Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Nanobiotechnology ; 21(1): 12, 2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36635755

RESUMEN

Despite the development of therapeutic modalities to treat cancer, multidrug resistance (MDR) and incomplete destruction of deeply embedded lung tumors remain long-standing problems responsible for tumor recurrence and low survival rates. Therefore, developing therapeutic approaches to treat MDR tumors is necessary. In this study, nanodrugs with enhanced intracellular drug internalization were identified by the covalent bonding of carbon nanotubes of a specific nano size and doxorubicin (DOX). In addition, carbon nanotube conjugated DOX (CNT-DOX) sustained in the intracellular environment in multidrug-resistant tumor cells for a long time causes mitochondrial damage, suppresses ATP production, and results in the effective therapeutic effect of drug-resistant tumors. This study identified that H69AR lung cancer cells, an adriamycin (DOX) drug-resistant tumor cell line, did not activate drug resistance function on designed nano-anticancer drugs with a specific nano size. In summary, this study identified that the specific size of the nanodrug in combination with DOX overcame multidrug-resistant tumors by inducing selective accumulation in tumor cells and inhibiting ATP by mitochondrial damage.


Asunto(s)
Neoplasias Pulmonares , Nanopartículas , Nanotubos de Carbono , Humanos , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Línea Celular Tumoral , Nanopartículas/uso terapéutico , Adenosina Trifosfato
2.
Nanotechnology ; 28(46): 465102, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-29053471

RESUMEN

Although activating apoptosis in cancer cells by targeting the mitochondria is an effective strategy for cancer therapy, insufficient targeting of the mitochondria in cancer cells restricts the availability in clinical treatment. Here, we report on a polyethylene glycol-coated carbon nanotube (CNT)-ABT737 nanodrug that improves the mitochondrial targeting of lung cancer cells. The polyethylene glycol-coated CNT-ABT737 nanodrug internalized into the early endosomes via macropinocytosis and clathrin-mediated endocytosis in advance of early endosomal escape and delivered into the mitochondria. Cytosol release of the nanodrug led to apoptosis of lung cancer cells by abruption of the mitochondrial membrane potential, inducing Bcl-2-mediated apoptosis and generating intracellular reactive oxygen species. As such, this study provides an effective strategy for increasing the anti-lung cancer efficacy by increasing mitochondria accumulation rate of cytosol released anticancer nanodrugs.


Asunto(s)
Compuestos de Bifenilo/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Mitocondrias/metabolismo , Nitrofenoles/farmacología , Polietilenglicoles/química , Sulfonamidas/farmacología , Células A549 , Compuestos de Bifenilo/química , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Endosomas/química , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Nanotubos de Carbono/química , Nitrofenoles/química , Piperazinas/química , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/química
3.
Acta Pharmacol Sin ; 38(1): 90-99, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27890918

RESUMEN

Mast cells are important effector cells in immunoglobulin (Ig) E-mediated allergic reactions such as asthma, atopic dermatitis and rhinitis. Vanillic acid, a natural product, has shown anti-oxidant and anti-inflammatory activities. In the present study, we investigated the anti-allergic inflammatory effects of ortho-vanillic acid (2-hydroxy-3-methoxybenzoic acid, o-VA) that was a derivative of vanillic acid isolated from Amomum xanthioides. In mouse anaphylaxis models, oral administration of o-VA (2, 10, 50 mg/kg) dose-dependently attenuated ovalbumin-induced active systemic anaphylaxis and IgE-mediated cutaneous allergic reactions such as hypothermia, histamine release, IgE production and vasodilation; administration of o-VA also suppressed the mast cell degranulator compound 48/80-induced anaphylaxis. In cultured mast cell line RBL-2H3 and isolated rat peritoneal mast cells in vitro, pretreatment with o-VA (1-100 µmol/L) dose-dependently inhibited DNP-HSA-induced degranulation of mast cells by decreasing the intracellular free calcium level, and suppressed the expression of pro-inflammatory cytokines TNF-α and IL-4. Pretreatment of RBL-2H3 cells with o-VA suppressed DNP-HSA-induced phosphorylation of Lyn, Syk, Akt, and the nuclear translocation of nuclear factor-κB. In conclusion, o-VA suppresses the mast cell-mediated allergic inflammatory response by blocking the signaling pathways downstream of high affinity IgE receptor (FcεRI) on the surface of mast cells.


Asunto(s)
Anafilaxia/tratamiento farmacológico , Anafilaxia/inmunología , Benzoatos/farmacología , Benzoatos/uso terapéutico , Hipersensibilidad/tratamiento farmacológico , Mastocitos/efectos de los fármacos , Receptores de IgE/inmunología , Ácido Vanílico/análogos & derivados , Anafilaxia/inducido químicamente , Animales , Calcio/metabolismo , Degranulación de la Célula/efectos de los fármacos , Células Cultivadas , Dinitrofenoles/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Hipersensibilidad/inmunología , Inmunoglobulina E/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Masculino , Mastocitos/inmunología , Ratones , FN-kappa B/metabolismo , Ovalbúmina/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Ratas , Receptores de IgE/antagonistas & inhibidores , Albúmina Sérica/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , p-Metoxi-N-metilfenetilamina/antagonistas & inhibidores
4.
J Appl Toxicol ; 37(5): 554-562, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27682001

RESUMEN

Perfluorooctanoic acid (PFOA) has wide applications, including as a raw material for converted paper and packaging products. With the widespread use of PFOA, concerns regarding its potential environmental and health impacts have increased. In spite of the known hepatotoxicity and genotoxicity of PFOA, correlation with PFOA and allergic inflammation is not well known. In this study, the effect of PFOA on the degranulation of mast cells and mast cell-mediated allergic inflammation in the presence of FcεRI cross-linking was evaluated. In immunoglobulin (Ig) E-stimulated mast cells, PFOA increased the release of histamine and ß-hexosaminidase by the up-regulation of intracellular calcium levels. PFOA enhanced gene expression of several pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and IL-8 by the activation of nuclear factor (NF)-κB in IgE-stimulated mast cells. Also, PFOA exacerbated allergic symptoms via hypothermia, and an increase of serum histamine, TNF-α, IgE and IgG1 in the ovalbumin-induced systemic anaphylaxis. The present data indicate that PFOA aggravated FcɛRI-mediated mast cell degranulation and allergic symptoms. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Caprilatos/toxicidad , Degranulación de la Célula/efectos de los fármacos , Fluorocarburos/toxicidad , Hipersensibilidad/patología , Inflamación/patología , Mastocitos/efectos de los fármacos , Anafilaxia/inducido químicamente , Anafilaxia/patología , Animales , Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocinas/biosíntesis , Liberación de Histamina/efectos de los fármacos , Inmunoglobulina E/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Masculino , Mastocitos/patología , Ratones , Ratones Endogámicos ICR , beta-N-Acetilhexosaminidasas/metabolismo
5.
Toxicol Appl Pharmacol ; 287(2): 119-127, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25981167

RESUMEN

As the importance of allergic disorders such as atopic dermatitis and allergic asthma, research on potential drug candidates becomes more necessary. Mast cells play an important role as initiators of allergic responses through the release of histamine; therefore, they should be the target of pharmaceutical development for the management of allergic inflammation. In our previous study, anti-allergic effect of extracts of Amomum xanthioides was demonstrated. To further investigate improved candidates, 1,2,4,5-tetramethoxybenzene (TMB) was isolated from methanol extracts of A. xanthioides. TMB dose-dependently attenuated the degranulation of mast cells without cytotoxicity by inhibiting calcium influx. TMB decreased the expression of pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin (IL)-4 at both the transcriptional and translational levels. Increased expression of these cytokines was caused by translocation of nuclear factor-κB into the nucleus, and it was hindered by suppressing activation of IκB kinase complex. To confirm the effect of TMB in vivo, the ovalbumin (OVA)-induced active systemic anaphylaxis (ASA) and IgE-mediated passive cutaneous anaphylaxis (PCA) models were used. In the ASA model, hypothermia was decreased by oral administration of TMB, which attenuated serum histamine, OVA-specific IgE, and IL-4 levels. Increased pigmentation of Evans blue was reduced by TMB in a dose-dependent manner in the PCA model. Our results suggest that TMB is a possible therapeutic candidate for allergic inflammatory diseases that acts through the inhibition of mast cell degranulation and expression of pro-inflammatory cytokines.


Asunto(s)
Anisoles/farmacología , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Amomum , Animales , Degranulación de la Célula/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Hipersensibilidad , Quinasa I-kappa B/biosíntesis , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , Mastocitos/efectos de los fármacos , Ratones , Extractos Vegetales/farmacología , Ratas , Ratas Sprague-Dawley
6.
Nanotechnology ; 26(32): 325101, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26202827

RESUMEN

Surface chemistry and nanoscale surface morphology are both influential factors for cell adhesion, growth, and differentiation. In particular, cell migration is one of the major markers of initial immune response activation to implanted biomaterials. Despite their indication, it has been difficult to directly examine macrophages on nanoscale materials, because most nanomaterials possess greater thicknesses than nanoscale. This study developed transparent films comprising a carbon nanotube and polymer composite with controlled surface stiffness and nanoscale roughness. As nanoscale surface topography can incite immune cell activation, analysis of the real-time cell migration (including velocity) of macrophages due to changes in nanoscale surface topography of a biopolymer can support the direct relationship between initial macrophage dynamics and corresponding pro-inflammatory responses. Through real-time analysis, we have identified that surface chemistry and surface nanoscale topography are both independent factors mediating macrophage interactions, and, thus, immune cell behavior can be further controlled by the systematic variation of nanoscale surface topography for a given surface chemistry. Considering that the initial immune response can determine the fate and lifetime of implanted biomaterials, this study presents the direct relationship between initial macrophage dynamics and subsequent inflammatory cytokine release on transparent carbon nanotube polymer composites.


Asunto(s)
Movimiento Celular , Citocinas/análisis , Macrófagos/fisiología , Nanotubos de Carbono , Animales , Adhesión Celular , Línea Celular , Supervivencia Celular , Mediadores de Inflamación/análisis , Ratones Endogámicos BALB C , Propiedades de Superficie
7.
Int J Nanomedicine ; 19: 2529-2552, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38505170

RESUMEN

The blood-brain barrier (BBB) and blood-tumor barrier (BTB) pose substantial challenges to efficacious drug delivery for glioblastoma multiforme (GBM), a primary brain tumor with poor prognosis. Nanoparticle-based combinational strategies have emerged as promising modalities to overcome these barriers and enhance drug penetration into the brain parenchyma. This review discusses various nanoparticle-based combinatorial approaches that combine nanoparticles with cell-based drug delivery, viral drug delivery, focused ultrasound, magnetic field, and intranasal drug delivery to enhance drug permeability across the BBB and BTB. Cell-based drug delivery involves using engineered cells as carriers for nanoparticles, taking advantage of their intrinsic migratory and homing capabilities to facilitate the transport of therapeutic payloads across BBB and BTB. Viral drug delivery uses engineered viral vectors to deliver therapeutic genes or payloads to specific cells within the GBM microenvironment. Focused ultrasound, coupled with microbubbles or nanoparticles, can temporarily disrupt the BBB to increase drug permeability. Magnetic field-guided drug delivery exploits magnetic nanoparticles to facilitate targeted drug delivery under an external magnetic field. Intranasal drug delivery offers a minimally invasive avenue to bypass the BBB and deliver therapeutic agents directly to the brain via olfactory and trigeminal pathways. By combining these strategies, synergistic effects can enhance drug delivery efficiency, improve therapeutic efficacy, and reduce off-target effects. Future research should focus on optimizing nanoparticle design, exploring new combination strategies, and advancing preclinical and clinical investigations to promote the translation of nanoparticle-based combination therapies for GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Humanos , Barrera Hematoencefálica , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Sistemas de Liberación de Medicamentos , Encéfalo/metabolismo , Nanopartículas/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Microambiente Tumoral
8.
Adv Mater ; 36(11): e2304333, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38096399

RESUMEN

Current clinical therapeutic efficacy for the treatment of osteo- and rheumatoid-arthritis is obviously limited. Although mesenchymal stem cells (MSCs) are considered as a source of promising regenerative therapy, un-modified or genetically engineered MSCs injected in vivo restrict their clinical utility because of the low drug efficacy and unpredicted side effect, respectively. Herein, a strategy to enhance the migration efficacy of MSCs to inflamed joints via an inflammation-mediated education process is demonstrated. To reinforce the limited anti-inflammatory activity of MSCs, gold nanostar loaded with triamcinolone is conjugated to MSC. Furthermore, near-infrared laser-assisted photothermal therapy (PTT) induced by gold nanostar significantly elevates the anti-inflammatory efficacy of the developed drugs, even in advanced stage arthritis model. An immunological regulation mechanism study of PTT is first suggested in this study; the expression of the interleukin 22 receptor, implicated in the pathogenesis of arthritis, is downregulated in T lymphocytes by PTT, and Th17 differentiation from naïve CD4 T cell is inhibited. Collectively, inflammation-targeting MSCs conjugated with triamcinolone-loaded gold nanostar (Edu-MSCs-AuS-TA) promote the repolarization of macrophages and decrease neutrophil recruitment in joints. In addition, Edu-MSCs-AuS-TA significantly alleviate arthritis-associated pain, improve general locomotor activity, and more importantly, induce cartilage regeneration even for severe stages of arthritis model.


Asunto(s)
Artritis Reumatoide , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Inflamación/metabolismo , Triamcinolona/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antiinflamatorios/metabolismo , Oro
9.
ACS Appl Mater Interfaces ; 16(10): 12217-12231, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480984

RESUMEN

Psoriasis, a prevalent chronic inflammatory skin ailment affecting approximately 2-3% of the global population, is characterized by persistent symptoms. Dexamethasone, a primary corticosteroid for treating psoriasis, demonstrates notable efficacy; however, its limited skin permeation results in documented adverse effects. To address this, the presented study employed a novel strategy to conjugate gold nanorod and dexamethasone and evaluate their potential for mitigating psoriatic inflammation using an imiquimod-induced mouse model and human skin cells. Our findings revealed enhanced cutaneous penetration of gold nanorod and dexamethasone conjugates compared with that of dexamethasone, owing to superior skin penetration. Gold nanorod and dexamethasone conjugates demonstrated an optimal pharmacological impact at minimal dosages without toxicity during extended use. To further enhance the effectiveness of gold nanorod and dexamethasone conjugates, 808 nm near-infrared laser irradiation, which reacts to gold, was additionally applied to achieve thermal elevation to expedite drug skin penetration. Supplementary laser irradiation at 808 nm significantly ameliorated psoriatic symptoms following deep gold nanorod and dexamethasone conjugates penetration. This corresponded with restored peroxisome proliferator-activated receptor-γ levels and accelerated dexamethasone release from the gold nanorod and dexamethasone conjugates complex. These findings highlight the potential of gold nanorod and dexamethasone conjugates to enhance drug penetration through dermal layers, thereby aiding psoriasis treatment. Moreover, its compatibility with photothermal therapy offers prospects for novel therapeutic interventions across various inflammatory skin disorders.


Asunto(s)
Nanotubos , Psoriasis , Animales , Ratones , Humanos , Terapia Fototérmica , Oro/farmacología , Oro/uso terapéutico , Psoriasis/tratamiento farmacológico , Dexametasona/farmacología , Dexametasona/uso terapéutico , Inflamación/tratamiento farmacológico
10.
Inflammation ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38777857

RESUMEN

Mast cells are primary cells initiating allergic inflammation by the release of various allergic mediators, such as histamine and pro-inflammatory cytokines. Aspalathin (ASP) is the predominant flavonoid found exclusively in rooibos, an herb that has been traditionally used in allergy relief therapy. In the present study, we investigated the beneficial effects of ASP on mast cell-mediated allergic inflammation. For in vivo study, two well-known mast cell-mediated local and systemic allergic inflammation mouse models were used: passive cutaneous anaphylaxis (PCA) and active systemic anaphylaxis mouse models (ASA). Oral administration of ASP dose-dependently suppressed immunoglobulin (Ig)E-mediated PCA responses evidenced by Evans blue extravasation, ear thickening, and mast cell degranulation. ASP also significantly mitigated ovalbumin-induced ASA responses, including hypothermia, histamine secretion, and the production of IgE and interleukin-4. Notably, ASP was more effective in suppressing allergic inflammation than nothofagin, another prominent flavonoid known as an anti-allergic component of rooibos. The regulatory mechanism of mast cell activation by ASP was clarified using mast cell line and primary cultured mast cells (RBL-2H3 and mouse bone marrow-derived mast cells). ASP reduced IgE-stimulated mast cells degranulation and intracellular calcium influx by the inhibition of FcεRI signaling pathway (Lyn, Fyn, and Syk). Moreover, ASP reduced pro-inflammatory cytokine expressions by inhibiting two major transcription factors, nuclear factor of activated T cells and nuclear factor-κB. Collectively, we proposed that ASP could be a potential therapeutic candidate for the treatment of mast cell-mediated allergic inflammatory diseases.

11.
Biomed Pharmacother ; 176: 116911, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38861857

RESUMEN

Atopic dermatitis (AD) is a globally increasing chronic inflammatory skin disease with limited and potentially side-effect-prone treatment options. Monotropein is the predominant iridoid glycoside in Morinda officinalis How roots, which has previously shown promise in alleviating AD symptoms. This study aimed to systematically investigate the pharmacological effects of monotropein on AD using a 2, 4-dinitrochlorobenzene (DNCB)/Dermatophagoides farinae extract (DFE)-induced AD mice and tumor necrosis factor (TNF)-α/interferon (IFN)-γ-stimulated keratinocytes. Oral administration of monotropein demonstrated a significant reduction in AD phenotypes, including scaling, erythema, and increased skin thickness in AD-induced mice. Histological analysis revealed a marked decrease in immune cell infiltration in skin lesions. Additionally, monotropein effectively downregulated inflammatory markers, encompassing pro-inflammatory cytokines, T helper (Th)1 and Th2 cytokines, and pro-inflammatory chemokines in skin tissues. Notably, monotropein also led to a considerable decrease in serum immunoglobulin (Ig)E and IgG2a levels. At a mechanistic level, monotropein exerted its anti-inflammatory effects by suppressing the phosphorylation of Janus kinase / signal transducer and activator of transcription proteins in both skin tissues of AD-induced mice and TNF-α/IFN-γ-stimulated keratinocytes. In conclusion, monotropein exhibited a pronounced alleviation of AD symptoms in the experimental models used. These findings underscore the potential application of monotropein as a therapeutic agent in the context of AD, providing a scientific basis for further exploration and development.


Asunto(s)
Dermatitis Atópica , Quinasas Janus , Queratinocitos , Transducción de Señal , Piel , Animales , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/patología , Dermatitis Atópica/inducido químicamente , Transducción de Señal/efectos de los fármacos , Ratones , Quinasas Janus/metabolismo , Piel/efectos de los fármacos , Piel/patología , Piel/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Citocinas/metabolismo , Ratones Endogámicos BALB C , Factores de Transcripción STAT/metabolismo , Humanos , Dinitroclorobenceno , Antiinflamatorios/farmacología , Femenino , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/patología , Inmunoglobulina E/sangre , Dermatophagoides farinae/inmunología , Iridoides/farmacología
12.
Toxicol Appl Pharmacol ; 269(1): 72-80, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23499868

RESUMEN

Atopic dermatitis (AD) and allergic contact dermatitis (ACD) are common allergic and inflammatory skin diseases caused by a combination of eczema, scratching, pruritus, and cutaneous sensitization with allergens. This paper examines whether oleanolic acid acetate (OAA) modulates AD and ACD symptoms by using an existing AD model based on the repeated local exposure of mite extract (Dermatophagoides farinae extract, DFE) and 2,4-dinitrochlorobenzene to the ears of BALB/c mice. In addition, the paper uses a 2,4-dinitrofluorobenzene-sensitized local lymph node assay (LLNA) for the ACD model. The oral administration of OAA over a four-week period attenuated AD symptoms in terms of decreased skin lesions, epidermal thickness, the infiltration of immune cells (CD4⁺ cells, eosinophils, and mast cells), and serum IgE, IgG2a, and histamine levels. The gene expression of Th1, Th2, Th17, and Th22 cytokines was reduced by OAA in the lymph node and ear tissue, and the LLNA verified that OAA suppressed ACD. The oral administration of OAA over a three-day period attenuated ACD symptoms in terms of ear thickness, lymphocyte proliferation, and serum IgG2a levels. The gene expression of Th1, Th2, and Th17 cytokines was reduced by OAA in the thymus and ear tissue. Finally, to define the underlying mechanism, this paper uses a TNF-α/IFN-γ-activated human keratinocyte (HaCaT) model. OAA inhibited the expression of cytokines and chemokines through the downregulation of NF-κB and MAPKs in HaCaT cells. Taken together, the results indicate that OAA inhibited AD and ACD symptoms, suggesting that OAA may be effective in treating allergic skin disorders.


Asunto(s)
Antialérgicos/farmacología , Dermatitis Alérgica por Contacto/prevención & control , Dermatitis Atópica/prevención & control , Ácido Oleanólico/farmacología , Piel/efectos de los fármacos , Administración Oral , Animales , Antialérgicos/administración & dosificación , Antígenos Dermatofagoides , Línea Celular , Citocinas/metabolismo , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/inmunología , Dermatitis Alérgica por Contacto/patología , Dermatitis Atópica/genética , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Dinitroclorobenceno , Modelos Animales de Enfermedad , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Ensayo del Nódulo Linfático Local , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Ácido Oleanólico/administración & dosificación , Piel/inmunología , Piel/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factores de Tiempo
13.
Int J Nanomedicine ; 18: 7267-7285, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38090362

RESUMEN

Purpose: Rheumatoid arthritis (RA) is a highly prevalent form of autoimmune disease that affects nearly 1% of the global population by causing severe cartilage damage and inflammation. Despite its prevalence, previous efforts to prevent the perpetuation of RA have been hampered by therapeutics' cytotoxicity and poor delivery to target cells. The present study exploited drug repositioning and nanotechnology to convert metformin, a widely used antidiabetic agent, into an anti-rheumatoid arthritis drug by designing poly(lactic-co-glycolic acid) (PLGA)-based spheres. Moreover, this study also explored the thermal responsiveness of the IL-22 receptor, a key regulator of Th-17, to incorporate photothermal therapy (PTT) into the nanodrug treatment. Materials and Methods: PLGA nanoparticles were synthesized using the solvent evaporation method, and metformin and indocyanine green (ICG) were encapsulated in PLGA in a dropwise manner. The nanodrug's in vitro anti-inflammatory properties were examined in J744 and FLS via real-time PCR. PTT was induced by an 808 nm near-infrared (NIR) laser, and the anti-RA effects of the nanodrug with PTT were evaluated in DBA/1 collagen-induced arthritis (CIA) mice models. Further evaluation of anti-RA properties was carried out using flow cytometry, immunofluorescence analysis, and immunohistochemical analysis. Results: The encapsulation of metformin into PLGA allowed the nanodrug to enter the target cells via macropinocytosis and clathrin-mediated endocytosis. Metformin-encapsulated PLGA (PLGA-MET) demonstrated promising anti-inflammatory effects by decreasing the expression of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α), increasing the expression of anti-inflammatory cytokines (IL-10 and IL-4), and promoting the polarization of M1 to M2 macrophages in J774 cells. The treatment of the nanodrug with PTT exhibited more potent anti-inflammatory effects than free metformin or PLGA-MET in CIA mice models. Conclusion: These results demonstrated that PLGA-encapsulated metformin treatment with PTT can effectively ameliorate inflammation in a spatiotemporal manner.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Metformina , Ratones , Animales , Terapia Fototérmica , Metformina/farmacología , Reposicionamiento de Medicamentos , Ratones Endogámicos DBA , Artritis Reumatoide/tratamiento farmacológico , Citocinas/metabolismo , Artritis Experimental/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
14.
Adv Mater ; 35(30): e2300934, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37114740

RESUMEN

Owing to the intrinsic ability of stem cells to target the tumor environment, stem-cell-membrane-functionalized nanocarriers can target and load active anticancer drugs. In this work, a strategy that focuses on stem cells that self-target pancreatic cancer cells is developed. In particular, malignant deep tumors such as pancreatic cancer cells, one of the intractable tumors that currently have no successful clinical strategy, are available for targeting and destruction. By gaining the targeting ability of stem cells against pancreatic tumor cells, stem cell membranes can encapsulate nano-polylactide-co-glycolide loaded with doxorubicin to target and reduce deep pancreatic tumor tissues. Considering the lack of known target proteins on pancreatic tumor cells, the suggested platform technology can be utilized for targeting any malignant tumors in which surface target receptors are unavailable.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias Pancreáticas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Células Madre , Línea Celular Tumoral , Neoplasias Pancreáticas
15.
Cells ; 12(11)2023 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-37296614

RESUMEN

(1) Background: This study aims to elucidate a novel non-transcriptional action of IRF3 in addition to its role as a transcription factor in mast cell activation and associated allergic inflammation; (2) Methods: For in vitro experiments, mouse bone-marrow-derived mast cells (mBMMCs) and a rat basophilic leukemia cell line (RBL-2H3) were used for investigating the underlying mechanism of IRF3 in mast-cell-mediated allergic inflammation. For in vivo experiments, wild-type and Irf3 knockout mice were used for evaluating IgE-mediated local and systemic anaphylaxis; (3) Results: Passive cutaneous anaphylaxis (PCA)-induced tissues showed highly increased IRF3 activity. In addition, the activation of IRF3 was observed in DNP-HSA-treated mast cells. Phosphorylated IRF3 by DNP-HSA was spatially co-localized with tryptase according to the mast cell activation process, and FcεRI-mediated signaling pathways directly regulated that activity. The alteration of IRF3 affected the production of granule contents in the mast cells and the anaphylaxis responses, including PCA- and ovalbumin-induced active systemic anaphylaxis. Furthermore, IRF3 influenced the post-translational processing of histidine decarboxylase (HDC), which is required for granule maturation; and (4) Conclusion: Through this study, we demonstrated the novel function of IRF3 as an important factor inducing mast cell activation and as an upstream molecule for HDC activity.


Asunto(s)
Anafilaxia , Receptores de IgE , Ratas , Ratones , Animales , Receptores de IgE/metabolismo , Mastocitos/metabolismo , Línea Celular , Inflamación/metabolismo
16.
Acta Pharm Sin B ; 2022 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-36438851

RESUMEN

Despite the global administration of approved COVID-19 vaccines (e.g., ChAdOx1 nCoV-19®, mRNA-1273®, BNT162b2®), the number of infections and fatalities continue to rise at an alarming rate because of the new variants such as Omicron and its subvariants. Including COVID-19 vaccines that are licensed for human use, most of the vaccines that are currently in clinical trials are administered via parenteral route. However, it has been proven that the parenteral vaccines do not induce localized immunity in the upper respiratory mucosal surface, and administration of the currently approved vaccines does not necessarily lead to sterilizing immunity. This further supports the necessity of a mucosal vaccine that blocks the main entrance route of COVID-19: nasal and oral mucosal surfaces. Understanding the mechanism of immune regulation of M cells and dendritic cells and targeting them can be another promising approach for the successful stimulation of the mucosal immune system. This paper reviews the basic mechanisms of the mucosal immunity elicited by mucosal vaccines and summarizes the practical aspects and challenges of nanotechnology-based vaccine platform development, as well as ligand hybrid nanoparticles as potentially effective target delivery agents for mucosal vaccines.

17.
Adv Sci (Weinh) ; 9(34): e2202800, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36180414

RESUMEN

Although plasma is a promising technology in various fields, its clinical application is restricted by several limitations. A cold atmospheric plasma (CAP) patch is fabricated to help overcome hurdles, especially when treating skin diseases. This patch has surface dielectric barrier discharge, which generates reactive oxygen species (ROS) and reactive nitrogen species (RNS) on a flexible polymer film surface on which the embedded electrode induces a locally strong electric field. The effect of the CAP patch on psoriasis is also evaluated. The distinct characteristics of psoriasis between the lesion and non-lesion area allow the CAP patch to be suitable for only lesion area for its treatment. The CAP patch induces the opening of calcium channels in keratinocytes, thereby restoring abnormal keratinocyte differentiation and the collapse of the tight junction; thus, alleviating psoriatic symptoms. In addition, the favorable effect is due to the induction of ROS/RNS by the CAP patch, not the electric field generated during plasma generation. The findings indicate that the proposed portable CAP patch can help treat inflammatory skin disorders, especially psoriasis. As this can be used easily as a combination therapy with existing drugs, it may help reduce side effects caused by existing drugs.


Asunto(s)
Gases em Plasma , Gases em Plasma/uso terapéutico , Antiinflamatorios
18.
Int J Nanomedicine ; 17: 4599-4617, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36199478

RESUMEN

Purpose: The protein corona surrounding nanoparticles has attracted considerable attention as it induces subsequent inflammatory responses. Although mesoporous silica nanoparticles (MSN) are commonly used in medicines, cosmetics, and packaging, the inflammatory effects of the MSN protein corona on the cutaneous system have not been investigated till date. Methods: We examined the greater plasma protein adsorption on MSN leads to serious inflammatory reactions in Dermatophagoides farinae extract (DFE)-induced mouse atopic dermatitis (AD)-like skin inflammation because of increased uptake by keratinocytes. Results: We compare the AD lesions induced by MSN and colloidal (non-porous) silica nanoparticles (CSN), which exhibit different pore architectures but similar dimensions and surface chemistry. MSN-corona treatment of severe skin inflammation in a DFE-induced in vivo AD model greatly increases mouse ear epidermal thickness and infiltration of immune cells compared with the CSN-corona treatment. Moreover, MSN-corona significantly increase AD-specific immunoglobulins, serum histamine, and Th1/Th2/Th17 cytokines in the ear and lymph nodes. MSN-corona induce more severe cutaneous inflammation than CSN by significantly decreasing claudin-1 expression. Conclusion: This study demonstrates the novel impact of the MSN protein corona in inducing inflammatory responses through claudin-1 downregulation and suggests useful clinical guidelines for MSN application in cosmetics and drug delivery systems.


Asunto(s)
Dermatitis Atópica , Nanopartículas , Corona de Proteínas , Adsorción , Animales , Claudina-1/uso terapéutico , Citocinas/metabolismo , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/tratamiento farmacológico , Histamina , Inmunoglobulina E , Inflamación/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/farmacología , Dióxido de Silicio/uso terapéutico
19.
Int J Nanomedicine ; 16: 8485-8507, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35002240

RESUMEN

Mesenchymal stem cells (MSCs) are considered a promising regenerative therapy due to their ability to migrate toward damaged tissues. The homing ability of MSCs is unique compared with that of non-migrating cells and MSCs are considered promising therapeutic vectors for targeting major cells in many pathophysiological sites. MSCs have many advantages in the treatment of malignant diseases, particularly rheumatoid arthritis (RA). RA is a representative autoimmune disease that primarily affects joints, and secreted chemokines in the joints are well recognized by MSCs following their migration to the joints. Furthermore, MSCs can regulate the inflammatory process and repair damaged cells in the joints. However, the functionality and migration ability of MSCs injected in vivo still show insufficient. The targeting ability and migration efficiency of MSCs can be enhanced by genetic engineering or modification, eg, overexpressing chemokine receptors or migration-related genes, thus maximizing their therapeutic effect. However, there are concerns about genetic changes due to the increased probability of oncogenesis resulting from genome integration of the viral vector, and thus, clinical application is limited. Furthermore, it is suspected that administering MSCs can promote tumor growth and metastasis in xenograft and orthotopic models. For this reason, MSC mimicking nanoencapsulations are an alternative strategy that does not involve using MSCs or bioengineered MSCs. MSC mimicking nanoencapsulations consist of MSC membrane-coated nanoparticles, MSC-derived exosomes and artificial ectosomes, and MSC membrane-fused liposomes with natural or genetically engineered MSC membranes. MSC mimicking nanoencapsulations not only retain the targeting ability of MSCs but also have many advantages in terms of targeted drug delivery. Specifically, MSC mimicking nanoencapsulations are capable of encapsulating drugs with various components, including chemotherapeutic agents, nucleic acids, and proteins. Furthermore, there are fewer concerns over safety issues on MSC mimicking nanoencapsulations associated with mutagenesis even when using genetically engineered MSCs, because MSC mimicking nanoencapsulations use only the membrane fraction of MSCs. Genetic engineering is a promising route in clinical settings, where nano-encapsulated technology strategies are combined. In this review, the mechanism underlying MSC homing and the advantages of MSC mimicking nanoencapsulations are discussed. In addition, genetic engineering of MSCs and MSC mimicking nanoencapsulation is described as a promising strategy for the treatment of immune-related diseases.


Asunto(s)
Artritis Reumatoide , Exosomas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Nanopartículas , Sistemas de Liberación de Medicamentos , Humanos
20.
Int J Nanomedicine ; 16: 4943-4957, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326636

RESUMEN

BACKGROUND: Understanding structural interactions between the active drug and conjugated nanoparticles is critical for optimizing intracellular drug transport and for increasing nano drug efficacy. In this regard, analyzing the conformational deformation of conjugated drugs surrounding nanoparticles is essential to understand the corresponding nanodrug efficacy. PURPOSE: The objective of this study is to present an optimal synthesis method for efficient drug delivery through a clear structural analysis of nanodrugs according to the type of conjugation. METHODS AND RESULTS: In this study, the structural variation of methotrexate (MTX) surrounding carbon nanotubes, depending on the type of conjugation style, such as covalent and non-covalent (PEGylation) bonds, was investigated. Specifically, covalent bonds of MTX surrounding CNTs induced greater structural deformation compared to non-covalent bonds (ie, PEGylated CNT). CONCLUSION: Greater changes in the structural variations of MTX analyzed by nuclear magnetic resonance (NMR) significantly improved the anti-inflammatory drug efficacy of human fibroblast-like synovial cells (FLS) via stable drug release in the extracellular environment and burst drug release under intracellular conditions.


Asunto(s)
Nanopartículas , Nanotubos de Carbono , Preparaciones Farmacéuticas , Sistemas de Liberación de Medicamentos , Humanos , Metotrexato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA