Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int Immunopharmacol ; 114: 109487, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36493694

RESUMEN

Systemic lupus erythematosus (SLE) is a multisystemic, inflammatory autoimmune disease. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells participated in the pathogenesis of SLE. MDSCs has been considered a potential therapeutic target for lupus. As traditional Chinese medicine, Halofuginone (HF) has the extensive immunomodulatory effects on some autoimmune disorders. Our research was dedicated to discovering therapeutic efficacy of HF for lupus to explore novel mechanisms on MDSCs. We found that HF prominently alleviated the systemic symptoms especially nephritis in Imiquimod-induced lupus mice, and simultaneously repaired the immune system, reflected in the alteration of autoantibodies. HF diminished the quantity of MDSCs in lupus mice, and induced apoptosis of MDSCs. Through RNA sequencing performed on the sorted MDSC from lupus mice and HF-treated lupus mice, B lymphoid tyrosine kinase (Blk, a non-receptor cytoplasmic tyrosine kinase) was screened as the target molecule of HF. It's proven that HF had two independent effects on Blk. On the one hand, HF increased the mRNA expression of Blk in MDSCs by inhibiting the nuclear translocation of p65/p50 heterodimer. On the other hand, HF enhanced the kinase activity of Blk in MDSCs through direct molecular binding. We further investigated that Blk suppressed the phosphorylation of downstream ERK signaling pathway to increase the apoptosis of MDSCs. In conclusion, our study illustrated that HF alleviated the disease progression of lupus mice by targeting Blk to promote the apoptosis of MDSCs, which indicated the immunotherapeutic potential of HF to treat lupus.


Asunto(s)
Lupus Eritematoso Sistémico , Células Supresoras de Origen Mieloide , Ratones , Animales , Piperidinas/farmacología , Piperidinas/uso terapéutico , Quinazolinonas/farmacología , Quinazolinonas/uso terapéutico
2.
Sci Rep ; 12(1): 2003, 2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35132095

RESUMEN

The present article describes the design, synthesis, in vitro urease inhibition, and in silico molecular docking studies of a novel series of nitrothiazolacetamide conjugated to different thioquinazolinones. Fourteen nitrothiazolacetamide bearing thioquinazolinones derivatives (8a-n) were synthesized through the reaction of isatoic anhydride with different amine, followed by reaction with carbon disulfide and KOH in ethanol. The intermediates were then converted into final products by treating them with 2-chloro-N-(5-nitrothiazol-2-yl)acetamide in DMF. All derivatives were then characterized through different spectroscopic techniques (1H, 13C-NMR, MS, and FTIR). In vitro screening of these molecules against urease demonstrated the potent urease inhibitory potential of derivatives with IC50 values ranging between 2.22 ± 0.09 and 8.43 ± 0.61 µM when compared with the standard thiourea (IC50 = 22.50 ± 0.44 µM). Compound 8h as the most potent derivative exhibited an uncompetitive inhibition pattern against urease in the kinetic study. The high anti-ureolytic activity of 8h was confirmed against two urease-positive microorganisms. According to molecular docking study, 8h exhibited several hydrophobic interactions with Lys10, Leu11, Met44, Ala47, Ala85, Phe87, and Pro88 residues plus two hydrogen bound interactions with Thr86. According to the in silico assessment, the ADME-Toxicity and drug-likeness profile of synthesized compounds were in the acceptable range.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos , Quinazolinonas , Ureasa , Aminas/química , Disulfuro de Carbono/química , Simulación por Computador , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Etanol/química , Hidróxidos/química , Simulación del Acoplamiento Molecular , Oxazinas/química , Compuestos de Potasio/química , Ureasa/antagonistas & inhibidores , Quinazolinonas/síntesis química , Quinazolinonas/química , Quinazolinonas/farmacología
3.
Nat Commun ; 12(1): 3697, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140482

RESUMEN

Despite the clinical success of the third-generation EGFR inhibitor osimertinib as a first-line treatment of EGFR-mutant non-small cell lung cancer (NSCLC), resistance arises due to the acquisition of EGFR second-site mutations and other mechanisms, which necessitates alternative therapies. Dacomitinib, a pan-HER inhibitor, is approved for first-line treatment and results in different acquired EGFR mutations than osimertinib that mediate on-target resistance. A combination of osimertinib and dacomitinib could therefore induce more durable responses by preventing the emergence of resistance. Here we present an integrated computational modeling and experimental approach to identify an optimal dosing schedule for osimertinib and dacomitinib combination therapy. We developed a predictive model that encompasses tumor heterogeneity and inter-subject pharmacokinetic variability to predict tumor evolution under different dosing schedules, parameterized using in vitro dose-response data. This model was validated using cell line data and used to identify an optimal combination dosing schedule. Our schedule was subsequently confirmed tolerable in an ongoing dose-escalation phase I clinical trial (NCT03810807), with some dose modifications, demonstrating that our rational modeling approach can be used to identify appropriate dosing for combination therapy in the clinical setting.


Asunto(s)
Acrilamidas/administración & dosificación , Acrilamidas/farmacología , Compuestos de Anilina/administración & dosificación , Compuestos de Anilina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos , Neoplasias Pulmonares/dietoterapia , Quinazolinonas/administración & dosificación , Quinazolinonas/farmacología , Acrilamidas/farmacocinética , Acrilamidas/toxicidad , Compuestos de Anilina/farmacocinética , Compuestos de Anilina/toxicidad , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Estudios de Cohortes , Simulación por Computador , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Modelos Estadísticos , Modelos Teóricos , Mutación , Quinazolinonas/farmacocinética , Quinazolinonas/toxicidad
4.
Drug Res (Stuttg) ; 71(6): 335-340, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33535253

RESUMEN

Sleeping sickness, caused by trypanosomes, is a debilitating, neglected tropical disease wherein current treatments suffer from several drawbacks such as toxicity, low activity, and poor pharmacokinetic properties, and hence the need for alternative treatment is apparent. To this effect, we screened in vitro a library of 2-quinazolinone derivatives for antitrypanosomal activity against T.b. brucei and cytotoxicity against HeLa cells. Seven compounds having no overt cytotoxicity against HeLa cells exhibited antitrypanosomal activity in the range of 0.093-45 µM were identified. The activity data suggests that the antitrypanosomal activity of this compound class is amenable to substituents at N1 and C6 positions. Compound 14: having a molecular weight of 238Da, ClogP value of 1 and a total polar surface area of 49 was identified as the most active, exhibiting an IC50 value of 0.093 µM Graphical Abstract.


Asunto(s)
Quinazolinonas/farmacología , Tripanocidas/farmacología , Tripanosomiasis Africana/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Células HeLa , Humanos , Pruebas de Sensibilidad Parasitaria , Quinazolinonas/química , Quinazolinonas/uso terapéutico , Pruebas de Toxicidad Aguda , Tripanocidas/química , Tripanocidas/uso terapéutico , Trypanosoma brucei gambiense/efectos de los fármacos , Trypanosoma brucei rhodesiense/efectos de los fármacos , Tripanosomiasis Africana/parasitología
5.
Oxid Med Cell Longev ; 2020: 7151946, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32963699

RESUMEN

Vascular endothelial senescence induced by high glucose and palmitate (HG/PA) contributes to endothelial dysfunction, which leads to diabetic cardiovascular complications. Reduction of endothelial senescence may attenuate these pathogenic processes. This study is aimed at determining whether Ginseng-Sanqi-Chuanxiong (GSC) extracts, traditional Chinese medicine, can ameliorate human aortic endothelial cell (HAEC) senescence under HG/PA-stressed conditions and further explore the underlying mechanism. We found that GSC extracts significantly increased antisenescent activity by reducing the HG/PA-induced mitochondrial ROS (mtROS) levels in senescent HAECs. GSC extracts also induced cellular mitophagy formation, which mediated the effect of GSC extracts on mtROS reduction. Apart from this, the data showed that GSC extracts stimulated mitophagy via the AMPK pathway, and upon inhibition of AMPK by pharmacological and genetic inhibitors, GSC extract-mediated mitophagy was abolished which further led to reverse the antisenescence effect. Taken together, these data suggest that GSC extracts prevent HG/PA-induced endothelial senescence and mtROS production by mitophagy regulation via the AMPK pathway. Thus, the induction of mitophagy by GSC extracts may provide a novel therapeutic candidate for cardiovascular protection in metabolic syndrome.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Senescencia Celular/efectos de los fármacos , Diabetes Mellitus/patología , Medicamentos Herbarios Chinos/farmacología , Células Endoteliales/enzimología , Células Endoteliales/patología , Mitofagia/efectos de los fármacos , Extractos Vegetales/farmacología , Transducción de Señal , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/química , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glucosa/toxicidad , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Ácido Palmítico/toxicidad , Extractos Vegetales/química , Quinazolinonas/farmacología , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos
6.
J Biosci ; 452020.
Artículo en Inglés | MEDLINE | ID: mdl-32457283

RESUMEN

Halofuginone (HF) is an extract from the widely used traditional Chinese medicine (TCM) Dichroa febrifuga that facilitates the recovery of wounds and attenuates hepatic fibrosis. However, the role of HF in the epithelial-mesenchymal transition (EMT) of IPEC-J2 cells remains unclear. The current study explored the anti-EMT effect of HF in IPEC-J2 cells and illustrates its molecular mechanism. Transforming growth factor ß1 (TGF-ß1), as a recognized profibrogenic cytokine, decreased the level of the epithelial marker E-cadherin and increased the level of the mesenchymal markers, such as N-cadherin, fibronectin (FN), vimentin (Vim), and α-smooth muscle actin (α-SMA), in IPEC-J2 cells depending on the exposure time and dose. HF markedly prevented the EMT induced by TGF-ß1. Dissection of the mechanism revealed that HF inhibited IPEC-J2 cell EMT via modulating the phosphorylation of SMAD2/3 and the SMAD2/3-SMAD4 complex nuclear translocation. Furthermore, HF could promote the phosphorylation of eukaryotic translation initiation factor-2α (eIF2α), which modulates the SMAD signaling pathway. These results suggested that HF inhibits TGF-ß1-induced EMT in IPEC-J2 cells through the eIF2α/SMAD signaling pathway. Our findings suggest that HF can serve as a potential anti-EMT agent in intestinal fibrosis therapy.


Asunto(s)
Antineoplásicos/farmacología , Enterocitos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/genética , Piperidinas/farmacología , Quinazolinonas/farmacología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Actinas/genética , Actinas/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Enterocitos/citología , Enterocitos/metabolismo , Transición Epitelial-Mesenquimal/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Porcinos , Factor de Crecimiento Transformador beta1/farmacología , Vimentina/genética , Vimentina/metabolismo
7.
J Med Chem ; 63(8): 4256-4292, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32212730

RESUMEN

A series of quinazolin-4-one based hydroxamic acids was rationally designed and synthesized as novel dual PI3K/HDAC inhibitors by incorporating an HDAC pharmacophore into a PI3K inhibitor (Idelalisib) via an optimized linker. Several of these dual inhibitors were highly potent (IC50 < 10 nM) and selective against PI3Kγ, δ and HDAC6 enzymes and exhibited good antiproliferative activity against multiple cancer cell lines. The lead compound 48c, induced necrosis in several mutant and FLT3-resistant AML cell lines and primary blasts from AML patients, while showing no cytotoxicity against normal PBMCs, NIH3T3, and HEK293 cells. Target engagement of PI3Kδ and HDAC6 by 48c was demonstrated in MV411 cells using the cellular thermal shift assay (CETSA). Compound 48c showed good pharmacokinetics properties in mice via intraperitoneal (ip) administration and provides a means to examine the biological effects of inhibiting these two important enzymes with a single molecule, either in vitro or in vivo.


Asunto(s)
Diseño de Fármacos , Inhibidores de Histona Desacetilasas/síntesis química , Ácidos Hidroxámicos/síntesis química , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/síntesis química , Quinazolinonas/síntesis química , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Femenino , Células HEK293 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos BALB C , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Estructura Terciaria de Proteína , Quinazolinonas/farmacología , Ratas
8.
Neuromolecular Med ; 21(4): 484-492, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31152363

RESUMEN

Stroke significantly affects white matter in the brain by impairing axon function, which results in clinical deficits. Axonal mitochondria are highly dynamic and are transported via microtubules in the anterograde or retrograde direction, depending upon axonal energy demands. Recently, we reported that mitochondrial division inhibitor 1 (Mdivi-1) promotes axon function recovery by preventing mitochondrial fission only when applied during ischemia. Application of Mdivi-1 after injury failed to protect axon function. Interestingly, L-NIO, which is a NOS3 inhibitor, confers post-ischemic protection to axon function by attenuating mitochondrial fission and preserving mitochondrial motility via conserving levels of the microtubular adaptor protein Miro-2. We propose that preventing mitochondrial fission protects axon function during injury, but that restoration of mitochondrial motility is more important to promote axon function recovery after injury. Thus, Miro-2 may be a therapeutic molecular target for recovery following a stroke.


Asunto(s)
Transporte Axonal , Axones/patología , Accidente Cerebrovascular Isquémico/patología , Mitocondrias/ultraestructura , Dinámicas Mitocondriales , Quinazolinonas/uso terapéutico , Sustancia Blanca/patología , Adenosina Trifosfato/biosíntesis , Envejecimiento/patología , Animales , Transporte Axonal/efectos de los fármacos , Axones/efectos de los fármacos , Axones/ultraestructura , Calcio/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Hipoxia-Isquemia Encefálica/patología , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Dinámicas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/fisiología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ornitina/análogos & derivados , Ornitina/farmacología , Quinazolinonas/farmacología , Daño por Reperfusión/patología , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/ultraestructura , Proteínas de Unión al GTP rho/fisiología
9.
Cell Commun Signal ; 17(1): 39, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31046771

RESUMEN

BACKGROUND: In the event of amino acid starvation, the cell activates two main protective pathways: Amino Acid starvation Response (AAR), to inhibit global translation, and autophagy, to recover the essential substrates from degradation of redundant self-components. Whether and how AAR and autophagy (ATG) are cross-regulated and at which point the two regulatory pathways intersect remain unknown. Here, we provide experimental evidence that the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) specifically located at the lysosome level links the AAR with the autophagy pathway. METHODS: As an inducer of the AAR, we used halofuginone (HF), an alkaloid that binds to the prolyl-tRNA synthetase thus mimicking the unavailability of proline (PRO). Induction of AAR was determined assessing the phosphorylation of the eukaryotic translation initiation factor (eIF) 2α. Autophagy was monitored by assessing the processing and accumulation of microtubule-associated protein 1 light chain 3 isoform B (LC3B) and sequestosome-1 (p62/SQSTM1) levels. The activity of mTORC1 was monitored through assessment of the phosphorylation of mTOR, (rp)S6 and 4E-BP1. Global protein synthesis was determined by puromycin incorporation assay. mTORC1 presence on the membrane of the lysosomes was monitored by cell fractionation and mTOR expression was determined by immunoblotting. RESULTS: In three different types of human cancer cells (thyroid cancer WRO cells, ovarian cancer OAW-42 cells, and breast cancer MCF-7 cells), HF induced both the AAR and the autophagy pathways time-dependently. In WRO cells, which showed the strongest induction of autophagy and of AAR, global protein synthesis was little if any affected. Consistently, 4E-BP1 and (rp)S6 were phosphorylated. Concomitantly, mTOR expression and activation declined along with its detachment from the lysosomes and its degradation by the proteasome, and with the nuclear translocation of transcription factor EB (TFEB), a transcription factor of many ATG genes. The extra supplementation of proline rescued all these effects. CONCLUSIONS: We demonstrate that the AAR and autophagy are mechanistically linked at the level of mTORC1, and that the lysosome is the central hub of the cross-talk between these two metabolic stress responses.


Asunto(s)
Autofagia/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Piperidinas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Quinazolinonas/farmacología , Aminoácidos/deficiencia , Aminoácidos/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Células MCF-7 , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Sequestosoma-1/metabolismo
10.
Carbohydr Res ; 478: 10-17, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31039450

RESUMEN

A series of novel tricyclic quinazolinone-iminosugars 1 (a-c) were synthesized from the benzyl protected sugars through three steps. Firstly, the benzyl protected sugar (aldehyde) 5 reacted with o-aminobenzamide by the iodine-induced oxidative condensation to afford the corresponding aldo-quizanolinone 6. Secondly, through the intramolecular cyclization of the unprotected OH and the amide NH in 6, the tricyclic compounds 7 and 8 were constructed by the key Mitsunobu reaction. Finally, removal of the benzyl group gave the target tricyclic quinazolinone-iminosugars 1. The protocol was effective for the preparation of the tricyclic iminosugars in satisfactory yield. Interestingly, an unusual C-2 epimerization was observed with d-mannose and d-ribose compounds under the conditions of the Mitsunobu reaction that generated the products having the trans configuration at the C-2 and C-3 positions. Unfortunately, such tricyclic quinazolinone-iminosugars showed no inhibitory effects on the tested five glycosidases.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glicósido Hidrolasas/antagonistas & inhibidores , Iminoazúcares/farmacología , Quinazolinonas/farmacología , Aspergillus niger/enzimología , Canavalia/enzimología , Conformación de Carbohidratos , Café/enzimología , Ciclización , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Escherichia coli/enzimología , Glicósido Hidrolasas/metabolismo , Iminoazúcares/síntesis química , Iminoazúcares/química , Prunus dulcis/enzimología , Quinazolinonas/síntesis química , Quinazolinonas/química
11.
J Cell Physiol ; 234(10): 18970-18984, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30916359

RESUMEN

Traditional Chinese medicine (TCM) are both historically important therapeutic agents and important source of new drugs. Halofuginone (HF), a small molecule alkaloid derived from febrifugine, has been shown to exert strong antiproliferative effects that differ markedly among various cell lines. However, whether HF inhibits MCF-7 cell growth in vitro and underlying mechanisms of this process are not yet clear. Here, we offer the strong evidence of the connection between HF treatment, exosome production and proliferation of MCF-7 cells. Our results showed that HF inhibits MCF-7 cell growth in both time- and dose-dependent manner. Further microRNA (miRNA) profiles analysis in HF treated and nontreated MCF-7 cell and exosomes observed that six miRNAs are particularly abundant and sorted in exosomes. miRNAs knockdown experiment in exosomes and the MCF-7 growth inhibition assay showed that exosomal microRNA-31 (miR-31) modulates MCF-7 cells growth by specially targeting the histone deacetylase 2 (HDAC2), which increases the levels of cyclin-dependent kinases 2 (CDK2) and cyclin D1 and suppresses the expression of p21. In conclusion, these data indicate that inhibition of exosome production reduces exosomal miR-31, which targets the HDAC2 and further regulates the level of cell cycle regulatory proteins, contributing to the anticancer functions of HF. Our data suggest a new role for HF and the exosome production in tumorigenesis and may provide novel insights into prevention and treatment of breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Exosomas/genética , Histona Desacetilasa 2/metabolismo , MicroARNs/genética , Piperidinas/farmacología , Quinazolinonas/farmacología , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Células MCF-7 , Medicina Tradicional China
12.
J Cell Biochem ; 119(5): 4009-4020, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29231257

RESUMEN

Traditional Chinese medicines have been recognized as especially promising anticancer agents in modern anticancer research. Halofuginone (HF), an analog of quinazolinone alkaloid extracted from Dichroa febrifuga, is widely used in traditional medicine. However, whether HF inhibits the growth of breast cancer cells and/or reduces the migration and invasion of MCF-7 human breast cancer cells, as well as the underlying mechanisms in vitro, remains unclear. In this study, we report that an HF extract inhibits the growth of MCF-7 cells and reduces their migration and invasion, an important feature of potential anticancer agents. In addition, HF significantly increases the activation of autophagy, which is closely associated with tumor metastasis. As STMN1 and p53 have been closely implicated in breast cancer progression, we analyzed their expression in the context of HF extract treatment. Western blot analysis showed that HF suppresses STMN1 and p53 expression and activity in an autophagy-dependent manner. Collectively, these data indicate that activation of autophagy reduces expression of STMN1 and p53, and the migration and invasion of cancer cells contributes to the anti-cancer effects of the HF. These findings may provide new insight into breast cancer prevention and therapy.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Piperidinas/farmacología , Quinazolinonas/farmacología , Estatmina/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Femenino , Humanos , Células MCF-7 , Invasividad Neoplásica , Piperidinas/química , Quinazolinonas/química
13.
Bioorg Med Chem Lett ; 27(20): 4601-4605, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28943043

RESUMEN

The discovery of antiviral activity of 2,3-disubstituted quinazolinones, prepared by a one-pot, three-component condensation of isatoic anhydride with amines and aldehydes, against Herpes Simplex Virus (HSV)-1 is reported. Sequential iterative synthesis/antiviral assessment allowed structure-activity relationship (SAR) generation revealing synergistic structural features required for potent anti-HSV-1 activity. The most potent derivatives show greater efficacy than acyclovir against acute HSV-1 infections in neurons and minimal toxicity to the host.


Asunto(s)
Herpesvirus Humano 1/efectos de los fármacos , Quinazolinonas/química , Quinazolinonas/farmacología , Aciclovir/farmacología , Animales , Antivirales/química , Antivirales/farmacología , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Inmunoprecipitación de Cromatina , Evaluación Preclínica de Medicamentos , Humanos , Relación Estructura-Actividad , Células Vero
14.
Fitoterapia ; 119: 136-149, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28495308

RESUMEN

Quinazolinones belong to a family of heterocyclic nitrogen compounds that have attracted increasing interest because of their broad spectrum of biological functions. This review describes three types of natural quinazolinones and their synthesized derivatives and summarizes their various pharmacological activities, including antifungal, anti-tumor, anti-malaria, anticonvulsant, anti-microbial, anti-inflammatory and antihyperlipidemic activities. In addition, structure-activity relationships of quinazolinone derivatives are also reviewed.


Asunto(s)
Quinazolinonas/farmacología , Antiinfecciosos/farmacología , Antiinflamatorios/farmacología , Anticonvulsivantes/farmacología , Antifúngicos/farmacología , Antimaláricos/farmacología , Antineoplásicos/farmacología , Hipolipemiantes/farmacología , Estructura Molecular , Relación Estructura-Actividad
15.
J Am Heart Assoc ; 6(5)2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28487390

RESUMEN

BACKGROUND: The amino acid response (AAR) is an evolutionarily conserved protective mechanism activated by amino acid deficiency through a key kinase, general control nonderepressible 2. In addition to mobilizing amino acids, the AAR broadly affects gene and protein expression in a variety of pathways and elicits antifibrotic, autophagic, and anti-inflammatory activities. However, little is known regarding its role in cardiac stress. Our aim was to investigate the effects of halofuginone, a prolyl-tRNA synthetase inhibitor, on the AAR pathway in cardiac fibroblasts, cardiomyocytes, and in mouse models of cardiac stress and failure. METHODS AND RESULTS: Consistent with its ability to inhibit prolyl-tRNA synthetase, halofuginone elicited a general control nonderepressible 2-dependent activation of the AAR pathway in cardiac fibroblasts as evidenced by activation of known AAR target genes, broad regulation of the transcriptome and proteome, and reversal by l-proline supplementation. Halofuginone was examined in 3 mouse models of cardiac stress: angiotensin II/phenylephrine, transverse aortic constriction, and acute ischemia reperfusion injury. It activated the AAR pathway in the heart, improved survival, pulmonary congestion, left ventricle remodeling/fibrosis, and left ventricular function, and rescued ischemic myocardium. In human cardiac fibroblasts, halofuginone profoundly reduced collagen deposition in a general control nonderepressible 2-dependent manner and suppressed the extracellular matrix proteome. In human induced pluripotent stem cell-derived cardiomyocytes, halofuginone blocked gene expression associated with endothelin-1-mediated activation of pathologic hypertrophy and restored autophagy in a general control nonderepressible 2/eIF2α-dependent manner. CONCLUSIONS: Halofuginone activated the AAR pathway in the heart and attenuated the structural and functional effects of cardiac stress.


Asunto(s)
Aminoácidos/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Insuficiencia Cardíaca/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Quinazolinonas/farmacología , Estrés Fisiológico , Aminoácidos/deficiencia , Aminoacil-ARNt Sintetasas/antagonistas & inhibidores , Aminoacil-ARNt Sintetasas/metabolismo , Animales , Autofagia/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/prevención & control , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
16.
Biochem J ; 474(11): 1867-1877, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28381646

RESUMEN

Until recently, one of the major limitations of hydrogen/deuterium exchange mass spectrometry (HDX-MS) was the peptide-level resolution afforded by proteolytic digestion. This limitation can be selectively overcome through the use of electron-transfer dissociation to fragment peptides in a manner that allows the retention of the deuterium signal to produce hydrogen/deuterium exchange tandem mass spectrometry (HDX-MS/MS). Here, we describe the application of HDX-MS/MS to structurally screen inhibitors of the oncogene phosphoinositide 3-kinase catalytic p110α subunit. HDX-MS/MS analysis is able to discern a conserved mechanism of inhibition common to a range of inhibitors. Owing to the relatively minor amounts of protein required, this technique may be utilised in pharmaceutical development for screening potential therapeutics.


Asunto(s)
Antineoplásicos/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Inhibidores Enzimáticos/metabolismo , Modelos Moleculares , Fragmentos de Péptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Fosfatidilinositol 3-Quinasa Clase I , Fosfatidilinositol 3-Quinasa Clase Ia/química , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Medición de Intercambio de Deuterio , Evaluación Preclínica de Medicamentos/métodos , Transporte de Electrón , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Indazoles/química , Indazoles/metabolismo , Indazoles/farmacología , Peso Molecular , Oligonucleótidos/antagonistas & inhibidores , Oligonucleótidos/química , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Conformación Proteica , Purinas/química , Purinas/metabolismo , Purinas/farmacología , Piridazinas , Quinazolinonas/química , Quinazolinonas/metabolismo , Quinazolinonas/farmacología , Quinolinas/química , Quinolinas/metabolismo , Quinolinas/farmacología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Reproducibilidad de los Resultados , Procesamiento de Señales Asistido por Computador , Sulfonamidas/química , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Espectrometría de Masas en Tándem , Triazinas/química , Triazinas/metabolismo , Triazinas/farmacología
17.
Drug Discov Ther ; 10(4): 188-94, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27594295

RESUMEN

In this study, a series of novel scaffold-based 3-(((substituted phenyl)amino)methyl)-2-methylquinazolin-4(3H)-one compounds, 3a-3r, was synthesized, characterized, and screened for its in vitro activity against the H37Ra strain of Mycobacterium tuberculosis. A number of analogs were found to have highly potent anti-tuberculosis activity. Compound 3m in particular had potent activity equal to that of the standard anti-tuberculosis drug rifampicin. New leads can be generated with the model developed in this study and this model will be optimized with the eventual goal of preparing new anti-tuberculosis agents.


Asunto(s)
Antituberculosos/síntesis química , Diseño de Fármacos , Simulación del Acoplamiento Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Quinazolinonas/síntesis química , Antituberculosos/farmacología , Simulación por Computador , Evaluación Preclínica de Medicamentos , Pruebas de Sensibilidad Microbiana , Quinazolinonas/farmacología , Rifampin/farmacología , Relación Estructura-Actividad
18.
J Med Chem ; 59(17): 7915-35, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27531604

RESUMEN

Bruton's tyrosine kinase (BTK) belongs to the TEC family of nonreceptor tyrosine kinases and plays a critical role in multiple cell types responsible for numerous autoimmune diseases. This article will detail the structure-activity relationships (SARs) leading to a novel second generation series of potent and selective reversible carbazole inhibitors of BTK. With an excellent pharmacokinetic profile as well as demonstrated in vivo activity and an acceptable safety profile, 7-(2-hydroxypropan-2-yl)-4-[2-methyl-3-(4-oxo-3,4-dihydroquinazolin-3-yl)phenyl]-9H-carbazole-1-carboxamide 6 (BMS-935177) was selected to advance into clinical development.


Asunto(s)
Antirreumáticos/química , Carbazoles/química , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinonas/química , Administración Oral , Agammaglobulinemia Tirosina Quinasa , Animales , Antirreumáticos/síntesis química , Antirreumáticos/farmacocinética , Antirreumáticos/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Disponibilidad Biológica , Carbazoles/síntesis química , Carbazoles/farmacocinética , Carbazoles/farmacología , Línea Celular , Cristalografía por Rayos X , Perros , Humanos , Macaca fascicularis , Ratones , Microsomas Hepáticos/metabolismo , Permeabilidad , Proteínas Tirosina Quinasas/química , Quinazolinonas/síntesis química , Quinazolinonas/farmacocinética , Quinazolinonas/farmacología , Relación Estructura-Actividad
19.
J Invest Dermatol ; 136(10): 1990-2002, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27369778

RESUMEN

Phototherapy with UV light is a standard treatment for psoriasis, yet the mechanisms underlying the therapeutic effects are not well understood. Studies in human and mouse keratinocytes and in the skin tissues from human patients and mice showed that UV treatment triggers ubiquitination and downregulation of the type I IFN receptor chain IFNAR1, leading to suppression of IFN signaling and an ensuing decrease in the expression of inflammatory cytokines and chemokines. The severity of imiquimod-induced psoriasiform inflammation was greatly exacerbated in skin of mice deficient in IFNAR1 ubiquitination (Ifnar1(SA)). Furthermore, these mice did not benefit from UV phototherapy. Pharmacologic induction of IFNAR1 ubiquitination and degradation by an antiprotozoal agent halofuginone also relieved psoriasiform inflammation in wild-type but not in Ifnar1(SA) mice. These data identify downregulation of IFNAR1 by UV as a major mechanism of the UV therapeutic effects against the psoriatic inflammation and provide a proof of principle for future development of agents capable of inducing IFNAR1 ubiquitination and downregulation for the treatment of psoriasis.


Asunto(s)
Inflamación/terapia , Piperidinas/farmacología , Psoriasis/terapia , Quinazolinonas/farmacología , Receptor de Interferón alfa y beta/metabolismo , Terapia Ultravioleta/métodos , Animales , Línea Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de la radiación , Humanos , Inflamación/patología , Queratinocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Psoriasis/patología , Receptor de Interferón alfa y beta/genética , Transducción de Señal , Piel/patología , Ubiquitinación/efectos de los fármacos , Ubiquitinación/efectos de la radiación
20.
Blood ; 128(7): 934-47, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27297795

RESUMEN

Novel agents such as the Bcl-2 inhibitor venetoclax (ABT-199) are changing treatment paradigms for chronic lymphocytic leukemia (CLL) but important problems remain. Although some patients exhibit deep and durable responses to venetoclax as a single agent, other patients harbor subpopulations of resistant leukemia cells that mediate disease recurrence. One hypothesis for the origin of resistance to venetoclax is by kinase-mediated survival signals encountered in proliferation centers that may be unique for individual patients. An in vitro microenvironment model was developed with primary CLL cells that could be incorporated into an automated high-content microscopy-based screen of kinase inhibitors (KIs) to identify agents that may improve venetoclax therapy in a personalized manner. Marked interpatient variability was noted for which KIs were effective; nevertheless, sunitinib was identified as the most common clinically available KI effective in overcoming venetoclax resistance. Examination of the underlying mechanisms indicated that venetoclax resistance may be induced by microenvironmental signals that upregulate antiapoptotic Bcl-xl, Mcl-1, and A1, which can be counteracted more efficiently by sunitinib than by ibrutinib or idelalisib. Although patient-specific drug responses are common, for many patients, combination therapy with sunitinib may significantly improve the therapeutic efficacy of venetoclax.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Inhibidores de Proteínas Quinasas/análisis , Inhibidores de Proteínas Quinasas/uso terapéutico , Sulfonamidas/uso terapéutico , Adenina/análogos & derivados , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Microambiente Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Imagenología Tridimensional , Indoles/farmacología , Mutación/genética , Piperidinas , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Quinazolinonas/farmacología , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Sulfonamidas/farmacología , Sunitinib , Regulación hacia Arriba/efectos de los fármacos , Proteína bcl-X/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA