Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 173
Filtrar
1.
Circ Res ; 126(8): e37-e52, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32089086

RESUMEN

RATIONALE: Cholesterol crystal embolism can be a life-threatening complication of advanced atherosclerosis. Pathophysiology and molecular targets for treatment are largely unknown. OBJECTIVE: We aimed to develop a new animal model of cholesterol crystal embolism to dissect the molecular mechanisms of cholesterol crystal (CC)-driven arterial occlusion, tissue infarction, and organ failure. METHODS AND RESULTS: C57BL/6J mice were injected with CC into the left kidney artery. Primary end point was glomerular filtration rate (GFR). CC caused crystal clots occluding intrarenal arteries and a dose-dependent drop in GFR, followed by GFR recovery within 4 weeks, that is, acute kidney disease. In contrast, the extent of kidney infarction was more variable. Blocking necroptosis using mixed lineage kinase domain-like deficient mice or necrostatin-1s treatment protected from kidney infarction but not from GFR loss because arterial obstructions persisted, identifying crystal clots as a primary target to prevent organ failure. CC involved platelets, neutrophils, fibrin, and extracellular DNA. Neutrophil depletion or inhibition of the release of neutrophil extracellular traps had little effects, but platelet P2Y12 receptor antagonism with clopidogrel, fibrinolysis with urokinase, or DNA digestion with recombinant DNase I all prevented arterial occlusions, GFR loss, and kidney infarction. The window-of-opportunity was <3 hours after CC injection. However, combining Nec-1s (necrostatin-1s) prophylaxis given 1 hour before and DNase I 3 hours after CC injection completely prevented kidney failure and infarcts. In vitro, CC did not directly induce plasmatic coagulation but induced neutrophil extracellular trap formation and DNA release mainly from kidney endothelial cells, neutrophils, and few from platelets. CC induced ATP release from aggregating platelets, which increased fibrin formation in a DNase-dependent manner. CONCLUSIONS: CC embolism causes arterial obstructions and organ failure via the formation of crystal clots with fibrin, platelets, and extracellular DNA as critical components. Therefore, our model enables to unravel the pathogenesis of the CC embolism syndrome as a basis for both prophylaxis and targeted therapy.


Asunto(s)
Colesterol/toxicidad , Embolia por Colesterol/patología , Riñón/irrigación sanguínea , Riñón/patología , Insuficiencia Renal/patología , Animales , Embolia por Colesterol/inducido químicamente , Células Endoteliales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Renal/inducido químicamente
2.
Arterioscler Thromb Vasc Biol ; 41(1): 302-316, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33028096

RESUMEN

OBJECTIVE: Vascular smooth muscle cells (SMCs) dedifferentiate and initiate expression of macrophage markers with cholesterol exposure. This phenotypic switching is dependent on the transcription factor Klf4 (Krüppel-like factor 4). We investigated the molecular pathway by which cholesterol induces SMC phenotypic switching. Approach and Results: With exposure to free cholesterol, SMCs decrease expression of contractile markers, activate Klf4, and upregulate a subset of macrophage and fibroblast markers characteristic of modulated SMCs that appear with atherosclerotic plaque formation. These phenotypic changes are associated with activation of all 3 pathways of the endoplasmic reticulum unfolded protein response (UPR), Perk (protein kinase RNA-like endoplasmic reticulum kinase), Ire (inositol-requiring enzyme) 1α, and Atf (activating transcription factor) 6. Blocking the movement of cholesterol from the plasma membrane to the endoplasmic reticulum prevents free cholesterol-induced UPR, Klf4 activation, and upregulation of the majority of macrophage and fibroblast markers. Cholesterol-induced phenotypic switching is also prevented by global UPR inhibition or specific inhibition of Perk signaling. Exposure to chemical UPR inducers, tunicamycin and thapsigargin, is sufficient to induce these same phenotypic transitions. Finally, analysis of published single-cell RNA sequencing data during atherosclerotic plaque formation in hyperlipidemic mice provides preliminary in vivo evidence of a role of UPR activation in modulated SMCs. CONCLUSIONS: Our data demonstrate that UPR is necessary and sufficient to drive phenotypic switching of SMCs to cells that resemble modulated SMCs found in atherosclerotic plaques. Preventing a UPR in hyperlipidemic mice diminishes atherosclerotic burden, and our data suggest that preventing SMC transition to dedifferentiated cells expressing macrophage and fibroblast markers contributes to this decreased plaque burden.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Colesterol/toxicidad , Fibroblastos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Factor de Transcripción Activador 4/metabolismo , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Línea Celular , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Placa Aterosclerótica , eIF-2 Quinasa/metabolismo
3.
Angew Chem Int Ed Engl ; 60(10): 5193-5198, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33252821

RESUMEN

All-nitrogenated sugars (ANSs), in which all hydroxy groups in a carbohydrate are replaced with amino groups, are anticipated to be privileged structures with useful biological activities. However, ANS synthesis has been challenging due to the difficulty in the installation of multi-amino groups. We report herein the development of a concise synthetic route to peracetylated ANSs in seven steps from commercially available monosaccharides. The key to success is the use of the sequential Overman rearrangement, which enables formal simultaneous substitution of four or five hydroxy groups in monosaccharides with amino groups. A variety of ANSs are available through the same reaction sequence starting from different initial monosaccharides by chirality transfer of secondary alcohols. Transformations of the resulting peracetylated ANSs such as glycosylation and deacetylation are also demonstrated. Biological studies reveal that ANS-modified cholesterol show cytotoxicity against human cancer cell lines, whereas each ANS and cholesterol have no cytotoxicity.


Asunto(s)
Amino Azúcares/síntesis química , Amino Azúcares/farmacología , Amino Azúcares/toxicidad , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colesterol/análogos & derivados , Colesterol/farmacología , Colesterol/toxicidad , Glicosilación , Humanos
4.
Int J Mol Sci ; 21(6)2020 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-32182973

RESUMEN

BACKGROUND: Atherosclerotic plaques are unstable, and their release may result in thrombosis; therefore, currently, antiplatelet therapy with anticoagulants is recommended for the treatment of acute coronary syndrome. The aim of this study was to assess the effect of oxidized cholesterol on human umbilical vascular endothelial cells (HUVECs). The study also examines the protective and repairing effect of dabigatran and rivaroxaban in a model of vascular endothelial damage with 25-hydroxycholesterol (25-OHC). METHODS: HUVECs were treated with compounds induce DNA single-strand breaks (SSBs) using the comet assay. Oxidative DNA damage was detected using endonuclease III (Nth) or human 8 oxoguanine DNA glycosylase (hOOG1). Reactive oxygen species (ROS) formation was determined using flow cytometry. RESULTS: 25-hydroxycholesterol caused DNA SSBs, induced oxidative damage and increased ROS in the HUVECs; ROS level was lowered by dabigatran and rivaroxaban. Only dabigatran was able to completely repair the DNA SSBs induced by oxysterol. Dabigatran was able to reduce the level of oxidative damage of pyrimidines induced by oxysterol to the level of control cells. CONCLUSIONS: Observed changes strongly suggest that the tested anticoagulants induced indirect repair of DNA by inhibiting ROS production. Furthermore, dabigatran appears to have a higher antioxidant activity than rivaroxaban.


Asunto(s)
Anticoagulantes/farmacología , Antioxidantes/farmacología , Daño del ADN , Dabigatrán/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Rivaroxabán/farmacología , Colesterol/análogos & derivados , Colesterol/toxicidad , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Estrés Oxidativo
5.
Dig Dis Sci ; 64(5): 1238-1256, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30511198

RESUMEN

BACKGROUND: There is a marked need for improved animal models of nonalcoholic steatohepatitis (NASH) to facilitate the development of more efficacious drug therapies for the disease. METHODS: Here, we investigated the development of fibrotic NASH in male Wistar rats fed a choline-deficient L-amino acid-defined (CDAA) diet with or without cholesterol supplementation for subsequent assessment of drug treatment efficacy in NASH biopsy-confirmed rats. The metabolic profile and liver histopathology were evaluated after 4, 8, and 12 weeks of dieting. Subsequently, rats with biopsy-confirmed NASH were selected for pharmacological intervention with vehicle, elafibranor (30 mg/kg/day) or obeticholic acid (OCA, 30 mg/kg/day) for 5 weeks. RESULTS: The CDAA diet led to marked hepatomegaly and fibrosis already after 4 weeks of feeding, with further progression of collagen deposition and fibrogenesis-associated gene expression during the 12-week feeding period. Cholesterol supplementation enhanced the stimulatory effect of CDAA on gene transcripts associated with fibrogenesis without significantly increasing collagen deposition. Pharmacological intervention with elafibranor, but not OCA, significantly reduced steatohepatitis scores, and fibrosis-associated gene expression, however, was unable to prevent progression in fibrosis scores. CONCLUSION: CDAA-fed rats develop early-onset progressive NASH, which offers the opportunity to probe anti-NASH compounds with potential disease-modifying properties.


Asunto(s)
Chalconas/uso terapéutico , Ácido Quenodesoxicólico/análogos & derivados , Colesterol/toxicidad , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Nutrientes/deficiencia , Propionatos/uso terapéutico , Animales , Ácido Quenodesoxicólico/uso terapéutico , Colesterol/administración & dosificación , Progresión de la Enfermedad , Masculino , Enfermedad del Hígado Graso no Alcohólico/patología , Ratas , Ratas Wistar
6.
J Appl Toxicol ; 39(9): 1348-1361, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31231834

RESUMEN

CJ-12,918, a 5-lipoxygenase (5-LO) inhibitor, caused cataracts during a 1-month safety assessment studies in rats whereas the structurally similar ZD-2138 was without effect. For CJ-12,918 analogs, blocking different sites of metabolic liability reduced (CJ-13,454) and eliminated (CJ-13,610) cataract formation in both rats and dogs. Using this chemical series as a test set, models and mechanisms of toxicity were first explored by testing the utility of ex vivo rat lens explant cultures as a safety screen. This model overpredicted the cataractogenic potential of ZD-2138 due to appreciably high lens drug levels and was abandoned in favor of a mechanism-based screen. Perturbations in lens sterol content, from a decline in lathosterol content, preceded cataract formation suggesting CJ-12,918 inhibited lens cholesterol biosynthesis (LCB). A 2-day bioassay in rats using ex vivo LCB assessments showed that the level of LCB inhibition was correlated with incidence of cataract formation in animal studies by these 5-LO inhibitors. Thereafter, this 2-day bioassay was applied to other pharmaceutical programs (neuronal nitric oxide synthase, sorbitol dehydrogenase inhibitor, squalene synthetase inhibitor and stearoyl-CoA desaturase-1 inhibitors/D4 antagonists) that demonstrated cataract formation in either rats or dogs. LCB inhibition >40% was associated with a high incidence of cataract formation in both rats and dogs that was species specific. Bioassay sensitivity/specificity were further explored with positive (RGH-6201/ciglitazone/U18666A) and negative (tamoxifen/naphthalene/galactose) mechanistic controls. This body of work over two decades shows that LCB inhibition was a common mechanism of cataract formation by pharmaceutical agents and defined a level of inhibition >40% that was typically associated with causing cataracts in safety assessment studies typically ≥1 month.


Asunto(s)
Catarata/inducido químicamente , Colesterol/biosíntesis , Colesterol/toxicidad , Inhibidores Enzimáticos/toxicidad , Cristalino/efectos de los fármacos , Cristalino/metabolismo , Tiazolidinedionas/toxicidad , Animales , Animales de Laboratorio , Catarata/metabolismo , Perros , Femenino , Masculino , Preparaciones Farmacéuticas , Ratas , Ratas Sprague-Dawley
7.
J Biol Chem ; 292(1): 146-160, 2017 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-27881673

RESUMEN

The liver plays a key role in cholesterol metabolism. Impaired hepatic cholesterol homeostasis causes intracellular free cholesterol accumulation and hepatocyte injury. Sortilin 1 (SORT1) is a lysosomal trafficking receptor that was identified by genome-wide association studies (GWAS) as a novel regulator of cholesterol metabolism in humans. Here we report that SORT1 deficiency protected against cholesterol accumulation-induced liver injury and inflammation in mice. Using an LC-MS/MS-based proteomics approach, we identified liver carboxylesterase 1 (CES1) as a novel SORT1-interacting protein. Mechanistic studies further showed that SORT1 may regulate CES1 lysosomal targeting and degradation and that SORT1 deficiency resulted in higher liver CES1 protein abundance. Previous studies have established an important role of hepatic CES1 in promoting intracellular cholesterol mobilization, cholesterol efflux, and bile acid synthesis. Consistently, high cholesterol atherogenic diet-challenged Sort1 knock-out mice showed less hepatic free cholesterol accumulation, increased bile acid synthesis, decreased biliary cholesterol secretion, and the absence of gallstone formation. SORT1 deficiency did not alter hepatic ceramide and fatty acid metabolism in high cholesterol atherogenic diet-fed mice. Finally, knockdown of liver CES1 in mice markedly increased the susceptibility to high cholesterol diet-induced liver injury and abolished the protective effect against cholesterol lipotoxicity in Sort1 knock-out mice. In summary, this study identified a novel SORT1-CES1 axis that regulates cholesterol-induced liver injury, which provides novel insights that improve our current understanding of the molecular links between SORT1 and cholesterol metabolism. This study further suggests that therapeutic inhibition of SORT1 may be beneficial in improving hepatic cholesterol homeostasis in metabolic and inflammatory liver diseases.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Hidrolasas de Éster Carboxílico/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Colesterol/toxicidad , Hepatocitos/patología , Inflamación/patología , Animales , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Hidrolasas de Éster Carboxílico/genética , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Femenino , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Inflamación/etiología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética
8.
FASEB J ; 31(1): 356-367, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27737897

RESUMEN

The contribution of metabolic factors on the severity of osteoarthritis (OA) is not fully appreciated. This study aimed to define the effects of hypercholesterolemia on the progression of OA. Apolipoprotein E-deficient (ApoE-/-) mice and rats with diet-induced hypercholesterolemia (DIHC) rats were used to explore the effects of hypercholesterolemia on the progression of OA. Both models exhibited OA-like changes, characterized primarily by a loss of proteoglycans, collagen and aggrecan degradation, osteophyte formation, changes to subchondral bone architecture, and cartilage degradation. Surgical destabilization of the knees resulted in a dramatic increase of degradative OA symptoms in animals fed a high-cholesterol diet compared with controls. Clinically relevant doses of free cholesterol resulted in mitochondrial dysfunction, overproduction of reactive oxygen species (ROS), and increased expression of degenerative and hypertrophic markers in chondrocytes and breakdown of the cartilage matrix. We showed that the severity of diet-induced OA changes could be attenuated by treatment with both atorvastatin and a mitochondrial targeting antioxidant. The protective effects of the mitochondrial targeting antioxidant were associated with suppression of oxidative damage to chondrocytes and restoration of extracellular matrix homeostasis of the articular chondrocytes. In summary, our data show that hypercholesterolemia precipitates OA progression by mitochondrial dysfunction in chondrocytes, in part by increasing ROS production and apoptosis. By addressing the mitochondrial dysfunction using antioxidants, we were able attenuate the OA progression in our animal models. This approach may form the basis for novel treatment options for this OA risk group in humans.-Farnaghi, S., Prasadam, I., Cai, G., Friis, T., Du, Z., Crawford, R., Mao, X., Xiao, Y. Protective effects of mitochondria-targeted antioxidants and statins on cholesterol-induced osteoarthritis.


Asunto(s)
Anticolesterolemiantes/farmacología , Atorvastatina/farmacología , Colesterol/toxicidad , Hipercolesterolemia/inducido químicamente , Mitocondrias/efectos de los fármacos , Osteoartritis/etiología , Animales , Antioxidantes , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Remodelación Ósea , Colesterol/sangre , Condrocitos/efectos de los fármacos , Grasas de la Dieta , Hipercolesterolemia/sangre , Hipercolesterolemia/complicaciones , Masculino , Ratones , Ratones Noqueados , Osteoartritis/patología , Osteoartritis/prevención & control , Ratas , Ratas Wistar
9.
Adv Exp Med Biol ; 975 Pt 2: 1093-1102, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28849525

RESUMEN

Taurine is thought to affect bone in rats favorably. However, studies on the actions of this estrogen deficiency and high cholesterol diet factors on the bone metabolism are limited. In this study, the protective effect of taurine on bone was determined. Thirty-two 42 days old female SD rats were placed in individual stainless cages. Given to rats was fed to chow (Samyang Corporation, South Korea) and deionized water for a 4 days adaptation period. After the period of adaptation, Half of the rats were induced estrogen deficiency model by ovariectomy (OVX), and the left rats with sham-operated were used control (SHAM). For six weeks, the OVX and SHAM rats had separately a 2% taurine supplemented diet with ad libitum in both the water and the food. DEXA for small animals (PIXImus, GE Lunar co, Wisconsin) was used to determine spinal and femoral bone. The concentrations of serum calcium and phosphorus were also measured. The monitoring of bone formation was done by determining the serum ALP and osteocalcin. Urinary DPD the values were determined as index of bone resorption. Statistical measure was done with SAS (version 9.3). A lower overall intake of the daily food was observed in non-ovariectomized rats than in the OVX rats. At sacrifice, a much greater body weight was observed in ovariectomized group compare to non-operated group. That difference was absent in both fed taurine SHAM and OVX rats. Serum calcium and phosphorus were not statistically different by taurine supplementation. Urinary excretion of calcium was not effected by taurine supplementation. Serum ALP and was significantly decreased by taurine in OVX rats (p < 0.05). For the spine BMD and BMC, there was no difference among SHAM and OVX rats by taurine. Spine BMC per body weight of taurine groups were higher than control groups (p < 0.1). No significant difference was observed after taurine supplementation in femur BMD and BMC. The analysis of the results suggest that taurine supplementation modulates the bone mineral contents in postmenopausal model rats fed with high cholesterol diet.


Asunto(s)
Huesos/efectos de los fármacos , Colesterol/toxicidad , Taurina/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Estrógenos/deficiencia , Femenino , Humanos , Osteoporosis Posmenopáusica , Ovariectomía , Ratas , Ratas Sprague-Dawley
10.
Proc Natl Acad Sci U S A ; 111(46): E4972-80, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25368174

RESUMEN

The HDL receptor scavenger receptor, class B type I (SR-BI) controls the structure and fate of plasma HDL. Female SR-BI KO mice are infertile, apparently because of their abnormal cholesterol-enriched HDL particles. We examined the growth and meiotic progression of SR-BI KO oocytes and found that they underwent normal germinal vesicle breakdown; however, SR-BI KO eggs, which had accumulated excess cholesterol in vivo, spontaneously activated, and they escaped metaphase II (MII) arrest and progressed to pronuclear, MIII, and anaphase/telophase III stages. Eggs from fertile WT mice were activated when loaded in vitro with excess cholesterol by a cholesterol/methyl-ß-cyclodextrin complex, phenocopying SR-BI KO oocytes. In vitro cholesterol loading of eggs induced reduction in maturation promoting factor and MAPK activities, elevation of intracellular calcium, extrusion of a second polar body, and progression to meiotic stages beyond MII. These results suggest that the infertility of SR-BI KO females is caused, at least in part, by excess cholesterol in eggs inducing premature activation and that cholesterol can activate WT mouse eggs to escape from MII arrest. Analysis of SR-BI KO female infertility raises the possibility that abnormalities in cholesterol metabolism might underlie some cases of human female infertility of unknown etiology.


Asunto(s)
HDL-Colesterol/metabolismo , Colesterol/toxicidad , Infertilidad Femenina/etiología , Meiosis/efectos de los fármacos , Oocitos/efectos de los fármacos , Receptores Depuradores de Clase B/deficiencia , Animales , Supervivencia Celular , Ácido Egtácico/farmacología , Femenino , Sistema de Señalización de MAP Quinasas , Meiosis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/citología , Cuerpos Polares , Receptores Depuradores de Clase B/fisiología , Estroncio/farmacología , beta-Ciclodextrinas/farmacología
11.
Anal Biochem ; 492: 49-55, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26366784

RESUMEN

Free cholesterol in mammalian cells resides mostly in the plasma membrane, where it plays an important role in cellular homeostasis. We synthesized a new fluorescent cholesterol analogue that retained an intact alkyl chain and the sterane backbone of cholesterol. The hydroxyl group of cholesterol was converted into an amino group that was covalently linked to the fluorophore tetramethylrhodamine to retain the ability to form hydrogen bonds with adjacent molecules. Incubating live MDCK (Madin-Darby canine kidney) cells with our fluorescent cholesterol analogue resulted in the generation of intense signals that were detected by microscopy at the plasma membrane. Incubation with the analogue exerted minimal, if any, influence on cell growth, indicating that it could serve as a useful tool for analyzing free cholesterol at the plasma membrane.


Asunto(s)
Membrana Celular/metabolismo , Colesterol/análogos & derivados , Colorantes Fluorescentes/química , Animales , Compuestos de Boro/química , Membrana Celular/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colesterol/metabolismo , Colesterol/toxicidad , Perros , Células de Riñón Canino Madin Darby , Microscopía Fluorescente
12.
Langmuir ; 32(39): 10126-10135, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27597442

RESUMEN

Cholesterol-containing molecules or nanoparticles play a significant role in achieving favorable plasma membrane imaging and efficient cellular uptake of drugs by the excellent membrane anchoring capability of the cholesterol moiety. By linking cholesterol to a water-soluble component (such as poly(ethylene glycol), PEG), the resulting cholesterol-PEG conjugate can form micelles in aqueous solution through self-assembly, and such a micellar structure represents an important drug delivery vehicle in which hydrophobic drugs can be encapsulated. However, the understanding of the subcellular fate and cytotoxicity of cholesterol-PEG conjugates themselves remains elusive. Herein, by using cholesterol-PEG2000-fluorescein isothiocyanate (Chol-PEG-FITC) as a model system, we found that the Chol-PEG-FITC molecules could attach to the plasma membranes of mammalian cells within 10 min and such a firm membrane attachment could last at least 1 h, displaying excellent plasma membrane staining performance that surpassed that of commonly used commercial membrane dyes such as DiD and CellMask. Besides, we systematically studied the endocytosis pathway and intracellular distribution of Chol-PEG-FITC and found that the cell surface adsorption and endocytosis processes of Chol-PEG-FITC molecules were lipid-raft-dependent. After internalization, the Chol-PEG-FITC molecules gradually reached many organelles with membrane structures. At 5 h, they were mainly distributed in lysosomes and the Golgi apparatus, with some in the endoplasmic reticulum (ER) and very few in the mitochondrion. At 12 h, the Chol-PEG-FITC molecules mostly aggregated in the Golgi apparatus and ER close to the nucleus. Finally, we demonstrated that Chol-PEG-FITC was toxic to mammalian cells only at concentrations above 50 µM. In summary, Chol-PEG-FITC can be a promising plasma membrane imaging reagent to avoid the fast cellular internalization and quick membrane detachment problems faced by commercial membrane dyes. We believe that the investigation of the dynamic subcellular fate of Chol-PEG-FITC can provide important knowledge to facilitate the use of cholesterol-PEG conjugates in fields such as cell surface engineering and drug delivery.


Asunto(s)
Membrana Celular/metabolismo , Colesterol/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/metabolismo , Polietilenglicoles/metabolismo , Colesterol/análogos & derivados , Colesterol/química , Colesterol/toxicidad , Endocitosis/efectos de los fármacos , Citometría de Flujo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/toxicidad , Colorantes Fluorescentes/química , Colorantes Fluorescentes/toxicidad , Humanos , Células MCF-7 , Microdominios de Membrana/metabolismo , Micelas , Microscopía Confocal , Tamaño de la Partícula , Polietilenglicoles/química , Polietilenglicoles/toxicidad
13.
Lab Invest ; 95(8): 914-24, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26097999

RESUMEN

Accumulation of cholesterol in the liver is associated with the development of non-alcoholic steatohepatitis-related fibrosis. However, underlying mechanisms are not well understood. The present study investigated the role of inducible nitric oxide synthase (iNOS) in cholesterol-induced liver fibrosis by feeding wild-type (WT) and iNOS-deficient mice with control or high-cholesterol diet (HCD) for 6 weeks. WT mice fed with HCD developed greater liver fibrosis, compared with iNOS-deficient mice, as evident by Sirius red staining and higher expression levels of profibrotic genes. Enhanced liver fibrosis in the presence of iNOS was associated with hypoxia-inducible factor-1α stabilization, matrix metalloproteinase-9 expression, and enhanced hepatic DNA damage. The profibrotic role of iNOS was also demonstrated in vivo using a selective inhibitor of iNOS as well as in vitro in a rat liver stellate cell line (HSC-T6). In conclusion, these findings suggest that iNOS is an important mediator in HCD-induced liver fibrosis.


Asunto(s)
Colesterol/toxicidad , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Animales , Línea Celular , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Dieta Alta en Grasa , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas
14.
Eur J Neurosci ; 41(10): 1345-55, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25847620

RESUMEN

Elevations in neuronal cholesterol have been associated with several degenerative diseases. An enhanced excitability and synchronous firing in surviving neurons are among the sequels of neuronal death in these diseases and also in some epileptic syndromes. Here, we attempted to increase neuronal cholesterol levels, using a short hairpin RNA to suppress expression of the enzyme cytochrome P450 family 46, subfamily A, polypeptide 1 gene (CYP46A1). This protein hydroxylates cholesterol and so facilitates transmembrane extrusion. A short hairpin RNA CYP46A1construction coupled to the adeno-associated virus type 5 was injected focally and unilaterally into mouse hippocampus. It was selectively expressed first in neurons of the cornu ammonis (hippocampus) (CA)3a region. Cytoplasmic and membrane cholesterol increased, and the neuronal soma volume increased and then decreased before pyramidal cells died. As CA3a pyramidal cells died, interictal electroencephalographic (EEG) events occurred during exploration and non-rapid eye movement sleep. With time, neuronal death spread to involve pyramidal cells and interneurons of the CA1 region. CA1 neuronal death was correlated with a delayed local expression of phosphorylated tau. Astrocytes were activated throughout the hippocampus and microglial activation was specific to regions of neuronal death. CA1 neuronal death was correlated with distinct aberrant EEG activity. During exploratory behaviour and rapid eye movement sleep, EEG oscillations at 7-10 Hz (theta) could accelerate to 14-21 Hz (beta) waves. They were accompanied by low-amplitude, high-frequency oscillations of peak power at ~300 Hz and a range of 250-350 Hz. Although episodes of EEG acceleration were not correlated with changes in exploratory behaviour, they were followed in some animals by structured seizure-like discharges. These data strengthen links between increased cholesterol, neuronal sclerosis and epileptic behaviour.


Asunto(s)
Región CA3 Hipocampal/patología , Región CA3 Hipocampal/fisiopatología , Colesterol/toxicidad , Epilepsia/patología , Células Piramidales/patología , Células Piramidales/fisiología , Animales , Astrocitos/metabolismo , Región CA3 Hipocampal/metabolismo , Muerte Celular , Colesterol/metabolismo , Colesterol 24-Hidroxilasa , Dependovirus/fisiología , Electroencefalografía , Epilepsia/etiología , Femenino , Vectores Genéticos , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Fosforilación , Células Piramidales/metabolismo , ARN Interferente Pequeño/genética , Esclerosis , Esteroide Hidroxilasas/farmacología , Proteínas tau/metabolismo
15.
Org Biomol Chem ; 13(14): 4310-20, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25762431

RESUMEN

Herein, we present the design and synthesis of new redox-active monomeric and dimeric (gemini) cationic lipids based on ferrocenylated cholesterol derivatives for gene delivery. The cationic cholesterols are shown to be transfection efficient after being formulated with the neutral helper lipid DOPE in the presence of serum (FBS). The redox activity of the resulting co-liposomes and their lipoplexes could be regulated using the alkanyl ferrocene moiety attached to the ammonium head groups of the cationic cholesterols. Atomic force microscopy (AFM), dynamic light scattering (DLS) and zeta potential measurements were performed to characterize the co-liposomal aggregates and their complexes with pDNA. The transfection efficiency of lipoplexes could be tuned by changing the oxidation state of the ferrocene moiety. The gene transfection capability was assayed in terms of green fluorescence protein (GFP) expression using pEGFP-C3 plasmid DNA in three cell lines of different origins, namely Caco-2, HEK293T and HeLa, in the presence of serum. The vesicles possessing ferrocene in the reduced state induced an efficient transfection, even better than a commercial reagent Lipofectamine 2000 (Lipo 2000) as evidenced by flow cytometry and fluorescence microscopy. All the co-liposomes containing the oxidized ferrocene displayed diminished levels of gene expression. Gene transfection events from the oxidized co-liposomes were further potentiated by introducing ascorbic acid (AA) as a reducing agent during lipoplex incubation with cells, leading to the resumption of transfection activity. Assessment of transfection capability of both reduced and oxidized co-liposomes was also undertaken following cellular internalization of labelled pDNA using confocal microscopy and flow cytometry. Overall, we demonstrate here controlled gene transfection activities using redox-driven, transfection efficient cationic monomeric and dimeric cholesterol lipids. Such systems could be used in gene delivery applications where transfection needs to be performed spatially or temporally.


Asunto(s)
Colesterol/química , Dimerización , Portadores de Fármacos/química , Compuestos Ferrosos/química , Suero/metabolismo , Transfección/métodos , Animales , Ácido Ascórbico/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colesterol/metabolismo , Colesterol/toxicidad , Portadores de Fármacos/metabolismo , Portadores de Fármacos/toxicidad , Humanos , Liposomas/química , Metalocenos , Oxidación-Reducción
16.
J Nanosci Nanotechnol ; 15(6): 4058-69, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26369013

RESUMEN

In this study, three types of galactosylated cholesterol (i.e., gal-PEG194-chol, gal-PEG1000-chol and gal-PEG2000-chol) were synthesized with one terminal of polyethylene glycol of various chain lengths conjugated to the galactoside moiety, and the other terminal conjugated to the cholesterol. The galactose-modified liposomes were prepared by thin film-hydration method and doxorubicin (DOX) was loaded to the liposomes by using a ammonium sulfate gradient procedure. The liposomal formulations with galactosylated cholesterol were characterized. Flow cytometry and laser confocal scanning microscopy analyses showed that the galactose-modified liposomes facilitated the intracellular uptake of liposomes into HepG2 via asialoglycoprotein receptor (ASGP-R) mediated endocytosis. Cytotoxicity assay showed that the cell proliferation inhibition effect of galactose-modified liposomes was higher than that of the unmodified liposomes. Additionally, the study on frozen section of liver showed that the galactose-modified liposomes enhanced the intracellular uptake of liposomes into hepatocytes. Taken together, these results suggested that liposomes containing such galactosylated cholesterol (i.e., gal-PEG-chol), had a great potential as drug delivery carriers for hepatocyte-selective targeting.


Asunto(s)
Colesterol/análogos & derivados , Galactosa/química , Hepatocitos/metabolismo , Liposomas/química , Polietilenglicoles/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Receptor de Asialoglicoproteína/metabolismo , Proliferación Celular/efectos de los fármacos , Colesterol/química , Colesterol/farmacocinética , Colesterol/toxicidad , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Células HeLa , Células Hep G2 , Humanos , Liposomas/farmacocinética , Liposomas/toxicidad , Hígado/química , Hígado/metabolismo , Ratones , Polietilenglicoles/farmacocinética , Polietilenglicoles/toxicidad , Distribución Tisular
17.
Artículo en Inglés | MEDLINE | ID: mdl-24561378

RESUMEN

The organoselenium compound, dicholesteroyl diselenide (DCDS) is a structural analogue of diphenyl diselenide (DPDS) and may be considered as a promising antioxidant drug in vivo. Nevertheless, little is known about the toxicological properties of DCDS. In the present study we evaluated the cytotoxic, genotoxic and mutagenic properties of DCDS in Chinese hamster lung fibroblasts (V79) and in strains of the yeast Saccharomyces cerevisiae, proficient and deficient in several DNA-repair pathways. The results with V79 cells show that DCDS induced cytotoxicity, GSH depletion and elevation of lipid peroxidation at lower concentrations than did DPDS. DCDS also generated single- and double-strand DNA breaks in V79 cells, both in the presence and in the absence of metabolic activation, as revealed by alkaline and neutral comet assays. Moreover, the induction of oxidative DNA base-damage was demonstrated by means of a modified comet assay with formamidopyrimidine-DNA glycosylase and endonuclease III. Treatment with DCDS also induced micronucleus formation in V79 cells as well as point and frame-shift mutations in a haploid wild-type strain of S. cerevisiae. Yeast mutants defective in base excision-repair proteins were the most sensitive to DCDS. Pre-incubation with N-acetylcysteine reduced DCDS's oxidative, genotoxic and mutagenic effects in yeast and in V79 cells. Our findings indicate that the presence of cholesteroyl substituents in DCDS results in elevation of its cytotoxic and genotoxic potential compared with that of DPDS in yeast and in V79 cells. However, due to dose-dependent contrasting behaviour of organoselenium compounds and differences in their toxicity in in vitro and in vivo systems, further studies are needed in order to establish the non-toxic concentration range for treatment in mammals.


Asunto(s)
Colesterol/análogos & derivados , Daño del ADN , Micronúcleos con Defecto Cromosómico/inducido químicamente , Mutágenos/toxicidad , Compuestos de Organoselenio/toxicidad , Saccharomyces cerevisiae/efectos de los fármacos , Animales , Biomarcadores/análisis , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colesterol/toxicidad , Ensayo Cometa , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Mutación del Sistema de Lectura/efectos de los fármacos , Humanos , Peroxidación de Lípido/efectos de los fármacos , Pruebas de Micronúcleos , Estrés Oxidativo/efectos de los fármacos , Saccharomyces cerevisiae/genética , Pruebas de Toxicidad/métodos
18.
Lipids Health Dis ; 13: 194, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25515296

RESUMEN

BACKGROUND: Comparable to commercial expensive high-fat diets, cholesterol-cholate-butterfat (CCB) diet has also been used to induce hyperlipidemia in rats. Our objective was to explore its influence on multiple organs. Consequence of fasting was also analysed. METHODS: Rats in groups 1 and 2 received normal diet (ND) whereas groups 3 and 4 received CCB-diet. Food was withdrawn daily for two hours from groups 2 (ND-F) and 4 (CCB-F). Blood was collected at fourth and sixth week for biochemical estimation; Morris water maze was done in the sixth week for learning ability and memory; after which aortae were isolated for vascular reactivity. RESULTS: Apart from hyperlipidemia, CCB also induced hyperglycemia with marked increase in hepatic enzymes: gamma-glutamyl transferase (GGT), alanine and aspartate aminotransferase (ALT and AST); and vascular biomarkers: uric acid (UA), phosphorus and alkaline phosphatase (ALP). Isolated aortae, pre-contracted with phenylephrine, were less responsive to acetylcholine indicating endothelial dysfunction--serum nitric oxide (NO) production was limited with subsequent inhibition of endothelial NO synthase. CCB diet also compromised learning ability. CCB-coupled fasting potentiated hyperlipidemia but prevented memory-loss. CONCLUSION: We introduce CCB-diet for multi-organ dysfunction in rats, and propose its use for research on cardiovascular diseases and associated manifestations involving immense interplay of integrated pathways.


Asunto(s)
Colatos/toxicidad , Colesterol/toxicidad , Dieta Alta en Grasa/efectos adversos , Animales , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatología , Mantequilla , Femenino , Hiperlipidemias/etiología , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Aprendizaje por Laberinto , Ratas Sprague-Dawley
19.
Int J Mol Sci ; 15(11): 20339-54, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25383677

RESUMEN

Here, three novel cholesterol (Ch)/low molecular weight polyethylene glycol (PEG) conjugates, termed α, ω-cholesterol-functionalized PEG (Ch2-PEGn), were successfully synthesized using three kinds of PEG with different average molecular weight (PEG600, PEG1000 and PEG2000). The purpose of the study was to investigate the potential application of novel cationic liposomes (Ch2-PEGn-CLs) containing Ch2-PEGn in gene delivery. The introduction of Ch2-PEGn affected both the particle size and zeta potential of cationic liposomes. Ch2-PEG2000 effectively compressed liposomal particles and Ch2-PEG2000-CLs were of the smallest size. Ch2-PEG1000 and Ch2-PEG2000 significantly decreased zeta potentials of Ch2-PEGn-CLs, while Ch2-PEG600 did not alter the zeta potential due to the short PEG chain. Moreover, the in vitro gene transfection efficiencies mediated by different Ch2-PEGn-CLs also differed, in which Ch2-PEG600-CLs achieved the strongest GFP expression than Ch2-PEG1000-CLs and Ch2-PEG2000-CLs in SKOV-3 cells. The gene delivery efficacy of Ch2-PEGn-CLs was further examined by addition of a targeting moiety (folate ligand) in both folate-receptor (FR) overexpressing SKOV-3 cells and A549 cells with low expression of FR. For Ch2-PEG1000-CLs and Ch2-PEG2000-CLs, higher molar ratios of folate ligand resulted in enhanced transfection efficacies, but Ch2-PEG600-CLs had no similar in contrast. Additionally, MTT assay proved the reduced cytotoxicities of cationic liposomes after modification by Ch2-PEGn. These findings provide important insights into the effects of Ch2-PEGn on cationic liposomes for delivering genes, which would be beneficial for the development of Ch2-PEGn-CLs-based gene delivery system.


Asunto(s)
Cationes/química , Colesterol/análogos & derivados , Técnicas de Transferencia de Gen , Liposomas/química , Polietilenglicoles/química , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Colesterol/síntesis química , Colesterol/química , Colesterol/toxicidad , Fluorescencia , Receptor 1 de Folato/metabolismo , Ácido Fólico/química , Humanos , Ligandos , Espectrometría de Masas , Peso Molecular , Tamaño de la Partícula , Espectroscopía de Fotoelectrones , Polietilenglicoles/síntesis química , Polietilenglicoles/toxicidad , Espectroscopía de Protones por Resonancia Magnética , Electricidad Estática , Transfección , Temperatura de Transición
20.
Biomacromolecules ; 14(11): 4135-49, 2013 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24125032

RESUMEN

A library of cholesterol-derived ionic copolymers were previously synthesized via reversible addition-fragmentation chain transfer (RAFT) polymerization as 'smart' gene delivery vehicles that hold diverse surface charges. Polyplex systems formed with anionic poly(methacrylic acid-co-cholesteryl methacrylate) (P(MAA-co-CMA)) and cationic poly(dimethylamino ethyl methacrylate-co-cholesteryl methacrylate) (Q-P(DMAEMA-co-CMA)) copolymer series were evaluated for their therapeutic efficiency. Cell viability assays, conducted on SHEP, HepG2, H460, and MRC5 cell lines, revealed that alterations in the copolymer composition (CMA mol %) affected the cytotoxicity profile. Increasing the number of cholesterol moieties in Q-P(DMAEMA-co-CMA) copolymers reduced the overall toxicity (in H460 and HepG2 cells) while P(MAA-co-CMA) series displayed no significant toxicity regardless of the CMA content. Agarose gel electrophoresis was employed to investigate the formation of stable polyplexes and determine their complete conjugation ratios. P(MAA-co-CMA) copolymer series were conjugated to DNA through a cationic linker, oligolysine, while Q-P(DMAEMA-co-CMA)-siRNA complexes were readily formed via electrostatic interactions at conjugation ratios beginning from 6:1:1 (oligolysine-P(MAA-co-CMA)-DNA) and 20:1 (Q-P(DMAEMA-co-CMA)-siRNA), respectively. The hydrodynamic diameter, ζ potential and complex stability of the polyplexes were evaluated in accordance to complexation ratios and copolymer composition by dynamic light scattering (DLS). The therapeutic efficiency of the conjugates was assessed in SHEP cells via transfection and imaging assays using RT-qPCR, Western blotting, flow cytometry, and confocal microscopy. DNA transfection studies revealed P(MAA-co-CMA)-oligolysine-DNA ternary complexes to be ineffective transfection vehicles that mostly adhere to the cell surface as opposed to internalizing and partaking in endosomal disrupting activity. The transfection efficiency of Q-P(DMAEMA-co-CMA)-GFP siRNA complexes were found to be polymer composition and N/P ratio dependent, with Q-2% CMA-GFP siRNA polyplexes at N/P ratio 20:1 showing the highest gene suppression in GFP expressing SHEP cells. Cellular internalization studies suggested that Q-P(DMAEMA-co-CMA)-siRNA conjugates efficiently escaped the endolysosomal pathway and released siRNA into the cytoplasm. The gene delivery profile, reported herein, illuminates the positive and negative attributes of each therapeutic design and strongly suggests Q-P(DMAEMA-co-CMA)-siRNA particles are extremely promising candidates for in vivo applications of siRNA therapy.


Asunto(s)
Colesterol/química , ADN/administración & dosificación , Polímeros/química , ARN Interferente Pequeño/administración & dosificación , Transfección/métodos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colesterol/administración & dosificación , Colesterol/farmacología , Colesterol/toxicidad , Ésteres del Colesterol/administración & dosificación , Ésteres del Colesterol/química , Ésteres del Colesterol/toxicidad , Relación Dosis-Respuesta a Droga , Terapia Genética/métodos , Células Hep G2 , Humanos , Iones/administración & dosificación , Iones/química , Iones/farmacología , Iones/toxicidad , Metacrilatos/administración & dosificación , Metacrilatos/química , Metacrilatos/toxicidad , Modelos Moleculares , Estructura Molecular , Tamaño de la Partícula , Polímeros/administración & dosificación , Polímeros/toxicidad , Ácidos Polimetacrílicos/administración & dosificación , Ácidos Polimetacrílicos/química , Ácidos Polimetacrílicos/toxicidad , Relación Estructura-Actividad , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA