Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Adv Sci (Weinh) ; 11(12): e2306499, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38229201

RESUMO

EZH2 is the catalytic subunit of the histone methyltransferase Polycomb Repressive Complex 2 (PRC2), and its somatic activating mutations drive lymphoma, particularly the germinal center B-cell type. Although PRC2 inhibitors, such as tazemetostat, have demonstrated anti-lymphoma activity in patients, the clinical efficacy is not limited to EZH2-mutant lymphoma. In this study, Activin A Receptor Type 1 (ACVR1), a type I Bone Morphogenetic Protein (BMP) receptor, is identified as critical for the anti-lymphoma efficacy of PRC2 inhibitors through a whole-genome CRISPR screen. BMP6, BMP7, and ACVR1 are repressed by PRC2-mediated H3K27me3, and PRC2 inhibition upregulates their expression and signaling in cell and patient-derived xenograft models. Through BMP-ACVR1 signaling, PRC2 inhibitors robustly induced cell cycle arrest and B cell lineage differentiation in vivo. Remarkably, blocking ACVR1 signaling using an inhibitor or genetic depletion significantly compromised the in vitro and in vivo efficacy of PRC2 inhibitors. Furthermore, high levels of BMP6 and BMP7, along with ACVR1, are associated with longer survival in lymphoma patients, underscoring the clinical relevance of this study. Altogether, BMP-ACVR1 exhibits anti-lymphoma function and represents a critical PRC2-repressed pathway contributing to the efficacy of PRC2 inhibitors.


Assuntos
Linfoma de Células B , Linfoma , Humanos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Transdução de Sinais/fisiologia , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo
3.
J Biol Chem ; 299(4): 103073, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36858198

RESUMO

Polycomb repressive complex 2 (PRC2) suppresses gene transcription by methylating lysine 27 of histone H3 (H3K27) and plays critical roles in embryonic development. Among the core PRC2 subunits, EZH2 is the catalytic subunit and EED allosterically activates EZH2 upon binding trimethylated H3K27 (H3K27me3). Activating mutations on Y641, A677, and A687 within the enzymatic SET (Su(Var)3 to 9, Enhancer-of-zeste, and Trithorax) domain of EZH2 have been associated with enhanced H3K27me3 and tumorigenicity of many cancers including B-cell lymphoma and melanoma. To tackle the critical residues outside the EZH2 SET domain, we examined EZH2 mutations in lymphoma from cancer genome databases and identified a novel gain-of-function mutation W113C, which increases H3K27me3 in vitro and in vivo and promotes CDKN2A silencing to a similar level as EZH2 Y641F. Different from other gain-of-function mutations, this mutation is located in the SET-activation loop at the EZH2 N terminus, which stabilizes the SET domain and facilitates substrate binding. This may explain how the W113C mutation increases PRC2 activity. Tazemetostat is a Food and Drug Administration-approved EZH2-binding inhibitor for follicular lymphoma treatment. Intriguingly, the W113C mutation leads to tazemetostat resistance in both H3K27 methylation and tumor proliferation. Another class of allosteric PRC2 inhibitor binding EED overcomes the resistance, effectively decreases H3K27me3, and blocks tumor proliferation in cells expressing EZH2 W113C. As this mutation is originally identified from lymphoma samples, our results demonstrated its activating characteristic and the deleterious consequence, provide insights on PRC2 regulation, and support the continued exploration of treatment optimization for lymphoma patients.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Mutação com Ganho de Função , Linfoma de Células B , Humanos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histonas/metabolismo , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/genética , Mutação , Complexo Repressor Polycomb 2/genética
4.
Cell Death Dis ; 13(2): 155, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35169119

RESUMO

The methyltransferase Polycomb Repressive Complex 2 (PRC2), composed of EZH2, SUZ12, and EED subunits, is associated with transcriptional repression via tri-methylation of histone H3 on lysine 27 residue (H3K27me3). PRC2 is a valid drug target, as the EZH2 gain-of-function mutations identified in patient samples drive tumorigenesis. PRC2 inhibitors have been discovered and demonstrated anti-cancer efficacy in clinic. However, their pharmacological mechanisms are poorly understood. MAK683 is a potent EED inhibitor in clinical development. Focusing on MAK683-sensitive tumors with SMARCB1 or ARID1A loss, we identified a group of PRC2 target genes with high H3K27me3 signal through epigenomic and transcriptomic analysis. Multiple senescence-associated secretory phenotype (SASP) genes, such as GATA4, MMP2/10, ITGA2 and GBP1, are in this group besides previously identified CDKN2A/p16. Upon PRC2 inhibition, the de-repression of SASP genes is detected in multiple sensitive models and contributes to decreased Ki67+, extracellular matrix (ECM) reorganization, senescence associated inflammation and tumor regression even in CDKN2A/p16 knockout tumor. And the combination of PRC2 inhibitor and CDK4/6 inhibitor leads to better effect. The genes potential regulated by PRC2 in neuroblastoma samples exhibited significant enrichment of ECM and senescence associated inflammation, supporting the clinical relevance of our results. Altogether, our results unravel the pharmacological mechanism of PRC2 inhibitors and propose a combination strategy for MAK683 and other PRC2 drugs.


Assuntos
Neoplasias , Complexo Repressor Polycomb 2 , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histonas/química , Humanos , Inflamação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Fenótipo Secretor Associado à Senescência
5.
Front Cell Dev Biol ; 9: 757747, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869346

RESUMO

miR-92a-3p (microRNA-92a-3p) has been reported to be dysregulated in several cancers, and as such, it is considered to be a cancer-related microRNA. However, the influence of miR-92a-3p on biological behaviors in cervical cancer (CC) still remains unclear. Quantitative real-time PCR was used to detect miR-92a-3p levels in CC stem cells. Here, Cell Counting Kit-8 (CCK8) assay, Transwell cell invasion assay and flow cytometry assay were used to characterize the effects that miR-92a-3p and large tumor suppressor l (LATS1) had on proliferation, invasion and cell cycle transition. The luciferase reporter gene assay was used to verify the targeting relationship between miR-92a-3p and LATS1. Western Blotting was used to investigate the related signaling pathways and proteins. Data from The Cancer Genome Atlas (TCGA) showed that miR-92a-3p was upregulated in CC tissues and closely associated with overall survival. miR-92a-3p promoted proliferation, invasion and cell cycle transition in CC stem cells. The luciferase reporter assay showed that miR-92a-3p bound to the 3'-untranslated region (3'-UTR) of the LATS1 promoter. LATS1 inhibited proliferation, invasion and cell cycle transition. Results measured by Western Blotting showed that LATS1 downregulated expressions of transcriptional co-activator with PDZ-binding motif (TAZ), vimentin and cyclin E, but upregulated the expression of E-cadherin. Re-expression of LATS1 partly reversed the effects of miR-92a-3p on proliferation, invasion and cell cycle transition, as well as on TAZ, E-cadherin, vimentin, and cyclin E. miR-92a-3p promoted the malignant behavior of CC stem cells by targeting LATS1, which regulated TAZ and E-cadherin.

6.
Front Chem ; 8: 500, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850613

RESUMO

The utilization of nanotechnology to deliver vaccines and modulate immunity has shown great potential in cancer therapy. Peptide-based supramolecular hydrogels as novel vaccine adjuvants have been found to effectively improve the immune response and tumor curative effect. In this study, we designed a set of reduction-responsive self-assembled peptide precursors (Fbp-GDFDFDYD(E, S, or K)-ss-ERGD), which can be reduced by glutathione (GSH) into Fbp-GDFDFDYD(E, S or K)-SH for forming of hydrogel with different surface properties (E-gel, S-gel, and K-gel, respectively). Using the same method, co-assembled hydrogel vaccines (E-vac, S-vac, and K-vac, respectively) can also be prepared by mixing different precursors with antigens before GSH reduction. Through TEM observation of the nanostructure, we found that all the co-assembled hydrogels, especially K-vac, possessed much denser and more unified nanofiber networks as compared with antigen-free hydrogels, which were very suitable for antigen storage and vaccine delivery. Although the three peptides adopted similar ß-sheet secondary structures, the mechanical properties of their resulted co-assembled hydrogel vaccines were obviously different. Compared to E-vac, S-vac had a much weaker mechanical property, while K-vac had a much higher. In vivo experiments, co-assembled hydrogel vaccines, especially K-vac, also promoted antibody production and anti-tumor immune responses more significantly than the other two vaccines. Our results demonstrated that co-assembled hydrogels formed by peptides and antigens co-assembly could act as effective vaccine delivery systems for boosting antibody production, and different immune effects can be acquired by tuning the surface properties of the involved self-assembling peptides.

7.
Nat Commun ; 11(1): 379, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31953408

RESUMO

Insig-2 is an ER membrane protein negatively controlling lipid biosynthesis. Here, we find that Insig-2 is increased in the tissues, including liver, but unaltered in the muscle of gp78-deficient mice. In hepatocytes and undifferentiated C2C12 myoblasts, Insig-2 is ubiquitylated on Cys215 by gp78 and degraded. However, the C215 residue is oxidized by elevated reactive oxygen species (ROS) during C2C12 myoblasts differentiating into myotubes, preventing Insig-2 from ubiquitylation and degradation. The stabilized Insig-2 downregulates lipogenesis through inhibiting the SREBP pathway, helping to channel the carbon flux to ATP generation and protecting myotubes from lipid over-accumulation. Evolutionary analysis shows that the YECK (in which C represents Cys215 in human Insig-2) tetrapeptide sequence in Insig-2 is highly conserved in amniotes but not in aquatic amphibians and fishes, suggesting it may have been shaped by differential selection. Together, this study suggests that competitive oxidation-ubiquitylation on Cys215 of Insig-2 senses ROS and prevents muscle cells from lipid accumulation.


Assuntos
Cisteína/metabolismo , Proteínas de Membrana/metabolismo , Receptores do Fator Autócrino de Motilidade/metabolismo , Ubiquitinação , Anfíbios , Animais , Células CHO , Linhagem Celular , Cricetulus , Regulação para Baixo , Evolução Molecular , Peixes , Hepatócitos/metabolismo , Humanos , Metabolismo dos Lipídeos , Lipogênese , Fígado/metabolismo , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Receptores do Fator Autócrino de Motilidade/genética , Análise de Sequência de Proteína , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Transcriptoma
8.
Oncol Lett ; 18(2): 1689-1696, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31423236

RESUMO

Anoikis is a specific form of programmed cell death induced by loss of contact between cells and extracellular matrices or other cells. Only tumor cells that are resistant to anoikis can survive in the state of detachment from the primary tissue during the early stages of metastasis. The ability to resist anoikis is crucial for cancer cell metastasis. ILF2 is a proto-oncogene previously studied in glioma, NSCLC, esophageal cancer and pancreatic ductal carcinoma. The results from the present study revealed that the transcription factor interleukin enhancer-binding factor 2 (ILF2) was highly expressed in non-small cell lung cancer (NSCLC) cell lines compared with in normal cell lines. ChIP and luciferase reporter gene assays demonstrated that ILF2 inhibited the expression level of the tumor suppressor gene phosphatase and tensin homolog (PTEN) by directly binding to its upstream regulatory region. Furthermore, the results from the detection of cell adhesion and apoptosis in cell suspension culture demonstrated that this mechanism enabled NSCLC cells to reduce adherence to the matrix and to survive in this abnormal state. These results suggested that ILF2 may promote the anchorage-independence of NSCLC cells through the suppression of PTEN.

9.
J Biomed Nanotechnol ; 14(2): 312-320, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-31352927

RESUMO

Combination therapy with different functional drugs has been proved to be an effective strategy for cancer treatments by achieving cooperative therapeutic efficacy and reducing drug side effects. In this study, a novel combination anticancer system was designed by coating silver nanoparticles in the poly(aspartic acid) (PAsp) shell of polymer micelle via the in situ reduction of silver ions and encapsulating doxorubicin (DOX) into the micellar poly(ε-caprolactone) (PCL) core through hydrophobic interaction. This combination system not only avoided aggregation of the silver nanoparticles but also improved the bioavailability of DOX. As a result, it exhibited enhanced antitumor activity against HepG2 tumor cells compared with single silver-coated or DOX-loaded nanoparticles because of the combined anticancer effects of nanosilver and DOX. Therefore, this combination therapeutic system could be a potential candidate for enhanced anticancer therapy.


Assuntos
Nanopartículas Metálicas , Doxorrubicina , Portadores de Fármacos , Células Hep G2 , Humanos , Micelas , Prata
10.
ACS Appl Mater Interfaces ; 9(15): 13016-13028, 2017 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-28378992

RESUMO

Chemo-photodynamic combination therapy has been received widespread attention in cancer treatment due to its excellent characteristics, such as reducing the adverse side effects of chemo-drugs and improving the therapeutic effects for various cancers. In this study, RGD and DOX was conjugated to PEG by thiol-ene addition and Schiff's base reaction, respectively, to prepare the targeted and pH-sensitive antitumor prodrug nanoparticles (RGD-PEG-DOX NPs, RGD-NPs). Subsequently, the photosensitizer chlorin e6 (Ce6) was encapsulated into RGD-NPs, thus obtaining a simple and efficient chemo-photodynamic combination platform (RGD-PEG-DOX/Ce6 NPs, RGD-NPs/Ce6). This nanoparticle possessed high drug loading property of both the chemo-drug and photosensitizer and could simultaneously release them under the mild acidic microenvironment of cancer cells, which was expected to realize the synchronization therapy of chemotherapy and photodynamic therapy (PDT). Compared with free DOX and Ce6, RGD-NPs/Ce6 could significantly improve the cellular uptake capacities of DOX and Ce6, resulting in the increased contents of ROS in cancer cells and effective cytotoxicity for tumor cells (MDA-MB-231 cells and MCF-7 cells) upon a laser radiation. The in vivo experiment showed that RGD-NPs/Ce6 displayed superior tumor targeting, accumulation, and retention ability than the other groups (free DOX, free Ce6 and NPs/Ce6), and thus significantly enhancing the antitumor effect in vivo with a laser radiation. In addition, the cardiotoxicity induced by DOX was thoroughly wiped out after being loaded and delivered by the nanoparticles according to the pathological analysis. Therefore, the targeted chemo-photodynamic combination therapeutic platform may be a promising candidate for enhanced cancer therapy.


Assuntos
Nanopartículas , Linhagem Celular Tumoral , Humanos , Neoplasias , Fotoquimioterapia , Fármacos Fotossensibilizantes , Pró-Fármacos
11.
Int J Nanomedicine ; 11: 1119-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27051287

RESUMO

Although the shortcomings of small molecular antitumor drugs were efficiently improved by being entrapped into nanosized vehicles, premature drug release and insufficient tumor targeting demand innovative approaches that boost the stability and tumor responsiveness of drug-loaded nanocarriers. Here, we show the use of the core cross-linking method to generate a micelle with enhanced drug encapsulation ability and sensitivity of drug release in tumor. This kind of micelle could increase curcumin (Cur) delivery to HeLa cells in vitro and improve tumor accumulation in vivo. We designed and synthesized the core cross-linked micelle (CCM) with polyethylene glycol and folic acid-polyethylene glycol as the hydrophilic units, pyridyldisulfide as the cross-linkable and hydrophobic unit, and disulfide bond as the cross-linker. CCM showed spherical shape with a diameter of 91.2 nm by the characterization of dynamic light scattering and transmission electron microscope. Attributed to the core cross-linking, drug-loaded CCM displayed higher Nile Red or Cur-encapsulated stability and better sensitivity to glutathione than noncross-linked micelle (NCM). Cellular uptake and in vitro antitumor studies proved the enhanced endocytosis and better cytotoxicity of CCM-Cur against HeLa cells, which had a high level of glutathione. Meanwhile, the folate receptor-mediated drug delivery (FA-CCM-Cur) further enhanced the endocytosis and cytotoxicity. Ex vivo imaging studies showed that CCM-Cur and FA-CCM-Cur possessed higher tumor accumulation until 24 hours after injection. Concretely, FA-CCM-Cur exhibited the highest tumor accumulation with 1.7-fold of noncross-linked micelle Cur and 2.8-fold of free Cur. By combining cross-linking of the core with active tumor targeting of FA, we demonstrated a new and effective way to design nanocarriers for enhanced drug encapsulation, smart tumor responsiveness, and elevated tumor accumulation.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Curcumina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Ácido Fólico/química , Micelas , Animais , Antineoplásicos/química , Reagentes de Ligações Cruzadas/química , Curcumina/química , Dissulfetos/química , Estabilidade de Medicamentos , Endocitose/efeitos dos fármacos , Células HeLa/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos Endogâmicos BALB C , Polietilenoglicóis/química , Distribuição Tecidual
12.
ACS Appl Mater Interfaces ; 8(17): 10726-36, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27058429

RESUMO

Finding a smart cancer drug delivery carrier with long blood circulation, enhanced cancer targeting, and quick drug release in tumors is critical for efficient cancer chemotherapy. Herein, we design a cRGD-polycarboxybetaine methacrylate-b-polybenzimidazole methacrylate (cRGD-PCB-b-PBBMZ) copolymer to self-assemble into smart drug-loaded nanoparticles (cRGD-PCM NPs) which can target αvß3 integrin overexpressed cancer tissue by cRGD peptide unit and release drug quickly in cancer cells by protonation of benzimidazole groups. The outer PCB layer can resist protein adhesion, and there are only about 10% of proteins in mouse serum adhered to the surface of PCM NPs. With the pKa value of 5.08 of the benzimidazole units, DOX can be released from NPs in pH 5.0 PBS. cRGD-PCM NPs can bring more DOX into HepG2 cells than nontargeting PCM NPs, and there has high DOX release rate in HepG2 cells because of the protonation of benzimidazole groups in endosome and lysosome. MTT assay verifies that higher cellular uptake of DOX causes higher cytotoxicity. Furthermore, the results of ex vivo imaging studies confirm that cRGD-PCM/DOX NPs can successfully deliver DOX into tumor tissue from the injection site. Therefore, the multifunctional cRGD-PCM NPs show great potential as novel nanocarriers for targeting cancer chemotherapy.


Assuntos
Neoplasias , Animais , Benzimidazóis , Doxorrubicina , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Nanopartículas
13.
Sci Rep ; 6: 21225, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26876480

RESUMO

Ample attention has focused on cancer drug delivery via prodrug nanoparticles due to their high drug loading property and comparatively lower side effects. In this study, we designed a PEG-DOX-Cur prodrug nanoparticle for simultaneous delivery of doxorubicin (DOX) and curcumin (Cur) as a combination therapy to treat cancer. DOX was conjugated to PEG by Schiff's base reaction. The obtained prodrug conjugate could self-assemble in water at pH 7.4 into nanoparticles (PEG-DOX NPs) and encapsulate Cur into the core through hydrophobic interaction (PEG-DOX-Cur NPs). When the PEG-DOX-Cur NPs are internalized by tumor cells, the Schiff's base linker between PEG and DOX would break in the acidic environment that is often observed in tumors, causing disassembling of the PEG-DOX-Cur NPs and releasing both DOX and Cur into the nuclei and cytoplasma of the tumor cells, respectively. Compared with free DOX, free Cur, free DOX-Cur combination, or PEG-DOX NPs, PEG-DOX-Cur NPs exhibited higher anti-tumor activity in vitro. In addition, the PEG-DOX-Cur NPs also showed prolonged blood circulation time, elevated local drug accumulation and increased tumor penetration. Enhanced anti-tumor activity was also observed from the PEG-DOX-Cur-treated animals, demonstrating better tumor inhibitory property of the NPs. Thus, the PEG-DOX-Cur prodrug nanoparticle system provides a simple yet efficient approach of drug delivery for chemotherapy.


Assuntos
Curcumina/administração & dosagem , Doxorrubicina/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/química , Linhagem Celular Tumoral , Curcumina/química , Doxorrubicina/química , Sistemas de Liberação de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Nanopartículas/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química
14.
Nanoscale ; 7(10): 4451-60, 2015 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-25679795

RESUMO

The key to developing more nanocarriers for the delivery of drugs in clinical applications is to consider the route of the carrier from the administration site to the target tissue and to look for a simple design to complete this whole journey. We synthesized the amphiphilic copolymer cRGDfK-poly(ethylene glycol)-b-poly(2,4,6-trimethoxybenzylidene-1,1,1-tris(hydroxymethyl) ethane methacrylate) (cRGD-PETM) to construct multifunctional micelles. These micelles combined enhanced drug-loading efficiency with tumor-targeting properties, visual detection and controllable intracellular drug release, resulting in an improved chemotherapeutic effect in vivo. Doxorubicin (DOX) was encapsulated within the cRGD-PETM micelles as a model drug (termed as cRGD-PETM/DOX Ms). The size and morphology of the micelles were characterized systematically. As a result of the hydrophobic interaction and the π-π conjugation between the DOX molecules and the PTTMA copolymers, the cRGD-PETM/DOX Ms showed an excellent drug-loading capacity. The results of in vitro drug-release studies indicated that the cumulative release of DOX from cRGD-PETM/DOX Ms at pH 5.0 was twice that at pH 7.4. The results of fluorescent microscopic analysis showed that the cRGD-PETM/DOX Ms could be internalized by 4T1 and HepG2 cells via receptor-mediated endocytosis with rapid intracellular drug release, which resulted in increased cytotoxicity compared with free DOX. Ex vivo imaging studies showed that the cRGD-PETM/DOX Ms improved the accumulation and retention of the drug in tumor tissues. Studies of the in vivo anticancer effects showed that the cRGD-PETM/DOX Ms had a significantly higher therapeutic efficacy with lower side-effects than free DOX and PETM/DOX Ms. These results show that the multifunctional cRGD-PETM/DOX Ms have great potential as vehicles for the delivery of hydrophobic anticancer drugs.


Assuntos
Antibióticos Antineoplásicos , Doxorrubicina , Sistemas de Liberação de Medicamentos/métodos , Integrinas/agonistas , Proteínas de Neoplasias/agonistas , Neoplasias/tratamento farmacológico , Animais , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/química , Doxorrubicina/farmacologia , Endocitose/efeitos dos fármacos , Células Hep G2 , Humanos , Integrinas/metabolismo , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Propilenoglicóis
15.
ACS Appl Mater Interfaces ; 7(4): 2735-44, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25555064

RESUMO

Self-assembling peptide nanofibers (including naturally L-amino acid-based and unnaturally D-amino acid-based ones) have been widely utilized in biomedical research. However, there has been no systematic study on their in vivo stability, distribution, and toxicity. Herein we systematically study the in vivo dynamic biostability, biodistribution, and toxicity of supramolecular nanofibers formed by Nap-GFFYGRGD (L-amino acid-based, L-fibers) and Nap-G(D)F(D)F(D)YGRGD (D-amino acid-based, D-fibers), respectively. The D-fibers have better in vitro and in vivo biostabilities than L-fibers. It is found that D-fibers keep a good integrity in plasma during 24 h, while half of l-fibers are digested upon incubation in plasma for 6 h. The biodistributions of L- and D-fibers are also studied using the iodine-125 radiolabeling technique. The results reveal that L-fibers mainly accumulate in stomach, whereas d-fibers preferentially distribute in liver. Successive administrations of both L- and D-fibers with the dose of 30 mg/kg/dose cause no significant inflammation, liver and kidney function damages, immune reaction, and dysfunction of hematopoietic system. This study will provide fundamental guidelines for utilization of self-assembling peptide-based supramolecular nanomaterials in biomedical applications, such as drug delivery, bioimaging, and regenerative medicine.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Nanofibras/química , Peptídeos/metabolismo , Aminoácidos/química , Animais , Feminino , Mucosa Gástrica/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Nanofibras/toxicidade , Peptídeos/química , Peptídeos/toxicidade , Distribuição Tecidual
16.
Biomacromolecules ; 15(10): 3634-42, 2014 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-25308336

RESUMO

Exploring ideal nanocarriers for drug delivery systems has encountered unavoidable hurdles, especially the conflict between enhanced cellular uptake and prolonged blood circulation, which have determined the final efficacy of cancer therapy. Here, based on controlled self-assembly, surface structure variation in response to external environment was constructed toward overcoming the conflict. A novel micelle with mixed shell of hydrophilic poly(ethylene glycol) PEG and pH responsive hydrophobic poly(ß-amino ester) (PAE) was designed through the self-assembly of diblock amphiphilic copolymers. To avoid the accelerated clearance from blood circulation caused by the surface exposed targeting group c(RGDfK), here c(RGDfK) was conjugated to the hydrophobic PAE and hidden in the shell of PEG at pH 7.4. At tumor pH, charge conversion occurred, and c(RGDfK) stretched out of the shell, leading to facilitated cellular internalization according to the HepG2 cell uptake experiments. Meanwhile, the heterogeneous surface structure endowed the micelle with prolonged blood circulation. With the self-regulated multifunctional collaborated properties of enhanced cellular uptake and prolonged blood circulation, successful inhibition of tumor growth was achieved from the demonstration in a tumor-bearing mice model. This novel nanocarrier could be a promising candidate in future clinical experiments.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Feminino , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Micelas , Polietilenoglicóis/química , Polímeros/química
17.
ACS Appl Mater Interfaces ; 6(8): 5558-65, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24660962

RESUMO

D-peptides, which consist of D-amino acids and can resist the hydrolysis catalyzed by endogenous peptidases, are one of the promising candidates for construction of peptide materials with enhanced biostability in vivo. In this paper, we report on a self-assembling supramolecular nanostructure of D-amino acid-based peptide Nap-G(D)F(D)F(D)YGRGD (D-fiber, (D)F meant D-phenylalanine, (D)Y meant D-tyrosine), which were used as carriers for 10-hydroxycamptothecin (HCPT). Transmission electron microscopy observations demonstrated the filamentous morphology of the HCPT-loaded peptides (d-fiber-HCPT). The better selectivity and antitumor activity of D-fiber-HCPT than L-fiber-HCPT were found in the in vitro and in vivo antitumor studies. These results highlight that this model D-fiber system holds great promise as vehicles of hydrophobic drugs for cancer therapy.


Assuntos
Aminoácidos/química , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/análogos & derivados , Portadores de Fármacos/química , Neoplasias/tratamento farmacológico , Peptídeos/química , Animais , Camptotecina/química , Camptotecina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Humanos , Camundongos , Camundongos Endogâmicos BALB C
18.
Int J Nanomedicine ; 9: 197-207, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24399876

RESUMO

The poor aqueous solubility and low bioavailability of curcumin restrict its clinical application for cancer treatment. In this study, a novel tumor-targeting nanofiber carrier was developed to improve the solubility and tumor-targeting ability of curcumin using a self-assembled Nap-GFFYG-RGD peptide. The morphologies of the peptide nanofiber and the curcumin-encapsulated nanofiber were visualized by transmission electron microscopy. The tumor-targeting activity of the curcumin-encapsulated Nap-GFFYG-RGD peptide nanofiber (f-RGD-Cur) was studied in vitro and in vivo, using Nap-GFFYG-RGE peptide nanofiber (f-RGE-Cur) as the control. Curcumin was encapsulated into the peptide nanofiber, which had a diameter of approximately 10-20 nm. Curcumin showed sustained-release behavior from the nanofibers in vitro. f-RGD-Cur showed much higher cellular uptake in αvß3 integrin-positive HepG2 liver carcinoma cells than did non-targeted f-RGE-Cur, thereby leading to significantly higher cytotoxicity. Ex vivo studies further demonstrated that curcumin could accumulate markedly in mouse tumors after administration of f-RGD-Cur via the tail vein. These results indicate that Nap-GFFYG-RGD peptide self-assembled nanofibers are a promising hydrophobic drug delivery system for targeted treatment of cancer.


Assuntos
Curcumina/administração & dosagem , Nanocápsulas/química , Nanofibras/química , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Oligopeptídeos/farmacocinética , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Curcumina/química , Difusão , Células Hep G2 , Humanos , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Nanocápsulas/administração & dosagem , Nanocápsulas/ultraestrutura , Nanofibras/administração & dosagem , Nanofibras/ultraestrutura , Neoplasias Experimentais/patologia , Oligopeptídeos/química , Ligação Proteica , Resultado do Tratamento
19.
Int J Nanomedicine ; 8: 4229-46, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24235825

RESUMO

Polyethylene glycol (PEG)-ylation is a widely used strategy to fabricate nanocarriers with a long blood circulation time. Further elaboration of the contribution of the surface PEGylation pattern to biodistribution is highly desirable. We fabricated a series of polyion complex (PIC) micelles PEGylated with different ratios (PEG2k and PEG550). The plasma protein adsorption, murine macrophage uptake, and in vivo biodistribution with iodine-125 as the tracer were systematically studied to elucidate the impact of PEGylation patterns on the biodistribution of micelles. We demonstrated that the PEGylated micelles with short hydrophilic PEG chains mixed on the surface were cleared quickly by the reticuloendothelial system (RES), and the single PEG2k PEGylated micelles could efficiently prolong the blood circulation time and increase their deposition in tumor sites. The present study extends the understanding of the PEGylation strategy to further advance the development of ideal nanocarriers for drug delivery and imaging applications.


Assuntos
Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Micelas , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , Adsorção , Análise de Variância , Animais , Proteínas Sanguíneas/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/farmacologia , Células Hep G2 , Humanos , Radioisótopos do Iodo/química , Radioisótopos do Iodo/farmacocinética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Peso Molecular , Células NIH 3T3 , Nanopartículas/química , Tamanho da Partícula , Polietilenoglicóis/farmacologia , Distribuição Tecidual
20.
Biomacromolecules ; 14(2): 460-7, 2013 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-23281663

RESUMO

The miserable targeting performance of nanocarriers for cancer therapy arises largely from the rapid clearance from blood circulation and the major accumulation in the organs of the reticuloendothelial system (RES), leading to inefficient enhanced permeability and retention (EPR) effect after intravenous injection (i.v.). Herein, we reported an efficient method to prolong the blood circulation of nanoparticles and decrease their deposition in liver and spleen. In this work, we fabricated a series of mixed shell micelles (MSMs) with approximately the same size, charge and core composition but with varied hydrophilic/hydrophobic ratios in the shell through spontaneously self-assembly of block copolymers poly(ethylene glycol)-block-poly(l-lysine) (PEG-b-PLys) and poly(N-isopropylacrylamide)-block-poly(aspartic acid) (PNIPAM-b-PAsp) in aqueous medium. The effect of the surface heterogeneity on the in vivo biodistribution was systematically investigated through in vivo tracking of the (125)I-labeled MSMs determined by Gamma counter. Compared with single PEGylated micelles, some MSMs were proved to be significantly efficient with more than 3 times lower accumulation in liver and spleen and about 6 times higher concentration in blood at 1 h after i.v.. The results provide us a novel strategy for future development of long-circulating nanocarriers for efficient cancer therapy.


Assuntos
Portadores de Fármacos , Nanopartículas , Polímeros/farmacologia , Distribuição Tecidual/efeitos dos fármacos , Transporte Biológico , Sobrevivência Celular/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas , Fígado/metabolismo , Micelas , Sistema Fagocitário Mononuclear/metabolismo , Neoplasias/tratamento farmacológico , Baço/metabolismo , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA