Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Plant Cell Rep ; 40(10): 1831-1844, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34230985

RESUMEN

KEY MESSAGE: MiR394 plays a negative role in tomato resistance to late blight. The lncRNA40787 severing as an eTM for miR394 to regulate LCR and exerting functions in tomato resistance. Tomato (Solanum lycopersicum), which was used as model species for studying the mechanism of plant disease defense, is susceptible to multiple pathogens. Non-coding RNA (ncRNA) has a pivotal role in plants response to biological stresses. It has previously been observed that the expression level of miR394 changed significantly after the infection of various pathogens. However, there has been no detailed investigation of the accumulated or suppressed mechanism of miR394. Our previous study predicted three lncRNAs (lncRNA40787, lncRNA27177, and lncRNA42566) that contain miR394 endogenous target mimics (eTM), which may exist as the competitive endogenous RNAs (ceRNAs) of miR394. In our study, the transcription levels of these three lncRNAs were strongly up-regulated in tomato upon infection with P. infestans. In contrast with the three lncRNAs, the accumulation of miR394 was significantly suppressed. Based on the expression pattern, and value of minimum free energy (mfes) that represents the binding ability between lncRNA and miRNA, lncRNA40787 was chosen for further investigation. Results showed that overexpression of lncRNA40787 reduced the expression of miR394 along with decreased lesion area and enhanced disease resistance. Overexpression of miR394, however, decreased the expression of its target gene Leaf Curling Responsiveness (LCR), and suppressed the synthesis components genes of jasmonic acid (JA), depressing the resistance of tomato to P. infestans infection. Taken together, our findings indicated that miR394 can be decoyed by lncRNA40787, and negatively regulated the expression of LCR to enhance tomato susceptibility under P. infestans infection. Our study provided detailed information on the lncRNA40787-miR394-LCR regulatory network and serves as a reference for future research.


Asunto(s)
MicroARNs/genética , Phytophthora infestans/patogenicidad , Enfermedades de las Plantas/genética , Solanum lycopersicum/genética , Solanum lycopersicum/microbiología , Ciclopentanos/metabolismo , Resistencia a la Enfermedad , Regulación de la Expresión Génica de las Plantas , Silenciador del Gen , Interacciones Huésped-Patógeno/genética , Solanum lycopersicum/metabolismo , Oxilipinas/metabolismo , Enfermedades de las Plantas/microbiología , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , ARN Largo no Codificante/genética , ARN de Planta/genética
2.
Zhongguo Zhong Yao Za Zhi ; 45(11): 2578-2585, 2020 Jun.
Artículo en Zh | MEDLINE | ID: mdl-32627492

RESUMEN

To explore whether paeonol can play an anti-atherosclerotic role by regulating the expression of aortic caveolin-1 and affecting NF-κB pathway, so as to inhibit the inflammatory response of vascular endothelium in atherosclerotic rats. The atherosclerotic model of rats was induced by high-fat diet and vitamin D_2. The primary culture of vascular endothelial cells(VECs) was carried out by tissue block pre-digestion and adherent method. The injury model of VECs was induced by lipopolysaccharide(LPS), and filipin, a small concave protein inhibitor, was added for control. HE staining was used to observe pathological changes of aorta. TNF-α, IL-6 and VCAM-1 were detected by ELISA. Western blot assay was used to detect the protein expression levels of caveolin-1 and p65 in aorta and VECs. The results showed that as compared with model group, paeonol significantly reduced aortic plaque area and lesion degree in rats, decreased the level of serum TNF-α, IL-6 and VCAM-1 in the rats and enhanced the relative expression level of caveolin-1, decreased p65 expression conversely(P<0.05 or P<0.01). In vitro, as compared to model group, paeonol obviously improved cell morphology, decreased the secretion of TNF-α, IL-6 and VCAM-1 in VECs, increased caveolin-1 expression, and decreased p65 protein expression(P<0.05 or P<0.01). Furthermore, filipin could reverse the effect of paeonol on expression of inflammatory factors and proteins(P<0.05 or P<0.01). According to the results, it was found that paeonol could play the role of anti-atherosclerosis by up-regulating the expression of caveolin-1 and inhibiting the activation of NF-κB pathway to reduce vascular inflammation in atherosclerotic rats.


Asunto(s)
Caveolina 1 , FN-kappa B , Acetofenonas , Animales , Células Endoteliales , Endotelio Vascular , Inflamación , Ratas , Transducción de Señal , Factor de Necrosis Tumoral alfa , Regulación hacia Arriba
3.
Phytother Res ; 32(4): 643-650, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29210118

RESUMEN

Alantolactone (ALA), a sesquiterpene lactone isolated from several medicinal plants such as Inula helenium, has been identified to have attractive anticancer activity. However, its role in the inhibition of angiogenesis during tumor development remains unclear. In this study, we found ALA can inhibit the proliferation, motility, migration, and tube formation of human umbilical vein endothelial cells. ALA also restrained angiogenesis in chick embryo chorioallantoic membrane and delayed the growth of human MDA-MB-231 breast cancer xenograft in mice through angiogenesis inhibition. Furthermore, ALA suppressed the phosphorylation of vascular endothelial growth factor receptor 2 and its downstream protein kinase including PLCγ1, FAK, Src, and Akt in endothelial cells. Taken together, the antiangiogenic activity of ALA and its molecular mechanism are identified for the first time, indicating that ALA may be a potential drug candidate or lead compound for antiangiogenic cancer therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Lactonas/química , Sesquiterpenos de Eudesmano/química , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Neoplasias de la Mama/patología , Embrión de Pollo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal
4.
Bioconjug Chem ; 26(8): 1702-12, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26076081

RESUMEN

Multidrug resistance is the main cause of clinical chemotherapeutic failure. Antiangiogenic cancer therapy with nanomedicine that allows the targeted delivery of antiangiogenic agents to tumor endothelial cells may contribute to innovative strategies for treating multidrug-resistant cancers. In this study, we developed a new nanodrug delivery system (nano-DDS), with improved antiangiogenic efficacy against multidrug resistant human breast cancer MCF-7/ADR cells. Here, the IF7 ligand was a peptide designed to bind the annexin 1 (Anxa 1), a highly specific marker of the tumor vasculature surface, with high affinity and specificity. IF7-conjugated Anxa 1-targeting nanoparticles containing paclitaxel (IF7-PTX-NP) allowed controlled drug release and displayed favorable prolonged circulation in vivo. IF7-PTX-NP was significantly internalized by human umbilical vein endothelial cells (HUVEC) through the IF7-Anxa 1 interaction, and this facilitated uptake enhanced the expected antiangiogenic activity of inhibiting HUVEC proliferation, migration, and tube formation in a Matrigel plug relative to those of Taxol and PTX-NP. As IF7-PTX-NP targeted the tumor vessels, more nanoparticles accumulated in MCF-7/ADR tumors, and more importantly, induced significant apoptosis of the tumor vascular endothelial cells and necrosis of the tumor tissues. Low dose paclitaxel (1 mg/kg) formulated in IF7-PTX-NP showed significant anticancer efficacy, delaying the growth of MCF-7/ADR tumors. The same efficacy was only obtained with an 8-fold dose of paclitaxel (8 mg/kg) as Taxol plus XR9576, a potent P-gp inhibitor. The anticancer efficacy of IF7-PTX-NP was strongly associated with the improved antiangiogenic effect, evident as a dramatic reduction in the tumor microvessel density and pronounced increase in apoptotic tumor cells, with no obvious toxicity to the mice. This nano-DDS, which targets the tumor neovasculature, offers a promising strategy for the treatment of multidrug-resistant cancer.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Anexina A1/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Neovascularización Patológica/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Animales , Anexina A1/metabolismo , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Cultivadas , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Paclitaxel/farmacología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Toxicol Appl Pharmacol ; 281(1): 118-24, 2014 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-25250884

RESUMEN

Platycodin D (PD) is an active component mainly isolated from the root of Platycodon grandiflorum. Recent studies proved that PD exhibited inhibitory effect on proliferation, migration, invasion and xenograft growth of diverse cancer cell lines. However, whether PD is suppressive for angiogenesis, an important hallmark in cancer development, remains unknown. Here, we found that PD could dose-dependently inhibit human umbilical vein endothelial cell (HUVEC) proliferation, motility, migration and tube formation. PD also significantly inhibited angiogenesis in the chick embryo chorioallantoic membrane (CAM). Moreover, the antiangiogenic activity of PD contributed to its in vivo anticancer potency shown in the decreased microvessel density and delayed growth of HCT-15 xenograft in mice with no overt toxicity. Western blot analysis indicated that PD inhibited the phosphorylation of VEGFR2 and its downstream protein kinase including PLCγ1, JAK2, FAK, Src, and Akt in endothelial cells. Molecular docking simulation showed that PD formed hydrogen bonds and hydrophobic interactions within the ATP binding pocket of VEGFR2 kinase domain. The present study firstly revealed the high antiangiogenic activity and the underlying molecular basis of PD, suggesting that PD may be a potential antiangiogenic agent for angiogenesis-related diseases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Triterpenos/farmacología , Carga Tumoral/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Campanulaceae , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Embrión de Pollo , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Membrana Corioalantoides/patología , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Saponinas/uso terapéutico , Transducción de Señal/fisiología , Triterpenos/uso terapéutico , Carga Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
Acta Pharmacol Sin ; 35(4): 483-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24562307

RESUMEN

AIM: Paeonol (2'-hydroxy-4'-methoxyacetophenone) from Cortex moutan root is a potential therapeutic agent for atherosclerosis. This study sought to investigate the mechanisms underlying anti-inflammatory effects of paeonol in rat vascular endothelial cells (VECs) in vitro. METHODS: VECs were isolated from rat thoracic aortas. The cells were pretreated with paeonol for 24 h, and then stimulated with ox-LDL for another 24 h. The expression of microRNA-21 (miR-21) and PTEN in VECs was analyzed using qRT-PCR. The expression of PTEN protein was detected by Western blotting. TNF-α release by VECs was measured by ELISA. RESULTS: Ox-LDL treatment inhibited VEC growth in dose- and time-dependent manners (the value of IC50 was about 20 mg/L at 24 h). Furthermore, ox-LDL (20 mg/L) significantly increased miR-21 expression and inhibited the expression of PTEN, one of downstream target genes of miR-21 in VECs. In addition, ox-LDL (20 mg/L) significantly increased the release of TNF-α from VECs. Pretreatment with paeonol increased the survival rate of ox-LDL-treated VECs in dose- and time-dependent manners. Moreover, paeonol (120 µmol/L) prevented ox-LDL-induced increases in miR-21 expression and TNF-α release, and ox-LDL-induced inhibition in PTEN expression. A dual-luciferase reporter assay showed that miR-21 bound directly to PTEN's 3'-UTR, thus inhibiting PTEN expression. In ox-LDL treated VECs, transfection with a miR-21 mimic significantly increased miR-21 expression and inhibited PTEN expression, and attenuated the protective effects of paeonol pretreatment, whereas transfection with an miR-21 inhibitor significantly decreased miR-21 expression and increased PTEN expression, thus enhanced the protective effects of paeonol pretreatment. CONCLUSION: miR-21 is an important target of paeonol for its protective effects against ox-LDL-induced VEC injury, which may play critical roles in development of atherosclerosis.


Asunto(s)
Acetofenonas/farmacología , Antiinflamatorios/farmacología , Aorta Torácica/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Lipoproteínas LDL/toxicidad , MicroARNs/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regiones no Traducidas 3' , Animales , Aorta Torácica/inmunología , Aorta Torácica/metabolismo , Aorta Torácica/patología , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoprotección , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación Enzimológica de la Expresión Génica , Genes Reporteros , Concentración 50 Inhibidora , Masculino , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Ratas Sprague-Dawley , Factores de Tiempo , Transfección
7.
Chem Asian J ; 14(15): 2685-2693, 2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31152680

RESUMEN

Two trinuclear copper phosphonate complexes, [Cu3 (pda)3 (tBuPO3 )]⋅2(Et3 NH) (1) and [Cu3 (pda)3 (PhPO3 )]⋅2(Et3 NH) (2), have been synthesized and investigated by a combination of X-ray crystallography, PXRD, magneto- and electrochemistry, EPR, in situ UV-vis spectroelectrochemistry and DLS. The two complexes feature almost identical crystal structures, the anions of which are both supported by pda2- and tBuPO3 2- /PhPO3 2- groups, bridging three five-coordinated CuII atoms to form a crown-like structure. This is the first time that trinuclear copper phosphonate complexes have been isolated and characterized. Magnetic susceptibility measurements reveal that complexes 1 and 2 both display overall ferromagnetic characters, but with different exchange interactions between the metal ions within the two clusters. The electrocatalytic activity for water oxidation of the two complexes was preliminarily investigated, which reveals that both of the two complexes can carry out electrocatalytic water oxidation in a neutral system owing to the introduction of phosphonate ligands into the complexes, with a TOF of about 0.82 s-1 (1) and 0.58 s-1 (2), respectively. We propose that the presence of phosphonate ligands may affect the magnetic property and catalytic activity of the complexes.

8.
J Cell Physiol ; 216(3): 835-43, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18551429

RESUMEN

Our previous studies have indicated that TGF-beta1 exerts its effect on the expression of A-type potassium channels (I(A)) in rat vascular myofibroblasts by activation of protein kinase C during the phenotypic transformation of vascular fibroblasts to myofibroblasts. In the present study, patch-clamp whole-cell recording and transwell-migration assays were used to examine the effects of TGF-beta1- and phorbol 12-myristate 13-acetate (PMA)-induced expression of I(A) channels on myofibroblast migration and its modulation by the protein kinase A (PKA) pathway. Our results reveal that incubation of fibroblasts with TGF-beta1 or PMA up-regulates the expression of I(A) channels and increases myofibroblast migration. Blocking I(A) channel expression by 4-aminopyridine (4-AP) significantly inhibits TGF-beta1- and PMA-induced myofibroblast migration. Incubation of fibroblasts with forskolin does not result in increased expression of I(A) channels but does cause a slight increase in fibroblast migration at higher concentrations. In addition, forskolin increases the TGF-beta1- and PMA-induced myofibroblast migration but inhibits TGF-beta1- and PMA-induced the expression of I(A) channels. Whole-cell current recordings showed that forskolin augments the delayed rectifier outward K(+) (I(K)) current amplitude of fibroblasts, but not the I(A) of myofibroblasts. Our results also indicate that TGF-beta1- and PMA-induced expression of I(A) channels might be related to increase TGF-beta1- or PMA-induced myofibroblast migration. Promoting fibroblast and myofibroblast migration via the PKA pathway does not seem to involve the expression of I(A) channels, but the modulation of I(K) and I(A) channels might be implicated.


Asunto(s)
Movimiento Celular/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Músculo Liso Vascular/citología , Canales de Potasio/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Colforsina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Fibroblastos/citología , Masculino , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/metabolismo , Canales de Potasio/genética , Ratas , Ratas Wistar , Sistemas de Mensajero Secundario/fisiología , Acetato de Tetradecanoilforbol/metabolismo
9.
Mini Rev Med Chem ; 16(16): 1290-1302, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26864555

RESUMEN

Angiogenesis is indispensible for tumor growth and metastasis. Antiangiogenic therapy is now a validated major strategy in cancer clinic; several small molecule angiogenic inhibitors have been successfully translated into clinic for multiple cancer indications. In the past decade, many natural products with potent antiangiogenic activity were explored and the underlying molecular mechanisms were revealed. One important mechanism is the inhibition of one or several steps in VEGF/VFGFR signaling pathway. Other factors (bFGF, HIF-1α, NF-κB etc.) capable of regulating angiogenesis, are also down-regulated by some natural products. Moreover, some of the antiangiogenic natural products also significantly inhibit vasculogenic mimicry (VM), another important vessel recruitment avenue in cancer, by regulating the key signaling of VM formation including VE-cadherin, EphA2, and Nodal signaling. In this mini-review, we summarized the natural products with suppressive effect on tumor angiogenesis and VM according to their diverse molecular mechanisms, and discussed the major direction of future research in this field.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Imitación Molecular , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/aislamiento & purificación , Animales , Productos Biológicos/química , Productos Biológicos/aislamiento & purificación , Humanos
10.
Biomaterials ; 95: 60-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27130953

RESUMEN

Normalization of the tumor microenvironment is a promising approach to render conventional chemotherapy more effective. Although passively targeted drug nanocarriers have been investigated to this end, actively targeted tumor priming remains to be explored. In this work, we demonstrate an effective tumor priming strategy using metronomic application of nanoparticles actively targeted to tumor neovasculature. F56 peptide-conjugated paclitaxel-loaded nanoparticles (F56-PTX-NP) were formulated from PEGylated polylactide using an oil in water emulsion approach. Metronomic F56-PTX-NP specifically targeted tumor vascular endothelial cells (ECs), pruned vessels with strong antiangiogenic activity and induced thrombospondin-1 (TSP-1) secretion from ECs. The treatment induced tumor vasculature normalization as evidenced by significantly increased coverage of basement membrane and pericytes. The tumor microenvironment was altered with enhanced pO2, lower interstitial fluid pressure, and enhanced vascular perfusion and doxorubicin delivery. A "normalization window" of at least 9 days was induced, which was longer than other approaches using antiangiogenic agents. Together, these results show that metronomic, actively-targeted nanomedicine can induce tumor vascular normalization and modulate the tumor microenvironment, opening a window of opportunity for effective combination chemotherapies.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Nanopartículas/química , Paclitaxel/administración & dosificación , Administración Metronómica , Inhibidores de la Angiogénesis/química , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos , Femenino , Xenoinjertos , Humanos , Lactatos/química , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Oligopéptidos/química , Paclitaxel/química , Polietilenglicoles/química , Microambiente Tumoral
11.
Phytomedicine ; 22(1): 103-10, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25636878

RESUMEN

Raddeanin A (RA) is an active triterpenoid saponin from a traditional Chinese medicinal herb, Anemone raddeana Regel. It was previously reported that RA possessed attractive antitumor activity through inhibiting proliferation and inducing apoptosis of multiple cancer cells. However, whether RA can inhibit angiogenesis, an essential step in cancer development, remains unknown. In this study, we found that RA could significantly inhibit human umbilical vein endothelial cell (HUVEC) proliferation, motility, migration, and tube formation. RA also dramatically reduced angiogenesis in chick embryo chorioallantoic membrane (CAM), restrained the trunk angiogenesis in zebrafish, and suppressed angiogenesis and growth of human HCT-15 colorectal cancer xenograft in mice. Western blot assay showed that RA suppressed VEGF-induced phosphorylation of VEGFR2 and its downstream protein kinases including PLCγ1, JAK2, FAK, Src, and Akt. Molecular docking simulation indicated that RA formed hydrogen bonds and hydrophobic interactions within the ATP binding pocket of VEGFR2 kinase domain. Our study firstly provides the evidence that RA has high antiangiogenic potency and explores its molecular basis, demonstrating that RA is a potential agent or lead candidate for antiangiogenic cancer therapy.


Asunto(s)
Anemone/química , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/farmacología , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Embrión de Pollo , Neoplasias Colorrectales/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Neovascularización Patológica/prevención & control , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/embriología
12.
Biomaterials ; 42: 161-71, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25542804

RESUMEN

Tumor angiogenesis is a multistep process involved with multiple molecular events in cancer microenvironment. Several molecular-targeted agents aiming to suppress tumor angiogenesis have been successfully translated into cancer clinic. However, new strategies are still urgently desired to be excavated to overcome the poor response and resistance in some antiangiogenic therapies. Recently, Delta-like ligand 4 (Dll4) is identified to be specifically over-expressed on tumor vascular endothelial cells (EC), and the Dll4-Notch pathway serves as a critical regulator in the development and maintenance of tumor angiogenesis. The intensively up-regulated phenotype of Dll4 on the membrane of tumor vascular EC implies that Dll4 may act as a targetable address for drug delivery system (DDS) to achieve targeted antiangiogenic cancer therapy. Here, a nano-DDS, GD16 peptide (H2N-GRCTNFHNFIYICFPD-CONH2, containing a disulfide bond between Cys3 and Cys13) conjugated nanoparticles loading paclitaxel (GD16-PTX-NP), which can specifically target the angiogenic marker Dll4, was fabricated for the investigation of antiangiogenic therapeutic efficacy in human head and neck cancer FaDu (Dll4-negative) xenograft in nude mice. The results demonstrate that GD16-PTX-NP achieved controlled drug release and exhibited favorable in vivo long-circulating feature. GD16-PTX-NP exerted enhanced antiangiogenic activity in the inhibition of human umbilical vein endothelial cell (HUVEC) viability, motility, migration, and tube formation, and in the Matrigel plug model as well, which can be definitely ascribed to the active internalization mediated by the interaction of GD16 and the over-expressed Dll4 on EC. GD16-PTX-NP showed accurate in vivo tumor neovasculature targeting property in FaDu tumor, where the paclitaxel was specifically delivered into the tumor vascular EC, leading to significant apoptosis of tumor vascular EC and necrosis of tumor tissues. The antiangiogenic activity of GD16-PTX-NP significantly contributed to its in vivo anticancer efficacy in Fadu tumor; moreover, no overt toxicity to the mice was observed. Our research firstly presents the potency and significance of a Dll4-targeted nanomedicine in antiangiogenic cancer therapy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Sistemas de Liberación de Medicamentos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Nanomedicina/métodos , Neovascularización Patológica/tratamiento farmacológico , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno , Cumarinas/metabolismo , Combinación de Medicamentos , Endocitosis/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Laminina , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Nanopartículas/química , Nanopartículas/ultraestructura , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/patología , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Péptidos/química , Proteoglicanos , Ratas Sprague-Dawley , Tejido Subcutáneo/efectos de los fármacos , Tejido Subcutáneo/patología , Tiazoles/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Biomaterials ; 35(4): 1215-26, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24231414

RESUMEN

Antiangiogenic therapy is a validated approach for colorectal cancer (CRC) treatment. However, diverse adverse effects inevitably appear due to the off-target effect of the approved antiangiogenic inhibitors on the physiological functions and homeostasis. This study was to investigate a new tumor vessel targeting nanoparticulate drug delivery system, F56 peptide conjugated nanoparticles loading vincristine (F56-VCR-NP), for the effective treatment of CRC subcutaneous xenograft and experimental lung metastasis model. The controlled release behavior and in vivo pharmacokinetic profile of F56-VCR-NP were characterized. The tumor vessel targeting and antiangiogenic activity of F56-VCR-NP was evaluated in human umbilical vein endothelial cells (HUVEC, a classical cell model mimicking tumor vascular EC), subcutaneous human HCT-15 xenograft in immunodeficient nude mice, and experimental CT-26 lung metastasis model in immunocompetent mice. The therapeutic efficacy (animal survival and toxicity) was further investigated in the model of CT-26 lung metastasis in mice. F56-VCR-NP could achieve 30-day controlled drug release in PBS (pH 7.4) and exhibited favorable long-circulating feature in vivo. F56-VCR-NP could accurately target the CRC neovasculature and elicit nanoparticle internalization in the tumor vascular EC, where the antiangiogenic VCR-induced dramatic EC apoptosis and necrosis of CRC tissue. F56-VCR-NP significantly prolonged the mouse survival with no obvious toxicity (weight loss and anepithymia) in the CT-26 lung metastasis mice model, and this pronounced antitumor effect was closely related with the decreased microvessel density in the metastases. The present nanoparticle-based targeted antiangiogenic therapy may provide a new promising approach for the therapy of CRC and lung metastasis, which deserves further translational research.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Vincristina/uso terapéutico , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Sistemas de Liberación de Medicamentos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Oligopéptidos/química , Ratas Sprague-Dawley , Vincristina/administración & dosificación , Vincristina/farmacocinética
14.
Biomaterials ; 35(9): 3060-70, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24393268

RESUMEN

Antiangiogenic cancer therapy based on nanoparticulate drug delivery systems (nano-DDS) is emerging as a promising new approach besides the proved molecular-targeted antiangiogenic agents. The current nano-DDS are restricted to the targeting to tumor vascular endothelial cells, but seldom efforts have been made to target the tumor vascular pericytes which are also actively involved in tumor angiogenesis. In this study, we developed a new nano-DDS, TH10 peptide (TAASGVRSMH) conjugated nanoparticles loading docetaxel (TH10-DTX-NP) that can target the NG2 proteoglycan highly expressed in tumor vascular pericytes, for the investigation of therapeutic efficacy in the mice bearing B16F10-luc-G5 melanoma experimental lung metastasis. The results demonstrated that TH10-DTX-NP achieved controlled drug release in PBS and the mixture of rat plasma and PBS (1:1, v/v), and exhibited favorable in vivo long-circulating feature. TH10 peptide conjugation facilitated the nanoparticle internalization in pericytes via the interaction between TH10 and NG2 receptor, leading to more inhibition of pericyte viability and migration. TH10-conjugated nanoparticles could accurately target the vascular pericytes of B16F10-luc-G5 lung metastasis, where DTX-induced pronounceable pericyte apoptosis. TH10-DTX-NP significantly prolonged the mice survival with no obvious toxicity, and this enhanced antitumor effect was closely related with the decreased pericyte density and microvessel density in the lung metastases. The present research reveals the potency and significance of targeting tumor vascular pericytes using nano-DDS in antiangiogenic cancer therapy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Nanopartículas/química , Péptidos/uso terapéutico , Pericitos/patología , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/farmacología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Docetaxel , Endocitosis/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Nanopartículas/ultraestructura , Neovascularización Patológica , Péptidos/química , Péptidos/farmacología , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Ratas , Ratas Sprague-Dawley , Taxoides/farmacología , Taxoides/uso terapéutico
15.
Artículo en Inglés | MEDLINE | ID: mdl-23455073

RESUMEN

A simple, rapid and sensitive LC-MS/MS analysis method was developed and validated for the determination of Raddeanin A (RA) in rat plasma. Protein precipitation with three volumes of methanol as the precipitation reagent was used as the sample preparation method. The analysis process was performed on a Thermo Syncronis C18 column with the mobile phase of methanol-water (containing 5mM ammonium formate, pH 2.2) (85:15, v/v). RA and glycyrrhetinic acid (internal standard) were monitored under negative electrospray ionization in multiple reaction monitoring (MRM) mode. Retention time of RA and IS were 2.1 min and 3.5 min, respectively. The limit of detection was 5 ng/mL and the linear range was 50-50,000 ng/mL. The intra-day and inter-day precision was 1.87-2.94% and 3.25-5.36%, and the intra-day and inter-day accuracy ranged from 5.9% to 10.5% and 5.6% to 11.1%, respectively. The absolute recovery was above 90.3%. The method has been successfully translated to the pharmacokinetic study of RA in rats after intravenous and intraperitoneal administration (0.75 mg/kg).


Asunto(s)
Cromatografía Liquida/métodos , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/sangre , Saponinas/sangre , Espectrometría de Masas en Tándem/métodos , Anemone/química , Animales , Límite de Detección , Masculino , Ácido Oleanólico/química , Ácido Oleanólico/farmacocinética , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Saponinas/química , Saponinas/farmacocinética
16.
Biomaterials ; 34(16): 3925-3937, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23465835

RESUMEN

Metronomic chemotherapy aiming at inhibiting tumor angiogenesis with conventional chemotherapeutics is a promising strategy for antiangiogenic cancer therapy. However, current metronomic chemotherapy mainly focuses on free small-molecule drugs, without any effort to achieve tumor-specific biodistribution, which may lead to long-term toxicity concerns. Metronomic chemotherapy using nanoparticulate drug delivery system (DDS) offers significant upside to reduce off-target side effects, decrease accumulated dose, and enhance the efficacy of tumor vessel targeting without compromising antitumor efficacy; but there has been a lack of thorough experimental data describing the targeted metronomic chemotherapy. Here, we develop a new nanoparticulate DDS, SP5.2 peptide conjugated, Flt-1 (VEGFR-1) targeted nanoparticles for docetaxel (SP5.2-DTX-NP), as a model for the investigation of targeted metronomic chemotherapy with respect to both antitumor efficacy and toxicity. The results demonstrate that metronomic SP5.2-DTX-NP exerts antitumor activity mainly through the antiangiogenic effect of docetaxel, which is specifically delivered into the tumor vascular endothelial cells through the nanoparticle internalization mediated by the interaction of SP5.2 and over-expressed Flt-1 receptors on tumor vessels. Moreover, the antitumor efficacy of targeted metronomic chemotherapy is better than that of the treatment with the DDS given in the maximum tolerated dose (MTD) regimen, which is shown in significantly prolonged mice survival and minimal drug-associated toxicity (bone marrow suppression, hematological toxicity, and mucosal injury of small intestine). The present research reveals and highlights the significance of targeted metronomic therapy with nanoparticulate DDS in antiangiogenic cancer therapy.


Asunto(s)
Administración Metronómica , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Animales , Antineoplásicos/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Docetaxel , Endocitosis/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/patología , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/química , Péptidos/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Taxoides/administración & dosificación , Taxoides/farmacología , Distribución Tisular/efectos de los fármacos , Resultado del Tratamiento , Receptor 1 de Factores de Crecimiento Endotelial Vascular/química , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
17.
Biomaterials ; 34(26): 6163-74, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23706689

RESUMEN

Anticancer drug resistance is a common intractable obstacle in clinical cancer chemotherapy. Here, we hypothesize that antiangiogenic cancer therapy through the targeted delivery of antiangiogenic agents to the tumor endothelial cells (EC), not the resistant cancer cells, may have the potential of combating multidrug resistant cancer. The K237 peptide-conjugated paclitaxel loaded nanoparticles (K237-PTX-NP), which can target KDR receptors highly expressed in the tumor vasculature, were fabricated for this investigation and the human colorectal adenocarcinoma HCT-15 with naturally expressed P-gp on the cell surface was adopted as the resistant tumor model. The human umbilical vein endothelial cells (HUVEC, a classical cell model mimicking tumor EC) were much more sensitive, in the cytotoxicity and apoptosis test, to K237-PTX-NP than Taxol and non-targeted PTX-NP. The enhanced antiangiogenic feature of K237-PTX-NP can be ascribed to the active internalization mediated by the interaction of K237 and KDR specifically highly expressed on the HUVEC, and the significantly extended intracellular drug retention. The tumor vessel targeting of K237-PTX-NP led to increased nanoparticle accumulation in HCT-15 tumors, and more importantly, induced significant apoptosis of tumor vascular EC and necrosis of tumor tissues. Low dose paclitaxel formulated in K237-PTX-NP (1 mg/kg) achieved significant anticancer efficacy of inhibiting the growth of HCT-15 tumors, but the same efficacy could be only obtained with 8 fold dose paclitaxel (8 mg/kg) in Taxol plus XR9576, a potent P-gp inhibitor. The anticancer efficacy of K237-PTX-NP was well related with the improved antiangiogenic effect shown in the dramatically decreased intratumoral microvessel density and pronouncedly increased apoptotic tumor cells, and such approach did not lead to obvious toxicity in the mice. These results suggest that the nanoparticles targeting drug to tumor neovasculature may be a promising strategy for the treatment of multidrug resistant cancer.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Paclitaxel/administración & dosificación , Paclitaxel/uso terapéutico , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Paclitaxel/química , Péptidos/química , Péptidos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-24184832

RESUMEN

A sensitive, rapid and simple LC-MS/MS analysis method was developed and validated for the determination of pristimerin (PR) in rat plasma. Protein precipitation with four volumes of acetonitrile as the precipitation reagent was used as the sample preparation method. The analysis process was performed on a Merck ZIC-HILIC column with the mobile phase of acetonitrile-water (containing 5mM ammonium formate, pH 2.8) (85:15, v/v). PR (m/z 465.3-201.1) and glycyrrhetinic acid (internal standard, m/z 471.5-177.1) were monitored under positive electrospray ionization in multiple reaction monitoring (MRM) mode. Retention time of PR and IS was 2.45min and 2.4min, respectively. The limit of detection was 0.5ng/mL and the linear range was 1-500ng/mL. The intra-day and inter-day precision were 2.89-6.27% and 4.91-8.98%, and the intra-day and inter-day accuracy ranged from -5.81% to 8.64% and -7.37% to 9.57%, respectively. The matrix effects and absolute recovery ranged from 89.3% to 92.4% and 88.7% to 92.8%, respectively. The method has been successfully applied to the determination of PR concentration in rat plasma after intravenous administration (0.5mg/kg).


Asunto(s)
Cromatografía Liquida/métodos , Espectrometría de Masas en Tándem/métodos , Triterpenos/sangre , Animales , Femenino , Triterpenos Pentacíclicos , Ratas , Ratas Sprague-Dawley , Triterpenos/farmacocinética
19.
Toxicol Appl Pharmacol ; 226(3): 225-35, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17961620

RESUMEN

The effect of non-steroidal anti-inflammatory drugs (NSAIDs) on ion channels has been widely studied in several cell models, but less is known about their modulatory mechanisms. In this report, the effect of mefenamic acid on voltage-activated transient outward K(+) current (I(A)) in cultured rat cerebellar granule cells was investigated. At a concentration of 5 microM to 100 microM, mefenamic acid reversibly inhibited I(A) in a dose-dependent manner. However, mefenamic acid at a concentration of 1 microM significantly increased the amplitude of I(A) to 113+/-1.5% of the control. At more than 10 microM, mefenamic acid inhibited the amplitude of I(A) without any effect on activation or inactivation. In addition, a higher concentration of mefenamic acid induced a significant acceleration of recovery from inactivation with an increase of the peak amplitude elicited by the second test pulse. Intracellular application of mefenamic acid could significantly increase the amplitude of I(A), but had no effect on the inhibition induced by extracellular mefenamic acid, implying that mefenamic acid may exert its effect from both inside and outside the ion channel. Furthermore, the activation of current induced by intracellular application of mefenamic acid was mimicked by other cyclooxygenase inhibitors and arachidonic acid. Our data demonstrate that mefenamic acid is able to bi-directionally modulate I(A) channels in neurons at different concentrations and by different methods of application, and two different mechanisms may be involved.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Corteza Cerebelosa/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/toxicidad , Canal de Potasio Kv.1.1/efectos de los fármacos , Ácido Mefenámico/toxicidad , Neuronas/efectos de los fármacos , Animales , Ácido Araquidónico/toxicidad , Células Cultivadas , Corteza Cerebelosa/metabolismo , Relación Dosis-Respuesta a Droga , Canal de Potasio Kv.1.1/fisiología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA