Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mater Sci Eng C Mater Biol Appl ; 98: 419-436, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30813043

RESUMO

In the present investigation, FePt alloy nanoparticles were synthesized with controlled size and elemental composition followed by surface modification using (3-Aminopropyl) triethoxysilane (APTES). Lenalidomide was covalently bound to FePt-NH2 by pH sensitive hydrazone bonding. Hyaluronic acid was conjugated to amino groups of APTES while lactoferrin (Lf) was directly conjugated to excess carboxylic group present on hyaluronic acid (HA) to form surface modified pH sensitive alloy-drug nanoconjugates (SPANs). The multifunctional nanoconjugates were characterized and evaluated using extensive in vitro and in vivo techniques. The nanoconjugates demonstrated excellent heating ability on exposure to alternating magnetic field and near-infrared laser irradiation. The acidic microenvironment of lysozome triggered release of LND from SPANs. Owing to leaching of Fe and Pt contents, SPANs demonstrated ability to generate reactive oxygen species (ROS) in U87MG cell line which further enhanced therapeutic effect of SPANs. Significant difference in cell viability suppression was observed in in vitro photothermal, chemo-photothermal and chemo-magnetophotothermal killing of cancer cells using SPANs in U87MG cell lines. Significant difference in heating ability and cell cytotoxicity of SPANs in comparison to alternative magnetic field and NIR irradiation was observed for DUAL-mode exposure of SPANs. The results of cellular internalization study showed efficient internalization of SPANs inside U87MG cells. The in vivo results (both qualitative and quantitative) confirmed enhanced uptake of SPANs in brain after intranasal administration with enhanced nasal and mucus penetration owing to presence of Lf. No significant interaction was observed with ECM and mucin due to presence of carboxyl group on SPANs.


Assuntos
Ligas/química , Glioblastoma/terapia , Ácido Hialurônico/química , Nanoconjugados/química , Administração Intranasal , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cães , Liberação Controlada de Fármacos , Endocitose , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Concentração de Íons de Hidrogênio , Hipertermia Induzida , Ferro/química , Lactoferrina/química , Lenalidomida/administração & dosagem , Lenalidomida/farmacologia , Lenalidomida/uso terapêutico , Masculino , Mucinas/metabolismo , Nanoconjugados/ultraestrutura , Ácido Oleico/química , Espectroscopia Fotoeletrônica , Fototerapia , Platina/química , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Suínos
2.
Food Funct ; 9(4): 2015-2024, 2018 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-29541738

RESUMO

The effect of chitosan decoration on the transport of epigallocatechin (EGC)-encapsulated ferritin cage across the Caco-2 cells was investigated. After the encapsulation of EGC in apo-red bean (adzuki) ferritin (apoRBF), the EGC-loaded apoRBF nanoparticle (ER) was fabricated with an encapsulation ratio of 11.6% (w/w). The results indicated that different chitosan molecules (with molecular weights of 980, 4600, 46 000, and 210 000 Da) could attach onto the apoRBF via electrostatic interactions to form ER-chitosan complexes (ERCs) (ERCs980, ERCs4600, ERCs46000, and ERCs210000). ERCs980 and ERCs4600 retained the typical shell-like morphology of ferritin with a size distribution of 12 nm and showed weak negative zeta-potentials at pH 6.7, while ERCs46000 and ERCs210000 significantly aggregated. Furthermore, the transport of EGC in ERCs980 and ERCs4600 across the Caco-2 cells was enhanced by the transferrin receptor 1 (TfR-1)-mediated absorption pathway, demonstrating that chitosan molecules with low molecular weights of 980 and 4600 Da were beneficial to the absorption of EGC based on the ferritin cage. This study will facilitate the application of ferritin-chitosan materials for fabricating the core-shell platform for encapsulation and bioavailability enhancement of bioactive molecules.


Assuntos
Apoferritinas/metabolismo , Catequina/análogos & derivados , Quitosana/metabolismo , Enterócitos/metabolismo , Absorção Intestinal , Nanoconjugados/química , Receptores da Transferrina/metabolismo , Absorção Fisiológica , Algoritmos , Apoferritinas/química , Apoferritinas/ultraestrutura , Catequina/administração & dosagem , Catequina/química , Catequina/metabolismo , Quitosana/química , Suplementos Nutricionais/análise , Difusão Dinâmica da Luz , Humanos , Microscopia Eletrônica de Transmissão , Peso Molecular , Nanoconjugados/ultraestrutura , Tamanho da Partícula , Proteínas de Vegetais Comestíveis/metabolismo , Sementes/química , Eletricidade Estática , Propriedades de Superfície , Vigna/química
3.
Biomaterials ; 124: 35-46, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28187393

RESUMO

Superparamagnetic Fe3O4 nanoparticles (NPs)-based hyperthermia is a promising non-invasive approach for cancer therapy. However, the heat transfer efficiency of Fe3O4 NPs is relative low, which hinders their practical clinical applications. Therefore, it is promising to improve the magnetic hyperthermia efficiency by exploring the higher performance magnetic NPs-based hybrid nanostructures. In the current study, it presents a straightforward in situ reduction method for the shape-controlled preparation of magnetite (Fe3O4) silver (Ag) hybrid NPs designed as magnetic hyperthermia heat mediators. The magnetite silver hybrid NPs with core-shell (Fe3O4@Ag) or heteromer (Fe3O4-Ag) structures exhibited a higher biocompatibility with SMMC-7721 cells and L02 cells than the individual Ag NPs. Importantly, in the magnetic hyperthermia, with the exposure to alternating current magnetic field, the Fe3O4@Ag and Fe3O4-Ag hybrid NPs indicated much better tumor suppression effect against SMMC-7721 cells than the individual Fe3O4 NPs in vitro and in vivo. These results demonstrate that the hybridisation of Fe3O4 and Ag NPs could greatly enhance the magnetic hyperthermia efficiency of Fe3O4 NPs. Therefore, the Fe3O4@Ag and Fe3O4-Ag hybrid NPs can be used to be as high performance magnetic hyperthermia mediators based on a simple and effective preparation approach.


Assuntos
Hipertermia Induzida/métodos , Magnetoterapia/métodos , Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/ultraestrutura , Neoplasias Experimentais/terapia , Prata/administração & dosagem , Animais , Linhagem Celular Tumoral , Cristalização/métodos , Composição de Medicamentos/métodos , Feminino , Humanos , Nanopartículas de Magnetita/química , Camundongos Endogâmicos C57BL , Camundongos Nus , Nanoconjugados/administração & dosagem , Nanoconjugados/química , Nanoconjugados/ultraestrutura , Neoplasias Experimentais/patologia , Tamanho da Partícula , Prata/química , Resultado do Tratamento
4.
Biosens Bioelectron ; 86: 958-965, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27498321

RESUMO

Herein, integrated with DNAzyme highly specific to metal ions, hemin@reduced graphene oxide (hemin@rGO) functionalized with flower-like MnO2 and hollow AuPd (hAuPd-fMnO2-hemin@rGO) was used as electroactive probe and electrocatalyst to construct a universal platform for metal ion detection (lead ion Pb(2+) as the model). The proposed strategy with generality was mainly based on two aspects. Firstly, the designed probe not only showed high stability and excellent peroxidase-like activity originating from hemin, fMnO2 and hAuPd, but also possessed intrinsic redox performance from hemin, which resulted in the promotion of electron transfer and the enhancement of the response signal readout. Secondly, due to the introduction of Pb(2+), Pb(2+)-dependent DNAzyme bound in the electrode surface could be specifically identified and cleaved by Pb(2+), and the remained fragment (its supplementary sequence is a single-strand DNA S3) captured the nanocomposites S3-hAuPd-fMnO2-hemin@rGO by the hybridization reaction. Therefore, combined the cooperative catalysis of fMnO2, hAuPd and hemin to H2O2 reduction with highly specific interaction of Pb(2+)-dependent DNAzyme, the proposed Pb(2+) biosensor showed significant improvement of electrochemical analytical performance, which was involved in wide dynamic response in the range of 0.1pM-200nM, low detection limit of 0.034pM, high sensitivity and high specificity. This could facilitate the universal strategy to be a promising method for detection of other metal ions, only changing the corresponding DNAzyme specific to them.


Assuntos
Condutometria/instrumentação , DNA Catalítico , Hemina/química , Imunoensaio/instrumentação , Chumbo/análise , Nanopartículas Metálicas/química , Adsorção , DNA/química , Desenho de Equipamento , Análise de Falha de Equipamento , Ouro/química , Grafite/química , Íons/análise , Compostos de Manganês/química , Nanopartículas Metálicas/ultraestrutura , Nanoconjugados/química , Nanoconjugados/ultraestrutura , Nanoporos/ultraestrutura , Óxidos/química , Paládio/química , Tamanho da Partícula
5.
J Colloid Interface Sci ; 482: 121-130, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27497233

RESUMO

We report that through facile lipid self-assembly, biomimetic membrane-conjugated mesoporous silica-coated graphene oxide is constructed as targeting nanocarrier toward efficient combination of photothermal therapy and chemotherapy. Impressively, the simple surface modification with folate-contained lipid bilayer allows the graphene-based nanoarchitecture above to be selectively internalized by tumor cells overexpressing relevant receptors. Compared to pure drug, 7-fold doxorubicin is delivered into tumor cells by the nanoarchitecture. After cellular internalization, upon near infrared light illumination, graphene oxide in the nanoarchitecture can convert light energy into heat to kill cancer cells partially. Simultaneously, hyperthermia will drive rapid release of doxorubicin from the nanoarchitecture above to further cause the death of more cancer cells. Thus, integrated cancer treatment with higher efficacy is achieved in vitro compared to that of individual therapy.


Assuntos
Materiais Biomiméticos/síntese química , Portadores de Fármacos , Nanoconjugados/química , Fototerapia/métodos , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacologia , Materiais Biomiméticos/farmacologia , Materiais Biomiméticos/efeitos da radiação , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Terapia Combinada , Doxorrubicina/química , Doxorrubicina/farmacologia , Composição de Medicamentos , Endocitose , Receptores de Folato com Âncoras de GPI/genética , Receptores de Folato com Âncoras de GPI/metabolismo , Ácido Fólico/química , Ácido Fólico/metabolismo , Expressão Gênica , Grafite/química , Células HeLa , Humanos , Raios Infravermelhos , Bicamadas Lipídicas/química , Células MCF-7 , Nanoconjugados/efeitos da radiação , Nanoconjugados/ultraestrutura , Dióxido de Silício/química
6.
Front Biosci (Landmark Ed) ; 21(6): 1211-21, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27100501

RESUMO

The aim of this study was to enhance the effectiveness of photo thermal therapy (PTT) in the targeting of superficial bladder cancers using a green light laser in conjunction with gold nanoparticles (GNPs) conjugated to antibody fragments (anti-EGFR). GNPs conjugated with anti-EGFR-antibody fragments were used as probes in the targeting of tumor cells and then exposed to a green laser (532nm), resulting in the production of sufficient thermal energy to kill urothelial carcinomas both in vitro and in vivo. Nanoparticles conjugated with antibody fragments are capable of damaging cancer cells even at relatively very low energy levels, while non-conjugated nanoparticles would require an energy level of 3 times under the same conditions. The lower energy required by the nanoparticles allows this method to destroy cancerous cells while preserving normal cells when applied in vivo. Nanoparticles conjugated with antibody fragments (anti-EGFR) require less than half the energy of non-conjugated nanoparticles to kill cancer cells. In an orthotopic bladder cancer model, the group treated using PTT presented significant differences in tumor development.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Terapia com Luz de Baixa Intensidade/métodos , Nanoconjugados/uso terapêutico , Neoplasias da Bexiga Urinária/terapia , Animais , Anticorpos/administração & dosagem , Linhagem Celular Tumoral , Ouro , Humanos , Fragmentos de Imunoglobulinas/administração & dosagem , Nanopartículas Metálicas/uso terapêutico , Nanopartículas Metálicas/ultraestrutura , Camundongos , Camundongos Endogâmicos C3H , Nanoconjugados/ultraestrutura , Nanotecnologia , Neoplasias da Bexiga Urinária/ultraestrutura
7.
J Biomed Nanotechnol ; 11(8): 1431-41, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26295143

RESUMO

Generation 5 (G5) poly(amidoamide) (PAMAM) dendrimers are synthetic polymers that have been broadly applied as drug delivery carriers. Methotrexate (MTX), an anti-folate metabolite, has been successfully used as an anti-inflammatory drug to treat rheumatoid arthritis (RA) in the clinic. In this study, we examine the therapeutic efficacy of G5 PAMAM dendrimer methotrexate conjugates (G5-MTX) that also have folic acid (FA) conjugated to the G5-MTX (G5-FA-MTX) to target inflammation-activated folate receptors overexpressing macrophages. These cells are thought to play an important role in the development of RA. With G5 serving as a control, the in vitro binding affinities of G5-FA-MTX and G5-MTX to activated macrophages were assessed in RAW264.7, NR8383 and primary rat peritoneal macrophages. The results indicated that the binding of either conjugate to macrophages was concentration- and temperature-dependent and could be blocked by the presence of 6.25 mM free FA (p < 0.005). The preventive effects of G5-MTX and G5-FA-MTX conjugates on the development of arthritis were explored on an adjuvant-induced inflammatory arthritis model and had similar preventive effects in inflammatory arthritis at a MTX equivalent dose of 4.95 µmol/kg. These studies indicated that when multiples of MTX are conjugated on dendritic polymers, they specifically bind to folate receptor overexpressing macrophages and have comparable anti-inflammatory effects to folate targeted MTX conjugated polymers.


Assuntos
Artrite/tratamento farmacológico , Artrite/metabolismo , Dendrímeros/química , Ácido Fólico/farmacocinética , Metotrexato/administração & dosagem , Nanocápsulas/química , Animais , Linhagem Celular , Difusão , Feminino , Receptores de Folato com Âncoras de GPI/metabolismo , Ácido Fólico/química , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Metotrexato/química , Nanocápsulas/ultraestrutura , Nanoconjugados/química , Nanoconjugados/ultraestrutura , Ratos , Ratos Endogâmicos Lew , Resultado do Tratamento
8.
Biomaterials ; 63: 115-27, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26093792

RESUMO

To integrate biological imaging and multimodal therapies into one platform for enhanced anti-cancer efficacy, we have designed a novel core/shell structured nano-theranostic by conjugating photosensitive Au25(SR)18 - (SR refers to thiolate) clusters, pH/temperature-responsive polymer P(NIPAm-MAA), and anti-cancer drug (doxorubicin, DOX) onto the surface of mesoporous silica coated core-shell up-conversion nanoparticles (UCNPs). It is found that the photodynamic therapy (PDT) derived from the generated reactive oxygen species and the photothermal therapy (PTT) arising from the photothermal effect can be simultaneously triggered by a single 980 nm near infrared (NIR) light. Furthermore, the thermal effect can also stimulate the pH/temperature sensitive polymer in the cancer sites, thus realizing the targeted and controllable DOX release. The combined PDT, PTT and pH/temperature responsive chemo-therapy can markedly improve the therapeutic efficacy, which has been confirmed by both in intro and in vivo assays. Moreover, the doped rare earths endow the platform with dual-modal up-conversion luminescent (UCL) and computer tomography (CT) imaging properties, thus achieving the target of imaging-guided synergistic therapy under by a single NIR light.


Assuntos
Acrilamidas/química , Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Substâncias Luminescentes/química , Nanoconjugados/química , Neoplasias/diagnóstico , Neoplasias/terapia , Ácidos Polimetacrílicos/química , Acrilamidas/uso terapêutico , Animais , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Preparações de Ação Retardada/química , Preparações de Ação Retardada/uso terapêutico , Doxorrubicina/uso terapêutico , Liberação Controlada de Fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Hipertermia Induzida/métodos , Substâncias Luminescentes/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Nanoconjugados/uso terapêutico , Nanoconjugados/ultraestrutura , Imagem Óptica , Fotoquimioterapia/métodos , Ácidos Polimetacrílicos/uso terapêutico , Temperatura , Nanomedicina Teranóstica/métodos , Tomografia Computadorizada por Raios X
9.
J Biomed Nanotechnol ; 10(7): 1267-76, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24804547

RESUMO

Nanomaterial-directed, photothermal ablation is a practical future approach for the treatment of early-stage bladder cancer. Using a new PEGylation technique with bi-functional nitrophenyl carbonate PEG (bi-NPC-PEG) that promotes uniform suspension of the nanomaterial in solution, we have shown that gold nanorods conjugated to an anti-EGFR antibody (nano-alphaEGFR) bind effectively to EGFR-expressing bladder cancer cells. The subsequent application of infrared light, specifically tuned to the plasmon resonance of the nanorods used in this work, allows for the specific heating of nano-alphaEGFR to the point of localized cellular death. Such an approach, administering nano-alphaEGFR intravesically via a urinary catheter and infrared light via a modified cystoscope, represents a novel, future clinical application of this technology, which avoids the problem of systemic exposure and clearance of nanoparticles from body.


Assuntos
Técnicas de Ablação , Ouro/uso terapêutico , Hipertermia Induzida , Nanotubos/química , Neoplasias da Bexiga Urinária/terapia , Morte Celular , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Microscopia de Fluorescência , Nanoconjugados/ultraestrutura , Nanotubos/ultraestrutura , Polietilenoglicóis/química , Espectrofotometria Atômica
10.
Nanomedicine (Lond) ; 9(9): 1389-402, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24364872

RESUMO

AIM: Discovering which anticancer drugs attack which organelle(s) of cancer cells is essential and significant, not only for understanding their therapeutic and adverse effects, but also to enable the development of new-generation therapeutics. Here, we show that novel Fe3O4-carboxymethyl cellulose-5-fluorouracil (Fe3O4-CMC-5FU) nanomedicine can apparently enhance the antitumor effect on gastric cancer cells, and its mechanism of killing the SGC-7901 gastric cancer cells can be directly observed at the atomic scale. MATERIALS & METHODS: The novel nanomedicine was prepared using the traditional antitumor drug 5FU to chemically bond onto the functionalized Fe3O4 nanoparticles (Fe3O4-CMC-5FU nanomedicine), and then was fed into SGC-7901 gastric cancer cells. The inorganic Fe3O4 nanoparticles were used to track the distribution and antitumor effect of the nanomedicine within individual SGC-7901 gastric cancer cells. RESULTS & DISCUSSION: Atomic-level observation and tracking the elemental distribution inside individual cells proved that the magnetic nanomedicine killed the gastric cells mainly by attacking their mitochondria. The enhanced therapeutic efficacy derives from the localized high concentration and poor mobility of the aggregated Fe3O4-CMC-5FU nanomedicine in the cytoplasm. CONCLUSION: A brand new mechanism of Fe3O4-CMC-5FU nanomedicine killing SGC-7901 gastric cancer cells by attacking their mitochondria was discovered, which is different from the classical mechanism utilized by traditional medicine 5FU, which kills gastric cancer cells by damaging their DNA. Our work might provide a partial solution in nanomedicines or even modern anticancer medicine for the visualized investigation of their antitumor effect.


Assuntos
Antineoplásicos/administração & dosagem , Fluoruracila/administração & dosagem , Nanopartículas de Magnetita/administração & dosagem , Neoplasias Gástricas/tratamento farmacológico , Antineoplásicos/farmacocinética , Carboximetilcelulose Sódica/química , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Fluoruracila/farmacocinética , Humanos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestrutura , Microscopia Eletrônica de Transmissão e Varredura , Mitocôndrias/efeitos dos fármacos , Nanoconjugados/administração & dosagem , Nanoconjugados/química , Nanoconjugados/ultraestrutura , Nanomedicina , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
11.
Methods Enzymol ; 509: 127-42, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22568904

RESUMO

The design of antigen delivery systems, particularly for mucosal surfaces, has been a focus of interest in recent years. In this chapter, we describe the preparation of chitosan-based particles as promising antigen delivery systems for mucosal surfaces already tested by our group with hepatitis B surface antigen. The final proof of the concept is always carried out with immunization studies performed in an appropriate animal model. However, before these important studies, it is advisable that the delivery system should be submitted to a variety of in vitro tests. Among several tests, the characterization of the particles (size, morphology, and zeta potential), the studies of antigen adsorption onto particles, the evaluation of toxicity of the particles, and the studies of particle uptake into lymphoid organs are the most important and will be described in this chapter.


Assuntos
Adjuvantes Imunológicos/química , Quitosana/química , Antígenos de Superfície da Hepatite B/química , Vacinas contra Hepatite B/química , Nanoconjugados/química , Poliésteres/química , Adjuvantes Imunológicos/administração & dosagem , Adsorção , Alginatos , Animais , Sobrevivência Celular , Células Cultivadas , Ácido Glucurônico , Hepatite B/imunologia , Antígenos de Superfície da Hepatite B/administração & dosagem , Vacinas contra Hepatite B/administração & dosagem , Ácidos Hexurônicos , Proteínas Imobilizadas/administração & dosagem , Proteínas Imobilizadas/química , Absorção Intestinal , Camundongos , Nanoconjugados/ultraestrutura , Tamanho da Partícula , Nódulos Linfáticos Agregados/metabolismo , Ratos , Baço/citologia
12.
J Nanobiotechnology ; 9: 13, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21477294

RESUMO

BACKGROUND: The use of silica coated magnetic nanoparticles as contrast agents has resulted in the production of highly stable, non-toxic solutions that can be manipulated via an external magnetic field. As a result, the interaction of these nanocomposites with cells is of vital importance in understanding their behaviour and biocompatibility. Here we report the preparation, characterisation and potential application of new "two-in-one" magnetic fluorescent nanocomposites composed of silica-coated magnetite nanoparticles covalently linked to a porphyrin moiety. METHOD: The experiments were performed by administering porphyrin functionalised silica-coated magnetite nanoparticles to THP-1 cells, a human acute monocytic leukaemia cell line. Cells were cultured in RPMI 1640 medium with 25 mM HEPES supplemented with heat-inactivated foetal bovine serum (FBS). RESULTS: We have synthesised, characterised and analysed in vitro, a new multimodal (magnetic and fluorescent) porphyrin magnetic nanoparticle composite (PMNC). Initial co-incubation experiments performed with THP-1 macrophage cells were promising; however the PMNC photobleached under confocal microscopy study. ß-mercaptoethanol (ß-ME) was employed to counteract this problem and resulted not only in enhanced fluorescence emission, but also allowed for elongated imaging and increased exposure times of the PMNC in a cellular environment. CONCLUSION: Our experiments have demonstrated that ß-ME visibly enhances the emission intensity. No deleterious effects to the cells were witnessed upon co-incubation with ß-ME alone and no increases in background fluorescence were recorded. These results should present an interest for further development of in vitro biological imaging techniques.


Assuntos
Nanopartículas de Magnetita/química , Nanoconjugados/química , Porfirinas/síntese química , Linhagem Celular Tumoral , Diagnóstico por Imagem/métodos , HEPES/administração & dosagem , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Nanopartículas de Magnetita/ultraestrutura , Mercaptoetanol/administração & dosagem , Microscopia Confocal/métodos , Nanoconjugados/ultraestrutura , Fotodegradação , Porfirinas/metabolismo , Coloração e Rotulagem/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA