Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Invest New Drugs ; 37(4): 796, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30937691

RESUMEN

The blots of control and docetaxel for caspase-9, caspase-3, caspase-8, Bcl-XL, and tubulin in the Figure 4f were reused from Figure 4 of our previous paper published in Journal of Urology in 2010 ( https://doi.org/10.1016/j.juro.2010.07.035 ).

2.
Exp Mol Med ; 50(6): 1-12, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29884820

RESUMEN

Mitsugumin 53 (MG53) is an E3 ligase that induces insulin receptor substrate-1 (IRS-1) ubiquitination and degradation in skeletal muscle. We previously demonstrated that the pharmaceutical disruption of the MG53-IRS-1 interaction improves insulin sensitivity by abrogating IRS-1 ubiquitination and increasing IRS-1 levels in C2C12 myotubes. Here, we developed a novel MG53-IRS-1 interaction disruptor (MID-00935) that ameliorates insulin resistance in diet-induced obese (DIO) mice. MID-00935 disrupted the molecular interaction of MG53 and IRS-1, abrogated MG53-induced IRS-1 ubiquitination and degradation and improved insulin signaling in C2C12 myotubes. Oral administration of MID-00935 increased insulin-induced IRS-1, Akt, and Erk phosphorylation via increasing IRS-1 levels in the skeletal muscle of DIO mice. In DIO mice, MID-00935 treatment lowered fasting blood glucose levels and improved glucose disposal in glucose and insulin tolerance tests. These results suggest that MID-00935 may be a potential muscle-targeting drug candidate for treating insulin resistance.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Resistencia a la Insulina , Insulina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Obesidad/metabolismo , Animales , Proteínas Portadoras/metabolismo , Células HEK293 , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas de la Membrana , Fibras Musculares Esqueléticas/patología , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico , Obesidad/patología , Transducción de Señal/efectos de los fármacos
3.
Sci Rep ; 7: 41615, 2017 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-28134290

RESUMEN

Pancreatic cancer is predominantly lethal, and is primarily treated using gemcitabine, with increasing resistance. Therefore, novel agents that increase tumor sensitivity to gemcitabine are needed. Histone deacetylase (HDAC) inhibitors are emerging therapeutic agents, since HDAC plays an important role in cancer initiation and progression. We evaluated the antitumor effect of a novel HDAC inhibitor, CG200745, combined with gemcitabine/erlotinib on pancreatic cancer cells and gemcitabine-resistant pancreatic cancer cells. Three pancreatic cancer-cell lines were used to evaluate the antitumor effect of CG200745 combined with gemcitabine/erlotinib. CG200745 induced the expression of apoptotic proteins (PARP and caspase-3) and increased the levels of acetylated histone H3. CG200745 with gemcitabine/erlotinib showed significant growth inhibition and synergistic antitumor effects in vitro. In vivo, gemcitabine/erlotinib and CG200745 reduced tumor size up to 50%. CG200745 enhanced the sensitivity of gemcitabine-resistant pancreatic cancer cells to gemcitabine, and decreased the level of ATP-binding cassette-transporter genes, especially multidrug resistance protein 3 (MRP3) and MRP4. The novel HDAC inhibitor, CG200745, with gemcitabine/erlotinib had a synergistic anti-tumor effect on pancreatic cancer cells. CG200745 significantly improved pancreatic cancer sensitivity to gemcitabine, with a prominent antitumor effect on gemcitabine-resistant pancreatic cancer cells. Therefore, improved clinical outcome is expected in the future.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Naftalenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Humanos , Ratones , Neoplasias Pancreáticas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
4.
Korean J Physiol Pharmacol ; 20(5): 477-85, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27610034

RESUMEN

CG200745 is a novel inhibitor of histone deacetylases (HDACs), initially developed for treatment of various hematological and solid cancers. Because it is water-soluble, it can be administered orally. We hypothesized that the HDAC inhibitor, CG200745, attenuates cardiac hypertrophy and fibrosis in deoxycorticosterone acetate (DOCA)-induced hypertensive rats. For establishment of hypertension, 40 mg/kg of DOCA was subcutaneously injected four times weekly into Sprague-Dawley rats. All the rats used in this study including those in the sham group had been unilaterally nephrectomized and allowed free access to drinking water containing 1% NaCl. Systolic blood pressure was measured by the tail-cuff method. Blood chemistry including sodium, potassium, glucose, triglyceride, and cholesterol levels was analyzed. Sections of the heart were visualized after trichrome and hematoxylin and eosin stain. The expression of hypertrophic genes such as atrial natriuretic peptide A (Nppa) and atrial natriuretic peptide B (Nppb) in addition to fibrotic genes such as Collagen-1, Collagen-3, connective tissue growth factor (Ctgf), and Fibronectin were measured by quantitative real-time PCR (qRT-PCR). Injection of DOCA increased systolic blood pressure, heart weight, and cardiac fibrosis, which was attenuated by CG200745. Neither DOCA nor CG200745 affected body weight, vascular contraction and relaxation responses, and blood chemistry. Injection of DOCA increased expression of both hypertrophic and fibrotic genes, which was abrogated by CG200745. These results indicate that CG200745 attenuates cardiac hypertrophy and fibrosis in DOCA-induced hypertensive rats.

5.
Invest New Drugs ; 33(5): 1048-57, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26076682

RESUMEN

PURPOSE: The aim of the present study was to assess the safety, maximum tolerated dose (MTD), pharmacokinetics, pharmacodynamics, and efficacy of single and multiple doses of intravenous CG200745, a novel histone deacetylase (HDAC) inhibitor, in patients with advanced solid malignancies. EXPERIMENTAL DESIGN: Two to six patients received intravenous CG200745 according to the 2 + 4 dose-escalating method. This first-in-human trial was comprised of two parts: Part 1 was a single ascending dose, and Part 2 was multiple ascending doses weekly for 3 weeks, and then 1 week off. For the first cycle, pharmacokinetic sampling for CG200745 and pharmacodynamic sampling for acetylated histone H4 in peripheral blood mononuclear cells (PBMCs) were performed on day 1 for Part 1 and on days 1 and 15 for Part 2. Examination of acetylated histone H4 in pre- and post-biopsy samples was performed in accessible patients. RESULTS: In all, 28 patients were treated at 13 dose levels (1.8-250 mg/m(2)) and received a total of 71 cycles of CG200745. Hematologic toxicities included grade 3/4 neutropenia (22.2 %) that did not last a week and non-hematologic toxicities included fatigue (22.2 %) and anorexia (16.7 %) that did not exceed grade 2. No dose-limiting toxic effects were noted. Dose proportionality was observed for both the maximum concentration and area under the curve. The elimination half-life was 5.67 ± 2.69 h (mean ± standard deviation). An increase in PBMC acetylated histone H4 was observed at dose levels up to 51 mg/m(2), which plateaued at higher dose levels. At 24 h, 75 % of patients (6/8) showed higher relative acetylation in tumor tissue compared to PBMCs. Although there was no partial or complete response, 57.1 % of patients (16/28) had stable disease that lasted at least 6 weeks. CONCLUSIONS: CG200745 can be safely administered at effective dose levels that inhibit HDAC in PBMCs and tumor tissue. Although MTD was not reached, further escalation was not performed because acetylated histone H4 plateaued at dose levels higher than 51 mg/m(2). Additional phase II trials are recommended at 250 mg/m(2).


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Naftalenos/farmacología , Naftalenos/uso terapéutico , Neoplasias/tratamiento farmacológico , Adolescente , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Semivida , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/efectos adversos , Ácidos Hidroxámicos/farmacocinética , Leucocitos Mononucleares/metabolismo , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Naftalenos/efectos adversos , Naftalenos/farmacocinética , Adulto Joven
6.
Oncol Rep ; 33(4): 1947-55, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25672292

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) have been suggested as the potential new class of preventive or therapeutic antitumor agents. The aim of the present study was to evaluate the antitumor activity of the novel NSAID, CG100649. CG100649 is a novel NSAID dual inhibitor for COX-2 and carbonic anhydrase (CA)-I/-II. In the present study, we investigated the alternative mechanism by which CG100649 mediated suppression of the colon cancer growth and development. The anchorage­dependent and -independent clonogenic assay showed that CG100649 inhibited the clonogenicity of human colon cancer cells. The flow cytometric analysis showed that CG100649 induced the G2/M cell cycle arrest in colon cancer cells. Animal studies showed that CG100649 inhibited the tumor growth in colon cancer xenograft in nude mice. Furthermore, quantitative PCR and FACS analysis demonstrated that CG100649 upregulated the expression of TNF-related apoptosis-inducing ligand (TRAIL) receptors (DR4 and DR5) but decreased the expression of decoy receptors (DcR1 and DcR2) in colon cancer cells. The results showed that CG100649 treatment sensitized TRAIL­mediated growth suppression and apoptotic cell death. The combination treatment resulted in significant repression of the intestinal polyp formation in APCmin/+ mice. Our data clearly demonstrated that CG100649 contains preventive and therapeutic activity for colon cancer. The present study may be useful for identification of the potential benefit of the NSAID CG100649, for the achievement of a better treatment response in colon cancer.


Asunto(s)
Anticarcinógenos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Furanos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Sulfonamidas/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Anticarcinógenos/farmacología , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/patología , Sinergismo Farmacológico , Femenino , Furanos/administración & dosificación , Furanos/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Pólipos Intestinales/prevención & control , Intestino Delgado/patología , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Miembro 10c de Receptores del Factor de Necrosis Tumoral/biosíntesis , Miembro 10c de Receptores del Factor de Necrosis Tumoral/genética , Organismos Libres de Patógenos Específicos , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Receptores Señuelo del Factor de Necrosis Tumoral/biosíntesis , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Urol ; 193(5): 1660-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25433307

RESUMEN

PURPOSE: Despite several therapeutic options renal cell carcinoma is associated with a poor clinical outcome. Therefore, we investigated whether combining 5-fluorouracil with the histone deacetylase inhibitor belinostat would exert a synergistic effect on renal cell carcinoma cells in vitro and in vivo. MATERIALS AND METHODS: We used SN12C cells treated with 5-fluorouracil and/or belinostat in vitro and in xenograft experiments in vivo. Cell viability and death mechanisms were assessed by MTS assay and Western blot. To investigate the role of reactive oxygen species we used H2DCF-DA, reactive oxygen species scavengers and the roGFP2 construct. RESULTS: Belinostat potentiated the anticancer effect of 5-fluorouracil. It synergistically induced apoptosis by activating caspases and increasing the subG1 cell population. Effects on reactive oxygen species mediated DNA damage included decreased thioredoxin expression and increased levels of TBP-2, γ-H2AX and Ac-H3. Furthermore, belinostat attenuated the 5-fluorouracil mediated induction of thymidylate synthase via HSP90 hyperacetylation. Co-administration of 5-fluorouracil with belinostat similarly reduced tumor volume and weight, and increased γ-H2AX and Ac-H3 levels in the SN12C xenograft model. CONCLUSIONS: In combination with 5-fluorouracil the targeted inhibitor of histone deacetylase synergistically inhibited renal cancer cell growth by the blockade of thymidylate synthase induction and the induction of reactive oxygen species mediated DNA damage in vitro and in vivo. Our results suggest that combined treatment with belinostat and 5-fluorouracil may represent a promising new approach to renal cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Daño del ADN/efectos de los fármacos , Fluorouracilo/administración & dosificación , Proteínas HSP90 de Choque Térmico/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Sulfonamidas/administración & dosificación , Timidilato Sintasa/efectos de los fármacos , Animales , Quimioterapia Combinada , Humanos , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa , Células Tumorales Cultivadas
8.
Sci Rep ; 4: 5546, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24989033

RESUMEN

Glioblastoma multiforme is the most common malignant brain tumor in adults, with an average survival of less than one year due to its resistance to therapy. Recent studies reported that GBM initiates from CD133-expressing cancer stem cells (CSC). However, the efficacy of CSC targeting is limited. A newly developed approach in cancer treatment is the forced differentiation of cancer cells. Here, we show that the treatment of the novel small molecule, CG500354, into CD133-expressing human primary GBM cells induces growth arrest by cell cycle regulators, p53, p21, p27 and phase-specific cyclins, and neural differentiation, as confirmed by neural progenitor/precursor markers, nestin, GFAP and Tuj1. When GBM-derived cells caused the tumors in NOD/SCID mice, CG500354 induced GBM-derived cells differentiation into Tuj1 and GFAP expressing cells. We next demonstrated that CG500354 plays a tumor-suppressive role via cAMP/CREB signaling pathway. CG500354 increases not only the extracellular cAMP level but also the protein level of PKA and CREB. Additionally, both mimetic substances, Forskolin and Rolipram, revealed comparable results with CG500354. Our findings indicate that induction of growth arrest and neural differentiation via cAMP/CREB signaling pathway by CG500354 treatment suggests the novel targeting of PDE4D in the development of new drugs for brain tumor therapy.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Tiazoles/farmacología , Compuestos de Anilina/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/patología , Diferenciación Celular , Línea Celular Tumoral , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Glioblastoma/patología , Humanos , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal , Esferoides Celulares/efectos de los fármacos , Tiazoles/uso terapéutico , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Bioorg Med Chem Lett ; 24(14): 3142-5, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24894560

RESUMEN

The design, synthesis, and capacity to inhibit HIF prolyl 4-hydroxylases (PHDs) are described for 2-[2-(3-hydroxy-pyridin-2-yl)-thiazol-4-yl]-acetamide analogs. These analogs revealed two kinds of novel scaffolds as PHD2 inhibitors. Synthetic routes were developed for the preparation of their analogs containing the new scaffolds. In addition, the structure-activity relationship (SAR) of the 2-[2-(3-hydroxy-pyridin-2-yl)-thiazol-4-yl]-acetamide derivatives and their biological activities were reported. The complex structure of compound 18 with PHD2 was also obtained for the purpose of more efficient lead optimization.


Asunto(s)
Acetamidas/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Piridinas/farmacología , Tiazoles/farmacología , Acetamidas/síntesis química , Acetamidas/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Modelos Moleculares , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/química
10.
Bioorg Med Chem Lett ; 23(21): 5953-7, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24042008

RESUMEN

A new series of PHD (HIF prolyl 4-hydroxylase) inhibitors was designed based on the X-ray co-crystal structure of FG-2216. Using a lead generation process, a series of [(4-Hydroxyl-benzo[4,5]thieno[3,2-c]pyridine-3-carbonyl)-amino]-acetic acid derivatives was developed as potent PHD2 inhibitors. This class of compounds also showed the ability to stabilize HIF-α, to stimulate EPO secretion in in vitro studies, and to increase hematocrit, red blood cell count, and hemoglobin levels in an animal efficacy study.


Asunto(s)
Ácido Acético/química , Ácido Acético/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Eritropoyetina/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Ácido Acético/farmacocinética , Animales , Línea Celular , Inhibidores Enzimáticos/farmacocinética , Eritropoyesis/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/química , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Masculino , Modelos Moleculares , Piridinas/química , Piridinas/farmacocinética , Piridinas/farmacología
11.
PLoS One ; 8(4): e60383, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23565238

RESUMEN

Glycogen synthase kinase 3α/ß (GSK3α/ß) is a constitutively active serine/threonine kinase involved in multiple physiological processes, such as protein synthesis, stem cell maintenance and apoptosis, and acts as a key suppressor of the Wnt-ß-catenin pathway. In the present study, we examined the therapeutic potential of a novel GSK3 inhibitor, CG0009, in the breast cancer cell lines, BT549, HS578T, MDA-MB-231, NCI/ADR-RES, T47D, MCF7 and MDA-MB-435, from the NCI-60 cancer cell line panel. Assessment of cytotoxicity, apoptosis and changes in estrogen-signaling proteins was performed using cell viability assays, Western blotting and quantitative real-time PCR. CG0009 enhanced the inactivating phosphorylation of GSK3α at Ser21 and GSK3ß at Ser9 and simultaneously decreased activating phosphorylation of GSK3ß at Tyr216, and induced caspase-dependent apoptosis independently of estrogen receptor α (ERα) expression status, which was not observed with the other GSK3 inhibitors examined, including SB216763, kenpaullone and LiCl. CG0009 treatment (1 µmol/L) completely ablated cyclin D1 expression in a time-dependent manner in all the cell lines examined, except T47D. CG0009 alone significantly activated p53, leading to relocation of p53 and Bax to the mitochondria. GSK3 inhibition by CG0009 led to slight upregulation of the ß-catenin target genes, c-Jun and c-Myc, but not cyclin D1, indicating that CG0009-mediated cyclin D1 depletion overwhelms the pro-survival signal of ß-catenin, resulting in cell death. Our findings suggest that the novel GSK3 inhibitor, CG0009, inhibits breast cancer cell growth through cyclin D1 depletion and p53 activation, and may thus offer an innovative therapeutic approach for breast cancers resistant to hormone-based therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Ciclina D1/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Apoptosis/efectos de los fármacos , Benzazepinas/farmacología , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Humanos , Indoles/farmacología , Cloruro de Litio/farmacología , Maleimidas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Anticancer Res ; 32(8): 3161-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22843888

RESUMEN

AIM: To investigate the anticancer effect of histone deacetylase inhibitors (HDACIs) in combination with sorafenib in wild-type and mutant von Hippel-Lindau (VHL)-expressing renal cell carcinomas (RCCs). MATERIALS AND METHODS: We exposed clear cell RCC cells to HDACIs (vorinostat or belinostat) or sorafenib. We performed 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assays, western blotting, flow cytometry and enzyme-linked immunosorbent assays (ELISA) to evaluate mechanisms of cell death, and used CalcuSyn to analyze the potential synergism. RESULTS: HDACIs alone inhibited the growth of clear cell RCC cell lines, increased acetylation of histone 3 and of tubulin, activated caspases-8, -9, and -3, and augmented the sub-G(1) population, independently of VHL and permeability glycoprotein (P-gp). Moreover, pre-treatment of Caki-1 (wild-type VHL) and 786-O (mutant VHL) with HDACIs followed by sorafenib reduced cell viability synergistically via activation of caspases and downregulation of the levels of myeloid leukemia cell differentiation protein (MCL1), phospho-extracellular signal-regulated kinase (ERK), and secreted vascular endothelial growth factor (VEGF). CONCLUSION: Sorafenib is more effective in combination with HDACIs even for clear cell RCCs harboring mutant VHL.


Asunto(s)
Antineoplásicos/farmacología , Bencenosulfonatos/farmacología , Carcinoma de Células Renales/patología , Proliferación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Renales/patología , Piridinas/farmacología , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Ácidos Hidroxámicos/farmacología , Neoplasias Renales/genética , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Sorafenib , Sulfonamidas , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Vorinostat
13.
Invest New Drugs ; 30(2): 435-42, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20978925

RESUMEN

Histone deacetylase (HDAC) plays an important role in cancer onset and progression. Therefore, inhibition of HDAC offers potential as an effective cancer treatment regimen. CG200745, (E)-N(1)-(3-(dimethylamino)propyl)-N(8)-hydroxy-2-((naphthalene-1-loxy)methyl)oct-2-enediamide, is a novel HDAC inhibitor presently undergoing a phase I clinical trial. Enhancement of p53 acetylation by HDAC inhibitors induces cell cycle arrest, differentiation, and apoptosis in cancer cells. The purpose of the present study was to investigate the role of p53 acetylation in the cancer cell death caused by CG200745. CG200745-induced clonogenic cell death was 2-fold greater in RKO cells expressing wild-type p53 than in p53-deficient RC10.1 cells. CG200745 treatment was also cytotoxic to PC-3 human prostate cancer cells, which express wild-type p53. CG200745 increased acetylation of p53 lysine residues K320, K373, and K382. CG200745 induced the accumulation of p53, promoted p53-dependent transactivation, and enhanced the expression of MDM2 and p21(Waf1/Cip1) proteins, which are encoded by p53 target genes. An examination of CG200745 effects on p53 acetylation using cells transfected with various p53 mutants showed that cells expressing p53 K382R mutants were significantly resistant to CG200745-induced clonogenic cell death compared with wild-type p53 cells. Moreover, p53 transactivation in response to CG200745 was suppressed in all cells carrying mutant forms of p53, especially K382R. Taken together, these results suggest that acetylation of p53 at K382 plays an important role in CG200745-induced p53 transactivation and clonogenic cell death.


Asunto(s)
Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Naftalenos/farmacología , Neoplasias de la Próstata/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Semivida , Histonas/metabolismo , Humanos , Lisina , Masculino , Mutación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factores de Tiempo , Activación Transcripcional/efectos de los fármacos , Transfección , Ensayo de Tumor de Célula Madre , Proteína p53 Supresora de Tumor/genética
14.
Invest New Drugs ; 30(4): 1434-42, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21773733

RESUMEN

We synthesized a novel hydroxamate-based pan-histone deacetylase inhibitor (HDACI), CG200745 {(E)-2-(Naphthalen-1-yloxymethyl)-oct-2-enedioic acid 1-[(3-dimethylamino-propyl)-amide] 8-hydroxyamide]}. Like other inhibitors, for example vorinostat and belinostat, CG200745 has the hydroxamic acid moiety to bind zinc at the bottom of catalytic pocket. Firstly, we analyzed its inhibitory activity against histone deacetylase (HDAC) in hormone-dependent LNCaP cells and hormone-independent DU145 and PC3 cells. CG200745 inhibited deacetylation of histone H3 and tubulin as much as vorinostat and belinostat did. CG200745 also inhibited growth of prostate cancer cells, increased sub-G1 population, and activated caspase-9, -3 and -8 in LNCaP, DU145 and PC3 cells. These results indicate that CG200745 induces apoptosis. Next, we examined the effect of CG200745 on cell death induced by docetaxel in DU145 cells in vitro and in vivo. Compared to mono-treatment with each drug, pre-treatment of DU145 cells with docetaxel followed by CG200745 showed synergistic cytotoxicity, and increased the apoptotic sub-G1 population, caspase activation, and tubulin acetylation. Moreover, the combination treatment decreased Mcl-1 and Bcl-(XL). Docetaxel and CG200745 combination reduced tumor size in the DU145 xenograft model. These preclinical results show that combination treatment with docetaxel and new HDACI, CG200745, potentiated anti-tumor effect in hormone-refractory prostate cancer (HRPC) cells via activation of apoptosis.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Naftalenos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Taxoides/uso terapéutico , Proteína bcl-X/metabolismo , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Docetaxel , Sinergismo Farmacológico , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Concentración 50 Inhibidora , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Naftalenos/química , Naftalenos/farmacología , Neoplasias de la Próstata/patología , Taxoides/farmacología , Tubulina (Proteína)/metabolismo
15.
Diabetes ; 60(2): 496-506, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21270261

RESUMEN

OBJECTIVE: Peroxisome proliferator-activated receptor (PPAR)-α/γ dual agonists have been developed to alleviate metabolic disorders. However, several PPARα/γ dual agonists are accompanied with unwanted side effects, including body weight gain, edema, and tissue failure. This study investigated the effects of a novel PPARα/γ dual agonist, CG301269, on metabolic disorders both in vitro and in vivo. RESEARCH DESIGN AND METHODS: Function of CG301269 as a PPARα/γ dual agonist was assessed in vitro by luciferase reporter assay, mammalian one-hybrid assay, and analyses of PPAR target genes. In vitro profiles on fatty acid oxidation and inflammatory responses were acquired by fatty acid oxidation assay and quantitative (q)RT-PCR of proinflammatory genes. In vivo effect of CG301269 was examined in db/db mice. Total body weight and various tissue weights were measured, and hepatic lipid profiles were analyzed. Systemic glucose and insulin tolerance were measured, and the in vivo effect of CG301269 on metabolic genes and proinflammatory genes was examined by qRT-PCR. RESULTS: CG301269 selectively stimulated the transcriptional activities of PPARα and PPARγ. CG301269 enhanced fatty acid oxidation in vitro and ameliorated insulin resistance and hyperlipidemia in vivo. In db/db mice, CG301269 reduced inflammatory responses and fatty liver, without body weight gain. CONCLUSIONS: We demonstrate that CG301269 exhibits beneficial effects on glucose and lipid metabolism by simultaneous activation of both PPARα and PPARγ. Our data suggest that CG301269 would be a potential lead compound against obesity and related metabolic disorders.


Asunto(s)
Peso Corporal/efectos de los fármacos , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Oxazoles/farmacología , PPAR alfa/agonistas , PPAR gamma/agonistas , Tiazolidinas/farmacología , Análisis de Varianza , Animales , Línea Celular , Simulación por Computador , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Hígado/metabolismo , Ratones , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
16.
J Struct Biol ; 174(1): 173-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21094257

RESUMEN

Triclosan (5-chloro-2-(2,4-dichloro-phenoxy)-phenol, TCL) is a well known inhibitor against enoyl-acyl carrier protein reductase (ENR), an enzyme critical for cell-wall synthesis of bacteria. The inhibitory concentration at 50% inhibition (IC(50)) of TCL against the Escherichia coli ENR is 150nM for wild type (WT), 380, 470 and 68,500nM for Ala, Ser and Val mutants, respectively. To understand this high TCL resistance in the G93V mutant, we obtained the crystal structures of mutated ENRs complexed with TCL and NAD(+). The X-ray structural analysis along with the ab initio calculations and molecular dynamics simulations explains the serious consequence in the G93V mutant complex. The major interactions around TCL due to the aromatic(cation)-aromatic and hydrogen bonding interactions are found to be conserved both in WT and mutant complexes. Thus, the overall structural change of protein is minimal except that a flexible α-helical turn around TCL is slightly pushed away due to the presence of the bulky valine group. However, TCL shows substantial edge-to-face aromatic (π)-interactions with both the flexible R192-F203 region and the residues in the close vicinity of G93. The weakening of some edge-to-face aromatic interactions around TCL in the G93V mutant results in serious resistance to TCL. This understanding is beneficial to design new generation of antibiotics which will effectively act on the mutant ENRs.


Asunto(s)
Enoil-ACP Reductasa (NADH)/química , Enoil-ACP Reductasa (NADH)/metabolismo , Triclosán/farmacología , Cristalografía por Rayos X , Farmacorresistencia Bacteriana/genética , Enoil-ACP Reductasa (NADH)/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Simulación de Dinámica Molecular , Mutación , Estructura Secundaria de Proteína , Relación Estructura-Actividad
17.
Breast Cancer Res Treat ; 130(2): 365-75, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21184271

RESUMEN

We previously reported that CG0006, a novel hydroxamate-based pan-histone deacetylase inhibitor (HDACI), suppresses the growth of human cancer cells. Here, we tested the ability of CG0006 to inhibit breast cancer cell proliferation in relation to estrogen receptor (ER) status, and examined changes in the expression of cell-cycle regulatory proteins. CG0006 effects on the proliferation of multiple human cancer cell lines were tested using MTT and MTS assays. Changes in estrogen-signaling proteins and cell-cycle regulatory proteins were examined by western blotting and quantitative RT-PCR, and cell-cycle effects were tested using flow cytometry. CG0006 increased histone H3 and H4 acetylation, up-regulated p21 protein, and promoted cell-cycle arrest, inducing G(2)/M-phase accumulation in ER-positive MCF7 cells, and G(1)- and G(2)/M-phase accumulation in ER-negative MDA-MB-231 cells. In both cell types, CG0006 treatment (1 µM) reduced the levels of the estrogen-signaling proteins ERα and cyclin D1, and promoted massive degradation of cell-cycle regulatory proteins. CG0006 down-regulated the histone deacetylase HDAC6 at the protein level in association with a subsequent increase in Hsp90 and α-tubulin acetylation. HDAC6 depletion using small interfering RNA produced a protein-degradation phenotype similar to that of CG0006 treatment. These findings suggest that CG0006 inhibits breast cancer cell growth by two different pathways: a histone acetylation-dependent pathway, and a non-epigenetic pathway that disrupts chaperone function.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Apoptosis/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Ácidos Hidroxámicos/farmacología , Piperidinas/farmacología , Acetilación , Neoplasias de la Mama , Caspasa 9/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Activación Enzimática , Receptor alfa de Estrógeno/genética , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Biosíntesis de Proteínas , Proteolisis , Interferencia de ARN , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transducción de Señal
18.
Oncol Rep ; 24(6): 1509-14, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21042746

RESUMEN

Chemotherapies for colon cancer have recently advanced. However, there is still a need to develop agents and identify effective regimens for better treatments of colon cancer. Histone deacetylase inhibitors (HDACIs) have shown potential as anti-cancer agents. We investigated the anti-tumor effects of CG2 (an HDACI) in combination with irinotecan, 5-FU, or oxaliplatin. Combinations of CG2 with SN38 (the active form of irinotecan), 5FU, or oxaliplatin were more effective than the agents alone when used to inhibit the growth of HCT116 cells. The protein expressions of acetyl-H3, p21, caspase-3, -8, and -9, PARP, and XIAP were affected in a time- and dose-dependent manner in HCT116 cells treated with the CG2 alone or combined CG2 and SN-38. In HCT116 xenografts, the HDACI CG2 in combination with irinotecan dramatically inhibited tumor growth without showing additive toxicity. These data indicate that CG2 together with irinotecan is a promising combination novel treatment for colon cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Camptotecina/análogos & derivados , Carcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Fluorouracilo/administración & dosificación , Inhibidores de Histona Desacetilasas/administración & dosificación , Ácidos Hidroxámicos/administración & dosificación , Naftalenos/administración & dosificación , Compuestos Organoplatinos/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Camptotecina/administración & dosificación , Carcinoma/patología , Neoplasias del Colon/patología , Relación Dosis-Respuesta a Droga , Células HCT116 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Irinotecán , Ratones , Ratones Desnudos , Modelos Biológicos , Naftalenos/farmacología , Oxaliplatino , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Pharmacol ; 78(5): 877-85, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20724462

RESUMEN

Activation of peroxisome proliferator-activated receptors (PPARs) have been implicated in the treatment of metabolic disorders with different mechanisms; PPARα agonists promote fatty acid oxidation and reduce hyperlipidemia, whereas PPARγ agonists regulate lipid redistribution from visceral fat to subcutaneous fat and enhance insulin sensitivity. To achieve combined benefits from activated PPARs on lipid metabolism and insulin sensitivity, a number of PPARα/γ dual agonists have been developed. However, several adverse effects such as weight gain and organ failure of PPARα/γ dual agonists have been reported. By use of virtual ligand screening, we identified and characterized a novel PPARα/γ dual agonist, (R)-1-(4-(2-(5-methyl-2-p-tolyloxazol-4-yl)ethoxy)benzyl)piperidine-2-carboxylic acid (CG301360), exhibiting the improvement in insulin sensitivity and lipid metabolism. CG301360 selectively stimulated transcriptional activities of PPARα and PPARγ and induced expression of their target genes in a PPARα- and PPARγ-dependent manner. In cultured cells, CG301360 enhanced fatty acid oxidation and glucose uptake and it reduced pro-inflammatory gene expression. In db/db mice, CG301360 also restored insulin sensitivity and lipid homeostasis. Collectively, these data suggest that CG301360 would be a novel PPARα/γ agonist, which might be a potential lead compound to develop against insulin resistance and hyperlipidemia.


Asunto(s)
Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Oxazoles/farmacología , PPAR alfa/agonistas , PPAR delta/agonistas , Ácidos Pipecólicos/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Células Cultivadas , Ciclooxigenasa 2/biosíntesis , Citocinas/biosíntesis , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Ratones Obesos , Oxidación-Reducción , PPAR alfa/fisiología , PPAR delta/fisiología , Estereoisomerismo , Transcripción Genética
20.
Hepatogastroenterology ; 57(99-100): 657-62, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20698245

RESUMEN

BACKGROUND/AIMS: This study was performed to determine the efficacy of histone deacetylase inhibitors in gastric cancer, together with other established regimens. METHODOLOGY: The chemosensitivities of 93 gastric cancer patients to established drugs, and three histone deacetylase inhibitors (SAHA, PXD101, and a novel candidate, CG-2) were evaluated using the histoculture drug response assay. RESULTS: Tumor growth inhibition rates were the highest with cisplatin, followed by PXD101, taxol, docetaxel, and TS-1, in descending order. The response rates were 41.9-68.8%, and 37.6-47.3%, respectively, at an inhibition rate cutoff value of 30%. Synergistic activity was evident with most combinations of established drugs and histone deacetylase inhibitors. Diffuse- or mixed-type carcinomas on Lauren classification were closely associated with increased chemosensitivity to TS-1 (p = 0.044). Node-positive and "other than tubular type" tumors on WHO classification were chemosensitive to cisplatin (p = 0.011 and 0.014, respectively). CG-2 chemosensitivity was markedly associated with low preoperative CA724 level (< or = 4 U/ml) (p = 0.046). CONCLUSIONS: This in vitro chemosensitivity assay validates the comparable chemo-response of gastric cancers to histone deacetylase inhibitors and established drugs, indicating considerable therapeutic efficacy of these agents. Additionally, a number of clinicopathological parameters are significantly associated with specific regimens.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Adenocarcinoma/patología , Adulto , Anciano , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA