Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Arthritis Res Ther ; 23(1): 265, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34696809

RESUMEN

BACKGROUND: The development and optimization of therapies for rheumatoid arthritis (RA) is currently hindered by a lack of methods for early non-invasive monitoring of treatment response. Annexin A2, an inflammation-associated protein whose presence and phosphorylation levels are upregulated in RA, represents a potential molecular target for tracking RA treatment response. METHODS: LS301, a near-infrared dye-peptide conjugate that selectively targets tyrosine 23-phosphorylated annexin A2 (pANXA2), was evaluated for its utility in monitoring disease progression, remission, and early response to drug treatment in mouse models of RA by fluorescence imaging. The intraarticular distribution and localization of LS301 relative to pANXA2 was determined by histological and immunohistochemical methods. RESULTS: In mouse models of spontaneous and serum transfer-induced inflammatory arthritis, intravenously administered LS301 showed selective accumulation in regions of joint pathology including paws, ankles, and knees with positive correlation between fluorescent signal and disease severity by clinical scoring. Whole-body near-infrared imaging with LS301 allowed tracking of spontaneous disease remission and the therapeutic response after dexamethasone treatment. Histological analysis showed preferential accumulation of LS301 within the chondrocytes and articular cartilage in arthritic mice, and colocalization was observed between LS301 and pANXA2 in the joint tissue. CONCLUSIONS: We demonstrate that fluorescence imaging with LS301 can be used to monitor the progression, remission, and early response to drug treatment in mouse models of RA. Given the ease of detecting LS301 with portable optical imaging devices, the agent may become a useful early treatment response reporter for arthritis diagnosis and drug evaluation.


Asunto(s)
Anexina A2 , Artritis Experimental , Artritis Reumatoide , Animales , Artritis Experimental/diagnóstico por imagen , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/diagnóstico por imagen , Artritis Reumatoide/tratamiento farmacológico , Condrocitos , Ratones , Imagen Óptica , Tirosina
3.
Nanomedicine ; 11(3): 601-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25652900

RESUMEN

High-relaxivity T1-weighted (T1w) MR molecular imaging nanoparticles typically present high surface gadolinium payloads that can elicit significant acute complement activation (CA). The objective of this research was to develop a high T1w contrast nanoparticle with improved safety. We report the development, optimization, and characterization of a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC; 138±10 (Dav)/nm; PDI: 0.06; zeta: -27±2 mV). High r1 particulate relaxivity with minute additions of Gd-DOTA-lipid conjugate to the MnOL nanocolloid surface achieved an unexpected paramagnetic synergism. This hybrid MnOL-Gd NC provided optimal MR TSE signal intensity at 5 nM/voxel and lower levels consistent with the level expression anticipated for sparse biomarkers, such as neovascular integrins. MnOL NC produced optimal MR TSE signal intensity at 10 nM/voxel concentrations and above. Importantly, MnOL-Gd NC avoided acute CA in vitro and in vivo while retaining minimal transmetallation risk. From the clinical editor: The authors developed a gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC) in this study. These were used as a high-relaxivity paramagnetic MR molecular imaging agent in experimental models. It was shown that MnOL-Gd NC could provide high T1w MR contrast for targeted imaging. As the level of gadolinium used was reduced, there was also reduced risk of systemic side effects from complement activation.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Medios de Contraste , Gadolinio , Imagen por Resonancia Magnética , Manganeso , Nanopartículas , Animales , Biomarcadores/sangre , Coloides , Medios de Contraste/efectos adversos , Medios de Contraste/química , Medios de Contraste/farmacología , Evaluación Preclínica de Medicamentos , Gadolinio/efectos adversos , Gadolinio/química , Gadolinio/farmacología , Manganeso/efectos adversos , Manganeso/química , Manganeso/farmacología , Ratones , Nanopartículas/efectos adversos , Nanopartículas/química
4.
ACS Nano ; 8(7): 7305-17, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-24941020

RESUMEN

Antiangiogenesis has been extensively explored for the treatment of a variety of cancers and certain inflammatory processes. Fumagillin, a mycotoxin produced by Aspergillus fumigatus that binds methionine aminopeptidase 2 (MetAP-2), is a potent antiangiogenic agent. Native fumagillin, however, is poorly soluble and extremely unstable. We have developed a lipase-labile fumagillin prodrug (Fum-PD) that eliminated the photoinstability of the compound. Using αvß3-integrin-targeted perfluorocarbon nanocarriers to deliver Fum-PD specifically to angiogenic vessels, we effectively suppressed clinical disease in an experimental model of rheumatoid arthritis (RA). The exact mechanism by which Fum-PD-loaded targeted nanoparticles suppressed inflammation in experimental RA, however, remained unexplained. We herein present evidence that Fum-PD nanotherapy indirectly suppresses inflammation in experimental RA through the local production of endothelial nitric oxide (NO). Fum-PD-induced NO activates AMP-activated protein kinase (AMPK), which subsequently modulates macrophage inflammatory response. In vivo, NO-induced AMPK activation inhibits mammalian target of rapamycin (mTOR) activity and enhances autophagic flux, as evidenced by p62 depletion and increased autolysosome formation. Autophagy in turn mediates the degradation of IkappaB kinase (IKK), suppressing the NF-κB p65 signaling pathway and inflammatory cytokine release. Inhibition of NO production by N(G)-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, reverses the suppression of NF-κB-mediated inflammatory response induced by Fum-PD nanotherapy. These unexpected results uncover an activity of Fum-PD nanotherapy that may be further explored in the treatment of angiogenesis-dependent diseases.


Asunto(s)
Ciclohexanos/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Ácidos Grasos Insaturados/metabolismo , Macrófagos/efectos de los fármacos , Nanomedicina , Óxido Nítrico/metabolismo , Profármacos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/metabolismo , Animales , Artritis/tratamiento farmacológico , Artritis/inmunología , Artritis/metabolismo , Artritis/patología , Ciclohexanos/química , Citocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Ácidos Grasos Insaturados/química , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Lipasa/metabolismo , Macrófagos/citología , Masculino , Ratones , Nanopartículas , Profármacos/metabolismo , Profármacos/uso terapéutico , Sesquiterpenos/química , Sesquiterpenos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo
5.
Biomaterials ; 33(33): 8632-40, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22922023

RESUMEN

Nanoparticle-based therapeutics are emerging technologies that have the potential to greatly impact the treatment of many human diseases. However, drug instability and premature release from the nanoparticles during circulation currently preclude clinical translation. Herein, we use a lipase-labile (Sn 2) fumagillin prodrug platform coupled with a unique lipid surface-to-surface targeted delivery mechanism, termed contact-facilitated drug delivery, to counter the premature drug release and overcome the inherent photo-instability of fumagillin, an established anti-angiogenic agent. We show that α(v)ß(3)-integrin targeted fumagillin prodrug nanoparticles, administered at 0.3 mg of fumagillin prodrug/kg of body weight suppress the clinical disease indices of KRN serum-mediated arthritis in a dose-dependent manner when compared to treatment with the control nanoparticles with no drug. This study demonstrates the effectiveness of this lipase-labile prodrug nanocarrier in a relevant preclinical model that approximates human rheumatoid arthritis. The lipase-labile prodrug paradigm offers a translatable approach that is broadly applicable to many targeted nanosystems and increases the translational potential of this platform for many diseases.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Ciclohexanos/metabolismo , Ciclohexanos/uso terapéutico , Ácidos Grasos Insaturados/metabolismo , Ácidos Grasos Insaturados/uso terapéutico , Lipasa/metabolismo , Nanopartículas/química , Animales , Ciclohexanos/química , Electroforesis , Ensayo de Inmunoadsorción Enzimática , Ácidos Grasos Insaturados/química , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL , Sesquiterpenos/química , Sesquiterpenos/metabolismo , Sesquiterpenos/uso terapéutico
6.
FASEB J ; 23(9): 2978-85, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19376816

RESUMEN

The purpose of this study was to assess whether an alternative treatment approach that targets angiogenesis, delivered through ligand-targeted nanotherapy, would ameliorate inflammatory arthritis. Arthritis was induced using the K/BxN mouse model of inflammatory arthritis. After arthritis was clearly established, mice received three consecutive daily doses of alpha(v)beta(3)-targeted fumagillin nanoparticles. Control groups received no treatment or alpha(v)beta(3)-targeted nanoparticles without drugs. Disease score and paw thickness were measured daily. Mice that received alpha(v)beta(3)-targeted fumagillin nanoparticles showed a significantly lower disease activity score (mean score of 1.4+/-0.4; P<0.001) and change in ankle thickness (mean increase of 0.17+/-0.05 mm; P<0.001) 7 d after arthritis induction, whereas the group that received alpha(v)beta(3)-targeted nanoparticles without drugs exhibited a mean arthritic score of 9.0 +/- 0.3 and mean change in ankle thickness of 1.01 +/- 0.09 mm. Meanwhile, the group that received no treatment showed a mean arthritic score of 9.8 +/- 0.5 and mean change in ankle thickness of 1.05 +/- 0.10 mm. Synovial tissues from animals treated with targeted fumagillin nanoparticles also showed significant decrease in inflammation and angiogenesis and preserved proteoglycan integrity. Ligand-targeted nanotherapy to deliver antiangiogenic agents may represent an effective way to treat inflammatory arthritis.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Ciclohexanos/uso terapéutico , Ácidos Grasos Insaturados/uso terapéutico , Integrina alfaVbeta3/efectos de los fármacos , Animales , Artritis Experimental/patología , Ciclohexanos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Ácidos Grasos Insaturados/farmacología , Inflamación/tratamiento farmacológico , Ratones , Nanopartículas/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Sesquiterpenos/farmacología , Sesquiterpenos/uso terapéutico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA