Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
J Immunother Cancer ; 11(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37419511

RESUMEN

BACKGROUND: Most immunotherapies approved for clinical use rely on the use of recombinant proteins and cell-based approaches, rendering their manufacturing expensive and logistics onerous. The identification of novel small molecule immunotherapeutic agents might overcome such limitations. METHOD: For immunopharmacological screening campaigns, we built an artificial miniature immune system in which dendritic cells (DCs) derived from immature precursors present MHC (major histocompatibility complex) class I-restricted antigen to a T-cell hybridoma that then secretes interleukin-2 (IL-2). RESULTS: The screening of three drug libraries relevant to known signaling pathways, FDA (Food and Drug Administration)-approved drugs and neuroendocrine factors yielded two major hits, astemizole and ikarugamycin. Mechanistically, ikarugamycin turned out to act on DCs to inhibit hexokinase 2, hence stimulating their antigen presenting potential. In contrast, astemizole acts as a histamine H1 receptor (H1R1) antagonist to activate T cells in a non-specific, DC-independent fashion. Astemizole induced the production of IL-2 and interferon-γ (IFN-γ) by CD4+ and CD8+ T cells both in vitro and in vivo. Both ikarugamycin and astemizole improved the anticancer activity of the immunogenic chemotherapeutic agent oxaliplatin in a T cell-dependent fashion. Of note, astemizole enhanced the CD8+/Foxp3+ ratio in the tumor immune infiltrate as well as IFN-γ production by local CD8+ T lymphocytes. In patients with cancer, high H1R1 expression correlated with low infiltration by TH1 cells, as well as with signs of T-cell exhaustion. The combination of astemizole and oxaliplatin was able to cure the majority of mice bearing orthotopic non-small cell lung cancers (NSCLC), then inducing a state of protective long-term immune memory. The NSCLC-eradicating effect of astemizole plus oxaliplatin was lost on depletion of either CD4+ or CD8+ T cells, as well as on neutralization of IFN-γ. CONCLUSIONS: These findings underscore the potential utility of this screening system for the identification of immunostimulatory drugs with anticancer effects.


Asunto(s)
Linfocitos T CD8-positivos , Interleucina-2 , Estados Unidos , Ratones , Animales , Interleucina-2/metabolismo , Astemizol/farmacología , Astemizol/uso terapéutico , Astemizol/metabolismo , Oxaliplatino , Inmunidad Celular , Antígenos de Histocompatibilidad Clase I , Interferón gamma/metabolismo
2.
J Med Chem ; 65(24): 16695-16715, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36507890

RESUMEN

Iterative medicinal chemistry optimization of an ester-containing astemizole (AST) analogue 1 with an associated metabolic instability liability led to the identification of a highly potent 3-trifluoromethyl-1,2,4-oxadiazole analogue 23 (PfNF54 IC50 = 0.012 µM; PfK1 IC50 = 0.040 µM) displaying high microsomal metabolic stability (HLM CLint < 11.6 µL·min-1·mg-1) and > 1000-fold higher selectivity over hERG compared to AST. In addition to asexual blood stage activity, the compound also shows activity against liver and gametocyte life cycle stages and demonstrates in vivo efficacy in Plasmodium berghei-infected mice at 4 × 50 mg·kg-1 oral dose. Preliminary interrogation of the mode of action using live-cell microscopy and cellular heme speciation revealed that 23 could be affecting multiple processes in the parasitic digestive vacuole, with the possibility of a novel target at play in the organelles associated with it.


Asunto(s)
Antimaláricos , Malaria , Ratones , Animales , Plasmodium berghei , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Astemizol/farmacología , Astemizol/uso terapéutico , Plasmodium falciparum/metabolismo , Malaria/tratamiento farmacológico , Malaria/parasitología , Modelos Animales de Enfermedad
3.
Int J Mol Sci ; 23(18)2022 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-36142445

RESUMEN

The hEag1 (Kv10.1) K+ channel is normally found in the brain, but it is ectopically expressed in tumor cells, including osteosarcoma. Based on the pivotal role of ion channels in osteogenesis, we tested whether pharmacological modulation of hEag1 may affect osteogenic differentiation of osteosarcoma cell lines. Using molecular biology (RT-PCR), electrophysiology (patch-clamp) and pharmacology (astemizole sensitivity, IC50 = 0.135 µM) we demonstrated that SaOS-2 osteosarcoma cells also express hEag1 channels. SaOS-2 cells also express to KCa1.1 K+ channels as shown by mRNA expression and paxilline sensitivity of the current. The inhibition of hEag1 (2 µM astemizole) or KCa1.1 (1 mM TEA) alone did not induce Ca2+ deposition in SaOS-2 cultures, however, these inhibitors, at identical concentrations, increased Ca2+ deposition evoked by the classical or pathological (inorganic phosphate, Pi) induction pathway without causing cytotoxicity, as reported by three completer assays (LDH release, MTT assay and SRB protein assay). We observed a similar effect of astemizole on Ca2+ deposition in MG-63 osteosarcoma cultures as well. We propose that the increase in the osteogenic stimuli-induced mineral matrix formation of osteosarcoma cell lines by inhibiting hEag1 may be a useful tool to drive terminal differentiation of osteosarcoma.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Astemizol/farmacología , Línea Celular Tumoral , Canales de Potasio Éter-A-Go-Go , Humanos , Osteogénesis , Osteosarcoma/tratamiento farmacológico , Fosfatos/metabolismo , ARN Mensajero/genética
4.
Eur J Pharmacol ; 928: 175086, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35714693

RESUMEN

The transient receptor potential (TRP) channel TRPV2 is widely expressed in a variety of different cell types and tissues. However, elucidating the exact biological functions of TRPV2 is significantly hampered by the lack of selective pharmacological tools to modulate channel activity in vitro and in vivo. This study aimed to identify new compounds that modify TRPV2 activity via the use of a plate-based calcium imaging approach to screen a drug repurposing library. Three antihistaminic drugs, loratadine, astemizole and clemizole were identified to reduce calcium-influx evoked by the TRPV2 agonist tetrahydrocannabivarin in HEK293 cells expressing murine TRPV2. Using single-cell calcium-microfluorimetry and whole-cell patch clamp recordings, we further confirmed that all three compounds induced a concentration-dependent block of TRPV2-mediated Ca2+ influx and whole-cell currents, with loratadine being the most potent antagonist of TRPV2. Moreover, this study demonstrated that loratadine was able to block both the human and mouse TRPV2 orthologs, without inhibiting the activity of other closely related members of the TRPV superfamily. Finally, loratadine inhibited TRPV2-dependent responses in a primary culture of mouse endometrial stromal cells and attenuated cell proliferation and migration in in vitro cell proliferation and wound healing assays. Taken together, our study revealed that the antihistaminic drugs loratadine, astemizole and clemizole target TRPV2 in a concentration-dependent manner. The identification of these antihistaminic drugs as blockers of TRPV2 may form a new starting point for the synthesis of more potent and selective TRPV2 antagonists, which could further lead to the unravelling of the physiological role of the channel.


Asunto(s)
Bloqueadores de los Canales de Calcio , Canales Catiónicos TRPV , Canales de Potencial de Receptor Transitorio , Animales , Astemizol/farmacología , Bencimidazoles/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio , Proliferación Celular , Células HEK293 , Antagonistas de los Receptores Histamínicos , Humanos , Loratadina/farmacología , Ratones , Células del Estroma , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores
5.
J Med Chem ; 64(12): 8194-8207, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34077206

RESUMEN

Disruption of EZH2-embryonic ectoderm development (EED) protein-protein interaction (PPI) is a new promising cancer therapeutic strategy. We have previously reported the discovery of astemizole, a small-molecule inhibitor targeting the EZH2-EED PPI. Herein, we report the cocrystal structure of EED in complex with astemizole at 2.15 Å. The structure elucidates the detailed binding mode of astemizole to EED and provides a structure-guided design for the discovery of a novel EZH2-EED interaction inhibitor, DC-PRC2in-01, with an affinity Kd of 4.56 µM. DC-PRC2in-01 destabilizes the PRC2 complex, thereby leading to the degradation of PRC2 core proteins and the decrease of global H3K27me3 levels in cancer cells. The proliferation of PRC2-driven lymphomas cells is effectively inhibited, and the cell cycle is arrested in the G0/G1 phase. Together, these data demonstrate that DC-PRC2in-01 could be an effective chemical probe for investigating the PRC2-related physiology and pathology and providing a promising chemical scaffold for further development.


Asunto(s)
Astemizol/análogos & derivados , Astemizol/farmacología , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Unión Proteica/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Reposicionamiento de Medicamentos , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Inhibidores Enzimáticos/síntesis química , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Complejo Represivo Polycomb 2/metabolismo , Relación Estructura-Actividad
6.
PLoS Negl Trop Dis ; 15(5): e0009432, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34033658

RESUMEN

BACKGROUND: Anthelminthic treatment options against schistosomiasis are limited. The current treatment relies almost exclusively on a single drug, praziquantel (PZQ). As a consequence, the development of resistance to PZQ and limited activity of PZQ against earlier development stages are respectively a risk and a limitation to achieving the goals of the new WHO roadmap towards elimination. For the discovery of new chemical starting points, the in vitro drug screening on Schistosoma mansoni (S. mansoni) against newly transformed schistosomula (NTS) is still the most predominant approach. The use of only NTS in the initial screening limits sensitivity to potential new compounds which are predominantly active in later developmental stages. Using our recently described highly standardized, straightforward and reliable culture method that generates high rates of juvenile worms, we aimed to repurpose a subset of the National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection (340 compounds) to identify new hits with an in vitro worm culture assay. METHODOLOGY/PRINCIPAL FINDINGS: Cercariae were mechanically transformed into skin-stage (SkS) schistosomula and continuously cultured for 3-6 weeks to the liver stage (LiS). A commercial source of serum was identified, and decrease of NTS/well along with optimal drug testing conditions was established to test compounds on early and late LiS worms. The library was screened in 96-well format assays using praziquantel (PZQ) as a positive control. Primary screening allowed a 5.9% hit rate and generated two confirmed hits on adult worms; a prophylactic antianginal agent and an antihistaminic drug. CONCLUSION: With this standardized and reliable in vitro assay, important S. mansoni developmental stages up to LiS worms can be generated and cultured over an extended period. When exposed to a subset of the NCATS Pharmaceutical Collection, 3 compounds yielded a defined anti-schistosomal phenotype on juvenile worms. Translation of activity on perfused adult S. mansoni worms was achieved only for perhexiline (a prophylactic antianginal agent) and astemizole (an antihistaminic drug).


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Schistosoma mansoni/efectos de los fármacos , Esquistosomicidas/farmacología , Animales , Astemizol/farmacología , Técnicas In Vitro , Perhexilina/farmacología , Schistosoma mansoni/crecimiento & desarrollo , Esquistosomiasis mansoni/tratamiento farmacológico
7.
Microb Pathog ; 156: 104929, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33932547

RESUMEN

Since the beginning of December 2019, a novel Coronavirus severe respiratory disease, caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) which also been termed 2019-new CoV (2019-nCoV), has continued to spread worldwide. As of August 27, 2020, a total of 24,232,429 people have been infected and 826,518 people have died. In our study, we found that astemizole can antagonize ACE2 and inhibit the entry of SARS-COV-2 spike pseudovirus into ACE2-expressed HEK293T cells (ACE2hi cells). We analysied the binding character of astemizole to ACE2 by molecular docking and surface plasmon resonance (SPR) assays and molecule docking, SARS-COV-2 spike pseudotype virus was also taken to investigate the suppression viropexis effect of astemizole. The results showed that astemizole can bind to the ACE2 receptor and inhibit the invasion of SARS-COV-2 Spike pseudoviruses. Thus astemizole represent potential drug candidates that can be re-used in anti-coronavirus therapies.


Asunto(s)
COVID-19 , Preparaciones Farmacéuticas , Astemizol/farmacología , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Internalización del Virus
8.
Structure ; 29(3): 203-212.e4, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33450182

RESUMEN

The hERG channel is a voltage-gated potassium channel involved in cardiac repolarization. Off-target hERG inhibition by drugs has become a critical issue in the pharmaceutical industry. The three-dimensional structure of the hERG channel was recently reported at 3.8-Å resolution using cryogenic electron microscopy (cryo-EM). However, the drug inhibition mechanism remains unclear because of the scarce structural information regarding the drug- and potassium-bound hERG channels. In this study, we obtained the cryo-EM density map of potassium-bound hERG channel complexed with astemizole, a well-known hERG inhibitor that increases risk of potentially fatal arrhythmia, at 3.5-Å resolution. The structure suggested that astemizole inhibits potassium conduction by binding directly below the selectivity filter. Furthermore, we propose a possible binding model of astemizole to the hERG channel and provide insights into the unusual sensitivity of hERG to several drugs.


Asunto(s)
Astemizol/química , Canal de Potasio ERG1/química , Bloqueadores de los Canales de Potasio/química , Astemizol/farmacología , Sitios de Unión , Microscopía por Crioelectrón , Canal de Potasio ERG1/antagonistas & inhibidores , Canal de Potasio ERG1/metabolismo , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Bloqueadores de los Canales de Potasio/farmacología , Unión Proteica
9.
Sci Rep ; 10(1): 22267, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33335233

RESUMEN

Cytochrome P450 2J2 (CYP2J2) is responsible for the epoxidation of endogenous arachidonic acid, and is involved in the metabolism of exogenous drugs. To date, no crystal structure of CYP2J2 is available, and the proposed structural basis for the substrate recognition and specificity in CYP2J2 varies with the structural models developed using different computational protocols. In this study, we developed a new structural model of CYP2J2, and explored its sensitivity to substrate binding by molecular dynamics simulations of the interactions with chemically similar fluorescent probes. Our results showed that the induced-fit binding of these probes led to the preferred active poses ready for the catalysis by CYP2J2. Divergent conformational dynamics of CYP2J2 due to the binding of each probe were observed. However, a stable hydrophobic clamp composed of residues I127, F310, A311, V380, and I487 was identified to restrict any substrate access to the active site of CYP2J2. Molecular docking of a series of compounds including amiodarone, astemizole, danazol, ebastine, ketoconazole, terfenadine, terfenadone, and arachidonic acid to CYP2J2 confirmed the role of those residues in determining substrate binding and specificity of CYP2J2. In addition to the flexibility of CYP2J2, the present work also identified other factors such as electrostatic potential in the vicinity of the active site, and substrate strain energy and property that have implications for the interpretation of CYP2J2 metabolism.


Asunto(s)
Astemizol/química , Butirofenonas/química , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/genética , Piperidinas/química , Ácido Araquidónico/genética , Ácido Araquidónico/metabolismo , Astemizol/farmacología , Butirofenonas/farmacología , Dominio Catalítico/efectos de los fármacos , Citocromo P-450 CYP2J2 , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Simulación del Acoplamiento Molecular , Oxidación-Reducción/efectos de los fármacos , Piperidinas/farmacología , Unión Proteica/efectos de los fármacos , Especificidad por Sustrato
10.
J Pharmacol Toxicol Methods ; 105: 106884, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32565325

RESUMEN

INTRODUCTION: Screening compounds for activity on the hERG channel using patch clamp is a crucial part of safety testing. Automated patch clamp (APC) is becoming widely accepted as an alternative to manual patch clamp in order to increase throughput whilst maintaining data quality. In order to standardize APC experiments, we have investigated the effects on IC50 values under different conditions using several devices across multiple sites. METHODS: APC instruments SyncroPatch 384i, SyncroPatch 384PE and Patchliner, were used to record hERG expressed in HEK or CHO cells. Up to 27 CiPA compounds were used to investigate effects of voltage protocol, incubation time, labware and time between compound preparation and experiment on IC50 values. RESULTS: All IC50 values of 21 compounds recorded on the SyncroPatch 384PE correlated well with IC50 values from the literature (Kramer et al., 2013) regardless of voltage protocol or labware, when compounds were used immediately after preparation, but potency of astemizole decreased if prepared in Teflon or polypropylene (PP) compound plates 2-3 h prior to experiments. Slow acting compounds such as dofetilide, astemizole, and terfenadine required extended incubation times of at least 6 min to reach steady state and therefore, stable IC50 values. DISCUSSION: Assessing the influence of different experimental conditions on hERG assay reliability, we conclude that either the step-ramp protocol recommended by CiPA or a standard 2-s step-pulse protocol can be used to record hERG; a minimum incubation time of 5 min should be used and although glass, Teflon, PP or polystyrene (PS) compound plates can be used for experiments, caution should be taken if using Teflon, PS or PP vessels as some adsorption can occur if experiments are not performed immediately after preparation. Our recommendations are not limited to the APC devices described in this report, but could also be extended to other APC devices.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Benchmarking/métodos , Fármacos Cardiovasculares/farmacología , Descubrimiento de Drogas/métodos , Corazón/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Animales , Arritmias Cardíacas/metabolismo , Astemizol/farmacología , Células CHO , Calibración , Fármacos Cardiovasculares/química , Línea Celular , Cricetulus , Evaluación Preclínica de Medicamentos/métodos , Canal de Potasio ERG1/metabolismo , Células HEK293 , Humanos , Fenetilaminas/farmacología , Polipropilenos/química , Politetrafluoroetileno/química , Estándares de Referencia , Reproducibilidad de los Resultados , Sulfonamidas/farmacología , Terfenadina/farmacología
11.
Biofabrication ; 12(2): 025017, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32101533

RESUMEN

Current practices in drug development have led to therapeutic compounds being approved for widespread use in humans, only to be later withdrawn due to unanticipated toxicity. These occurrences are largely the result of erroneous data generated by in vivo and in vitro preclinical models that do not accurately recapitulate human physiology. Herein, a human primary cell- and stem cell-derived 3D organoid technology is employed to screen a panel of drugs that were recalled from market by the FDA. The platform is comprised of multiple tissue organoid types that remain viable for at least 28 days, in vitro. For many of these compounds, the 3D organoid system was able to demonstrate toxicity. Furthermore, organoids exposed to non-toxic compounds remained viable at clinically relevant doses. Additional experiments were performed on integrated multi-organoid systems containing liver, cardiac, lung, vascular, testis, colon, and brain. These integrated systems proved to maintain viability and expressed functional biomarkers, long-term. Examples are provided that demonstrate how multi-organoid 'body-on-a-chip' systems may be used to model the interdependent metabolism and downstream effects of drugs across multiple tissues in a single platform. Such 3D in vitro systems represent a more physiologically relevant model for drug screening and will likely reduce the cost and failure rate associated with the approval of new drugs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Organoides/fisiología , Preparaciones Farmacéuticas/metabolismo , Astemizol/farmacología , Capecitabina/farmacología , Técnicas de Cultivo de Célula/instrumentación , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Dispositivos Laboratorio en un Chip , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Miocardio/citología , Miocardio/metabolismo , Organoides/citología , Organoides/efectos de los fármacos , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
12.
Genes (Basel) ; 11(2)2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31973216

RESUMEN

Retinoblastoma is the most common pediatric intraocular malignant tumor. Unfortunately, low cure rates and low life expectancy are observed in low-income countries. Thus, alternative therapies are needed for patients who do not respond to current treatments or those with advanced cases of the disease. Ether à-go-go-1 (Eag1) is a voltage-gated potassium channel involved in cancer. Eag1 expression is upregulated by the human papilloma virus (HPV) oncogene E7, suggesting that retinoblastoma protein (pRb) may regulate Eag1. Astemizole is an antihistamine that is suggested to be repurposed for cancer treatment; it targets proteins implicated in cancer, including histamine receptors, ATP binding cassette transporters, and Eag channels. Here, we investigated Eag1 regulation using pRb and Eag1 expression in human retinoblastoma. The effect of astemizole on the cell proliferation of primary human retinoblastoma cultures was also studied. HeLa cervical cancer cells (HPV-positive and expressing Eag1) were transfected with RB1. Eag1 mRNA expression was studied using qPCR, and protein expression was assessed using western blotting and immunochemistry. Cell proliferation was evaluated with an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. RB1 transfection down-regulated Eag1 mRNA and protein expression. The human retinoblastoma samples displayed heterogeneous Eag1 mRNA and protein expression. Astemizole decreased cell proliferation in primary retinoblastoma cultures. Our results suggest that Eag1 mRNA and protein expression was regulated by pRb in vitro, and that human retinoblastoma tissues had heterogeneous Eag1 mRNA and protein expression. Furthermore, our results propose that the multitarget drug astemizole may have clinical relevance in patients with retinoblastoma, for instance, in those who do not respond to current treatments.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Proteína de Retinoblastoma/metabolismo , Retinoblastoma/genética , Astemizol/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Preescolar , Canales de Potasio Éter-A-Go-Go/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Lactante , Masculino , Oncogenes , ARN Mensajero , Neoplasias de la Retina/genética , Retinoblastoma/metabolismo , Proteína de Retinoblastoma/genética , Transfección
13.
ACS Sens ; 4(10): 2623-2630, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31535848

RESUMEN

Detection of adverse effects of cardiac toxicity at an early stage by in vitro methods is crucial for the preclinical drug screening. Over the years, several kinds of biosensing platforms have been proposed by the scientific society for the detection of cardiac toxicity. However, the proposed tissue platforms have been optimized to measure either mechanophysiology or electrophysiology of the cardiomyocytes but not both. Herein, we demonstrate in detail our successful attempt toward developing a novel "multifunctional microphysiological system" also known as "organs-on-chips" to measure simultaneously the mechanical and electrical characteristics of cardiomyocytes in vitro. The proposed device can rapidly recognize drug-induced cardiovascular toxicity in real time, which is one of the most significant factors for drug discovery and postmarketing surveillance. We confirm that the proposed sensor delivers the direct relationship between the contraction force and cell impedance of cardiomyocytes under the influence of different cardiovascular drugs such as verapamil, astemizole, and lidocaine. The obtained assay results provide a great potential for a deep understanding of the drug effects on the cardiomyocytes in vitro.


Asunto(s)
Técnicas Biosensibles , Cardiotoxinas/farmacología , Evaluación Preclínica de Medicamentos/métodos , Miocitos Cardíacos/efectos de los fármacos , Animales , Astemizol/farmacología , Cardiotoxicidad , Células Cultivadas , Impedancia Eléctrica , Fenómenos Electrofisiológicos , Lidocaína/farmacología , Microelectrodos , Miocitos Cardíacos/fisiología , Ratas , Verapamilo/farmacología
14.
J Toxicol Sci ; 44(7): 441-457, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31270301

RESUMEN

The objective of this study is to assess the response of telemetered common marmosets to multiple cardiac ion channel inhibitors and to clarify the usefulness of this animal model in evaluating the effects of drug candidates on electrocardiogram (ECG). Six multiple cardiac ion channel inhibitors (sotalol, astemizole, flecainide, quinidine, verapamil and terfenadine) were orally administered to telemetered common marmosets and changes in QTc, PR interval and QRS duration were evaluated. Drugs plasma levels were determined to compare the sensitivity in common marmosets to that in humans. QTc prolongation was observed in the marmosets dosed with sotalol, astemizole, flecainide, quinidine, verapamil and terfenadine. PR prolongation was noted after flecainide and verapamil administration, and QRS widening occurred following treatment with flecainide and quinidine. Drugs plasma levels associated with ECG changes in marmosets were similar to those in humans, except for verapamil-induced QTc prolongation. Verapamil-induced change is suggested due to body temperature decrease. These results indicate that telemetered common marmoset is a useful animal for evaluation of the ECG effects of multiple cardiac ion channel inhibitors and the influence of body temperature change should be considered in the assessment.


Asunto(s)
Astemizol/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Callithrix , Electrocardiografía/efectos de los fármacos , Flecainida/farmacología , Modelos Animales , Quinidina/farmacología , Medición de Riesgo/métodos , Sotalol/farmacología , Telemetría , Terfenadina/farmacología , Verapamilo/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Animales , Astemizol/sangre , Temperatura Corporal/fisiología , Bloqueadores de los Canales de Calcio/sangre , Flecainida/sangre , Masculino , Quinidina/sangre , Sotalol/sangre , Terfenadina/sangre , Verapamilo/sangre , Bloqueadores del Canal de Sodio Activado por Voltaje/sangre
15.
Rev Invest Clin ; 71(3): 186-194, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31184333

RESUMEN

BACKGROUND: Expression and activity of the potassium channel ether-à-go-go-1 (EAG1) are strongly related to carcinogenesis and tumor progression, which can be exploited for therapeutic purposes. EAG1 activity may be reduced by preventing its phosphorylation with epidermal growth factor receptor (EGFR) kinase inhibitors and by astemizole, which blocks the channel pore and downregulates its gene expression. OBJECTIVE: We aimed to study the potential cooperative antiproliferative effect of the EGFR inhibitor gefitinib and the EAG1-blocker astemizole, in breast cancer cells. MATERIALS AND METHODS: The cells were characterized by immunocytochemistry. Inhibitory concentrations were determined by non-linear regression analysis using dose-response curves. The nature of the pharmacological effect was evaluated by the combination index equation while cell cycle analysis was studied by flow cy-tometry. RESULTS: Astemizole and gefitinib inhibited cell proliferation in a concentration-dependent manner, with inhibitory concentrations (IC 50) values of 1.72 µM and 0.51 µM, respectively. All combinations resulted in a synergistic antiproliferative effect. The combination of astemizole and gefitinib diminished the percentage of cells in G2/M and S phases, while increased accumulation in G0/G1 of the cell cycle. CONCLUSIONS: Astemizole and gefitinib synergistically inhibited proliferation in breast cancer cells expressing both EGFR and EAG1. Our results suggest that the combined treatment increased cell death by targeting the oncogenic activity of EAG1.


Asunto(s)
Antineoplásicos/farmacología , Astemizol/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Gefitinib/farmacología , Antineoplásicos/administración & dosificación , Astemizol/administración & dosificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Gefitinib/administración & dosificación , Regulación Neoplásica de la Expresión Génica , Humanos , Concentración 50 Inhibidora , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología
16.
Rev. invest. clín ; 71(3): 186-194, May.-Jun. 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1289686

RESUMEN

Abstract Background Expression and activity of the potassium channel ether-à-go-go-1 (EAG1) are strongly related to carcinogenesis and tumor progression, which can be exploited for therapeutic purposes. EAG1 activity may be reduced by preventing its phosphorylation with epidermal growth factor receptor (EGFR) kinase inhibitors and by astemizole, which blocks the channel pore and downregulates its gene expression. Objective We aimed to study the potential cooperative antiproliferative effect of the EGFR inhibitor gefitinib and the EAG1-blocker astemizole, in breast cancer cells. Materials and Methods The cells were characterized by immunocytochemistry. Inhibitory concentrations were determined by non-linear regression analysis using dose-response curves. The nature of the pharmacological effect was evaluated by the combination index equation while cell cycle analysis was studied by flow cytometry. Results Astemizole and gefitinib inhibited cell proliferation in a concentration-dependent manner, with inhibitory concentrations (IC 50) values of 1.72 µM and 0.51 µM, respectively. All combinations resulted in a synergistic antiproliferative effect. The combination of astemizole and gefitinib diminished the percentage of cells in G2/M and S phases, while increased accumulation in G0/G1 of the cell cycle. Conclusions Astemizole and gefitinib synergistically inhibited proliferation in breast cancer cells expressing both EGFR and EAG1. Our results suggest that the combined treatment increased cell death by targeting the oncogenic activity of EAG1.


Asunto(s)
Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Astemizol/farmacología , Gefitinib/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Astemizol/administración & dosificación , Concentración 50 Inhibidora , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Gefitinib/administración & dosificación , Antineoplásicos/administración & dosificación
17.
Int J Cancer ; 145(2): 415-426, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30628724

RESUMEN

Polycomb group proteins are important epigenetic regulators for cell proliferation and differentiation, organ development, as well as initiation and progression of lethal diseases, including cancer. Upregulated Polycomb group proteins, including Enhancer of zeste homolog 2 (EZH2), promote proliferation, migration, invasion and metastasis of cancer cells, as well as self-renewal of cancer stem cells. In our study, we report that EZH2 and embryonic ectoderm development (EED) indicate respective direct interaction with androgen receptor (AR). In the context of AR-positive prostate cancer, EZH2 and EED regulate AR expression levels and AR downstream targets. More importantly, we demonstrate that targeting EZH2 with the small-molecule inhibitor astemizole in cancer significantly represses the EZH2 and AR expression as well as the neoplastic capacities. These results collectively suggest that pharmacologically targeting EZH2 might be a promising strategy for advanced prostate cancer.


Asunto(s)
Astemizol/administración & dosificación , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Animales , Astemizol/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/genética , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int Immunopharmacol ; 65: 268-278, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30359933

RESUMEN

In this study, the immunomodulatory effects of astemizole (AST) against lipopolysaccharide (LPS) mediated T cell proliferation and induction of inflammation in RAW macrophages (in vitro), and zebrafish larvae (in vivo) were determined. AST significantly suppressed the phagocytic activity of macrophages (3.303 ±â€¯0.115) and inhibited lysosomal enzyme secretion (13.27 ±â€¯2.52) induced by LPS (100 ng/ml). Moreover, AST subdued the morphological deformities such as yolk sac edema (YSE) and spinal curvature curving (SC) by inhibiting ROS generation in zebrafish larvae 24 h after microinjection of LPS (0.5 mg/ml). AST was also shown to inhibit the production of the major cytokines TNF-α (150.8 ±â€¯0.6), IL-1ß (276.5 ±â€¯1.6), and PGE2 (194.6 ±â€¯0.6) pg/ml in RAW macrophages. It also subdued the ROS induced iNOS and COX-2 generated in response to LPS mediated immune dysfunctions in zebrafish larvae. These results suggested the immunosuppression effect of AST. Furthermore, induction of immune-suppression due to AST resulted in significant down-regulation of innate immunity directed by MAPK (p38, ERK and JNK), which was found to be associated with decreased production of acute inflammatory mediators both in vitro and in vivo. To confirm its activity, splenocytes were prepared using BALB/c mice and a mitogen activated splenocyte proliferation assay was also performed. Our findings suggest that AST has the ability to inhibit T cell proliferation and cytokine secretion both in vitro and in vivo by interfering with MAPK signaling pathway. Taken together, our results showed the potential of AST as a countermeasure to immune dysfunction and suggest its use as immunosuppressant compound in inflammatory disease.


Asunto(s)
Astemizol/farmacología , Inmunosupresores/farmacología , Macrófagos/inmunología , Bazo/inmunología , Linfocitos T/inmunología , Pez Cebra/inmunología , Animales , Proliferación Celular , Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Peces/metabolismo , Larva , Lipopolisacáridos/inmunología , Activación de Linfocitos , Ratones , Células RAW 264.7 , Transducción de Señal , Bazo/patología , Linfocitos T/efectos de los fármacos
19.
Biomed Pharmacother ; 107: 1682-1691, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30257386

RESUMEN

Hepatocellular carcinoma (HCC) accounts for the fifth most common cancer worldwide. Vitamin D and antihistamines have been shown to play an anti-tumor role in various tumors. In the present study, we ought to investigate the synergistic effect of astemizole and Vitamin D in HCC cells. We showed that astemizole enhanced the anti-tumor effect of Vitamin D in HCC both in vitro and in vivo. Astemizole enhanced Vitamin D-induced decrease of cell viability and proliferation, increase of apoptosis, decrease of cell migration and invasion in HCC cells in vitro and decrease of tumor number, mass and incidence in HCC in vivo. Astemizole increased VDR expression both in HCC cells in vitro and in tumor tissues in vivo. Downregulation of VDR significantly inhibited the synergistic effect of Vitamin D and astemizole on HCC cell viability, proliferation, apoptosis, migration and invasion. Bioinformatics analysis identified that miR-125a-5p had a putative binding site in the 3'-UTR of VDR. miR-125a-5p mimics inhibited astemizole-induced increase of VDR and enhancement of the anti-tumor effect of Vitamin D in HCC. Reporter gene assay has confirmed that VDR was regulated by miR-125a-5p. miR-125a-5p inhibitors increased VDR expression and decreased cell viability and proliferation in HCC cells. Moreover, VDR and miR-125a-5p expression in tumor tissues in HCC patients were negatively correlated. We identified that inhibition of miR-125a-5p and subsequent upregulation of VDR was involved in astemizole-induced enhancement of the anti-tumor effect of Vitamin D in HCC. These results highlight the importance of combined treatment of astemizole and Vitamin D and provide novel insights into the role of miR-125a-5p-VDR signaling in HCC.


Asunto(s)
Astemizol/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , MicroARNs/genética , Vitamina D/farmacología , Regiones no Traducidas 3'/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Astemizol/administración & dosificación , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Receptores de Calcitriol/genética , Regulación hacia Arriba/efectos de los fármacos , Vitamina D/administración & dosificación
20.
Sci Rep ; 8(1): 1153, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29348631

RESUMEN

Voltage-gated ion channels (VGCs) are prime targets for the pharmaceutical industry, but drug profiling on VGCs is challenging, since drug interactions are confined to specific conformational channel states mediated by changes in transmembrane potential. Here we combined various optogenetic tools to develop dynamic, high-throughput drug profiling assays with defined light-step protocols to interrogate VGC states on a millisecond timescale. We show that such light-induced electrophysiology (LiEp) yields high-quality pharmacological data with exceptional screening windows for drugs acting on the major cardiac VGCs, including hNav1.5, hKv1.5 and hERG. LiEp-based screening remained robust when using a variety of optogenetic actuators (ChR2, ChR2(H134R), CatCh, ChR2-EYFP-ßArchT) and different types of organic (RH421, Di-4-ANBDQPQ, BeRST1) or genetic voltage sensors (QuasAr1). The tractability of LiEp allows a versatile and precise alternative to state-of-the-art VGC drug screening platforms such as automated electrophysiology or FLIPR readers.


Asunto(s)
Canal de Potasio ERG1/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Optogenética/métodos , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Astemizol/farmacología , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Flecainida/farmacología , Expresión Génica , Células HEK293 , Humanos , Lidocaína/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Optogenética/instrumentación , Técnicas de Placa-Clamp , Fosfinas/farmacología , Plásmidos/química , Plásmidos/metabolismo , Quinidina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA