Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Nanobiotechnology ; 20(1): 248, 2022 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-35641956

RESUMEN

From the global perspective, stroke refers to a highly common cause of disability and death. Ischemic stroke (IS), attributed to blood vessel blockage, preventing the flow of blood to brain, acts as the most common form of stroke. Thus far, thrombolytic therapy is the only clinical treatment for IS with the approval from the FDA. Moreover, the physiology barrier complicates therapeutically and diagnostically related intervention development of IS. Accordingly, developing efficient and powerful curative approaches for IS diagnosis and treatment is urgently required. The advent of nanotechnology has brought dawn and hope to better curative and imaging forms for the management of IS. This work reviews the recent advances and challenges correlated with the nano drug delivery system for IS therapy and diagnosis. The overview of the current knowledge of the important molecular pathological mechanisms in cerebral ischemia and how the drugs cross the blood brain barrier will also be briefly summarized.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Isquemia Encefálica/tratamiento farmacológico , Humanos , Sistema de Administración de Fármacos con Nanopartículas , Medicina de Precisión , Accidente Cerebrovascular/tratamiento farmacológico
3.
Mol Pharm ; 13(10): 3506-3517, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27636161

RESUMEN

Combining treatment of anticancer cells and antiangiogenesis is considered to be a potential targeted strategy for brain glioblastoma therapy. In this study, by utilizing the overexpression of Interleukin 13 receptor α2 (IL-13Rα2) on the glioma cells and heparan sulfate on neovascular endothelial cells, we developed a paclitaxel (PTX) loaded Pep-1 and CGKRK peptide-modified PEG-PLGA nanoparticle (PC-NP-PTX) for glioma cells and neovasculature dual-targeted chemotherapy to enhance the antiglioma efficacy. There were significant differences both on the enhancement of cellular uptake in HUVEC and C6 cells and on the improvement of in vitro antiglioma activity in the respect of proliferation, tumor spheroid growth, tube formation, and migration between PC-NP-PTX and Taxol and NP-PTX. As for C6 cells, the IC50 were 3.59 ± 0.056, 2.37 ± 0.044, 1.38 ± 0.028, 1.82 ± 0.035, and 1.00 ± 0.016 µg/mL of Taxol, NP-PTX, Pep-NP-PTX, CGKRK-NP-PTX, and PC-NP-PTX, and for HUVEC cells, the IC50 were 0.44 ± 0.006, 0.33 ± 0.005, 0.25 ± 0.005, 0.19 ± 0.004, and 0.16 ± 0.004 µg/mL of Taxol, NP-PTX, Pep-NP-PTX, CGKRK-NP-PTX, and PC-NP-PTX, respectively. In vivo distribution assays confirmed that PC-NP-PTX targeted and accumulated effectively at glioma site. PC-NP-PTX showed a longer median survival time of 61 days when compared with Taxol (22 days), NP-PTX (24 days), Pep-NP-PTX (32 days), and CGKRK-NP-PTX (34 days). The in vivo antiglioma efficacy and safety evaluation showed PC-NP-PTX significantly enhanced the antiglioma efficacy and displayed negligible acute toxicity.


Asunto(s)
Glioma/tratamiento farmacológico , Nanopartículas/química , Paclitaxel/química , Paclitaxel/uso terapéutico , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Glioblastoma/tratamiento farmacológico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Poliésteres/química , Polietilenglicoles/química , Ratas
4.
Cell Biochem Funct ; 33(8): 534-40, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26559153

RESUMEN

Dysregulation of microRNAs has been demonstrated to contribute to malignant progression of cancers, including nasopharyngeal carcinoma (NPC). miR-539 was previously reported to be significantly downregulated in osteosarcoma. However, the potential role and mechanism of action of miR-539 in the initiation and progression of NPC remain largely unknown. Quantitative reverse transcription (RT)-PCR demonstrated that miR-539 was significantly downregulated in NPC tumour tissues compared with nontumour tissues. The cell viability, colony formation assay and tumourigenicity assays in nude mice showed that miR-539 could inhibit NPC cell growth in vitro and in vivo. The cyclin-dependent kinase 4 (CDK4) was verified as a miR-539 target gene using dual-luciferase reporter assays, quantitative RT-PCR and Western blotting and was involved in miR-539-regulated NPC cell growth. These results indicated that miR-539 plays an important role in the initiation and progression of NPC by targeting CDK4 and the miR-539/CDK4 pathway may contribute to the development of novel therapeutic strategies for NPC in the future.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , MicroARNs/metabolismo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Animales , Secuencia de Bases , Carcinoma , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Datos de Secuencia Molecular , Carcinoma Nasofaríngeo
5.
Cell Biochem Funct ; 33(1): 23-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25529528

RESUMEN

Epithelial barrier dysfunction is involved in the pathogenesis of allergic disorders, such as nasal allergy. TWIK-related K(+) 1 (Trek1) potassium channels are required in the maintenance of the epithelial barrier function. This study aims to investigate the role of antigen-specific immunotherapy (SIT) in the regulation of Trek1 expression in the nasal mucosa. In this study, patients with nasal allergy were treated with SIT and/or Clostridium butyricum. The expression of Trek1 and histone demethylase 1 (HDAC1) in the nasal epithelia was assessed by real-time reverse transcription polymerase chain reaction and Western blotting. Serum cytokines were assessed by enzyme-linked immunosorbent assay. The results showed that Trek1 and HDAC1 were detected in the nasal epithelia. Trek1 was lower, whereas HDAC1 was higher in patients with allergic rhinitis as compared with healthy controls. Trek1-null RPMI2650 monolayers showed a markedly compromised epithelial barrier function. Treatment with SIT significantly increased the Trek1 levels in the nasal epithelia of allergic rhinitis patients that were further improved in conjunction of SIT and administration of probiotic C. butyricum. In conclusion, nasal epithelia express Trek1 that can be suppressed by allergic response. SIT can restore the expression of Trek1 in the nasal epithelia and can be further improved by conjunction with administration of C. butyricum.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inmunoterapia , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Rinitis Alérgica/tratamiento farmacológico , Animales , Anticuerpos Bloqueadores/farmacología , Butiratos/farmacología , Línea Celular Tumoral , Infecciones por Clostridium/metabolismo , Clostridium butyricum/química , Clostridium butyricum/metabolismo , Histona Desacetilasa 1/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Masculino , Ratones Endogámicos BALB C , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Rinitis Alérgica/metabolismo , Células Th2/metabolismo
6.
Heliyon ; 9(11): e21219, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37964842

RESUMEN

Background: In the Qing dynasty, Yanghe decoction was as a therapeutic soup for effectively treating chronic inflammatory disorders. It was used as a therapeutic soup for effectively treating chronic inflammatory disorders. In the clinical use of Yanghe decoction, the adjustment of the medication for a variety of inflammatory diseases have therapeutic effect, including mastitis. Therefore, Jiawei Yanghe decoction (JWYHD) may be used to treat inflammatory breast diseases. Methods: First, LM- and JWYHD-related components were retrieved from the database and analysis platform. Next, protein-protein interaction networks were constructed to screen the key targets, and gene ontology and Kyoto encyclopedia of gene and genome enrichment analyses were performed to predict the potential biological functions and mechanisms of JWYHD. Simultaneously, the JWYHD samples were collected and analyzed by UPLC-HRMS. Finally, in vivo and in vitro experiments were conducted to construct animal and cellular inflammation models of mastitis with LPS. Pathological changes in the mammary tissues were detected. Enzyme-linked immunosorbent assay, reverse transcription-polymerase chain reaction, and Western blotting was performed to determine the mRNA and protein levels of inflammatory cytokines and toll-like receptor 4/myeloid differentiation primary response 88/nuclear factor kappa B signaling pathway in the breast tissues to elucidate the potential underlying mechanisms of anti-mastitis effects of JWYHD from different aspects. Results: In total, 103 compounds were detected in JWYHD by UPLC-HRMS. 691 active ingredients of JWYHD were screened by network pharmacology, and 47 LM-related targets were identified. The PPI network analysis of the targets revealed the 5 core targets. The KEGG enrichment results established the NF-κB signaling pathways as the core. After JWYHD intervention, low inflammatory enrichment and mild inflammatory damage in breast tissues were observed. Furthermore, JWYHD treatment affected mammary gland inflammatory cytokines and the TLR4/Myd88/NF-κB signaling pathway by considerably reducing the respective protein levels and gene expression; thus, JWYHD alleviated LM symptoms. Conclusions: We hypothesized and demonstrated the anti-inflammatory effects of JWYHD by cytokine regulation via the TLR4/Myd88/NF-κB signaling pathway. In conclusion, JWYHD showed its potential in LM treatment and in treating other acute and chronic inflammatory diseases.

7.
J Control Release ; 359: 224-233, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37290721

RESUMEN

Infiltrative glioma growth makes surgical excision incomplete, and the residual tumor cells proliferate rapidly. Residual glioma cells evade phagocytosis by macrophages through upregulating anti-phagocytosis molecule CD47, which binds to the signal regulatory protein alpha (SIRPα) of macrophages. Specifically, blocking the CD47-SIRPα pathway is a potential strategy for post-resection glioma treatment. In addition, the anti-CD47 antibody (α-CD47) in combination with temozolomide (TMZ) caused an enhanced pro-phagocytic effect due to the TMZ not only destroying DNA but also inducing endoplasmic reticulum stress response of glioma cells. However, the obstruction of the blood-brain barrier makes systemic combination therapy not ideal for post-resection glioma treatment. Herein, we designed a temperature-sensitive hydrogel system based on a moldable thermosensitive hydroxypropyl chitin (HPCH) copolymer to encapsulate both α-CD47 and TMZ as α-CD47&TMZ@Gel for in situ postoperative cavity administration. Through the in vitro and in vivo evaluations, α-CD47&TMZ@Gel significantly inhibited glioma recurrence post-resection through enhancement of pro-phagocytosis of macrophages, recruitment, and activation of CD8+ T cells and NK cells.


Asunto(s)
Glioblastoma , Glioma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/cirugía , Glioblastoma/metabolismo , Temozolomida/uso terapéutico , Linfocitos T CD8-positivos/patología , Receptores Inmunológicos , Glioma/tratamiento farmacológico
8.
Cells ; 11(15)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35954205

RESUMEN

Follicular atresia is primarily caused by breakdown to granulosa cells (GCs) due to oxidative stress (OS). MicroRNAs (miRNAs) elicit a defense response against environmental stresses, such as OS, by acting as gene-expression regulators. However, the association between miRNA expression and OS in porcine GCs (PGCs) is unclear. Here, we examined the impact of H2O2-mediated OS in PGCs through miRNA-Seq. We identified 22 (14 upregulated and 8 downregulated) and 33 (19 upregulated and 14 downregulated) differentially expressed miRNAs (DEmiRNAs) at 100 µM and 300 µM H2O2, respectively, compared with the control group. Among the DEmiRNAs, mi-192 was most induced by H2O2-mediated OS, and the downregulation of miR-192 alleviated PGC oxidative injury. The dual-luciferase reporter assay results revealed that miR-192 directly targeted Acvr2a. The Acvr2a level was found to be remarkably decreased after OS. Furthermore, grape seed procyanidin B2 (GSPB2) treatment significantly reduced the H2O2-induced upregulation of miR-192, and decreased PGC apoptosis and oxidative damage. Meanwhile, GSPB2 prevented an H2O2-induced increase in caspase-3 activity, which was enhanced by the application of the miR-192 inhibitor. These results indicate that GSPB2 protects against PGC oxidative injury via the downregulation of miR-192, the upregulation of Acvr2a expression, and the suppression of the caspase-3 apoptotic signaling pathway.


Asunto(s)
Peróxido de Hidrógeno , MicroARNs , Animales , Caspasa 3/metabolismo , Regulación hacia Abajo/genética , Femenino , Atresia Folicular/genética , Células de la Granulosa/metabolismo , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Estrés Oxidativo , Porcinos
9.
Acta Biomater ; 147: 314-326, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35588994

RESUMEN

Reperfusion injury is one of the major causes of disability and death caused by ischemic stroke, and drug development focuses mainly on single neuron protection. However, different kinds of cells in the neurovascular units (NVUs), including neurons, microglia and vascular endothelial cells, are pathologically changed after cerebral ischemia-reperfusion injury, resulting in an urgent need to develop a drug delivery system to comprehensively protect the kinds of cells involved in the NVU. Herein, we have constructed a c(RGDyK) peptide modified, NF-κB inhibitor caffeic acid phenethyl ester (CAPE)-loaded and reactive nitrogen species (RNS) stimuli-responsive liposomal nanocarrier (R-Lipo-CAPE) to target ischemic lesions and then remodel the NVU to reduce the progression of cerebral ischemia-reperfusion injury. The R-Lipo-CAPE liposomes were approximately 170 nm with a zeta potential of -30.8 ± 0.2 mV. The in vitro CAPE release behavior from R-Lipo-CAPE showed an RNS-dependent pattern. For in vivo studies, transient middle cerebral artery occlusion/reperfusion (MCAO) model mice treated with R-Lipo-CAPE had the least neurological impairment and decreased brain tissue damage, with an infarct area of 13%, compared with those treated with saline of 53% or free CAPE of 38%. Furthermore, microglia in the ischemic brain were polarized to the tissue-repairing M2 phenotype after R-Lipo-CAPE treatment. In addition, R-Lipo-CAPE-treated mice displayed a prominent down-regulated expression of MMP-9 and restored expression of the tight junction protein claudin-5. This proof-of-concept indicates that R-Lipo-CAPE is a promising nanomedicine for the treatment of cerebral ischemia-reperfusion injury through the regulation of neurovascular units. STATEMENT OF SIGNIFICANCE: Based on the complex mechanism and difficulty in treatment of cerebral ischemia-reperfusion injury, the overall regulation of neurovascular unit has become an extremely important target. However, little nanomedicine has been directed to remodel the neurovascular units in targeted cerebral ischemia-reperfusion injury therapy. Here, c(RGDyK) peptide modified reactive nitrogen species (RNS) stimuli-responsive liposomal nanocarrier loaded with a NF-κB inhibitor (CAPE), was designed to simultaneously regulate various cells in the microenvironment of cerebral ischemia-reperfusion injury to remodel the neurovascular units. Our in vitro and in vivo data showed that the intelligent nanocarrier exerted the ability of pathological signal stimuli-responsive drug release, cerebral ischemia-reperfusion injury site targeting and neurovascular units remodeling through reducing neuron apoptosis, regulating microglia polarization and repairing vascular endothelial cell. Overall, the intelligent liposomal drug delivery system was a promising and safe nanomedicine in the perspective of cerebral ischemia-reperfusion injury treatment.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Células Endoteliales/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ratones , FN-kappa B/metabolismo , Especies de Nitrógeno Reactivo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología
10.
ACS Appl Mater Interfaces ; 14(24): 27623-27633, 2022 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-35673881

RESUMEN

Glioma is the most lethal brain tumor with a poor prognosis, and a combination of multiple therapeutic strategies is critical for postoperative glioma treatment. Herein, a multifunctional hybrid hydrogel system (designated as CP&CL@RNPPTX-Gel) was developed for local treatment of postoperative glioma. The system was composed of self-illuminating chlorin e6 (Ce6) conjugated with luminol molecule (CL)-loaded glioma-targeting paclitaxel prodrug nanoparticles and copper peroxide nanodots (CP NDs) coembedded into a three-dimensional thermosensitive hydroxypropyl chitin hydrogel frame. After injection of CP&CL@RNPPTX-Gel into the cavity of postoperative glioma, the solution could be cross-linked into the gel as a drug reservoir under body temperature stimulation. Then, the sustained-released CP NDs decomposed into Cu2+ and H2O2 in the acidic microenvironment of the glioma cells to exert chemodynamic therapy (CDT). Meanwhile, Cu2+ could catalyze the self-luminescence of CL to induce photodynamic therapy (PDT) without external excitation light. Moreover, paclitaxel prodrug nanoparticles degraded into paclitaxel to restrain residual glioma cells in response to intracellular reduced glutathione (GSH). The in vitro and in vivo results showed that CP&CL@RNPPTX-Gel had great potential as a multifunctional hybrid hydrogel system with remarkable therapeutic effects for postoperative glioma treatment via a combination of chemotherapy, CDT, and PDT.


Asunto(s)
Glioma , Nanopartículas , Fotoquimioterapia , Profármacos , Línea Celular Tumoral , Cobre/farmacología , Glioma/tratamiento farmacológico , Glioma/cirugía , Humanos , Hidrogeles/farmacología , Peróxido de Hidrógeno/farmacología , Nanopartículas/uso terapéutico , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Profármacos/farmacología , Microambiente Tumoral
11.
J Control Release ; 345: 786-797, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35367277

RESUMEN

It is well known that glioma is currently the most malignant brain tumor. Because of the existence of blood-brain barrier (BBB) and tumor cell heterogeneity, systemic chemotherapy exerts unsatisfied therapeutic effect for the treatment of glioma after surgical resection and may even damage the body's immune system. Here, we developed an in situ sustained-release hydrogel delivery system for combined chemo-immunotherapy of glioma by combined chemotherapy drug and immunoadjuvant through the resection cavity local delivery. Briefly, glioma homing peptide modified paclitaxel targeting nanoparticles (PNPPTX) and mannitolated immunoadjuvant CpG targeting nanoparticles (MNPCpG) were embedded into PLGA1750-PEG1500-PLGA1750 thermosensitive hydrogel framework (PNPPTX&MNPCpG@Gel). The in vitro and in vivo results showed that the targeting nanoparticles-hydrogel hybrid system could cross-link into a gel drug reservoir when injected into the resection cavity of glioma. And then, the sustained-release PNPPTX could target the residual infiltration glioma cells and produce tumor antigens. Meanwhile, MNPCpG targeted and activated the antigen-presenting cells, which enhanced the tumor antigen presentation ability and activated CD8+T and NK cells to reverse immunosuppression of glioma microenvironment. This study indicated that the PNPPTX&MNPCpG@Gel system could enhance the therapeutic effect of glioma by chemo-immunotherapy.


Asunto(s)
Neoplasias Encefálicas , Glioma , Nanopartículas , Adyuvantes Inmunológicos/uso terapéutico , Antígenos de Neoplasias/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Preparaciones de Acción Retardada/uso terapéutico , Sistemas de Liberación de Medicamentos , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Hidrogeles/uso terapéutico , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Microambiente Tumoral
12.
Reprod Sci ; 28(9): 2630-2640, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33725312

RESUMEN

It remained unknown whether HDAC6 affected the histone deacetylation of in vitro maturation oocytes and the reprogramming of nuclear transplantation in pig. Our results indicated that HDAC6 specific inhibition did not affect overall HDAC activity and meiosis process, which increased histone H3K9/K14 and H4K8 acetylation of porcine in vitro maturation oocytes and pseudo-pronucleus embryos. HDAC6 inhibition also significantly enhanced the cleavage and blastocyst of nuclear transfer embryos (0.81 ± 0.12 vs. 0.68 ± 0.12 and 0.46 ± 0.19; 0.73 ± 0.13 vs. 0.63 ± 0.18 and 0.40 ± 0.16, P<0.05). The inhibition of HDAC6 significantly enhanced histone H3K9/K14 and H4K8 acetylation, and upregulated the OCT4 and CDX2 expressions (1.83 ± 0.16 vs. 1.00 ± 0.00 %; 2.07 ± 0.09 vs. 1.00 ± 0.00; P<0.05) in porcine SCNT blastocysts. Interestingly, HDAC6 inhibition significantly increased the pseudo-pronucleus volume during somatic cell reprogramming. Thus, HDAC6 was required for porcine histone deacetylation during the in vitro maturation and pseudo-pronucleus stages. HDAC6 inhibition improved the in vitro development of nuclear transfer embryos. HDAC6 may restrict the reprogramming of somatic nuclear transfer by regulating pseudo-pronucleus expansion. We need further research to confirm this in the future.


Asunto(s)
Blastocisto/enzimología , Reprogramación Celular , Histona Desacetilasa 6/metabolismo , Histonas/metabolismo , Técnicas de Maduración In Vitro de los Oocitos , Técnicas de Transferencia Nuclear , Oocitos/enzimología , Acetilación , Animales , Blastocisto/efectos de los fármacos , Bufexamac/farmacología , Células Cultivadas , Técnicas de Cultivo de Embriones , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Oocitos/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Sus scrofa
13.
Front Pharmacol ; 11: 558, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425792

RESUMEN

The serious therapeutic obstacles to glioma treatment include poor penetration across the blood-brain barrier (BBB) and low accumulation of therapeutic drugs at tumor sites. In this study, borneol combined with CGKRK peptide (a ligand of the heparan sulfate which overexpress on the glioma cells) modified paclitaxel prodrug self-assembled redox-responsive nanoparticles (CGKRK-PSNPs) were hypothesized to enhance the BBB penetration ability and active tumor targeting efficiency, respectively. The resulting CGKRK-PSNPs possessed a spherical shape with a small particle size (105.61 ± 1.53 nm) and high drug loading for PTX (54.18 ± 1.13%). The drug release behavior proved that CGKRK-PSNPs were highly sensitive to glutathione (GSH) redox environment. The in vitro cell experiments suggested that CGKRK-PSNPs significantly increased the cellular uptake and cytotoxicity of U87MG cells, meanwhile CGKRK-PSNPs showed the low cytotoxicity against BCEC cells. Combined with borneol, CGKRK-PSNPs exhibited enhanced transportation across in vitro BBB model. In intracranial U87MG glioma-bearing nude mice, the higher accumulation of CGKRK-PSNPs combined with borneol was observed through real-time fluorescence image. Moreover, the in vivo anti-glioma results confirmed that CGKRK-PSNPs combined with borneol could improve the anti-glioma efficacy with the prolonged medium survival time (39 days). In conclusion, the collaborative strategy of CGKRK-PSNPs combined with borneol provided a promising drug delivery routine for glioblastoma therapy.

14.
Onco Targets Ther ; 12: 509-518, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30666129

RESUMEN

BACKGROUND: There are few clinical challenges associated with the treatment of colorectal cancer (CRC). Studies have shown that TGF-ß plays a crucial role in CRC. Importantly, celastrol, a major components of the root extract of the traditional Chinese herb Tripterygium wilfordii Hook F, has been shown to inhibit the growth, adhesion, and metastasis of human CRC cells through the inhibition of TGF-ß1/Smad signaling. MATERIALS AND METHODS: Real-time PCR and Western blot tests were proceeded to present TGF-ß1, TGF-ß receptor type I (TGFßRI), TGF-ß receptor type II (TGFßRII), Smad2/3, p-Smad2/3, Smad4, and glyceraldehyde-3-phosphate dehydrogenase expression in human colon cancer cell samples. RESULTS: Our results indicated that celastrol can reduce the expression levels of TGF-ß1, TGFßRI, and TGFßRII in HCT116 and SW620 cells. Furthermore, celastrol could also prevent the increase in Smad4 and p-Smad2/3 in HCT116 and SW620 cells. CONCLUSION: Celastrol could inhibit tumor growth through TGF-ß1/Smad signaling and might be a promising therapeutic component against CRC.

15.
Drug Deliv ; 24(1): 1401-1409, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28933201

RESUMEN

Drug delivery systems based on nanoparticles (nano-DDS) have aroused attentions for the treatment of glioblastoma (GBM), the most malignant brain cancer with a dismal prognosis. However, there are still numerous unmet challenges for traditional nano-DDS, such as the poor nanoparticle penetration, short retention in the GBM parenchyma and low glioma targeting ability. Herein, we used Pep-1 and CREKA peptides to construct a novel multifunctional GBM targeting nano-DDS (PC-NP). Pep-1 was used to overcome the blood-brain tumor barrier (BBTB) and home to glioma cells via interleukin-13 receptor-α2-mediated endocytosis, and CREKA was used to bind to fibrin-fibronectin complexes abundantly expressed in tumor microenvironment for enhanced retention in the GBM. Biological studies showed that the cellular uptake of PC-NP by U87MG cells was significantly enhanced compared with the non-targeting NP. Furthermore, CREKA modification increased the binding capacity of PC-NP to fibrin-fibronectin complexes as confirmed by the competition experiment. In accordance with the increased cellular uptake, PC-NP remarkably increased the cytotoxicity of its payload paclitaxel (PTX) against U87MG cells with an IC50 of 0.176 µg/mL. In vivo fluorescence imaging and antiglioma efficacy evaluation further confirmed that PC-NP accumulated effectively and penetrated deeply into GBM tissue. PC-NP-PTX exhibited a median survival time as long as 61 days in intracranial GBM-bearing mice. In conclusion, our findings indicated PC-NP as a promising nano-DDS for GBM targeting delivery of anticancer drugs.


Asunto(s)
Glioblastoma , Animales , Neoplasias Encefálicas , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas , Paclitaxel
16.
Colloids Surf B Biointerfaces ; 147: 242-249, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27518456

RESUMEN

Glioblastoma multiforme (GBM) is the most common and aggressive primary central nervous system (CNS) tumor with a short survival time. The failure of chemotherapy is ascribed to the low transport of chemotherapeutics across the Blood Brain Tumor Barrier (BBTB) and poor penetration into tumor tissue. In order to overcome the two barriers, small nanoparticles with active targeted capability are urgently needed for GBM drug delivery. In this study, we proposed PEGylated Polyamidoamine (PAMAM) dendrimer nanoparticles conjugated with glioma homing peptides (Pep-1) as potential glioma targeting delivery system (Pep-PEG-PAMAM), where PEGylated PAMAM dendrimer nanoparticle was utilized as carrier due to its small size and perfect penetration into tumor and Pep-1 was used to overcome BBTB via interleukin 13 receptor α2 (IL-13Rα2) mediated endocytosis. The preliminary availability and safety of Pep-PEG-PAMAM as a nanocarrier for glioma was evaluated. In vitro results indicated that a significantly higher amount of Pep-PEG-PAMAM was endocytosed by U87 MG cells. In vivo fluorescence imaging of U87MG tumor-bearing mice confirmed that the fluorescence intensity at glioma site of targeted group was 2.02 folds higher than that of untargeted group (**p<0.01), and glioma distribution experiment further revealed that Pep-PEG-PAMAM exhibited a significantly enhanced accumulation and improved penetration at tumor site. In conclusion, Pep-1 modified PAMAM was a promising nanocarrier for targeted delivery of brain glioma.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cisteamina/análogos & derivados , Dendrímeros/química , Sistemas de Liberación de Medicamentos , Glioma/patología , Subunidad alfa2 del Receptor de Interleucina-13/química , Péptidos/administración & dosificación , Polietilenglicoles/química , Animales , Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Cisteamina/administración & dosificación , Cisteamina/química , Cisteamina/farmacocinética , Dendrímeros/administración & dosificación , Endocitosis , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Subunidad alfa2 del Receptor de Interleucina-13/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Péptidos/química , Péptidos/farmacocinética , Polietilenglicoles/administración & dosificación , Distribución Tisular , Células Tumorales Cultivadas
17.
J Control Release ; 233: 64-71, 2016 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-27142584

RESUMEN

Focal cerebral ischemia, known as stroke, causes serious long-term disabilities globally. Effective therapy for cerebral ischemia demands a carrier that can penetrate the blood-brain barrier (BBB) and subsequently target the ischemia area in brain. Here, we designed a novel neuroprotectant (ZL006) loaded dual targeted nanocarrier based on liposome (T7&SHp-P-LPs/ZL006) conjugated with T7 peptide (T7) and stroke homing peptide (SHp) for penetrating BBB and targeting ischemia area, respectively. Compared with non-targeting liposomes, T7&SHp-P-LPs/ZL006 could transport across BCEC cells and significantly enhance cellular uptake and reduce cells apoptosis of excitatory amino acid stimulated PC-12 cells. However, there was no significant difference in cellular uptake between SHp-modified and plain liposomes when PC-12 cells were incubated without excitatory amino acid. Besides, ex vivo fluorescent images indicated that DiR labeled T7&SHp-P-LPs could efficiently transport across BBB and mostly accumulated in ischemic region rather than normal cerebral hemisphere of MCAO rats. Furthermore, T7&SHp-P-LPs/ZL006 could enhance the ability of in vivo anti-ischemic stroke of MCAO rats. These results demonstrated that T7&SHp-P-LPs could be used as a safe and effective dual targeted nanocarrier for ischemic stroke treatment.


Asunto(s)
Colágeno Tipo IV/administración & dosificación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Nanopartículas/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Línea Celular Tumoral , Colágeno Tipo IV/química , Colágeno Tipo IV/uso terapéutico , Liberación de Fármacos , Ácido Glutámico/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Liposomas , Masculino , Ratones Endogámicos ICR , Nanopartículas/química , Nanopartículas/uso terapéutico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/química , Fragmentos de Péptidos/uso terapéutico , Polietilenglicoles/química , Ratas Sprague-Dawley , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
18.
Sci Rep ; 5: 16589, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26567528

RESUMEN

Glioma presents one of the most malignant brain tumors, and the therapeutic effect is often limited due to the existence of brain tumor barrier. Based on interleukin-13 receptor α2 (IL-13Rα2) over-expression on glioma cell, it was demonstrated to be a potential receptor for glioma targeting. In this study, Pep-1-conjugated PEGylated nanoparticles loaded with paclitaxel (Pep-NP-PTX) were developed as a targeting drug delivery system for glioma treatment. The Pep-NP-PTX presented satisfactory size of 95.78 nm with narrow size distribution. Compared with NP-PTX, Pep-NP-PTX exhibited significantly enhanced cellular uptake in C6 cells (p < 0.001). The in vitro anti-proliferation evaluation showed that the IC50 were 146 ng/ml and 349 ng/ml of Pep-NP-PTX and NP-PTX, respectively. The in vivo fluorescent image results indicated that Pep-NP had higher specificity and efficiency in intracranial tumor accumulation. Following intravenous administration, Pep-NP-PTX could enhance the distribution of PTX in vivo glioma section, 1.98, 1.91 and 1.53-fold over that of NP-PTX group after 0.5, 1 and 4 h, respectively. Pep-NP-PTX could improve the anti-glioma efficacy with a median survival time of 32 days, which was significantly longer than that of PTX-NP (23 days) and Taxol(®) (22 days). In conclusion, Pep-NP-PTX is a potential targeting drug delivery system for glioma treatment.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Portadores de Fármacos/administración & dosificación , Glioblastoma/tratamiento farmacológico , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Nanopartículas/administración & dosificación , Paclitaxel/administración & dosificación , Animales , Antineoplásicos Fitogénicos/farmacocinética , Encéfalo/metabolismo , Línea Celular Tumoral , Portadores de Fármacos/farmacocinética , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Paclitaxel/farmacocinética , Ratas , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Sci Rep ; 5: 12651, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26219474

RESUMEN

The treatment for ischemic stroke is one of the most challenging problems and the therapeutic effect remains unsatisfied due to the poor permeation of drugs across the blood brain barrier (BBB). In this study, HAIYPRH (T7), a peptide that targeted to transferrin receptor (TfR) can mediate the transport of nanocarriers across the BBB, was conjugated to liposomes for ischemic stroke targeting treatment of a novel neuroprotectant (ZL006). T7-conjugated PEGylated liposomes (T7-P-LPs) loaded with ZL006 (T7-P-LPs/ZL006) were showed satisfactory vesicle size and size distribution. Furthermore, the cellular uptake results showed that T7 modification increased liposomes uptake by the brain capillary endothelial cells (BCECs) and little cytotoxicity of liposomes with or without ZL006 was observed. The in vivo biodistribution and near-infrared fluorescence imaging evidenced that T7 modification rendered liposomes significantly enhanced the transport of liposomes across the BBB. The pharmacodynamic study suggested that, T7-P-LPs/ZL006 exhibited reduced infarct volume and ameliorated neurological deficit compared with unmodified liposomes or free ZL006. T7-P-LPs/ZL006 could be targeted to brain and displayed remarkable neuroprotective effects. They could be used as a potential targeted drug delivery system of ischemic stroke treatment.


Asunto(s)
Ácidos Aminosalicílicos/farmacología , Bencilaminas/farmacología , Sistemas de Liberación de Medicamentos/métodos , Péptidos/farmacología , Accidente Cerebrovascular/prevención & control , Secuencia de Aminoácidos , Ácidos Aminosalicílicos/administración & dosificación , Ácidos Aminosalicílicos/química , Animales , Bencilaminas/administración & dosificación , Bencilaminas/química , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia Encefálica/complicaciones , Células Cultivadas , Células Endoteliales/metabolismo , Liposomas/química , Liposomas/farmacocinética , Liposomas/ultraestructura , Masculino , Ratones Endogámicos ICR , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Tamaño de la Partícula , Péptidos/administración & dosificación , Péptidos/química , Polietilenglicoles/química , Ratas Sprague-Dawley , Accidente Cerebrovascular/etiología , Distribución Tisular
20.
Biomaterials ; 35(22): 5897-907, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24743033

RESUMEN

The treatment for glioma is one of the most challenging problems and therapeutic effect of glioma is often limited due to poor penetration into the tumor tissue. Interleukin 13 receptor α2 (IL-13Rα2) is over-expressed on tumor including established glioma cell lines and primary glioblastoma cell cultures. However, it will not cause activation of its signaling pathways. So it could be served as a promising targeted moiety for anti-glioma drug delivery. Pep-1, one specific ligand of IL-13Rα2, was identified to exhibit excellent capacity of crossing the blood tumor barrier (BTB) and homing to giloma. In this study, based on the IL-13Rα2-mediated endocytosis, Pep-1 was exploited as a potential ligand for effective glioma-targeting delivery. Pep-1 was functionalized to the surface of PEG-PLGA nanoparticles (Pep-NP) to evaluate its glioma homing, by taking advantage of the excessive expression of the IL-13Rα2 on the surface of glioma cells. Compared with non-targeting nanoparticles, Pep-NP exhibited a significantly enhanced cellular association in rat C6 glioma cells and improved penetration in 3D avascular C6 glioma spheroids. Following intravenous administration, Pep-NP could facilitate the distribution of the coumarin-6 in vivo glioma region, 2.21 times higher than that of NP for quantitative analysis. In conclusion, the Pep-NP could precisely target to the brain glioma, which was a potential targeting drug delivery system for glioma treatment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Cisteamina/análogos & derivados , Sistemas de Liberación de Medicamentos , Glioma/metabolismo , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Nanopartículas/metabolismo , Péptidos/metabolismo , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular , Cumarinas/administración & dosificación , Cumarinas/farmacocinética , Cisteamina/química , Cisteamina/metabolismo , Glioma/tratamiento farmacológico , Masculino , Ratones , Nanopartículas/química , Péptidos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA