Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 278
Filtrar
1.
PLoS One ; 8(4): e61432, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23630589

RESUMEN

The endocrine hormone FGF21 has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to the native cytokine, we generated bispecific Avimer polypeptides that bind with high affinity and specificity to one of the receptor and coreceptor pairs used by FGF21, FGFR1c and ß-Klotho. These Avimers exhibit FGF21-like activity in in vitro assays with potency greater than FGF21. In a study conducted in obese male cynomolgus monkeys, animals treated with an FGFR1c/ß-Klotho bispecific Avimer showed improved metabolic parameters and reduced body weight comparable to the effects seen with FGF21. These results not only demonstrate the essential roles of FGFR1c and ß-Klotho in mediating the metabolic effects of FGF21, they also describe a first bispecific activator of this unique receptor complex and provide validation for a novel therapeutic approach to target this potentially important pathway for treating diabetes and obesity.


Asunto(s)
Fármacos Antiobesidad/farmacología , Factores de Crecimiento de Fibroblastos/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Péptidos/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Fármacos Antiobesidad/farmacocinética , Sitios de Unión , Unión Competitiva , Peso Corporal/efectos de los fármacos , Línea Celular , Evaluación Preclínica de Medicamentos , Factores de Crecimiento de Fibroblastos/química , Insulina/sangre , Proteínas Klotho , Macaca fascicularis , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Imitación Molecular , Datos de Secuencia Molecular , Obesidad/sangre , Péptidos/farmacocinética , Unión Proteica , Ratas , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/biosíntesis , Albúmina Sérica/farmacocinética , Albúmina Sérica/farmacología , Transducción de Señal , Triglicéridos/sangre
2.
Structure ; 21(3): 493-9, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23473668

RESUMEN

Necroptosis is a cellular mechanism that mediates necrotic cell death. The receptor-interacting serine/threonine protein kinase 1 (RIP1) is an essential upstream signaling molecule in tumor-necrosis-factor-α-induced necroptosis. Necrostatins, a series of small-molecule inhibitors, suppress necroptosis by specifically inhibiting RIP1 kinase activity. Both RIP1 structure and the mechanisms by which necrostatins inhibit RIP1 remain unknown. Here, we report the crystal structures of the RIP1 kinase domain individually bound to necrostatin-1 analog, necrostatin-3 analog, and necrostatin-4. Necrostatin, caged in a hydrophobic pocket between the N- and C-lobes of the kinase domain, stabilizes RIP1 in an inactive conformation through interactions with highly conserved amino acids in the activation loop and the surrounding structural elements. Structural comparison of RIP1 with the inhibitor-bound oncogenic kinase B-RAF reveals partially overlapping binding sites for necrostatin and for the anticancer compound PLX4032. Our study provides a structural basis for RIP1 inhibition by necrostatins and offers insights into potential structure-based drug design.


Asunto(s)
Imidazoles/química , Indoles/química , Simulación del Acoplamiento Molecular , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Baculoviridae/genética , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/genética , Cinética , Mutación , Necrosis , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Relación Estructura-Actividad , Termodinámica
3.
Methods Enzymol ; 524: 105-22, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23498737

RESUMEN

The microtubule (MT) plus-end-tracking proteins (+TIPs) belonging to the end binding (EB) protein family have been studied extensively in the context of cytoplasmic MTs and were shown to play pivotal roles in regulating MT dynamics and in recruiting other +TIPs to MT ends. Early studies in the green alga Chlamydomonas reinhardtii showed that EB1 localizes to the distal flagellum tip and the basal body, and subsequent studies using green fluorescent protein-tagged fusion proteins have demonstrated similar localization of EBs in other ciliated organisms, including mammalian cells as demonstrated here. Functional analysis of EBs in cultured mammalian cells revealed that EB1 and EB3 are required for biogenesis of primary cilia. Although mammalian EB3 localizes to the tip of some cilia and induces cilium elongation, EBs primarily seem to promote ciliogenesis via MT minus-end anchoring at the basal body, in turn facilitating vesicular trafficking to the cilium base. Moreover, mammalian EBs were shown to interact with several proteins implicated in MT minus-end anchoring and/or vesicular trafficking to cilia. Recent work suggests that apart from EBs, additional +TIPs may be present at the distal tip of cilia where they could regulate axoneme assembly, stability, or disassembly.


Asunto(s)
Chlamydomonas reinhardtii/metabolismo , Cilios/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Flagelos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Células Cultivadas , Chlamydomonas reinhardtii/química , Cilios/química , Células Epiteliales/química , Fibroblastos/química , Flagelos/química , Expresión Génica , Proteínas Fluorescentes Verdes/antagonistas & inhibidores , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos , Imagen Molecular , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
4.
FEBS Lett ; 587(4): 311-6, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23313255

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is a common type of cancer. Better understanding of molecular aberrations associated with HNSCC might identify new diagnostic and therapeutic strategies for this disease. In this study, we found ubiquitin-specific protease 4 (USP4) was significantly upregulated in HNSCC. USP4 negatively regulates RIP1-mediated NF-κB activation and promotes TNF-α-induced apoptosis in FaDu cells. USP4 directly interacts with receptor-interacting protein 1 (RIP1) and deubiquitinates K63-linked ubiquitination from RIP1. Therefore, our results indicate that USP4 has tumor suppressor roles in HNSCC and suggest USP4 as a potential therapeutic target for HNSCC.


Asunto(s)
Apoptosis , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Proteínas Ubiquitinadas/metabolismo , Carcinoma de Células Escamosas/enzimología , Línea Celular Tumoral , Regulación hacia Abajo , Silenciador del Gen , Células HEK293 , Neoplasias de Cabeza y Cuello/enzimología , Humanos , Inmunoprecipitación , Lisina/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/genética , Proteasas Ubiquitina-Específicas , Ubiquitinación , Regulación hacia Arriba
5.
Cell Signal ; 25(4): 999-1010, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23275032

RESUMEN

Zinc is an essential micronutrient and cytoprotectant involved in many types of apoptosis. The zinc transporter family SLC30A (ZnTs) is an important factor in the regulation of zinc homeostasis; however, its function in apoptosis in peritoneal mesothelial cells (PMCs) remains unknown. This study explores the regulation of zinc transporters and how they play a role in cell survival, particularly in rat peritoneal mesothelial cells (RPMCs), surrounding glucose concentrations, and the molecular mechanism involved. The messenger RNA (mRNA) transcripts were quantitatively measured by real-time polymerase chain reaction for all known nine zinc transport exporters (SLC30A1-8,10), as well as in primary RPMCs and the cells cultured under nonstimulated and HG-stimulated conditions. While many zinc transporters were constitutively expressed, ZnT5 mRNA and ZnT7 mRNA were strongly induced by HG. Overexpression of ZnT5 and ZnT7 respectively resulted in a decrease in the expression of caspace 3, caspace 8, BAX, and AIF and coincided with cell survival in the presence of HG. Inhibition of ZnT5 and ZnT7 expression using considerable siRNA-mediated knockdown of RPMCs was examined and, afterwards, the impact on cell apoptosis was investigated. Increased levels of apoptosis were observed after knockdown of ZnT5 and ZnT7. Furthermore, overexpression of ZnT5 and ZnT7 is accompanied by activation of PI3K/Akt pathway and inhibiting HG-induced apoptosis. This study suggests that the zinc transporting system in RPMCs is influenced by exposure to HG, particularly ZnT5 and ZnT7. This may account for the inhibition of HG-induced RPMC apoptosis and peritoneum injury, likely through targeting PI3K/Akt pathway-mediated cell survival.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Transporte de Catión/metabolismo , Glucosa/farmacología , Animales , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/genética , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/efectos de los fármacos , Zinc/farmacología
6.
J Mol Neurosci ; 49(3): 507-11, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23065353

RESUMEN

DJ-1 deficiency is a cause of genetic Parkinson's disease (PARK7 PD). In sporadic Parkinson's disease (PD), however, DJ-1 is abundantly expressed in reactive astrocytes. This may represent a compensatory protective response. In initial support of this hypothesis, we have shown in vitro that DJ-1-overexpressing astrocytes protect neurons against rotenone-induced death. Rotenone, a pesticide linked to increased PD risk, can stimulate oxidative stress. This process is implicated in PD pathogenesis. Since DJ-1 can enhance antioxidant systems, we hypothesized that augmenting its expression in astrocytes would protect cocultured neurons against oxidative stress. We report here that DJ-1-overexpressing astrocytes were significantly more protective against rotenone-induced neuronal thiol oxidation than wild-type astrocytes in neuron-astrocyte cocultures. DJ-1-knockdown astrocytes, on the other hand, were significantly impaired in their capacity to protect neurons against thiol oxidation. Each of these findings was replicated using astrocyte-conditioned media on neuron-enriched cultures. Thus, DJ-1-modulated, astrocyte-released soluble factors must be involved in the mechanism. This is the first demonstration that the manipulation of a PD-causing gene in astrocytes affects their ability to protect neurons against oxidative stress.


Asunto(s)
Astrocitos/fisiología , Regulación de la Expresión Génica , Neuronas/efectos de los fármacos , Proteínas Oncogénicas/fisiología , Estrés Oxidativo , Animales , Células Cultivadas/metabolismo , Corteza Cerebral/citología , Técnicas de Cocultivo , Ratones , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Enfermedad de Parkinson/metabolismo , Peroxirredoxinas , Proteína Desglicasa DJ-1 , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/fisiología , Compuestos de Sulfhidrilo/toxicidad , Transfección
7.
PLoS One ; 7(10): e48537, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23119051

RESUMEN

Tannerella forsythia is a gram-negative bacteria, which is strongly associated with the development of periodontal disease. Karilysin is a newly identified metalloprotease-like enzyme, that is secreted from T. forsythia. Karilysin modulates the host immune response and is therefore considered a likely drug target. In this study peptides were selected towards the catalytic domain from Karilysin (Kly18) by phage display. The peptides were linear with low micromolar binding affinities. The two best binders (peptide14 and peptide15), shared the consensus sequence XWFPXXXGGG. A peptide15 fusion with Maltose Binding protein (MBP) was produced with peptide15 fused to the N-terminus of MBP. The peptide15-MBP was expressed in E. coli and the purified fusion-protein was used to verify Kly18 specific binding. Chemically synthesised peptide15 (SWFPLRSGGG) could inhibit the enzymatic activity of both Kly18 and intact Karilysin (Kly48). Furthermore, peptide15 could slow down the autoprocessing of intact Kly48 to Kly18. The WFP motif was important for inhibition and a truncation study further demonstrated that the N-terminal serine was also essential for Kly18 inhibition. The SWFP peptide had a Ki value in the low micromolar range, which was similar to the intact peptide15. In conclusion SWFP is the first reported inhibitor of Karilysin and can be used as a valuable tool in structure-function studies of Karilysin.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Bacteroidetes/enzimología , Inhibidores Enzimáticos/farmacología , Péptidos/farmacología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Inhibidores Enzimáticos/química , Cinesis , Metaloproteinasas de la Matriz/química , Metaloproteinasas de la Matriz/metabolismo , Modelos Moleculares , Biblioteca de Péptidos , Péptidos/química , Unión Proteica , Conformación Proteica , Proteolisis/efectos de los fármacos , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Solubilidad
8.
PLoS One ; 7(9): e45412, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23049798

RESUMEN

BACKGROUND: COMMD7 is a newly identified gene overexpressed in hepatocellular carcinoma (HCC) and associated with tumor invasion and poor prognosis. We aim to examine the biological function of COMMD7 in HCC by shRNA silencing. METHODS: COMMD7 expressions were examined in human HCC cell lines HepG2, Huh7, Hep3B, HLE, HLF, SK-Hep-1 and PLC/PRF/5 cells. Recombinant pGenesil-COMMD7-shRNA was transfected into COMMD7-abundant HepG2 cells to silence COMMD7 expression. The effects of COMMD7 silencing on HepG2 cell proliferation in vitro and xenograft tumor growth in vivo were evaluated. Flow cytometry profiling was used to detect the presence of apoptosis in COMMD7-silenced HepG2 cells and to differentiate cell cycle distribution. Electrophoretic mobility shift assay and luciferase reporter assays to examine the activities of nuclear factor-kappaB (NF-κB) signaling pathways in response to tumor necrosis factor (TNF)-α in COMMD7-silenced HepG2 cells. RESULTS: COMMD7 expression level was abundance in HepG2 and SK-Hep-1 cells. COMMD7 was aberrantly overexpressed in HepG2 cells, whilst pGenesil-COMMD7-shRNA exhibited a maximal inhibition rate of 75%. COMMD7 silencing significantly reduced HepG2 cell proliferation and colony formation. The knockdown of COMMD7 resulted in an increased apoptosis and cell cycle arrest at S-phase. COMMD7 knockdown also exhibited an antineoplastic effect in vivo, which manifested as tumor xenograft growth retardation. COMMD7 silencing also suppressed the responsiveness of NF-κB signaling pathway to the stimulation with TNF-α in vitro. Moreover, the similar suppressive effects of COMMD7 silence on SK-Hep-1 cells were also observed. CONCLUSIONS: COMMD7 contributes to HCC progression by reducing cell apoptosis and overcoming cell cycle arrest. The proliferative and antiapoptotic effects of COMMD7 may be mediated by NF-κB signaling pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , ARN Interferente Pequeño/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Trasplante de Neoplasias , Plásmidos , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Carga Tumoral , Factor de Necrosis Tumoral alfa/farmacología
9.
Protein Expr Purif ; 86(2): 120-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23036358

RESUMEN

Glucosamine 6-phosphate N-acetyltransferase (GNA1; EC 2.3.1.4) is required for the de novo synthesis of N-acetyl-d-glucosamine-6-phosphate (GlcNAc-6P), which is an essential precursor in Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) biosynthesis pathway. Therefore, GNA1 is indispensable for the viability of organisms. Here, a novel cell-free expression strategy was developed to efficiently produce large amounts of human GNA1(HsGNA1) and HsGNA1-sGFP for throughput inhibitor screening. The binding site of inhibitor glucose-6-phosphate (G6P) to hGNA was identified by simulated annealing. Subtle differences to the binding site of Aspergillius GNA1(AfGNA1) can be harnessed for inhibitor design. HsGNA1 may be also useful as an antimicrobial and chemotherapeutic target against cancer. Additionally HsGNA1 inhibitors/modulators can possibly be administered with other drugs in the next generation of personalized medicine.


Asunto(s)
Sistema Libre de Células/metabolismo , Glucosamina 6-Fosfato N-Acetiltransferasa/antagonistas & inhibidores , Glucosamina 6-Fosfato N-Acetiltransferasa/biosíntesis , Secuencia de Aminoácidos , Sitios de Unión , Biotecnología/métodos , Glucosamina 6-Fosfato N-Acetiltransferasa/química , Glucosamina 6-Fosfato N-Acetiltransferasa/genética , Glucosa-6-Fosfato/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Alineación de Secuencia
10.
Eur J Med Chem ; 57: 1-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23041456

RESUMEN

Protein kinases are widely recognized as important therapeutic targets due to their involvement in signal transduction pathways. These pathways are tightly controlled and regulated, notably by the ability of kinases to selectively phosphorylate a defined set of substrates. A wide variety of disorders can arise as a consequence of abnormal kinase-mediated phosphorylation and numerous kinase inhibitors have earned their place as key components of the modern pharmacopeia. Although "traditional" kinase inhibitors typically act by preventing the interaction between the kinase and ATP, thus stopping substrate phosphorylation, an alternative approach consists in disrupting the protein-protein interaction between the kinase and its downstream partners. In order to facilitate the identification of potential chemical starting points for substrate-site inhibition approaches, we desired to investigate the application of Substrate Activity Screening to kinases. We herein report a proof-of-concept study demonstrating, on a model tyrosine kinase, that the key requirements of this methodology can be met. Namely, using peptides as model substrates, we show that a simple ADP-accumulation assay can be used to monitor substrate efficiency and that efficiency can be optimized in a modular manner. More importantly, we demonstrate that structure-efficiency relationships translate into structure-activity relationships upon conversion of the substrates into inhibitors.


Asunto(s)
Péptidos/química , Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Quinasas Receptoras/química , Adenosina Trifosfato/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Péptidos/antagonistas & inhibidores , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Relación Estructura-Actividad , Especificidad por Sustrato
11.
Diabetes ; 61(11): 2922-31, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22872237

RESUMEN

Congenital generalized lipodystrophy (CGL), secondary to AGPAT2 mutation is characterized by the absence of adipocytes and development of severe insulin resistance. In the current study, we investigated the adipogenic defect associated with AGPAT2 mutations. Adipogenesis was studied in muscle-derived multipotent cells (MDMCs) isolated from vastus lateralis biopsies obtained from controls and subjects harboring AGPAT2 mutations and in 3T3-L1 preadipocytes after knockdown or overexpression of AGPAT2. We demonstrate an adipogenic defect using MDMCs from control and CGL human subjects with mutated AGPAT2. This defect was rescued in CGL MDMCs with a retrovirus expressing AGPAT2. Both CGL-derived MDMCs and 3T3-L1 cells with knockdown of AGPAT2 demonstrated an increase in cell death after induction of adipogenesis. Lack of AGPAT2 activity reduces Akt activation, and overexpression of constitutively active Akt can partially restore lipogenesis. AGPAT2 modulated the levels of phosphatidic acid, lysophosphatidic acid, phosphatidylinositol species, as well as the peroxisome proliferator-activated receptor γ (PPARγ) inhibitor cyclic phosphatidic acid. The PPARγ agonist pioglitazone partially rescued the adipogenic defect in CGL cells. We conclude that AGPAT2 regulates adipogenesis through the modulation of the lipome, altering normal activation of phosphatidylinositol 3-kinase (PI3K)/Akt and PPARγ pathways in the early stages of adipogenesis.


Asunto(s)
Aciltransferasas/metabolismo , Lipodistrofia Generalizada Congénita/genética , Lipodistrofia Generalizada Congénita/metabolismo , PPAR gamma/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células 3T3-L1 , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/genética , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis , Animales , Células Cultivadas , Humanos , Metabolismo de los Lípidos , Lipodistrofia Generalizada Congénita/patología , Ratones , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/patología , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa/genética , Proteínas Proto-Oncogénicas c-akt/genética , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo
12.
FEBS Lett ; 586(19): 3208-14, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22884421

RESUMEN

Myosin-6 is an actin-based motor protein that moves its cargo towards the minus-end of actin filaments. Mutations in the gene encoding the myosin-6 heavy chain and changes in the cellular abundance of the protein have been linked to hypertrophic cardiomyopathy, neurodegenerative diseases, and cancer. Here, we present a detailed kinetic characterization of the human myosin-6 motor domain, describe the effect of 2,4,6-triiodophenol on the interaction of myosin-6 with F-actin and nucleotides, and show how addition of the drug reduces the number of myosin-6-dependent vesicle fusion events at the plasma membrane during constitutive secretion.


Asunto(s)
Cadenas Pesadas de Miosina/antagonistas & inhibidores , Cadenas Pesadas de Miosina/metabolismo , Actinas/metabolismo , N-Acetiltransferasa de Aminoácidos , Animales , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Técnicas In Vitro , Cinética , Modelos Biológicos , Modelos Moleculares , Proteínas Motoras Moleculares/antagonistas & inhibidores , Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Mutagénesis Sitio-Dirigida , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Fenoles/farmacología , Dominios y Motivos de Interacción de Proteínas , ARN Interferente Pequeño/genética , Conejos , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
13.
Cancer Lett ; 325(2): 139-46, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-22781393

RESUMEN

This study focuses on determining whether the combination of NYP-BKM120 (BKM120) and RAD001 exerts enhanced therapeutic effect against lung cancer. The combination of BKM120 and RAD001 exerted synergistic inhibitory effects on the growth of lung cancer cells both in culture and in mouse xenograft model. This combination abrogated RAD001-induced Akt phosphorylation and exerted enhanced suppressive effect on 4EBP1 phosphorylation. Collectively, we suggest that the combination of RAD001 and BKM120 may be an effective regimen for treatment of lung cancer, hence warranting further evaluation of the combination in the clinic.


Asunto(s)
Aminopiridinas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inmunosupresores/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Morfolinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Sirolimus/análogos & derivados , Aminopiridinas/administración & dosificación , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Medio de Cultivo Libre de Suero , Sinergismo Farmacológico , Everolimus , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunosupresores/administración & dosificación , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Morfolinas/administración & dosificación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Hematol Oncol ; 5: 39, 2012 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-22800464

RESUMEN

BACKGROUND: Gain-of-function mutations of tyrosine kinase FLT3 are frequently found in acute myeloid leukemia (AML). This has made FLT3 an important marker for disease diagnosis and a highly attractive target for therapeutic drug development. This study is intended to generate a sensitive substrate for assays of the FLT3 enzymatic activity. METHODS: We expressed in Escherichia coli cells a glutathione S-transferase (GST) fusion protein designated GST-FLT3S, which contains a peptide sequence derived from an autophosphorylation site of FLT3. The protein was used to analyze tyrosine kinase activity of baculovirus-expressed FLT3 and crude cell extracts of bone marrow cells from AML patients. It was also employed to perform FLT3 kinase assays for FLT3 inhibitor screening. RESULTS: GST-FLT3S in solution or on beads was strongly phosphorylated by recombinant proteins carrying the catalytic domain of wild type FLT3 and FLT3D835 mutants, with the latter exhibiting much higher activity and efficiency. GST-FLT3S was also able to detect elevated tyrosine kinase activity in bone marrow cell extracts from AML patients. A small-scale inhibitor screening led to identification of several potent inhibitors of wild type and mutant forms of FLT3. CONCLUSIONS: GST-FLT3S is a sensitive protein substrate for FLT3 assays. It may find applications in diagnosis of diseases related to abnormal FLT3 activity and in inhibitor screening for drug development.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Leucemia Mieloide Aguda/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Médula Ósea/metabolismo , Médula Ósea/patología , Evaluación Preclínica de Medicamentos , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación/genética , Fosforilación/efectos de los fármacos , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Especificidad por Sustrato , Células Tumorales Cultivadas , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética
15.
Biochem J ; 446(2): 203-12, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22671145

RESUMEN

A critical cis-regulatory element for the CFTR (cystic fibrosis transmembrane conductance regulator) gene is located in intron 11, 100 kb distal to the promoter, with which it interacts. This sequence contains an intestine-selective enhancer and associates with enhancer signature proteins, such as p300, in addition to tissue-specific TFs (transcription factors). In the present study we identify critical TFs that are recruited to this element and demonstrate their importance in regulating CFTR expression. In vitro DNase I footprinting and EMSAs (electrophoretic mobility-shift assays) identified four cell-type-selective regions that bound TFs in vitro. ChIP (chromatin immunoprecipitation) identified FOXA1/A2 (forkhead box A1/A2), HNF1 (hepatocyte nuclear factor 1) and CDX2 (caudal-type homeobox 2) as in vivo trans-interacting factors. Mutation of their binding sites in the intron 11 core compromised its enhancer activity when measured by reporter gene assay. Moreover, siRNA (small interfering RNA)-mediated knockdown of CDX2 caused a significant reduction in endogenous CFTR transcription in intestinal cells, suggesting that this factor is critical for the maintenance of high levels of CFTR expression in these cells. The ChIP data also demonstrate that these TFs interact with multiple cis-regulatory elements across the CFTR locus, implicating a more global role in intestinal expression of the gene.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Elementos de Facilitación Genéticos , Enterocitos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Transactivadores/metabolismo , Factor de Transcripción CDX2 , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Genes Reporteros , Factor Nuclear 1-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 1-alfa del Hepatocito/química , Factor Nuclear 1-alfa del Hepatocito/genética , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Humanos , Intrones , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/química , Transactivadores/genética
16.
Neuropharmacology ; 63(4): 538-46, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22580377

RESUMEN

Drug development for nicotinic acetylcholine receptors (nAChR) is challenged by subtype diversity arising from variations in subunit composition. On-target activity for neuronal heteromeric receptors is typically associated with CNS receptors that contain α4 and other subunits, while off-target activity could be associated with ganglionic-type receptors containing α3ß4 binding sites and other subunits, including ß4, ß2, α5, or α3 as a structural subunit in the pentamer. Additional interest in α3 ß4 α5-containing receptors arises from genome-wide association studies linking these genes, and a single nucleotide polymorphism (SNP) in α5 in particular, to lung cancer and heavy smoking. While α3 and ß4 readily form receptors in expression system such as the Xenopus oocyte, since α5 is not required for function, simple co-expression approaches may under-represent α5-containing receptors. We used a concatamer of human α3 and ß4 subunits to form ligand-binding domains, and show that we can force the insertions of alternative structural subunits into the functional pentamers. These α3ß4 variants differ in sensitivity to ACh, nicotine, varenicline, and cytisine. Our data indicated lower efficacy for varenicline and cytisine than expected for ß4-containing receptors, based on previous studies of rodent receptors. We confirm that these therapeutically important α4 receptor partial agonists may present different autonomic-based side-effect profiles in humans than will be seen in rodent models, with varenicline being more potent for human than rat receptors and cytisine less potent. Our initial characterizations failed to find functional effects of the α5 SNP. However, our data validate this approach for further investigations.


Asunto(s)
Acetilcolina/metabolismo , Ganglios/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Agonistas Nicotínicos/metabolismo , Antagonistas Nicotínicos/metabolismo , Subunidades de Proteína/metabolismo , Receptores Nicotínicos/metabolismo , Acetilcolina/agonistas , Acetilcolina/antagonistas & inhibidores , Alcaloides/metabolismo , Alcaloides/farmacología , Animales , Azocinas/metabolismo , Azocinas/farmacología , Benzazepinas/metabolismo , Benzazepinas/farmacología , Agonismo Parcial de Drogas , Potenciales Evocados/efectos de los fármacos , Humanos , Ligandos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/agonistas , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Nicotina/agonistas , Nicotina/antagonistas & inhibidores , Nicotina/metabolismo , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Subunidades de Proteína/agonistas , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/genética , Quinolizinas/metabolismo , Quinolizinas/farmacología , Quinoxalinas/metabolismo , Quinoxalinas/farmacología , Ratas , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Proteínas Recombinantes de Fusión/agonistas , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo , Especificidad de la Especie , Vareniclina , Xenopus laevis
17.
Protein Pept Lett ; 19(5): 485-91, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22486643

RESUMEN

The Janus kinase (JAK) family consists of four members: JAK-1, -2, -3 and tyrosine kinase 2 (TYK-2). Recent work suggests that cytokine signaling through TYK-2 may play a critical role in a number of inflammatory processes. We recently described the purification and characterization of phosphorylated isoforms of the TYK-2 kinase domain (TYK-2 KD) and its high resolution 3D structure in the presence of inhibitors. We now report the expression and a two-step purification procedure for the doubly tagged full-length construct, H6-FL-TYK-2-FLAG, and examine its properties compared to those of TYK-2 KD. In the presence of ATP and a peptide substrate, H6-FL-TYK-2-FLAG showed a marked lag in phosphopeptide product formation, while TYK-2 KD showed no such lag. This lag could be eliminated by ATP pretreatment, suggesting that the H6-FL-TYK-2-FLAG enzyme was activated by phosphorylation. The potencies of several nanomolar inhibitors were similar for TYK-2 KD and H6-FL-TYK-2-FLAG. However, these same inhibitors were about 1000 times less potent inhibiting the autophosphorylation of H6-FL-TYK-2-FLAG than they were inhibiting the phosphorylation of a peptide substrate modeled after the activation loop sequence of TYK-2. This intriguing result suggests that autophosphorylation and, thus, activation of H6-FL-TYK-2-FLAG is relatively insensitive to inhibition and that present inhibitors act to inhibit TYK-2 subsequent to activation. Inhibition of TYK-2 autophosphorylation may represent a new area of investigation for the JAK family.


Asunto(s)
TYK2 Quinasa/antagonistas & inhibidores , TYK2 Quinasa/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Dominio Catalítico , Electroforesis en Gel de Poliacrilamida , Histidina/química , Humanos , Cinética , Oligopéptidos/química , Fosforilación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , TYK2 Quinasa/química , TYK2 Quinasa/aislamiento & purificación
18.
Blood ; 119(18): 4253-63, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22411871

RESUMEN

Chronic myeloid leukemia in chronic phase (CML-CP) is induced by BCR-ABL1 oncogenic tyrosine kinase. Tyrosine kinase inhibitors eliminate the bulk of CML-CP cells, but fail to eradicate leukemia stem cells (LSCs) and leukemia progenitor cells (LPCs) displaying innate and acquired resistance, respectively. These cells may accumulate genomic instability, leading to disease relapse and/or malignant progression to a fatal blast phase. In the present study, we show that Rac2 GTPase alters mitochondrial membrane potential and electron flow through the mitochondrial respiratory chain complex III (MRC-cIII), thereby generating high levels of reactive oxygen species (ROS) in CML-CP LSCs and primitive LPCs. MRC-cIII-generated ROS promote oxidative DNA damage to trigger genomic instability, resulting in an accumulation of chromosomal aberrations and tyrosine kinase inhibitor-resistant BCR-ABL1 mutants. JAK2(V617F) and FLT3(ITD)-positive polycythemia vera cells and acute myeloid leukemia cells also produce ROS via MRC-cIII. In the present study, inhibition of Rac2 by genetic deletion or a small-molecule inhibitor and down-regulation of mitochondrial ROS by disruption of MRC-cIII, expression of mitochondria-targeted catalase, or addition of ROS-scavenging mitochondria-targeted peptide aptamer reduced genomic instability. We postulate that the Rac2-MRC-cIII pathway triggers ROS-mediated genomic instability in LSCs and primitive LPCs, which could be targeted to prevent the relapse and malignant progression of CML.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Inestabilidad Genómica , Leucemia Mieloide de Fase Crónica/patología , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al GTP rac/fisiología , Animales , Catalasa/metabolismo , Daño del ADN , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Progresión de la Enfermedad , Transporte de Electrón , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Potencial de la Membrana Mitocondrial , Metacrilatos/farmacología , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Policitemia Vera/metabolismo , Policitemia Vera/patología , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/fisiología , Superóxido Dismutasa/metabolismo , Tiazoles/farmacología , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/genética , Proteína RCA2 de Unión a GTP
19.
Cell Res ; 22(5): 873-85, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22410793

RESUMEN

The serine/threonine kinase Akt functions in multiple cellular processes, including cell survival and tumor development. Studies of the mechanisms that negatively regulate Akt have focused on dephosphorylation-mediated inactivation. In this study, we identified a negative regulator of Akt, MULAN, which possesses both a RING finger domain and E3 ubiquitin ligase activity. Akt was found to directly interact with MULAN and to be ubiquitinated by MULAN in vitro and in vivo. Other molecular assays demonstrated that phosphorylated Akt is a substantive target for both interaction with MULAN and ubiquitination by MULAN. The results of the functional studies suggest that the degradation of Akt by MULAN suppresses cell proliferation and viability. These data provide insight into the Akt ubiquitination signaling network.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proliferación Celular , Supervivencia Celular , Células HEK293 , Células HeLa , Humanos , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
20.
J Biol Chem ; 287(16): 13228-38, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22383527

RESUMEN

The Rho GTPase Rac regulates actin cytoskeleton reorganization to form cell surface extensions (lamellipodia) required for cell migration/invasion during cancer metastasis. Rac hyperactivation and overexpression are associated with aggressive cancers; thus, interference of the interaction of Rac with its direct upstream activators, guanine nucleotide exchange factors (GEFs), is a viable strategy for inhibiting Rac activity. We synthesized EHop-016, a novel inhibitor of Rac activity, based on the structure of the established Rac/Rac GEF inhibitor NSC23766. Herein, we demonstrate that EHop-016 inhibits Rac activity in the MDA-MB-435 metastatic cancer cells that overexpress Rac and exhibits high endogenous Rac activity. The IC(50) of 1.1 µM for Rac inhibition by EHop-016 is ∼100-fold lower than for NSC23766. EHop-016 is specific for Rac1 and Rac3 at concentrations of ≤5 µM. At higher concentrations, EHop-016 inhibits the close homolog Cdc42. In MDA-MB-435 cells that demonstrate high active levels of the Rac GEF Vav2, EHop-016 inhibits the association of Vav2 with a nucleotide-free Rac1(G15A), which has a high affinity for activated GEFs. EHop-016 also inhibits the Rac activity of MDA-MB-231 metastatic breast cancer cells and reduces Rac-directed lamellipodia formation in both cell lines. EHop-016 decreases Rac downstream effects of PAK1 (p21-activated kinase 1) activity and directed migration of metastatic cancer cells. Moreover, at effective concentrations (<5 µM), EHop-016 does not affect the viability of transformed mammary epithelial cells (MCF-10A) and reduces viability of MDA-MB-435 cells by only 20%. Therefore, EHop-016 holds promise as a targeted therapeutic agent for the treatment of metastatic cancers with high Rac activity.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carbazoles/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Pirimidinas/farmacología , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Aminoquinolinas/farmacología , Sitios de Unión/efectos de los fármacos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Carbazoles/síntesis química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Femenino , Humanos , Pirimidinas/síntesis química , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/genética , Proteínas de Unión al GTP rho/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA