Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
iScience ; 24(4): 102270, 2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33817575

RESUMEN

Voltage-gated sodium (NaV) channels, initially characterized in excitable cells, have been shown to be aberrantly expressed in non-excitable cancer tissues and cells from epithelial origins such as in breast, lung, prostate, colon, and cervix, whereas they are not expressed in cognate non-cancer tissues. Their activity was demonstrated to promote aggressive and invasive potencies of cancer cells, both in vitro and in vivo, whereas their deregulated expression in cancer tissues has been associated with metastatic progression and cancer-related death. This review proposes NaV channels as pharmacological targets for anticancer treatments providing opportunities for repurposing existing NaV-inhibitors or developing new pharmacological and nutritional interventions.

2.
ACS Appl Bio Mater ; 4(8): 6244-6255, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35006910

RESUMEN

Brain-derived neurotrophic factor (BDNF) and its receptor tyrosine receptor kinase B (TrkB) have been shown to play an important role in numerous neurological disorders, such as Alzheimer's disease. The identification of biologically active compounds interacting with TrkB serves as a drug discovery strategy to identify drug leads for neurological disorders. Here, we report effective immobilization of functional TrkB on magnetic iron oxide nanoclusters, where TrkB receptors behave as "smart baits" to bind compounds from mixtures and magnetic nanoclusters enable rapid isolation through magnetic separation. The presence of the immobilized TrkB was confirmed by specific antibody labeling. Subsequently, the activity of the TrkB on iron oxide nanoclusters was evaluated with ATP/ADP conversion experiments using a known TrkB agonist. The immobilized TrkB receptors can effectively identify binders from mixtures containing known binders, synthetic small molecule mixtures, and Gotu Kola (Centella asiatica) plant extracts. The identified compounds were analyzed by an ultrahigh-performance liquid chromatography system coupled with a quadrupole time-of-flight mass spectrometer. Importantly, some of the identified TrkB binders from Gotu Kola plant extracts matched with compounds previously linked to neuroprotective effects observed for a Gotu Kola extract approved for use in a clinical trial. Our studies suggest that the possible therapeutic effects of the Gotu Kola plant extract in dementia treatment, at least partially, might be associated with compounds interacting with TrkB. The unique feature of this approach is its ability to fast screen potential drug leads using less explored transmembrane targets. This platform works as a drug-screening funnel at early stages of the drug discovery pipeline. Therefore, our approach will not only greatly benefit drug discovery processes using transmembrane proteins as targets but also allow for evaluation and validation of cellular pathways targeted by drug leads.


Asunto(s)
Centella , Evaluación Preclínica de Medicamentos , Fenómenos Magnéticos , Extractos Vegetales , Proteínas Tirosina Quinasas Receptoras
3.
Sci Rep ; 7(1): 5974, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28729722

RESUMEN

Streptococcus mutans employs a key virulence factor, three glucosyltransferase (GtfBCD) enzymes to establish cariogenic biofilms. Therefore, the inhibition of GtfBCD would provide anti-virulence therapeutics. Here a small molecule library of 500,000 small molecule compounds was screened in silico against the available crystal structure of the GtfC catalytic domain. Based on the predicted binding affinities and drug-like properties, small molecules were selected and evaluated for their ability to reduce S. mutans biofilms, as well as inhibit the activity of Gtfs. The most potent inhibitor was further characterized for Gtf binding using OctetRed instrument, which yielded low micromolar KD against GtfB and nanomolar KD against GtfC, demonstrating selectivity towards GtfC. Additionally, the lead compound did not affect the overall growth of S. mutans and commensal oral bacteria, and selectively inhibit the biofilm formation by S. mutans, indicative of its selectivity and non-bactericidal nature. The lead compound also effectively reduced cariogenicity in vivo in a rat model of dental caries. An analog that docked poorly in the GtfC catalytic domain failed to inhibit the activity of Gtfs and S. mutans biofilms, signifying the specificity of the lead compound. This report illustrates the validity and potential of structure-based design of anti-S. mutans virulence inhibitors.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Caries Dental/microbiología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Benzamidas/farmacología , Biopelículas/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/metabolismo , Cinética , Viabilidad Microbiana/efectos de los fármacos , Boca/microbiología , Mutación/genética , Streptococcus mutans/efectos de los fármacos , Streptococcus mutans/crecimiento & desarrollo , Streptococcus mutans/patogenicidad , Streptococcus mutans/fisiología , Relación Estructura-Actividad , Virulencia/efectos de los fármacos
4.
Chin J Nat Med ; 13(7): 554-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26233847

RESUMEN

We have recently designed and synthesized several novel iminoquinone anticancer agents that have entered preclinical development for the treatment of human cancers. Herein we developed and validated a quantitative HPLC-MS/MS analytical method for one of the lead novel anticancer makaluvamine analog, TCBA-TPQ, and conducted a pharmacokinetic study in laboratory rats. Our results indicated that the HPLC-MS/MS method was precise, accurate, and specific. Using this method, we carried out in vitro and in vivo evaluations of the pharmacological properties of TCBA-TPQ and plasma pharmacokinetics in rats. Our results provide a basis for future preclinical and clinical development of this promising anticancer marine analog.


Asunto(s)
Antineoplásicos/farmacocinética , Cromatografía Líquida de Alta Presión/métodos , Pirroles/farmacocinética , Quinolonas/farmacocinética , Espectrometría de Masas en Tándem/métodos , Animales , Antineoplásicos/sangre , Pirroles/sangre , Quinolonas/sangre , Ratas Sprague-Dawley
5.
Chin J Nat Med ; 13(8): 561-77, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26253489

RESUMEN

The ocean continues to provide a plethora of unique scaffolds capable of remarkable biological applications. A large number of pyrroloiminoquinone alkaloids, including discorhabdins, epinardins, batzellines, makaluvamines, and veiutamine, have been isolated from various marine organisms. A class of pyrroloiminoquinone-related alkaloids, known as bispyrroloquinones, is the focus of this review article. This family of marine alkaloids, which contain an aryl substituted bispyrroloquinone ring system, includes three subclasses of alkaloids namely, wakayin, tsitsikammamines A-B, and zyzzyanones A-D. Both wakayin and the tsitsikammamines contain a tetracyclic fused bispyrroloiminoquinone ring system, while zyzzyanones contain a fused tricyclic bispyrroloquinone ring system. The unique chemical structures of these marine natural products and their diverse biological properties, including antifungal and antimicrobial activity, as well as the potent, albeit generally nonspecific and universal cytotoxicities, have attracted great interest of synthetic chemists over the past three decades. Tsitsikammamines, wakayin, and several of their analogs show inhibition of topoisomerases. One additional possible mechanism of anticancer activity of tsitsikammamines analogs that has been discovered recently is through the inhibition of indoleamine 2, 3-dioxygenase, an enzyme involved in tumoral immune resistance. This review discusses the isolation, synthesis, and evaluation of bioactivities of bispyrroloquinone alkaloids and their analogs.


Asunto(s)
Alcaloides/farmacología , Antiinfecciosos/farmacología , Antineoplásicos/farmacología , Productos Biológicos/farmacología , Pirroles/farmacología , Quinolinas/farmacología , Quinonas/farmacología , Alcaloides/química , Animales , Antiinfecciosos/química , Antineoplásicos/química , Productos Biológicos/química , Humanos , Alcaloides Indólicos/química , Alcaloides Indólicos/farmacología , Indoles/química , Indoles/farmacología , Pirroles/química , Quinolinas/química , Quinonas/química
6.
Int J Antimicrob Agents ; 46(2): 174-82, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26022931

RESUMEN

Streptococcus mutans is a major aetiological agent of dental caries. Formation of biofilms is a key virulence factor of S. mutans. Drugs that inhibit S. mutans biofilms may have therapeutic potential. Dihydrofolate reductase (DHFR) plays a critical role in regulating the metabolism of folate. DHFR inhibitors are thus potent drugs and have been explored as anticancer and antimicrobial agents. In this study, a library of analogues based on a DHFR inhibitor, trimetrexate (TMQ), an FDA-approved drug, was screened and three new analogues that selectively inhibited S. mutans were identified. The most potent inhibitor had a 50% inhibitory concentration (IC50) of 454.0±10.2nM for the biofilm and 8.7±1.9nM for DHFR of S. mutans. In contrast, the IC50 of this compound for human DHFR was ca. 1000nM, a >100-fold decrease in its potency, demonstrating the high selectivity of the analogue. An analogue that exhibited the least potency for the S. mutans biofilm also had the lowest activity towards inhibiting S. mutans DHFR, further indicating that inhibition of biofilms is related to reduced DHFR activity. These data, along with docking of the most potent analogue to the modelled DHFR structure, suggested that the TMQ analogues indeed selectively inhibited S. mutans through targeting DHFR. These potent and selective small molecules are thus promising lead compounds to develop new effective therapeutics to prevent and treat dental caries.


Asunto(s)
Antibacterianos/farmacología , Antagonistas del Ácido Fólico/farmacología , Streptococcus mutans/efectos de los fármacos , Trimetrexato/farmacología , Biopelículas/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Antagonistas del Ácido Fólico/química , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Simulación de Dinámica Molecular , Streptococcus mutans/fisiología , Tetrahidrofolato Deshidrogenasa/química , Trimetrexato/análogos & derivados
7.
Oncotarget ; 6(25): 21268-82, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26020804

RESUMEN

Here, we report the chemotherapeutic effect of honokiol, a phytochemical from Magnolia plant, on human head and neck squamous cell carcinoma (HNSCC). Treatment of HNSCC cell lines from different sub-sites, SCC-1 (oral cavity), SCC-5 (larynx), OSC-19 (tongue) and FaDu (pharynx) with honokiol inhibited their cell viability, which was associated with the: (i) induction of apoptosis, (ii) correction of dysregulatory cell cycle proteins of G0/G1 phase. Honokiol decreased the expression levels of epidermal growth factor receptor (EGFR), mTOR and their downstream signaling molecules. Treatment of FaDu and SCC-1 cell lines with rapamycin, an inhibitor of mTOR pathway, also reduced cell viability of HNSCC cells. Administration of honokiol by oral gavage (100 mg/kg body weight) significantly (P < 0.01-0.001) inhibited the growth of SCC-1 and FaDu xenografts in athymic nude mice, which was associated with: (i) inhibition of tumor cell proliferation, (ii) induction of apoptosis, (iii) reduced expressions of cyclins and Cdks, and (iv) inhibition of EGFR signaling pathway. Molecular docking analysis of honokiol in EGFR binding site indicated that the chemotherapeutic effect of honokiol against HNSCC is mediated through its firm binding with EGFR, which is better than that of gefitinib, a commonly used drug for HNSCC treatment.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Compuestos de Bifenilo/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Lignanos/farmacología , Animales , Apoptosis , Sitios de Unión , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Separación Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Receptores ErbB/química , Femenino , Citometría de Flujo , Humanos , Magnolia/química , Ratones , Ratones Desnudos , Simulación del Acoplamiento Molecular , Fitoterapia/métodos , Extractos Vegetales/química , Transducción de Señal , Sirolimus/química
8.
Mar Drugs ; 10(5): 1138-1155, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22822362

RESUMEN

We have recently designed and synthesized a novel iminoquinone anticancer agent, 7-(4-fluorobenzylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one (FBA-TPQ) and initiated its preclinical development. Herein we investigated its efficacy, safety, and pharmacokinetics in in vitro and in vivo models of human pancreatic cancer. Our results demonstrated that FBA-TPQ inhibited pancreatic cancer cell growth, induced apoptosis, and caused cell cycle arrest in vitro. It inhibited the growth of xenograft tumors with minimal host toxicity. To facilitate future preclinical and clinical development of the agent, we also developed and validated a Rapid Resolution Liquid Chromatography (RRLC) method for quantitative analysis of FBA-TPQ in plasma and tissue samples. The method was found to be precise, accurate, and specific. Using this method, we carried out in vitro and in vivo evaluations of the pharmacological properties of FBA-TPQ, including stability in plasma, plasma protein binding, metabolism by S9 enzymes, plasma pharmacokinetics, and tissue distribution. Our results indicate that FBA-TPQ is a potential therapeutic agent for pancreatic cancer, providing a basis for future preclinical and clinical development.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Pirroles/farmacología , Pirroles/farmacocinética , Quinolonas/farmacología , Quinolonas/farmacocinética , Animales , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Unión Proteica/efectos de los fármacos , Pirroles/efectos adversos , Quinolonas/efectos adversos , Distribución Tisular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Mar Drugs ; 8(7): 2129-41, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20714427

RESUMEN

Marine natural products and their synthetic derivatives represent a major source of novel candidate anti-cancer compounds. We have recently tested the anti-cancer activity of more than forty novel compounds based on an iminoquinone makaluvamine scaffold, and have found that many of the compounds exert potent cytotoxic activity against human cancer cell lines. One of the most potent compounds, BA-TPQ [(11,12),7-(benzylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one], was active against a variety of human cancer cell lines, and inhibited the growth of breast and prostate xenograft tumors in mice. However, there was some toxicity noted in the mice following administration of the compound. In order to further the development of BA-TPQ, and in a search for potential sites of accumulation that might underlie the observed toxicity of the compound, we accomplished preclinical pharmacological studies of the compound. We herein report the in vitro and in vivo pharmacological properties of BA-TPQ, including its stability in plasma, plasma protein binding, metabolism by S9 enzymes, and plasma and tissue distribution. We believe these studies will be useful for further investigations, and may be useful for other investigators examining the use of similar compounds for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Pirroles/farmacología , Quinolonas/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Ratones , Neoplasias/patología , Unión Proteica , Pirroles/farmacocinética , Pirroles/toxicidad , Quinolonas/farmacocinética , Quinolonas/toxicidad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Med Chem ; 5(3): 227-36, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19442212

RESUMEN

The high mortality rate and lack of effective therapies make lung cancer an ideal target for novel therapeutic agents. The present study was designed to implement a novel chemical synthesis pathway and to determine the biological activities of synthetic makaluvamine analogs in human lung cancer. Seventeen compounds were synthesized and purified, and their chemical structures were elucidated on the basis of physicochemical constants and NMR spectra. Their in vitro activity was determined in human lung cancer cell lines. Based on initial screens, compound Ic was found to be the most potent, and was therefore used as a model for further studies in lung cancer cells. Ic induced both apoptosis and S-phase cell cycle arrest. Furthermore, it activated p53 and induced cleavage of PARP and caspases 8 and 9. Our preclinical data indicate that the makaluvamine analogs are potential therapeutic agents against lung cancer, providing a basis for further development of Ic (and perhaps other analogs) as a novel anti-cancer agent.


Asunto(s)
Alcaloides/síntesis química , Alcaloides/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Quinolonas/síntesis química , Quinolonas/farmacología , Alcaloides/química , Alcaloides/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Quinolonas/química , Quinolonas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA