Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
mBio ; 15(9): e0138524, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39162560

RESUMEN

Infection with respiratory syncytial virus (RSV) is a major cause of acute lower respiratory tract disease in young children and older people. Despite intensive efforts over the past few decades, no direct-acting small-molecule agents against RSV are available. Most small-molecule candidates targeting the RSV fusion (F) protein pose a considerable risk of inducing drug-resistant mutations. Here, we explored the in vitro and in vivo virological properties of the K394R variant, a cross-resistant mutant capable of evading multiple RSV fusion inhibitors. Our results demonstrated that the K394R variant is highly fusogenic in vitro and more pathogenic than the parental strain in vivo. The small molecule (2E,2'E)-N,N'-((1R,2S,3S)-3-hydroxycyclohexane-1,2-diyl)bis(3-(2-bromo-4-fluorophenyl) acrylamide) (CL-A3-7), a structurally optimized compound derived from a natural caffeoylquinic acid derivative, substantially reduced in vitro and in vivo infections of both wild-type RSV and the K394R variant. Mechanistically, CL-A3-7 significantly inhibited virus-cell fusion during RSV entry by blocking the interaction between the viral F protein and the cellular insulin-like growth factor 1 receptor (IGF1R). Collectively, these results indicate severe disease risks caused by the K394R variant and reveal a new anti-RSV mechanism to overcome K394R-associated resistance. IMPORTANCE: Respiratory syncytial virus (RSV) infection is a major public health concern, and many small-molecule candidates targeting the viral fusion (F) protein are associated with a considerable risk of inducing drug-resistant mutations. This study investigated virological features of the K394R variant, a mutant strain conferring resistance to multiple RSV fusion inhibitors. Our results demonstrated that the K394R variant is highly fusogenic in cell cultures and more pathogenic than the parental strain in mice. The small-molecule inhibitor CL-A3-7 substantially reduced in vitro and in vivo infections of both wild-type RSV and the K394R variant by blocking the interaction of viral F protein with its cellular receptor, showing a new mechanism of action for small-molecules to inhibit RSV infection and overcome K394R-associated resistance.


Asunto(s)
Antivirales , Farmacorresistencia Viral , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Proteínas Virales de Fusión , Internalización del Virus , Internalización del Virus/efectos de los fármacos , Animales , Humanos , Antivirales/farmacología , Farmacorresistencia Viral/genética , Farmacorresistencia Viral/efectos de los fármacos , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Proteínas Virales de Fusión/antagonistas & inhibidores , Ratones , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/fisiología , Ratones Endogámicos BALB C , Línea Celular , Femenino
2.
J Nanobiotechnology ; 22(1): 459, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085911

RESUMEN

BACKGROUND: Sunitinib is a multikinase inhibitor used to treat patients with advanced renal cell carcinoma (RCC). However, sunitinib toxicity makes it a double-edged sword. Potent immune modulation by sunitinib extends to nuclear interactions. To address these issues, there is an urgent need for delivery vectors suitable for sunitinib treatment. METHODS: We developed PEGylated liposomes as delivery vectors to precisely target sunitinib (lipo-sunitinib) to RCC tumors. Further investigations, including RNA sequencing (RNA-seq), were performed to evaluate transcriptomic changes in these pathways. DiI/DiR-labeled lipo-sunitinib was used for the biodistribution analysis. Flow cytometry and immunofluorescence (IF) were used to examine immune modulation in orthotopic RCC models. RESULTS: The evaluation of results indicated that lipo-sunitinib precisely targeted the tumor site to induce autophagy and was readily taken up by RCC tumor cells. In addition, transcriptomic assays revealed that following lipo-sunitinib treatment, autophagy, antigen presentation, cytokine, and chemokine production pathways were upregulated, whereas the epithelial-mesenchymal transition (EMT) pathway was downregulated. In vivo data provided evidence supporting the inhibitory effect of lipo-sunitinib on RCC tumor progression and metastasis. Flow cytometry further demonstrated that liposunitinib increased the infiltration of effector T cells (Teffs) and conventional type 1 dendritic cells (cDC1s) into the tumor. Furthermore, systemic immune organs such as the tumor-draining lymph nodes, spleen, and bone marrow exhibited upregulated anticancer immunity following lipo-sunitinib treatment. CONCLUSION: Our findings demonstrated that lipo-sunitinib is distributed at the RCC tumor site, concurrently inducing potent autophagy, elevating antigen presentation, activating cytokine and chemokine production pathways, and downregulating EMT in RCC cells. This comprehensive approach significantly enhanced tumor inhibition and promoted anticancer immune modulation.


Asunto(s)
Autofagia , Carcinoma de Células Renales , Neoplasias Renales , Liposomas , Polietilenglicoles , Sunitinib , Carcinoma de Células Renales/tratamiento farmacológico , Sunitinib/farmacología , Autofagia/efectos de los fármacos , Animales , Liposomas/química , Neoplasias Renales/tratamiento farmacológico , Ratones , Línea Celular Tumoral , Polietilenglicoles/química , Humanos , Inmunomodulación/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Distribución Tisular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino
3.
Drug Dev Res ; 85(2): e22170, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38481011

RESUMEN

A four-step synthetic process has been developed to prepare 1,3,5,8-tetrahydroxyxanthone (2a) and its isomer 1,3,7,8-tetrahydroxyxanthone (2b). 25 more xanthones were also synthesized by a modified scheme. Xanthone 2a was identified as the most active inhibitor against both α-glucosidase and aldose reductase (ALR2), with IC50 values of 7.8 ± 0.5 µM and 63.2 ± 0.6 nM, respectively, which was far active than acarbose (35.0 ± 0.1 µM), and a little more active than epalrestat (67.0 ± 3.0 nM). 2a was also confirmed as the most active antioxidant in vitro with EC50 value of 8.9 ± 0.1 µM. Any structural modification including methylation, deletion, and position change of hydroxyl group in 2a will cause an activity loss in inhibitory and antioxidation. By applying a H2 O2 -induced oxidative stress nematode model, it was confirmed that xanthone 2a can be absorbed by Caenorhabditis elegans and is bioavailable to attenuate in vivo oxidative stress, including the effects on lifespan, superoxide dismutase, Catalase, and malondialdehyde. 2a was verified with in vivo hypoglycemic effect and mitigation of embryo malformations in high glucose. All our data support that xanthone 2a behaves triple roles and is a potential agent to treat diabetic mellitus, gestational diabetes mellitus, and diabetic complications.


Asunto(s)
Complicaciones de la Diabetes , Diabetes Mellitus , Xantonas , Humanos , Relación Estructura-Actividad , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Hipoglucemiantes/química , alfa-Glucosidasas/química , alfa-Glucosidasas/metabolismo , Complicaciones de la Diabetes/tratamiento farmacológico , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Xantonas/farmacología , Xantonas/uso terapéutico , Simulación del Acoplamiento Molecular , Diabetes Mellitus/tratamiento farmacológico
5.
Bioorg Chem ; 130: 106223, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36356372

RESUMEN

Eight hybrids of amantadine (ATD) with a natural modulator gardenamide A (GA) via an alkylene carbonyl bridge or alkylene bridge have been designed and synthesized. Evaluated by electrophysiological assay, compound 5b was confirmed an enhanced NMDAR antagonist compared to ATD with IC50 value of 10.2 ± 1.2 µM. 5b has been demonstrated to reverse the damages of behavioral performance, the loss of dopaminergic neurons, the reduction of TH positive, and the increase of α-synuclein in both MPTP-treated mice and zebrafish models. In both ethological and ecological experiments, the activity of 5b was confirmed better than ATD or ATD/GA combination, and was almost equal to the positive selegiline. In vivo and in vitro, 5b is shown to reverse the ascend of NR1 and i-NOS levels. This candidate was also demonstrated the activity to down-regulated MPTP-increased Ca2+ influx in SH-SY5Y cells in a steep and sharp mode. It is displayed that 5b exerts neuroprotective effect partly by activating the PI3K/Akt signaling pathway. Taken all together, our data support that 5b is a more promising agent against PD than ATD.


Asunto(s)
N-Metilaspartato , Neuroblastoma , Humanos , Ratones , Animales , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Fosfatidilinositol 3-Quinasas/metabolismo , Pez Cebra/metabolismo , Ratones Endogámicos C57BL , Amantadina/farmacología
6.
Bioorg Chem ; 131: 106300, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36455484

RESUMEN

The primary objective of this study was to investigate the structure-activity relationship of a new series of 5F-like Aldose Reductase Inhibitors (ARIs) using in silico docking method. In this perspective, 6 novel ARIs have been designed and synthesized. Evaluation of the inhibition of these compounds to ALR2 was carried on with epalrestat and 5F as the references. It was found that the spacer of 5F-like ARIs has a great influence on their inhibitory activity. Rigid spacer with length equal to 3 âˆ¼ 4 carbon alkyl chain brings about better inhibitory activity. Among them, compound 4b was verified as the most active ARIs, where its IC50 value was 16.8 ± 1.3 nM. Furthermore, in silico docking studies using AutoDock 4.2 as well as molecular simulation using GROMACS 2022.1 showed that 5F-like ARIs adopt a dual-occupation mode. The interaction energy (-25 to -74 kcal/mol), as well as MM-GBSA binding free energy (-37 to -65 kcal/mol) was positively correlated with their ALR2 inhibition constant (2000 to 16.8 nM). Docking interaction explained well the structure-activity relationship. A pharmacophore model has been set up for 5F-like ARIs thereafter. This model indicates that as an effective ARI, the entity should have four characteristics: an aromatic center, two hydrogen bond donors, and one hydrogen bond acceptor. By the way, all the 5F-like ARIs reported here are good to mild antioxidant with EC50 value between 13.6 ± 1.2 and 71.1 ± 3.2 µM. All our data direct the further development of more optimal ARIs for the treatment of diabetic complication in the future.


Asunto(s)
Aldehído Reductasa , Complicaciones de la Diabetes , Humanos , Inhibidores Enzimáticos/química , Relación Estructura-Actividad , Complicaciones de la Diabetes/tratamiento farmacológico , Simulación del Acoplamiento Molecular
7.
Bioorg Chem ; 120: 105624, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35065464

RESUMEN

A series of 5f-based new compounds has been designed and synthesized. In vitro screening demonstrated that the binding affinity and selectivity on aldose reductase (AR) were positively correlated with its antioxidation capacity. Compound 6d was verified the most active candidate, where its IC50, selective index (SI), and EC50 value was 22.3 ± 1.6 nM, 236.2, and 8.7 µM respectively. 6d was confirmed as both an excellent antioxidant and aldose reductase inhibitor (ARI). It was identified as a mixed type ARI with Ki and Kis values of 23.94 and 1.20 nM. When evaluated by a high-glucose impaired chicken embryo model, it was found that 6d attenuated the incidence of neural tube defect (NTD) and death rate in a dose-dependent manner. It significantly improved the hyperglycemia-induced abnormalities of body weight and morphology of chicken embryos. 6d reversed the hyperglycemia-raised AR activity, sorbitol accumulation, reactive oxygen species (ROS) and malondialdehyde (MDA) levels. It restored the high-glucose-reduced Pax3 protein expression. At the same dose (0.5 µM), 6d showed better effects than 5f in all the above detections. By the way, 6d did not affect hyperglycemia-elevated aldehyde reductase (ALR1) activity. This evidence together with its kinetic properties, implicated that 6d is a high selective ARI without the suspicion of promiscuity. 6d was proved here an effective agent to treat diabetic peripheral neuropathy (DPN). Whether 6d has potential to treat other types of diabetic complications (DC) needs to be further investigation.


Asunto(s)
Aldehído Reductasa , Hiperglucemia , Animales , Antioxidantes/farmacología , Embrión de Pollo , Inhibidores Enzimáticos/uso terapéutico , Glucosa , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes
8.
Bioorg Chem ; 110: 104769, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33677247

RESUMEN

Three hybrids of dihydro-artemisinin (DHA) with ß-aminopropionic acid, γ-aminobutyric acid, and histamine have been designed and synthesized. The conjugate of DHA with GABA labelled as 5b was confirmed the most active candidate against both Cort- and SNP-induced PC12 cell impairments with EC50 value of 8.04 ± 0.35, and 9.38 ± 0.56 µM, respectively. 5b was clearly highlighted as a good modulator on protein expression of Akt, Bcl-2, and Bax, indicating its functions against programmed cell apoptosis. 5b significantly reversed the Cort-induced excessive calcium influx and release from internal organelles. It was demonstrated the ability to express increased levels of ß-tubulin III and to up-regulate phosphorylation level of cAMP response element-binding protein (CREB), leading to cell differentiation. It can penetrate blood - brain barrier (BBB) with propriate stability. Altogether, these data strongly support that 5b is a potential anti-depressant.


Asunto(s)
Antidepresivos/química , Antidepresivos/farmacología , Artemisininas/química , Artemisininas/farmacología , Ácido gamma-Aminobutírico/química , Ácido gamma-Aminobutírico/farmacología , Animales , Barrera Hematoencefálica , Calcio/metabolismo , Cortisona/metabolismo , Membranas Artificiales , Estructura Molecular , Células PC12 , Permeabilidad , Ratas
9.
Eur J Med Chem ; 211: 113067, 2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33338868

RESUMEN

Seven tacrine/CHR21 conjugates have been designed and synthesized. Compound 8-7 was confirmed as the most active AChE inhibitor with IC50 value of 5.8 ± 1.4 nM, which was 7.72-fold stronger than tacrine. It was also shown as a strong BuChE inhibitor (IC50 value of 3.7 ± 1.3 nM). 8-7 was clearly highlighted not only as an excellent ChEs inhibitor, but also as a good modulator on protein expression of AChE, p53, Bax, Bcl-2, LC3, p62, and ULK, indicating its functions against programmed cell apoptosis and decrease of autophagy. 8-7 significantly reversed the glutamate-induced dysfunctions including excessive calcium influx and release from internal organelles, overproduction of nitric oxide (NO) and Aß high molecular weight oligomer. This compound can penetrate blood-brain barrier (BBB). The in vivo hepatotoxicity assay indicated that 8-7 was much less toxic than tacrine. Altogether, these data strongly support that 8-7 is a potential multitarget-directed ligand (MTDL) for treating Alzheimer's disease (AD).


Asunto(s)
Acetilcolinesterasa/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Iridoides/uso terapéutico , Tacrina/uso terapéutico , Acetilcolinesterasa/farmacología , Enfermedad de Alzheimer/patología , Autofagia , Diseño de Fármacos , Humanos , Iridoides/farmacología , Estructura Molecular , Relación Estructura-Actividad , Tacrina/farmacología
10.
Arch Pharm (Weinheim) ; 352(9): e1900100, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31313862

RESUMEN

The antioxidative effects of 30 xanthone derivatives (XDs) (XD-n, n = 1-30) in HepG2 cells were evaluated by the cellular antioxidant activity assay. Results showed that all XDs were antioxidants and 1,3,5,8-tetrahydroxy-9H-xanthen-9-one (XD-2) was the most active antioxidant. The all-oxygenated substituted xanthones extended the lifespan of wild-type N2 nematodes under normal culture conditions and XD-2 was the best one. XD-2 eliminated excessive intracellular reactive oxygen species and enhanced the expression levels and activities of the antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase. XD-2 inhibited the H2 O2 -increased phosphorylation levels of c-JUN N-terminal kinase, extracellular signal-regulated kinase, and p38 in HepG2 cells. In vivo, XD-2 also extended the lifespan of wild-type N2 nematodes under oxidative stress induced by paraquat, but failed in extending the lifespan of CF1038 (daf-16 deletion) and AY102 (pmk-1 deletion) mutant nematodes. It was revealed by real-time polymerase chain reaction that the genes daf-16, sir-2.1, akt-1, and age-1 were all inhibited by paraquat stimuli, while XD-2 reversed these inhibitions; in contrast, paraquat stimuli upregulated both the skn-1 and pmk-1 genes. However, treatment by XD-2 further increased the levels of both genes. These pieces of evidence implied that XD-2 promotes longevity through endogenous signaling pathways rather than through the antioxidative activity alone. Taken all together, it may be concluded that XD-2 is a promising antiageing agent.


Asunto(s)
Antioxidantes/farmacología , Proteínas de Caenorhabditis elegans/genética , Regulación de la Expresión Génica/efectos de los fármacos , Longevidad/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Xantonas/farmacología , Animales , Antioxidantes/química , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Células Hep G2 , Humanos , Longevidad/genética , Sistema de Señalización de MAP Quinasas/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Xantonas/química
11.
Arch Pharm (Weinheim) ; 352(6): e1800354, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31081964

RESUMEN

Nineteen antioxidant pseudopeptides were designed and synthesized. They were confirmed as mild antioxidants, in which L1-11 was the most active antioxidant with a cellular antioxidant activity (CAA) value of 5.65 ± 0.64 µmol QE/g, and L1-12 was the second most active one (5.58 ± 0.66 µmol QE/g). The existence of nonnatural amino acids in L1-12 increased its stability. Pretreatment with L1-12 dose-dependently extended the lifespan of Caenorhabditis elegans. L1-12 improved resistance against UVB irradiation, oxidative stress induced by paraquat, and thermal shock. It decreased the reactive oxygen species level and upregulated the superoxide dismutase activity inside C. elegans. This pseudopeptide sensitively enhanced the expressions of the Cat-1 and Nhr-8 genes to reduce oxidative damage, leading to an extension of the lifespan. All the evidence support that L1-12 may probably be a potential antiageing agent.


Asunto(s)
Aminoácidos/síntesis química , Antioxidantes/síntesis química , Caenorhabditis elegans/efectos de los fármacos , Diseño de Fármacos , Longevidad/efectos de los fármacos , Péptidos/síntesis química , Aminoácidos/química , Aminoácidos/farmacología , Animales , Antioxidantes/química , Antioxidantes/farmacología , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/efectos de la radiación , Proteínas de Caenorhabditis elegans/genética , Transportador de Aminoácidos Catiónicos 1/genética , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Expresión Génica/efectos de los fármacos , Longevidad/genética , Estructura Molecular , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Péptidos/química , Péptidos/farmacología , Receptores Citoplasmáticos y Nucleares/genética , Relación Estructura-Actividad
12.
AJR Am J Roentgenol ; 212(4): 717-726, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30699010

RESUMEN

OBJECTIVE: This study aimed to investigate the feasibility of intravoxel incoherent motion (IVIM) DWI and R2* (transverse relaxation rate) mapping to monitor the hyperacute therapeutic efficacy of desacetylvinblastine monohydrazide (DAVLBH) on an experimental hepatocellular carcinoma mouse model within 24 hours. MATERIALS AND METHODS: Forty-four mice were implanted with hepatocellular carcinoma and divided into three random groups. A treatment group and a control group underwent IVIM-DWI and R2* mapping examinations before and after a single injection of DAVLBH or saline at 1, 2, 4, and 24 hours. The pathology group was set for pathologic analysis, including H and E staining and CD31 and hypoxia-inducible factor (HIF)-1α immunohistochemical staining. RESULTS: DAVLBH caused hyperacute disruptions on the tumor capillaries in the treatment group. Water molecule diffusion (D), microcirculation perfusion (D*), and perfusion fraction (f) decreased initially but then gradually recovered to the baseline level by 24 hours after the first injection of DAVLBH. In contrast, R2* increased dramatically at 1 hour and then gradually decreased from 1 hour to 24 hours after treatment. D*, f, and D showed similar trends and were positively correlated with CD31 expression (r = 0.868, 0.721, and 0.730, respectively), but were negatively correlated with HIF-1α expression (r = -0.784, -0.737, and -0.673, respectively). R2* showed a negative correlation with CD31 expression (r = -0.823) and a positive correlation with HIF-1α expression (r = 0.791). CONCLUSION: Both IVIM-DWI and R2* mapping can adequately detect the vascular-disrupting effect of DAVLBH as early as 1 hour after injection in a mouse xenograft model. Moreover, D* and R2* are the two most sensitive hemodynamic parameters and can monitor the hyperacute changes associated with DAVLBH treatment in vivo.


Asunto(s)
Carcinoma Hepatocelular , Imagen de Difusión por Resonancia Magnética , Neoplasias Hepáticas , Vindesina , Animales , Femenino , Humanos , Ratones , Permeabilidad Capilar/efectos de los fármacos , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/tratamiento farmacológico , Imagen de Difusión por Resonancia Magnética/métodos , Modelos Animales de Enfermedad , Estudios de Factibilidad , Xenoinjertos , Procesamiento de Imagen Asistido por Computador , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/tratamiento farmacológico , Ratones Endogámicos BALB C , Microcirculación , Células Tumorales Cultivadas , Vindesina/farmacología
13.
Bioorg Chem ; 83: 461-467, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30448724

RESUMEN

Incorporation of carbobenzoxy-glycylprolyl (Z-GP) to either α or ß position of the hydrazine moiety in procarbazine (Pcb) has been carried on in 5-steps process. The overall yield was 32.7%. The new entity Z-GP-Pcb was confirmed targeting to fibroblast activation protein-α (FAPα). Z-GP-Pcb may be hydrolyzed by either isolated rhFAPα or tumor homogenate. It was shown far less cytotoxicity against NCI-H460 cell line than Pcb. Z-GP-Pcb was displayed the potency to reduce spermatoxcity in H22-bearing mice. The mechanism may be ascribed to the blockade of dehydrogenation by α-glycerolphosphate dehydrogenase. This candidate was further proved equal antitumor activity to Pcb. However, the introduction of Z-GP scaffold decreased myelosuppression. All the evidences support that Z-GP-Pcb is a better antitumor agent than Pcb.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Sanguíneas/efectos de los fármacos , Dipéptidos/uso terapéutico , Procarbazina/uso terapéutico , Profármacos/uso terapéutico , Espermatozoides/efectos de los fármacos , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Recuento de Células Sanguíneas , Plaquetas/efectos de los fármacos , Línea Celular Tumoral , Dipéptidos/síntesis química , Dipéptidos/farmacología , Dipéptidos/toxicidad , Diseño de Fármacos , Endopeptidasas , Eritrocitos/efectos de los fármacos , Gelatinasas/metabolismo , Humanos , Hidrólisis , Leucocitos/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Tamaño de los Órganos , Procarbazina/síntesis química , Procarbazina/farmacología , Procarbazina/toxicidad , Profármacos/síntesis química , Profármacos/farmacología , Profármacos/toxicidad , Serina Endopeptidasas/metabolismo , Recuento de Espermatozoides , Testículo/efectos de los fármacos
14.
Eur J Med Chem ; 151: 158-172, 2018 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-29609121

RESUMEN

Fifty 1,3-dioxyxanthone nitrates (4a ∼ i-n, n = 1-6) were designed and synthesized based on molecular similarity strategy. Incorporation of nitrate into 1,3-dioxyxanthones with electron-donating groups at 6-8 position brought about synergistic anticancer effect. Among them, compound 4g-4 was confirmed the most active agent against HepG-2 cells growth with an IC50 of 0.33 ±â€¯0.06 µM. It dose-dependently increased intramolecular NO levels. This activity was attenuated by either NO scavenger PTIO or mitochondrial aldehyde dehydrogenase (mtADH) inhibitor PCDA. Apoptosis analysis indicated different contributions of early/late apoptosis and necrosis to cell death for different dose of 4g-4. 4g-4 arrested more cells on S phase. Results from Western Blot implied that 4g-4 regulated p53/MDM2 to promote cancer cell apoptosis. All the evidences support that 4g-4 is a promising anti-cancer agent.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Donantes de Óxido Nítrico/química , Donantes de Óxido Nítrico/farmacología , Xantonas/química , Xantonas/farmacología , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Nitratos/síntesis química , Nitratos/química , Nitratos/farmacología , Donantes de Óxido Nítrico/síntesis química , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Proteína p53 Supresora de Tumor/metabolismo , Xantonas/síntesis química
15.
Theranostics ; 8(2): 384-398, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29290815

RESUMEN

Vinca alkaloids, the well-known tubulin-binding agents, are widely used for the clinical treatment of malignant tumors. However, little attention has been paid to their vascular disrupting effects, and the underlying mechanisms remain largely unknown. This study aims to investigate the vascular disrupting effect and the underlying mechanisms of vinca alkaloids. Methods: The capillary disruption assay and aortic ring assay were performed to evaluate the in vitro vascular disrupting effect of desacetylvinblastine monohydrazide (DAVLBH), a derivate of vinblastine, and the in vivo vascular disrupting effect was assessed on HepG2 xenograft model using magnetic resonance imaging, hematoxylin and eosin staining and immunohistochemistry. Tubulin polymerization, endothelial cell monolayer permeability, western blotting and immunofluorescence assays were performed to explore the underlying mechanisms of DAVLBH-mediated tumor vascular disruption. Results: DAVLBH has potent vascular disrupting activity both in vitro and in vivo. DAVLBH disrupts tumor vessels in a different manner than classical tubulin-targeting VDAs; it inhibits microtubule polymerization, promotes the internalization of vascular endothelial cadherin (VE-cadherin) and inhibits the recycling of internalized VE-cadherin to the cell membrane, thus increasing endothelial cell permeability and ultimately resulting in vascular disruption. DAVLBH-mediated promotion of VE-cadherin internalization and inhibition of internalized VE-cadherin recycling back to the cell membrane are partly dependent on inhibition of microtubule polymerization, and Src activation is involved in DAVLBH-induced VE-cadherin internalization. Conclusions: This study sheds light on the tumor vascular disrupting effect and underlying mechanisms of vinca alkaloids and provides new insight into the molecular mechanism of tubulin-targeting VDAs.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Vindesina/farmacología , Animales , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Cancer Lett ; 418: 239-249, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337108

RESUMEN

Multidrug resistance (MDR) mediated by ATP-binding cassette (ABC) transporters is the major obstacle for chemotherapeutic success. Although attempts have been made to circumvent ABC transporter-mediated MDR in past decades, there is still no effective agent in clinic. Here, we identified a vascular disrupting agent, Z-GP-DAVLBH, that significantly inhibited the growth of multidrug-resistant human hepatoma HepG2/ADM and human breast cancer MCF-7/ADR tumor xenografts, although these cells were insensitive to Z-GP-DAVLBH in vitro. Z-GP-DAVLBH increased the secretion of granulocyte-macrophage colony-stimulating factor in tumor tissues and serum of tumor-bearing mice to skew tumor-associated macrophages from the pro-tumor M2 phenotype to the antitumor M1 phenotype, thereby contributing to the induction of HepG2/ADM and MCF-7/ADR cell apoptosis. Our findings shed new light on the underlying mechanisms of VDAs in the treatment of drug-resistant tumors and provide strong evidence that Z-GP-DAVLBH should be a promising agent for overcoming MDR.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Femenino , Células Hep G2 , Humanos , Células MCF-7 , Macrófagos/clasificación , Macrófagos/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Fenotipo , Células THP-1 , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biochem Pharmacol ; 150: 54-63, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29371030

RESUMEN

(R,E)-N-(3-(2-acetamido-3-(benzyloxy) propanamido)propyl)-2-cyano-3-(4-hydroxy phenyl)acrylamide (hr5F) was design-synthesized based on bioactivity focus strategy as a potential agent to treat diabetic complicates. With in vitro enzyme assay, it is confirmed that hr5F is an effective ALR2 inhibitor with IC50 value of 2.60 ±â€¯0.15 nM, and selectivity index of 86.0 over ALR1, which is a little bit better than the reference Epalrestat (Epa). hr5F inhibits the increase of ALR2 enzyme activity and expression in human lens epithelial cells (HLECs) induced by high glucose. By applying western blot, it was found that hr5F alleviates the high glucose-induced superoxide overproduction insults by regulating SIRT1-PGC-1α/Nrf2 pathway, together with regulating NRF-1, mtTFA, Bax/Bcl-2 to ameliorate cell apoptosis. The in vivo effects of hr5F on short term streptozocin (STZ)-induced diabetic mice confirm the same functions disclosed in vitro. All the evidences support that hr5F may serve as a promising agent in the treatment of diabetic complications with close efficacy and broader indication than the reference Epa.


Asunto(s)
Aldehído Reductasa/metabolismo , Diabetes Mellitus Experimental/metabolismo , Hipoglucemiantes/uso terapéutico , Factor 2 Relacionado con NF-E2/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/fisiología , Sirtuina 1/fisiología , Aldehído Reductasa/antagonistas & inhibidores , Animales , Células Cultivadas , Complicaciones de la Diabetes/tratamiento farmacológico , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Hipoglucemiantes/farmacología , Masculino , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Resultado del Tratamiento
18.
J Chromatogr Sci ; 56(1): 18-24, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977348

RESUMEN

Four flavonoids including apigenin-7,4'-dimethylether, genkwanin, quercetin, and kaempferol were isolated in a preparative or semi-preparative scale from the leaves of wild Aquilaria sinensis using an improved preparative high-speed counter-current chromatography apparatus. The separations were performed with a two-phase solvent system composed of hexane-ethyl acetate, methanol-water at suitable volume ratios. The obtained fractions were analyzed by HPLC, and the identification of each target compound was carried out by ESI-MS and NMR. The yields of the above four target flavonoids were 4.7, 10.0, 11.0 and 4.4%, respectively. All these four flavonoids exhibited nitrite scavenging activities with the clearance rate of 12.40 ± 0.20%, 5.84 ± 0.03%, 28.10 ± 0.17% and 5.19 ± 0.11%, respectively. Quercetin was originally isolated from the Thymelaeaceae family, while kaempferol was isolated from the Aquilaria genus for the first time. In cytotoxicity test these two flavonoids exhibited moderate inhibitory activities against HepG2 cells with the IC50 values of 12.54 ± 1.37 and 38.63 ± 4.05 µM, respectively.


Asunto(s)
Distribución en Contracorriente/métodos , Flavonoides/análisis , Flavonoides/aislamiento & purificación , Thymelaeaceae/química , Antineoplásicos/análisis , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Flavonoides/química , Flavonoides/farmacología , Células Hep G2 , Humanos , Hojas de la Planta/química
19.
Eur J Med Chem ; 143: 1768-1778, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29129511

RESUMEN

Four 5,6-dimethylxanthone-4-acetic acid (D) and pyranoxanthone (P) hybrids (D-P-n) were design-synthesized based on multi-target-addressed strategy. D-P-4 was confirmed as the most active agent against HepG-2 cell line growth with an IC50 of 0.216 ± 0.031 µM. Apoptosis analysis indicated different contributions of early/late apoptosis/necrosis to cell death for both monomers, the combination (D + P in 1:1 mol ratio) and D-P-4. They all arrested more cells on S phase. Western Blot implied that D-P-4 regulated p53/MDM2 to a better healthy state. Moreover, it improved Bax/Bcl-2 signaling pathway to increase cancer cell apoptosis. In all cases studied, D-P-4 showed the best activity and synergistic effect. All the evidences support that D-P-4 is a better anti-cancer therapy with multi-target functions.


Asunto(s)
Antineoplásicos/farmacología , Piranos/farmacología , Xantonas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células Hep G2 , Humanos , Estructura Molecular , Piranos/química , Relación Estructura-Actividad , Células Tumorales Cultivadas , Xantonas/química
20.
J Clin Invest ; 127(10): 3689-3701, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28846068

RESUMEN

Blood vessels in the tumor periphery have high pericyte coverage and are resistant to vascular disrupting agents (VDAs). VDA treatment resistance leads to a viable peripheral tumor rim that contributes to treatment failure and disease recurrence. Here, we provide evidence to support a hypothesis that shifting the target of VDAs from tumor vessel endothelial cells to pericytes disrupts tumor peripheral vessels and the viable rim, circumventing VDA treatment resistance. Through chemical engineering, we developed Z-GP-DAVLBH (from the tubulin-binding VDA desacetylvinblastine monohydrazide [DAVLBH]) as a prodrug that can be selectively activated by fibroblast activation protein α (FAPα) in tumor pericytes. Z-GP-DAVLBH selectively destroys the cytoskeleton of FAPα-expressing tumor pericytes, disrupting blood vessels both within the core and around the periphery of tumors. As a result, Z-GP-DAVLBH treatment eradicated the otherwise VDA-resistant tumor rim and led to complete regression of tumors in multiple lines of xenografts without producing the drug-related toxicity that is associated with similar doses of DAVLBH. This study demonstrates that targeting tumor pericytes with an FAPα-activated VDA prodrug represents a potential vascular disruption strategy in overcoming tumor resistance to VDA treatments.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Profármacos/farmacología , Vinblastina , Células A549 , Animales , Células 3T3 BALB , Endopeptidasas , Gelatinasas/biosíntesis , Células HeLa , Células Hep G2 , Humanos , Proteínas de la Membrana/biosíntesis , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Pericitos , Serina Endopeptidasas/biosíntesis , Vinblastina/análogos & derivados , Vinblastina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA