Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 112(37): 11612-7, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26305967

RESUMEN

Epstein-Barr virus (EBV) infects germinal center (GC) B cells and establishes persistent infection in memory B cells. EBV-infected B cells can cause B-cell malignancies in humans with T- or natural killer-cell deficiency. We now find that EBV-encoded latent membrane protein 2A (LMP2A) mimics B-cell antigen receptor (BCR) signaling in murine GC B cells, causing altered humoral immune responses and autoimmune diseases. Investigation of the impact of LMP2A on B-cell differentiation in mice that conditionally express LMP2A in GC B cells or all B-lineage cells found LMP2A expression enhanced not only BCR signals but also plasma cell differentiation in vitro and in vivo. Conditional LMP2A expression in GC B cells resulted in preferential selection of low-affinity antibody-producing B cells despite apparently normal GC formation. GC B-cell-specific LMP2A expression led to systemic lupus erythematosus-like autoimmune phenotypes in an age-dependent manner. Epigenetic profiling of LMP2A B cells found increased H3K27ac and H3K4me1 signals at the zinc finger and bric-a-brac, tramtrack domain-containing protein 20 locus. We conclude that LMP2A reduces the stringency of GC B-cell selection and may contribute to persistent EBV infection and pathogenesis by providing GC B cells with excessive prosurvival effects.


Asunto(s)
Centro Germinal/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas de la Matriz Viral/metabolismo , Animales , Autoanticuerpos/química , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/virología , Diferenciación Celular , Linaje de la Célula , Cruzamientos Genéticos , Epigénesis Genética , Citometría de Flujo , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Heterocigoto , Inmunidad Humoral , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Fluorescente , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Bazo/citología , Dedos de Zinc
2.
Antimicrob Agents Chemother ; 57(11): 5366-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23959316

RESUMEN

Group A streptococcus (GAS) infection may cause severe life-threatening diseases, including necrotizing fasciitis and streptococcal toxic shock syndrome. Despite the availability of effective antimicrobial agents, there has been a worldwide increase in the incidence of invasive GAS infection. Kallistatin (KS), originally found to be a tissue kallikrein-binding protein, has recently been shown to possess anti-inflammatory properties. However, its efficacy in microbial infection has not been explored. In this study, we transiently expressed the human KS gene by hydrodynamic injection and investigated its anti-inflammatory and protective effects in mice via air pouch inoculation of GAS. The results showed that KS significantly increased the survival rate of GAS-infected mice. KS treatment reduced local skin damage and bacterial counts compared with those in mice infected with GAS and treated with a control plasmid or saline. While there was a decrease in immune cell infiltration of the local infection site, cell viability and antimicrobial factors such as reactive oxygen species actually increased after KS treatment. The efficiency of intracellular bacterial killing in neutrophils was directly enhanced by KS administration. Several inflammatory cytokines, including tumor necrosis factor alpha, interleukin 1ß, and interleukin 6, in local infection sites were reduced by KS. In addition, KS treatment reduced vessel leakage, bacteremia, and liver damage after local infection. Therefore, our study demonstrates that KS provides protection in GAS-infected mice by enhancing bacterial clearance, as well as reducing inflammatory responses and organ damage.


Asunto(s)
Inmunomodulación , Neutrófilos/inmunología , Serpinas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Expresión Génica , Humanos , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Ratones , Neutrófilos/microbiología , Serpinas/genética , Serpinas/metabolismo , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/mortalidad , Streptococcus pyogenes/patogenicidad , Análisis de Supervivencia , Transgenes , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis
3.
Eur Cell Mater ; 23: 170-81; discussion 181, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22415803

RESUMEN

Cellular behaviour is controlled by numerous processes, including intracellular signalling pathways that are triggered by the binding of ligands with cell surface receptors. Multivalent ligands have multiple copies of a recognition element that binds to receptors and influences downstream signals. Nanoparticle-ligand complexes may form multivalent structures to crosslink receptors with high avidity and specificity. After conjugation onto gold nanoparticles, galectin-1 (Au-Gal1) bound with higher affinity to Jurkat cells to promote CD45 clustering and inhibition of its phosphatase activity, resulting in enhancement of apoptosis via caspase-dependent pathways. Au-Gal1 injected intra-articularly into rats with collagen-induced arthritis (CIA) promoted apoptosis of CD4+ T cells and reduced pro-inflammatory cytokine levels in the ankle joints as well as ameliorated clinical symptoms of arthritis. These observed therapeutic effects indicate that the multivalent structure of nanoparticle-ligands can regulate the distribution of cell surface receptors and subsequent intracellular signalling, and this may provide new insights into nanoparticle applications.


Asunto(s)
Apoptosis/efectos de los fármacos , Artritis Experimental/tratamiento farmacológico , Galectina 1/administración & dosificación , Oro/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Receptores de Superficie Celular/efectos de los fármacos , Animales , Articulación del Tobillo/diagnóstico por imagen , Articulación del Tobillo/patología , Artritis Experimental/inducido químicamente , Artritis Experimental/diagnóstico por imagen , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Galectina 1/química , Oro/química , Humanos , Inyecciones Intraarticulares , Células Jurkat/efectos de los fármacos , Células Jurkat/metabolismo , Masculino , Nanopartículas del Metal/química , Radiografía , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/metabolismo
4.
J Immunol ; 188(1): 68-76, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22156340

RESUMEN

Gold nanoparticles (GNPs), which are generally thought to be bio-inert and non-cytotoxic, have become one of the most ideal nanomaterials for medical applications. Once engulfed by phagocytes, the immunological effects of GNPs are still of concern and require detailed investigation. Therefore, this study explored the immunological significance of GNPs on TLR-mediated innate immunity in murine macrophages. GNP causes specific inhibition of TLR9 (CpG oligodeoxynucleotides; CpG-ODNs) signal in macrophages. The impaired CpG-ODN-induced TNF-α production is GNP concentration- and size-dependent in murine Raw264.7 cells: a GNP of 4 nm in size is more potent than a GNP of 11, 19, 35, or 45 nm in size. Consistent with cytokine inhibition, the CpG-ODN-induced phosphorylation of NF-κB and JNK as well as NF-κB activation are suppressed by GNPs. GNPs accumulate in lysosomes after phagocytosis and also increase TLR9-associated lysosomal cathepsin expression and activities, but this is irrelevant to TLR9 inhibition by GNPs in our studies. In addition, GNPs affected TLR9 translocation in response to CpG-ODNs and to phagosomes. Further exploring how GNPs inhibited TLR9 function, we found that GNPs could bind to high-mobility group box-1 (which is involved in the regulation of TLR9 signaling) inside the lysosomes. The current studies demonstrate that size-dependent inhibition of TLR9 function by GNP may be attributed to its binding to high-mobility group box-1.


Asunto(s)
Oro , Macrófagos/inmunología , Nanopartículas del Metal , Fagocitosis/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 9/inmunología , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Femenino , Proteína HMGB1/inmunología , Lisosomas/inmunología , Macrófagos/citología , Ratones , FN-kappa B/inmunología , Oligodesoxirribonucleótidos/farmacología , Tamaño de la Partícula , Fagocitosis/efectos de los fármacos , Fagosomas/inmunología , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
5.
ACS Nano ; 5(5): 3905-16, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21513334

RESUMEN

A new magnetic nanoparticle was synthesized in the form of Gd(3+)-chelated Fe(3)O(4)@SiO(2). The Fe(3)O(4) nanoparticle was octahedron-structured, was highly magnetic (∼94 emu/g), and was the core of an encapsulating mesoporous silica shell. DOTA-NHS molecules were anchored to the interior channels of the porous silica to chelate Gd(3+) ions. Because there were Gd(3+) ions within the silica shell, the transverse relaxivity increased 7-fold from 97 s(-1) mM(-1) of Fe(3)O(4) to 681 s(-1) mM(-1) of Gd(3+)-chelated Fe(3)O(4)@SiO(2) nanoparticles with r(2)/r(1) = 486. The large transversal relaxivity of the Gd(3+)-chelated Fe(3)O(4)@SiO(2) nanoparticles had an effective magnetic resonance imaging effect and clearly imaged lymph nodes. Physiological studies of liver, spleen, kidney, and lung tissue in mice infused with these new nanoparticles showed no damage and no cytotoxicity in Kupffer cells, which indicated that Gd(3+)-chelated Fe(3)O(4)@SiO(2) nanoparticles are biocompatible.


Asunto(s)
Gadolinio/química , Nanopartículas de Magnetita/química , Dióxido de Silicio/química , Animales , Quelantes/química , Medios de Contraste , Gadolinio/toxicidad , Nanopartículas de Magnetita/toxicidad , Ratones , Porosidad
6.
Mol Pharm ; 4(5): 713-22, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17708653

RESUMEN

Methotrexate (MTX), a stoichiometric inhibitor of dihydrofolate reductase, is a chemotherapeutic agent for treating a variety of neoplasms. Impairment of drug import into cells and increase in drug export from cells may render cells resistant to MTX. MTX, when locally administered in a soluble form, is rapidly absorbed through capillaries into the circulatory system, which may also account for therapeutic failure in patients. To retain MTX within tumor cells for longer duration and alter its pharmacokinetic behavior, we proposed a new formulation of MTX bound to the gold nanoparticle (AuNP) that serves as drug carriers. In this study, we developed the MTX-AuNP conjugate and examined its cytotoxic effect in vitro and antitumor effect in vivo. Spectroscopic examinations revealed that MTX can be directly bound onto AuNP via the carboxyl group (-COOH) to form the MTX-AuNP complex and kinetically released from the nanoparticles. The accumulation of MTX is faster and higher in tumor cells treated with MTX-AuNP than that treated with free MTX. Notably, MTX-AuNP shows higher cytotoxic effects on several tumor cell lines compared with an equal dose of free MTX. This can be attributed to the "concentrated effect" of MTX-AuNP. Administration of MTX-AuNP suppresses tumor growth in a mouse ascites model of Lewis lung carcinoma (LL2), whereas an equal dose of free MTX had no antitumor effect. In conclusion, these results suggest that by combining nanomaterials with anticancer drugs MTX-AuNP may be more effective than free MTX for cancer treatment.


Asunto(s)
Oro/química , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas del Metal/química , Metotrexato/química , Metotrexato/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Concentración 50 Inhibidora , Neoplasias Pulmonares/patología , Nanopartículas del Metal/ultraestructura , Metotrexato/farmacología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Estructura Molecular , Trasplante de Neoplasias
7.
Arthritis Rheum ; 56(2): 544-54, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17265489

RESUMEN

OBJECTIVE: Angiogenesis plays a part in the pathogenesis of rheumatoid arthritis (RA), and nanogold inhibits the activity of an angiogenic factor, vascular endothelial growth factor (VEGF). We therefore investigated whether intraarticular delivery of nanogold ameliorates collagen-induced arthritis (CIA) in rats. METHODS: Binding of 13-nm nanogold to VEGF in human RA synovial fluid (SF) and its effects on RA SF-induced endothelial cell proliferation and migration were assessed. Nanogold was administered intraarticularly to rats with CIA before the onset of arthritis. Progression of CIA was monitored by measures of clinical, radiologic, and histologic changes. In addition, the microvessel density and extent of infiltrating macrophages as well as levels of tumor necrosis factor alpha (TNFalpha) and interleukin-1beta (IL-1beta) in the ankle joints were determined. RESULTS: Nanogold bound to VEGF in RA SF, resulting in inhibition of RA SF-induced endothelial cell proliferation and migration. Significant reductions in ankle circumference, articular index scores, and radiographic scores were observed in the nanogold-treated rats with CIA compared with their control counterparts. In addition, the histologic score (of synovial hyperplasia, cartilage erosion, and leukocyte infiltration), microvessel density, macrophage infiltration, and levels of TNFalpha and IL-1beta were also significantly reduced in the ankle joints of nanogold-treated rats. CONCLUSION: Our results are the first to demonstrate that intraarticular administration of nanogold ameliorates the clinical course of CIA in rats. Nanogold exerted antiangiogenic activities and subsequently reduced macrophage infiltration and inflammation, which resulted in attenuation of arthritis. These results demonstrate proof of principle for the use of nanogold as a novel therapeutic agent for the treatment of RA.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Oro/uso terapéutico , Nanopartículas del Metal/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Animales , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/diagnóstico por imagen , Artritis Reumatoide/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Oro/administración & dosificación , Oro/farmacología , Humanos , Inflamación/tratamiento farmacológico , Inyecciones Intraarticulares , Macrófagos/efectos de los fármacos , Masculino , Nanopartículas del Metal/administración & dosificación , Neovascularización Patológica/patología , Radiografía , Ratas , Ratas Sprague-Dawley , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
8.
Biomaterials ; 26(34): 7183-91, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15964622

RESUMEN

In the current study, amine surface modified iron-oxide nanoparticles of 6 nm diameter without polymer coating were fabricated in an aqueous solution by organic acid modification as an adherent following chemical coprecipitation. Structure and the superparamagnetic property of magnetite nanoparticles were characterized by selected area electron diffraction (SAED) and superconducting quantum interference measurement device (SQUID). X-ray photoelectron spectrometer (XPS) and zeta potential measurements revealed cationic surface mostly decorated with terminal -NH(3)(+). This feature enables them to function as a magnetic carrier for nucleotides via electrostatic interaction. In addition, Fe(3)O(4)/trypsin conjugates with well-preserved functional activity was demonstrated. The nanoparticles displayed excellent in vitro biocompatibility. The NMR and the in vitro MRI measurements showed significantly reduced water proton relaxation times of both T(1) and T(2). Significantly reduced T(2) and T(2)*-weighted signal intensity were observed in a 1.5 T clinical MR imager. In vivo imaging contrast effect showed a fast and prolonged inverse contrast effect in the liver that lasted for more than 1 week. In addition, it was found that the spherical Fe(3)O(4) assembled as rod-like configuration through an aging process in aqueous solution at room temperature. Interestingly, TEM observation of the liver tissue revealed the rod-like shape but not the spherical-type nanoparticles being taken up by the Kupffer cells 120 h after tail vein infusion. Combining these results, we have demonstrated the potential applications of the newly synthesized magnetite nanoparticles in a broad spectrum of biomedical applications.


Asunto(s)
Medios de Contraste/química , Separación Inmunomagnética/métodos , Imagen por Resonancia Magnética/métodos , Micromanipulación/métodos , Nanotubos/química , Compuestos de Amonio Cuaternario/química , Animales , Biopolímeros/análisis , Biopolímeros/química , Células COS , Chlorocebus aethiops , Materiales Biocompatibles Revestidos/química , Coloides/química , Aumento de la Imagen/métodos , Ensayo de Materiales , Nanotubos/ultraestructura , Tamaño de la Partícula , Soluciones , Agua/química
9.
Biomaterials ; 26(7): 729-38, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15350777

RESUMEN

A newly developed non-polymer coated Fe(3)O(4) nanoparticles showing well-dispersion were synthesized using Fe(II) and Fe(III) salt chemical coprecipitation with tetramethylammonium hydroxide (N(CH(3))(4)OH) in an aqueous solution. Transmission electron microscopy (TEM), X-ray diffraction (XRD), Fourier transform infrared spectrometer (FT-IR), X-ray photoelectron spectrometer (XPS) and superconducting quantum interference measurement device (SQUID) measurements were employed to investigate the iron oxide properties. The resulting iron oxide particles were manipulated to be as small as 9 nm diameter in size. Based on FT-IR and X-ray photoelectron spectrometer results, it is suggested that the surfaces of the magnetite (Fe(3)O(4)) particles are covered with hydroxide (-OH) groups incorporated with (CH(3))(4)N(+) through electrostatic interaction. The in vitro cytotoxicity test revealed that the magnetite particles exhibited excellent biocompatibility, suggesting that they may be further explored for biomedical applications. NMR measurements revealed significantly reduced water proton relaxation times T1 and T2. The MR images of the nanoparticles in water, serum, and whole blood were investigated using a 1.5 T clinical MR imager. Significant reduction of the background medium signal was achieved in the T2-weighted and the T2*-weighted sequence especially in the serum and whole blood. Combining the advantage of MRI signal contrast, the non-polymer-coated surface chemistry for distinct bioconjugation and the homogenous nanometer size for better controlled biodistribution, these preliminary experiments demonstrated the potential of the as-synthesized magnetite material in functional molecular imaging for biomedical research and clinical diagnosis.


Asunto(s)
Materiales Biocompatibles/química , Compuestos Férricos/química , Ensayo de Materiales , Nanotubos/química , Nanotubos/ultraestructura , Agua/química , Animales , Ingeniería Biomédica/métodos , Células COS , Chlorocebus aethiops , Coloides/química , Compuestos Férricos/toxicidad , Hemólisis/efectos de los fármacos , Humanos , Nanotubos/toxicidad , Tamaño de la Partícula
10.
Vaccine ; 20(7-8): 1186-95, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11803081

RESUMEN

We describe a simple, efficient two-step method for construction of glycoprotein D (gD)-negative pseudorabies virus (PrV) carrying transgenes inserted in place of the gD gene. The first step was the use of the thymidine kinase (TK) gene of herpes simplex virus (HSV) for insertional inactivation of the gD gene in a PrV mutant deficient in both TK and glycoprotein E (gE). The gD-negative, HSV-TK-positive mutant could be selected in HAT medium. The second step was substitution of HSV-TK with other genes of interest. The resultant gD/gE/TK-negative mutant was easily isolated by acyclovir selection. The expression of the transgene was detectable in vivo and the antibody responses against both inserted antigens and PrV were induced. The protective efficacy of the gD/gE/TK-negative PrV against lethal PrV challenge was also demonstrated. This PrV mutant carrying immunogenic proteins from unrelated porcine pathogens may be tested as a multivalent vaccine candidate for swine.


Asunto(s)
Vectores Genéticos , Herpesvirus Suido 1/genética , Timidina Quinasa/genética , Vacunas Sintéticas/biosíntesis , Proteínas del Envoltorio Viral/genética , Animales , Anticuerpos Antivirales/análisis , Línea Celular , Herpesvirus Suido 1/inmunología , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Músculo Esquelético/metabolismo , Simplexvirus/enzimología , Porcinos , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...