlcNAc transferase knockdown cells, DR4-preferred versus DR5-preferred cancer cells, and a DR5-neutralizing antibody, we evaluated the essential role of DR4-specific O-GlcNAc modification in TRAIL cytotoxicity. In contrast to DR4, DR5 was not O-GlcNAcylated by TRAIL treatment, discriminating DR4 from DR5-mediated signaling. Apart from genetic changes in DR4-Ser424, we further classified various cancer cell lines originated from stomach, colon, lung, and glioblastoma according to their sensitivity to and receptor preference upon TRAIL death signaling and generated TRAIL-tolerant persister-derived DLD-1PER cells. Among these, we discovered that DR4 was not modified by O-GlcNAc in most of the TRAIL-resistant cancer cells and DLD-1PER cells. Interestingly, promoting DR4 O-GlcNAcylation intentionally using 2-deoxy-d-glucose or a high concentration of glucose sensitized those resistant cancer cells to TRAIL. The O-GlcNAcylation-defective DR4 failed to form DISC/necrosome and could not translocate to aggregated platforms for receptor clustering. Our findings demonstrate that DR4 O-GlcNAcylation is crucial for TRAIL death signaling, providing new opportunities for TRAIL therapy overcoming TRAIL resistance in cancers. SIGNIFICANCE: This study reports that a novel posttranslational modification by O-GlcNAcylation of one of the two human TRAIL receptors with a death domain, TRAIL-R1 (DR4), plays a crucial role in enabling both apoptotic and necroptotic cell death induction by TRAIL.


Asunto(s)
Resistencia a Antineoplásicos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Serina/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Acetilglucosamina/metabolismo , Muerte Celular/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Neoplasias Endometriales/genética , Femenino , Glucosa/metabolismo , Humanos , Microdominios de Membrana/metabolismo , Mutación , N-Acetilglucosaminiltransferasas/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
9.
Pigment Cell Melanoma Res ; 32(2): 269-279, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30156010

RESUMEN

The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk of mesothelioma and melanocytic tumors. Here, we show that Bap1 deletion in melanocytes cooperates with the constitutively active, oncogenic form of BRAF (BRAFV600E ) and UV to cause melanoma in mice, albeit at very low frequency. In addition, Bap1-null melanoma cells derived from mouse tumors are more aggressive and colonize and grow at distant sites more than their wild-type counterparts. Molecularly, Bap1-null melanoma cell lines have increased DNA damage measured by γH2aX and hyperubiquitination of histone H2a. Therapeutically, these Bap1-null tumors are completely responsive to BRAF- and MEK-targeted therapies. Therefore, BAP1 functions as a tumor suppressor and limits tumor progression in melanoma.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Melanoma/genética , Melanoma/patología , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Daño del ADN , Transición Epitelial-Mesenquimal/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Melanocitos/metabolismo , Melanocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Transcripción Genética , Ubiquitinación , Melanoma Cutáneo Maligno
10.
Sci Signal ; 11(547)2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206136

RESUMEN

The Hippo signaling pathway regulates organ size and plays critical roles in maintaining tissue growth, homeostasis, and regeneration. Dysregulated in a wide spectrum of cancers, in mammals, this pathway is regulated by two key effectors, YAP and TAZ, that may functionally overlap. We found that TAZ promoted liver inflammation and tumor development. The expression of TAZ, but not YAP, in human liver tumors positively correlated with the expression of proinflammatory cytokines. Hyperactivated TAZ induced substantial myeloid cell infiltration into the liver and the secretion of proinflammatory cytokines through a TEAD-dependent mechanism. Furthermore, tumors with hyperactivated YAP and TAZ had distinct transcriptional signatures, which included the increased expression of inflammatory cytokines in TAZ-driven tumors. Our study elucidated a previously uncharacterized link between TAZ activity and inflammatory responses that influence tumor development in the liver.


Asunto(s)
Proteínas de Unión al ADN/genética , Inflamación/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/genética , Hígado/metabolismo , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/genética , Animales , Proteínas de Ciclo Celular , Citocinas/genética , Citocinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica/métodos , Vía de Señalización Hippo , Humanos , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones Endogámicos C57BL , Mutación , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/genética , Factores de Transcripción de Dominio TEA , Transactivadores , Factores de Transcripción/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Trasplante Heterólogo
11.
Mol Cell Biol ; 24(22): 9763-70, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15509781

RESUMEN

Apoptosis repressor with CARD (ARC) possesses the ability not only to block activation of caspase 8 but to modulate caspase-independent mitochondrial events associated with cell death. However, it is not known how ARC modulates both caspase-dependent and caspase-independent cell death. Here, we report that ARC is a Ca(2+)-dependent regulator of caspase 8 and cell death. We found that in Ca(2+) overlay and Stains-all assays, ARC protein bound to Ca(2+) through the C-terminal proline/glutamate-rich (P/E-rich) domain. ARC expression reduced not only cytosolic Ca(2+) transients but also cytotoxic effects of thapsigargin, A23187, and ionomycin, for which the Ca(2+)-binding domain of ARC was indispensable. Conversely, direct interference of endogenous ARC synthesis by targeting ARC enhanced such Ca(2+)-mediated cell death. In addition, binding and immunoprecipitation analyses revealed that the protein-protein interaction between ARC and caspase 8 was decreased by the increase of Ca(2+) concentration in vitro and by the treatment of HEK293 cells with thapsigargin in vivo. Caspase 8 activation was also required for the thapsigargin-induced cell death and suppressed by the ectopic expression of ARC. These results suggest that calcium binding mediates regulation of caspase 8 and cell death by ARC.


Asunto(s)
Apoptosis/fisiología , Calcio/metabolismo , Caspasas/metabolismo , Proteínas Musculares/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Células COS , Caspasa 8 , Línea Celular , Activación Enzimática/efectos de los fármacos , Células HeLa , Humanos , Técnicas In Vitro , Células Jurkat , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tapsigargina/farmacología
12.
Mol Cell Biol ; 37(2)2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27799292

RESUMEN

Fas-associated protein with death domain (FADD) plays a key role in extrinsic apoptosis. Here, we show that FADD is SUMOylated as an essential step during intrinsic necrosis. FADD was modified at multiple lysine residues (K120/125/149) by small ubiquitin-related modifier 2 (SUMO2) during necrosis caused by calcium ionophore A23187 and by ischemic damage. SUMOylated FADD bound to dynamin-related protein 1 (Drp1) in cells both in vitro and in ischemic tissue damage cores, thus promoting Drp1 recruitment by mitochondrial fission factor (Mff) to accomplish mitochondrial fragmentation. Mitochondrial-fragmentation-associated necrosis was blocked by FADD or Drp1 deficiency and SUMO-defective FADD expression. Interestingly, caspase-10, but not caspase-8, formed a ternary protein complex with SUMO-FADD/Drp1 on the mitochondria upon exposure to A23187 and potentiated Drp1 oligomerization for necrosis. Moreover, the caspase-10 L285F and A414V mutants, found in autoimmune lymphoproliferative syndrome and non-Hodgkin lymphoma, respectively, regulated this necrosis. Our study reveals an essential role of SUMOylated FADD in Drp1- and caspase-10-dependent necrosis, providing insights into the mechanism of regulated necrosis by calcium overload and ischemic injury.


Asunto(s)
Caspasa 10/metabolismo , Citosol/metabolismo , Dinaminas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Mitocondrias/metabolismo , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Hipoxia de la Célula , Células HEK293 , Células HeLa , Humanos , Lisina/metabolismo , Ratones Endogámicos C57BL , Complejos Multiproteicos/metabolismo , Proteínas Mutantes/metabolismo , Necrosis , Unión Proteica , Multimerización de Proteína , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo , Sumoilación
13.
Mol Cancer Ther ; 16(4): 694-704, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28138032

RESUMEN

Cancer cell line profiling to identify previously unrecognized kinase dependencies revealed a novel nonmutational dependency on the DNA damage response checkpoint kinase Chk1. Although Chk1 is a promising therapeutic target in p53-deficient cancers, we found that Ras-MEK signaling engages Chk1 in a subset of osteosarcoma, ovarian, and breast cancer cells to enable their survival upon DNA damage, irrespective of p53 mutation status. Mechanistically, Ras-MEK signaling drives Chk1 expression and promotes cancer cell growth that produces genotoxic stress that requires Chk1 to mediate a response to the consequent DNA damage. Reciprocally, Chk1 engages a negative feedback loop to prevent hyperactivation of Ras-MEK signaling, thereby limiting DNA damage. Furthermore, exogenous DNA damage promotes Chk1 dependency, and pharmacologic Chk1 inhibition combined with genotoxic chemotherapy potentiates a DNA damage response and tumor cell killing. These findings reveal a mechanism-based diagnostic strategy to identify cancer patients that may benefit from Chk1-targeted therapy. Mol Cancer Ther; 16(4); 694-704. ©2017 AACR.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias de la Mama/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Osteosarcoma/genética , Neoplasias Ováricas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Neoplasias Óseas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Osteosarcoma/tratamiento farmacológico , Piperidinas/administración & dosificación , Piperidinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
14.
Oncogene ; 24(4): 688-96, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15592525

RESUMEN

Caspase-8 is the most receptor-proximal, upstream caspase in the caspase cascade and plays a key role in cell death triggered by various death receptors. Here, we addressed the role of endogenous caspase-8 in tumor necrosis factor (TNF)-alpha-induced activation of NF-kappaB. Direct targeting of caspase-8 with siRNA and antisense (AS) approaches abolished TNF-alpha-induced activation of NF-kappaB in NIH3T3, HeLa, and HEK293 cells as determined with luciferase reporter gene and cell fractionation assays. Reconstitution of caspase-8-deficient C33A cells with processing-defective (P/D) mutant of caspase-8 sensitized the cells to TNF-alpha for NF-kappaB activation. In contrast to wild-type caspase-8, death effector domain mutant replacing Asp73 with Ala (caspase-8 (D73A)) failed to activate NF-kappaB and to bind FLICE-associated huge protein (FLASH) in vitro and in vivo. Instead, caspase-8 (D73A) mutant bound to caspase-8 and blocked NF-kappaB activation triggered by TNF-alpha and caspase-8. In addition, expression of an NF-kappaB-activating domain-deletion mutant of FLASH or transfection of FLASH AS oligonucleotides abolished TNF-alpha and caspase-8, but not phorbol 12-myristate 13-acetate, -induced activation of NF-kappaB. Further, immunoprecipitation assays showed that caspase-8 formed triple complex with TRAF2 and FLASH. Taken together, these results suggest that endogenous caspase-8 mediates TNF-alpha-induced activation of NF-kappaB via FLASH.


Asunto(s)
Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Caspasas/metabolismo , Mutación/genética , FN-kappa B/metabolismo , Transducción de Señal , Animales , Proteínas Reguladoras de la Apoptosis , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Caspasa 8 , Inhibidores de Caspasas , Caspasas/deficiencia , Caspasas/genética , Línea Celular , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Unión Proteica , Transducción de Señal/efectos de los fármacos , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
15.
Ann Dermatol ; 28(3): 344-51, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27274633

RESUMEN

BACKGROUND: Moisturizers with anti-inflammatory or anti-itch activity should be developed for the safe and effective management of atopic dermatitis (AD). OBJECTIVE: This study evaluated the efficacy of a newly developed moisturizer, CSP0510 lotion (Twolines Inc., Korea), containing citric acid (CA) and trisodium phosphate (TSP) as active ingredients, in mild to moderate AD. METHODS AND RESULTS: CSP0510 lotion applied twice daily for 4 weeks to eczematous lesions improved objective and subjective (itch) symptoms of AD. The physician's global assessment (PGA) score for objective symptoms decreased from 2.5±0.6 before application to 1.3±0.5 after application in the CSP0510-treated group (n=42, p<0.001). Also, the PGA score decreased from 2.3±0.6 to 1.9±0.5 by vehicle-treated (without CA and TSP) control group (p=0.001), but there was no statistical difference between CSP0510-treated and vehicle-treated groups (p=0.089). The visual analogue scale score for itch decreased from 4.8±1.3 to 2.0±0.9 in the CSP0510-treated group (p<0.001), and from 4.6±1.1 to 3.5±0.9 in the control group (p=0.075), showing a statistical significance between two groups (p=0.002). Our results in humans were further supported by in vitro and animal experiments. In HaCaT cells treated with compound 48/80 (7.5 µg/ml), CA:TSP (1:1, vol:vol) synergistically suppressed the compound 48/80-induced upregulation of thymic stromal lymphopoietin, nerve grow factor, and calcitonin gene-related peptide. Application of CSP0510 to the dorsal skin of hairless mice for 3 weeks suppressed the oxazolone-induced allergic skin inflammation. CONCLUSION: In conclusion, CSP0510 lotion has anti-itch and anti-inflammatory activity in the skin, which improves both objective and subjective symptoms of AD.

16.
Allergy Asthma Immunol Res ; 8(3): 230-8, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26922933

RESUMEN

PURPOSE: Questionnaire-based diagnostic criteria for atopic dermatitis (AD) have been proposed to detect the major group of AD with flexural dermatitis. We aimed to develop novel, questionnaire-based diagnostic criteria for childhood AD, which can detect more comprehensive AD including non-flexural type. METHODS: The draft version of questionnaire to detect childhood AD was prepared to be used for preliminary hospital- (n=1,756) and community-based (n=1,320) surveys. From analysis, the Reliable Estimation of Atopic dermatitis of ChildHood (REACH) was derived and verified in derivation (n=1,129) and validation (n=1,191) sets by community-based surveys. RESULTS: The REACH consists of 11 questions including 2 major and 9 minor criteria. AD is diagnosed as the major group of 'eczema on the antecubital or popliteal fossa' to fulfill the 2 major criteria (2M), and the minor group of 'eczema on the non-antecubital or popliteal fossa' to fulfill the 1 major plus 4 or more minor criteria (1M+4m). In the validation set, the overall 1-year AD prevalence by the REACH was estimated as 12.3% (95% CI, 10.5%-14.2%), and the REACH showed a sensitivity of 75.2%, a specificity of 96.1%, and an error rate of 6.4%. The REACH demonstrated better diagnostic performance than the ISAAC in terms of the number of misclassification (10.0%). CONCLUSIONS: We propose the REACH as new full, questionnaire-based diagnostic criteria for childhood AD in epidemiological surveys. Further studies are warranted to validate the REACH in different populations or countries in the context of large-scale, epidemiological surveys.

17.
FEBS Lett ; 578(3): 239-44, 2004 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-15637807

RESUMEN

We have identified a novel CARD-containing protein from EST database. BinCARD (Bcl10-interacting protein with CARD). BinCARD was ubiquitously expressed. Co-immunoprecipitation, In vitro binding, mammalian two-hybrid, and immunostaining assays revealed that BinCARD interacted with Bcl10 through CARD. BinCARD potently suppressed NF-kappa B activation induced by Bcl10 and decreased the amounts of phosphorylated Bcl10. Mutations at the residue Leu17 or Leu65, which is highly conserved in CARD, abolished the inhibitory effects of BinCARD on both Bcl10-induced activation of NF-kappa B and phosphorylation of Bcl10. Further, expression of BinCARD inhibited Bcl10 phosphorylation induced by T cell activation signal. These results suggest that BinCARD interacts with Bcl10 to inhibit Bcl10-mediated activation of NF-kappa B and to suppress Bcl10 phosphorylation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Portadoras/metabolismo , FN-kappa B/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteína 10 de la LLC-Linfoma de Células B , Western Blotting , Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Secuencia Conservada , Análisis Mutacional de ADN , Regulación hacia Abajo , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Inmunohistoquímica , Células Jurkat , Leucina/química , Leucina/genética , Luciferasas/metabolismo , Activación de Linfocitos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Pruebas de Precipitina , Unión Proteica , Estructura Secundaria de Proteína , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Linfocitos T/metabolismo , Transcripción Genética , Técnicas del Sistema de Dos Híbridos
18.
Cancer Cell ; 26(2): 207-21, 2014 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-25065853

RESUMEN

Pathway-targeted cancer drugs can produce dramatic responses that are invariably limited by the emergence of drug-resistant cells. We found that many drug-treated "oncogene-addicted" cancer cells engage a positive feedback loop leading to Stat3 activation, consequently promoting cell survival and limiting overall drug response. This was observed in cancer cells driven by diverse activated kinases, including EGFR, HER2, ALK, and MET, as well as mutant KRAS. Specifically, MEK inhibition led to autocrine activation of Stat3 via the FGF receptor and JAK kinases, and pharmacological inhibition of MEK together with JAK and FGFR enhanced tumor regression. These findings suggest that inhibition of a Stat3 feedback loop may augment the response to a broad spectrum of drugs that target pathways of oncogene addiction.


Asunto(s)
Adenocarcinoma/metabolismo , Resistencia a Antineoplásicos , Neoplasias Pulmonares/metabolismo , Factor de Transcripción STAT3/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/mortalidad , Adenocarcinoma del Pulmón , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Humanos , Imidazoles/farmacología , Interleucina-6/metabolismo , Janus Quinasa 1/metabolismo , Estimación de Kaplan-Meier , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Ratones , Ratones SCID , Mutación , Nitrilos , Oncogenes , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Pirazoles/farmacología , Piridazinas/farmacología , Pirimidinas , Quinazolinas/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/genética
19.
Nat Commun ; 5: 3351, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24548998

RESUMEN

Adenylate kinase 2 (AK2), which balances adenine nucleotide pool, is a multi-functional protein. Here we show that AK2 negatively regulates tumour cell growth. AK2 forms a complex with dual-specificity phosphatase 26 (DUSP26) phosphatase and stimulates DUSP26 activity independently of its AK activity. AK2/DUSP26 phosphatase protein complex dephosphorylates fas-associated protein with death domain (FADD) and regulates cell growth. AK2 deficiency enhances cell proliferation and induces tumour formation in a xenograft assay. This anti-growth function of AK2 is associated with its DUSP26-stimulating activity. Downregulation of AK2 is frequently found in tumour cells and human cancer tissues showing high levels of phospho-FADD(Ser194). Moreover, reconstitution of AK2 in AK2-deficient tumour cells retards both cell proliferation and tumourigenesis. Consistent with this, AK2(+/-) mouse embryo fibroblasts exhibit enhanced cell proliferation with a significant alteration in phospho-FADD(Ser191). These results suggest that AK2 is an associated activator of DUSP26 and suppresses cell proliferation by FADD dephosphorylation, postulating AK2 as a negative regulator of tumour growth.


Asunto(s)
Adenilato Quinasa/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Adenilato Quinasa/genética , Animales , Línea Celular , Proliferación Celular/genética , Proliferación Celular/fisiología , Fosfatasas de Especificidad Dual/genética , Electroforesis en Gel Bidimensional , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Células HeLa , Humanos , Técnicas In Vitro , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Fosforilación , Espectrometría de Masas en Tándem , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Discov ; 3(2): 168-81, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23229345

RESUMEN

UNLABELLED: Approximately half of EGFR-mutant non-small cell lung cancer (NSCLC) patients treated with small-molecule EGFR kinase inhibitors develop drug resistance associated with the EGF receptor (EGFR) T790M "gatekeeper" substitution, prompting efforts to develop covalent EGFR inhibitors, which can effectively suppress EGFR T790M in preclinical models. However, these inhibitors have yet to prove clinically efficacious, and their toxicity in skin, reflecting activity against wild-type EGFR, may limit dosing required to effectively suppress EGFR T790M in vivo. While profiling sensitivity to various kinase inhibitors across a large cancer cell line panel, we identified indolocarbazole compounds, including a clinically well-tolerated FLT3 inhibitor, as potent and reversible inhibitors of EGFR T790M that spare wild-type EGFR. These findings show the use of broad cancer cell profiling of kinase inhibitor efficacy to identify unanticipated novel applications, and they identify indolocarbazole compounds as potentially effective EGFR inhibitors in the context of T790M-mediated drug resistance in NSCLC. SIGNIFICANCE: EGFR-mutant lung cancer patients who respond to currently used EGFR kinase inhibitors invariably develop drug resistance, which is associated with the EGFR T790M resistance mutation in about half these cases. We unexpectedly identified a class of reversible potent inhibitors of EGFR T790M that do not inhibit wild-type EGFR, revealing a promising therapeutic strategy to overcome T790M-associated drug-resistant lung cancers.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Acrilamidas/farmacología , Sustitución de Aminoácidos , Animales , Área Bajo la Curva , Secuencia de Bases , Carbazoles/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Clorhidrato de Erlotinib , Humanos , Immunoblotting , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/farmacología , Quinazolinas/farmacología , Interferencia de ARN , Estaurosporina/análogos & derivados , Estaurosporina/farmacocinética , Estaurosporina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA