Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 360: 496-513, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37423524

RESUMO

CRISPR/Cas9-based genome editing is promising for therapy of cervical cancer by precisely targeting human papillomavirus (HPV). To develop CRISPR/Cas9-based genome editing nanotherapies, a pH-responsive hybrid nonviral nanovector was constructed for co-delivering Cas9 mRNA and guide RNAs (gRNAs) targeting E6 or E7 oncogenes. The pH-responsive nanovector was fabricated using an acetalated cyclic oligosaccharide (ACD), in combination with low molecular weight polyethyleneimine. Thus obtained hybrid ACD nanoparticles (defined as ACD NP) showed efficient loading for both Cas9 mRNA and E6 or E7 gRNA, giving rise to two pH-responsive genome editing nanotherapies E6/ACD NP and E7/ACD NP, respectively. Cellularly, ACD NP exhibited high transfection but low cytotoxicity in HeLa cervical carcinoma cells. Also, efficient genome editing of target genes was achieved in HeLa cells, with minimal off-target effects. In mice bearing HeLa xenografts, treatment with E6/ACD NP or E7/ACD NP afforded effective editing of target oncogenes and considerable antitumor activities. More importantly, treatment with E6/ACD NP or E7/ACD NP notably promoted CD8+ T cell survival by reversing the immunosuppressive microenvironment, thereby leading to synergistic antitumor effects by combination therapy using the gene editing nanotherapies and adoptive T-cell transfer. Consequently, our pH-responsive genome editing nanotherapies deserve further development for the treatment of HPV-associated cervical cancer, and they can also serve as promising nanotherapies to improve efficacies of other immune therapies against different advanced cancers by regulating the immunosuppressive tumor microenvironment.


Assuntos
Infecções por Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Camundongos , Animais , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/terapia , Neoplasias do Colo do Útero/patologia , Edição de Genes , Células HeLa , RNA Mensageiro/genética , Imunossupressores , Terapia Baseada em Transplante de Células e Tecidos , Proteínas E7 de Papillomavirus/genética , Microambiente Tumoral
2.
Nanotechnology ; 32(39)2021 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-34153956

RESUMO

Currently, a single treatment is less effective for triple-negative breast cancer (TNBC) therapy. Additionally, there are some limitations to the use of siRNA alone as a new method to treat breast cancer, such as its effective delivery into cells. In this study, we proposed a strategy that combines a siRNA-loaded DNA nanostructure and genistein for TNBC therapy. Both CD36 siRNA-loaded self-assembled DNA nanoprisms (NP-siCD36) and genistein knocked down CD36, resulting in enhanced anticancer efficacy through phosphorylation of the p38 MAPK pathway.In vitrostudies showed that combination therapy could effectively enhance cell apoptosis and reduce cell proliferation, achieving an antitumor effect in TNBC cells. The current study suggests that NP-siCD36 combined with genistein might be a promising strategy for breast cancer and treatment.


Assuntos
Antineoplásicos , Antígenos CD36/genética , Nanoestruturas/química , RNA Interferente Pequeno/genética , Neoplasias de Mama Triplo Negativas , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antígenos CD36/metabolismo , Linhagem Celular Tumoral , DNA/química , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Feminino , Genisteína/metabolismo , Genisteína/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Adv Mater ; 31(46): e1904607, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31583783

RESUMO

Inflammation is a common cause of many acute and chronic inflammatory diseases. A major limitation of existing anti-inflammatory therapeutics is that they cannot simultaneously regulate pro-inflammatory cytokine production, oxidative stress, and recruitment of neutrophils and macrophages. To overcome this limitation, nanoparticles (NPs) with multiple pharmacological activities are synthesized, using a chemically modified cyclic oligosaccharide. The manufacture of this type of bioactive, saccharide material-based NPs (defined as LCD NP) is straightforward, cost-effective, and scalable. Functionally, LCD NP effectively inhibits inflammatory response, oxidative stress, and cell migration for both neutrophils and macrophages, two major players of inflammation. Therapeutically, LCD NP shows desirable efficacies for the treatment of acute and chronic inflammatory diseases in mouse models of peritonitis, acute lung injury, and atherosclerosis. Mechanistically, the therapeutic benefits of LCD NP are achieved by inhibiting neutrophil-mediated inflammatory macrophage recruitment and by preventing subsequent pro-inflammatory events. In addition, LCD NP shows good safety profile in a mouse model. Thus, LCD NP can serve as an effective anti-inflammatory nanotherapy for the treatment of inflammatory diseases mainly associated with neutrophil and macrophage infiltration.


Assuntos
Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Nanopartículas/química , beta-Ciclodextrinas/química , beta-Ciclodextrinas/farmacologia , Doença Aguda , Animais , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/uso terapêutico , Transporte Biológico , Doença Crônica , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , beta-Ciclodextrinas/metabolismo , beta-Ciclodextrinas/uso terapêutico
4.
Theranostics ; 9(13): 3732-3753, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281510

RESUMO

Colitis-associated colon cancer (CAC) is a widely recognized cancer, while treatment with the existing chemotherapeutic drugs affords limited clinical benefits. Herein we proposed a site-specific, combination nanotherapy strategy for targeted treatment of CAC by the oral route. Methods: A reactive oxygen species (ROS)-responsive and hydrogen peroxide-eliminating material OCD was synthesized, which was further produced into a functional nanoparticle (OCD NP). The antioxidative stress and anti-inflammatory effects of OCD NP were examined by in vitro and in vivo experiments. By packaging an anticancer drug camptothecin-11 (CPT-11) into OCD NP, a ROS-responsive nanotherapy CPT-11/OCD NP was obtained, and its antitumor activity was evaluated by both in vitro and in vivo studies. Preliminary safety studies were also performed for CPT-11/OCD NP in mice. Results: OCD NP significantly attenuated oxidative stress and inhibited inflammatory response in different cells and mice with induced colitis. CPT-11/OCD NP could selectively release drug molecules under intestinal pH conditions and at high levels of ROS. In C26 murine colon carcinoma cells, this nanotherapy showed significantly higher antitumor activity compared to free CPT-11 and a non-responsive CPT-11 nanotherapy. Correspondingly, oral delivery of CPT-11/OCD NP notably inhibited tumorigenesis and tumor growth in mice with induced CAC. By combination therapy with the nanovehicle OCD NP in the inflammatory phase, more desirable therapeutic effects were achieved. Furthermore, CPT-11/OCD NP displayed excellent safety profile for oral administration at a dose that is 87.3-fold higher than that employed in therapeutic studies. Conclusions: Anticancer nanotherapies derived from intrinsic anti-inflammatory nanocarriers are promising for targeted combination treatment of inflammation-associated tumors by simultaneously shaping pro-inflammatory microenvironment toward a relatively normal niche sensitive to chemotherapy.


Assuntos
Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Nanopartículas Multifuncionais/química , Microambiente Tumoral , Administração Oral , Animais , Anti-Inflamatórios/farmacologia , Camptotecina/efeitos adversos , Camptotecina/farmacocinética , Camptotecina/farmacologia , Camptotecina/uso terapêutico , Linhagem Celular Tumoral , Colite/complicações , Neoplasias do Colo/etiologia , Progressão da Doença , Liberação Controlada de Fármacos , Endocitose/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/química , Hidrólise , Inflamação/patologia , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Distribuição Tecidual/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
5.
Sci Adv ; 5(1): eaat2953, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30662940

RESUMO

Nanoparticles have been extensively used for inflammation imaging and photodynamic therapy of cancer. However, the major translational barriers to most nanoparticle-based imaging and therapy applications are the limited depth of tissue penetration, inevitable requirement of external irradiation, and poor biocompatibility of the nanoparticles. To overcome these critical limitations, we synthesized a sensitive, specific, biodegradable luminescent nanoparticle that is self-assembled from an amphiphilic polymeric conjugate with a luminescent donor (luminol) and a fluorescent acceptor [chlorin e6 (Ce6)] for in vivo luminescence imaging and photodynamic therapy in deep tissues. Mechanistically, reactive oxygen species (ROS) and myeloperoxidase generated in inflammatory sites or the tumor microenvironment trigger bioluminescence resonance energy transfer and the production of singlet oxygen (1O2) from the nanoparticle, enabling in vivo imaging and cancer therapy, respectively. This self-illuminating nanoparticle shows an excellent in vivo imaging capability with suitable tissue penetration and resolution in diverse animal models of inflammation. It is also proven to be a selective, potent, and safe antitumor nanomedicine that specifically kills cancer cells via in situ 1O2 produced in the tumor microenvironment, which contains a high level of ROS.


Assuntos
Antineoplásicos/uso terapêutico , Substâncias Luminescentes/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/terapia , Fotoquimioterapia/métodos , Nanomedicina Teranóstica/métodos , Células A549 , Animais , Clorofilídeos , Humanos , Inflamação/diagnóstico por imagem , Luminescência , Luminol/química , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Nanopartículas/química , Polímeros/química , Porfirinas/química , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
ACS Nano ; 12(9): 8943-8960, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30114351

RESUMO

Atherosclerosis is a leading cause of vascular diseases worldwide. Whereas antioxidative therapy has been considered promising for the treatment of atherosclerosis in view of a critical role of reactive oxygen species (ROS) in the pathogenesis of atherosclerosis, currently available antioxidants showed considerably limited clinical outcomes. Herein, we hypothesize that a broad-spectrum ROS-scavenging nanoparticle can serve as an effective therapy for atherosclerosis, taking advantage of its antioxidative stress activity and targeting effects. As a proof of concept, a broad-spectrum ROS-eliminating material was synthesized by covalently conjugating a superoxide dismutase mimetic agent Tempol and a hydrogen-peroxide-eliminating compound of phenylboronic acid pinacol ester onto a cyclic polysaccharide ß-cyclodextrin (abbreviated as TPCD). TPCD could be easily processed into a nanoparticle (TPCD NP). The obtained nanotherapy TPCD NP could be efficiently and rapidly internalized by macrophages and vascular smooth muscle cells (VSMCs). TPCD NPs significantly attenuated ROS-induced inflammation and cell apoptosis in macrophages, by eliminating overproduced intracellular ROS. Also, TPCD NPs effectively inhibited foam cell formation in macrophages and VSMCs by decreasing internalization of oxidized low-density lipoprotein. After intravenous (i.v.) administration, TPCD NPs accumulated in atherosclerotic lesions of apolipoprotein E-deficient (ApoE-/-) mice by passive targeting through the dysfunctional endothelium and translocation via inflammatory cells. TPCD NPs significantly inhibited the development of atherosclerosis in ApoE-/- mice after i.v. delivery. More importantly, therapy with TPCD NPs afforded stabilized plaques with less cholesterol crystals, a smaller necrotic core, thicker fibrous cap, and lower macrophages and matrix metalloproteinase-9, compared with those treated with control drugs previously developed for antiatherosclerosis. The therapeutic benefits of TPCD NPs mainly resulted from reduced systemic and local oxidative stress and inflammation as well as decreased inflammatory cell infiltration in atherosclerotic plaques. Preliminary in vivo tests implied that TPCD NPs were safe after long-term treatment via i.v. injection. Consequently, TPCD NPs can be developed as a potential antiatherosclerotic nanotherapy.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aterosclerose/tratamento farmacológico , Nanopartículas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Anti-Inflamatórios não Esteroides/química , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Apoptose/efeitos dos fármacos , Aterosclerose/metabolismo , Ácidos Borônicos/química , Ácidos Borônicos/farmacologia , Óxidos N-Cíclicos/química , Óxidos N-Cíclicos/farmacologia , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nanopartículas/química , Polissacarídeos/química , Polissacarídeos/farmacologia , Marcadores de Spin , beta-Ciclodextrinas/química , beta-Ciclodextrinas/farmacologia
7.
Nano Lett ; 17(2): 1056-1064, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28075596

RESUMO

Targeting of nanoparticles to distant diseased sites after oral delivery remains highly challenging due to the existence of many biological barriers in the gastrointestinal tract. Here we report targeted oral delivery of diverse nanoparticles in multiple disease models, via a "Trojan horse" strategy based on a bioinspired yeast capsule (YC). Diverse charged nanoprobes including quantum dots (QDs), iron oxide nanoparticles (IONPs), and assembled organic fluorescent nanoparticles can be effectively loaded into YC through electrostatic force-driven spontaneous deposition, resulting in different diagnostic YC assemblies. Also, different positive nanotherapies containing an anti-inflammatory drug indomethacin (IND) or an antitumor drug paclitaxel (PTX) are efficiently packaged into YC. YCs containing either nanoprobes or nanotherapies may be rapidly endocytosed by macrophages and maintained in cells for a relatively long period of time. Post oral administration, nanoparticles packaged in YC are first transcytosed by M cells and sequentially endocytosed by macrophages, then transported to neighboring lymphoid tissues, and finally delivered to remote diseased sites of inflammation or tumor in mice or rats, all through the natural route of macrophage activation, recruitment, and deployment. For the examined acute inflammation model, the targeting efficiency of YC-delivered QDs or IONPs is even higher than that of control nanoprobes administered at the same dose via intravenous injection. Assembled IND or PTX nanotherapies orally delivered via YCs exhibit remarkably potentiated efficacies as compared to nanotherapies alone in animal models of inflammation and tumor, which is consistent with the targeting effect and enhanced accumulation of drug molecules at diseased sites. Consequently, through the intricate transportation route, nanoprobes or nanotherapies enveloped in YC can be preferentially delivered to desired targets, affording remarkably improved efficacies for the treatment of multiple diseases associated with inflammation.


Assuntos
Anti-Inflamatórios/administração & dosagem , Antineoplásicos/administração & dosagem , Portadores de Fármacos/química , Indometacina/química , Nanopartículas/química , Paclitaxel/química , Saccharomyces cerevisiae/química , Administração Oral , Animais , Anti-Inflamatórios/química , Antineoplásicos/química , Cápsulas , Carcinoma 256 de Walker/diagnóstico por imagem , Carcinoma 256 de Walker/tratamento farmacológico , Linhagem Celular , Liberação Controlada de Fármacos , Humanos , Indometacina/administração & dosagem , Inflamação/diagnóstico por imagem , Inflamação/tratamento farmacológico , Camundongos Endogâmicos BALB C , Paclitaxel/administração & dosagem , Ratos Sprague-Dawley , Eletricidade Estática
8.
Int J Mol Sci ; 17(7)2016 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-27399689

RESUMO

BACKGROUND: Atherosclerosis is a fundamental pathological process responded to some serious cardiovascular events. Although the cholesterol-lowering drugs are widely prescribed for atherosclerosis therapy, it is still the leading cause of death in the developed world. Here we measured the effects of compound K in atherosclerosis formation and investigated the probably mechanisms of the anti-antherosclerosis roles of compound K. METHODS: We treated the atherosclerotic model animals (apoE(-/-) mice on western diet) with compound K and measured the size of atherosclerotic lesions, inflammatory cytokine levels and serum lipid profile. Peritoneal macrophages were collected in vitro for the foam cell and inflammasome experiments. RESULTS: Our results show that treatment with compound K dose-dependently attenuates the formation of atherosclerotic plaques by 55% through activation of reverse cholesterol transport pathway, reduction of systemic inflammatory cytokines and inhibition of local inflammasome activity. Compound K increases the cholesterol efflux of macrophage-derived foam cells, and reduces the inflammasome activity in cholesterol crystal stimulated macrophages. The activation of LXRα may contribute to the athero-protective effects of compound K. CONCLUSION: These observations provide evidence for an athero-protective effect of compound K via LXRα activation, and support its further evaluation as a potential effective modulator for the prevention and treatment of atherosclerosis.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/tratamento farmacológico , Ginsenosídeos/uso terapêutico , Receptores X do Fígado/metabolismo , Animais , Aterosclerose/patologia , Caspase 1/metabolismo , Colesterol/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Fígado Gorduroso/tratamento farmacológico , Células Espumosas/metabolismo , Células Espumosas/patologia , Ginsenosídeos/farmacologia , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fator de Transcrição RelA/metabolismo , Triglicerídeos/sangue
9.
Guang Pu Xue Yu Guang Pu Fen Xi ; 36(10): 3114-8, 2016 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-30222254

RESUMO

In this paper, an Nd∶YAG laser with 10ns pulse width and output wavelength of 1 064 nm was employed to ablate Gd metal target and Gd-doped glass target for plasma generation. The out-of-band (OOB) radiation of extreme ultraviolet sources with the two target configurations was comparatively studied. It has been found that the continuous radiation emitted by the plasma is the main component of the out-of-band radiation. The spectral distribution of the continuum emission matches that of blackbody radiation with a temperature of about 5 eV. And it is also found that the intensity of OOB radiation can be considerably decreased by using Gd-doped glass target. Optical Emission Spectroscopy (OES) has been used to analyze the temporal and spatial behaviors of electron temperature (Te) and density (Ne) of the Gd-doped glass target plasma, and experimental results show that temporal evolution of electron temperature and density of the plasma are found to be decayed exponentially with the increasing of delay time. At 125 ns after laser irradiation, electron temperature and density were 4 eV and 1.2×1018 cm-3 respectively, and then decreased to 1.5 eV and 8×1017 cm-3 with delaying time of 250 ns. On the other hand, spatial evolution of electron temperature and density show that both of them first increase and then decrease in the region of 1~10 mm from the target surface. The electron temperature and electron density achieves the maximum of 2.6 eV and 8.5×1017 cm-3, respectively, when the probe location away from the target surface 6 mm.

10.
Acta Biomater ; 10(6): 2630-42, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24486911

RESUMO

There is still unmet demand for developing powerful approaches to produce polymeric nanoplatforms with versatile functions and broad applications, which are essential for the successful bench-to-bedside translation of polymeric nanotherapeutics developed in the laboratory. We have discovered a facile, convenient, cost-effective and easily scalable one-pot strategy to assemble various lipophilic therapeutics bearing carboxyl groups into nanomedicines, through which highly effective cargo loading and nanoparticle formation can be achieved simultaneously. Besides dramatically improving water solubility, the assembled nanopharmaceuticals showed significantly higher bioavailability and much better therapeutic activity. These one-pot assemblies may also serve as nanocontainers to effectively accommodate other highly hydrophobic drugs such as paclitaxel (PTX). PTX nanomedicines thus formulated display strikingly enhanced in vitro antitumor activity and can reverse the multidrug resistance of tumor cells to PTX therapy. The special surface chemistry offers these assembled entities the additional capability of efficiently packaging and efficaciously transfecting plasmid DNA, with a transfection efficiency markedly higher than that of commonly used positive controls. Consequently, this one-pot assembly approach provides a facile route to multifunctional nanoplatforms for simultaneous delivery of multiple therapeutics with improved therapeutic significance.


Assuntos
Sistemas de Liberação de Medicamentos , Nanotecnologia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
11.
Biomaterials ; 34(21): 5344-58, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23591391

RESUMO

Engineering of pH-responsive nanoplatforms can be facilely achieved from acetalated α-cyclodextrin materials. The hydrolysis period of nanoparticles can be precisely tailored by using materials with various acetal types that can be easily controlled by acetalation time. These nanomaterials with pH-modulated hydrolysis and pH-triggered drug delivery capability show good biocompatibility in vitro and in vivo. Incorporation of anticancer drug paclitaxel (PTX) into newly developed pH-sensitive nanosystems leads to nanotherapeutics with significantly improved cytotoxic activity against various tumor cells. Importantly, thus formulated nanomedicines can reverse the multidrug resistance of PTX-resistant cancer cells. In vivo antitumor studies also reveal the superior of pH-sensitive nanosystems over pristine PTX and pH-insensitive PLGA nanoformulations. Moreover, comparison with other two acid-labile materials evidenced the advantages of cyclodextrin-based nanovehicles with respect to drug loading capacity, in vitro and in vivo activity as well as alleviated adverse effects. These pH-responsive nanoparticles may serve as new generation nanocarriers for drug delivery.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Paclitaxel/farmacologia , alfa-Ciclodextrinas/química , Acetilação/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Portadores de Fármacos/química , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Testes de Toxicidade Aguda , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Biomaterials ; 34(16): 4159-4172, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23480956

RESUMO

The absence of safe, efficient, cost-effective, and easily scalable delivery platforms is one of the most significant hurdles and critical issues that limit the bench to bedside translation of oligonucleotides-based therapeutics. Acid-labile materials are of special interest in developing nonviral vectors due to their capability of intracellularly delivering therapeutic payload. In this study, a nanovector was designed by integrating a pH-responsive cyclodextrin material and low molecular weight polyethylenimine (PEI). Antisense oligonucleotide (ASON) Bcl-xl could be encapsulated into this hybrid nanosystem with extremely high loading efficiency by a nanoemulsion technique. The developed pH-responsive ASON nanotherapeutics could be efficiently transfected into human lung adenocarcinoma cells in a time- and dose-dependent manner, resulting in effective cell growth inhibition, significant suppression on the expression of Bcl-xl mRNA/protein, and efficient cell apoptosis. Importantly, the new nanovector showed drastically higher efficacy and lower cytotoxicity when compared with PLGA-based counterpart and commonly used cationic vectors like branched PEI (25,000 Da) and Lipofectamine 2000. This pH-responsive hybrid nanosystem may serve as a safe and efficient nonviral vector that may find wide applications in gene therapy.


Assuntos
Ciclodextrinas/química , Técnicas de Transferência de Genes , Nanopartículas/química , Acetilação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Ácido Láctico/química , Microscopia Confocal , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Oligonucleotídeos Antissenso/farmacologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção , Vírus/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
13.
Nanomedicine ; 9(3): 408-18, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22922571

RESUMO

The assembly of homostructured polypeptides bearing various side groups into well-defined nanostructures was presented, with their size and topology mainly dominated by the chemical structure and molecular weight of peptides. Pharmacokinetic and pharmacodynamic studies based on rat models suggested these newly constructed nanoassemblies with low cytotoxicity may function as novel nanoplatforms to efficiently and safely deliver therapeutics to achieve better efficacy but lower side effects. Other applications in biomedical fields, such as biotechnology, medical imaging, and tissue engineering, may also be expected. FROM THE CLINICAL EDITOR: This research team investigated the assembly of homostructured polypeptides bearing various side groups into well-defined nanostructures, and demonstrated low cytotoxicity in rat disease models, suggesting that these novel nanoplatforms may safely and efficiently deliver therapeutics with low side effects.


Assuntos
Sistemas de Liberação de Medicamentos , Nanoestruturas/química , Nanotecnologia , Peptídeos/química , Administração Oral , Animais , Morte Celular/efeitos dos fármacos , Células HeLa , Humanos , Indometacina/administração & dosagem , Indometacina/farmacocinética , Indometacina/farmacologia , Cinética , Masculino , Camundongos , Nanoestruturas/toxicidade , Nanoestruturas/ultraestrutura , Peptídeos/síntese química , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA