Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
J Med Chem ; 65(3): 2434-2457, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35043615

RESUMEN

A series of 2-phenylthiazole analogues were designed and synthesized as potential histone deacetylase 6 (HDAC6) inhibitors based on compound 12c (an HDAC6/tubulin dual inhibitor discovered by us recently) and CAY10603 (a known HDAC6 inhibitor). Among them, compound XP5 was the most potent HDAC6 inhibitor with an IC50 of 31 nM and excellent HDAC6 selectivity (SI = 338 for HDAC6 over HDAC3). XP5 also displayed high antiproliferative activity against various cancer cell lines including the HDACi-resistant YCC3/7 gastric cancer cells (IC50 = 0.16-2.31 µM), better than CAY10603. Further, XP5 (50 mg/kg) exhibited significant antitumor efficacy in a melanoma tumor model with a tumor growth inhibition (TGI) of 63% without apparent toxicity. Moreover, XP5 efficiently enhanced the in vivo antitumor immune response when combined with a small-molecule PD-L1 inhibitor, as demonstrated by the increased tumor-infiltrating lymphocytes and reduced PD-L1 expression levels. Taken together, the above results suggest that XP5 is a promising HDAC6 inhibitor deserving further investigation.


Asunto(s)
Antineoplásicos/uso terapéutico , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/uso terapéutico , Inmunidad/efectos de los fármacos , Melanoma/tratamiento farmacológico , Tiazoles/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacocinética , Ácidos Hidroxámicos/uso terapéutico , Ácidos Hidroxámicos/toxicidad , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Masculino , Melanoma/terapia , Ratones , Estructura Molecular , Ratas Sprague-Dawley , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/farmacocinética , Tiazoles/toxicidad
2.
Toxicol Lett ; 356: 121-131, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34923047

RESUMEN

Oxoguanine glycosylase 1 (OGG1) is both a DNA repair enzyme and an epigenetic modifier. We assessed behavioural abnormalities in OGG1-deficient progeny exposed once in utero to a low dose of ethanol (EtOH) and treated postnatally with a global histone deacetylase inhibitor, trichostatin A (TSA). The goal of this study was to determine if neurodevelopmental disorders initiated in the fetal brain by in utero exposure to EtOH could be mitigated by postnatal treatment with TSA. EtOH and TSA alone improved preference for novel location (short-term, 90 min) and novel object (long-term, 24 h) sex- and OGG1-dependently. Combined EtOH/TSA treatment reversed these effects in the short-term novel location test sex- and OGG1-dependently. In females but not males, the incidence of high shredders of nesting material was not altered by either TSA or EtOH alone, but was reduced by combined EtOH/TSA treatment in +/+ progeny. Similar but non-significant effects were observed in Ogg1 -/- females. Accelerated rotarod performance was enhanced by both EtOH and TSA alone in only male Ogg1 +/+ but not -/- progeny, and was not altered by combined EtOH/TSA exposure. The OGG1-dependent effects of EtOH and TSA particularly on novel location and the incidence of high shredders, and the reversal of EtOH effects on these parameters by combined EtOH/TSA treatment, suggests both xenobiotics may alter behaviour via a mechanism involving OGG1 acting as an epigenetic modifier, in addition to repairing DNA damage. These preliminary results suggest that the postnatal use of more selective epigenetic modifying agents may constitute a novel strategy for mitigating some components of ROS-initiated neurodevelopmental disorders.


Asunto(s)
ADN Glicosilasas/metabolismo , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/toxicidad , Animales , Conducta Animal/efectos de los fármacos , ADN Glicosilasas/genética , Etanol/administración & dosificación , Etanol/toxicidad , Femenino , Genotipo , Inhibidores de Histona Desacetilasas/administración & dosificación , Masculino , Ratones , Ratones Noqueados , Embarazo , Efectos Tardíos de la Exposición Prenatal
3.
Invest Ophthalmol Vis Sci ; 62(14): 8, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34757417

RESUMEN

Purpose: Current melphalan-based regimens for intravitreal chemotherapy for retinoblastoma vitreous seeds are effective but toxic to the retina. Thus, alternative agents are needed. Based on the known biology of histone deacetylases (HDACs) in the retinoblastoma pathway, we systematically studied whether the HDAC inhibitor belinostat is a viable, molecularly targeted alternative agent for intravitreal delivery that might provide comparable efficacy, without toxicity. Methods: In vivo pharmacokinetic experiments in rabbits and in vitro cytotoxicity experiments were performed to determine the 90% inhibitory concentration (IC90). Functional toxicity by electroretinography and structural toxicity by optical coherence tomography (OCT), OCT angiography, and histopathology were evaluated in rabbits following three injections of belinostat 350 µg (2× IC90) or 700 µg (4× IC90), compared with melphalan 12.5 µg (rabbit equivalent of the human dose). The relative efficacy of intravitreal belinostat versus melphalan to treat WERI-Rb1 human cell xenografts in rabbit eyes was directly quantified. RNA sequencing was used to assess belinostat-induced changes in RB cell gene expression. Results: The maximum nontoxic dose of belinostat was 350 µg, which caused no reductions in electroretinography parameters, retinal microvascular loss on OCT angiography, or retinal degeneration. Melphalan caused severe retinal structural and functional toxicity. Belinostat 350 µg (equivalent to 700 µg in the larger human eye) was equally effective at eradicating vitreous seeds in the rabbit xenograft model compared with melphalan (95.5% reduction for belinostat, P < 0.001; 89.4% reduction for melphalan, P < 0.001; belinostat vs. melphalan, P = 0.10). Even 700 µg belinostat (equivalent to 1400 µg in humans) caused only minimal toxicity. Widespread changes in gene expression resulted. Conclusions: Molecularly targeted inhibition of HDACs with intravitreal belinostat was equally effective as standard-of-care melphalan but without retinal toxicity. Belinostat may therefore be an attractive agent to pursue clinically for intravitreal treatment of retinoblastoma.


Asunto(s)
Modelos Animales de Enfermedad , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Siembra Neoplásica , Retina/efectos de los fármacos , Neoplasias de la Retina/tratamiento farmacológico , Retinoblastoma/tratamiento farmacológico , Sulfonamidas/uso terapéutico , Animales , Anexina A5 , Antineoplásicos Alquilantes/uso terapéutico , Electrorretinografía , Angiografía con Fluoresceína , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/farmacocinética , Ácidos Hidroxámicos/toxicidad , Inyecciones Intravítreas , Dosis Máxima Tolerada , Melfalán/uso terapéutico , Conejos , Retina/fisiología , Neoplasias de la Retina/diagnóstico , Neoplasias de la Retina/fisiopatología , Retinoblastoma/diagnóstico , Retinoblastoma/fisiopatología , Estudios Retrospectivos , Sulfonamidas/farmacocinética , Sulfonamidas/toxicidad , Tomografía de Coherencia Óptica , Cuerpo Vítreo/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Med Chem ; 64(2): 1116-1126, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33356256

RESUMEN

Due to the evolution and development of antifungal drug resistance, limited efficacy of existing drugs has led to high mortality in patients with serious fungal infections. To develop novel antifungal therapeutic strategies, herein a series of carboline fungal histone deacetylase (HDAC) inhibitors were designed and synthesized, which had potent synergistic effects with fluconazole against resistant Candida albicans infection. In particular, compound D12 showed excellent in vitro and in vivo synergistic antifungal efficacy with fluconazole to treat azole-resistant candidiasis. It cooperated with fluconazole in reducing the virulence of C. albicans by blocking morphological mutual transformation and inhibiting biofilm formation. Mechanism studies revealed that the reversion of drug resistance was due to downregulation of the expression of the azole target gene ERG11 and efflux gene CDR1. Taken together, fungal HDAC inhibitor D12 offered a promising lead compound for combinational treatment of azole-resistant candidiasis.


Asunto(s)
Azoles/uso terapéutico , Candida albicans/efectos de los fármacos , Candidiasis/tratamiento farmacológico , Carbolinas/síntesis química , Carbolinas/uso terapéutico , Farmacorresistencia Fúngica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/uso terapéutico , Animales , Biopelículas/efectos de los fármacos , Candida albicans/enzimología , Candidiasis/microbiología , Carbolinas/toxicidad , Quimioterapia Combinada , Femenino , Fluconazol/farmacología , Proteínas Fúngicas/efectos de los fármacos , Hongos/efectos de los fármacos , Hongos/enzimología , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Hígado/patología , Proteínas de Transporte de Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Pruebas de Sensibilidad Microbiana
6.
Toxicol Lett ; 338: 51-57, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33290829

RESUMEN

Belinostat is a pan-histone deacetylase (HDAC) inhibitor which recently approved for the treatment of relapsed/refractory Peripheral T-cell lymphomas (PTCL). To assess drug-drug interactions (DDIs) potential of belinostat via inhibition of UDP-glucuronosyltransferases (UGTs), the effects of belinostat on UGTs activities were investigated using the non-selective probe substrate 4-methylumbelliferone (4-MU) and trifluoperazine (TFP) by UPLC-MS/MS. Belinostat exhibited a wide range of inhibition against UGTs activities, particularly a potent non-competitive inhibition against UGT1A3, and weak inhibition against UGT1A1, 1A7, 1A8, 2B4 and 2B7. Further, in vitro-in vivo extrapolation (IVIVE) approaches were used to predict the risk of DDI arising from inhibition of UGTs. Our data indicate that the intravenous infusion of belinostat at clinical available dose can contribute a significant increase to the AUC of co-administrated drugs primarily cleared by UGT1A3 or UGT1A1, which will result in potential DDIs. In contrast, oral administrated belinostat is unlikely to cause significant DDIs through inhibition of glucuronidation.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Glucuronosiltransferasa/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Sulfonamidas/farmacología , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/toxicidad , Interacciones Farmacológicas , Glucuronosiltransferasa/metabolismo , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/toxicidad , Infusiones Intravenosas , Cinética , Sulfonamidas/administración & dosificación , Sulfonamidas/toxicidad
7.
Front Immunol ; 11: 579158, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33362765

RESUMEN

HIV/SIV persistence in latent reservoirs requires lifelong antiretroviral treatment and calls for effective cure strategies. Romidepsin (RMD), a histone deacetylase inhibitor, was reported to reactivate HIV/SIV from reservoirs in virus-suppressed individuals. We characterized in detail the pharmacokinetics and safety profile of RMD in three SIV-naïve rhesus macaques which received two rounds of treatment. In plasma, RMD mean terminal half-life was 15.3 h. In comparison, RMD mean terminal half-life was much longer in tissues: 110 h in the lymph nodes (LNs) and 28 h in gastrointestinal tract. RMD administration was accompanied by transient liver and systemic toxicity. Isoflurane anesthesia induced near-immediate transient lymphopenia, which was further exacerbated and extended with the extensive immune modifications by RMD. The effect of RMD on circulating immune cells was complex: (i) slight increase in lymphocyte death rates; (ii) transient, robust increase in neutrophils; (iii) massive downregulation of lymphocyte surface markers; (iv) important migration of CD3+ T cells to the gut and LNs; and (v) hindrance to CD8+ T cell functionality, yet without reaching significance. Our results show that, in contrast to transient plasma concentrations, RMD has a long-term presence in tissues, with multiple immunomodulatory effects and minimal to moderate kidney, liver, and lymphocyte toxicities. As such, we concluded that RMD can be used for "shock and kill" approaches, preferentially in combination with other latency reversal agents or cytotoxic T lymphocyte boosting strategies with consideration taken for adverse effects.


Asunto(s)
Depsipéptidos/farmacocinética , Inhibidores de Histona Desacetilasas/farmacocinética , Sistema Inmunológico/efectos de los fármacos , Animales , Quimiotaxis de Leucocito/efectos de los fármacos , Depsipéptidos/administración & dosificación , Depsipéptidos/toxicidad , Femenino , Semivida , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/toxicidad , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Infusiones Intravenosas , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/metabolismo , Linfopenia/inducido químicamente , Linfopenia/inmunología , Macaca mulatta , Masculino , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Distribución Tisular
8.
Int J Mol Sci ; 21(13)2020 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-32635356

RESUMEN

Histone deacetylase inhibitors (HDACi) are already approved for the therapy of leukemias. Since they are also emerging candidate compounds for the treatment of non-malignant diseases, HDACi with a wide therapeutic window and low hazard potential are desirable. Here, we investigated a panel of 12 novel hydroxamic acid- and benzamide-type HDACi employing non-malignant V79 hamster cells as toxicology guideline-conform in vitro model. HDACi causing a ≥10-fold preferential cytotoxicity in malignant neuroblastoma over non-malignant V79 cells were selected for further genotoxic hazard analysis, including vorinostat and entinostat for control. All HDACi selected, (i.e., KSK64, TOK77, DDK137 and MPK77) were clastogenic and evoked DNA strand breaks in non-malignant V79 cells as demonstrated by micronucleus and comet assays, histone H2AX foci formation analyses (γH2AX), DNA damage response (DDR) assays as well as employing DNA double-strand break (DSB) repair-defective VC8 hamster cells. Genetic instability induced by hydroxamic acid-type HDACi seems to be independent of bulky DNA adduct formation as concluded from the analysis of nucleotide excision repair (NER) deficient mutants. Summarizing, KSK64 revealed the highest genotoxic hazard and DDR stimulating potential, while TOK77 and MPK77 showed the lowest DNA damaging capacity. Therefore, these compounds are suggested as the most promising novel candidate HDACi for subsequent pre-clinical in vivo studies.


Asunto(s)
Benzamidas/toxicidad , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/toxicidad , Pruebas de Mutagenicidad/métodos , Mutágenos/toxicidad , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Ensayo Cometa , Cricetinae , Roturas del ADN de Doble Cadena , Roturas del ADN de Cadena Simple , Histonas/química , Histonas/metabolismo , Humanos , Pruebas de Micronúcleos , Fosforilación , Vorinostat/toxicidad
9.
J Med Chem ; 63(9): 4701-4715, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32267687

RESUMEN

While proteasome inhibitors such as bortezomib showed satisfactory clinical benefits in the initial treatment of multiple myeloma (MM), drug resistance and relapse are unavoidable. Recent studies suggested inhibition of histone deacetylases (HDACs) restored sensitivity of bortezomib-resistant MM. Hence, we designed dual inhibitors targeting both HDACs and proteasomes to address the resistance of bortezomib. The most potent inhibitors, ZY-2 and ZY-13 showed excellent inhibition against proteasome and good selectivity against HDACs. In particular, ZY-2 not only exhibited good antiproliferative activities on the MM cell lines RPMI-8226, U266, and KM3 (IC50 values of 6.66, 4.31, and 10.1 nM, respectively) but also showed more potent antiproliferative activities against the bortezomib-resistant MM cell line KM3/BTZ compared with bortezomib (IC50 values of 8.98 vs. 226 nM, P < 0.01) and even better than the combination of the HDAC inhibitor MS-275 and bortezomib (1:1) (IC50 values of 8.98 vs. 98.0 nM, P < 0.01).


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Péptidos/farmacología , Inhibidores de Proteasoma/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Ácidos Borónicos/síntesis química , Ácidos Borónicos/metabolismo , Ácidos Borónicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Histona Desacetilasa 1/química , Histona Desacetilasa 1/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/metabolismo , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Simulación del Acoplamiento Molecular , Mieloma Múltiple/tratamiento farmacológico , Péptidos/síntesis química , Péptidos/metabolismo , Péptidos/toxicidad , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/síntesis química , Inhibidores de Proteasoma/metabolismo , Inhibidores de Proteasoma/toxicidad , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos
10.
CNS Neurol Disord Drug Targets ; 19(1): 55-65, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31858907

RESUMEN

BACKGROUND & OBJECTIVE: The pharmacokinetics and acute toxicity of a histone deacetylase inhibitor, Scriptaid, was unknown in the mouse. The aim of this study was to determine the pharmacokinetics, acute toxicity, and tissue distribution of Scriptaid, a new histone deacetylase inhibitor, in mice, and its neuroprotective efficacy in a mouse intracranial hemorrhage (ICH) model. METHODS: The pharmacokinetics, acute toxicity, and tissue distribution were determined in C57BL/6 male and female mice after the intraperitoneal administration of a single dose. Behavioral tests, as well as investigations of brain atrophy and white matter injury, were used to evaluate the neuroprotective effect of Scriptaid after ICH. Western blotting was used to investigate if Scriptaid could offer antiinflammatory benefits after ICH. RESULTS: No significant differences were observed in body weight or brain histopathology between the group that received Scriptaid at 50 mg/kg and the group that received dimethyl sulfoxide (control). The pharmacokinetics of Scriptaid in mice was nonlinear, and it was cleared rapidly at low doses and slowly at higher doses. Consistent with the pharmacokinetic data, Scriptaid was found to distribute in several tissues, including the spleen and kidneys. In the ICH model, we found that Scriptaid could reduce neurological deficits, brain atrophy, and white matter injury in a dose-dependent manner. Western blotting results demonstrated that Scriptaid could decrease the expression of pro-inflammatory cytokines IL1ß and TNFα, as well as iNOS, after ICH. CONCLUSION: These findings indicate that Scriptaid is safe and can alleviate brain injury after ICH, thereby providing a foundation for the pharmacological action of Scriptaid in the treatment of brain injury after ICH.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacocinética , Hidroxilaminas/farmacocinética , Hemorragias Intracraneales/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Quinolinas/farmacocinética , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Inhibidores de Histona Desacetilasas/uso terapéutico , Inhibidores de Histona Desacetilasas/toxicidad , Hidroxilaminas/uso terapéutico , Hidroxilaminas/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroprotección/efectos de los fármacos , Quinolinas/uso terapéutico , Quinolinas/toxicidad
11.
Eur J Med Chem ; 176: 195-207, 2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31103900

RESUMEN

Previously, we focused on a series of 2-aminobenzamide-based histone deacetylase (HDAC) inhibitors, compound 9 of which displayed potent HDAC inhibitory activity against HDAC1 and HDAC2, and moderate anti-proliferative activity against several cancer cell lines. In the current study, we have designed and synthesized a series of novel HDAC inhibitors based on thioether moiety with 9 as a lead compound. Representative compounds12 g and 12 h showed apparently potent anti-proliferative activities against five solid cancer cell lines: A549, HCT116, Hela, A375 and SMMC7721, and low cytotoxicity against NIH 3T3 normal cells. Especially, 12 g and 12 h also revealed potent HDAC inhibitory activity against HDAC1, 2 and 3. In addition, the two compounds could arrest cell cycle in G2/M phase and promote cell apoptosis. Moreover, they showed extended inhibition of colony formation and effectively blocked cell migration towards A549 cancer cells. Furthermore, 12 g and 12 h possessed better pharmacokinetic properties than the lead compound 9. Benefiting from these results, we also explored 12 g and 12 h in the A549 xenografts model for in vivo antitumor activity. The in vivo experiment indicated that 12 g and 12 h could evidently augment antitumor activity (TGI = 56.9% and 62.7% respectively).


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Sulfuros/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Benzamidas/síntesis química , Benzamidas/farmacocinética , Benzamidas/toxicidad , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Ratones , Simulación del Acoplamiento Molecular , Células 3T3 NIH , Sulfuros/síntesis química , Sulfuros/farmacocinética , Sulfuros/toxicidad , Ensayos Antitumor por Modelo de Xenoinjerto
12.
ChemMedChem ; 14(9): 912-926, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30664827

RESUMEN

Novel malaria intervention strategies are of great importance, given the development of drug resistance in malaria-endemic countries. In this regard, histone deacetylases (HDACs) have emerged as new and promising malaria drug targets. In this work, we present the design, synthesis, and biological evaluation of 20 novel HDAC inhibitors with antiplasmodial activity. Based on a previously discovered peptoid-based hit compound, we modified all regions of the peptoid scaffold by using a one-pot multicomponent pathway and submonomer routes to gain a deeper understanding of the structure-activity and structure-toxicity relationships. Most compounds displayed potent activity against asexual blood-stage P. falciparum parasites, with IC50 values in the range of 0.0052-0.25 µm and promising selectivity over mammalian cells (SIPf3D7/HepG2 : 170-1483). In addition, several compounds showed encouraging sub-micromolar activity against P. berghei exo-erythrocytic forms (PbEEF). Our study led to the discovery of the hit compound N-(2-(benzylamino)-2-oxoethyl)-N-(4-(hydroxycarbamoyl)benzyl)-4-isopropylbenzamide (2 h) as a potent and parasite-specific dual-stage antiplasmodial HDAC inhibitor (IC50 Pf3D7=0.0052 µm, IC50 PbEEF=0.016 µm).


Asunto(s)
Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Peptoides/química , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Acetilación , Animales , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas/toxicidad , Histonas/metabolismo , Humanos , Concentración 50 Inhibidora , Relación Estructura-Actividad
13.
Drug Chem Toxicol ; 42(5): 526-535, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29681204

RESUMEN

The aims of the present research are to further validate the application of the improved three-dimensional (3 D) rat testicular cell co-culture model to evaluate the effects of various reprotoxic chemicals on the function of the main somatic cells, as well as on spermatogonial cell differentiation and even spermatogenesis, and to investigate the specific toxicant mechanisms in testes treated with HZ1006, a hydroxamate-based a hydroxamate-based histone deacetylase inhibitor (HDACI). Based on the characteristics of HZ1006, the appropriate exposure duration (8, 16, or 24 days), dosage (0, 3.125, 6.25, 12.5, or 25 µM) and toxic endpoints suitable for detection were selected in the experiments. The results showed inhibition of cell proliferation, reduced testosterone levels, and decreased spermatogonial cell meiosis-specific gene expression, as well as decreased protein levels of androgen receptor (AR) and decreased expression of the AR target gene PSA, accompanied by inhibition of Hdac6 expression after HZ1006 exposure in the 3 D rat testicular cell co-culture model. These findings indicate that the improved 3 D rat testicular cell co-culture model we have established has the potential to become a new testicular toxicity test system that can be used to test toxic characteristics and mechanisms of new compounds and has good application prospects, although more research on the model is required.


Asunto(s)
Cinamatos/toxicidad , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/toxicidad , Espermatogonias/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cinamatos/química , Técnicas de Cocultivo , Femenino , Inhibidores de Histona Desacetilasas/química , Ácidos Hidroxámicos/química , Masculino , Estructura Molecular , Ratas Sprague-Dawley , Espermatogonias/metabolismo , Espermatogonias/patología , Testículo/metabolismo , Testículo/patología , Testosterona/metabolismo
14.
CNS Neurosci Ther ; 25(2): 175-186, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29978554

RESUMEN

AIMS: Recently, histone deacetylase (HDAC) inhibitors are considered a possible therapeutic strategy in Alzheimer's disease (AD). However, HDACi treatments exhibit diverse functions with unfavorable effects in AD. Thus, the development of selective HDACi without side effects is urgently needed. METHODS: HDACi, namely, BML210, MGCD0103, PXD101, and Droxinostat, were screened in mouse hippocampal primary cultures incubated with oligomeric Aß25-35 (50 µmol/L). MGCD0103 was chosen for in vivo tests and was intraperitoneally injected into C57BL/6J mice (0.5 mg/kg, once per day) for 4 weeks following an intrahippocampal CA1 injection of oligomeric Aß25-35 . Brain samples were collected for pathological analyses after the behavioral analyses including open- field test (OFT), elevated plus maze (EPM), Y-maze, and Morris water maze (MWM). RESULTS: Among the HDACi, MGCD0103 exhibited significant neuroprotection against the Aß toxicity in primary cultures. MGCD0103 coattenuated cognitive deficits and anxiety against Aß damage in mice. MGCD0103 further ameliorated pathological features such as the levels of acetylated histone 3 at Lys 9 site (H3K9) and α-tubulin, synaptophysin, Aß, tau protein phosphorylation, and serotonergic neuron loss against Aß toxicity. Furthermore, chronic MGCD0103 treatment did not show liver or kidney toxicity in mice. CONCLUSIONS: These results reveal MGCD0103 could be a potential therapeutic agent against AD.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Ansiedad/tratamiento farmacológico , Benzamidas/uso terapéutico , Disfunción Cognitiva/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Fragmentos de Péptidos/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Animales , Ansiedad/inducido químicamente , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Benzamidas/toxicidad , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/efectos de los fármacos , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/psicología , Femenino , Hipocampo/citología , Hipocampo/efectos de los fármacos , Inhibidores de Histona Desacetilasas/toxicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/efectos de los fármacos , Cultivo Primario de Células , Pirimidinas/toxicidad
15.
Toxicol Appl Pharmacol ; 350: 43-51, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29733868

RESUMEN

Histone deacetylases (HDACs), which regulate transcription and specific functions such as tumor suppression by p53, are frequently altered in tumors and have a contentious role in carcinogenesis. HDAC inhibitors, which have a long history of use in psychiatry and neurology, have recently been tested as possible treatments for tumors. Belinostat received regulatory approval in the USA on July 3, 2014, for use against peripheral T-cell lymphoma. However, the unavailability of information on belinostat genotoxicity in normal cells and the molecular mechanisms involved in the genetic instability after exposure to belinostat encouraged us to conduct this study. Our data showed that the exposure of mice to belinostat at the recommended human doses induced chromosome breakage, whole-chromosome lagging, and oxidative DNA damage in bone marrow cells in a dose-dependent manner. The expression levels of 84 genes involved in the DNA damage signaling pathway were evaluated by using an RT2 Profiler PCR array. Belinostat exposure altered the expression of 25 genes, with statistically significant changes observed in 17 genes. The array results were supported by RT-PCR and western blotting experiments. Collectively, our results showed that belinostat exposure caused oxidative DNA damage and downregulated the expression of genes involved in DNA damage repair, which may be responsible for belinostat-induced genomic instability. Thus, the clinical usage of this drug should be weighed against the hazards of carcinogenesis, and the observed genotoxicity profile of belinostat may support further development of efficient HDAC inhibitors with weaker genotoxicity.


Asunto(s)
Análisis Citogenético/métodos , Perfilación de la Expresión Génica/métodos , Inestabilidad Genómica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/toxicidad , Transducción de Señal/efectos de los fármacos , Sulfonamidas/toxicidad , Animales , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Relación Dosis-Respuesta a Droga , Inestabilidad Genómica/fisiología , Masculino , Ratones , Transducción de Señal/fisiología
16.
J Med Chem ; 61(8): 3454-3477, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29589441

RESUMEN

Epigenetic modifiers of the histone deacetylase (HDAC) family contribute to autoimmunity, cancer, HIV infection, inflammation, and neurodegeneration. Hence, histone deacetylase inhibitors (HDACi), which alter protein acetylation, gene expression patterns, and cell fate decisions, represent promising new drugs for the therapy of these diseases. Whereas pan-HDACi inhibit all 11 Zn2+-dependent histone deacetylases (HDACs) and cause a broad spectrum of side effects, specific inhibitors of histone deacetylase 6 (HDAC6i) are supposed to have less side effects. We present the synthesis and biological evaluation of Marbostats, novel HDAC6i that contain the hydroxamic acid moiety linked to tetrahydro-ß-carboline derivatives. Our lead compound Marbostat-100 is a more potent and more selective HDAC6i than previously established well-characterized compounds in vitro as well as in cells. Moreover, Marbostat-100 is well tolerated by mice and effective against collagen type II induced arthritis. Thus, Marbostat-100 represents a most selective known HDAC6i and the possibility for clinical evaluation of a HDAC isoform-specific drug.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antirreumáticos/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Benzamidas/uso terapéutico , Histona Desacetilasa 6/metabolismo , Inhibidores de Histona Desacetilasas/uso terapéutico , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/farmacología , Antiinflamatorios/toxicidad , Antirreumáticos/síntesis química , Antirreumáticos/farmacología , Antirreumáticos/toxicidad , Artritis Experimental/inducido químicamente , Artritis Reumatoide/inducido químicamente , Benzamidas/líquido cefalorraquídeo , Benzamidas/farmacología , Benzamidas/toxicidad , Sitios de Unión , Carbolinas/síntesis química , Carbolinas/farmacología , Carbolinas/uso terapéutico , Carbolinas/toxicidad , Línea Celular Tumoral , Colágeno Tipo II , Células HEK293 , Histona Desacetilasa 6/química , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Ácidos Hidroxámicos/toxicidad , Masculino , Ratones Endogámicos DBA , Simulación del Acoplamiento Molecular , Pez Cebra
17.
Bioorg Med Chem ; 26(3): 747-757, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29317150

RESUMEN

Both histone deacetylase (HDAC) and fibroblast growth factor receptor (FGFR) are important targets for cancer therapy. Although combining dual HDAC pharmacophore with tyrosine kinase inhibitors (TKIs) had achieved a successful progress, dual HDAC/FGFR1 inhibitors haven't been reported yet. Herein, we designed a series of hybrids bearing 1H-indazol-3-amine and benzohydroxamic acids scaffold with scaffold hopping and molecular hybridization strategies. Among them, compound 7j showed the most potent inhibitory activity against HDAC6 with IC50 of 34 nM and exhibited the great inhibitory activities against a human breast cancer cell line MCF-7 with IC50 of 9 µM in vitro. Meanwhile, the compound also exhibited moderate FGFR1 inhibitory activities. This study provides new tool compounds for further exploration of dual HDAC/FGFR1 inhibition.


Asunto(s)
Diseño de Fármacos , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/síntesis química , Indazoles/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Histona Desacetilasa 6/metabolismo , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Indazoles/toxicidad , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Células MCF-7 , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Relación Estructura-Actividad
18.
Int J Mol Sci ; 19(1)2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29304031

RESUMEN

Neuroblastoma (NBL) originates from undifferentiated cells of the sympathetic nervous system. Chemotherapy is judged to be suitable for successful treatment of this disease. Here, the influence of histone deacetylase (HDAC) inhibitor valproate (VPA) combined with DNA-damaging chemotherapeutic, ellipticine, on UKF-NB-4 and SH-SY5Y neuroblastoma cells was investigated. Treatment of these cells with ellipticine in combination with VPA led to the synergism of their anticancer efficacy. The effect is more pronounced in the UKF-NB-4 cell line, the line with N-myc amplification, than in SH-SY5Y cells. This was associated with caspase-3-dependent induction of apoptosis in UKF-NB-4 cells. The increase in cytotoxicity of ellipticine in UKF-NB-4 by VPA is dictated by the sequence of drug administration; the increased cytotoxicity was seen only after either simultaneous exposure to these drugs or after pretreatment of cells with ellipticine before their treatment with VPA. The synergism of treatment of cells with VPA and ellipticine seems to be connected with increased acetylation of histones H3 and H4. Further, co-treatment of cells with ellipticine and VPA increased the formation of ellipticine-derived DNA adducts, which indicates an easier accessibility of ellipticine to DNA in cells by its co-treatment with VPA and also resulted in higher ellipticine cytotoxicity. The results are promising for in vivo studies and perhaps later for clinical studies of combined treatment of children suffering from high-risk NBL.


Asunto(s)
Elipticinas/toxicidad , Inhibidores de Histona Desacetilasas/toxicidad , Mutágenos/toxicidad , Neuronas/efectos de los fármacos , Ácido Valproico/toxicidad , Apoptosis , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Neuroblastoma/metabolismo , Neuronas/metabolismo
19.
Int J Mol Sci ; 18(12)2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29215575

RESUMEN

Effective DNA repair enables cancer cells to survive DNA damage induced by chemotherapeutic or radiotherapeutic treatments. Therefore, inhibiting DNA repair pathways is a promising therapeutic strategy for increasing the efficacy of such treatments. In this study, we found that dihydrocoumarin (DHC), a flavoring agent, causes deficiencies in double-stand break (DSB) repair and prolonged DNA damage checkpoint recovery in yeast. Following DNA damage, Rad52 recombinase was revealed to be inhibited by DHC, which results in deficiencies in DSB repair and prolonged DNA damage checkpoint recovery. The deletion of RPD3, a class I histone deacetylase (HDAC), was found to mimic DHC-induced suppression of Rad52 expression, suggesting that the HDAC inhibitor activity of DHC is critical to DSB repair and DNA damage sensitivity. Overall, our findings delineate the regulatory mechanisms of DHC in DSB repair and suggest that it might potentially be used as an inhibitor of the DNA repair pathway in human cells.


Asunto(s)
Cumarinas/toxicidad , Daño del ADN , Aromatizantes/toxicidad , Inhibidores de Histona Desacetilasas/toxicidad , Proteína Recombinante y Reparadora de ADN Rad52/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Cumarinas/farmacología , Aromatizantes/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Reparación del ADN por Recombinación , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
20.
Bioorg Med Chem Lett ; 27(21): 4885-4888, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28947154

RESUMEN

A series of hydroxamic acid-based HDACIs with 4-aminoquinazolinyl moieties as capping groups was profiled. Most compounds showed more potent HDACs inhibition activity than clinically used drug SAHA. Among them, compounds 5f and 5h selectively inhibited HDAC 1,2 over HDAC8, and showed strong activity in several cellular assays, not possessing significant toxicity to primary human cells and hERG inhibition. Strikingly, 5f possessed acceptable pharmacokinetic characteristics and exhibited significant antitumor activity in an A549 xenograft model study at well tolerated doses.


Asunto(s)
Diseño de Fármacos , Inhibidores de Histona Desacetilasas/síntesis química , Histona Desacetilasas/metabolismo , Quinazolinas/química , Células A549 , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ensayos de Selección de Medicamentos Antitumorales , Semivida , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/metabolismo , Inhibidores de Histona Desacetilasas/uso terapéutico , Inhibidores de Histona Desacetilasas/toxicidad , Histona Desacetilasas/química , Humanos , Concentración 50 Inhibidora , Ratones , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA