Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(9)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925510

RESUMEN

BACKGROUND: Hepatocyte growth factor (HGF) is an endogenously induced bioactive molecule that has strong anti-apoptotic and tissue repair activities. In this research, we identified APOA4 as a novel pharmacodynamic (PD) marker of the recombinant human HGF (rh-HGF), E3112. METHODS: rh-HGF was administered to mice, and their livers were investigated for the PD marker. Candidates were identified from soluble proteins and validated by using human hepatocytes in vitro and an animal disease model in vivo, in which its c-Met dependency was also ensured. RESULTS: Among the genes induced or highly enhanced after rh-HGF exposure in vivo, a soluble apolipoprotein, Apoa4, was found to be induced by rh-HGF in the murine liver. By using primary cultured human hepatocytes, the significant induction of human APOA4 was observed at the mRNA and protein levels, and it was inhibited in the presence of a c-Met inhibitor. Although mice constitutively expressed Apoa4 mRNA in the small intestine and the liver, the liver was the primary organ affected by administered rh-HGF to strongly induce APOA4 in a dose- and c-Met-dependent manner. Serum APOA4 levels were increased after rh-HGF administration, not only in normal mice but also in anti-Fas-induced murine acute liver failure (ALF), which confirmed the pharmacodynamic nature of APOA4. CONCLUSIONS: APOA4 was identified as a soluble PD marker of rh-HGF with c-Met dependency. It should be worthwhile to clinically validate its utility through clinical trials with healthy subjects and ALF patients.


Asunto(s)
Apolipoproteínas A/sangre , Biomarcadores Farmacológicos/sangre , Factor de Crecimiento de Hepatocito/farmacocinética , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Animales , Apolipoproteínas A/genética , Apolipoproteínas A/metabolismo , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/administración & dosificación , Hepatocitos/metabolismo , Humanos , Hígado/fisiología , Fallo Hepático Agudo/sangre , Fallo Hepático Agudo/etiología , Masculino , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética
2.
Cancer Res ; 81(4): 1052-1062, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33408116

RESUMEN

The Wnt/ß-catenin signaling pathway plays crucial roles in embryonic development and the development of multiple types of cancer, and its aberrant activation provides cancer cells with escape mechanisms from immune checkpoint inhibitors. E7386, an orally active selective inhibitor of the interaction between ß-catenin and CREB binding protein, which is part of the Wnt/ß-catenin signaling pathway, disrupts the Wnt/ß-catenin signaling pathway in HEK293 and adenomatous polyposis coli (APC)-mutated human gastric cancer ECC10 cells. It also inhibited tumor growth in an ECC10 xenograft model and suppressed polyp formation in the intestinal tract of ApcMin /+ mice, in which mutation of Apc activates the Wnt/ß-catenin signaling pathway. E7386 demonstrated antitumor activity against mouse mammary tumors developed in mouse mammary tumor virus (MMTV)-Wnt1 transgenic mice. Gene expression profiling using RNA sequencing data of MMTV-Wnt1 tumor tissue from mice treated with E7386 showed that E7386 downregulated genes in the hypoxia signaling pathway and immune responses related to the CCL2, and IHC analysis showed that E7386 induced infiltration of CD8+ cells into tumor tissues. Furthermore, E7386 showed synergistic antitumor activity against MMTV-Wnt1 tumor in combination with anti-PD-1 antibody. In conclusion, E7386 demonstrates clear antitumor activity via modulation of the Wnt/ß-catenin signaling pathway and alteration of the tumor and immune microenvironments, and its antitumor activity can be enhanced in combination with anti-PD-1 antibody. SIGNIFICANCE: These findings demonstrate that the novel anticancer agent, E7386, modulates Wnt/ß-catenin signaling, altering the tumor immune microenvironment and exhibiting synergistic antitumor activity in combination with anti-PD-1 antibody.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/patología , Fragmentos de Péptidos/metabolismo , Pirazinas/farmacología , Sialoglicoproteínas/metabolismo , Triazinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , Animales , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Genes APC , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Unión Proteica/efectos de los fármacos , Pirazinas/uso terapéutico , Sialoglicoproteínas/antagonistas & inhibidores , Triazinas/uso terapéutico , Vía de Señalización Wnt/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/antagonistas & inhibidores
3.
BMC Complement Med Ther ; 20(1): 136, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375753

RESUMEN

BACKGROUND: Type 2 diabetes mellitus is characterized by insulin resistance and pancreatic ß-cell dysfunction. A decrease in ß-cell mass, which occurs during the progression of Type 2 diabetes mellitus, contributes to impaired insulin secretion. Mulberry leaves contain various nutritional components that exert anti-diabetic and anti-atherogenic effects. The present study analyzed the effects of mulberry leaf intake on pancreatic ß-cells to clarify the mechanisms underlying its anti-diabetic function. METHODS: Mulberry leaves (Morus alba L.) were dried at 180 °C for 8 s in a hot-air mill and fed to obesity/Type 2 diabetes mellitus db/db mouse models at 5% (w/w) as part of a normal diet from 7 to 10, 15, or 20 weeks of age. An intraperitoneal glucose tolerance test was then performed on the mice. To evaluate the ß-cell mass, the pancreas was subjected to immunohistological analysis with an anti-insulin antibody. A TUNEL assay and immunohistological analysis with a proliferation marker was also performed. Expression levels of endoplasmic reticulum stress-responsible genes and proliferation markers were assessed by quantitative RT-PCR. RESULTS: Intake of mulberry leaves maintained the ß-cell function of db/db mice. Moreover, oral administration of mulberry leaves significantly decreased cell death by reducing endoplasmic reticulum stress in the pancreas. Mulberry leaves significantly increased proliferation of ß-cells and the expression of pancreatic duodenal homeobox1 mRNA in the pancreas. CONCLUSION: Considered together, these results indicate that dietary mulberry leaf administration can maintain insulin levels and pancreatic ß-cell mass, at least in part, by suppressing endoplasmic reticulum stress in Type 2 diabetes mellitus mouse models.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Células Secretoras de Insulina/citología , Morus , Fitoterapia , Hojas de la Planta , Administración Oral , Animales , Glucemia , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Insulina/sangre , Japón , Ratones , Ratones Obesos
4.
Toxicol Appl Pharmacol ; 329: 282-292, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28624441

RESUMEN

Sunitinib malate (sunitinib) is an orally available, multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities. Although sunitinib is effective for the treatment of patients with gastrointestinal stromal tumor, advanced renal cell carcinoma, or pancreatic neuroendocrine tumor, adverse cardiac events associated with sunitinib administration have been reported. Here, we examined the effect of geldanamycin, an inhibitor of heat shock protein (Hsp) 90, on sunitinib-induced cytotoxicity in cardiomyocytes. First, we found that treatment with geldanamycin or other Hsp90 inhibitors (tanespimycin, ganetespib, or BIIB021) significantly attenuated sunitinib-induced cytotoxicity in rat H9c2 cardiomyocytes, suggesting a drug-class effect of Hsp90 inhibitors. We then examined the mechanisms underlying sunitinib-induced cytotoxicity and found that sunitinib induced autophagy in H9c2 cells and that pretreatment with geldanamycin inhibited the induction of autophagy by promoting degradation of the autophagy-related proteins Atg7, Beclin-1, and ULK1. Pharmacological assessment with autophagy inhibitors confirmed that geldanamycin attenuated the cytotoxicity of sunitinib by interfering with autophagy. In addition, we found that the molecular chaperone Hsp70, which is induced by geldanamycin, was not involved in the attenuation of sunitinib-induced cytotoxicity. Finally, to provide more clinically relevant data, we confirmed that geldanamycin attenuated sunitinib-induced cytotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. Together, these data suggest that geldanamycin attenuates sunitinib-induced cytotoxicity in cardiomyocytes by inhibiting the autophagy pathway. Thus, the further investigation of combination or sequential treatment with an Hsp90 inhibitor and sunitinib is warranted as a potential strategy of attenuating the cardiotoxicity associated with sunitinib administration in the clinical setting.


Asunto(s)
Antineoplásicos/toxicidad , Autofagia/efectos de los fármacos , Benzoquinonas/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Indoles/toxicidad , Lactamas Macrocíclicas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/toxicidad , Pirroles/toxicidad , Animales , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Beclina-1/genética , Beclina-1/metabolismo , Cardiotoxicidad , Diferenciación Celular , Línea Celular , Linaje de la Célula , Citoprotección , Relación Dosis-Respuesta a Droga , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Interferencia de ARN , Ratas , Transducción de Señal/efectos de los fármacos , Sunitinib , Transfección
5.
Nat Chem Biol ; 13(6): 675-680, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28437394

RESUMEN

Target-protein degradation is an emerging field in drug discovery and development. In particular, the substrate-receptor proteins of the cullin-ubiquitin ligase system play a key role in selective protein degradation, which is an essential component of the anti-myeloma activity of immunomodulatory drugs (IMiDs), such as lenalidomide. Here, we demonstrate that a series of anticancer sulfonamides NSC 719239 (E7820), indisulam, and NSC 339004 (chloroquinoxaline sulfonamide, CQS) induce proteasomal degradation of the U2AF-related splicing factor coactivator of activating protein-1 and estrogen receptors (CAPERα) via CRL4DCAF15 mediated ubiquitination in human cancer cell lines. Both CRISPR-Cas9-based knockout of DCAF15 and a single amino acid substitution of CAPERα conferred resistance against sulfonamide-induced CAPERα degradation and cell-growth inhibition. Thus, these sulfonamides represent selective chemical probes for disrupting CAPERα function and designate DCAFs as promising drug targets for promoting selective protein degradation in cancer therapy.


Asunto(s)
Indoles/farmacología , Proteínas Nucleares/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Sulfonamidas/metabolismo , Antineoplásicos/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteolisis/efectos de los fármacos , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Sulfonamidas/farmacología
6.
Cancer Sci ; 105(10): 1334-42, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25060424

RESUMEN

Eribulin mesylate is a synthetic macrocyclic ketone analog of the marine sponge natural product halichondrin B and an inhibitor of microtubule dynamics. Some tubulin-binding drugs are known to have antivascular (antiangiogenesis or vascular-disrupting) activities that can target abnormal tumor vessels. Using dynamic contrast-enhanced MRI analyses, here we show that eribulin induces remodeling of tumor vasculature through a novel antivascular activity in MX-1 and MDA-MB-231 human breast cancer xenograft models. Vascular remodeling associated with improved perfusion was shown by Hoechst 33342 staining and by increased microvessel density together with decreased mean vascular areas and fewer branched vessels in tumor tissues, as determined by immunohistochemical staining for endothelial marker CD31. Quantitative RT-PCR analysis of normal host cells in the stroma of xenograft tumors showed that eribulin altered the expression of mouse (host) genes in angiogenesis signaling pathways controlling endothelial cell-pericyte interactions, and in the epithelial-mesenchymal transition pathway in the context of the tumor microenvironment. Eribulin also decreased hypoxia-associated protein expression of mouse (host) vascular endothelial growth factor by ELISA and human CA9 by immunohistochemical analysis. Prior treatment with eribulin enhanced the anti-tumor activity of capecitabine in the MDA-MB-231 xenograft model. These findings suggest that eribulin-induced remodeling of abnormal tumor vasculature leads to a more functional microenvironment that may reduce the aggressiveness of tumors due to elimination of inner tumor hypoxia. Because abnormal tumor microenvironments enhance both drug resistance and metastasis, the apparent ability of eribulin to reverse these aggressive characteristics may contribute to its clinical benefits.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Furanos/farmacología , Cetonas/farmacología , Moduladores de Tubulina/farmacología , Microambiente Tumoral/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos , Animales , Neoplasias de la Mama/patología , Capecitabina , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Femenino , Fluorouracilo/análogos & derivados , Fluorouracilo/farmacología , Humanos , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Ther ; 13(4): 842-54, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24563539

RESUMEN

Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Linfoma no Hodgkin/tratamiento farmacológico , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/genética , Piridonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo , Dominio Catalítico/genética , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Linfoma no Hodgkin/patología , Masculino , Ratones , Ratones SCID , Datos de Secuencia Molecular , Morfolinas , Mutación Puntual , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Pharmacol Toxicol Methods ; 69(2): 177-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24296355

RESUMEN

INTRODUCTION: Cardiac hypertrophy is a leading cause of many cardiovascular diseases, including heart failure, but its pathological mechanism is not fully understood. This study used highly purified human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes to produce an in vitro hypertrophy model and characterize its gene expression and electrophysiological properties. METHODS: For 7 days we cultured hiPSC-derived cardiomyocytes plated at high (2800-4800 cells/mm(2)) or low (500-1200 cells/mm(2)) cell density and assessed their cell size with confocal and fluorescence microscopy, their electrophysiological and pharmacological responses with multi-electrode array systems, and their gene expression patterns by using DNA microarray technology and quantitative PCR. We used quantitative PCR and Western blotting to compare the expression of potassium-channel genes between the hiPSC-derived cardiomyocytes and human fetal and adult hearts. RESULTS: The hiPSC-derived cardiomyocytes showed spontaneous beating and similar pattern of α-actinin molecules regardless of plating density. However, cells plated at low density had the following characteristics compared with those at high density: 1) significant enlargement in size; 2) significant increase or decrease in expression of the cardiac hypertrophy-characteristic genes NPPA, ATP2A2, ANKRD1 and MYL2 in accordance with the progression of hypertrophy; 3) significant reduction in responses to the inhibitors of cardiac slow delayed-rectifier K(+) current (IKs), chromanol 293B and HMR1556, in a cell-density-dependent manner; and 4) significant reduction in the expression of the KCNQ1 and KCNJ2 genes coding the K(+) ion channels conducting each IKs and cardiac inward rectifier outward K(+) current (IK1). DISCUSSION: The enlargement, hypertrophy-characteristic and potassium ion channels gene expression of hiPSC-derived cardiomyocytes suggest that low-density plating was sufficient to induce cardiac hypertrophy. This model may be useful in elucidating mechanisms underlying the onset and progress of cardiac hypertrophy, because these cells can be cultured for several weeks.


Asunto(s)
Cardiomegalia/patología , Cardiomegalia/fisiopatología , Técnicas de Cultivo de Célula , Fenómenos Electrofisiológicos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Adulto , Cardiomegalia/genética , Separación Celular , Electrodos , Perfilación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo
9.
J Recept Signal Transduct Res ; 33(4): 224-33, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23697547

RESUMEN

The effect of the intracerebroventricular (i.c.v.) injection of relaxin-3 (RLX3) was evaluated using anxiety-related behavioral tests in rats. RLX3-injected animals showed normal locomotion activity in a habituated environment and declined anxiety cognition in the elevated plus maze test and the shock probe-burying test. The measurement of spontaneous locomotor activity in a novel environment also suggested that RLX3 reduced the stress response. To elucidate the regulatory mechanisms of the downstream signaling pathways underlying RLX3 activity and its relation to anxiolytic and hyperphagic behavior phenotypes, RLX3-i.c.v.-injected rat hypothalamic responses were examined using a microarray analysis. Ingenuity Pathway Analysis software listed the phenotype-relating genes and they showed characteristic expression patterns in the rat hypothalamus. When peptidome data sets for the same listed genes was analyzed using a semi-quantitative approach, the expressions of two neuropeptides were found to have increased. One of these neuropeptides, oxytocin (Oxt), exhibited increased expression in both the microarray and the peptidomic analysis, and a Western blot analysis validated the mass spectrometry results. A cross-omics data analysis is useful for predicting downstream signaling pathways, and the anxiolytic-like behavior of RLX3 may be mediated by an oxytocin signaling pathway in rats. These results suggest that RLX3 acts as an anxiolytic peptide and that the downstream pathways mediated by its receptors may be potential candidates for the treatment of anxieties in the future.


Asunto(s)
Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Relaxina/metabolismo , Estrés Fisiológico/efectos de los fármacos , Animales , Ansiedad/fisiopatología , Conducta Animal/fisiología , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Aprendizaje por Laberinto , Análisis por Micromatrices , Proteínas del Tejido Nervioso/administración & dosificación , Neuropéptidos/aislamiento & purificación , Neuropéptidos/metabolismo , Oxitocina/metabolismo , Ratas , Relaxina/administración & dosificación , Transducción de Señal
10.
Exp Hematol ; 41(2): 189-97, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23085465

RESUMEN

DNA methyltransferase inhibitors (DNMTIs), including decitabine (DAC) and azacitidine (AZA), have recently been highlighted for the treatment of high-risk myelodysplastic syndrome (MDS); however, their action mechanisms have not been clearly defined. Therefore, we investigated the effects of DNMTIs on MDS-derived cell lines in vitro. An MDS-derived cell line MDS92 and its blastic subline MDS-L and HL-60 were used. All three cell lines were sensitive to DNMTIs, but MDS-L was the most susceptible. DAC-induced cell death in MDS-L was preceded by DNA damage-induced G2 arrest via a p53-independent pathway. AZA did not influence the pattern of cell cycle, although it induced DNA damage response. The IC(50) of DAC or AZA on MDS-L cells was associated with the dose inducing the maximal hypomethylation in long interspersed nuclear elements-1 (LINE-1) methylation assay. AZA suppressed the level of methylation in a time-dependent manner (days 4, 7, and 10), whereas DAC maintained the level of methylation from day 4 to 11. The protein expression of DNMT1 and DNMT3a decreased with the suppression of growth and methylation. We conclude that this study provides in vitro models for understanding the effects of DNMTIs on cell growth and gene regulation, including differences in the possible action mechanism of DAC and AZA.


Asunto(s)
Azacitidina/análogos & derivados , Azacitidina/farmacología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Síndromes Mielodisplásicos/patología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , Daño del ADN , Metilación de ADN/efectos de los fármacos , Decitabina , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Células HL-60/efectos de los fármacos , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora
11.
J Cardiovasc Pharmacol ; 59(4): 377-86, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22189896

RESUMEN

Indirect effects of drugs on ion channel expression levels on plasma membrane are focused as the cause of QT prolongation, and we explored the chronic effects of QT-prolonging drugs on the slow component of the delayed-rectifier potassium current (IKs). Chinese Hamster Ovary cells expressing IKs channels were constructed by transfecting KCNQ1/KCNE1 genes, and the IKs values were measured using IonWorks Quattro in the population patch-clamp mode. After 24 hours of treatment with IKs blockers (HMR1556, L-768673, or chromanol 293B) or human Ether-à-go-go related gene channel trafficking inhibitors (amiodarone,17-AAG, brefeldin A, pentamidine, thioridazine, or probucol), brefeldin A, pentamidine, and probucol decreased IKs. Probucol, which is a cholesterol-lowering drug and clinically reported to cause QT prolongation, potently inhibited the IKs in a concentration-dependent manner, with a half maximal inhibitory concentration of 149.1 nM. A reduction in the IKs by 1 µM of probucol was observed beginning 2 hours after treatment, and the current was reduced by about 80% at 24 hours. The activation and deactivation time constants of residual IKs currents became faster compared with that in the vehicle-treatment group. Acute application of probucol did not directly inhibit IKs channels at concentrations of up to 10 µM. Western blotting analysis indicated the reduction of multimeric complex of KCNQ1 proteins by probucol treatment but not monomeric form. These results suggest that chronic probucol treatment may contribute to QT prolongation in humans by decreasing the functional IKs channel complexes.


Asunto(s)
Anticolesterolemiantes/toxicidad , Síndrome de QT Prolongado/inducido químicamente , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Probucol/toxicidad , Animales , Anticolesterolemiantes/administración & dosificación , Western Blotting , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Canal de Potasio KCNQ1/genética , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Probucol/administración & dosificación , Factores de Tiempo , Transfección
12.
J Biol Chem ; 285(43): 33294-33306, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20720005

RESUMEN

One of the family of voltage-gated calcium channels (VGCC), the N-type Ca(2+) channel, is located predominantly in neurons and is associated with a variety of neuronal responses, including neurodegeneration. A precise mechanism for how the N-type Ca(2+) channel plays a role in neurodegenerative disease, however, is unknown. In this study, we immunized N-type Ca(2+) channel α(1B)-deficient (α(1B)(-/-)) mice and their wild type (WT) littermates with myelin oligodendrocyte glycoprotein 35-55 and analyzed the progression of experimental autoimmune encephalomyelitis (EAE). The neurological symptoms of EAE in the α(1B)(-/-) mice were less severe than in the WT mice. In conjunction with these results, sections of the spinal cord (SC) from α(1B)(-/-) mice revealed a reduction in both leukocytic infiltration and demyelination compared with WT mice. No differences were observed in the delayed-type hypersensitivity response, spleen cell proliferation, or cytokine production from splenocytes between the two genotypes. On the other hand, Western blot array analysis and RT-PCR revealed that a typical increase in the expression of MCP-1 in the SC showed a good correlation with the infiltration of leukocytes into the SC. Likewise, immunohistochemical analysis showed that the predominant source of MCP-1 was activated microglia. The cytokine-induced production of MCP-1 in primary cultured microglia from WT mice was significantly higher than that from α(1B)(-/-) mice and was significantly inhibited by a selective N-type Ca(2+) channel antagonist, ω-conotoxin GVIA or a withdrawal of extracellular Ca(2+). These results suggest that the N-type Ca(2+) channel is involved in the pathogenesis of EAE at least in part by regulating MCP-1 production by microglia.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Quimiocina CCL2/biosíntesis , Encefalomielitis Autoinmune Experimental/metabolismo , Glicoproteínas/metabolismo , Microglía/metabolismo , Fragmentos de Péptidos/metabolismo , Médula Espinal/metabolismo , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/genética , Quimiocina CCL2/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Glicoproteínas/genética , Leucocitos/metabolismo , Leucocitos/patología , Ratones , Ratones Endogámicos CBA , Ratones Mutantes , Microglía/patología , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/genética , Médula Espinal/patología , omega-Conotoxina GVIA/farmacología
13.
Curr Pharm Biotechnol ; 10(2): 261-7, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19199960

RESUMEN

Using gene knockout mice of particular genes is one of the most effective methods in conducting successful study on the mode of action of target gene products in targeted organs. So called the knockout technology is now a powerful tool that can lead us to find clear understanding on difficult questions such as the effects of full antagonist against target molecules. Cacna1b (alpha(1B)) gene knockout mouse was generated to study mechanisms of N-type calcium (Ca(2+)) channel. The model was able to overcome physiological obstacles in studies of N-type Ca(2+) channel selective blockers, such as unspecific binding to structurally similar molecules, and failed distribution to targeted organs. In the case of N-type Ca(2+) channel studies, knockout technology was successfully applied to various cardiovascular, sympathetic, nociceptive, sleep-awake cycles, metabolic and neurodegenerative experiments using homozygous mutants of the alpha(1B) gene that turned out to be viable. These studies were able to confirm not only the predicted phenotypes, but were able to present completely unexpected phenotypes that are great interest for future study. Thus the outputs from the knockout mouse studies lead to gain the proof of concept as a drug for specific inhibitors of the gene products and enabled us to make further prediction of side-effects of these inhibitors in the drug discovery and development process.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/fisiología , Diseño de Fármacos , Marcación de Gen , Modelos Biológicos , Animales , Bloqueadores de los Canales de Calcio/efectos adversos , Canales de Calcio Tipo N/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Ratones , Ratones Noqueados , Valor Predictivo de las Pruebas
14.
Nat Chem Biol ; 3(9): 570-5, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17643112

RESUMEN

Pladienolide is a naturally occurring antitumor macrolide that was discovered by using a cell-based reporter gene expression assay controlled by the human vascular endothelial growth factor promoter. Despite the unique mechanisms of action and prominent antitumor activities of pladienolides B and D in diverse in vitro and in vivo systems, their target protein has remained unclear. We used 3H-labeled, fluorescence-tagged and photoaffinity/biotin (PB)-tagged 'chemical probes' to identify a 140-kDa protein in splicing factor SF3b as the binding target of pladienolide. Immunoblotting of an enhanced green fluorescent protein fusion protein of SF3b subunit 3 (SAP130) revealed direct interaction between the PB probe and SAP130. The binding affinities of pladienolide derivatives to the SF3b complex were highly correlated with their inhibitory activities against reporter gene expression and cell proliferation. Furthermore, pladienolide B impaired in vivo splicing in a dose-dependent manner. Our results demonstrate that the SF3b complex is a pharmacologically relevant protein target of pladienolide and suggest that this splicing factor is a potential antitumor drug target.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Epoxi/farmacología , Macrólidos/farmacología , Proteínas de Unión al ARN/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Genes Reporteros , Humanos , Unión Proteica , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2
15.
J Neurochem ; 97(5): 1259-68, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16539653

RESUMEN

Endoplasmic reticulum (ER) stress is defined as an accumulation of unfolded proteins in the endoplasmic reticulum. 4-phenylbutyrate (4-PBA) has been demonstrated to promote the normal trafficking of the DeltaF508 cystic fibrosis transmembrane conductance regulator (CFTR) mutant from the ER to the plasma membrane and to restore activity. We have reported that 4-PBA protected against cerebral ischemic injury and ER stress-induced neuronal cell death. In this study, we revealed that 4-PBA possesses chemical chaperone activity in vitro, which prevents the aggregation of denatured alpha-lactalbumin and bovine serum albumin (BSA). Furthermore, we investigated the effects of 4-PBA on the accumulation of Parkin-associated endothelin receptor-like receptor (Pael-R) pathologically relevant to the loss of dopaminergic neurons in autosomal recessive juvenile parkinsonism (AR-JP). Interestingly, 4-PBA restored the normal expression of Pael-R protein and suppressed ER stress induced by the overexpression of Pael-R. In addition, we showed that 4-PBA attenuated the activation of ER stress-induced signal transduction pathways and subsequent neuronal cell death. Moreover, 4-PBA restored the viability of yeasts that fail to induce an ER stress response under ER stress conditions. These results suggest that 4-PBA suppresses ER stress by directly reducing the amount of misfolded protein, including Pael-R accumulated in the ER.


Asunto(s)
Retículo Endoplásmico/metabolismo , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/metabolismo , Fenilbutiratos/farmacología , Agregación de Receptores/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Estrés Fisiológico/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Lactalbúmina/efectos de los fármacos , Lactalbúmina/metabolismo , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/farmacología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/fisiopatología , Fenilbutiratos/metabolismo , Pliegue de Proteína , Receptores Acoplados a Proteínas G/efectos de los fármacos , Saccharomyces cerevisiae , Albúmina Sérica Bovina/efectos de los fármacos , Albúmina Sérica Bovina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tunicamicina/farmacología
16.
FEBS Lett ; 532(1-2): 147-52, 2002 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-12459480

RESUMEN

Stresses that impair the function of the endoplasmic reticulum (ER) lead to an accumulation of unfolded protein in the ER. Under these conditions, the expression of a variety of genes involved in preventing the accumulation of the unfolded proteins is induced. Yeast Hrd1p is an ER stress-inducible ER membrane protein that acts as a ubiquitin ligase (E3) with a RING finger motif and plays a role in the ubiquitination of proteins in the ER. We report here the identification and characterization of a human homolog to yeast Hrd1p. The predicted structures are highly conserved from yeast to humans. Indeed, human HRD1 was localized to the ER and ubiquitinated its substrates. Furthermore, it was found that human HRD1 was up-regulated by ER stress via IRE1 and ATF6, which are ER stress transducers. Interestingly, 293 cells stably expressing wild-type HRD1, but not the C329S mutant, afforded resistance to ER stress-induced apoptosis. These results suggest that the production of HRD1 is up-regulated to protect against ER stress-induced apoptosis by degrading unfolded proteins accumulated in the ER.


Asunto(s)
Apoptosis , Retículo Endoplásmico/enzimología , Ligasas/fisiología , Proteínas de Saccharomyces cerevisiae , Ubiquitina-Proteína Ligasas , Secuencia de Aminoácidos , Línea Celular , Citoprotección , Retículo Endoplásmico/metabolismo , Humanos , Cinética , Ligasas/análisis , Ligasas/genética , Datos de Secuencia Molecular , Pliegue de Proteína , Proteínas/metabolismo , Alineación de Secuencia , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...