Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 324
Filtrar
1.
Int J Nanomedicine ; 19: 6519-6546, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38957181

RESUMEN

Background: Salidroside (SAL) is the most effective component of Rhodiola rosea, a traditional Chinese medicine. Cryptotanshinone (CT) is the main fat-soluble extract of Salvia miltiorrhiza, exhibiting considerable potential for application in osteogenesis. Herein, a polycaprolactone/gelatin nanofiber membrane loaded with CT and SAL (PSGC membrane) was successfully fabricated via coaxial electrospinning and characterized. Methods and Results: This membrane capable of sustained and controlled drug release was employed in this study. Co-culturing the membrane with bone marrow mesenchymal stem cells and human umbilical vein endothelial cells revealed excellent biocompatibility and demonstrated osteogenic and angiogenic capabilities. Furthermore, drug release from the PSGC membrane activated the Wnt/ß-catenin signaling pathway and promoted osteogenic differentiation and vascularization. Evaluation of the membrane's vascularization and osteogenic capacities involved transplantation onto a rat's subcutaneous area and assessing rat cranium defects for bone regeneration, respectively. Microcomputed tomography, histological tests, immunohistochemistry, and immunofluorescence staining confirmed the membrane's outstanding angiogenic capacity two weeks post-operation, with a higher incidence of osteogenesis observed in rat cranial defects eight weeks post-surgery. Conclusion: Overall, the SAL- and CT-loaded coaxial electrospun nanofiber membrane synergistically enhances bone repair and regeneration.


Asunto(s)
Gelatina , Glucósidos , Células Endoteliales de la Vena Umbilical Humana , Células Madre Mesenquimatosas , Nanofibras , Neovascularización Fisiológica , Osteogénesis , Fenantrenos , Fenoles , Poliésteres , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Animales , Nanofibras/química , Gelatina/química , Poliésteres/química , Glucósidos/química , Glucósidos/farmacología , Fenoles/química , Fenoles/farmacología , Fenantrenos/química , Fenantrenos/farmacología , Fenantrenos/farmacocinética , Fenantrenos/administración & dosificación , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Ratas , Masculino , Regeneración Ósea/efectos de los fármacos , Membranas Artificiales , Técnicas de Cocultivo , Liberación de Fármacos , Diferenciación Celular/efectos de los fármacos
2.
Drug Deliv ; 31(1): 2354687, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38823413

RESUMEN

Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-associated death worldwide. Beside early detection, early diagnosis, and early surgery, it is urgent to try new strategies for the treatment of HCC. Triptolide (TPL) has been employed to treat HCC. However, its clinical applications were restricted by the narrow therapeutic window, severe toxicity, and poor water-solubility. In this study, we developed cancer cell membrane-camouflaged biomimetic PLGA nanoparticles loading TPL (TPL@mPLGA) with the homologous targeting property for the treatment of HCC. The TPL@mPLGA was successfully prepared with particle size of 195.5 ± 7.5 nm and zeta potential at -21.5 ± 0.2 mV with good stability. The drug loading (DL) of TPL@mPLGA was 2.94%. After Huh-7 cell membrane coating, the natural Huh-7 cell membrane proteins were found to be retained on TPL@mPLGA, thus endowing the TPL@mPLGA with enhanced accumulation at tumor site, and better anti-tumor activity in vitro and in vivo when compared with TPL or TPL@PLGA. The TPL@mPLGA showed enhanced anti-tumor effects and reduced toxicity of TPL, which could be adopted for the treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular , Diterpenos , Compuestos Epoxi , Neoplasias Hepáticas , Nanopartículas , Fenantrenos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Diterpenos/administración & dosificación , Diterpenos/farmacología , Diterpenos/química , Diterpenos/farmacocinética , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Compuestos Epoxi/química , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/farmacología , Fenantrenos/administración & dosificación , Fenantrenos/farmacología , Fenantrenos/química , Fenantrenos/farmacocinética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Humanos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Nanopartículas/química , Animales , Línea Celular Tumoral , Ratones , Membrana Celular/efectos de los fármacos , Tamaño de la Partícula , Portadores de Fármacos/química , Ratones Desnudos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Ratones Endogámicos BALB C
3.
Eur J Pharm Biopharm ; 201: 114389, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38945407

RESUMEN

Liposomes represent one of the most extensively studied nano-carriers due to their potential in targeted drug delivery. However, the complex in vivo fate, particularly under pathological conditions, presents challenges for clinical translation of liposomal therapeutics. Liver serves as the most important organ for liposome accumulation and metabolism. Unfortunately, the fate of liposomes under pathological liver conditions has been significantly overlooked. This study aimed to investigate the in vivo pharmacokinetic profile and biodistribution profile of liposomes under drug-induced liver injury (DILI) conditions. Two classic DILI animal models, i.e. acetaminophen-induced acute liver injury (AILI) and triptolide-induced subacute liver injury (TILI), were established to observe the effect of pathological liver conditions on the in vivo performance of liposomes. The study revealed significant changes in the in vivo fate of liposomes following DILI, including prolonged blood circulation and enhanced hepatic accumulation of liposomes. Changes in the composition of plasma proteins and mononuclear phagocyte system (MPS)-related cell subpopulations collectively led to the altered in vivo fate of liposomes under liver injury conditions. Despite liver injury, macrophages remained the primary cells responsible for liposomes uptake in liver, with the recruited monocyte-derived macrophages exhibiting enhanced ability to phagocytose liposomes under pathological conditions. These findings indicated that high capture of liposomes by the recruited hepatic macrophages not only offered potential solutions for targeted delivery, but also warned the clinical application of patients under pathological liver conditions.


Asunto(s)
Acetaminofén , Enfermedad Hepática Inducida por Sustancias y Drogas , Diterpenos , Liposomas , Hígado , Fenantrenos , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Acetaminofén/farmacocinética , Ratones , Masculino , Hígado/metabolismo , Hígado/efectos de los fármacos , Distribución Tisular , Fenantrenos/farmacocinética , Fenantrenos/administración & dosificación , Fenantrenos/toxicidad , Diterpenos/farmacocinética , Diterpenos/administración & dosificación , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/administración & dosificación , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Ratones Endogámicos C57BL
4.
Int J Pharm ; 656: 124096, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38583821

RESUMEN

Pulmonary fibrosis (PF) is a chronic, progressive and irreversible interstitial lung disease that seriously threatens human life and health. Our previous study demonstrated the unique superiority of traditional Chinese medicine cryptotanshinone (CTS) combined with sustained pulmonary drug delivery for treating PF. In this study, we aimed to enhance the selectivity, targeting efficiency and sustained-release capability based on this delivery system. To this end, we developed and evaluated CTS-loaded modified liposomes-chitosan (CS) microspheres SM(CT-lipo) and liposome-exosome hybrid bionic vesicles-CS microspheres SM(LE). The prepared nano-in-micro particles system integrates the advantages of the carriers and complements each other. SM(CT-lipo) and SM(LE) achieved lung myofibroblast-specific targeting through CREKA peptide binding specifically to fibronectin (FN) and the homing effect of exosomes on parent cells, respectively, facilitating efficient delivery of anti-fibrosis drugs to lung lesions. Furthermore, compared with daily administration of conventional microspheres SM(NC) and positive control drug pirfenidone (PFD), inhaled administration of SM(CT-lipo) and SM(LE) every two days still attained similar efficacy, exhibiting excellent sustained drug release ability. In summary, our findings suggest that the developed SM(CT-lipo) and SM(LE) delivery strategies could achieve more accurate, efficient and safe therapy, providing novel insights into the treatment of chronic PF.


Asunto(s)
Quitosano , Exosomas , Fibronectinas , Liposomas , Fibrosis Pulmonar , Animales , Humanos , Masculino , Administración por Inhalación , Antifibróticos/administración & dosificación , Antifibróticos/química , Quitosano/química , Quitosano/administración & dosificación , Preparaciones de Acción Retardada , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Exosomas/química , Fibronectinas/administración & dosificación , Liposomas/química , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Microesferas , Fenantrenos/administración & dosificación , Fenantrenos/química , Fenantrenos/farmacocinética , Fibrosis Pulmonar/tratamiento farmacológico , Piridonas , Ratas Sprague-Dawley , Ratas
5.
Mol Pharm ; 21(5): 2473-2483, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38579335

RESUMEN

In recent years, the drainage of fluids, immune cells, antigens, fluorescent tracers, and other solutes from the brain has been demonstrated to occur along lymphatic outflow pathways to the deep cervical lymph nodes in the neck. To the best of our knowledge, no studies have evaluated the lymphatic transport of therapeutics from the brain. The objective of this study was to determine the lymphatic transport of model therapeutics of different molecular weights and lipophilicity from the brain using cervical lymph cannulation and ligation models in rats. To do this, anesthetized Sprague-Dawley rats were cannulated at the carotid artery and cannulated, ligated, or left intact at the cervical lymph duct. Rats were administered 14C-ibuprofen (206.29 g/mol, logP 3.84), 3H-halofantrine HCl (536.89 g/mol, logP 8.06), or 3H-albumin (∼65,000 g/mol) via direct injection into the brain striatum at a rate of 0.5 µL/min over 16 min. Plasma or cervical lymph samples were collected for up to 6-8 h following dosing, and brain and lymph nodes were collected at 6 or 8 h. Samples were subsequently analyzed for radioactivity levels via scintillation counting. For 14C-ibuprofen, plasma concentrations over time (plasma AUC0-6h) were >2 fold higher in lymph-ligated rats than in lymph-intact rats, suggesting that ibuprofen is cleared from the brain primarily via nonlymphatic routes (e.g., across the blood-brain barrier) but that this clearance is influenced by changes in lymphatic flow. For 3H-halofantrine, >73% of the dose was retained at the brain dosing site in lymph-intact and lymph-ligated groups, and plasma AUC0-8h values were low in both groups (<0.3% dose.h/mL), consistent with the high retention in the brain. It was therefore not possible to determine whether halofantrine undergoes lymphatic transport from the brain within the duration of the study. For 3H-albumin, plasma AUC0-8h values were not significantly different between lymph-intact, lymph-ligated, and lymph-cannulated rats. However, >4% of the dose was recovered in cervical lymph over 8 h. Lymph/plasma concentration ratios of 3H-albumin were also very high (up to 53:1). Together, these results indicate that 3H-albumin is transported from the brain not only via lymphatic routes but also via the blood. Similar to other tissues, the lymphatics may thus play a significant role in the transport of macromolecules, including therapeutic proteins, from the brain but are unlikely to be a major transport pathway from the brain for small molecule drugs that are not lipophilic. Our rat cervical lymph cannulation model can be used to quantify the lymphatic drainage of different molecules and factors from the brain.


Asunto(s)
Encéfalo , Ibuprofeno , Ganglios Linfáticos , Ratas Sprague-Dawley , Animales , Ratas , Encéfalo/metabolismo , Masculino , Ganglios Linfáticos/metabolismo , Ibuprofeno/farmacocinética , Ibuprofeno/administración & dosificación , Ibuprofeno/química , Fenantrenos/farmacocinética , Fenantrenos/química , Fenantrenos/administración & dosificación , Transporte Biológico/fisiología , Albúminas/farmacocinética , Albúminas/metabolismo
6.
Eur J Med Chem ; 243: 114694, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36115206

RESUMEN

In this work, a series of water-soluble triptolide prodrugs were synthesized, and their triptolide release rate, pharmacokinetic characteristics and anti-tumor effect were measured. We found that inserting glycolic acid as a linker between triptolide and the cyclic amino acid accelerated the release of triptolide from prodrugs into the plasma while preserving its safety. Among them, prodrug TP-P1 was significantly better than Minnelide (the only water-soluble triptolide prodrug in clinical trials) in terms of release rate in plasma and synthetic yield. In mouse models of human acute myeloid leukemia (AML), TP-P1 was effective in reducing xenograft tumors at dose levels as low as 25 µg/kg, and eliminating tumors at dose 100 µg/kg. Furthermore, TP-P1 could significantly enhance the efficacy of FLT3 inhibitors in the treatment of AML. These experimental results showed the potential of TP-P1 as water-soluble prodrugs of triptolide.


Asunto(s)
Diterpenos , Leucemia Mieloide Aguda , Fenantrenos , Profármacos , Ratones , Animales , Humanos , Profármacos/uso terapéutico , Agua , Fenantrenos/uso terapéutico , Fenantrenos/farmacocinética , Diterpenos/uso terapéutico , Diterpenos/farmacocinética , Compuestos Epoxi/uso terapéutico , Compuestos Epoxi/farmacocinética , Leucemia Mieloide Aguda/tratamiento farmacológico
7.
J Nanobiotechnology ; 19(1): 409, 2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34876139

RESUMEN

BACKGROUND: Attenuating inflammatory response and relieving pain are two therapeutic therapeutical goals for rheumatoid arthritis (RA). Anti-inflammatory and analgesic drugs are often associated with many adverse effects due to nonspecific distribution. New drug delivery systems with practical targeting ability and other complementary strategies urgently need to be explored. To achieve this goal, an acupoint drug delivery system that can target deliver anti-inflammatory drugs and simulate acupuncture in relieving pain was constructed, which can co-deliver triptolide (TP) and 2-chloro-N (6)-cyclopentyl adenosine (CCPA). RESULTS: We have successfully demonstrated that acupoint nanocomposite hydrogel composed of TP-Human serum album nanoparticles (TP@HSA NPs) and CCPA could effectively treat RA. The result shows that CCPA-Gel can enhance analgesic effects specifically at the acupoint, while the mechanical and thermal pain threshold was 4.9 and 1.6 times compared with non-acupoint, respectively, and the nanocomposite gel further enhanced. Otherwise, the combination of acupoint and nanocomposite hydrogel exerted synergetic improvement of inflammation, bone erosion, and reduction of systemic toxicity. Furthermore, it could regulate inflammatory factors and restore the balance of Th17/Treg cells, which provided a novel and effective treatment strategy for RA. Interestingly, acupoint administration could improve the accumulation of the designed nanomedicine in arthritic paws (13.5% higher than those in non-acupoint at 48 h), which may explain the better therapeutic efficiency and low toxicity. CONCLUSION: This novel therapeutic approach-acupoint nanocomposite hydrogel, builds a bridge between acupuncture and drugs which sheds light on the combination of traditional and modern medicine.


Asunto(s)
Puntos de Acupuntura , Antiinflamatorios , Artritis Reumatoide/metabolismo , Diterpenos , Nanogeles , Fenantrenos , Terapia por Acupuntura , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Conducta Animal/efectos de los fármacos , Preparaciones de Acción Retardada , Diterpenos/química , Diterpenos/farmacocinética , Diterpenos/farmacología , Sistemas de Liberación de Medicamentos , Compuestos Epoxi/química , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/farmacología , Humanos , Masculino , Nanomedicina , Fenantrenos/química , Fenantrenos/farmacocinética , Fenantrenos/farmacología , Ratas , Ratas Sprague-Dawley
8.
J Nanobiotechnology ; 19(1): 360, 2021 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-34749742

RESUMEN

In addition to early detection, early diagnosis, and early surgery, it is of great significance to use new strategies for the treatment of hepatocellular carcinoma (HCC). Studies showed that the combination of sorafenib (SFN) and triptolide (TPL) could reduce the clinical dose of SFN and maintain good anti-HCC effect. But the solubility of SFN and TPL in water is low and both drugs have certain toxicity. Therefore, we constructed a biomimetic nanosystem based on cancer cell-platelet (PLT) hybrid membrane camouflage to co-deliver SFN and TPL taking advantage of PLT membrane with long circulation functions and tumor cell membrane with homologous targeting. The biomimetic nanosystem, SFN and TPL loaded cancer cell-PLT hybrid membrane-camouflaged liquid crystalline lipid nanoparticles ((SFN + TPL)@CPLCNPs), could simultaneously load SFN and TPL at the molar ratio of SFN to TPL close to 10:1. (SFN + TPL)@CPLCNPs achieved long circulation function and tumor targeting at the same time, promoting tumor cell apoptosis, inhibiting tumor growth, and achieving a better "synergy and attenuation effect", which provided new ideas for the treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Diterpenos , Liposomas , Neoplasias Hepáticas/metabolismo , Nanopartículas , Fenantrenos , Sorafenib , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Materiales Biomiméticos/química , Plaquetas/química , Línea Celular Tumoral , Membrana Celular/química , Diterpenos/química , Diterpenos/farmacocinética , Diterpenos/farmacología , Compuestos Epoxi/química , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/farmacología , Humanos , Liposomas/química , Liposomas/farmacocinética , Liposomas/toxicidad , Masculino , Ratones , Ratones Endogámicos BALB C , Nanomedicina , Nanopartículas/química , Nanopartículas/toxicidad , Fenantrenos/química , Fenantrenos/farmacocinética , Fenantrenos/farmacología , Células RAW 264.7 , Sorafenib/química , Sorafenib/farmacocinética , Sorafenib/farmacología
9.
J Nanobiotechnology ; 19(1): 381, 2021 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-34802453

RESUMEN

BACKGROUND: The prognosis of patients with advanced gastric cancer (GC) remains unsatisfactory owing to distant metastasis and resistance to concurrent systemic therapy. Cancer-associated fibroblasts (CAFs), as essential participators in the tumor microenvironment (TME), play a vital role in tumor progression. Thus, CAFs-targeting therapy is appealing for remodeling TME and sensitizing GC to conventional systemic therapy. METHODS: Amphiphilic SN38 prodrug polymeric micelles (PSN38) and encapsulated the hydrophobic esterase-responsive prodrug of Triptolide (TPL), triptolide-naphthalene sulfonamide (TPL-nsa), were synthesized to form PSN38@TPL-nsa nanoparticles. Then, CAFs were isolated from fresh GC tissues and immortalized. TPL at low dose concentration was used to investigate its effect on CAFs and CAFs-induced GC cells proliferation and migration. The synergistic mechanism and antitumor efficiency of SN38 and TPL co-delivery nanoparticle were investigated both in vitro and in vivo. RESULTS: Fibroblast activation protein (FAP), a marker of CAFs, was highly expressed in GC tissues and indicated poorer prognosis. TPL significantly reduced CAFs activity and inhibited CAFs-induced proliferation, migration and chemotherapy resistance of GC cells. In addition, TPL sensitized GC cells to SN38 treatment through attenuated NF-κB activation in both CAFs and GC cells. PSN38@TPL-nsa treatment reduced the expression of collagen, FAP, and α-smooth muscle actin (α-SMA) in tumors. Potent inhibition of primary tumor growth and vigorous anti-metastasis effect were observed after systemic administration of PSN38@TPL-nsa to CAFs-rich peritoneal disseminated tumor and patient-derived xenograft (PDX) model of GC. CONCLUSION: TPL suppressed CAFs activity and CAFs-induced cell proliferation, migration and chemotherapy resistance to SN38 of GC. CAFs-targeted TPL and SN38 co-delivery nanoparticles exhibited potent efficacy of antitumor and reshaping TME, which was a promising strategy to treat advanced GC.


Asunto(s)
Antineoplásicos , Micelas , Profármacos , Neoplasias Gástricas/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diterpenos/química , Diterpenos/farmacocinética , Diterpenos/farmacología , Sinergismo Farmacológico , Compuestos Epoxi/química , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Fenantrenos/química , Fenantrenos/farmacocinética , Fenantrenos/farmacología , Profármacos/química , Profármacos/farmacocinética , Profármacos/farmacología , Microambiente Tumoral/efectos de los fármacos
10.
Biomed Res Int ; 2021: 2310422, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34692827

RESUMEN

Isopimaric acid (IPA) exhibits a diverse array of pharmacological activities, having been shown to function as an antihypertensive, antitumor, antibacterial, and hypocholesterolemic agent. However, few studies of the pharmacokinetics of IPA have been performed to date, and such analyses are essential to explore the in vivo mechanisms governing the biological activity of this compound. As such, we herein designed a selective LC-MS approach capable of quantifying serum IPA levels in model rats using an Agilent HC-C18 column (250 mm × 4.6 mm, 5 µm) via isocratic elution with a mobile phase composed of methanol 0.5% formic acid (91 : 9, v/v) at a 1 mL/min flow rate. Ion monitoring at m/z 301.2 [M-H]- was used to quantify IPA levels in plasma samples from these rats, while internal standard (IS) levels were assessed at m/z 455.3 [M-H]-. After validation, this approach was employed to conduct a pharmacokinetic analysis of rats administered IPA via the oral (p.o. 50, 100, or 200 mg/kg) and intravenous (i.v. 5 mg/kg) routes. Analyses of noncompartmental pharmacokinetic parameters revealed that IPA underwent secondary absorption following oral administration to these animals, with the two tested oral doses (50 and 100 mg/kg) being associated with respective absolute bioavailability values of 11.9% and 17.5%. In summary, this study may provide a foundation for future efforts to explore the mechanistic basis for the pharmacological activity of IPA, offering insights to guide its subsequent clinical utilization.


Asunto(s)
Ácidos Carboxílicos/sangre , Cromatografía Líquida de Alta Presión/métodos , Fenantrenos/sangre , Espectrometría de Masas en Tándem/métodos , Administración Intravenosa , Administración Oral , Animales , Disponibilidad Biológica , Ácidos Carboxílicos/administración & dosificación , Ácidos Carboxílicos/farmacocinética , Ionóforos/administración & dosificación , Ionóforos/sangre , Ionóforos/farmacocinética , Masculino , Fenantrenos/administración & dosificación , Fenantrenos/farmacocinética , Ratas , Ratas Sprague-Dawley , Distribución Tisular
11.
Drug Deliv ; 28(1): 2127-2136, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34617835

RESUMEN

The aim of this study was to develop and evaluate a triptolide phospholipid complex (TPCX) for the treatment of rheumatoid arthritis (RA) by transdermal delivery. TPCX was prepared and characterized by differential scanning calorimetry (DSC), Fourier-transform infrared spectroscopy (FTIR) analysis, transmission electron microscope (TEM), and scanning electron microscope (SEM). The solubility of TPCX was determined. Then, a TPCX cream was prepared to evaluate its percutaneous permeability and the antiarthritis effect. The transdermal permeability was determined using the Franz method, and a microdialysis system was used for skin pharmacokinetic study. A rat model of RA was prepared to evaluate the pharmacological effects. TPCX increased the solubility of triptolide in water, and the percutaneous permeability of TPCX cream was greatly enhanced compared with triptolide cream. The skin pharmacokinetic study indicated that TPCX cream has a longer biological half-life (t1/2) and mean residence time (MRT), but it has a shorter Tmax than that of triptolide cream in vivo. The area under the curve (AUC0-t)/AUC0-∞) and the peak concentration (Cmax) of TPCX cream were obviously higher than those of triptolide cream. The TPCX-loaded cream alleviated paw swelling and slowed down the progression of arthritis by inhibiting the inflammatory response by down regulating the TNF-α, IL-1ß, and IL-6 levels, thus exhibiting excellent antiarthritic effects. In summary, the prepared TPCX effectively increases the hydrophilicity of triptolide, which is good for its percutaneous absorption and enhances its effect on RA rats. TPCX can be a good candidate for the transdermal delivery to treat RA.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Diterpenos/farmacología , Inmunosupresores/farmacología , Fenantrenos/farmacología , Fosfolípidos/química , Administración Cutánea , Animales , Área Bajo la Curva , Química Farmacéutica , Diterpenos/administración & dosificación , Diterpenos/farmacocinética , Relación Dosis-Respuesta a Droga , Liberación de Fármacos , Estabilidad de Medicamentos , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/farmacología , Semivida , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacocinética , Mediadores de Inflamación/metabolismo , Masculino , Fenantrenos/administración & dosificación , Fenantrenos/farmacocinética , Ratas , Ratas Wistar
12.
J Hepatol ; 75(1): 98-107, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33894327

RESUMEN

BACKGROUND & AIMS: Golexanolone is a novel small molecule GABA-A receptor-modulating steroid antagonist under development for the treatment of cognitive and vigilance disorders caused by allosteric over-activation of GABA-A receptors by neurosteroids. It restored spatial learning and motor coordination in animal models of hepatic encephalopathy (HE) and mitigated the effects of intravenous allopregnanolone in healthy adults in a dose-dependent fashion. Herein, we report data on the safety, pharmacokinetics (PK) and efficacy of golexanolone in adult patients with cirrhosis. METHODS: Following single/multiple ascending dose studies, adults with Child-Pugh A/B cirrhosis and abnormal continuous reaction time (CRT) on screening were randomized to 3 weeks' dosing with golexanolone (10, 40 or 80 mg BID) or placebo. CRT, psychometric hepatic encephalopathy score (PHES), animal naming test (ANT), Epworth sleepiness scale (ESS) and electroencephalogram (mean dominant frequency [MDF]; delta+theta/alpha+beta ratio [DT/AB]) were obtained at baseline, 10, and 21 days. RESULTS: Golexanolone exhibited satisfactory safety and PK. Baseline characteristics were similar between the 12 and 33 patients randomized to placebo or golexanolone, respectively. By prespecified analyses, golexanolone was associated with directionally favourable changes vs. placebo in ESS (p = 0.047), MDF (p = 0.142) and DT/AB (p = 0.021). All patients also showed directionally favourable changes in CRT, PHES and ANT, but with no statistical difference between golexanolone and placebo. Post hoc analyses taking into account the variability and improvement in CRT, PHES and ANT observed between screening and baseline suggested an efficacy signal by cognitive measures as well. CONCLUSION: Golexanolone was well tolerated and associated with improvement in cognitive performance. These results implicate GABA-A receptor-modulating neurosteroids in the pathogenesis of HE and support the therapeutic potential of golexanolone. LAY SUMMARY: Many patients with cirrhosis experience subtle but disabling cognitive problems, including sleepiness and poor attention span, that impair their ability to be gainfully employed or carry out activities of daily living. This pilot study tested the hypothesis that these problems with cognition, for which there is no approved treatment, might be improved by an experimental drug, golexanolone, designed to normalize the function of receptors which inhibit brain function. The results of this study suggest that golexanolone is well tolerated and may improve cognition, as reflected by measures of sleepiness, attention span and brain wave activity, paving the way for future larger studies of this promising experimental drug. CLINICAL TRIAL REGISTRATION NUMBER: EudraCT 2016-003651-30.


Asunto(s)
Cognición/efectos de los fármacos , Antagonistas de Receptores de GABA-A , Encefalopatía Hepática , Fenantrenos , Actividades Cotidianas , Nivel de Alerta/efectos de los fármacos , Atención/efectos de los fármacos , Método Doble Ciego , Drogas en Investigación , Electroencefalografía/métodos , Femenino , Antagonistas de Receptores de GABA-A/administración & dosificación , Antagonistas de Receptores de GABA-A/efectos adversos , Antagonistas de Receptores de GABA-A/farmacocinética , Encefalopatía Hepática/diagnóstico , Encefalopatía Hepática/tratamiento farmacológico , Encefalopatía Hepática/etiología , Encefalopatía Hepática/metabolismo , Humanos , Cirrosis Hepática/complicaciones , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Neuroesteroides/administración & dosificación , Neuroesteroides/efectos adversos , Neuroesteroides/farmacocinética , Fenantrenos/administración & dosificación , Fenantrenos/efectos adversos , Fenantrenos/farmacocinética , Proyectos Piloto , Somnolencia/efectos de los fármacos , Resultado del Tratamiento
13.
Biomed Res Int ; 2021: 8825640, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708996

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disease, which is associated with extracellular deposition of amyloid-ß proteins (Aß). It has been reported that triptolide (TP), an immunosuppressive and anti-inflammatory agent extracted from a Chinese herb Tripterygium wilfordii, shows potential neuroprotective effects pertinent to AD. However, the clinical use of TP for AD could be hampered due to its high toxicity, instability, poor water solubility, and nonspecific biodistribution after administration. In this paper, we reported a kind of multiple-coated PLGA nanoparticle with the entrapment of TP and surface coated by chitosan hydrochloride, Tween-80, PEG20000, and borneol/mentholum eutectic mixture (MC-PLGA-TP-NP) as a novel nasal brain targeting preparation for the first time. The obtained MC-PLGA-TP-NP was 147.5 ± 20.7 nm with PDI of 0.263 ± 0.075, zeta potential of 14.62 ± 2.47 mV, and the entrapment efficiency and loading efficiency of 93.14% ± 4.75% and 1.17 ± 0.08%, respectively. In comparison of TP, MC-PLGA-TP-NP showed sustained-release profile and better transcellular permeability to Caco-2 cells in vitro. In addition, our data showed that MC-PLGA-TP-NP remarkably reduced the cytotoxicity, attenuated the oxidative stress, and inhibited the increase of the intracellular Ca2+ influx in differentiated PC12 cells induced by Aß 1-42. Therefore, it can be concluded that MC-PLGA-TP-NP is a promising preparation of TP, which exerts a better neuroprotective activity in the AD cellular model.


Asunto(s)
Enfermedad de Alzheimer , Materiales Biocompatibles Revestidos , Diterpenos , Portadores de Fármacos , Modelos Neurológicos , Nanopartículas , Fenantrenos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Células CACO-2 , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacocinética , Materiales Biocompatibles Revestidos/farmacología , Diterpenos/química , Diterpenos/farmacocinética , Diterpenos/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Compuestos Epoxi/química , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/farmacología , Humanos , Nanopartículas/química , Nanopartículas/uso terapéutico , Células PC12 , Fenantrenos/química , Fenantrenos/farmacocinética , Fenantrenos/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacocinética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Ratas
14.
Toxicol Lett ; 342: 85-94, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33600922

RESUMEN

Triptolide (TP), an active component of Tripterygium wilfordii Hook. F, has been widely used in China for treating autoimmune and inflammatory diseases, and has also been validated by modern science and developed as a candidate anti-cancer treatment. However, liver toxicity of TP has seriously hindered its use and development, the clinical features and primary toxicological mechanism have been unclear. Considering the major target regulation mechanism of TP is the suppression of global transcription regulated by RNAPII, which is closed related with the detoxification of drugs. This paper tries to verify the synergistic liver injury and its mechanism of TP when co-administered with CYP3A4 substrate drug. The experiments showed that TP dose-dependently blocked transcriptional activation of CYP3A4 in both hPXR and hPXR-CYP3A4 reporter cell lines, lowered the mRNA and protein expression of PXR target genes such as CYP3A1, CYP2B1, and MDR1, and inhibited the functional activity of CYP3A in a time- and concentration-dependent manner in sandwich-cultured rat hepatocytes (SCRH) and female Sprague-Dawley (f-SD) rats. Furthermore, TP combined with atorvastatin (ATR), the substrate of CYP3A4, synergistically enhanced hepatotoxicity in cultured HepG2 and SCRH cells (CI is 0.38 and 0.29, respectively), as well as in f-SD rats, with higher exposure levels of both drugs. These results clearly indicate that TP inhibits PXR-mediated transcriptional activation of CYP3A4, leading to a blockade on the detoxification of itself and ATR, thereby greatly promoting liver injury. This study may implies the key cause of TP related liver injury and provides experimental data for the rational use of TP in a clinical scenario.


Asunto(s)
Atorvastatina/toxicidad , Citocromo P-450 CYP3A/metabolismo , Diterpenos/toxicidad , Hepatocitos/efectos de los fármacos , Fenantrenos/toxicidad , Receptor X de Pregnano/antagonistas & inhibidores , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacocinética , Antiinflamatorios no Esteroideos/toxicidad , Atorvastatina/administración & dosificación , Atorvastatina/farmacocinética , Citocromo P-450 CYP3A/genética , Diterpenos/administración & dosificación , Diterpenos/farmacocinética , Sinergismo Farmacológico , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/toxicidad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Fenantrenos/administración & dosificación , Fenantrenos/farmacocinética , Ratas , Ratas Sprague-Dawley
15.
Biomed Chromatogr ; 35(7): e5093, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33634891

RESUMEN

A rapid and specific UPLC-MS/MS method with a total run time of 3.5 min was developed for the determination of pravastatin, fexofenadine, rosuvastatin, and methotrexate in rat primary hepatocytes. After protein precipitation with 70% acetonitrile (containing 30% H2 O), these four analytes were separated under gradient conditions with a mobile phase consisting of 0.03% acetic acid (v/v) and methanol at a flow rate of 0.50 mL/min. The linearity, recovery, matrix effect, accuracy, precision, and stability of the method were well validated. We evaluated drug-drug interactions based on these four compounds in freshly suspended hepatocytes. The hepatic uptake of pravastatin, fexofenadine, rosuvastatin, and methotrexate at 4°C was significantly lower than that at 37°C, and the hepatocytes were saturable with increased substrate concentration and culture time, suggesting that the rat primary hepatocyte model was successfully established. Triptolide showed a significant inhibitory effect on the hepatic uptake of these four compounds. In conclusion, this method was successfully employed for the quantification of pravastatin, fexofenadine, rosuvastatin, and methotrexate and was used to verify the rat primary hepatocyte model for Oatp1, Oatp2, Oatp4, and Oat2 transporter studies. Then, we applied this model to explore the effect of triptolide on these four transporters.


Asunto(s)
Hepatocitos/metabolismo , Metotrexato , Pravastatina , Rosuvastatina Cálcica , Terfenadina/análogos & derivados , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión/métodos , Diterpenos/análisis , Diterpenos/farmacocinética , Interacciones Farmacológicas , Compuestos Epoxi/análisis , Compuestos Epoxi/farmacocinética , Modelos Lineales , Masculino , Metotrexato/análisis , Metotrexato/farmacocinética , Fenantrenos/análisis , Fenantrenos/farmacocinética , Pravastatina/análisis , Pravastatina/farmacocinética , Ratas Wistar , Reproducibilidad de los Resultados , Rosuvastatina Cálcica/análisis , Rosuvastatina Cálcica/farmacocinética , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem/métodos , Terfenadina/análisis , Terfenadina/farmacocinética
16.
Biomed Pharmacother ; 137: 111332, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33548911

RESUMEN

Cryptotanshinone (IUPAC name: (R)-1,2,6,7,8,9-hexahydro-1,6,6-trimethyl-phenanthro(1,2-b)furan-10,11-dione), a biologically active constituent extracted from the roots and rhizomes of the plant Salvia miltiorrhiza, has been studied in depth as a medicinally active compound and shown to have efficacy in the treatment of numerous diseases and disorders. In this review, we describe in detail the current status of cryptotanshinone research, including findings relating to the structure, pharmacokinetics, pharmacological activity, and derivatives of this compound. Cryptotanshinoneh as a diverse range of pharmacological effects, including anti-cancer, anti-inflammatory, immune regulatory, neuroprotective, and anti-fibrosis activities. Studies on the molecular mechanisms underlying the activities of cryptotanshinone have established that the JAK2/STAT3, PI3K/AKT, NF-κB, AMPK, and cell cycle pathways are involved in the inhibitory and pro-apoptotic effects of cryptotanshinone on different tumor cell lines, these molecular pathways interact in a coordinated manner to inhibit cell proliferation, migration and invasion,and induce transformation, autophagy, necrosis, and cellular immunity. The anti-inflammatory mechanisms of cryptotanshinone have been found to be associated with the TLR4-MyD88/PI3K/Nrf2 and TLR4-MyD88/NF-κB/MAPK pathways, whereasthe Hedgehog, NF-κB, and Nrf-2/HO-1 pathways are regulated by cryptotanshinone to reduce organ fibrosis, and its inhibitory effects on the PI3K/AKT-eNOS pathway have been linked to neuroprotective effects. Given the potential medicinal utility of cryptotanshinone, further research is needed to verify the efficacy and safety of this compound in clinical use, evaluate its pharmacological activity, and identify molecular targets.


Asunto(s)
Fenantrenos/farmacología , Salvia miltiorrhiza/química , Animales , Antiinflamatorios/farmacología , Antineoplásicos Fitogénicos/farmacología , Fibrosis/prevención & control , Humanos , Fármacos Neuroprotectores/farmacología , Fenantrenos/administración & dosificación , Fenantrenos/química , Fenantrenos/farmacocinética , Transducción de Señal/efectos de los fármacos
17.
Biomed Pharmacother ; 131: 110737, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32932044

RESUMEN

PURPOSE: To investigate whether triptolide-nanoliposome-APRPG (TP-nanolip-APRPG), a novel sustained-release nano-drug delivery system that targets vascular endothelial cells, could enhance the inhibition of triptolide (TP) on laser-induced choroidal neovascularization (CNV). METHODS: TP was encapsulated with or without APRPG (Ala-Pro-Arg-Pro-Gly) peptide-modified nanoliposomes. CNV was induced by laser photocoagulation in C57BL/6J mice. One microliter of 10 µg free TP monomer, TP-nanolip containing 10 µg TP, TP-nanolip-APRPG containing 10 µg TP, or an identical volume of PBS was intravitreally injected in mice immediately after laser photocoagulation. Seven days after laser photocoagulation, CNV volumes were calculated in each group. Infiltration of M2 macrophages as well as protein levels of vascular endothelial growth factor (VEGF) and inflammatory factors including ICAM-1 and MCP-1 in the RPE-choroid complex were determined. In vitro assays for cell proliferation, migration, and tube formation were also performed. RESULTS: TP-nanolip-APRPG was successfully synthesized and exhibited good TP delivery and enhanced the cellular uptake of TP in vitro. In vitro studies showed that TP-nanolip-APRPG was a better inhibitor of cell proliferation (31.34 ±â€¯3.89 % vs 41.25 ±â€¯4.67 % vs 53.55 ±â€¯5.76 %), migration (62.60 ±â€¯8.88 vs 104.60 ±â€¯13.32 vs 147.00 ±â€¯13.15), and tube formation (681.26 ±â€¯108.15 vs 926.75 ±â€¯54.01 vs 1189.84 ±â€¯157.14) than TP-nanolip or free TP (all P < 0.05). Intravitreal injections of free TP (77588.10±7719.28 µm3), TP-nanolip (64628.23 ±â€¯5857.96 µm3), and TP-nanolip-APRPG (50880.34 ±â€¯6606.56 µm3) inhibited the development of CNV compared with the PBS control group (120338.07 ±â€¯17428.90 µm3) (P < 0.01, n=6). TP-nanolip-APRPG and TP-nanolip significantly down-regulated the protein levels of VEGF (152.76±19.55 vs 182.24±19.98 vs 208.55±21.93 pg/mg total protein) and inflammatory factors including ICAM-1 (61.69±3.49 vs 72.04±3.49 vs 81.92±4.09 ng/mg total protein) and MCP-1 (40.14±3.50 vs 50.75±4.18 vs 60.27±5.23 pg/mg total protein) compared with the free TP monomer group (all P < 0.05, n=8), which paralleled the decreased infiltration of M2 macrophages in the CNV lesions. Moreover, no influence on retinal morphology and function was observed before or after treatment in each group (P > 0.05, n=6). CONCLUSIONS: TP-nanolip-APRPG, a novel sustained-release drug delivery system targeting endothelial cells of CNV lesions, could enhance TP inhibition of the development of CNV without toxicity in the retina, suggesting therapeutic potential for CNV-related diseases in future clinical practice.


Asunto(s)
Neovascularización Coroidal/prevención & control , Diterpenos/administración & dosificación , Células Endoteliales/efectos de los fármacos , Liposomas/química , Nanopartículas/química , Oligopéptidos/química , Fenantrenos/administración & dosificación , Animales , Movimiento Celular/efectos de los fármacos , Neovascularización Coroidal/etiología , Preparaciones de Acción Retardada , Diterpenos/química , Diterpenos/farmacocinética , Liberación de Fármacos , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/química , Compuestos Epoxi/farmacocinética , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenantrenos/química , Fenantrenos/farmacocinética , Retina/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/análisis
18.
Chemosphere ; 259: 127487, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32650165

RESUMEN

Polycyclic Aromatic Hydrocarbons (PAHs) have elicited increasing concern due to their ubiquitous occurrence in coastal marine environments and resultant toxicity in organisms. Due to their lipophilic nature, PAHs tend to accumulate in phytoplankton cells and thus subsequently transfer to other compartments of the marine ecosystem. The intrinsic fluorescence properties of PAHs in the ultraviolet (UV)/blue spectral range have recently been exploited to investigate their uptake modes, localization, and aggregation in various biological tissues. Here, we quantitatively evaluate the sorption of two model PAHs (phenanthrene and pyrene) in three marine phytoplankton species (Chaetoceros tenuissimus, Thalassiosira sp. and Proteomonas sp.) using a combined approach of UV excitation flow cytometry and fluorescence microscopy. Over a 48-h exposure to a gradient of PAHs, Thalassiosira sp. showed the highest proportion of PAH-sorbed cells (29% and 97% of total abundance for phenanthrene and pyrene, respectively), which may be attributed to its relatively high total lipid content (33.87 percent dry weight). Moreover, cell-specific pulse amplitude modulation (PAM) microscope fluorometry revealed that PAH sorption significantly reduced the photosynthetic quantum efficiency (Fv/Fm) of individual phytoplankton cells. We describe a rapid and precise hybrid method for the detection of sorption of PAHs on phytoplankton cells. Our results emphasize the ecologically relevant sub-lethal effects of PAHs in phytoplankton at the cellular level, even at concentrations where no growth inhibition was apparent. This work is the first study to address the cell-specific impacts of fluorescent toxicants in a more relevant toxicant-sorbed subpopulation; these cell-specific impacts have to date been unidentified in traditional population-based phytoplankton toxicity assays.


Asunto(s)
Fitoplancton/efectos de los fármacos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Análisis de la Célula Individual/métodos , Absorción Fisicoquímica , Ecosistema , Citometría de Flujo/métodos , Microscopía Fluorescente/métodos , Fenantrenos/química , Fenantrenos/farmacocinética , Fitoplancton/citología , Fitoplancton/metabolismo , Hidrocarburos Policíclicos Aromáticos/química , Hidrocarburos Policíclicos Aromáticos/farmacocinética , Pirenos/química , Pirenos/farmacocinética , Rayos Ultravioleta
19.
J Nanobiotechnology ; 18(1): 83, 2020 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-32473632

RESUMEN

BACKGROUND: Breast cancer lung metastasis occurs in more than 60% of all patients with breast cancer, and most of those afflicted by it eventually die of recurrence. The tumor microenvironment plays vital roles in metastasis. Modulating the tumor microenvironment via multiple pathways could efficiently prevent or inhibit lung metastasis. Silibinin and cryptotanshinone are natural plant products that demonstrate anti-metastasis effects and modulate the tumor microenvironment via different pathways. However, they have poor aqueous solubility, membrane permeability, and oral bioavailability. Oral drug administration may help improve the quality of life and compliance of patients with breast cancer, primarily under long-term and/or follow-up therapy. Herein, we developed poly-N-(2-hydroxypropyl) methacrylamide (pHPMA)-coated wheat germ agglutinin-modified lipid-polymer hybrid nanoparticles, co-loaded with silibinin and cryptotanshinone (S/C-pW-LPNs). We assessed their oral bioavailability, and evaluated their anti-metastasis efficacy in a 4T1 breast cancer tumor-bearing nude mouse model. RESULTS: An in vitro mucus diffusion study revealed that pHPMA enhanced W-LPN mucus penetration. After oral administration, pHPMA enhanced nanoparticle distribution in rat jejunum and substantially augmented oral bioavailability. S/C-W-LPNs markedly increased 4T1 cell toxicity and inhibited cell invasion and migration. Compared to LPNs loaded with either silibinin or cryptotanshinone alone, S/C-pW-LPNs dramatically slowed tumor progression in 4T1 tumor-bearing nude mice. S/C-pW-LPNs presented with the most robust anti-metastasis activity on smooth lung surfaces and mitigated lung metastasis foci. They also downregulated tumor microenvironment biomarkers such as CD31, TGF-ß1, and MMP-9 that promote metastasis. CONCLUSIONS: Silibinin- and cryptotanshinone-co-loaded pW-LPNs efficiently penetrate intestinal barriers, thereby enhancing the oral bioavailability of the drug loads. These nanoparticles exhibit favorable anti-metastasis effects in breast cancer-bearing nude mice. Hence, S/C-pW-LPNs are promising oral drug nanocarriers that inhibit breast cancer lung metastasis.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares , Nanopartículas , Fenantrenos , Silibina , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Disponibilidad Biológica , Neoplasias de la Mama/patología , Células CACO-2 , Movimiento Celular/efectos de los fármacos , Células HT29 , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Moco/química , Moco/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias Experimentales , Fenantrenos/química , Fenantrenos/farmacocinética , Fenantrenos/farmacología , Ratas Sprague-Dawley , Silibina/química , Silibina/farmacocinética , Silibina/farmacología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Pharm Pharmacol ; 72(12): 1854-1864, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32478421

RESUMEN

OBJECTIVES: We aimed to determine the diurnal rhythm of Tripterygium wilfordii (TW) hepatotoxicity and to investigate a potential role of metabolism and pharmacokinetics in generating chronotoxicity. METHODS: Hepatotoxicity was determined based on assessment of liver injury after dosing mice with TW at different circadian time points. Circadian clock control of metabolism, pharmacokinetics and hepatotoxicity was investigated using Clock-deficient (Clock-/- ) mice. KEY FINDINGS: Hepatotoxicity of TW displayed a significant circadian rhythm (the highest level of toxicity was observed at ZT2 and the lowest level at ZT14). Pharmacokinetic experiments showed that oral gavage of TW at ZT2 generated higher plasma concentrations (and systemic exposure) of triptolide (a toxic constituent) compared with ZT14 dosing. This was accompanied by reduced formation of triptolide metabolites at ZT2. Loss of Clock gene sensitized mice to TW-induced hepatotoxicity and abolished the time-dependency of toxicity that was well correlated with altered metabolism and pharmacokinetics of triptolide. Loss of Clock gene also decreased Cyp3a11 expression in mouse liver and blunted its diurnal rhythm. CONCLUSIONS: Tripterygium wilfordii chronotoxicity was associated with diurnal variations in triptolide pharmacokinetics and circadian expression of hepatic Cyp3a11 regulated by circadian clock. Our findings may have implications for improving TW treatment outcome with a chronotherapeutic approach.


Asunto(s)
Proteínas CLOCK/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Ritmo Circadiano/efectos de los fármacos , Diterpenos/toxicidad , Hígado/efectos de los fármacos , Fenantrenos/toxicidad , Extractos Vegetales/toxicidad , Tripterygium/toxicidad , Activación Metabólica , Animales , Proteínas CLOCK/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Diterpenos/aislamiento & purificación , Diterpenos/farmacocinética , Compuestos Epoxi/aislamiento & purificación , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/toxicidad , Hígado/metabolismo , Hígado/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenantrenos/aislamiento & purificación , Fenantrenos/farmacocinética , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacocinética , Toxicocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA