Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Mol Pharm ; 16(5): 1958-1970, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30912953

RESUMO

Fenofibrate is a peroxisome proliferator-activated receptor α (PPARα) agonist and has been shown to have therapeutic effects on diabetic retinopathy (DR). However, the effects of fenofibrate through systemic administration are not as potent as desired due to inefficient drug delivery to the retina. The present study aimed to explore the sustained therapeutic effects of fenofibrate-loaded biodegradable nanoparticles (NP) on both DR and neovascular age-related macular degeneration (AMD). Fenofibrate was successfully encapsulated into poly(lactic- co-glycolic acid) (PLGA) NP (Feno-NP), and Feno-NP were optimized by varying polymer composition to achieve high drug loading and prolonged drug release. The Feno-NP made of PLGA 34 kDa demonstrated a drug content of 6% w/w and a sustained drug release up to 60 days in vitro. Feno-NP (PLGA 34 kDa) was selected for following in vivo studies, and one single intravitreal (IVT) injection of Feno-NP into rat eyes with a 30G fine needle maintained sustained fenofibric acid drug level in the eye for more than 60 days. The efficacy of Feno-NP in DR and neovascular AMD was investigated using streptozotocin (STZ)-induced diabetic rats, laser-induced choroidal neovascularization (CNV) rats, and very low-density lipoprotein receptor knockout ( Vldlr -/-) mice. Therapeutic effects of Feno-NP were evaluated by measuring electroretinogram (ERG), retinal vascular leakage, leukostasis, CNV size, and retinal levels of vascular endothelial growth factor (VEGF) and intracellular adhesion molecule-1 (ICAM-1). In diabetic rats, Feno-NP ameliorated retinal dysfunctions, reduced retinal vascular leakage, inhibited retinal leukostasis, and downregulated the overexpression of VEGF and ICAM-1 at 8 weeks after one IVT injection. In addition, Feno-NP reduced retinal vascular leakage and CNV formation in both CNV rats and Vldlr -/- mice. Moreover, no toxicity of Feno-NP or Blank-NP to retinal structure and function was detected. Feno-NP exhibited good physiochemical characteristics and controlled drug release profile, conferring prolonged beneficial effects on DR and neovascular AMD.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Retinopatia Diabética/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Fenofibrato/análogos & derivados , Hipolipemiantes/uso terapêutico , Nanopartículas/química , Degeneração Macular Exsudativa/tratamento farmacológico , Animais , Permeabilidade Capilar , Neovascularização de Coroide/tratamento farmacológico , Diabetes Mellitus Experimental/induzido quimicamente , Liberação Controlada de Fármacos , Fenofibrato/química , Fenofibrato/farmacocinética , Fenofibrato/uso terapêutico , Hipolipemiantes/química , Molécula 1 de Adesão Intercelular/metabolismo , Leucostasia/tratamento farmacológico , Camundongos , Camundongos Knockout , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Ratos , Ratos Endogâmicos BN , Retina/efeitos dos fármacos , Retina/metabolismo , Estreptozocina/efeitos adversos , Estreptozocina/farmacologia , Distribuição Tecidual , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Diabetes ; 68(5): 1040-1053, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30765336

RESUMO

Fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist, reduces lower limb amputations in patients with type 2 diabetes. The mechanism is, however, unknown. In this study, we demonstrate that fenofibrate markedly attenuates diabetes-related impairment of ischemia-mediated angiogenesis. In a murine model of hindlimb ischemia, daily oral fenofibrate treatment restored diabetes-impaired blood flow recovery, foot movement, hindlimb capillary density, vessel diameter, and vascular endothelial growth factor signaling to nondiabetic levels in both wild-type and PPARα-knockout mice, indicating that these fenofibrate effects are largely PPARα independent. In vitro, fenofibric acid (FFA) rescued high glucose-induced (25 mmol/L) impairment of endothelial cell migration, tubulogenesis, and survival in a PPARα-independent manner. Interestingly, fenofibrate in vivo and FFA in vitro reversed high glucose-induced expression of thioredoxin-interacting protein (TXNIP), an exquisitely glucose-inducible gene previously identified as a critical mediator of diabetes-related impairment in neovascularization. Conversely, adenoviral overexpression of TXNIP abrogated the restorative effects of FFA on high glucose-impaired endothelial cell function in vitro, indicating that the effects of FFA are mediated by TXNIP. We conclude that fenofibrate rescues diabetic impairment in ischemia-mediated angiogenesis, in large part, by PPARα-independent regulation of TXNIP. These findings may therefore explain the reduction in amputations seen in patients with diabetes treated with fenofibrate.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Fenofibrato/uso terapêutico , Isquemia/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Animais , Proteínas de Transporte/metabolismo , Fenofibrato/análogos & derivados , Glucose/farmacologia , Membro Posterior/efeitos dos fármacos , Membro Posterior/patologia , Isquemia/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos
3.
Invest Ophthalmol Vis Sci ; 58(12): 5065-5075, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28980001

RESUMO

Purpose: This study was designed to evaluate effects of fenofibric acid (Feno-FA), a peroxisome proliferator-activated receptor-alpha (PPARα) agonist, on ocular neovascularization (NV) in models recapitulating neovascular age-related macular degeneration (AMD), and to explore whether the effects are PPARα dependent. Methods: Laser-induced choroidal NV (CNV) in rats and very low-density lipoprotein receptor knockout (Vldlr-/-) mice received daily intraperitoneal injections of Feno-FA or vehicle. Vascular leakage was examined by fundus fluorescein angiography and permeability assay using Evans blue as tracer. In CNV rats, severity of CNV was evaluated by CNV areas and CNV volume. In Vldlr-/- mice, subretinal NV (SRNV) and intraretinal NV (IRNV) were quantified in choroid flat mount and retina flat mount, respectively. Inflammatory factors were measured using Western blotting and retinal leukostasis assay. Further, Pparα-/- mice and age-matched wild-type (WT) mice were used for laser-induced CNV and treated with Feno-FA to explore the underlying mechanism. Results: Feno-FA significantly reduced vascular leakage in CNV rats and Vldlr-/- mice, reduced CNV volume in laser-induced CNV rats, and suppressed SRNV and IRNV in Vldlr-/- mice. In addition, Feno-FA downregulated the expression of inflammatory factors, including VEGF, TNF-α, and intercellular cell adhesion molecule-1 (ICAM-1), in the eyecups of CNV rats and decreased adherent retinal leukocytes in Vldlr-/- mice. Furthermore, Pparα-/- mice developed more severe CNV compared with WT mice, and PPARα knockout abolished the beneficial effects of Feno-FA on CNV. Conclusions: Feno-FA has therapeutic effects on ocular NV in models recapitulating neovascular AMD through a PPARα-dependent mechanism.


Assuntos
Neovascularização de Coroide/tratamento farmacológico , Modelos Animais de Doenças , Fenofibrato/análogos & derivados , Hipolipemiantes/uso terapêutico , PPAR alfa/agonistas , Degeneração Macular Exsudativa/tratamento farmacológico , Animais , Western Blotting , Permeabilidade Capilar , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Fenofibrato/uso terapêutico , Angiofluoresceinografia , Injeções Intraperitoneais , Molécula 1 de Adesão Intercelular/metabolismo , Leucostasia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR alfa/genética , PPAR alfa/metabolismo , Ratos , Ratos Endogâmicos BN , Receptores de LDL/genética , Receptores de LDL/metabolismo , Tomografia de Coerência Óptica , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Degeneração Macular Exsudativa/metabolismo , Degeneração Macular Exsudativa/patologia
4.
J Cell Biochem ; 118(4): 754-763, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27618583

RESUMO

Increasing apolipoproteinA-I (apoA-I) production may be anti-atherogenic. Thus, there is a need to identify regulatory factors involved. Transcription of apoA-I involves peroxisome-proliferator-activated-receptor-alpha (PPARα) activation, but endoplasmic reticulum (ER) -stress and inflammation also influence apoA-I production. To unravel why PPARα agonist GW7647 increased apoA-I production compared to PPARα agonist fenofibric acid (FeAc) in human hepatocellular carcinoma (HepG2) and colorectal adenocarcinoma (CaCo-2) cells, gene expression profiles were compared. Microarray analyses suggested CCAAT/enhancer-binding-protein-beta (C/EBP-ß) involvement in the FeAc condition. Therefore, C/EBP-ß silencing and isoform-specific overexpression experiments were performed under ER-stressed, inflammatory and non-inflammatory conditions. mRNA expression of C/EBP-ß, ATF3, NF-IL3 and GDF15 were upregulated by FeAc compared to GW7647 in both cell lines, while DDIT3 and DDIT4 mRNA were only upregulated in HepG2 cells. This ER-stress related signature was associated with decreased apoA-I secretion. After ER-stress induction by thapsigargin or FeAc addition, intracellular apoA-I concentrations decreased, while ER-stress marker expression (CHOP, XBP1s, C/EBP-ß) increased. Cytokine addition increased intracellular C/EBP-ß levels and lowered apoA-I concentrations. Although a C/EBP binding place is present in the apoA-I promoter, C/EBP-ß silencing or isoform-specific overexpression did not affect apoA-I production in inflammatory, non-inflammatory and ER-stressed conditions. Therefore, C/EBP-ß is not a target to influence hepatic apoA-I production. J. Cell. Biochem. 118: 754-763, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Apolipoproteína A-I/biossíntese , Butiratos/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Fenofibrato/análogos & derivados , PPAR alfa/agonistas , Compostos de Fenilureia/farmacologia , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Proteína beta Intensificadora de Ligação a CCAAT/antagonistas & inibidores , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células CACO-2 , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fenofibrato/farmacologia , Perfilação da Expressão Gênica , Inativação Gênica , Células Hep G2 , Humanos , Inflamação/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tapsigargina/farmacologia
5.
Molecules ; 21(12)2016 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-27973425

RESUMO

Zerumbone ameliorates retinal damage by blocking advanced glycation end products and their receptor system in streptozotocin-diabetic rats. Because of the multiple factors involved in diabetic retinopathy (DR) etiology, the mechanisms of zerumbone that are mainly responsible for its ameliorative effect on DR need to be further clarified. In the present study, zerumbone (20 mg or 40 mg/kg) or fenofibric acid (100 mg/kg) was orally administered to diabetic rats by intragastric gavage once daily for three consecutive months. Zerumbone displayed similar characteristics to fenofibric acid in reducing retinal vascular permeability and leukostasis in diabetic rats. Fundus photographs showed that large retinal vessel diameters were decreased in zerumbone-treated diabetic rats. Zerumbone not only down-regulated the gene expression of retinal angiogenic parameters, but also reduced the expression of inflammatory cytokines and chemokines in the retina of diabetic rats. Moreover, zerumbone reduced the p38 MAPK phosphorylation and abrogated the nuclear translocation of NF-κB p65 in the retina of diabetic rats. In conclusion, treatment of diabetic rats with zerumbone attenuates the severity of retinal inflammation and angiogenesis, via inhibition of p38 MAPK and NF-κB signaling pathways. These benefits of zerumbone for DR appear to be linked to its antihyperglycemic and antihyperlipidemic effects.


Assuntos
Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/prevenção & controle , Microvasos/efeitos dos fármacos , Vasos Retinianos/efeitos dos fármacos , Sesquiterpenos/uso terapêutico , Fator de Transcrição RelA/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Permeabilidade Capilar/efeitos dos fármacos , Fenofibrato/análogos & derivados , Fenofibrato/uso terapêutico , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Masculino , Microvasos/lesões , Ratos , Ratos Wistar , Retina/efeitos dos fármacos , Vasos Retinianos/lesões , Transdução de Sinais/efeitos dos fármacos , Estreptozocina , Fator de Transcrição RelA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Acta Diabetol ; 53(1): 109-18, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25936740

RESUMO

AIMS: There is clinical evidence that fenofibrate, a PPARα agonist, arrests the progression of diabetic macular edema (DME). However, the underlying mechanisms of this beneficial effect remain to be elucidated. We previously reported that fenofibric acid (FA), the active metabolite of fenofibrate, prevents the disorganization of tight junction proteins and the hyperpermeability provoked by the diabetic milieu in the retinal pigment epithelium (RPE). The aim of the present study was to evaluate whether this effect is mediated by inhibiting the proinflammatory transcription factor NF-κB, as well as the expression of several proinflammatory cytokines involved in the pathogenesis of DME. METHODS: Human RPE cells were cultured under standard conditions and under conditions leading to the disruption of the monolayer [IL-1ß (10 ng/ml)]. The effect of FA, QNZ (a NF-κB inhibitor), WY14643 (a PPARα agonist), and MK-866 (a PPARα antagonist) in the disruption of the monolayer was determined by dextran permeability and immunohistochemistry analyses. The effect of FA on NF-κB activity was assessed by EMSA and by NF-κB/p65 nuclear translocation analyses. The expression of cytokines (IL-6, IL-8, MCP-1) was measured by RT-PCR. RESULTS: FA prevented RPE monolayer disruption, and the consequent hyperpermeability induced by IL-1ß, through inhibition of NF-κB activity. This effect was due to PPARα activation and was associated with a significant downregulation of the expression of proinflammatory cytokines. CONCLUSIONS: Our findings suggest that the anti-inflammatory effects of FA through inhibition of NF-κB activity play a key role in the beneficial effect of fenofibrate for treating DME.


Assuntos
Barreira Hematorretiniana/efeitos dos fármacos , Barreira Hematorretiniana/metabolismo , Fenofibrato/farmacologia , Fator de Transcrição RelA/metabolismo , Barreira Hematorretiniana/patologia , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Regulação para Baixo/efeitos dos fármacos , Fenofibrato/análogos & derivados , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Edema Macular/metabolismo , Edema Macular/patologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
J Physiol Pharmacol ; 66(2): 233-47, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25903954

RESUMO

Fenofibrate, a well-known normolipidemic drug, has been shown to exert strong anticancer effects against tumors of neuroectodermal origin including glioblastoma. Although some pharmacokinetic studies were performed in the past, data are still needed about the detailed subcellular and tissue distribution of fenofibrate (FF) and its active metabolite, fenofibric acid (FA), especially in respect to the treatment of intracranial tumors. We used high performance liquid chromatography (HPLC) to elucidate the intracellular, tissue and body fluid distribution of FF and FA after oral administration of the drug to mice bearing intracranial glioblastoma. Following the treatment, FF was quickly cleaved to FA by blood esterases and FA was detected in the blood, urine, liver, kidney, spleen and lungs. We have also detected small amounts of FA in the brains of two out of six mice, but not in the brain tumor tissue. The lack of FF and FA in the intracranial tumors prompted us to develop a new method for intracranial delivery of FF. We have prepared and tested in vitro biodegradable poly-lactic-co-glycolic acid (PLGA) polymer wafers containing FF, which could ultimately be inserted into the brain cavity following resection of the brain tumor. HPLC-based analysis demonstrated a slow and constant diffusion of FF from the wafer, and the released FF abolished clonogenic growth of glioblastoma cells. On the intracellular level, FF and FA were both present in the cytosolic fraction. Surprisingly, we also detected FF, but not FA in the cell membrane fraction. Electron paramagnetic resonance spectroscopy applied to spin-labeled phospholipid model-membranes revealed broadening of lipid phase transitions and decrease of membrane polarity induced by fenofibrate. Our results indicate that the membrane-bound FF could contribute to its exceptional anticancer potential in comparison to other lipid-lowering drugs, and advocate for intracranial delivery of FF in the combined pharmacotherapy against glioblastoma.


Assuntos
Plásticos Biodegradáveis/farmacocinética , Neoplasias Encefálicas/tratamento farmacológico , Encéfalo/metabolismo , Portadores de Fármacos/farmacocinética , Fenofibrato/análogos & derivados , Glioblastoma/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Fenofibrato/farmacocinética , Fenofibrato/farmacologia , Humanos , Ácido Láctico/farmacocinética , Camundongos , Camundongos Nus , Ácido Poliglicólico/farmacocinética , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polímeros/farmacocinética , Distribuição Tecidual
8.
Discov Med ; 18(98): 105-12, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25227751

RESUMO

The earliest and most significant change in diabetic retinopathy (DR) is blood-retinal barrier (BRB) dysfunction, followed by two main pathologies that may cause severe visual impairment: Diabetic Macular Edema (DME) and Proliferative Diabetic Retinopathy (PDR). The pathological hallmarks of BRB dysfunction include loss of tight junction integrity, VEGF- and AGE-induced damage, oxidative stress, and inflammatory changes. Recently, several BRB protective factors have been reported. Our aim is to give a review of those protective factors and discuss new potential therapeutic targets for DR.


Assuntos
Barreira Hematorretiniana/fisiopatologia , Retinopatia Diabética/prevenção & controle , Retinopatia Diabética/fisiopatologia , Animais , Barreira Hematorretiniana/efeitos dos fármacos , Barreira Hematorretiniana/patologia , Retinopatia Diabética/etiologia , Eritropoetina/farmacologia , Fenofibrato/análogos & derivados , Fenofibrato/farmacologia , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Edema Macular/complicações , Edema Macular/patologia , Edema Macular/fisiopatologia , Fatores de Proteção , Pirazinas/farmacologia , Fosfato de Sitagliptina , Triazóis/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
9.
Ophthalmic Epidemiol ; 21(5): 307-17, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25133794

RESUMO

PURPOSE: Fenofibrate reduced progression of diabetic retinopathy in two large randomized studies. The effect of 135 mg fenofibric acid on diabetic macular edema (DME) was evaluated in subjects with existing DME. METHODS: In this double-blind, randomized, placebo-controlled study, 110 subjects with DME not requiring immediate photocoagulation or intraocular treatment with adequate diabetes and blood pressure control received either fenofibric acid or placebo once daily for 1 year. Total macula volume (TMV) and thickness were measured in the worse eye and all eligible eyes with time-domain optical coherence tomography at baseline and quarterly thereafter. RESULTS: TMV decreased by -0.35 mm(3) (within-group difference) after fenofibric acid treatment and by -0.11 mm(3) after placebo. The between-group comparison of the change was -0.25 mm(3) (95% confidence interval, CI, -0.645-0.155; p = 0.227, worse eye analysis). Weighted inner zone thickness and volume decreased by -18.7 µm and -0.13 mm(3), respectively, for within group difference after fenofibric acid and by -3.1 µm and -0.02 mm(3), respectively, after placebo. Considering all eligible eyes, thicknesses at central zone, mean inner zone, and entire retina decreased by -21.3 µm, -19.8 µm, and -20.4 µm, respectively, after fenofibric acid. No between-group difference in changes of these measurements was observed. Triglycerides decreased by 23% after fenofibric acid (vs 4% after placebo, p = 0.001) and high-density lipoprotein cholesterol increased by 8% (vs 0.3%, p = 0.014). No safety concern was identified. CONCLUSION: Subjects treated with fenofibric acid had a modest improvement in TMV, although the study was probably underpowered to detect a benefit over placebo after 1 year.


Assuntos
Anticolesterolemiantes/uso terapêutico , Retinopatia Diabética/tratamento farmacológico , Fenofibrato/análogos & derivados , Edema Macular/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Retinopatia Diabética/sangue , Retinopatia Diabética/diagnóstico , Método Duplo-Cego , Feminino , Fenofibrato/uso terapêutico , Humanos , Macula Lutea/patologia , Edema Macular/sangue , Edema Macular/diagnóstico , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Tomografia de Coerência Óptica , Triglicerídeos/sangue , Acuidade Visual/fisiologia
10.
Invest Ophthalmol Vis Sci ; 55(6): 3820-32, 2014 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-24845641

RESUMO

PURPOSE: The mechanism for the antiangiogenic activity of peroxisome proliferator-activated receptor alpha (PPARα) remains incompletely understood. Endothelial progenitor cells (EPC) are known to participate in neovascularization (NV). The purpose of this study was to investigate whether PPARα regulates EPC during retinal NV. METHODS: Retinal NV was induced by oxygen-induced retinopathy (OIR). Mice with OIR were injected intraperitoneally with the PPARα agonist fenofibric acid (FA) or with adenovirus expressing PPARα (Ad-PPARα). Flow cytometry was used to quantify circulating and retinal EPC. Serum stromal cell-derived factor 1 (SDF-1) levels were measured by ELISA. Hypoxia was induced in primary human retinal capillary endothelial cells (HRCEC) and mouse brain endothelial cells (MBEC) by CoCl2. Levels of SDF-1 and hypoxia-inducible factor 1 alpha (HIF-1α) were measured by Western blotting. RESULTS: Fenofibric acid and overexpression of PPARα attenuated the increase of circulating and retinal EPC, correlating with suppressed retinal NV in OIR mice at P17. The PPARα knockout enhanced the OIR-induced increase of circulating and retinal EPC. Fenofibric acid decreased retinal HIF-1α and SDF-1 levels as well as serum SDF-1 levels in the OIR model. In HRCEC, PPARα inhibited HIF-1α nuclear translocation and SDF-1 overexpression induced by hypoxia. Further, MBEC from PPARα(-/-) mice showed more prominent activation of HIF-1α and overexpression of SDF-1 induced by hypoxia, compared with the wild-type (WT) MBEC. PPARα failed to block SDF-1 overexpression induced by a constitutively active mutant of HIF-1α, suggesting that regulation of SDF-1 by PPARα was through blockade of HIF-1α activation. CONCLUSIONS: Peroxisome proliferator-activated receptor alpha suppresses ischemia-induced EPC mobilization and homing through inhibition of the HIF-1α/SDF-1 pathway. This represents a novel molecular mechanism for PPARα's antiangiogenic effects.


Assuntos
Quimiocina CXCL12/metabolismo , Endotélio Vascular/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , PPAR alfa/fisiologia , Células-Tronco/metabolismo , Adenoviridae/genética , Animais , Western Blotting , Células Cultivadas , Circulação Cerebrovascular , Modelos Animais de Doenças , Fenofibrato/análogos & derivados , Fenofibrato/farmacologia , Citometria de Fluxo , Angiofluoresceinografia , Expressão Gênica/fisiologia , Humanos , Hipolipemiantes/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/toxicidade , PPAR alfa/agonistas , Reação em Cadeia da Polimerase em Tempo Real , Neovascularização Retiniana/patologia , Neovascularização Retiniana/prevenção & controle , Vasos Retinianos/citologia
11.
Int J Pharm ; 445(1-2): 117-24, 2013 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-23384729

RESUMO

Based on the enhanced permeability and retention (EPR) effect, nanoparticles are believed to accumulate in tumors. In this conjunction, the stability of drug encapsulation is assumed to be sufficient. For clarification purposes, PEGylated poly-(D,L-lactic acid) (PEG-PDLLA) micelles which incorporated the hydrophobic model drug dechloro-4-iodo-fenofibrate (IFF) were investigated. H2N-PEG-PDLLA was synthesized, coupled to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and labeled with 111-indium. From this polymeric species, mixed micelles with H3CO-PEG-PDLLA were prepared which encapsulated the 125-iodine or 131-iodine labeled drug IFF. Bioimaging and biodistribution experiments in healthy and AR42J-tumor bearing mice were carried out to quantify the uptake of the drug and its carrier in single organs. As a result, upon injection of this system, a rapid dissociation of the polymeric carrier and the incorporated drug (<10 min post inj.) was revealed. Regardless of the premature release, the drug showed an enhanced tumor accumulation compared to the polymeric carrier. In conclusion, the self-assembling system allowed for successful solubilization of the hydrophobic drug by physical incorporation into micelles whereas the tumor targeting properties of the drug delivery system could not be sufficiently shown.


Assuntos
Portadores de Fármacos/química , Fenofibrato/farmacocinética , Neoplasias/metabolismo , Polietilenoglicóis/química , Animais , Linhagem Celular Tumoral , Feminino , Fenofibrato/administração & dosagem , Fenofibrato/análogos & derivados , Fenofibrato/química , Camundongos , Micelas , Neoplasias/tratamento farmacológico , Ratos , Distribuição Tecidual
12.
ChemMedChem ; 7(6): 1101-11, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22489042

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors that control the expression of genes involved in a variety of physiologic processes, through heterodimerization with retinoid X receptor and complex formation with various cofactors. Drugs or treatment regimens that combine the beneficial effects of PPARα and γ agonism present an attractive therapeutic strategy to reduce cardiovascular risk factors. Aleglitazar is a dual PPARα/γ agonist currently in phase III clinical development for the treatment of patients with type 2 diabetes mellitus who recently experienced an acute coronary event. The potency and efficacy of aleglitazar was evaluated in a head-to-head comparison with other PPARα, γ and δ ligands. A comprehensive, 12-concentration dose-response analysis using a cell-based assay showed aleglitazar to be highly potent, with EC(50) values of 5 nM and 9 nM for PPARα and PPARγ, respectively. Cofactor recruitment profiles confirmed that aleglitazar is a potent and balanced activator of PPARα and γ. The efficacy and potency of aleglitazar are discussed in relation to other dual PPARα/γ agonists, in context with the published X-ray crystal structures of both PPARα and γ.


Assuntos
Oxazóis/química , PPAR alfa/agonistas , PPAR gama/agonistas , Tiofenos/química , Sequência de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Fenofibrato/análogos & derivados , Fenofibrato/química , Ligantes , Dados de Sequência Molecular , Oxazóis/farmacologia , PPAR alfa/metabolismo , PPAR gama/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Pioglitazona , Tiazolidinedionas/química , Tiofenos/farmacologia , Transcrição Gênica/efeitos dos fármacos
13.
Invest Ophthalmol Vis Sci ; 52(9): 6348-54, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21715349

RESUMO

PURPOSE: To determine whether fenofibric acid (FA) reduces high glucose (HG)-induced basement membrane component overexpression and hyperpermeability in human retinal pigment epithelial (RPE) cells. METHODS: Retinal pigment epithelial cells (ARPE-19) were cultured for 18 days in normal glucose (5 mM) or HG (25 mM) medium and studied for the effects of FA on fibronectin (FN) and collagen IV (Coll IV) expression. During last 3 days of the experiment, 100 µM FA was added to cells grown in HG medium or in HG medium plus IL-1ß (HG + IL-1ß) to mimic, at least in part, the inflammatory aspect of the diabetic milieu. Real-time RT-PCR was performed to determine FN and Coll IV mRNA levels, whereas protein levels were assessed by Western blot analyses. Cell monolayer morphology and barrier function were analyzed by confocal microscopy using specific antibodies against tight junction proteins, ZO-1, and claudin-1 and by measuring apical-basolateral movements of FITC-dextran, respectively. RESULTS: FN and Coll IV expression were significantly increased in RPE cells grown in HG or HG + IL-1ß medium compared with cells grown in normal medium. When cells grown in HG or HG + IL-1ß medium were treated with FA, significant reductions in FN and Coll IV expression were observed. In addition, exposure to FA decreased excess permeability in a dose-dependent manner in cells grown in HG + IL-1ß medium. This effect was unrelated to changes in tight junction protein content. CONCLUSIONS: Findings from this study suggest that the downregulation of basement membrane components by FA may have a protective effect against outer blood-retinal barrier leakage associated with diabetic retinopathy.


Assuntos
Anticolesterolemiantes/farmacologia , Colágeno Tipo IV/metabolismo , Complicações do Diabetes , Fenofibrato/análogos & derivados , Fibronectinas/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Western Blotting , Permeabilidade Capilar/fisiologia , Células Cultivadas , Claudina-1 , Colágeno Tipo IV/genética , Dextranos/metabolismo , Relação Dose-Resposta a Droga , Fenofibrato/farmacologia , Fibronectinas/genética , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Glucose/toxicidade , Humanos , Interleucina-1beta/toxicidade , Proteínas de Membrana/metabolismo , Microscopia Confocal , Fosfoproteínas/metabolismo , RNA Mensageiro/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , ATPase Trocadora de Sódio-Potássio/metabolismo , Regulação para Cima , Proteína da Zônula de Oclusão-1
14.
Diabetologia ; 54(6): 1543-53, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21369818

RESUMO

AIMS/HYPOTHESIS: The mechanisms involved in the beneficial effects of fenofibrate on the development and progression of diabetic macular oedema (DMO) remain to be elucidated. To shed light on this issue we have explored the effect of fenofibric acid on the barrier function of human retinal pigment epithelium (RPE) cells. METHODS: ARPE-19 cells (a human RPE line) were cultured for 18 days under standard conditions and under conditions leading to the disruption of the monolayer (D-glucose, 25 mmol/l, with IL-1ß, 10 ng/ml, added at days 16 and 17). Fenofibric acid, 25 µmol/l and 100 µmol/l, was added on the last 3 days of the experiment (one application/day). RPE cell permeability was evaluated by measuring apical-basolateral movements of FITC-dextran (40 kDa). The production of tight junction proteins and AMP-activated protein kinase (AMPK) phosphorylation was assessed by western blot. Immunohistochemical studies of tight junction proteins and small interfering RNA transfection to AMPK were also performed in ARPE-19 monolayers. RESULTS: Treatment of ARPE-19 cells with fenofibric acid significantly reduced the increment of permeability and the breakdown of the ARPE-19 cell monolayer induced by D-glucose, 25 mmol/l, and IL-1ß, 10 ng/ml, in a dose-dependent manner. This effect was unrelated to changes in the content of tight junction proteins. Fenofibric acid prevented the activation of AMPK induced by IL-1ß and the hyperpermeability induced by IL-1ß was blocked by silencing AMPK. CONCLUSIONS/INTERPRETATION: Disruption of RPE induced by IL-1ß is prevented by fenofibric acid through its ability to suppress AMPK activation. This mechanism could be involved in the beneficial effects of fenofibrate on DMO development.


Assuntos
Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Fenofibrato/análogos & derivados , Interleucina-1beta/metabolismo , Edema Macular/prevenção & controle , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Idoso , Transporte Biológico/fisiologia , Barreira Hematorretiniana/efeitos dos fármacos , Barreira Hematorretiniana/metabolismo , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/fisiologia , Dextranos/metabolismo , Retinopatia Diabética/metabolismo , Retinopatia Diabética/prevenção & controle , Relação Dose-Resposta a Droga , Fenofibrato/farmacologia , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Glucose/farmacologia , Humanos , Hipolipemiantes/farmacologia , Edema Macular/metabolismo , Epitélio Pigmentado da Retina/patologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
17.
Drug Metab Dispos ; 37(11): 2236-43, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19661212

RESUMO

Fenofibric acid (FA), the active moiety of fenofibrate, is an agonist of the peroxisome proliferator-activated nuclear receptor alpha that modulates triglyceride and cholesterol profiles. Lipid response to fenofibrate and FA serum concentrations is highly variable. Although FA is reported to be almost exclusively inactivated by UDP-glucuronosyltransferases (UGTs) into FA-glucuronide (FA-G), the contribution of UGT isoenzymes has never been systematically assessed. Heterologously expressed human UGT1A and UGT2B and their coding variants were tested for FA glucuronidation using liquid chromatography/mass spectrometry. Recombinant UGT2B7 presented the highest V(max)/K(m) value (2.10 microl/min/mg), 16-fold higher than the activity of other reactive UGTs, namely, UGT1A3, UGT1A6, and UGT1A9 (0.13, 0.09, and 0.02 microl/min/mg, respectively). UGT2B7.1 (His(268)) and UGT2B7.2 (Tyr(268)) enzyme activity was similar, whereas UGT1A3.2 (R(11)A(47)), UGT1A3.3 (Trp(11)), and UGT1A9.3 (Thr(33)) showed 61 to 96% reduced V(max)/K(m) values compared with the respective (1) reference proteins. FA-G formation by a human liver bank (n = 48) varied by 10-fold, but the rate of formation was not associated with common genetic variations in UGT1A3, UGT1A6, UGT1A9, and UGT2B7. Correlation with activities for the probe substrates zidovudine (UGT2B7; r(2) = 0.75), mycophenolic acid (UGT1A9; r(2) = 0.42), fulvestrant (UGT1A3; r(2) = 0.36), but not serotonin (UGT1A6; r(2) = 0.06) indicated a primary role for UGT2B7 and lesser roles of UGT1A9 and UGT1A3 in hepatic FA glucuronidation. This was confirmed by a strong correlation of FA-G formation with UGT2B7 protein content and inhibition by fluconazole, a known UGT2B7 selective inhibitor. Additional studies are required to identify genetic factors contributing to the observed FA glucuronidation variability.


Assuntos
Fenofibrato/análogos & derivados , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Microssomos Hepáticos/metabolismo , Linhagem Celular , Fenofibrato/metabolismo , Glucuronídeos/química , Glucuronosiltransferase/química , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Microssomos Hepáticos/enzimologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
18.
Pharmacol Rep ; 61(6): 1134-45, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20081249

RESUMO

Because of methodological limitations and conflicting results of studies conducted thus far, the possible involvement of human adipose tissue in pleiotropic effects of statins and fibrates requires better understanding. Samples of visceral and subcutaneous adipose tissue obtained from 23 mixed dyslipidemic patients and 23 normolipidemic subjects were treated in vitro for 48 h with atorvastatin, fenofibric acid or both these agents. Visceral and subcutaneous fat of mixed dyslipidemic patients released more leptin, resistin, interleukin-6, tumor necrosis factor alpha (TNFalpha and plasminogen activator inhibitor-1 (PAI-1), and less adiponectin than respective adipose tissue of patients without lipid abnormalities. In both groups of patients, visceral and subcutaneous tissue varied in the amount of secreted adipokines. In dyslipidemic patients both drugs administered alone affected adipose tissue adiponectin and resistin secretion. Additionally, atorvastatin decreased PAI-1 while fenofibric acid reduced leptin release. A combined administration of atorvastatin and fenofibric acid changed the release of all studied markers by visceral fat but did not affect interleukin-6 and TNFalpha release by subcutaneous tissue. In normolipidemic subjects the effect on adipokine release was more pronounced in visceral fat, in which it was strongest if the drugs were given together. Adipose tissue hormonal activity differs between mixed dyslipidemic and normolipidemic patients and between visceral and subcutaneous adipose tissue. Atorvastatin and fenofibrate exhibit their pleiotropic effects in part by changing the adipokine release by human adipose tissue, regardless of its origin. These effects are stronger in patients with mixed dyslipidemia and are particularly pronounced if atorvastatin and fenofibric acid are given together.


Assuntos
Adipocinas/metabolismo , Anticolesterolemiantes/farmacologia , Fenofibrato/análogos & derivados , Ácidos Heptanoicos/farmacologia , Pirróis/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Adulto , Atorvastatina , Estudos de Casos e Controles , Sinergismo Farmacológico , Feminino , Fenofibrato/farmacologia , Humanos , Técnicas In Vitro , Gordura Intra-Abdominal/metabolismo , Masculino , Pessoa de Meia-Idade , Gordura Subcutânea/metabolismo
19.
J Biol Chem ; 283(15): 9666-73, 2008 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-18245819

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is associated with familial autosomal dominant hypercholesterolemia and is a natural inhibitor of the LDL receptor (LDLr). PCSK9 is degraded by other proprotein convertases: PC5/6A and furin. Both PCSK9 and the LDLr are up-regulated by the hypocholesterolemic statins. Thus, inhibitors or repressors of PCSK9 should amplify their beneficial effects. In the present study, we showed that PPARalpha activation counteracts PCSK9 induction by statins by repressing PCSK9 promoter activity and by increasing PC5/6A and furin expression. Quantification of mRNA and protein levels showed that various fibrates decreased PCSK9 and increased PC5/6A and furin expression. Fenofibric acid (FA) reduced PCSK9 protein content in immortalized human hepatocytes (IHH) as well as its cellular secretion. FA suppressed PCSK9 induction by statins or by the liver X receptor agonist TO901317. PCSK9 repression is occurring at the promoter level. We showed that PC5/6A and furin fibrate-mediated up-regulation is PPARalpha-dependent. As a functional test, we observed that FA increased by 30% the effect of pravastatin on the LDLr activity in vitro. In conclusion, fibrates simultaneously decreased PCSK9 expression while increasing PC5/6A and furin expression, indicating a broad action of PPARalpha activation in proprotein convertase-mediated lipid homeostasis. Moreover, this study validates the functional relevance of a combined therapy associating PCSK9 repressors and statins.


Assuntos
Anticolesterolemiantes/farmacologia , Ácido Clofíbrico/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hepatócitos/enzimologia , PPAR alfa/metabolismo , Serina Endopeptidases/biossíntese , Anticolesterolemiantes/uso terapêutico , Linhagem Celular Tumoral , Ácido Clofíbrico/uso terapêutico , Proteínas de Ligação a DNA/metabolismo , Fenofibrato/análogos & derivados , Fenofibrato/farmacologia , Furina/biossíntese , Furina/genética , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Hidrocarbonetos Fluorados , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/enzimologia , Hiperlipoproteinemia Tipo II/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Receptores X do Fígado , Receptores Nucleares Órfãos , PPAR alfa/genética , Pravastatina/farmacologia , Pravastatina/uso terapêutico , Regiões Promotoras Genéticas/genética , Pró-Proteína Convertase 5/biossíntese , Pró-Proteína Convertase 5/genética , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Serina Endopeptidases/genética , Sulfonamidas/farmacologia , Regulação para Cima/efeitos dos fármacos
20.
Gastroenterology ; 128(2): 334-42, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15685545

RESUMO

BACKGROUND AND AIMS: Liver inflammation, fibrosis, and dyslipidemia are common features in patients with chronic hepatitis C virus (HCV) infection. Because peroxisome proliferator-activated receptor alpha (PPARalpha) is highly expressed in the liver and is involved in the regulation of lipid metabolism and inflammation, we sought to determine whether HCV infection may locally impair PPARalpha expression and activity. METHODS: PPARalpha expression was investigated in liver biopsy specimens of 86 untreated patients with HCV infection and controls, by using real-time polymerase chain reaction (PCR), Western blot analysis, and immunohistochemistry. PPARalpha activity was assessed by quantification of the key gene target carnitine palmitoyl acyl-CoA transferase 1 (CPT1A) messenger RNA (mRNA). The influence of HCV core protein on PPARalpha mRNA expression was analyzed in vitro by real-time PCR in HCV core-expressing HepG2 cells activated with the PPARalpha ligand fenofibric acid. RESULTS: Hepatic concentrations of PPARalpha and CPT1A expressed by hepatocytes were impaired profoundly in the livers of untreated patients with HCV infection compared with controls. A mean decrease of 85% in PPARalpha mRNA expression paralleled with a lack of CPT1A mRNA induction also were observed in HCV core-expressing HepG2 cells compared with controls. CONCLUSIONS: HCV infection is related to altered expression and function of the anti-inflammatory nuclear receptor PPARalpha. These results identify hepatic PPARalpha as one mechanism underlying the pathogenesis of HCV infection, and as a new therapeutic target in traditional treatment of HCV-induced liver injury.


Assuntos
Fenofibrato/análogos & derivados , Regulação da Expressão Gênica/fisiologia , Hepatite C Crônica/patologia , PPAR alfa/genética , Biópsia , Carcinoma Hepatocelular , Linhagem Celular , Primers do DNA , Fenofibrato/farmacologia , Humanos , Imuno-Histoquímica , Fígado/patologia , Neoplasias Hepáticas , Transcrição Gênica/efeitos dos fármacos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA