Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Drug Dev Ind Pharm ; 50(7): 671-686, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39099436

RESUMEN

OBJECTIVE: This study was aimed to develop a cationic lipoplex formulation loaded with Nintedanib and miR-29b (LP-NIN-miR) as an alternative approach in the combination therapy of idiopathic pulmonary dibrosis (IPF) by proving its additive anti-fibrotic therapeutic effects through in vitro lung fibrosis model. SIGNIFICANCE: This is the first research article reported that the LP-NIN-MIR formulations in the treatment of IPF. METHODS: To optimize cationic liposomes (LPs), quality by design (QbD) approach was carried out. Optimized blank LP formulation was prepared with DOTAP, CHOL, DOPE, and DSPE-mPEG 2000 at the molar ratio of 10:10:1:1. Nintedanib loaded LP (LPs-NIN) were produced by microfluidization method and were incubated with miR-29b at room temperature for 30 min to obtain LP-NIN-miR. To evaluate the cellular uptake of LP-NIN-miR, NIH/3T3 cells were treated with 20 ng.mL-1 transforming growth factor-ß1 (TGF-ß1) for 96 h to establish the in vitro IPF model and incubated with LP-NIN-miR for 48 h. RESULTS: The hydrodynamic diameter, polydispersity index (PDI), and zeta potential of the LP-NIN-miR were 87.3 ± 0.9 nm, 0.184 ± 0.003, and +24 ± 1 mV, respectively. The encapsulation efficiencies of Nintedanib and miR-29b were 99.8% ± 0.08% and 99.7% ± 1.2%, respectively. The results of the cytotoxicity study conducted with NIH/3T3 cells indicated that LP-NIN-miR is a safe delivery system. CONCLUSIONS: The outcome of the transfection study proved the additive anti-fibrotic therapeutic effect of LP-NIN-miR and suggested that lipoplexes are effective delivery systems for drug and nucleic acid to the NIH/3T3 cells in the treatment of IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática , Indoles , Liposomas , MicroARNs , MicroARNs/administración & dosificación , Liposomas/química , Indoles/administración & dosificación , Indoles/química , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Animales , Ratones , Células 3T3 NIH , Humanos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética
2.
AAPS PharmSciTech ; 24(8): 235, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37973629

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive, fibrotic interstitial lung disease with unclear etiology and increasing prevalence. Pulmonary administration can make the drug directly reach the lung lesion location and reduce systemic toxic and side effects. The effectiveness of lenalidomide (Len) liposomal lung delivery in idiopathic pulmonary fibrosis was investigated. Len liposomes (Len-Lip) were prepared from soybean lecithin, cholesterol (Chol), and medicine in different weight ratios by thin film hydration method. The Len-Lip were spherical in shape with an average size of 226.7 ± 1.389 nm. The liposomes with a higher negative zeta potential of around - 34 mV, which was conducive to improving stability by repelling each other. The drug loading and encapsulation rate were 2.42 ± 0.07% and 85.47 ± 2.42%. Len-Lip had little toxicity at the cellular level and were well taken up by cells. At bleomycin-induced pulmonary fibrosis model mice, inhalation Len-Lip could improve lung function and decrease lung hydroxyproline contents, and alleviate pulmonary fibrosis state. Inhalation Len-Lip provided a reference for the treatment of idiopathic pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Liposomas , Ratones , Animales , Liposomas/farmacología , Bleomicina/efectos adversos , Lenalidomida/farmacología , Lenalidomida/uso terapéutico , Pulmón , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/patología
3.
J Nanobiotechnology ; 20(1): 213, 2022 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-35524280

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease with pathophysiological characteristics of transforming growth factor-ß (TGF-ß), and reactive oxygen species (ROS)-induced excessive fibroblast-to-myofibroblast transition and extracellular matrix deposition. Macrophages are closely involved in the development of fibrosis. Nuclear factor erythroid 2 related factor 2 (Nrf2) is a key molecule regulating ROS and TGF-ß expression. Therefore, Nrf2 signaling modulation might be a promising therapy for fibrosis. The inhalation-based drug delivery can reduce systemic side effects and improve therapeutic effects, and is currently receiving increasing attention, but direct inhaled drugs are easily cleared and difficult to exert their efficacy. Therefore, we aimed to design a ROS-responsive liposome for the Nrf2 agonist dimethyl fumarate (DMF) delivery in the fibrotic lung. Moreover, we explored its therapeutic effect on pulmonary fibrosis and macrophage activation. RESULTS: We synthesized DMF-loaded ROS-responsive DSPE-TK-PEG@DMF liposomes (DTP@DMF NPs). DTP@DMF NPs had suitable size and negative zeta potential and excellent capability to rapidly release DMF in a high-ROS environment. We found that macrophage accumulation and polarization were closely related to fibrosis development, while DTP@DMF NPs could attenuate macrophage activity and fibrosis in mice. RAW264.7 and NIH-3T3 cells coculture revealed that DTP@DMF NPs could promote Nrf2 and downstream heme oxygenase-1 (HO-1) expression and suppress TGF-ß and ROS production in macrophages, thereby reducing fibroblast-to-myofibroblast transition and collagen production by NIH-3T3 cells. In vivo experiments confirmed the above findings. Compared with direct DMF instillation, DTP@DMF NPs treatment presented enhanced antifibrotic effect. DTP@DMF NPs also had a prolonged residence time in the lung as well as excellent biocompatibility. CONCLUSIONS: DTP@DMF NPs can reduce macrophage-mediated fibroblast-to-myofibroblast transition and extracellular matrix deposition to attenuate lung fibrosis by upregulating Nrf2 signaling. This ROS-responsive liposome is clinically promising as an ideal delivery system for inhaled drug delivery.


Asunto(s)
Fibrosis Pulmonar Idiopática , Factor 2 Relacionado con NF-E2 , Animales , Fibrosis , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Liposomas , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
4.
Nanomedicine ; 41: 102517, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35032629

RESUMEN

Mesenchymal stem cells (MSCs) are promising in idiopathic pulmonary fibrosis (IPF) therapy. However, low survival rate and ambiguous behavior of MSCs after transplantation impede their clinical translation. To this end, we have developed a new strategy to improve the survival rate and monitor the behavior of the transplanted MSCs simultaneously. In our strategy, nintedanib, a tyrosine kinase inhibitor, is employed to protect the human MSCs (hMSCs) from excessive oxidative stress responses and inflammatory environment in the damaged lung. Moreover, by labeling of the transplanted hMSCs with a computed tomography (CT) nanotracer, Au nanoparticles functionalized with polyethylenimine (PEI) and polyethylene glycol (PEG) (Au@PEI@PEG), in combination with red-emitting firefly luciferase (RfLuc), in vivo CT/bioluminescence (BL) dual-modal imaging tracking of the location, distribution, and survival of the transplanted hMSCs in presence of nintedanib were achieved, which facilitates the profound understanding of the role the stem cells play in IPF therapy.


Asunto(s)
Fibrosis Pulmonar Idiopática , Células Madre Mesenquimatosas , Nanopartículas del Metal , Oro , Humanos , Fibrosis Pulmonar Idiopática/diagnóstico por imagen , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Indoles , Luciferasas de Luciérnaga , Nanopartículas del Metal/uso terapéutico , Polietilenglicoles , Polietileneimina , Tomografía Computarizada por Rayos X
5.
Mol Pharm ; 18(11): 4140-4147, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34657437

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease with poor prognosis. Evidence has shown that vimentin is a key regulator of lung fibrogenesis. 99mTc-labeled N-acetylglucosamine-polyethyleneimine (NAG-PEI), a vimentin-targeting radiotracer, was used for the early diagnosis of IPF, and NAG-PEI was also used as a therapeutic small interfering RNA (siRNA) delivery vector for the treatment of IPF in this study. Single-photon emission-computed tomography (SPECT) imaging of bleomycin (BM)- and silica-induced IPF mice with 99mTc-labeled NAG-PEI was performed to visualize pulmonary fibrosis and monitor the treatment efficiency of siRNA-loaded NAG-PEI, lipopolysaccharide (LPS, a tolerogenic adjuvant), or zymosan (ZYM, an immunostimulant). The lung uptakes of 99mTc-NAG-PEI in the BM- and silica-induced IPF mice were clearly and directly correlated with IPF progression. The lung uptake of 99mTc-NAG-PEI in the NAG-PEI/TGF-ß1-siRNA treatment group or LPS treatment group was evidently lower than that in the control group, while the lung uptake of 99mTc-NAG-PEI was significantly higher in the ZYM treatment group compared to that in the control group. These results demonstrate that NAG-PEI is a potent MicroSPECT imaging-guided theranostic platform for IPF diagnosis and therapy.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , Radiofármacos/administración & dosificación , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores , Acetilglucosamina/administración & dosificación , Acetilglucosamina/química , Animales , Biodiversidad , Bleomicina/administración & dosificación , Bleomicina/toxicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/patología , Pulmón/diagnóstico por imagen , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Polietileneimina/administración & dosificación , Polietileneimina/química , ARN Interferente Pequeño/genética , Radiofármacos/química , Radiofármacos/farmacocinética , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/toxicidad , Tecnecio , Tomografía Computarizada de Emisión de Fotón Único , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/metabolismo
6.
Int J Mol Sci ; 22(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070506

RESUMEN

Concentration of hyaluronic acid (HA) in the lungs increases in idiopathic pulmonary fibrosis (IPF). HA is involved in the organization of fibrin, fibronectin, and collagen. HA has been proposed to be a biomarker of fibrosis and a potential target for antifibrotic therapy. Hyaluronidase (HD) breaks down HA into fragments, but is a subject of rapid hydrolysis. A conjugate of poloxamer hyaluronidase (pHD) was prepared using protein immobilization with ionizing radiation. In a model of bleomycin-induced pulmonary fibrosis, pHD decreased the level of tissue IL-1ß and TGF-ß, prevented the infiltration of the lung parenchyma by CD16+ cells, and reduced perivascular and peribronchial inflammation. Simultaneously, a decrease in the concentrations of HA, hydroxyproline, collagen 1, total soluble collagen, and the area of connective tissue in the lungs was observed. The effects of pHD were significantly stronger compared to native HD which can be attributed to the higher stability of pHD. Additional spiperone administration increased the anti-inflammatory and antifibrotic effects of pHD and accelerated the regeneration of the damaged lung. The potentiating effects of spiperone can be explained by the disruption of the dopamine-induced mobilization and migration of fibroblast progenitor cells into the lungs and differentiation of lung mesenchymal stem cells (MSC) into cells of stromal lines. Thus, a combination of pHD and spiperone may represent a promising approach for the treatment of IPF and lung regeneration.


Asunto(s)
Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/farmacología , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/efectos de los fármacos , Espiperona/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/farmacocinética , Hidroxiprolina/metabolismo , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/enzimología , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Queratinas/metabolismo , Pulmón/enzimología , Pulmón/metabolismo , Pulmón/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Poloxámero/química , Receptores de IgG/metabolismo , Espiperona/administración & dosificación , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Exp Lung Res ; 43(9-10): 359-369, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29206498

RESUMEN

AIM: The capability of reducing fibrotic and inflammatory responses in lung tissues represents a gold standard for evaluating the efficacy of therapeutic interventions for treating idiopathic pulmonary fibrosis (IPF). A wide variety of therapeutic strategies have been employed in clinic to treat PF, but limited success has been obtained. Apigenin (4, 5, 7-trihydroxyflavone) is a member of flavonoid family that exerts anti-inflammatory and anti-fibrosis effects. In this study, we explore the potential therapeutic effect of apigenin in lung fibrosis. MATERIALS AND METHODS: Apigenin was employed to treat IPF in a bleomycin-induced PF rat model. Apigenin was loaded onto a biodegradable polymer carrier (nanoparticle, NP) to improve its bio-solubility and bio-availability. The properties (e.g. size, apigenin loading and release profile) of the apigenin loaded polymer carrier were well-characterized. In vitro study was performed to assess the impact of apigenin on pulmonary cell viability, growth, as well as inflammatory and pro-fibrosis responses in pulmonary cells. The impact of apigenin on the production of inflammatory cytokines (e.g. TGF-ß, TNF-α) and pro-fibrosis factors in bronchoalveolar lavage fluid and pulmonary cells from lung tissues was also investigated. RESULTS: Our results showed, apigenin has anti-fibrosis effect by inhibition fibrosis related cytokines expression. And compared with apigenin in soluble form, the strategic release of apigenin is more effective in inhibiting pulmonary fibrosis and inflammation. CONCLUSION: Our finding suggested that apigenin loaded on polymeric carrier might be an effective treatment for pulmonary fibrosis patients.


Asunto(s)
Apigenina/uso terapéutico , Portadores de Fármacos/uso terapéutico , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Apigenina/administración & dosificación , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Portadores de Fármacos/química , Pulmón/metabolismo , Pulmón/patología , Polímeros/uso terapéutico , Ratas
8.
Biol Pharm Bull ; 38(2): 270-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25747986

RESUMEN

In the present in vitro study, we assessed the delivery of pirfenidone incorporated into liposomes modified with truncated basic fibroblast growth factor (tbFGF) to lung fibroblasts and investigated the anti-fibrotic effect of the drug. The tbFGF peptide, KRTGQYKLC, was used to modify the surface of liposomes (tbFGF-liposomes). We used the thin-layer evaporation method, followed by sonication, to prepare tbFGF-liposomes containing pirfenidone. The cellular accumulation of tbFGF-liposomes was 1.7-fold greater than that of non-modified liposomes in WI-38 cells used as a model of lung fibroblasts. Confocal laser scanning microscopy showed that tbFGF-liposomes were widely localized in WI-38 cells. The inhibitory effects of pirfenidone incorporated into tbFGF-liposomes on transforming growth factor-ß1 (TGF-ß1)-induced collagen synthesis in WI-38 cells were evaluated by measuring the level of intracellular hydroxyproline, a major component of the protein collagen. Pirfenidone incorporated into tbFGF-liposomes at concentrations of 10, 30, and 100 µM significantly decreased the TGF-ß1-induced hydroxyproline content in WI-38 cells. The anti-fibrotic effect of pirfenidone incorporated into tbFGF-liposomes was enhanced compared with that of pirfenidone solution. These results indicate that tbFGF-liposomes are a useful drug delivery system of anti-fibrotic drugs to lung fibroblasts for the treatment of idiopathic pulmonary fibrosis.


Asunto(s)
Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/química , Fibroblastos/efectos de los fármacos , Piridonas/administración & dosificación , Línea Celular , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Fibroblastos/metabolismo , Células HEK293 , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Liposomas , Pulmón/citología
9.
J Control Release ; 371: 1-15, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38761856

RESUMEN

Since phospholipids have an important effect on the size, surface potential and hardness of liposomes that decide their in vivo fate after inhalation, this research has systematically evaluated the effect of phospholipids on pulmonary drug delivery by liposomes. In this study, liposomes composed of neutral saturated/unsaturated phospholipids, anionic and cationic phospholipids were constructed to investigate how surface potential and the degree of saturation of fatty acid chains determined their mucus and epithelium permeability both in vitro and in vivo. Our results clearly indicated that liposomes composed of saturated neutral and anionic phospholipids possessed high stability and permeability, compared to that of liposomes composed of unsaturated phospholipids and cationic phospholipids. Furthermore, both in vivo imaging of fluorescence-labeled liposomes and biodistribution of salvianolic acid B (SAB) that encapsulated in liposomes were performed to estimate the effect of phospholipids on the lung exposure and retention of inhaled liposomes. Finally, inhaled SAB-loaded liposomes exhibited enhanced therapeutic effects in a bleomycin-induced idiopathic pulmonary fibrosis mice model via inhibition of inflammation and regulation on coagulation-fibrinolytic system. Such findings will be beneficial to the development of inhalable lipid-based nanodrug delivery systems for the treatment of respiratory diseases where inhalation is the preferred route of administration.


Asunto(s)
Benzofuranos , Fibrosis Pulmonar Idiopática , Liposomas , Ratones Endogámicos C57BL , Fosfolípidos , Animales , Benzofuranos/administración & dosificación , Benzofuranos/farmacocinética , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fosfolípidos/química , Fosfolípidos/administración & dosificación , Administración por Inhalación , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Masculino , Distribución Tisular , Bleomicina/administración & dosificación , Ratones , Humanos , Depsidos
10.
Nat Commun ; 15(1): 6844, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39122711

RESUMEN

Lipid nanoparticle-assisted mRNA inhalation therapy necessitates addressing challenges such as resistance to shear force damage, mucus penetration, cellular internalization, rapid lysosomal escape, and target protein expression. Here, we introduce the innovative "LOOP" platform with a four-step workflow to develop inhaled lipid nanoparticles specifically for pulmonary mRNA delivery. iLNP-HP08LOOP featuring a high helper lipid ratio, acidic dialysis buffer, and excipient-assisted nebulization buffer, demonstrates exceptional stability and enhanced mRNA expression in the lungs. By incorporating mRNA encoding IL-11 single chain fragment variable (scFv), scFv@iLNP-HP08LOOP effectively delivers and secretes IL-11 scFv to the lungs of male mice, significantly inhibiting fibrosis. This formulation surpasses both inhaled and intravenously injected IL-11 scFv in inhibiting fibroblast activation and extracellular matrix deposition. The HP08LOOP system is also compatible with commercially available ALC0315 LNPs. Thus, the "LOOP" method presents a powerful platform for developing inhaled mRNA nanotherapeutics with potential for treating various respiratory diseases, including idiopathic pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Nanopartículas , ARN Mensajero , Anticuerpos de Cadena Única , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/inmunología , Animales , Administración por Inhalación , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Nanopartículas/química , Masculino , Ratones , Anticuerpos de Cadena Única/administración & dosificación , Humanos , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Lípidos/química , Modelos Animales de Enfermedad , Liposomas
11.
Sci Adv ; 10(24): eado4791, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38865465

RESUMEN

The stemness loss-associated dysregeneration of impaired alveolar type 2 epithelial (AT2) cells abolishes the reversible therapy of idiopathic pulmonary fibrosis (IPF). We here report an inhalable mucus-penetrating lipid nanoparticle (LNP) for codelivering dual mRNAs, promoting realveolarization via restoring AT2 stemness for IPF treatment. Inhalable LNPs were first formulated with dipalmitoylphosphatidylcholine and our in-house-made ionizable lipids for high-efficiency pulmonary mucus penetration and codelivery of dual messenger RNAs (mRNAs), encoding cytochrome b5 reductase 3 and bone morphogenetic protein 4, respectively. After being inhaled in a bleomycin model, LNPs reverses the mitochondrial dysfunction through ameliorating nicotinamide adenine dinucleotide biosynthesis, which inhibits the accelerated senescence of AT2 cells. Concurrently, pathological epithelial remodeling and fibroblast activation induced by impaired AT2 cells are terminated, ultimately prompting alveolar regeneration. Our data demonstrated that the mRNA-LNP system exhibited high protein expression in lung epithelial cells, which markedly extricated the alveolar collapse and prolonged the survival of fibrosis mice, providing a clinically viable strategy against IPF.


Asunto(s)
Bleomicina , Moco , Nanopartículas , Animales , Nanopartículas/química , Ratones , Moco/metabolismo , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/metabolismo , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Modelos Animales de Enfermedad , Administración por Inhalación , Lípidos/química , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Humanos , Liposomas
12.
J Control Release ; 370: 421-437, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701884

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with a high mortality rate due to limited treatment options. Current therapies cannot effectively reverse the damage caused by IPF. Research suggests that promoting programmed cell death (apoptosis) in myofibroblasts, the key cells driving fibrosis, could be a promising strategy. However, inducing apoptosis in healthy cells like epithelial and endothelial cells can cause unwanted side effects. This project addresses this challenge by developing a targeted approach to induce apoptosis specifically in myofibroblasts. We designed liposomes (LPS) decorated with peptides that recognize VCAM-1, a protein highly expressed on myofibroblasts in fibrotic lungs. These VCAM1-targeted LPS encapsulate Venetoclax (VNT), a small molecule drug that inhibits BCL-2, an anti-apoptotic protein. By delivering VNT directly to myofibroblasts, we hypothesize that VCAM1-VNT-LPS can selectively induce apoptosis in these cells, leading to reduced fibrosis and improved lung function. We successfully characterized VCAM1-VNT-LPS for size, surface charge, and drug loading efficiency. Additionally, we evaluated their stability over three months at different temperatures. In vitro and in vivo studies using a bleomycin-induced mouse model of lung fibrosis demonstrated the therapeutic potential of VCAM1-VNT-LPS. These studies showed a reduction in fibrosis-associated proteins (collagen, α-SMA, VCAM1) and BCL-2, while simultaneously increasing apoptosis in myofibroblasts. These findings suggest that VCAM1-targeted delivery of BCL-2 inhibitors using liposomes presents a promising and potentially selective therapeutic approach for IPF.


Asunto(s)
Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes , Liposomas , Ratones Endogámicos C57BL , Nanopartículas , Proteínas Proto-Oncogénicas c-bcl-2 , Sulfonamidas , Molécula 1 de Adhesión Celular Vascular , Animales , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Nanopartículas/administración & dosificación , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Molécula 1 de Adhesión Celular Vascular/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Apoptosis/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Humanos , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Masculino , Ratones , Bleomicina/administración & dosificación
13.
Bull Exp Biol Med ; 155(4): 501-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24143378

RESUMEN

Antifibrotic activity of testicular hyaluronidase, immobilized on polyethylenoxide and obtained by electron beam synthesis, was studied on the model of bleomycin injuries to the alveolar epithelium (irreversible pneumofibrosis) in C57Bl/6 mice and compared to the effect of testicular hyaluronidase. Intranasal therapy with immobilized and testicular hyaluronidases prevented the deposition of fibrotic mass in the parenchyma of "bleomycin" lungs. The effect of immobilized hyaluronidase was more pronounced than that of testicular hyaluronidase. The studied compounds were virtually inessential for infiltration of the alveolar interstitium and alveolar tracts by lymphocytes, macrophages, neutrophils, and plasma cells. Unchanged histoarchitectonics of bleomycin-damaged lungs in immobilized hyaluronidase therapy was due to suppression of the progenitor fibroblast cells (CD45(-)).


Asunto(s)
Antiinflamatorios/administración & dosificación , Hialuronoglucosaminidasa/administración & dosificación , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Animales , Antiinflamatorios/química , Bleomicina , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Enzimas Inmovilizadas/administración & dosificación , Enzimas Inmovilizadas/química , Hialuronoglucosaminidasa/química , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/patología , Ratones , Ratones Endogámicos C57BL , Polietilenos/química , Alveolos Pulmonares/patología , Mucosa Respiratoria/patología
14.
J Colloid Interface Sci ; 636: 388-400, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-36640550

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is the most common form of idiopathic interstitial pneumonia, where M2 macrophages play an irreplaceable role in the anti-inflammatory progress. Targeting M2 macrophages and regulating their polarization may be a potential treatment strategy for IPF. Herein, we designed a magnetic liposome based dual-targeting delivery system for the IPF treatment, constructed by mannose-modified magnetic nanoparticles (MAN-MNPs) loaded on the surface of the liposome (MAN-MNPs@LP). The delivery system is capable of responding to a static magnetic field (SMF) and then recognizing in situ of M2 macrophages through the mannose receptor-dependent internalization. Firstly, a series of physical and chemical assays were used to characterize these nanoparticles. Subsequently, magnetic liposomes accumulation in the damaged lung with/without mannose modification and SMF were compared by in vivo imaging system. Finally, the reduction of M2 macrophages and inhibition of their polarization confirmed that the development of IPF was retarded due to the in situ release of encapsulated dexamethasone (Dex) in lungs under the SMF. Further investigation demonstrated that the expression of α-SMA and collagen deposition was reduced. Altogether, this dual-targeting delivery system can effectively deliver Dex into M2 macrophages in the lung, making it a novel and promising therapeutic system for the IPF treatment.


Asunto(s)
Fibrosis Pulmonar Idiopática , Liposomas , Humanos , Liposomas/metabolismo , Manosa/metabolismo , Manosa/farmacología , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Fenómenos Magnéticos
15.
Biomaterials ; 303: 122404, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37992600

RESUMEN

Idiopathic pulmonary fibrosis (IPF) stands as a highly heterogeneous and deadly lung disease, yet the available treatment options remain limited. Combining myofibroblast inhibition with ROS modulation in damaged AECs offers a comprehensive strategy to halt IPF progression, but delivering drugs separately to these cell types is challenging. Inspired by the successful application of pulmonary surfactant (PS) replacement therapy in lung disease treatment, we have developed PS nano-biomimetic liposomes (PSBs) to utilize its natural transport pathway for targeting AECs while reducing lung tissue clearance. In this collaborative pulmonary drug delivery system, PSBs composed of DPPC/POPG/DPPG/CHO (20:9:5:4) were formulated for inhalation. These PSBs loaded with ROS-scavenger astaxanthin (AST) and anti-fibrosis drug pirfenidone (PFD) were aerosolized for precise quantification and mimicking patient inhalation. Through aerosol inhalation, the lipid membrane of PSBs gradually fused with natural PS, enabling AST delivery to AECs by hitchhiking with PS circulation. Simultaneously, PFD was released within the PS barrier, effectively penetrating lung tissue to exert therapeutic effects. In vivo results have shown that PSBs offer numerous therapeutic advantages in mice with IPF, particularly in terms of lung function recovery. This approach addresses the challenges of drug delivery to specific lung cells and offers potential benefits for IPF patients.


Asunto(s)
Fibrosis Pulmonar Idiopática , Surfactantes Pulmonares , Humanos , Ratones , Animales , Surfactantes Pulmonares/uso terapéutico , Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/farmacología , Liposomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Biomimética , Aerosoles y Gotitas Respiratorias , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Piridonas/farmacología
16.
Lung ; 190(5): 523-7, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22810758

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis is a progressive interstitial lung disease with no current effective therapies. Treatment has focused on antifibrotic agents to stop proliferation of fibroblasts and collagen deposition in the lung. We present the first clinical trial data on the use of losartan, an antifibrotic agent, to treat idiopathic pulmonary fibrosis. The primary objective was to evaluate the effect of losartan on progression of idiopathic pulmonary fibrosis measured by the change in percentage of predicted forced vital capacity (%FVC) after 12 months. Secondary outcomes included the change in forced expiratory volume at 1 second, diffusing capacity of carbon monoxide, 6-minute walk test distance, and baseline/transition dyspnea index. METHODS: Patients with idiopathic pulmonary fibrosis and a baseline %FVC of ≥50 % were treated with losartan 50 mg by mouth daily for 12 months. Pulmonary function testing, 6-minute walk, and breathlessness indices were measured every 3 months. RESULTS: Twenty participants with idiopathic pulmonary fibrosis were enrolled and 17 patients were evaluable for response. Twelve patients had a stable or improved %FVC at study month 12. Similar findings were observed in secondary end-point measures, including 58, 71, and 65 % of patients with stable or improved forced expiratory volume at 1 second, diffusing capacity for carbon monoxide, and 6-minute walk test distance, respectively. No treatment-related adverse events that resulted in early study discontinuation were reported. CONCLUSION: Losartan stabilized lung function in patients with idiopathic pulmonary fibrosis over 12 months. Losartan is a promising agent for the treatment of idiopathic pulmonary fibrosis and has a low toxicity profile.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Losartán/uso terapéutico , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Disnea/tratamiento farmacológico , Prueba de Esfuerzo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Pruebas de Función Respiratoria , Resultado del Tratamiento
17.
J Control Release ; 351: 623-637, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36191673

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fibrotic interstitial lung disease in which collagen progressively deposits in the supporting framework of the lungs. The pathological collagen creates a recalcitrant barrier in mesenchyme for drug penetration, thus greatly restricting the therapeutical efficacy. On the other hand, this overloaded collagen is gradually exposed to the bloodstream at fibrotic sites because of the vascular hyperpermeability, thus serving as a potential target. Herein, pathological collagen targeting and penetrating liposomes (DP-CC) were constructed to deliver anti-fibrotic dual drugs including pirfenidone (PFD) and dexamethasone (DEX) deep into injured alveoli. The liposomes were co-decorated with collagen binding peptide (CBP) and collagenase (COL). CBP could help vehicle recognize the pathological collagen and target the fibrotic lungs efficiently because of its high affinity to collagen, and COL assisted in breaking through the collagen barrier and delivering vehicle to the center of injured sites. Then, the released dual drugs developed a synergistic anti-fibrotic effect to repair the damaged epithelium and remodel the extracellular matrix (ECM), thus rebuilding the lung architecture. This study provides a promising strategy to deliver drugs deep into pathological collagen accumulated sites for the enhanced treatment of IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Liposomas/metabolismo , Colágeno/metabolismo , Pulmón/metabolismo , Matriz Extracelular/metabolismo , Fibrosis , Fibroblastos/metabolismo
18.
Curr Drug Deliv ; 18(7): 947-954, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33372874

RESUMEN

BACKGROUND: Idiopathic Pulmonary Fibrosis (IPF) is a chronic and progressive respiratory disease characterized by the destruction of the alveolar structure. In pulmonary fibrosis, aerosolized drugs are easily transferred to the systemic circulation via leakage through the injured alveolar epithelium. Therefore, pulmonary drug delivery systems for sustained distribution in fibrotic lungs are needed. OBJECTIVE: We evaluated the intrapulmonary pharmacokinetics of aerosolized liposomes as pulmonary drug delivery systems in mice with bleomycin-induced pulmonary fibrosis. METHODS: The aerosolized liposomal formulations and solutions of model compounds, including indocyanine green and 6-carboxyfluorescein (6-CF), were intrapulmonary administered to mice with bleomycin-induced pulmonary fibrosis. In vivo imaging for indocyanine green and 6-CF measurements in lung tissues and plasma were performed. Additionally, in vitro permeation experiments using NCI-H441 cell monolayers as a model of alveolar epithelial cells were performed. RESULTS: The fluorescence signals of indocyanine green following the administration of liposomal formulations were observed longer in the lungs than those in solution-treated mice. Compared with the solution, the 6-CF concentrations in lung tissues after the administration of liposomal formulations were determined higher, whereas those in the plasma were lower. 6-CF permeability was significantly increased by transforming growth factor-ß1 in NCI-H441 cell monolayers treated with the solution but unchanged in the presence of the liposomal formulation. CONCLUSION: The aerosolized liposomal formulation can prevent enhanced drug transfer from fibrotic lungs into the systemic circulation via the injured alveolar epithelium. This system may be useful for the sustained distribution of anti-fibrotic agents in fibrotic lungs and the optimization of IPF therapy.


Asunto(s)
Fibrosis Pulmonar Idiopática , Liposomas , Animales , Antifibróticos , Bleomicina , Modelos Animales de Enfermedad , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Pulmón , Ratones , Ratones Endogámicos C57BL
19.
Drug Deliv ; 28(1): 1419-1431, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34223777

RESUMEN

Glucocorticoid (GC) hormone has been commonly used to treat systemic inflammation and immune disorders. However, the side effects associated with long-term use of high-dose GC hormone limit its clinical application seriously. GC hormone that can specifically target the lung might decrease the effective dosage and thus reduce GC-associated side effects. In this study, we successfully prepared human lung-targeting liposomal methylprednisolone crosslinked with nanobody (MPS-NSSLs-SPANb). Our findings indicate that MPS-NSSLs-SPANb may reduce the effective therapeutic dosage of MPS, achieve better efficacy, and reduce GC-associated side effects. In addition, MPS-NSSLs-SPANb showed higher efficacy and lower toxicity than conventional MPS.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Metilprednisolona/administración & dosificación , Metilprednisolona/farmacología , Proteína A Asociada a Surfactante Pulmonar/administración & dosificación , Proteína A Asociada a Surfactante Pulmonar/farmacología , Animales , Química Farmacéutica , Portadores de Fármacos/química , Ensayo de Inmunoadsorción Enzimática , Humanos , Liposomas/química , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Anticuerpos de Dominio Único/administración & dosificación , Anticuerpos de Dominio Único/farmacología
20.
Front Immunol ; 12: 613907, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679754

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is serious chronic lung disease with limited therapeutic approaches. Inflammation and immune disorders are considered as the main factors in the initiation and development of pulmonary fibrosis. Inspired by the key roles of macrophages during the processes of inflammation and immune disorders, here, we report a new method for direct drug delivery into the in-situ fibrotic tissue sites in vitro and in vivo. First, liposomes containing dexamethasone (Dex-L) are prepared and designed to entry into the macrophages in the early hours, forming the macrophages loaded Dex-L delivery system (Dex-L-MV). Chemokine and cytokine factors such as IL-6, IL-10, Arg-1 are measured to show the effect of Dex-L to the various subtypes of macrophages. Next, we mimic the inflammatory and anti-inflammatory microenvironment by co-culture of polarized/inactive macrophage and fibroblast cells to show the acute inflammation response of Dex-L-MV. Further, we confirm the targeted delivery of Dex-L-MV into the inflammatory sites in vivo, and surprisingly found that injected macrophage containing Dex can reduce the level of macrophage infiltration and expression of the markers of collagen deposition during the fibrotic stage, while causing little systematic toxicity. These data demonstrated the suitability and immune regulation effect of Dex-L-MV for the anti-pulmonary process. It is envisaged that these findings are a step forward toward endogenous immune targeting systems as a tool for clinical drug delivery.


Asunto(s)
Microambiente Celular/efectos de los fármacos , Microambiente Celular/inmunología , Dexametasona/administración & dosificación , Inmunosupresores/administración & dosificación , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Animales , Biomarcadores , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Liposomas , Activación de Macrófagos/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones , Células 3T3 NIH , Células RAW 264.7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA