Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Anal Chem ; 89(10): 5357-5363, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28345878

RESUMO

Extracellular vesicles (EVs), including exosomes, are circulating nanoscale particles heavily implicated in cell signaling and can be isolated in vast numbers from human biofluids. Study of their molecular profiling and materials properties is currently underway for purposes of describing a variety of biological functions and diseases. However, the large, and as yet largely unquantified, variety of EV subpopulations differing in composition, size, and likely function necessitates characterization schemes capable of measuring single vesicles. Here we describe the first application of multispectral optical tweezers (MS-OTs) to single vesicles for molecular fingerprinting of EV subpopulations. This versatile imaging platform allows for sensitive measurement of Raman chemical composition (e.g., variation in protein, lipid, cholesterol, nucleic acids), coupled with discrimination by fluorescence markers. For exosomes isolated by ultracentrifugation, we use MS-OTs to interrogate the CD9-positive subpopulations via antibody fluorescence labeling and Raman spectra measurement. We report that the CD9-positive exosome subset exhibits reduced component concentration per vesicle and reduced chemical heterogeneity compared to the total purified EV population. We observed that specific vesicle subpopulations are present across exosomes isolated from cell culture supernatant of several clonal varieties of mesenchymal stromal cells and also from plasma and ascites isolated from human ovarian cancer patients.


Assuntos
Exossomos/metabolismo , Pinças Ópticas , Tetraspanina 29/análise , Animais , Anticorpos/imunologia , Feminino , Corantes Fluorescentes/química , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Nanopartículas/química , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Análise de Componente Principal , Ratos , Análise Espectral Raman , Tetraspanina 29/imunologia
2.
Adv Biosyst ; 1(5)2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-29911169

RESUMO

All cells expel a variety of nano-sized extracellular vesicles (EVs), including exosomes, with composition reflecting the cells' biological state. Cancer pathology is dramatically mediated by EV trafficking via key proteins, lipids, metabolites, and microRNAs. Recent proteomics evidence suggests that tumor-associated exosomes exhibit distinct expression of certain membrane proteins, rendering those proteins as attractive targets for diagnostic or therapeutic application. Yet, it is not currently feasible to distinguish circulating EVs in complex biofluids according to their tissue of origin or state of disease. Here we demonstrate peptide binding to tumor-associated EVs via overexpressed membrane protein. We find that SKOV-3 ovarian tumor cells and their released EVs express α3ß1 integrin, which can be targeted by our in-house cyclic nonapeptide, LXY30. After measuring bulk SKOV-3 EV association with LXY30 by flow cytometry, Raman spectral analysis of laser-trapped single exosomes with LXY30-dialkyne conjugate enabled us to differentiate cancer-associated exosomes from non-cancer exosomes. Furthermore, we introduce the foundation for a highly specific detection platform for tumor-EVs in solution with biosensor surface-immobilized LXY30. LXY30 not only exhibits high specificity and affinity to α3ß1 integrin-expressing EVs, but also reduces EV uptake into SKOV-3 parent cells, demonstrating the possibility for therapeutic application.

3.
J Extracell Vesicles ; 4: 28533, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26649679

RESUMO

Current analysis of exosomes focuses primarily on bulk analysis, where exosome-to-exosome variability cannot be assessed. In this study, we used Raman spectroscopy to study the chemical composition of single exosomes. We measured spectra of individual exosomes from 8 cell lines. Cell-line-averaged spectra varied considerably, reflecting the variation in total exosomal protein, lipid, genetic, and cytosolic content. Unexpectedly, single exosomes isolated from the same cell type also exhibited high spectral variability. Subsequent spectral analysis revealed clustering of single exosomes into 4 distinct groups that were not cell-line specific. Each group contained exosomes from multiple cell lines, and most cell lines had exosomes in multiple groups. The differences between these groups are related to chemical differences primarily due to differing membrane composition. Through a principal components analysis, we identified that the major sources of spectral variation among the exosomes were in cholesterol content, relative expression of phospholipids to cholesterol, and surface protein expression. For example, exosomes derived from cancerous versus non-cancerous cell lines can be largely separated based on their relative expression of cholesterol and phospholipids. We are the first to indicate that exosome subpopulations are shared among cell types, suggesting distributed exosome functionality. The origins of these differences are likely related to the specific role of extracellular vesicle subpopulations in both normal cell function and carcinogenesis, and they may provide diagnostic potential at the single exosome level.

4.
PLoS One ; 10(12): e0141655, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26657215

RESUMO

BACKGROUND: HCV replication in persistently infected cell culture remains resistant to IFN-α/RBV combination treatment, whereas IFN-λ1 induces viral clearance. The antiviral mechanisms by which IFN-λ1 induces sustained HCV clearance have not been determined. AIM: To investigate the mechanisms by which IFN-λ clears HCV replication in an HCV cell culture model. METHODS: IFN-α sensitive (S3-GFP) and resistant (R4-GFP) cells were treated with equivalent concentrations of either IFN-α or IFN-λ. The relative antiviral effects of IFN-α and IFN-λ1 were compared by measuring the HCV replication, quantification of HCV-GFP expression by flow cytometry, and viral RNA levels by real time RT-PCR. Activation of Jak-Stat signaling, interferon stimulated gene (ISG) expression, and miRNA-122 transcription in S3-GFP and R4-GFP cells were examined. RESULTS: We have shown that IFN-λ1 induces HCV clearance in IFN-α resistant and sensitive replicon cell lines in a dose dependent manner through Jak-Stat signaling, and induces STAT 1 and STAT 2 activation, ISRE-luciferase promoter activation and ISG expression. Stat 3 activation is also involved in IFN-λ1 induced antiviral activity in HCV cell culture. IFN-λ1 induced Stat 3 phosphorylation reduces the expression of hepatocyte nuclear factor 4 alpha (HNF4α) through miR-24 in R4-GFP cells. Reduced expression of HNF4α is associated with decreased expression of miR-122 resulting in an anti-HCV effect. Northern blot analysis confirms that IFN-λ1 reduces miR-122 levels in R4-GFP cells. Our results indicate that IFN-λ1 activates the Stat 3-HNF4α feedback inflammatory loop to inhibit miR-122 transcription in HCV cell culture. CONCLUSIONS: In addition to the classical Jak-Stat antiviral signaling pathway, IFN-λ1 inhibits HCV replication through the suppression of miRNA-122 transcription via an inflammatory Stat 3-HNF4α feedback loop. Inflammatory feedback circuits activated by IFNs during chronic inflammation expose non-responders to the risk of hepatocellular carcinoma.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Fator 4 Nuclear de Hepatócito/genética , Interleucinas/farmacologia , MicroRNAs/antagonistas & inibidores , RNA Viral/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Hepacivirus/genética , Hepacivirus/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/virologia , Interações Hospedeiro-Patógeno , Humanos , Interferon-alfa/farmacologia , Interferons , Janus Quinase 1/genética , Janus Quinase 1/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fosforilação , RNA Viral/biossíntese , Ribavirina/farmacologia , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Replicação Viral/efeitos dos fármacos
5.
Nanoscale ; 7(20): 9290-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25939587

RESUMO

Thin silver film coated nanobowl Surface Enhanced Raman Spectroscopy (SERS) substrates are used to capture exosomes in solution for SERS measurements that can provide biochemical analysis of intact and ruptured exosomes. Exosomes derived via Total Exosome Isolation Reagent (TEIR) as well as ultracentrifugation (UC) from the SKOV3 cell line were analyzed. Spectra of exosomes derived via TEIR are dominated by a signal characteristic for the TEIR kit that needs to be subtracted for all measurements. Differences in SERS spectra recorded at different times during the drying of the exosome solution are statistically analyzed with Principal Component Analysis (PCA). At the beginning of the drying process, SERS spectra of exosomes exhibit peaks characteristic for both lipids and proteins. Later on during the drying process, new SERS peaks develop, suggesting that the initially intact exosome ruptures over time. This time-dependent evolution of SERS peaks enables analysis of exosomal membrane contents and the contents inside the exosomes.


Assuntos
Exossomos/química , Ressonância de Plasmônio de Superfície , Linhagem Celular , Humanos
6.
PLoS One ; 9(9): e108616, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25265476

RESUMO

PURPOSE: Chronic Hepatitis C Virus (HCV)-infected patients with liver cirrhosis (LC) respond poorly to interferon-alpha (IFN-α) and ribavirin (RBV) combination therapy, but the reason for this is unclear. We previously reported that HCV-infection induces endoplasmic reticulum (ER) stress and autophagy response that selectively down regulates the type I IFN-α receptor-1 (IFNAR1) and RBV transporters (CNT1 and ENT1), leading to IFN-α/RBV resistance. The goal of this study is to verify whether an increase in ER stress and autophagy response is also associated with the reduced expression of IFNAR1 and RBV transporters in chronic HCV-infected patients. METHODS: Primary human hepatocytes (PHH) were infected with cell culture grown HCV particles (JFH-ΔV3-Rluc). HCV replication was confirmed by the detection of viral RNA by RT-qPCR and HCV-core protein by Western blotting. The ER stress and autophagy response and expression of IFN receptors and RBV transporters in HCV infected PHH and liver tissues derived from patients were measured by Western blotting. RESULT: HCV infection of PHH showed impaired expression of IFNAR1, IFNγR1 (Type II IFN receptor) and RBV transporters but not IL10Rß (Type III IFN-λ receptor). ER stress markers (BiP, IRE1α and peIF2α) and autophagy response (LC3II, Beclin 1 and ATG5) were induced in HCV infected chronic liver disease (CLD) and LC patients. Liver biopsies (CLD) show a 50% reduced expression of IFNAR1 and RBV transporters. Furthermore, the expression of IFNAR1 and RBV transporters was impaired in almost all LC patients. CONCLUSION: HCV infection induces ER stress and autophagy response in infected PHH and chronically infected liver tissues. The expression of IFNAR1, IFNγR1 and RBV transporters were significantly impaired in CLD and cirrhotic livers. Our study provides a potential explanation for the reduced response rate of IFN-α and RBV combination therapy in HCV infected patients with liver cirrhosis.


Assuntos
Hepacivirus/fisiologia , Hepatite C Crônica/metabolismo , Hepatite C Crônica/virologia , Cirrose Hepática/metabolismo , Cirrose Hepática/virologia , Receptor de Interferon alfa e beta/metabolismo , Receptores de Interferon/metabolismo , Autofagia , Transporte Biológico , Biópsia , Células Cultivadas , Regulação para Baixo , Estresse do Retículo Endoplasmático , Etanol/farmacologia , Ácidos Graxos não Esterificados/farmacologia , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/patologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Fígado/patologia , Fígado/virologia , Cirrose Hepática/patologia , Ribavirina , Replicação Viral
7.
PLoS One ; 8(8): e72791, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24009705

RESUMO

PURPOSE: Although chronic hepatitis C virus (HCV) infection has been treated with the combination of interferon alpha (IFN-α) and ribavirin (RBV) for over a decade, the mechanism of antiviral synergy is not well understood. We aimed to determine the synergistic antiviral mechanisms of IFN-α and RBV combination treatment using HCV cell culture. METHODS: The antiviral efficacy of IFN-α, RBV alone and in combination was quantitatively measured using HCV infected and replicon cell culture. Direct antiviral activity of these two drugs at the level of HCV internal ribosome entry site (IRES) mediated translation in Huh-7 cell culture was investigated. The synergistic antiviral effect of IFN-α and RBV combination treatment was verified using both the CalcuSyn Software and MacSynergy Software. RESULTS: RBV combination with IFN-α efficiently inhibits HCV replication cell culture. Our results demonstrate that IFN-α, interferon lambda (IFN-λ) and RBV each inhibit the expression of HCV IRES-GFP and that they have a minimal effect on the expression of GFP in which the translation is not IRES dependent. The combination treatments of RBV along with IFN-α or IFN-λ were highly synergistic with combination indexes <1. We show that IFN-α treatment induce levels of PKR and eIF2α phosphorylation that prevented ribosome loading of the HCV IRES-GFP mRNA. Silencing of PKR expression in Huh-7 cells prevented the inhibitory effect of IFN-α on HCV IRES-GFP expression. RBV also blocked polyribosome loading of HCV-IRES mRNA through the inhibition of cellular IMPDH activity, and induced PKR and eIF2α phosphorylation. Knockdown of PKR or IMPDH prevented RBV induced HCV IRES-GFP translation. CONCLUSIONS: We demonstrated both IFN-α and RBV inhibit HCV IRES through prevention of polyribosome formation. The combination of IFN-α and RBV treatment synergistically inhibits HCV IRES translation via using two different mechanisms involving PKR activation and depletion of intracellular guanosine pool through inhibition of IMPDH.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Interferons/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/genética , RNA Viral/genética , Ribavirina/farmacologia , Técnicas de Cultura de Células , Linhagem Celular , Sinergismo Farmacológico , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Genoma Viral , Humanos , IMP Desidrogenase/metabolismo , Polirribossomos/metabolismo , Ligação Proteica/efeitos dos fármacos , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Replicon , Replicação Viral/efeitos dos fármacos , eIF-2 Quinase/metabolismo
8.
Virol J ; 9: 143, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22863531

RESUMO

BACKGROUND: Hepatic steatosis is recognized as a major risk factor for liver disease progression and impaired response to interferon based therapy in chronic hepatitis C (CHC) patients. The mechanism of response to interferon-alpha (IFN-α) therapy under the condition of hepatic steatosis is unexplored. We investigated the effect of hepatocellular steatosis on hepatitis C virus (HCV) replication and IFN-α antiviral response in a cell culture model. METHODS: Sub-genomic replicon (S3-GFP) and HCV infected Huh-7.5 cells were cultured with a mixture of saturated (palmitate) and unsaturated (oleate) long-chain free fatty acids (FFA). Intracytoplasmic fat accumulation in these cells was visualized by Nile red staining and electron microscopy then quantified by microfluorometry. The effect of FFA treatment on HCV replication and IFN-α antiviral response was measured by flow cytometric analysis, Renilla luciferase activity, and real-time RT-PCR. RESULTS: FFA treatment induced dose dependent hepatocellular steatosis and lipid droplet accumulation in the HCV replicon cells was confirmed by Nile red staining, microfluorometry, and by electron microscopy. Intracellular fat accumulation supports replication more in the persistently HCV infected culture than in the sub-genomic replicon (S3-GFP) cell line. FFA treatment also partially blocked IFN-α response and viral clearance by reducing the phosphorylation of Stat1 and Stat2 dependent IFN-ß promoter activation. We show that FFA treatment induces endoplasmic reticulum (ER) stress response and down regulates the IFNAR1 chain of the type I IFN receptor leading to defective Jak-Stat signaling and impaired antiviral response. CONCLUSION: These results suggest that intracellular fat accumulation in HCV cell culture induces ER stress, defective Jak-Stat signaling, and attenuates the antiviral response, thus providing an explanation to the clinical observation regarding how hepatocellular steatosis influences IFN-α response in CHC.


Assuntos
Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Ácidos Graxos não Esterificados/toxicidade , Hepacivirus/imunologia , Hepatócitos/imunologia , Hepatócitos/virologia , Interferon-alfa/imunologia , Linhagem Celular , Citofotometria , Ácidos Graxos não Esterificados/análise , Citometria de Fluxo , Hepatócitos/química , Humanos , Luciferases/análise , Microscopia Eletrônica , Oxazinas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Coloração e Rotulagem , Estresse Fisiológico
9.
Mol Ther ; 20(9): 1724-36, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22617108

RESUMO

Sustained antiviral responses of chronic hepatitis C virus (HCV) infection have improved recently by the use of direct-acting antiviral agents along with interferon (IFN)-α and ribavirin. However, the emergence of drug-resistant variants is expected to be a major problem. We describe here a novel combinatorial small interfering RNA (siRNA) nanosome-based antiviral approach to clear HCV infection. Multiple siRNAs targeted to the highly conserved 5'-untranslated region (UTR) of the HCV genome were synthesized and encapsulated into lipid nanoparticles called nanosomes. We show that siRNA can be repeatedly delivered to 100% of cells in culture using nanosomes without toxicity. Six siRNAs dramatically reduced HCV replication in both the replicon and infectious cell culture model. Repeated treatments with two siRNAs were better than a single siRNA treatment in minimizing the development of an escape mutant, resulting in rapid inhibition of viral replication. Systemic administration of combinatorial siRNA-nanosomes is well tolerated in BALB/c mice without liver injury or histological toxicity. As a proof-of-principle, we showed that systemic injections of siRNA nanosomes significantly reduced HCV replication in a liver tumor-xenotransplant mouse model of HCV. Our results indicate that systemic delivery of combinatorial siRNA nanosomes can be used to minimize the development of escape mutants and inhibition of HCV infection.


Assuntos
Hepacivirus/genética , Hepatite C/terapia , Lipossomos/farmacologia , Fígado/virologia , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/genética , RNA Viral/antagonistas & inibidores , Regiões 5' não Traduzidas , Animais , Linhagem Celular Tumoral , Colesterol/química , Ácidos Graxos Monoinsaturados/química , Hepatite C/virologia , Lipossomos/química , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Transplante de Neoplasias , Compostos de Amônio Quaternário/química , RNA Viral/genética , Replicon , Transfecção , Replicação Viral
10.
Am J Pathol ; 180(6): 2490-503, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22521303

RESUMO

MicroRNAs (miRNAs) are short, noncoding RNA molecules that regulate the expression of a number of genes involved in cancer; therefore, they offer great diagnostic and therapeutic targets. We have developed doxorubicin-resistant and -sensitive metastatic human breast cancer cell lines (MDA-MB-231) to study the chemoresistant mechanisms regulated by miRNAs. We found that doxorubicin localized exclusively to the cytoplasm and was unable to reach the nuclei of resistant tumor cells because of the increased nuclear expression of MDR1/P-glycoprotein (P-gp). An miRNA array between doxorubicin-sensitive and -resistant breast cancer cells showed that reduced expression of miR-298 in doxorubicin-resistant human breast cancer cells was associated with increased expression of P-gp. In a transient transfection experiment, miR-298 directly bound to the MDR1 3' untranslated region and regulated the expression of firefly luciferase reporter in a dose-dependent manner. Overexpression of miR-298 down-regulated P-gp expression, increasing nuclear accumulation of doxorubicin and cytotoxicity in doxorubicin-resistant breast cancer cells. Furthermore, down-regulation of miR-298 increased P-gp expression and induced doxorubicin resistance in sensitive breast cancer cells. In summary, these results suggest that miR-298 directly modulates P-gp expression and is associated with the chemoresistant mechanisms of metastatic human breast cancer. Therefore, miR-298 has diagnostic and therapeutic potential for predicting doxorubicin chemoresistance in human breast cancer.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Doxorrubicina/farmacologia , MicroRNAs/fisiologia , Regiões 3' não Traduzidas/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Transporte Biológico/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/fisiologia , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , RNA Neoplásico/fisiologia , Células Tumorais Cultivadas/efeitos dos fármacos
11.
Eur J Pharm Biopharm ; 80(2): 257-67, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22119665

RESUMO

The objective of this study is to develop an effective siRNA delivery system for successful delivery to the liver for the treatment of HCV. Nanosize liposomes (nanosomes) have been prepared using a mixture of cholesterol and DOTAP. A functional siRNA was encapsulated into nanosomes following condensation with protamine sulfate. The delivery of siRNA was optimized in an in vitro cell culture system. The efficacy of the formulations was evaluated by measuring functional gene silencing and cytotoxicity. Encapsulation of siRNA ≥ 7.4 nM resulted in successful delivery of siRNA to nearly 100% of cells. The formulations containing lipid-to-siRNA ratio ≥ 10.56:1 instantly cleared approximately 85% of HCV while maintaining cell viability at about 90%. The formulations were sonicated to further reduce the particle size. The size of these formulations was decreased up to 100 nm. However, there were no significant changes observed in zeta potential, or in siRNA encapsulation and integrity following sonication. The sonicated formulations also showed higher liver hepatocytes deposition and gene silencing properties. This study therefore provides a novel approach of siRNA delivery to liver hepatocytes, which can also be applied to treat HCV in chronic liver diseases.


Assuntos
Inativação Gênica , Fígado/metabolismo , Nanopartículas , RNA Interferente Pequeno/administração & dosagem , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Sobrevivência Celular , Colesterol/química , Ácidos Graxos Monoinsaturados/química , Hepacivirus/efeitos dos fármacos , Hepatite C/terapia , Hepatócitos/metabolismo , Humanos , Lipossomos , Fígado/virologia , Compostos de Amônio Quaternário/química , RNA Interferente Pequeno/farmacocinética , RNA Interferente Pequeno/farmacologia , Sonicação
12.
J Pharm Pharmacol ; 64(1): 61-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22150673

RESUMO

OBJECTIVES: The objective of this study was to investigate the efficiency of uptake of PEGylated polylactide-co-gycolide (PLGA) nanoparticles by breast cancer cells. METHODS: Nanoparticles of PLGA containing various amounts of polyethylene glycol (PEG, 5%-15%) were prepared using a double emulsion solvent evaporation method. The nanoparticles were loaded with coumarin-6 (C6) as a fluorescence marker. The particles were characterized for surface morphology, particle size, zeta potential, and for cellular uptake by 4T1 murine breast cancer cells. KEY FINDINGS: Irrespective of the amount of PEG, all formulations yielded smooth spherical particles. However, a comparison of the particle size of various formulations showed bimodal distribution of particles. Each formulation was later passed through a 1.2 µm filter to obtain target size particles (114-335 nm) with zeta potentials ranging from -2.8 mV to -26.2 mV. While PLGA-PEG di-block (15% PEG) formulation showed significantly higher 4T1 cellular uptake than all other formulations, there was no statistical difference in cellular uptake among PLGA, PLGA-PEG-PLGA tri-block (10% PEG), PLGA-PEG di-block (5% PEG) and PLGA-PEG di-block (10% PEG) nanoparticles. CONCLUSION: These preliminary findings indicated that the nanoparticle formulation prepared with 15% PEGylated PLGA showed maximum cellular uptake due to it having the smallest particle size and lowest zeta potential.


Assuntos
Materiais Biocompatíveis/química , Cumarínicos/farmacocinética , Portadores de Fármacos , Ácido Láctico/química , Nanopartículas/química , Ácido Poliglicólico/química , Compostos de Sulfidrila/farmacocinética , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Citometria de Fluxo , Camundongos , Microscopia de Fluorescência , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
13.
Int J Pharm ; 423(2): 525-34, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22172291

RESUMO

The goal of this study is to evaluate the stability of lyophilized siRNA formulations. The gene silencing efficiency of a stored lyophilized siRNA formulation (i.e. siRNA nanosomes) was evaluated in interferon-α (IFN-α) resistant hepatitis C virus (HCV) at different time points up to three months in an in vitro cell culture model and compared with freshly prepared siRNA formulations. Novel siRNA sequences were encapsulated within nanosize liposomes following condensation with protamine sulfate. The siRNA encapsulated nanosomes were lyophilized and stored at 4 °C for 3 months, along with liquid liposomes (L) and lyophilized liposome powder (P) which were subsequently used to prepare siRNA nanosomes (L) and siRNA nanosomes (P), respectively at different time points. Physiochemical and biological properties of all three formulations were compared at different time points up to 3 months. The particle size of the stored siRNA nanosomes (642 ± 25 nm) was considerably larger initially in comparison with the liquid liposomes (134 ± 5 nm) and lyophilized liposomes (118 ± 3). However, the particle size gradually became smaller over time (413 ± 128 nm by the third month). The zeta potential of all three formulations was initially very high (> +40 mV), followed by a gradual decrease over time. The amount of siRNA in the stored siRNA nanosomes decreased ∼18 % during the 3 month storage period (1.16 ± 0.03 nmol initially on day 1 vs. 0.95 ± 0.04 nmol after 3 months). With respect to biological potency, all three formulations were significantly effective to knock-down HCV throughout the storage time. The cell viability was well-maintained throughout this period. Thus, this study indicates that the stored lyophilized siRNA formulation is as effective as the fresh preparation and that long-term storage could be a viable option to treat deadly diseases such as cancer and viral infection.


Assuntos
Liofilização , Terapia Genética/métodos , Nanopartículas , Interferência de RNA , Estabilidade de RNA , RNA Interferente Pequeno/química , Antivirais/farmacologia , Linhagem Celular Tumoral , Farmacorresistência Viral , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Humanos , Interferon-alfa/farmacologia , Lipídeos/química , Lipossomos , Nanotecnologia , Conformação de Ácido Nucleico , Tamanho da Partícula , Protaminas/química , Fatores de Tempo , Transfecção , Replicação Viral/efeitos dos fármacos
14.
Virol J ; 8: 351, 2011 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-21756311

RESUMO

The mechanisms underlying the Hepatitis C virus (HCV) resistance to interferon alpha (IFN-α) are not fully understood. We used IFN-α resistant HCV replicon cell lines and an infectious HCV cell culture system to elucidate the mechanisms of IFN-α resistance in cell culture. The IFN-α resistance mechanism of the replicon cells were addressed by a complementation study that utilized the full-length plasmid clones of IFN-α receptor 1 (IFNAR1), IFN-α receptor 2 (IFNAR2), Jak1, Tyk2, Stat1, Stat2 and the ISRE-luciferase reporter plasmid. We demonstrated that the expression of the full-length IFNAR1 clone alone restored the defective Jak-Stat signaling as well as Stat1, Stat2 and Stat3 phosphorylation, nuclear translocation and antiviral response against HCV in all IFN-α resistant cell lines (R-15, R-17 and R-24) used in this study. Moreover RT-PCR, Southern blotting and DNA sequence analysis revealed that the cells from both R-15 and R-24 series of IFN-α resistant cells have 58 amino acid deletions in the extracellular sub domain 1 (SD1) of IFNAR1. In addition, cells from the R-17 series have 50 amino acids deletion in the sub domain 4 (SD4) of IFNAR1 protein leading to impaired activation of Tyk2 kinase. Using an infectious HCV cell culture model we show here that viral replication in the infected Huh-7 cells is relatively resistant to exogenous IFN-α. HCV infection itself induces defective Jak-Stat signaling and impairs Stat1 and Stat2 phosphorylation by down regulation of the cell surface expression of IFNAR1 through the endoplasmic reticulum (ER) stress mechanisms. The results of this study suggest that expression of cell surface IFNAR1 is critical for the response of HCV to exogenous IFN-α.


Assuntos
Expressão Gênica , Hepacivirus/imunologia , Interferon-alfa/imunologia , Receptor de Interferon alfa e beta/biossíntese , Linhagem Celular , Hepatócitos/imunologia , Hepatócitos/virologia , Humanos , Receptor de Interferon alfa e beta/genética , Deleção de Sequência , Transdução de Sinais , Cultura de Vírus
15.
Am J Pathol ; 178(2): 838-52, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21281816

RESUMO

Development of drug resistance is one of the major causes of breast cancer treatment failure. The goal of this study was to understand the chemoresistance mechanism using the highly metastatic 4T1 breast cancer model, which emulates stage IV breast cancer in humans. The metastatic 4T1 breast cancer cell line treated with either doxorubicin or 5-FU showed a concentration-dependent reduced cell proliferation, with induced G2-phase growth arrest (doxorubicin) or G1-phase growth arrest (5-FU). Doxorubicin treatment partially suppressed the multiorgan metastasis of 4T1 breast cancer cells in the lung, heart, liver, and bone, compared with either 5-FU or cyclophosphamide. We isolated and characterized 4T1 breast cancer cells from doxorubicin-resistant metastatic tumors (cell line 4T1-R). Multiorgan metastasis of drug-resistant 4T1 breast tumors was totally resistant to doxorubicin treatment. Our results indicate that doxorubicin is localized exclusively in the cytoplasm of resistant 4T1 breast cancer cells and that it cannot reach the nucleus because of increased nuclear expression of P-glycoprotein. Pretreatment of doxorubicin-resistant 4T1-R breast cancer cells with verapamil, a general inhibitor of P-glycoprotein, increased nuclear translocation of doxorubicin and cellular cytotoxicity. Thus, impaired nuclear translocation of doxorubicin due to increased expression of P-glycoprotein is associated with doxorubicin resistance of highly metastatic 4T1 breast cancer.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Citometria de Fluxo , Fluoruracila/farmacologia , Fase G2/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Mitose/efeitos dos fármacos , Metástase Neoplásica , Análise de Sobrevida , Verapamil/farmacologia
16.
World J Gastroenterol ; 17(3): 300-12, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21253388

RESUMO

AIM: To develop a hepatocellular carcinoma (HCC) xenograft model for studying hepatitis C virus (HCV) replication in a mice, and antiviral treatment. METHODS: We developed a stable S3-green fluorescence protein (GFP) cell line that replicated the GFP-tagged HCV sub-genomic RNA derived from a highly efficient JFH1 virus. S3-GFP replicon cell line was injected subcutaneously into γ-irradiated SCID mice. We showed that the S3-GFP replicon cell line formed human HCC xenografts in SCID mice. Cells were isolated from subcutaneous tumors and then serially passaged multiple times in SCID mice by culturing in growth medium supplemented with G-418. The mouse-adapted S3-GFP replicon cells were implanted subcutaneously and also into the liver of SCID mice via intrasplenic infusion to study the replication of HCV in the HCC xenografts. The tumor model was validated for antiviral testing after intraperitoneal injection of interferon-α (IFN-α). RESULTS: A highly tumorigenic S3-GFP replicon cell line was developed that formed subcutaneous tumors within 2 wk and diffuse liver metastasis within 4 wk in SCID mice. Replication of HCV in the subcutaneous and liver tumors was confirmed by cell colony assay, detection of the viral RNA by ribonuclease protection assay and real-time quantitative reverse transcription polymerase chain reaction. High-level replication of HCV sub-genomic RNA in the tumor could be visualized by GFP expression using fluorescence microscopy. IFN-α cleared HCV RNA replication in the subcutaneous tumors within 2 wk and 4 wk in the liver tumor model. CONCLUSION: A non-infectious mouse model allows us to study replication of HCV in subcutaneous and metastatic liver tumors. Clearance of HCV by IFN-α supports use of this model to test other anti-HCV drugs.


Assuntos
Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Hepacivirus/fisiologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Transplante Heterólogo , Replicação Viral/fisiologia , Animais , Antivirais/uso terapêutico , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hepatite C/tratamento farmacológico , Humanos , Interferon-alfa/farmacologia , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , RNA Viral/genética , Replicação Viral/efeitos dos fármacos
17.
PLoS One ; 5(9)2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20949125

RESUMO

BACKGROUND: We have developed multiple stable cell lines containing subgenomic HCV RNA that are resistant to treatment with interferon alpha (IFN-α. Characterization of these IFN-α resistant replicon cells showed defects in the phosphorylation and nuclear translocation of STAT1 and STAT2 proteins due to a defective Jak-STAT pathway. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we have developed an alternative strategy to overcome interferon resistance in a cell culture model by improving intracellular STAT1 signaling. An engineered STAT1-CC molecule with double cysteine substitutions in the Src-homology 2 (SH2) domains of STAT1 (at Ala-656 and Asn-658) efficiently phosphorylates and translocates to the nucleus of IFN-resistant cells in an IFN-γ dependent manner. Transfection of a plasmid clone containing STAT1-CC significantly activated the GAS promoter compared to wild type STAT1 and STAT3. The activity of the engineered STAT1-CC is dependent upon the phosphorylation of tyrosine residue 701, since the construct with a substituted phenylalanine residue at position 701 (STAT1-CC-Y701F) failed to activate GAS promoter in the replicon cells. Intracellular expression of STAT1-CC protein showed phosphorylation and nuclear translocation in the resistant cell line after IFN-γ treatment. Transient transfection of STAT1-CC plasmid clone into an interferon resistant cell line resulted in inhibition of viral replication and viral clearance in an IFN-γ dependent manner. Furthermore, the resistant replicon cells transfected with STAT1-CC constructs significantly up regulated surface HLA-1 expression when compared to the wild type and Y to F mutant controls. CONCLUSIONS: These results suggest that modification of the SH2 domain of the STAT1 molecule allows for improved IFN-γ signaling through increased STAT1 phosphorylation, nuclear translocation, HLA-1 surface expression, and prolonged interferon antiviral gene activation.


Assuntos
Farmacorresistência Viral , Antígenos HLA/genética , Hepacivirus/genética , Hepatite C/metabolismo , Interferon gama/metabolismo , Replicon , Fator de Transcrição STAT1/química , Transdução de Sinais , Antivirais/farmacologia , Linhagem Celular , Expressão Gênica , Antígenos HLA/metabolismo , Hepacivirus/efeitos dos fármacos , Hepacivirus/fisiologia , Hepatite C/genética , Hepatite C/virologia , Humanos , Interferon-alfa/farmacologia , Interferon gama/genética , Fosforilação , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Replicação Viral/efeitos dos fármacos , Domínios de Homologia de src
18.
Virol J ; 7: 265, 2010 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-20939906

RESUMO

Interferon alpha (IFN-α) binds to a cell surface receptor that activates the Jak-Stat signaling pathway. A critical component of this pathway is the translocation of interferon stimulated gene factor 3 (a complex of three proteins Stat1, Stat2 and IRF9) to the nucleus to activate antiviral genes. A stable sub-genomic replicon cell line resistant to IFN-α was developed in which the nuclear translocation of Stat1 and Stat2 proteins was prevented due to the lack of phosphorylation; whereas the nuclear translocation of IRF9 protein was not affected. In this study, we sought to overcome defective Jak-Stat signaling and to induce an antiviral state in the IFN-α resistant replicon cell line by developing a chimera IRF9 protein fused with the trans activating domain (TAD) of either a Stat1 (IRF9-S1C) or Stat2 (IRF9-S2C) protein. We show here that intracellular expression of fusion proteins using the plasmid constructs of either IRF9-S1C or IRF9-S2C, in the IFN-α resistant cells, resulted in an increase in Interferon Stimulated Response Element (ISRE) luciferase promoter activity and significantly induced HLA-1 surface expression. Moreover, we show that transient transfection of IRF9-S1C or IRF9-S2C plasmid constructs into IFN-α resistant replicon cells containing sub-genomic HCV1b and HCV2a viruses resulted in an inhibition of viral replication and viral protein expression independent of IFN-α treatment. The results of this study indicate that the recombinant fusion proteins of IRF9-S1C, IRF9-S2C alone, or in combination, have potent antiviral properties against the HCV in an IFN-α resistant cell line with a defective Jak-Stat signaling.


Assuntos
Hepacivirus/imunologia , Hepacivirus/fisiologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT2/imunologia , Transdução de Sinais , Replicação Viral , Linhagem Celular , Genes Reporter , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Interferon-alfa/imunologia , Luciferases/genética , Luciferases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT2/genética
19.
J Pharm Pharmacol ; 62(9): 1103-11, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20796188

RESUMO

OBJECTIVES: The aim of this project was to develop a novel lipid-based formulation suitable for gene therapy. METHODS: Novel nanosize liposome (nanosome) formulations containing pDNA (plasmid DNA) were developed using high-pressure homogenization (HPH). The effect of lipid concentration was studied at two levels: 3 mm and 20 mm. The preformed nanosomes were incubated for 18-20 h with pDNA or pDNA/protamine sulfate (PS) complex. The physical properties of the pDNA nanosomes were compared by particle size distribution and zeta-potential measurements. Their biological properties were also compared by pDNA efficiency of encapsulation/complexation, integrity, nuclease digestion, transfection efficiency and cell cytotoxicity. KEY FINDINGS: pDNA nanosomes prepared with 20 mM lipid (nanosomes:pDNA:PS at a ratio of 8.6:1:2) had particle sizes of 170-422 nm (90% confidence). The zeta-potential of the formulation was 49.2 +/- 1.5 mV, and the pDNA encapsulation/complexation efficiency was approximately 98%. pDNA nanosomes prepared with 3 mM lipid (nanosomes:pDNA PS at a ratio of 2.09:1:2) had particle sizes of 140-263 nm (90% confidence). The zeta-potential of this formulation was 36.4 +/- 1.2 mV, and the pDNA encapsulation/complexation efficiency was approximately 100%. However, a comparison of the efficiency of transfection indicated that pDNA nanosomes prepared with low-concentration lipids (3 mM) showed significantly higher transfection efficiency compared with the pDNA nanosomes prepared with high-concentration lipids (20 mM), as well as those prepared with Fugene-6 (a commercially available transfection reagent). This particular formulation (pDNA nanosomes, 3 mM lipids) also showed significantly less cytotoxicity compared with the other pDNA nanosome formulations. CONCLUSIONS: To conclude, these results indicate that condensing pDNA with PS followed by subsequent complexation with low-concentration nanosomes generated from HPH can produce a pDNA nanosome formulation that will boost transfection efficiency, while minimizing cytotoxicity. This new technology appears to be an efficient tool for future commercial or large-scale manufacture of DNA delivery systems for gene therapy.


Assuntos
Química Farmacêutica/métodos , DNA/química , Terapia Genética/métodos , Vetores Genéticos , Lipídeos/química , Lipossomos , Nanocápsulas , Animais , Linhagem Celular Tumoral , Técnicas de Transferência de Genes , Camundongos , Plasmídeos , Pressão , Protaminas/química , Transfecção
20.
J Pharm Pharmacol ; 62(4): 422-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20604830

RESUMO

OBJECTIVE: The aim was to develop biodegradable nanoparticles suitable for cellular delivery of chemotherapeutic drugs. METHODS: Poly (lactide-co-glycolide) (PLGA) nanoparticles were prepared using a modified solvent evaporation method. Chitosan and calcium chloride were tested as surface modifiers. Coumarin-6 was incorporated into some formulations as a fluorescent marker. KEY FINDINGS: The median size of the particles was between 400 nm and 7 microm, and scanning electron microscope pictures showed that the particles were smooth and spherical. The zeta potentials of the particles with and without surface modifier ranged between -25.7 mV and -7.0 mV, respectively. Fluorescence microscopy and flow cytometry (FACS) analysis showed that smaller surface-modified particles were efficiently internalised by neoplastic 4T1 cells. Image analysis of frozen tissue sections from Balb/c mice given nanoparticles via the tail vein showed that the particles were distributed preferentially into the lungs, followed by the liver, spleen, kidney and heart. CONCLUSIONS: Chitosan-modified PLGA nanoparticles showed significant uptake by neoplastic 4T1 cells, and were distributed to several major organs frequently seen as sites of cancer metastasis in mice.


Assuntos
Neoplasias da Mama/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Fluorescência , Ácido Láctico/química , Nanopartículas , Ácido Poliglicólico/química , Tecnologia Farmacêutica/métodos , Animais , Transporte Biológico , Linhagem Celular Tumoral , Quitosana , Cumarínicos , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Varredura , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Espectrometria de Fluorescência , Propriedades de Superfície , Tiazóis , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA