Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(25): e2218896120, 2023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-37327313

RESUMEN

Programmed ferroptotic death eliminates cells in all major organs and tissues with imbalanced redox metabolism due to overwhelming iron-catalyzed lipid peroxidation under insufficient control by thiols (Glutathione (GSH)). Ferroptosis has been associated with the pathogenesis of major chronic degenerative diseases and acute injuries of the brain, cardiovascular system, liver, kidneys, and other organs, and its manipulation offers a promising new strategy for anticancer therapy. This explains the high interest in designing new small-molecule-specific inhibitors against ferroptosis. Given the role of 15-lipoxygenase (15LOX) association with phosphatidylethanolamine (PE)-binding protein 1 (PEBP1) in initiating ferroptosis-specific peroxidation of polyunsaturated PE, we propose a strategy of discovering antiferroptotic agents as inhibitors of the 15LOX/PEBP1 catalytic complex rather than 15LOX alone. Here we designed, synthesized, and tested a customized library of 26 compounds using biochemical, molecular, and cell biology models along with redox lipidomic and computational analyses. We selected two lead compounds, FerroLOXIN-1 and 2, which effectively suppressed ferroptosis in vitro and in vivo without affecting the biosynthesis of pro-/anti-inflammatory lipid mediators in vivo. The effectiveness of these lead compounds is not due to radical scavenging or iron-chelation but results from their specific mechanisms of interaction with the 15LOX-2/PEBP1 complex, which either alters the binding pose of the substrate [eicosatetraenoyl-PE (ETE-PE)] in a nonproductive way or blocks the predominant oxygen channel thus preventing the catalysis of ETE-PE peroxidation. Our successful strategy may be adapted to the design of additional chemical libraries to reveal new ferroptosis-targeting therapeutic modalities.


Asunto(s)
Ferroptosis , Proteínas de Unión a Fosfatidiletanolamina , Glutatión/metabolismo , Hierro/metabolismo , Peroxidación de Lípido , Lípidos , Oxidación-Reducción , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores
2.
Dig Dis Sci ; 64(9): 2570-2580, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30874989

RESUMEN

BACKGROUND AND AIMS: Liver fibrosis is featured with excessive deposition of extracellular matrix and fibrous connective tissue hyperplasia. The specific inhibitor of Raf-1 kinase inhibitor protein, locostatin, inhibits the migration of hepatic stellate cells. In this study, we investigated the effect of locostatin on liver fibrosis and its underlying mechanism. METHODS: Carbon tetrachloride (CCl4) was used to induce liver fibrosis in mice, and locostatin was injected intraperitoneally. Liver fibrosis was assessed by Masson and Sirius red staining, hydroxyproline (HYP) assay, and collagen percentage area. Collagen I, collagen III, and α-SMA were detected by RT-PCR and western blot. The levels of MMP-13, MMP-2, TIMP-1, and TIMP-2 were estimated by ELISA. Liver inflammation was evaluated by HE staining and immunohistochemistry; liver myeloperoxidase (MPO), superoxide dismutase, and malondialdehyde were measured by ELISA; and cytokines were by Mouse Cytokine Array Q4000. RESULTS: Compared to the CCl4 group, HYP (208.56 ± 6.12) µg/g, percentage of total collagen at overall region (1.91 ± 0.13), MMP-13/TIMP-1 (0.19 ± 0.01), MPO (1.45 ± 0.04) U/g, TGF-ß (2652 ± 91.20), PDGF-AA (3897 ± 290.69), and E-selectin (1569 ± 66.48) in the liver tissues were decreased significantly in the locostatin-treated group. CONCLUSIONS: Locostatin mitigated liver fibrosis and inflammation induced by CCl4. The mechanism is via inhibition inflammatory cytokines, TGF-ß, PDGF-AA, and E-selectin.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Oxazolidinonas/uso terapéutico , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Actinas/genética , Actinas/metabolismo , Animales , Tetracloruro de Carbono , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Selectina E/metabolismo , Células Estrelladas Hepáticas/fisiología , Hidroxiprolina/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Masculino , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Oxazolidinonas/farmacología , Peroxidasa/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , ARN Mensajero/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
J Pharmacol Exp Ther ; 368(3): 446-461, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30622172

RESUMEN

Targeted inhibition of RAF and MEK by molecularly targeted agents has been employed as a strategy to block aberrant mitogen-activated protein kinase (MAPK) signaling in melanoma. While the use of BRAF and MEK inhibitors, either as a single agent or in combination, improved efficacy in BRAF-mutant melanoma, initial responses are often followed by relapse due to acquired resistance. Moreover, some BRAF inhibitors are associated with paradoxical activation of the MAPK pathway, causing the development of secondary malignancies. The use of panRAF inhibitors, i.e., those that target all isoforms of RAF, may overcome paradoxical activation and resistance. The purpose of this study was to perform a quantitative assessment and evaluation of the influence of efflux mechanisms at the blood-brain barrier (BBB), in particular, Abcb1/P-glycoprotein (P-gp) and Abcg2/breast cancer resistance protein (Bcrp), on the brain distribution of three panRAF inhibitors: CCT196969 [1-(3-(tert-butyl)-1-phenyl-1H-pyrazol-5-yl)-3-(2-fluoro-4-((3-oxo-3,4-dihydropyrido[2,3-b]pyrazin-8-yl)oxy)phenyl)urea], LY3009120 1-(3,3-Dimethylbutyl)-3-(2-fluoro-4-methyl-5-(7-methyl-2-(methylamino)pyrido(2,3-d)pyrimidin-6-yl)phenyl)urea, and MLN2480 [4-pyrimidinecarboxamide, 6-amino-5-chloro-N-[(1R)-1-[5-[[[5-chloro-4-(trifluoromethyl)-2-pyridinyl]amino]carbonyl]-2-thiazolyl]ethyl]-]. In vitro studies using transfected Madin-Darby canine kidney II cells indicate that only LY3009120 and MLN2480 are substrates of Bcrp, and none of the three inhibitors are substrates of P-gp. The three panRAF inhibitors show high nonspecific binding in brain and plasma. In vivo studies in mice show that the brain distribution of CCT196969, LY3009120, and MLN2480 is limited, and is enhanced in transgenic mice lacking P-gp and Bcrp. While MLN2480 has a higher brain distribution, LY3009120 exhibits superior in vitro efficacy in patient-derived melanoma cell lines. The delivery of a drug to the site of action residing behind a functionally intact BBB, along with drug potency against the target, collectively play a critical role in determining in vivo efficacy outcomes.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Compuestos Heterocíclicos con 3 Anillos/metabolismo , Melanoma/metabolismo , Compuestos de Fenilurea/metabolismo , Pirazinas/metabolismo , Pirimidinas/metabolismo , Animales , Encéfalo/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Perros , Relación Dosis-Respuesta a Droga , Femenino , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Humanos , Células de Riñón Canino Madin Darby , Masculino , Melanoma/tratamiento farmacológico , Ratones , Ratones Noqueados , Compuestos de Fenilurea/administración & dosificación , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Pirazinas/administración & dosificación , Pirimidinas/administración & dosificación
4.
Leuk Lymphoma ; 59(12): 2917-2928, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29911936

RESUMEN

The Raf-1 kinase inhibitory protein (RKIP) is an important regulatory element in multiple signaling pathways, including MAPK-ERK1/2. We investigated whether targeted disruption of RKIP is a therapeutic option for chronic lymphocytic leukemia (CLL). The RKIP inhibitor locostatin-induced apoptosis of CLL cells, irrespective of poor prognostic indications or treatment history. Locostatin down-regulated MAPK-ERK1/2 and AKT phosphorylation, decreased expression of the chemokine receptor CXCR4 (p = .04) and reduced the migratory capacity of CLL cells toward stroma-derived factor 1α (SDF-1α, p = .02). Immuno-blotting and immuno-precipitation showed that RKIP is constitutively phosphorylated and highly expressed in CLL cells and that the actions of locostatin may be mediated by binding of G-protein receptor kinase-2 (GRK2) to MEK1 and AKT. Collectively, our data suggest that inhibition of RKIP may be effective against CLL, reducing the survival and migratory capacity of the leukemic cells through down-regulation of MAPK-ERK1/2 and AKT-mediated signaling.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Oxazolidinonas/farmacología , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Linfocítica Crónica de Células B/sangre , Leucemia Linfocítica Crónica de Células B/patología , Leucocitos Mononucleares , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Oxazolidinonas/uso terapéutico , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células
5.
Int J Cancer ; 142(11): 2323-2334, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29315556

RESUMEN

Oncogenic KRAS plays a crucial role in pancreatic ductal adenocarcinoma (PDAC) development and progression. However, the mechanism has not been clearly elucidated. RKIP is a tumor repressor, and loss of RKIP has been shown in PDAC. Here, we found that KRAS expression was inversely correlated with RKIP expression in PDAC fresh tissue regardless of the KRAS mutant status. The negative correlation between KRAS and RKIP was further confirmed in our PDAC tissue microarray. KRAS overexpression and RKIP downregulation were associated with poor clinical outcomes. Knockdown or overexpression of KRAS in PDAC cell lines robustly increased or decreased, respectively, RKIP protein and mRNA levels. Furthermore, the MAPK-ERK pathway was involved in the regulation of RKIP. KRAS-regulated RKIP expression, which in turn affected the expression of pivotal epithelial-mesenchymal transition (EMT) and apoptosis factors. The biological function of the KRAS-RKIP axis was demonstrated in human pancreatic cancer cells in vitro and in vivo. KRAS knockdown increased RKIP expression and inhibited metastasis and chemoresistance. Moreover, the feature of metastasis and chemoresistance was rescued in the KRAS-knockdown cells through the inhibition of RKIP by RNA interference. In conclusion, our studies demonstrate how KRAS inhibits the tumor suppressor RKIP, thus offering novel justification for targeting RKIP as a strategy to overcome KRAS-induced tumor metastasis and chemoresistance in PDAC.


Asunto(s)
Resistencia a Antineoplásicos/genética , Sistema de Señalización de MAP Quinasas , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Inmunohistoquímica , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
6.
Int Immunopharmacol ; 42: 130-138, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27912149

RESUMEN

A flavone was isolated from Origanum vulgare and identified as didymin (O. vulgare didymin, OVD). The protective effect and mechanism of OVD on acute liver injury was then assessed in vivo and in vitro. Our results showed that OVD significantly alleviated CCl4-induced liver injury in mice and markedly decreased serum ALT and AST activities. OVD treatment significantly reduced CYP2E1 activity, lipid peroxidation level, ROS generation, NO production and pro-inflammatory cytokines (such as TNF-α, IL-6 and IL-1ß) in liver tissues and RAW 264.7 cells, but enhanced the hepatic antioxidative enzymes activities. Further study showed that OVD significantly inhibited the NF-κB and MAPK pathways. Interestingly, OVD notably enhanced Raf kinase inhibitor protein (RKIP) expression, and the effects of OVD on histological changes, oxidative stress and inflammation was largely abolished by the RKIP specific inhibitor locostatin. Our findings indicate that OVD can ameliorate CCl4-induced liver injury, which may be ascribed to its radical scavenging action, antioxidant activity, and modulation of MAPK and NF-κB signaling pathways.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Flavonoides/uso terapéutico , Glicósidos/uso terapéutico , Origanum , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Tetracloruro de Carbono , Citocromo P-450 CYP2E1/metabolismo , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Oxazolidinonas/farmacología , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo
7.
Eur J Med Chem ; 127: 997-1011, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27839788

RESUMEN

Beginning with our previously reported ERK inhibitor BL-EI001, we found Raf1 to be an important regulator in the ERK interactive network, and then we designed and synthesized a novel series of Raf1/ERK dual inhibitors against human breast cancers through integrative computational, synthetic and biological screening methods. Moreover, we found that compound 9d suppressed the proliferation of breast cancer cell lines and induced cellular apoptosis via a mitochondrial pathway with only partial dependence on Raf1 and ERK. Our results suggest that an integrative method including in silico design, chemical synthesis, biological screening and bioinformatics analysis could be an attractive strategy for the discovery of multi-target inhibitors against breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Biología Computacional , Descubrimiento de Drogas/métodos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Diseño de Fármacos , Quinasas MAP Reguladas por Señal Extracelular/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Células MCF-7 , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Simulación del Acoplamiento Molecular , Proteínas de Unión a Fosfatidiletanolamina/química , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Dominios Proteicos , Homología de Secuencia de Aminoácido
8.
World J Surg Oncol ; 14(1): 221, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27553494

RESUMEN

BACKGROUND: Human phosphatidylethanolamine-binding protein 4 (hPEBP4) is a well-established antiapoptosis molecule in recent years. It has also been demonstrated to be involved in the radioresistance of rectal cancer. The objective of this study was to determine whether IOI-42, a chemical inhibitor of hPEBP4, could sensitize rectal cancer cells. METHODS: Rectal cancer cells were treated with IOI-42 alone or in combination with irradiation. Clonogenic survival assays and tumor volume growth analysis were used, respectively, to study the effect of IOI-42 in vitro and in vivo. Western blot was adopted to measure the activation of signal pathway. RESULTS: Clonogenic survival assays showed that IOI-42, combined with irradiation, caused a significant decrease in colony formation compared with radiation alone, which was associated with the downregulation of Akt activation. And we also confirmed the effect of IOI-42 in nude mice transplanted with human rectal cancer subcutaneously. CONCLUSIONS: These data suggest that IOI-42 has a potential to enhance the radiosensitivity of rectal cancer cells, providing a rationale to further investigate the feasibility of combining of IOI-42 with radiation, keeping in mind that this may result in unexpected toxicities.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias del Recto/tratamiento farmacológico , Animales , Apoptosis/efectos de la radiación , Rayos gamma , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias del Recto/metabolismo , Neoplasias del Recto/patología , Neoplasias del Recto/radioterapia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
9.
Fertil Steril ; 106(6): 1530-1538.e1, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27565262

RESUMEN

OBJECTIVE: To investigate the presence of Raf kinase inhibitor protein (RKIP) in human myometrium and leiomyoma as well as to determine the effect of locostatin (RKIP inhibitor) on extracellular matrix (ECM) production, proliferation, and migration in human myometrial and leiomyoma cells. DESIGN: Laboratory study. SETTING: Human myometrium and leiomyoma. PATIENT(S): Thirty premenopausal women who were admitted to the hospital for myomectomy or hysterectomy. INTERVENTION(S): Myometrial and leiomyoma tissues were used to investigate the localization and the expression level of RKIP through immunohistochemistry and Western blotting. Myometrial and leiomyoma cells were treated with locostatin (10 µM) to measure ECM expression by real-time polymerase chain reaction, GSK3ß expression by Western blotting, cell migration by wound-healing assay, and cell proliferation by MTT assay and immunocytochemistry. MAIN OUTCOME MEASURE(S): The expression of RKIP in human myometrial and leiomyoma tissue; ECM components and GSK3ß expression, migration, and proliferation in myometrial and leiomyoma cells. RESULT(S): RKIP is expressed in human myometrial and leiomyoma tissue. Locostatin treatment resulted in the activation of the mitogen-activated protein kinase (MAPK) signal pathway (ERK phosphorylation), providing a powerful validation of our targeting protocol. Further, RKIP inhibition by locostatin reduces ECM components. Moreover, the inhibition of RKIP by locostatin impaired cell proliferation and migration in both leiomyoma and myometrial cells. Finally, locostatin treatment reduced GSK3ß expression. Therefore, even if the activation of MAPK pathway should increase proliferation and migration, the destabilization of GSK3ß leads to the reduction of proliferation and migration of myometrial and leiomyoma cells. CONCLUSION(S): Our results indicate that RKIP may be involved in leiomyoma pathophysiology.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Leiomioma/tratamiento farmacológico , Miocitos del Músculo Liso/efectos de los fármacos , Miometrio/efectos de los fármacos , Oxazolidinonas/farmacología , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Neoplasias Uterinas/tratamiento farmacológico , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Leiomioma/genética , Leiomioma/metabolismo , Leiomioma/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Miometrio/metabolismo , Miometrio/patología , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología
10.
Oncotarget ; 7(17): 23512-20, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-26992205

RESUMEN

A tobacco-specific component, 4-methylnitrosamino-1-3-pyridyl-1-butanone (NNK), is a major risk factor for many cancers. Recent reports have demonstrated that NNK exposure may be associated with tumor progression and chemoresistance in certain cancers. However, the underlying NNK-induced mechanism contributing to the aggressiveness of colorectal cancer (CRC) has not been thoroughly studied. In this study, we used HT29 cells treated with NNK to simulate the long-term exposure of cigarette smoke. A comparative analysis was performed to evaluate cell proliferation, migration, and invasion as well as epithelial-mesenchymal transition (EMT) markers and drug-resistance genes expression, cancer stem cell (CSC) properties, and anti-apoptotic activity. Signaling pathways related to chemoresistance were also investigated. As a result, NNK exposure dose-dependently stimulates cell proliferation, enhance abilities of migration and invasion, induce EMT phenomenon, and attenuate apoptosis. Furthermore, NNK exposure also promotes the capabilities of sphere formation, upregulation of Snail, and overexpression of CD133, Nanog, OCT4, and the drug-resistant genes. Knockdown of Snail results in upregulation of Raf kinase inhibitor protein (RKIP), increased apoptosis, reversal of EMT phenomenon, and reducation of expression of CSC markers, all of which contribute to a decrease of chemoresistance. Our study demonstrates a number of related mechanisms that mediate the effect of NNK exposure on increasing CRC therapeutic resistance via the Snail signaling pathway. Targeting Snail may provide a feasible strategy for the treatment of CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos , Nitrosaminas/farmacología , Compuestos Organoplatinos/farmacología , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Carcinógenos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Oxaliplatino , Transducción de Señal , Células Tumorales Cultivadas
11.
Sci Rep ; 6: 21810, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26911452

RESUMEN

Protein arginine methyltransferase 1 (PRMT1) catalyzes methylation of histones and other cellular proteins, and thus regulates gene transcription and protein activity. In antigen-induced pulmonary inflammation (AIPI) PRMT1 was up-regulated in the epithelium, while in chronic AIPI, increased PRMT1 shifted to fibroblasts. In this study we investigated the cell type specific regulatory mechanism of PRMT1. Epithelial cells and fibroblasts were stimulated with IL-4 or IL-1ß. Gene and protein expression were determined by RT-qPCR, immunohistochemistry staining and Western blotting. Signaling pathway inhibitors, siRNAs and shRNA were used to determine the regulatory mechanism of PRMT1. The results showed that IL-4 up-regulated PRMT1 through STAT6 signaling in epithelial cells, while IL-1ß regulated PRMT1 through NF-κB in fibroblasts. The NF-kB inhibitor protein RKIP was highly expressed in epithelial cells and blocked IL-1ß induced PRMT1 up-regulation; while the STAT6 inhibitor protein PIAS1 was expressed in fibroblasts and suppressed IL-4 induced PRMT1 expression. Furthermore, IL-4 stimulated epithelial cells to release IL-1ß which up-regulated PRMT1 expression in fibroblasts. In conclusion, the inhibitor proteins RKIP and PIAS1 regulated the cell type and signaling specific expression of PRMT1. Thus PRMT1 expression in structural lung cells in asthma can be considered as potential target for new therapeutic intervention.


Asunto(s)
Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Células A549 , Sitios de Unión , Western Blotting , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Inmunoprecipitación , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-4/genética , Interleucina-4/metabolismo , Interleucina-4/farmacología , Pulmón/metabolismo , Pulmón/patología , Microscopía Fluorescente , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/genética , Fosforilación/efectos de los fármacos , Neumonía/metabolismo , Neumonía/patología , Regiones Promotoras Genéticas , Proteínas Inhibidoras de STAT Activados/antagonistas & inhibidores , Proteínas Inhibidoras de STAT Activados/genética , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Factor de Transcripción STAT6/antagonistas & inhibidores , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/antagonistas & inhibidores , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética
12.
Drug Metab Dispos ; 44(1): 115-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26558823

RESUMEN

Prasugrel is a thienopyridine antiplatelet prodrug that undergoes rapid hydrolysis in vivo to a thiolactone metabolite by human carboxylesterase-2 (hCE2) during gastrointestinal absorption. The thiolactone metabolite is further converted to a pharmacologically active metabolite by cytochrome P450 isoforms. The aim of the current study was to elucidate hydrolases other than hCE2 involved in the bioactivation step of prasugrel in human intestine. Using size-exclusion column chromatography of a human small intestinal S9 fraction, another peak besides the hCE2 peak was observed to have prasugrel hydrolyzing activity, and this protein was found to have a molecular weight of about 20 kDa. This prasugrel hydrolyzing protein was successfully purified from a monkey small intestinal cytosolic fraction by successive four-step column chromatography and identified as Raf-1 kinase inhibitor protein (RKIP) by liquid chromatography-tandem mass spectrometry. Second, we evaluated the enzymatic kinetic parameters for prasugrel hydrolysis using recombinant human RKIP and hCE2 and estimated the contributions of these two hydrolyzing enzymes to the prasugrel hydrolysis reaction in human intestine, which were approximately 40% for hRKIP and 60% for hCE2. Moreover, prasugrel hydrolysis was inhibited by anti-hRKIP antibody and carboxylesterase-specific chemical inhibitor (bis p-nitrophenyl phosphate) by 30% and 60%, respectively. In conclusion, another protein capable of hydrolyzing prasugrel to its thiolactone metabolite was identified as RKIP, and this protein may play a significant role with hCE2 in prasugrel bioactivation in human intestine. RKIP is known to have diverse functions in many intracellular signaling cascades, but this is the first report describing RKIP as a hydrolase involved in drug metabolism.


Asunto(s)
Hidrolasas/metabolismo , Intestino Delgado/enzimología , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Inhibidores de Agregación Plaquetaria/metabolismo , Clorhidrato de Prasugrel/metabolismo , Activación Metabólica , Animales , Carboxilesterasa/antagonistas & inhibidores , Carboxilesterasa/metabolismo , Catálisis , Cromatografía en Gel , Cromatografía Liquida , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Hidrolasas/antagonistas & inhibidores , Hidrólisis , Intestino Delgado/efectos de los fármacos , Cinética , Macaca fascicularis , Masculino , Modelos Biológicos , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Espectrometría de Masas en Tándem
13.
Cell Signal ; 25(5): 1156-65, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23416466

RESUMEN

MAP kinase (MAPK) signaling results from activation of Raf kinases in response to external or internal stimuli. Here, we demonstrate that Raf kinase inhibitory protein (RKIP) regulates the activation of MAPK when B-Raf signaling is defective. We used multiple models including mouse embryonic fibroblasts (MEFs) and primary keratinocytes from RKIP- or Raf-deficient mice as well as allografts in mice to investigate the mechanism. Loss of B-Raf protein or activity significantly reduces MAPK activation in these cells. We show that RKIP depletion can rescue the compromised ERK activation and promote proliferation, and this rescue occurs through a Raf-1 dependent mechanism. These results provide formal evidence that RKIP is a bona fide regulator of Raf-1. We propose a new model in which RKIP plays a key role in regulating the ability of cells to signal through Raf-1 to ERK in B-Raf compromised cells.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Activación Enzimática , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal
14.
J Phys Chem B ; 116(34): 10176-81, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22861375

RESUMEN

The present work is aimed to provide detail on the binding process between Raf kinase inhibitor protein (RKIP) and locostatin, the only exogenous compound known to alter the function of RKIP. Understanding the basis of RKIP inhibition for use in pharmacological applications is of considerable interest, as dysregulated RKIP expression has the potential to contribute to pathophysiological processes. Herein, we report a series of atomistic models to describe the protein-ligand recognition step and the subsequent reactivity steps. Modeling approaches include ligand docking, molecular dynamics, and quantum mechanics/molecular mechanics calculations. We expect that such a computational assay will serve to study similar complexes in which potency is associated with recognition and reactivity. Although previous data suggested a single amino acid residue (His86) to be involved in the binding of locostatin, the actual ligand conformation and the steps involved in the reactivity process remain elusive from a detailed atomistic description. We show that the first reaction step, consisting of a nucleophilic attack of the nitrogen (Nε) of His86 at the sp(2)-hybridized carbon (C2) of locostatin, presents a late transition state (almost identical to the product). The reaction is followed by a hydrogen abstraction and hydrolysis. The theoretically predicted overall rate constant (6 M(-1) s(-1)) is in a very good agreement with the experimentally determined rate constant (13 M(-1) s(-1)).


Asunto(s)
Oxazolidinonas/química , Oxazolidinonas/farmacología , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/química , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
15.
Cancer Res ; 72(12): 3091-104, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22505648

RESUMEN

Epigenetic modifications such as histone methylation play an important role in human cancer metastasis. Enhancer of zeste homolog 2 (EZH2), which encodes the histone methyltransferase component of the polycomb repressive complex 2 (PRC2), is overexpressed widely in breast and prostate cancers and epigenetically silences tumor suppressor genes. Expression levels of the novel tumor and metastasis suppressor Raf-1 kinase inhibitor protein (RKIP) have been shown to correlate negatively with those of EZH2 in breast and prostate cell lines as well as in clinical cancer tissues. Here, we show that the RKIP/EZH2 ratio significantly decreases with the severity of disease and is negatively associated with relapse-free survival in breast cancer. Using a combination of loss- and gain-of-function approaches, we found that EZH2 negatively regulated RKIP transcription through repression-associated histone modifications. Direct recruitment of EZH2 and suppressor of zeste 12 (Suz12) to the proximal E-boxes of the RKIP promoter was accompanied by H3-K27-me3 and H3-K9-me3 modifications. The repressing activity of EZH2 on RKIP expression was dependent on histone deacetylase promoter recruitment and was negatively regulated upstream by miR-101. Together, our findings indicate that EZH2 accelerates cancer cell invasion, in part, via RKIP inhibition. These data also implicate EZH2 in the regulation of RKIP transcription, suggesting a potential mechanism by which EZH2 promotes tumor progression and metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Invasividad Neoplásica/patología , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Neoplasias de la Próstata/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Histonas/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Metástasis de la Neoplasia , Proteínas de Neoplasias , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/biosíntesis , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Interferencia de ARN , ARN Interferente Pequeño
16.
PLoS One ; 7(1): e30769, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22292035

RESUMEN

Malignant gliomas are highly infiltrative and invasive tumors, which precludes the few treatment options available. Therefore, there is an urgent need to elucidate the molecular mechanisms underlying gliomas aggressive phenotype and poor prognosis. The Raf Kinase Inhibitory protein (RKIP), besides regulating important intracellular signaling cascades, was described to be associated with progression, metastasis and prognosis in several human neoplasms. Its role in the prognosis and tumourigenesis of gliomas remains unclear. In the present study, we found that RKIP protein is absent in a low frequency (10%, 20/193) of glioma tumors. Nevertheless, the absence of RKIP expression was an independent prognostic marker in glioma. Additionally, by in vitro downregulation of RKIP, we found that RKIP inhibition induces a higher viability and migration of the cells, having no effect on cellular proliferation and angiogenesis, as assessed by in vivo CAM assay. In conclusion, this is the largest series studied so far evaluating the expression levels of this important cancer suppressor protein in glioma tumors. Our results suggest that in a subset of tumors, the absence of RKIP associates with highly malignant behavior and poor survival of patients, which may be a useful biomarker for tailored treatment of glioma patients.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Glioma/diagnóstico , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Adolescente , Adulto , Anciano , Animales , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/fisiología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Células Cultivadas , Embrión de Pollo , Niño , Preescolar , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/metabolismo , Glioma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/efectos de los fármacos , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteínas de Unión a Fosfatidiletanolamina/fisiología , Pronóstico , ARN Interferente Pequeño/farmacología , Transfección , Adulto Joven
17.
Cancer Prev Res (Phila) ; 5(3): 473-83, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22174364

RESUMEN

Neuroblastomas arise from the neural crest cells and represent the most common solid tumors outside the nervous system in children. The amplification of N-Myc plays a primary role in the pathogenesis of neuroblastomas, whereas acquired mutations of p53 lead to refractory and relapsed cases of neuroblastomas. In this regard, dietary compounds which can target N-Myc and exert anticancer effects independent of p53 status acquire significance in the management of neuroblastomas. Hence, we investigated the anticancer properties of the flavonoid didymin in neuroblastomas. Didymin effectively inhibited proliferation and induced apoptosis irrespective of p53 status in neuroblastomas. Didymin downregulated phosphoinositide 3-kinase, pAkt, Akt, vimentin, and upregulated RKIP levels. Didymin induced G(2)/M arrest along with decreasing the levels of cyclin D1, CDK4, and cyclin B1. Importantly, didymin inhibited N-Myc as confirmed at protein, mRNA, and transcriptional level by promoter-reporter assays. High-performance liquid chromatography analysis of didymin-treated (2 mg/kg b.w.) mice serum revealed effective oral absorption with free didymin concentration of 2.1 µmol/L. Further in vivo mice xenograft studies revealed that didymin-treated (2 mg/kg b.w.) animals had significant reductions in tumors size compared with controls. Didymin strongly inhibited the proliferation (Ki67) and angiogenesis (CD31) markers, as well as N-Myc expression, as revealed by the histopathologic examination of paraffin-embedded section of resected tumors. Collectively, our in vitro and in vivo studies elucidated the anticancer properties and mechanisms of action of a novel, orally active, and palatable flavonoid didymin, which makes it a potential new approach for neuroblastoma therapy (NANT) to target pediatric neuroblastomas.


Asunto(s)
Apoptosis/efectos de los fármacos , Flavonoides/farmacología , Glicósidos/farmacología , Neuroblastoma/patología , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Desnudos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Tumorales Cultivadas , Regulación hacia Arriba
18.
J Cell Physiol ; 227(5): 1821-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21732351

RESUMEN

Raf kinase inhibitor protein (RKIP) regulates growth and differentiation signaling of mitogen-activated protein kinases (MAPK), GRK2 and NF-kappaB pathways each of which regulates cytotrophoblast differentiation and normal placental development. We show here that RKIP is expressed in human normal and preeclampic placentas as detected by immunostaining. RKIP was detected in villous cytotrophoblast in normal placenta and switched to syncytiotrophoblast in pre-eclampsia (PE)-complicated pregnancies. RKIP was also localized in extravillous cytotrophoblast of cell islands and cell columns both in normal and in PE placentas, although staining was less uniform in the latter specimens. In order to test RKIP involvement in cytotrophoblast function, we performed in vitro studies on HTR-8/SVneo cells, a first trimester cytotrophoblast cell line. We show that the RKIP inhibitor locostatin reduces ERK phosphorylation and impairs HTR-8/SV neo cells motility in wound closure experiments. We also document the presence of GRK2 mRNA, the reduction of phosphorylated RKIP expression by locostatin and the induction of PAI mRNA expression in HTR-8/SV neo cells, suggesting the involvement of GRK2 and NF-kappaB pathways in these cells. In conclusion, our work provides evidence that RKIP is a novel factor expressed in cytotrophoblast cells where it likely regulates cell migration.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Preeclampsia/metabolismo , Trofoblastos/fisiología , Línea Celular , Movimiento Celular/efectos de los fármacos , Femenino , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Oxazolidinonas/farmacología , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/genética , Placenta/citología , Placenta/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Preeclampsia/patología , Embarazo , Transducción de Señal/fisiología , Trofoblastos/citología
19.
J Neurochem ; 118(2): 176-86, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21554319

RESUMEN

ß-N-Oxalyl-L-α,ß-diaminopropionic acid (l-ODAP) an α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor agonist activates protein kinase C in white leghorn chick brain. The current study focuses on the protein kinase C downstream signaling targets associated with L-ODAP excitotoxicity in SK-N-MC human neuroblastoma cells and white leghorn male chick (Gallus domesticus) brain extracts. L-ODAP treatment in SK-N-MC cells (1.5 mM) and chicks (0.5 mg/g body weight) results in a decreased expression and increased phosphorylation of phosphatidylehthanolamine-binding protein 1 (PEBP1) up to 4 h which however, returns to normal by 8 h. D-ODAP, the non-toxic enantiomer however, did not affect PEBP1 levels in either chick brain or SK-N-MC cells. Decreased PEBP1 expression correlated with subsequent activation of Raf-1, MEK and ERK signaling components of the mitogen-activated protein kinase cascade and nuclear translocation of hypoxia inducible factor-1α (HIF-1α) in chick brain nuclear extracts and SK-N-MC cells. SK-N-MC cells over-expressing PEBP1 inhibited nuclear translocation of HIF-1α when treated with l-ODAP, indicating that down-regulation of PEBP1 is responsible for HIF-1α stabilization and nuclear localization. Excitotoxicity of L-ODAP may thus be the result of phosphorylation and down-regulation of PEBP1, a crucial signaling protein regulating diverse signaling cascades. L-ODAP induced convulsions and seizures in chicks could be the result of a hypoxic insult to brain.


Asunto(s)
Aminoácidos Diaminos/fisiología , Regulación hacia Abajo/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Animales , Línea Celular Tumoral , Pollos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratas
20.
Mol Biosyst ; 6(12): 2498-507, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20941418

RESUMEN

This work describes the identification and characterization of a Sulfolobus solfataricus acylpeptide hydrolase, named APEH(Ss), recognised as a new protease target of the endogenous PEBP inhibitor, SsCEI. APEH is one of the four members of the prolyl oligopeptidase (POP) family, which removes acylated amino acid residues from the N terminus of oligopeptides. APEH(Ss) is a cytosolic homodimeric protein with a molecular mass of 125 kDa. It displays a similar exopeptidase and endopeptidase activity to the homologous enzymes from Aeropyrum pernix and Pyrococcus horikoshii. Herein we demonstrate that SsCEI is the first PEBP protein found to efficiently inhibit APEH from both S. solfataricus and mammalian sources with IC(50) values in the nanomolar range. The 3D model of APEH(Ss) shows the typical structural features of the POP family including an N-terminal ß-propeller and a C-terminal α/ß hydrolase domain. Moreover, to gain insights into the binding mode of SsCEI toward APEH(Ss), a structural model of the inhibition complex is proposed, suggesting a mechanism of steric blockage on substrate access to the active site or on product release. Like other POP enzymes, APEH may constitute a new therapeutic target for the treatment of a number of pathologies and this study may represent a starting point for further medical research.


Asunto(s)
Péptido Hidrolasas/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/antagonistas & inhibidores , Inhibidores de Proteasas/metabolismo , Sulfolobus solfataricus/química , Sulfolobus solfataricus/enzimología , Secuencia de Aminoácidos , Electroforesis en Gel de Poliacrilamida , Concentración 50 Inhibidora , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Péptido Hidrolasas/química , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Inhibidores de Proteasas/química , Inhibidores de Proteasas/aislamiento & purificación , Multimerización de Proteína , Transporte de Proteínas , Alineación de Secuencia , Homología Estructural de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA