Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Appl Mater Interfaces ; 14(36): 40599-40611, 2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36052562

RESUMO

Amyloidosis pathologically proceeds via production of amyloidogenic proteins by organs, formation of protein aggregates through structural changes, and their deposition on tissues. A growing body of evidence demonstrates that amyloidosis generally develops through three critical pathological steps: (1) production of amyloid precursor proteins, (2) amyloid formation, and (3) amyloid deposition. However, no clinically effective therapy that is capable of targeting each pathological step of amyloidosis independently is currently available. Here, we combined therapeutic effects and developed a short hairpin RNA expression vector (shRNA) complex with a cyclodextrin-appended cationic dendrimer (CDE) as a novel multitarget therapeutic drug that is capable of simultaneously suppressing these three steps. We evaluated its therapeutic effects on systemic transthyretin (ATTR) amyloidosis and Alzheimer's disease (AD) as localized amyloidosis, by targeting TTR and amyloid ß, respectively. CDE/shRNA exhibited RNAi effects to suppress amyloid protein production and also achieved both inhibition of amyloid formation and disruption of existing amyloid fibrils. The multitarget therapeutic effects of CDE/shRNA were confirmed by evaluating TTR deposition reduction in early- and late-onset human ATTR amyloidosis model rats and amyloid ß deposition reduction in AppNL-G-F/NL-G-F AD model mice. Thus, the CDE/shRNA complex exhibits multifunctional therapeutic efficacy and may reveal novel strategies for establishing curative treatments for both systemic and localized amyloidosis.


Assuntos
Doença de Alzheimer , Amiloidose , Ciclodextrinas , Dendrímeros , Doença de Alzheimer/tratamento farmacológico , Amiloide , Peptídeos beta-Amiloides , Proteínas Amiloidogênicas , Amiloidose/tratamento farmacológico , Amiloidose/metabolismo , Animais , Ciclodextrinas/farmacologia , Dendrímeros/farmacologia , Humanos , Camundongos , RNA Interferente Pequeno , Ratos
2.
ACS Nano ; 14(2): 1665-1681, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-31922724

RESUMO

Despite the common knowledge that the reticuloendothelial system is largely responsible for blood clearance of systemically administered nanoparticles, the sequestration mechanism remains a "black box". Using transgenic zebrafish embryos with cell type-specific fluorescent reporters and fluorescently labeled model nanoparticles (70 nm SiO2), we here demonstrate simultaneous three-color in vivo imaging of intravenously injected nanoparticles, macrophages, and scavenger endothelial cells (SECs). The trafficking processes were further revealed at ultrastructural resolution by transmission electron microscopy. We also find, using a correlative light-electron microscopy approach, that macrophages rapidly sequester nanoparticles via membrane adhesion and endocytosis (including macropinocytosis) within minutes after injection. In contrast, SECs trap single nanoparticles via scavenger receptor-mediated endocytosis, resulting in gradual sequestration with a time scale of hours. Inhibition of the scavenger receptors prevented SECs from accumulating nanoparticles but enhanced uptake in macrophages, indicating the competitive nature of nanoparticle clearance in vivo. To directly quantify the relative contributions of the two cell types to overall nanoparticle sequestration, the differential sequestration kinetics was studied within the first 30 min post-injection. This revealed a much higher and increasing relative contribution of SECs, as they by far outnumber macrophages in zebrafish embryos, suggesting the importance of the macrophage:SECs ratio in a given tissue. Further characterizing macrophages on their efficiency in nanoparticle clearance, we show that inflammatory stimuli diminish the uptake of nanoparticles per cell. Our study demonstrates the strength of transgenic zebrafish embryos for intravital real-time and ultrastructural imaging of nanomaterials that may provide mechanistic insights into nanoparticle clearance in rodent models and humans.


Assuntos
Células Endoteliais/química , Macrófagos/química , Nanopartículas/metabolismo , Dióxido de Silício/metabolismo , Animais , Células Endoteliais/metabolismo , Cinética , Macrófagos/metabolismo , Nanopartículas/química , Tamanho da Partícula , Dióxido de Silício/química , Propriedades de Superfície , Fatores de Tempo , Peixe-Zebra/embriologia
3.
Biol Pharm Bull ; 42(10): 1679-1688, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31582656

RESUMO

Targeted drug delivery system (DDS) is required for RNA interference (RNAi) therapy to increase the therapeutic effect and to reduce the adverse effect. Especially in transthyretin (TTR)-related amyloidosis, hepatocyte specific delivery is desired because TTR mainly expresses in hepatocyte. Herein, we report on a hepatocyte-specific small interfering RNA (siRNA) delivery system using polyethylene glycol (PEG)-modified lactosylated dendrimer (generation 3; G3) conjugates with α-cyclodextrin (PEG-LαCs (G3)) for TTR-related amyloidosis therapy, and investigated the in vitro and in vivo gene silencing effect of PEG-LαCs (G3)/siRNA polyplexes. PEG-LαC (G3, average degree of substitution of PEG (DSP) 2)/TTR siRNA (siTTR) polyplex exhibited the asialoglycoprotein receptor (ASGPR)-mediated cellular uptake, high endosomal escaping ability and localization of the siRNA in cytoplasm, resulting in significant TTR silencing in HepG2 cells. In vivo studies showed that PEG-LαC (G3, DSP2)/siTTR polyplex led to a significant TTR silencing effect in liver after systemic administration to mice. Furthermore, safety evaluation revealed that PEG-LαC (G3, DSP2)/siTTR polyplex had no significant toxicity both in vitro and in vivo. These findings suggest the utility of PEG-LαC (G3, DSP2) as a promising hepatocyte-specific siRNA delivery system both in vitro and in vivo, and as a therapeutic approach for TTR-related amyloidosis.


Assuntos
Neuropatias Amiloides Familiares/tratamento farmacológico , Ciclodextrinas/administração & dosagem , Dendrímeros/administração & dosagem , Hepatócitos/metabolismo , Polietilenoglicóis/administração & dosagem , Pré-Albumina/genética , RNA Interferente Pequeno/administração & dosagem , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/metabolismo , Animais , Dendrímeros/farmacocinética , Células Hep G2 , Humanos , Masculino , Camundongos Endogâmicos BALB C , Polietilenoglicóis/farmacocinética , Pré-Albumina/metabolismo , RNA Interferente Pequeno/farmacocinética
4.
Biol Pharm Bull ; 41(12): 1737-1744, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30504675

RESUMO

Transthyretin (TTR) amyloidosis, also known as transthyretin-related familial amyloidotic polyneuropathy (ATTR-FAP), is a fatal hereditary systemic amyloidosis caused by mutant forms of TTR. Although conventional treatments for ATTR-FAP, such as liver transplantation (LT) and TTR tetramer stabilizer, reportedly halt the progression of clinical manifestation, these therapies have several limitations. Oligonucleotide-based therapy, e.g. small interfering RNA (siRNA)- and antisense oligonucleotides (ASOs)-based therapy, hold enormous potential for the treatment of intractable diseases such as ATTR-FAP, by specifically regulating the gene responsible for the disease. Clinical evidence strongly suggests that LT inhibits mutant TTR production, thus improving the manifestation of ATTR-FAP. Therefore, an oligonucleotide-based therapy for ATTR-FAP, which reduces the production of TTR by the liver, has recently been developed in preclinical and clinical studies. This review focuses on recent advances in oligonucleotide-based therapy and future prospects of next-generation oligonucleotide-based drugs for therapeutic use against ATTR-FAP.


Assuntos
Neuropatias Amiloides Familiares/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Pré-Albumina/genética , RNA Interferente Pequeno/uso terapêutico , Neuropatias Amiloides Familiares/genética , Animais , Ensaios Clínicos como Assunto , Edição de Genes/métodos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Mutação , Oligonucleotídeos Antissenso/administração & dosagem , Pré-Albumina/biossíntese , RNA Interferente Pequeno/administração & dosagem
5.
Int J Biol Macromol ; 107(Pt A): 1113-1121, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28964842

RESUMO

In this study, we newly developed the ternary complexes consisting of lactosylated dendrimer (generation 3)/α-cyclodextrin conjugate (Lac-α-CDE), siRNA and the anionic polysaccharide sacrans, and evaluated their utility as siRNA transfer carriers. Three kinds of the low-molecular-weight sacrans, i.e. sacran (100) (Mw 44,889Da), sacran (1000) (Mw 943,692Da) and sacran (10,000) (Mw 1,488,281Da) were used. Lac-α-CDE/siRNA/sacran ternary complexes were prepared by adding the low-molecular-weight sacrans to the Lac-α-CDE/siRNA binary complex solution. Cellular uptake of the ternary complex with sacran (100) was higher than that of the binary complex or the other ternary complexes with sacran (1000) and sacran (10,000) in HepG2 cells. Additionally, the ternary complex possessed high serum resistance and endosomal escaping ability in HepG2 cells. High liver levels of siRNA and Lac-α-CDE were observed after the intravenous administration of the ternary complex rather than that of the binary complex. Moreover, intravenous administration of the ternary complex (siRNA 5mg/kg) induced the significant RNAi effect in the liver of mice with negligible change of blood chemistry values. Therefore, a ternary complexation of the Lac-α-CDE/siRNA binary complex with sacran is useful as a hepatocyte-specific siRNA delivery system.


Assuntos
Ciclodextrinas/química , Dendrímeros/química , Polissacarídeos/química , RNA Interferente Pequeno/química , Animais , Ciclodextrinas/genética , Dendrímeros/farmacologia , Portadores de Fármacos , Técnicas de Transferência de Genes , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Lactose/química , Camundongos , Polissacarídeos/genética , Polissacarídeos/farmacologia , RNA Interferente Pequeno/genética , Fatores de Complexo Ternário/química , Fatores de Complexo Ternário/genética
6.
Nat Commun ; 8: 13991, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28071661

RESUMO

Formation of organ-specific vasculatures requires cross-talk between developing tissue and specialized endothelial cells. Here we show how developing zebrafish spinal cord neurons coordinate vessel growth through balancing of neuron-derived Vegfaa, with neuronal sFlt1 restricting Vegfaa-Kdrl mediated angiogenesis at the neurovascular interface. Neuron-specific loss of flt1 or increased neuronal vegfaa expression promotes angiogenesis and peri-neural tube vascular network formation. Combining loss of neuronal flt1 with gain of vegfaa promotes sprout invasion into the neural tube. On loss of neuronal flt1, ectopic sprouts emanate from veins involving special angiogenic cell behaviours including nuclear positioning and a molecular signature distinct from primary arterial or secondary venous sprouting. Manipulation of arteriovenous identity or Notch signalling established that ectopic sprouting in flt1 mutants requires venous endothelium. Conceptually, our data suggest that spinal cord vascularization proceeds from veins involving two-tiered regulation of neuronal sFlt1 and Vegfaa via a novel sprouting mode.


Assuntos
Neurônios/fisiologia , Medula Espinal/embriologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Veias/embriologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Biomarcadores/metabolismo , Embrião não Mamífero/citologia , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Mutação , Neovascularização Fisiológica , Receptores Notch/genética , Receptores Notch/metabolismo , Medula Espinal/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Veias/metabolismo , Proteínas de Peixe-Zebra/genética
7.
Amyloid ; 22(3): 181-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26204452

RESUMO

Localized insulin-derived amyloid masses occasionally form at the site of repeated insulin injections in patients with insulin-dependent diabetes and cause subcutaneous insulin resistance. Various kinds of insulin including porcine insulin, human insulin, and insulin analogues reportedly formed amyloid fibrils in vitro and in vivo, but the impact of the amino acid replacement in insulin molecules on amyloidogenicity is largely unknown. In the present study, we demonstrated the difference in amyloid fibril formation kinetics of human insulin and insulin analogues, which suggests an important role of the C-terminal domain of the insulin B chain in nuclear formation of amyloid fibrils. Furthermore, we determined that cyclodextrins, which are widely used as drug carriers in the pharmaceutical field, had an inhibitory effect on the nuclear formation of insulin amyloid fibrils. These findings have significant implications for the mechanism underlying insulin amyloid fibril formation and for developing optimal additives to prevent this subcutaneous adverse effect.


Assuntos
Amiloide/antagonistas & inibidores , Ciclodextrinas/química , Insulina Aspart/química , Insulina Detemir/química , Insulina/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Amiloide/química , Benzotiazóis , Corantes Fluorescentes , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Recombinantes/química , Soluções , Espectrometria de Fluorescência , Tiazóis
8.
J Drug Target ; 22(10): 883-90, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25051178

RESUMO

We previously reported that 6-O-α-(4-O-α-D-glucuronyl)-D-glucosyl-ß-cyclodextrin (GUG-ß-CyD) conjugate with polyamidoamine dendrimer (dendrimer, generation 2; G2) (GUG-ß-CDE (G2)) is useful as a gene transfer carrier. In the present study, to investigate the potentials of GUG-ß-CDE (G2) as a siRNA carrier, we evaluated the RNAi effect of its complex with siRNA against transthyretin (TTR) mRNA (siTTR) for the treatment of familial amyloidotic polyneuropathy (FAP). Among the various GUG-ß-CDEs (G2) having the different degrees of substitution of GUG-ß-CyD (degree of substation (DS) 1.8, 2.5, 3.0 and 5.0), GUG-ß-CDE (G2, DS 1.8) showed the highest siTTR transfer activity. GUG-ß-CDE (G2, DS 1.8)/siTTR complex showed no cytotoxicity in HepG2 cells. After intravenous administration of GUG-ß-CDE (G2, DS 1.8)/siTTR complex to BALB/c mice, TTR mRNA expression was tended to reduce with negligible change of blood chemistry data. Particle size, ζ-potential and cellular association of the GUG-ß-CDE (G2, DS 1.8) complex were almost the same as those of the other CDEs complexes. Meanwhile, GUG-ß-CDE (G2, DS 1.8)/siTTR complex showed high endosomal escaping ability of siTTR in cytoplasm. These findings suggest the potential of GUG-ß-CDE (G2, DS 1.8) as a siRNA carrier for the FAP treatment.


Assuntos
Neuropatias Amiloides Familiares/terapia , Dendrímeros/química , Oligossacarídeos/química , Poliaminas/química , RNA Interferente Pequeno/administração & dosagem , Administração Intravenosa , Neuropatias Amiloides Familiares/genética , Animais , Endossomos/metabolismo , Técnicas de Transferência de Genes , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Tamanho da Partícula , Pré-Albumina/genética , RNA Mensageiro/metabolismo
9.
Toxicol Lett ; 222(1): 55-63, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23872614

RESUMO

Investigation of the genotoxic potential of nanomaterials is essential to evaluate if they pose a cancer risk for exposed workers and consumers. The Chinese hamster ovary cell line CHO-K1 is recommended by the OECD for use in the micronucleus assay and is commonly used for genotoxicity testing. However, studies investigating if this cell line is suitable for the genotoxic evaluation of nanomaterials, including induction of DNA adduct and micronuclei formation, are rare and for silver nanoparticles (Ag NPs) missing. Therefore, we here systematically investigated DNA and chromosomal damage induced by BSA coated Ag NPs (15.9±7.6 nm) in CHO-K1 cells in relation to cellular uptake and intracellular localization, their effects on mitochondrial activity and production of reactive oxygen species (ROS), cell cycle, apoptosis and necrosis. Ag NPs are taken up by CHO-K1 cells and are presumably translocated into endosomes/lysosomes. Our cytotoxicity studies demonstrated a concentration-dependent decrease of mitochondrial activity and increase of intracellular reactive oxygen species (ROS) in CHO-K1 cells following exposure to Ag NPs and Ag⁺ (0-20 µg/ml) for 24h. Annexin V/propidium iodide assay showed that Ag NPs and Ag⁺ induced apoptosis and necrosis, which is in agreement with an increased fraction of cells in subG1 phase of the cell cycle. Genotoxicity studies showed that Ag NPs but also silver ions (Ag⁺) induced bulky-DNA adducts, 8-oxodG and micronuclei formation in a concentration-dependent manner, however, there were quantitative and qualitative differences between the particulate and ionic form of silver. Taken together, our multi-platform genotoxicity and cytotoxicity analysis demonstrates that CHO-K1 cells are suitable for the investigation of genotoxicity of nanoparticles like Ag NPs.


Assuntos
Testes de Mutagenicidade/métodos , Mutagênicos , Nanopartículas/toxicidade , Prata/toxicidade , 8-Hidroxi-2'-Desoxiguanosina , Animais , Apoptose/efeitos dos fármacos , Células CHO , Ciclo Celular/efeitos dos fármacos , Corantes , Cricetinae , Cricetulus , Adutos de DNA , Desoxiguanosina/análogos & derivados , Citometria de Fluxo , Espectrometria de Massas , Testes para Micronúcleos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Nanopartículas/administração & dosagem , Necrose , Espécies Reativas de Oxigênio/metabolismo , Prata/administração & dosagem , Sais de Tetrazólio , Tiazóis
10.
J Drug Target ; 21(5): 487-96, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23570609

RESUMO

To develop a novel hepatocyte-selective gene carrier, we prepared polyamidoamine starburst dendrimer (generation 3, G3) conjugates with three functional molecules, i.e. α-cyclodextrin, polyethylene glycol (PEG, molecular weight = 2170) and lactose (PEG-LαCs), and evaluated gene delivery efficiency of these conjugates in vitro and in vivo. PEG-LαC (G3, degrees of substitution of the PEG moiety (DSP) 2.1) showed higher gene transfer activity than other PEG-LαCs (G3, DSP4.0, 6.2) in HepG2 cells, expressing asialoglycoprotein receptor, and the activity decreased in HeLa cells, non-expressing the receptor and in the presence of asialofetuin. High gene transfer activity of PEG-LαC (G3, DSP2.1) was retained even in the presence of 50% serum, although the activity of α-cyclodextrin/lactosylated dendrimer (G3) conjugate (Lac-α-CDE (G3)), which is lacking a PEG moiety, was severely decreased in the presence of 20% serum. PEG-LαC (G3, DSP2.1) provided negligible cytotoxicity up to a charge ratio of 50 (carrier/pDNA) in HepG2 cells and less acute organ toxicity. PEG-LαC (G3, DSP2.1) showed selective gene transfer activity to hepatic parenchymal cells rather than hepatic non-parenchymal cells. These results suggest that PEG-LαC (G3, DSP2.1) is useful as a hepatocyte-selective gene carrier in vitro and in vivo.


Assuntos
Dendrímeros/administração & dosagem , Hepatócitos/metabolismo , Lactose/administração & dosagem , Poliaminas/administração & dosagem , Polietilenoglicóis/administração & dosagem , alfa-Ciclodextrinas/administração & dosagem , Animais , Receptor de Asialoglicoproteína/genética , Assialoglicoproteínas/administração & dosagem , Linhagem Celular Tumoral , Dendrímeros/química , Dendrímeros/farmacocinética , Fetuínas/administração & dosagem , Técnicas de Transferência de Genes , Terapia Genética/métodos , Células HeLa , Células Hep G2 , Humanos , Lactose/química , Lactose/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Poliaminas/química , Poliaminas/farmacocinética , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , alfa-Ciclodextrinas/química , alfa-Ciclodextrinas/farmacocinética
11.
Toxicol Sci ; 130(1): 145-57, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22831968

RESUMO

The toxic effects of silver nanoparticles (AgNPs) on cells are well established, but only limited studies on the effect of AgNPs and silver ions on the cellular transcriptome have been performed. In this study, the effect of AgNPs on the gene expression in the human lung epithelial cell line A549 exposed to 12.1 µg/ml AgNPs (EC20) for 24 and 48h was compared with the response to control and silver ion (Ag(+)) treated cells (1.3 µg/ml) using microarray analysis. Twenty-four hours to AgNP altered the regulation of more than 1000 genes (more than twofold regulation), whereas considerably fewer genes responded to Ag(+) (133 genes). The upregulated genes included members of the metallothionein, heat shock protein, and histone families. As expected from the induction of meta l lothionein and heat shock protein genes, Ag(+) and AgNP treatment resulted in intracellular production of reactive oxygen species but did not induce apoptosis or necrosis at the concentrations used in this study. In addition, the exposure to AgNPs influenced the cell cycle and led to an arrest in the G2/M phase as shown by cell cycle studies by flow cytometry and microscopy. In conclusion, although the transcriptional response to Ag(+) exposure was highly related to the response caused by AgNPs, our findings suggest that AgNPs, due to their particulate form, affect exposed cells in a more complex way.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Estudo de Associação Genômica Ampla , Nanopartículas Metálicas/toxicidade , Mucosa Respiratória/efeitos dos fármacos , Nitrato de Prata/toxicidade , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares , Nanopartículas Metálicas/ultraestrutura , Necrose/induzido quimicamente , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho da Partícula , Análise de Componente Principal , Espécies Reativas de Oxigênio/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
12.
Amyloid ; 19 Suppl 1: 47-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22519861

RESUMO

RNA interference (RNAi) is a sequence-specific gene-silencing mechanism triggered by double-stranded RNA and powerful tools for a gene function study and RNAi therapy. Although siRNAs offer several advantages as potential new drugs to treat various diseases, the efficient delivery system of siRNAs in vivo remains a crucial challenge for achieving the desired RNAi effect in clinical development. In particular, when considering the siRNA therapeutics for familial amyloidotic polyneuropathy (FAP) caused by the deposition of variant transthyretin (TTR) in various organs, hepatocyte-selective siRNA delivery is desired because TTR is predominantly synthesized by hepatocytes. In this study, to reveal the potential use of lactosylated dendrimer (G3)/α-cyclodextrin conjugate (Lac-α-CDE (G3)) as novel hepatocyte-selective siRNA carriers in order to treat FAP, we evaluated the RNAi effect of siRNA complex with Lac-α-CDE (G3) both in vitro and in vivo.


Assuntos
Neuropatias Amiloides Familiares/terapia , Dendrímeros/química , Hepatócitos/metabolismo , Pré-Albumina/genética , RNA Interferente Pequeno/genética , alfa-Ciclodextrinas/química , Animais , Humanos , Interferência de RNA
13.
Mol Pharm ; 9(6): 1645-53, 2012 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-22510029

RESUMO

To reveal the potential use of lactosylated-dendrimer (G3) conjugates with α-cyclodextrin (Lac-α-CDE (G3)) as novel hepatocyte-specific siRNA carriers in order to treat transthyretin (TTR)-related familial amyloidotic polyneuropathy (FAP), we evaluated the RNAi effect of siRNA complexes with Lac-α-CDE (G3) both in vitro and in vivo. Herein, we targeted TTR gene expression because TTR-related FAP was often caused by amyloidogenic TTR (ATTR), which mainly expresses in hepatocytes. Lac-α-CDE (G3, average degree of substitution of lactose (DSL) 1.2)/siRNA complex had a potent RNAi effect against TTR gene expression through adequate physicochemical properties, asialoglycoprotein receptor (ASGP-R)-mediated cellular uptake, efficient endosomal escape and the delivery of the siRNA complex to cytoplasm, but not nucleus, with negligible cytotoxicity. Lac-α-CDE (G3, DSL 1.2)/siRNA complex had the potential to induce the in vivo RNAi effect after intravenous administration in the liver of mice. The blood chemistry values in the α-CDE (G3) and Lac-α-CDE (G3, DSL 1.2) systems were almost equivalent to those in the control system (5% mannitol solution). Taken together, these results suggest that Lac-α-CDE (G3, DSL 1.2) has the potential for a novel hepatocyte-selective siRNA carrier in vitro and in vivo, and has a possibility as a therapeutic tool for FAP to the liver transplantation.


Assuntos
Neuropatias Amiloides Familiares/terapia , Dendrímeros/química , Vetores Genéticos/química , Hepatócitos/metabolismo , Lactose/química , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , alfa-Ciclodextrinas/química , Neuropatias Amiloides Familiares/metabolismo , Animais , Western Blotting , Citometria de Fluxo , Vetores Genéticos/administração & dosagem , Células Hep G2 , Humanos , Masculino , Camundongos , Microscopia de Fluorescência , Pré-Albumina/genética , Pré-Albumina/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
14.
Toxicol Lett ; 208(3): 286-92, 2012 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-22101214

RESUMO

The toxicity of silver nanoparticles (AgNPs) has been shown in many publications. Here we investigated to which degree the silver ion fraction of AgNP suspensions, contribute to the toxicity of AgNPs in A549 lung cells. Cell viability assays revealed that AgNP suspensions were more toxic when the initial silver ion fraction was higher. At 1.5µg/ml total silver, A549 cells exposed to an AgNP suspension containing 39% silver ion fraction showed a cell viability of 92%, whereas cells exposed to an AgNP suspension containing 69% silver ion fraction had a cell viability of 54% as measured by the MTT assay. In addition, at initial silver ion fractions of 5.5% and above, AgNP-free supernatant had the same toxicity as AgNP suspensions. Flow-cytometric analyses of cell cycle and apoptosis confirmed that there is no significant difference between the treatment with AgNP suspension and AgNP supernatant. Only AgNP suspensions with silver ion fraction of 2.6% or less were significantly more toxic than their supernatant as measured by MTT assays. From our data we conclude that at high silver ion fractions (≥5.5%) the AgNPs did not add measurable additional toxicity to the AgNP suspension, whereas at low silver ion fractions (≤2.6%) AgNP suspensions are more toxic than their supernatant.


Assuntos
Nanopartículas Metálicas/toxicidade , Prata/toxicidade , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Formazans/química , Humanos , Nanopartículas Metálicas/ultraestrutura , Microscopia Eletrônica de Transmissão , Prata/química , Espectrofotometria Atômica , Sais de Tetrazólio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA