Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Dev Res ; 85(4): e22217, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38845214

RESUMEN

As a hybrid weapon, two novel series of pyrazoles, 16a-f and 17a-f, targeting both COX-2 and ACE-1-N-domain, were created and their anti-inflammatory, anti-hypertensive, and anti-fibrotic properties were evaluated. In vitro, 17b and 17f showed COX-2 selectivity (SI = 534.22 and 491.90, respectively) compared to celecoxib (SI = 326.66) and NF-κB (IC50 1.87 and 2.03 µM, respectively). 17b (IC50 0.078 µM) and 17 f (IC50 0.094 µM) inhibited ACE-1 comparable to perindopril (PER) (IC50 0.048 µM). In vivo, 17b decreased systolic blood pressure by 18.6%, 17b and 17f increased serum NO levels by 345.8%, and 183.2%, respectively, increased eNOS expression by 0.97 and 0.52 folds, respectively and reduced NF-κB-p65 and P38-MAPK expression by -0.62, -0.22, -0.53, and -0.24 folds, respectively compared to  l-NAME (-0.34, -0.45 folds decline in NF-κB-p65 and P38-MAPK, respectively). 17b reduced ANG-II expression which significantly reversed the cardiac histological changes induced by L-NAME.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina , Antiinflamatorios , Antihipertensivos , Inhibidores de la Ciclooxigenasa 2 , Pirazoles , Tetrazoles , Pirazoles/farmacología , Pirazoles/química , Animales , Antihipertensivos/farmacología , Antihipertensivos/química , Antihipertensivos/síntesis química , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/química , Inhibidores de la Enzima Convertidora de Angiotensina/síntesis química , Tetrazoles/farmacología , Tetrazoles/química , Antiinflamatorios/farmacología , Antiinflamatorios/química , Antiinflamatorios/síntesis química , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa 2/síntesis química , Inhibidores de la Ciclooxigenasa 2/química , Ratas , Diseño de Fármacos , Masculino , Antifibróticos/farmacología , Antifibróticos/química , Ciclooxigenasa 2/metabolismo , Presión Sanguínea/efectos de los fármacos , Humanos , Peptidil-Dipeptidasa A/metabolismo
2.
J Med Chem ; 67(13): 11389-11400, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38938102

RESUMEN

Hepatic stellate cells (HSCs) activation is a key event in the development of liver fibrosis, and blockage of the activation of HSCs has been shown to alleviate liver fibrosis. Sophoridine, a bioactive alkaloid found in many Chinese herbs, exhibits a broad spectrum of pharmacological effects, but its activities are not strong. In this study, a series of structurally modified derivatives of sophoridine were designed and synthesized. Among them, sophoridine α-aryl propionamide derivative ZM600 displayed a significant inhibitory effect on the activation of HSCs. The in vivo experiment demonstrated that ZM600 markedly ameliorated carbon tetrachloride (CCl4) and bile duct ligation (BDL)-induced liver fibrosis with a significant improvement of extracellular matrix deposition. Mechanism investigations revealed that ZM600 specifically inhibited the activation of NF-κB, PI-3K/AKT, and TGF-ß/Smads signaling pathways. These results suggest that ZM600 has a protective effect on liver fibrosis, which provides a new candidate for the treatment of liver fibrosis.


Asunto(s)
Alcaloides , Células Estrelladas Hepáticas , Cirrosis Hepática , Matrinas , Quinolizinas , Animales , Quinolizinas/farmacología , Quinolizinas/síntesis química , Quinolizinas/química , Quinolizinas/uso terapéutico , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Alcaloides/farmacología , Alcaloides/química , Alcaloides/síntesis química , Alcaloides/uso terapéutico , Masculino , FN-kappa B/metabolismo , FN-kappa B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tetracloruro de Carbono , Ratones , Relación Estructura-Actividad , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Descubrimiento de Drogas , Antifibróticos/farmacología , Antifibróticos/uso terapéutico , Antifibróticos/química , Antifibróticos/síntesis química , Ratas Sprague-Dawley
3.
Int J Pharm ; 656: 124096, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38583821

RESUMEN

Pulmonary fibrosis (PF) is a chronic, progressive and irreversible interstitial lung disease that seriously threatens human life and health. Our previous study demonstrated the unique superiority of traditional Chinese medicine cryptotanshinone (CTS) combined with sustained pulmonary drug delivery for treating PF. In this study, we aimed to enhance the selectivity, targeting efficiency and sustained-release capability based on this delivery system. To this end, we developed and evaluated CTS-loaded modified liposomes-chitosan (CS) microspheres SM(CT-lipo) and liposome-exosome hybrid bionic vesicles-CS microspheres SM(LE). The prepared nano-in-micro particles system integrates the advantages of the carriers and complements each other. SM(CT-lipo) and SM(LE) achieved lung myofibroblast-specific targeting through CREKA peptide binding specifically to fibronectin (FN) and the homing effect of exosomes on parent cells, respectively, facilitating efficient delivery of anti-fibrosis drugs to lung lesions. Furthermore, compared with daily administration of conventional microspheres SM(NC) and positive control drug pirfenidone (PFD), inhaled administration of SM(CT-lipo) and SM(LE) every two days still attained similar efficacy, exhibiting excellent sustained drug release ability. In summary, our findings suggest that the developed SM(CT-lipo) and SM(LE) delivery strategies could achieve more accurate, efficient and safe therapy, providing novel insights into the treatment of chronic PF.


Asunto(s)
Quitosano , Exosomas , Fibronectinas , Liposomas , Fibrosis Pulmonar , Animales , Humanos , Masculino , Administración por Inhalación , Antifibróticos/administración & dosificación , Antifibróticos/química , Quitosano/química , Quitosano/administración & dosificación , Preparaciones de Acción Retardada , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Exosomas/química , Fibronectinas/administración & dosificación , Liposomas/química , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Microesferas , Fenantrenos/administración & dosificación , Fenantrenos/química , Fenantrenos/farmacocinética , Fibrosis Pulmonar/tratamiento farmacológico , Piridonas , Ratas Sprague-Dawley , Ratas
4.
Eur J Med Chem ; 245(Pt 1): 114886, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36347091

RESUMEN

Sixty-one palmatine (PMT) derivatives, of which twenty-eight were new, were synthesized and evaluated for their anti-fibrogenic activities via collagen type I α 1 (COL1A1)-promoter based luciferase model in LX-2 cells, taking 2,3,10-trimethoxy-9-p-isopropyloxyprotopalmatine bromide (1) as the lead. Among them, compound 3a exerted the highest potency with the IC50 value of 8.19 µmol/L and SI value of 8.59, and reduced the expressions of multiple fibrogenic biomarkers, including COL1A1, TGF-ß1, α-SMA and TIMP1 in a dose-dependent manner. In addition, it significantly reduced liver steatosis and inflammation, and especially attenuated the degree of liver fibrosis in choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD)-induced NASH mice model in vivo. Mechanism study indicated that it significantly ameliorated liver injury by activating farnesoid X receptor (FXR). BDL-induced fibrosis rats model further verified its liver-protective and anti-fibrosis activities. Therefore, PMT derivatives constituted a new family of non-steroidal FXR agonists as anti-NASH candidates, with the advantage of good safety profile, and are worthy for further investigation.


Asunto(s)
Antifibróticos , Alcaloides de Berberina , Hígado , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Ratas , Alcaloides de Berberina/química , Alcaloides de Berberina/farmacología , Alcaloides de Berberina/uso terapéutico , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Antifibróticos/química , Antifibróticos/farmacología , Antifibróticos/uso terapéutico
5.
J Med Chem ; 65(1): 163-190, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34939411

RESUMEN

DCN1, a co-E3 ligase, interacts with UBC12 and activates cullin-RING ligases (CRLs) by catalyzing cullin neddylation. Although DCN1 has been recognized as an important therapeutic target for human diseases, its role in the cardiovascular area remains unknown. Here, we first found that DCN1 was upregulated in isolated cardiac fibroblasts (CFs) treated by angiotensin (Ang) II and in mouse hearts after pressure overload. Then, structure-based optimizations for DCN1-UBC12 inhibitors were performed based on our previous work, yielding compound DN-2. DN-2 specifically targeted DCN1 at molecular and cellular levels as shown by molecular modeling studies, HTRF, cellular thermal shift and co-immunoprecipitation assays. Importantly, DN-2 effectively reversed Ang II-induced cardiac fibroblast activation, which was associated with the inhibition of cullin 3 neddylation. Our findings indicate a potentially unrecognized role of DCN1 inhibition for anticardiac fibrotic effects. DN-2 may be used as a lead compound for further development.


Asunto(s)
Antifibróticos , Descubrimiento de Drogas , Inhibidores Enzimáticos , Fibrosis , Cardiopatías , Péptidos y Proteínas de Señalización Intracelular , Pirimidinas , Enzimas Ubiquitina-Conjugadoras , Animales , Masculino , Ratones , Ratas , Antifibróticos/química , Antifibróticos/farmacología , Proteínas Cullin/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Fibrosis/patología , Cardiopatías/tratamiento farmacológico , Cardiopatías/metabolismo , Cardiopatías/patología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Ratones Endogámicos C57BL , Proteína NEDD8/metabolismo , Pirimidinas/química , Ratas Sprague-Dawley , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Ubiquitinas
6.
Bioorg Chem ; 117: 105409, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34749117

RESUMEN

Phosphodiesterase (PDE) inhibitors are currently an extensively studied group of compounds that can bring many benefits in the treatment of various inflammatory and fibrotic diseases, including asthma. Herein, we describe a series of novel N'-phenyl- or N'-benzylbutanamide and N'-arylidenebutanehydrazide derivatives of 8-aminopurine-2,6-dione (27-43) and characterized them as prominent pan-PDE inhibitors. Most of the compounds exhibited antioxidant and anti-inflammatory activity in lipopolysaccharide (LPS)-induced murine macrophages RAW264.7. The most active compounds (32-35 and 38) were evaluated in human bronchial epithelial cells (HBECs) derived from asthmatics. To better map the bronchial microenvironment in asthma, HBECs after exposure to selected 8-aminopurine-2,6-dione derivatives were incubated in the presence of two proinflammatory and/or profibrotic factors: transforming growth factor type ß (TGF-ß) and interleukin 13 (IL-13). Compounds 32-35 and 38 significantly reduced both IL-13- and TGF-ß-induced expression of proinflammatory and profibrotic mediators, respectively. Detailed analysis of their inhibition preferences for selected PDEs showed high affinity for isoenzymes important in the pathogenesis of asthma, including PDE1, PDE3, PDE4, PDE7, and PDE8. The presented data confirm that structural modifications within the 7 and 8 positions of the purine-2,6-dione core result in obtaining preferable pan-PDE inhibitors which in turn exert an excellent anti-inflammatory and anti-fibrotic effect in the bronchial epithelial cells derived from asthmatic patients. This dual-acting pan-PDE inhibitors constitute interesting and promising lead structures for further anti-asthmatic agent discovery.


Asunto(s)
Antiasmáticos/farmacología , Antiinflamatorios/farmacología , Antifibróticos/farmacología , Antioxidantes/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Animales , Antiasmáticos/síntesis química , Antiasmáticos/química , Antiinflamatorios/síntesis química , Antiinflamatorios/química , Antifibróticos/síntesis química , Antifibróticos/química , Antioxidantes/síntesis química , Antioxidantes/química , Humanos , Ratones , Inhibidores de Fosfodiesterasa/síntesis química , Inhibidores de Fosfodiesterasa/química , Células RAW 264.7
7.
Int J Mol Sci ; 22(21)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34769206

RESUMEN

Exposure to metallic nanoparticles (NPs) can result in inadvertent NP accumulation in body tissues. While their subsequent cellular interactions can lead to unintended consequences and are generally regarded as detrimental for health, they can on occasion mediate biologically beneficial effects. Among NPs, cerium oxide nanoparticles (CeO2 NP) possess strong antioxidant properties and have shown to alleviate certain pathological conditions. Herein, we show that the presence of cubic 25 nm CeO2 NP was able to reduce TGF-ß-mediated activation in the cultured hepatic stellate cell line LX2 by reducing oxidative stress levels and TGF-ß-mediated signalling. These cells displayed reduced classical liver fibrosis phenotypes, such as diminished fibrogenesis, altered matrix degradation, decreased cell motility, modified contractability and potentially lowered autophagy. These findings demonstrate that CeO2 NP may be able to ameliorate hepatic fibrosis and suggest a possible therapeutic pathway for an otherwise difficult-to-treat condition.


Asunto(s)
Antifibróticos/farmacología , Antioxidantes/farmacología , Cerio/farmacología , Cirrosis Hepática/tratamiento farmacológico , Antifibróticos/química , Antioxidantes/química , Línea Celular , Cerio/química , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Nanopartículas/química , Estrés Oxidativo/efectos de los fármacos
8.
Eur J Pharmacol ; 913: 174618, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34762934

RESUMEN

Fibrosis is the formation of scar tissue due to injury or long-term inflammation and is a leading cause of morbidity and mortality. Activation of the pro-fibrotic cytokine transforming growth factor-ß (TGFß) via the alpha-V beta-6 (αvß6) integrin has been identified as playing a key role in the development of fibrosis. Therefore, a drug discovery programme to identify an orally bioavailable small molecule αvß6 arginyl-glycinyl-aspartic acid (RGD)-mimetic was initiated. As part of a medicinal chemistry programme GSK3335103 was identified and profiled in a range of pre-clinical in vitro and in vivo systems. GSK3335103 was shown to bind to the αvß6 with high affinity and demonstrated fast binding kinetics. In primary human lung epithelial cells, GSK3335103-induced concentration- and time-dependent internalisation of αvß6 with a rapid return of integrin to the cell surface observed after washout. Following sustained engagement of the αvß6 integrin in vitro, lysosomal degradation was induced by GSK3335103. GSK3335103 was shown to engage with the αvß6 integrin and inhibit the activation of TGFß in both ex vivo IPF tissue and in a murine model of bleomycin-induced lung fibrosis, as measured by αvß6 engagement, TGFß signalling and collagen deposition, with a prolonged duration of action observed in vivo. In summary, GSK3335103 is a potent αvß6 inhibitor that attenuates TGFß signalling in vitro and in vivo with a well-defined pharmacokinetic/pharmacodynamic relationship. This translates to a significant reduction of collagen deposition in vivo and therefore GSK3335103 represents a potential novel oral therapy for fibrotic disorders.


Asunto(s)
Antifibróticos/farmacología , Integrinas/antagonistas & inhibidores , Fibrosis Pulmonar/tratamiento farmacológico , Administración Oral , Animales , Antifibróticos/química , Antifibróticos/uso terapéutico , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Disponibilidad Biológica , Bleomicina/administración & dosificación , Bleomicina/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Integrinas/química , Integrinas/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Lisosomas/metabolismo , Masculino , Ratones , Oligopéptidos/química , Cultivo Primario de Células , Proteolisis/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Factor de Crecimiento Transformador beta/metabolismo
9.
Int Immunopharmacol ; 101(Pt A): 108166, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34628270

RESUMEN

Our previous studies demonstrated that Curc-mPEG454, a curcumin derivative modified with short-chain polyethylene glycol (PEG), not only increased the blood concentration of curcumin, but also retained its anti-inflammatory activity. Here, we aimed to evaluate the anti-fibrotic effect of Curc-mPEG454 on a rat liver fibrosis model induced by carbon tetrachloride (CCl4), and to explore the underlying mechanisms by integrating our total liver RNA sequencing (RNA-seq) data with recent liver single-cell sequencing (scRNA-seq) studies. 50 mg/kg and 100 mg/kg Curc-mPEG454 treatment significantly reduced the elevation of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) induced by CCl4, and the incidence of liver cirrhosis decreased from 75% to 37% and 35%, respectively. RNA-seq analysis revealed that Curc-mPEG454 significantly upregulated aldehyde oxidase 1 (AOX1) while downregulated cytochrome p450 26A1 (CYP26A1) and cytochrome p450 26B1 (CYP26B1) resulting in restoring liver retinoic acid (RA) level, increased glutamate-cysteine ligase catalytic subunit (GCLC) and glutamate-cysteine ligase modifier subunit (GCLM) expression to synthesize hepatic glutathione (GSH), and inhibited liver inflammation via down-regulating the Prostaglandin E Synthase 2 (PTGES2)/prostacyclin E2 (PGE2) signaling. Integrating scRNA-seq data revealed that Curc-mPEG454 effectively inhibited the expansion of scar-associated macrophage subpopulation and scar-producing myofibroblasts in the damaged liver, and remodeled the fibrotic niche via regulation of ligand-receptor interactions including platelet-derived growth factor-B (PDGF-B)/platelet-derived growth factor receptor-α (PDGFR-α) signaling. As a multi-target prodrug, PEGylated curcumin deserves further attention and research.


Asunto(s)
Antifibróticos/farmacología , Curcumina/farmacología , Cirrosis Hepática/tratamiento farmacológico , Hígado/efectos de los fármacos , Profármacos/farmacología , Animales , Antifibróticos/química , Antifibróticos/uso terapéutico , Tetracloruro de Carbono/administración & dosificación , Tetracloruro de Carbono/toxicidad , Curcumina/química , Curcumina/uso terapéutico , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inyecciones Intravenosas , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Polietilenglicoles/química , Profármacos/química , Profármacos/uso terapéutico , RNA-Seq , Ratas , Análisis de la Célula Individual
10.
J Pharmacol Exp Ther ; 379(3): 290-300, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34593558

RESUMEN

Metabolic reprogramming of the myofibroblast plays a fundamental role in the pathogenesis of fibrosing interstitial lung diseases. Here, we characterized the in vitro and in vivo metabolic and antifibrotic effects of IM156, an oxidative phosphorylation (OXPHOS) modulator that acts by inhibiting protein complex 1. In vitro, IM156 inhibited transforming growth factor ß (TGFß)-dependent increases in mitochondrial oxygen consumption rate and expression of myofibroblast markers in human pulmonary fibroblasts without altering cell viability or adding to TGFß-induced increases in the extracellular acidification rate. IM156 significantly increased cellular AMP-activated protein kinase (AMPK) phosphorylation and was 60-fold more potent than metformin. In vivo, chronic oral administration of IM156 was highly distributed to major peripheral organs (i.e., lung, liver, kidney, heart) and had significant dose-related effects on the plasma metabolome consistent with OXPHOS modulation and AMPK activation. IM156 increased glycolysis, lipolysis, ß-oxidation, and amino acids and decreased free fatty acids, tricarboxylic acid cycle activity, and protein synthesis. In the murine bleomycin model of pulmonary fibrosis, daily oral administration of IM156, administered 7 days after lung injury, attenuated body/lung weight changes and reduced lung fibrosis and inflammatory cell infiltration. The plasma exposures of IM156 were comparable to well tolerated doses in human studies. In conclusion, the metabolic and antifibrotic effects of IM156 suggest that OXPHOS modulation can attenuate myofibroblast metabolic reprogramming and support testing IM156 as a therapy for idiopathic pulmonary fibrosis and other fibrotic diseases. SIGNIFICANCE STATEMENT: Fibrosing interstitial lung diseases have a poor prognosis, and current antifibrotic treatments have significant limitations. This study demonstrates that attenuation of fibrogenic metabolic remodeling, by modulation of oxidative phosphorylation with IM156, prevents myofibroblast phenotype/collagen deposition and is a potentially effective and translational antifibrotic strategy.


Asunto(s)
Antifibróticos/farmacología , Reprogramación Celular/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Fibrosis Pulmonar/metabolismo , Animales , Antifibróticos/química , Antifibróticos/uso terapéutico , Línea Celular , Reprogramación Celular/fisiología , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/prevención & control
11.
Bioorg Med Chem ; 46: 116362, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34428714

RESUMEN

A series of imidazo[1,2-a]pyridine compounds bearing urea moiety (8-27) were designed, synthesized and evaluated for their ATX inhibitory activities in vitro by FS-3 based enzymatic assay. Delightfully, benzylamine derivatives (14-27) exhibited higher ATX inhibitory potency with IC50 value ranging from 1.72 to 497 nM superior to benzamide analogues (8-13). Remarkably, benzylamine derivative 20 bearing 4-hydroxypiperidine exerted an amazing inhibitory activity (IC50 = 1.72 nM) which exceeded the positive control GLPG1690 (IC50 = 2.90 nM). Simultaneously, the binding model of 20 with ATX was established which rationalized the well performance of 20 in enzymatic assay. Accordingly, further in vivo studies were carried out to evaluate direct anti-fibrotic effects of 20 through Masson staining. Notably, 20 effectively alleviated lung structural damage with fewer fibrotic lesions at an oral dose of 60 mg/kg, qualifying 20 as a promising ATX inhibitor for IPF treatment.


Asunto(s)
Antifibróticos/farmacología , Diseño de Fármacos , Fibrosis/tratamiento farmacológico , Hidrolasas Diéster Fosfóricas/metabolismo , Piridinas/farmacología , Urea/farmacología , Animales , Antifibróticos/síntesis química , Antifibróticos/química , Relación Dosis-Respuesta a Droga , Fibrosis/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Relación Estructura-Actividad , Urea/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...