Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Br J Clin Pharmacol ; 80(6): 1350-61, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26261033

RESUMEN

AIMS: LDL-receptor expression is inhibited by the protease proprotein convertase subtilisin/kexin type 9 (PCSK9), which is considered a pharmacological target to reduce LDL-C concentrations in hypercholesterolaemic patients. We performed a first-in-human trial with SPC5001, a locked nucleic acid antisense inhibitor of PCSK9. METHODS: In this randomized, placebo-controlled trial, 24 healthy volunteers received three weekly subcutaneous administrations of SPC5001 (0.5, 1.5 or 5 mg kg(-1)) or placebo (SPC5001 : placebo ratio 6 : 2). End points were safety/tolerability, pharmacokinetics and efficacy of SPC5001. RESULTS: SPC5001 plasma exposure (AUC(0,24 h)) increased more than dose-proportionally. At 5 mg kg(-1), SPC5001 decreased target protein PCSK9 (day 15 to day 35: -49% vs. placebo, P < 0.0001), resulting in a reduction in LDL-C concentrations (maximal estimated difference at day 28 compared with placebo -0.72 mmol l(-1), 95% confidence interval - 1.24, -0.16 mmol l(-1); P < 0.01). SPC5001 treatment (5 mg kg(-1)) also decreased ApoB (P = 0.04) and increased ApoA1 (P = 0.05). SPC5001 administration dose-dependently induced mild to moderate injection site reactions in 44% of the subjects, and transient increases in serum creatinine of ≥20 µmol l(-1) (15%) over baseline with signs of renal tubular toxicity in four out of six subjects at the highest dose level. One subject developed biopsy-proven acute tubular necrosis. CONCLUSIONS: SPC5001 treatment dose-dependently inhibited PCSK9 and decreased LDL-C concentrations, demonstrating human proof-of-pharmacology. However, SPC5001 caused mild to moderate injection site reactions and renal tubular toxicity, and clinical development of SPC5001 was terminated. Our findings underline the need for better understanding of the molecular mechanisms behind the side effects of compounds such as SPC5001, and for sensitive and relevant renal toxicity monitoring in future oligonucleotide studies.


Asunto(s)
Oligonucleótidos Antisentido/farmacología , Proproteína Convertasas/antagonistas & inhibidores , Adulto , Anciano , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Riñón/efectos de los fármacos , Masculino , Persona de Mediana Edad , Oligonucleótidos Antisentido/efectos adversos , Proproteína Convertasa 9 , Serina Endopeptidasas
2.
Sci Transl Med ; 5(212): 212ra162, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24259050

RESUMEN

MicroRNAs (miRNAs) regulate many aspects of human biology. They target mRNAs for translational repression or degradation through base pairing with 3' untranslated regions, primarily via seed sequences (nucleotides 2 to 8 in the mature miRNA sequence). A number of individual miRNAs and miRNA families share seed sequences and targets, but differ in the sequences outside of the seed. miRNAs have been implicated in the etiology of a wide variety of human diseases and therefore represent promising therapeutic targets. However, potential redundancy of different miRNAs sharing the same seed sequence and the challenge of simultaneously targeting miRNAs that differ significantly in nonseed sequences complicate therapeutic targeting approaches. We recently demonstrated effective inhibition of entire miRNA families using seed-targeting 8-mer locked nucleic acid (LNA)-modified antimiRs in short-term experiments in mammalian cells and in mice. However, the long-term efficacy and safety of this approach in higher organisms, such as humans and nonhuman primates, have not been determined. We show that pharmacological inhibition of the miR-33 family, key regulators of cholesterol/lipid homeostasis, by a subcutaneously delivered 8-mer LNA-modified antimiR in obese and insulin-resistant nonhuman primates results in derepression of miR-33 targets, such as ABCA1, increases circulating high-density lipoprotein cholesterol, and is well tolerated over 108 days of treatment. These findings demonstrate the efficacy and safety of an 8-mer LNA-antimiR against an miRNA family in a nonhuman primate metabolic disease model, suggesting that this could be a feasible approach for therapeutic targeting of miRNA families sharing the same seed sequence in human diseases.


Asunto(s)
Silenciador del Gen , MicroARNs/antagonistas & inhibidores , Animales , HDL-Colesterol/sangre , Femenino , Células Hep G2 , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Primates
3.
Cardiovasc Pathol ; 20(1): 36-43, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-19919900

RESUMEN

INTRODUCTION: Transcription factor activator protein-1 regulates genes involved in inflammation and repair. The aim of this study was to determine whether transcription factor activator protein-1 activity in carotid plaques is related to symptoms, lipid accumulation, or extracellular matrix composition. METHODS: Twenty-eight atherosclerotic carotid plaques were removed by endarterectomy and divided into two groups based on the presence or absence of ipsilateral symptoms (<1 month ago). Activator protein-1 DNA binding activity was assessed, and subunit (c-Jun, JunD, JunB, c-Fos, FosB, Fra-1, Fra-2) protein levels analyzed by immunoblotting. Distribution of c-Jun in plaques was analyzed by immunohistochemistry. RESULTS: Plaques associated with symptoms had increased activator protein-1 activity and increased expression of c-Jun and JunD, as compared to asymptomatic plaques. Fra-1 and Fra-2 were present in equal amounts in both groups, whereas JunB, FosB, and c-Fos were undetectable. Activator protein-1 activity correlated with cholesteryl ester and elastin in plaques and decreased with age. Activator protein-1 activity did not correlate with collagen, calcified tissue, or proteoglycan content. CONCLUSIONS: Activator protein-1 is increased in plaques associated with symptoms. The correlation between activator protein-1 and cholesteryl esters suggests that high activator protein-1 is a marker of plaque vulnerability. Activator protein-1 expression can also reflect the activation of repair processes.


Asunto(s)
Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Ésteres del Colesterol/metabolismo , Factor de Transcripción AP-1/metabolismo , Anciano , Estenosis Carotídea/cirugía , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/metabolismo , Endarterectomía Carotidea , Femenino , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Subunidades de Proteína , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción AP-1/química
4.
Nucleic Acids Res ; 38(20): 7100-11, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20615897

RESUMEN

The potency and specificity of locked nucleic acid (LNA) antisense oligonucleotides was investigated as a function of length and affinity. The oligonucleotides were designed to target apolipoprotein B (apoB) and were investigated both in vitro and in vivo. The high affinity of LNA enabled the design of short antisense oligonucleotides (12- to 13-mers) that possessed high affinity and increased potency both in vitro and in vivo compared to longer oligonucleotides. The short LNA oligonucleotides were more target specific, and they exhibited the same biodistribution and tissue half-life as longer oligonucleotides. Pharmacology studies in both mice and non-human primates were conducted with a 13-mer LNA oligonucleotide against apoB, and the data showed that repeated dosing of the 13-mer at 1-2 mg/kg/week was sufficient to provide a significant and long lasting lowering of non-high-density lipoprotein (non-HDL) cholesterol without increasing serum liver toxicity markers. The data presented here show that oligonucleotide length as a parameter needs to be considered in the design of antisense oligonucleotide and that potent short oligonucleotides with sufficient target affinity can be generated using the LNA chemistry. Conclusively, we present a 13-mer LNA oligonucleotide with therapeutic potential that produce beneficial cholesterol lowering effect in non-human primates.


Asunto(s)
Apolipoproteínas B/metabolismo , Colesterol/sangre , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos/farmacología , Animales , Apolipoproteínas B/genética , Autorradiografía , Disparidad de Par Base , Línea Celular Tumoral , Femenino , Humanos , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Oligonucleótidos/química , Oligonucleótidos/farmacocinética , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/farmacocinética , ARN Mensajero/metabolismo
5.
BMC Immunol ; 9: 70, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19032770

RESUMEN

BACKGROUND: Pro-inflammatory cytokines can affect intracellular lipid metabolism. A variety of effects have been described for different cell types; hepatocyte lipid turnover pathways are inhibited during inflammation, whereas interleukin-1beta (IL-1beta) reduces intracellular cholesterol levels in fibroblasts. Levels of the pro-inflammatory cytokines IL-1beta and tumour necrosis factor-alpha (TNF-alpha) are up-regulated at sites of formation of atherosclerotic plaques. Plaque formation is though to begin with infiltration of monocytes to the intimal layer of the vascular wall, followed by differentiation to macrophages and macrophage uptake of modified lipoproteins, resulting in accumulation of intracellular lipids. The lipid-filled cells are referred to as macrophage foam cells, a key feature of atherosclerotic plaques. We have investigated the effects of IL-1beta and TNF-alpha on macrophage foam cells in order to assess whether presence of the pro-inflammatory cytokines improves or aggravates macrophage foam cell formation by affecting lipid accumulation and lipid turn-over in the cells. RESULTS: Differentiated primary human macrophages or THP-1 cells were lipid loaded by uptake of aggregated low density lipoproteins (AgLDL) or very low density lipoproteins (VLDL), and then incubated with IL-1beta (0 - 5000 pg/ml) in lipoprotein-free media for 24 h. Cells incubated in absence of cytokine utilized accumulated neutral lipids, in particular triglycerides. Addition of exogenous IL-1beta resulted in a dose-dependent retention of intracellular cholesterol and triglycerides. Exchanging IL-1beta with TNF-alpha gave a similar response. Analysis of fatty acid efflux and intracellular fatty acid activation revealed a pattern of decreased lipid utilization in cytokine-stimulated cells. CONCLUSION: IL-1beta and TNF-alpha enhance macrophage foam cell formation, in part by inhibition of macrophage intracellular lipid catabolism. If present in vivo, these mechanisms will further augment the pro-atherogenic properties of the two cytokines.


Asunto(s)
Células Espumosas/efectos de los fármacos , Interleucina-1beta/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Células Espumosas/citología , Células Espumosas/metabolismo , Humanos , Lipoproteínas VLDL/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Factores de Tiempo , Triglicéridos/metabolismo
6.
Biochem Biophys Res Commun ; 363(4): 1020-6, 2007 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-17927964

RESUMEN

Lipid-filled macrophages (foam cells) are a defining feature of atherosclerotic plaques. Foam cells contain lipid droplet-associated proteins that in other cell types regulate lipid turnover. In foam cell such proteins may directly affect lipid droplet formation and lipid efflux. Differentiated primary human monocytes or THP-1 cells were lipid loaded by incubation with aggregated low density lipoproteins (AgLDL) or VLDL resulting in macrophage foam cells with predominantly cholesterol ester or triglyceride-rich lipid droplets, respectively. Lipid droplets were isolated and major proteins identified by mass spectrometry, among them the apolipoprotein B-48 receptor that has not previously been recognized in this context. Expression of two proteins, perilipin and adipophilin, was quantified by Western blots of cell lysates. Perilipin content decreased and adipophilin increased with lipoprotein lipid loading regardless of intracellular neutral lipid composition. This protein expression pattern may hinder lipid turnover in macrophage foam cells, thereby increasing lipid content of atherosclerotic plaques.


Asunto(s)
Metabolismo de los Lípidos , Macrófagos/metabolismo , Péptidos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Portadoras , Células Cultivadas , Humanos , Lipoproteínas LDL/metabolismo , Espectrometría de Masas , Proteínas de la Membrana , Perilipina-1 , Perilipina-2
7.
Autoimmunity ; 40(2): 122-30, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17453711

RESUMEN

BACKGROUND: Oxidation of LDL is associated with generation of autoantibodies against a large number of different aldehyde-modified peptide sequences in apo B-100. Autoantibodies recognizing peptide sequences in the LDL receptor-binding region of apo B-100 could potentially affect both cholesterol metabolism and atherosclerosis. The aim of the present study was to determine physiological effects of induction of immune responses against the apo B-100 LDL receptor-binding site in mice deficient for the LDL receptor. METHODS AND RESULTS: Mice received three immunizations, beginning at 6 weeks of age, with aldehyde-modified or non-modified peptides corresponding to the amino acid sequence of the LDL receptor-binding site. Analysis of antibody response by ELISA unexpectedly revealed high titers of pre-existing IgG against both native and aldehyde-modified binding site sequences in non-immunized mice. Immunization with aldehyde-modified binding site sequences resulted in an almost complete down-regulation of this autoimmune response. It was also associated with a rapid increase in lipid-rich plaques in the aorta and a substantial depletion of the lipid content of the liver, whereas plasma lipid and apo B values were similar in all groups. CONCLUSIONS: These observations demonstrate existence of an endogenous T cell-dependent autoimmune response against the LDL receptor-binding site in LDL receptor(-/-) mice and suggest that this may help to prevent accumulation of lipoprotein lipids in the artery wall, whereas immunization with the corresponding aldehyde modified sequence down-regulates this response and induces substantial atherosclerotic development.


Asunto(s)
Aorta Torácica/metabolismo , Apolipoproteína B-100/metabolismo , Autoinmunidad , Metabolismo de los Lípidos , Lípidos/sangre , Receptores de LDL/inmunología , Aldehídos/química , Secuencia de Aminoácidos , Animales , Aorta Torácica/patología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Sitios de Unión/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Receptores de LDL/genética , Receptores de LDL/metabolismo , Proteína Amiloide A Sérica/metabolismo
8.
Vascul Pharmacol ; 46(2): 91-6, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16942919

RESUMEN

Statin treatment inhibits oxidized lipoprotein-induced intracellular lipid accumulation (foam cell formation) and reduces plasma levels of inflammatory markers such as interleukin-1beta (IL-1beta). The aim of the present study was to determine if simvastatin affected lipid accumulation in macrophages incubated with aggregated low density lipoproteins (AgLDL) and whether simvastatin had a direct effect on cytokine secretion from macrophages. Simvastatin treatment did not inhibit AgLDL-induced macrophage lipid accumulation, but significantly increased the secretion of IL-1beta and IL-8 from macrophages, whilst inhibiting the secretion of tumor necrosis factor-alpha (TNF-alpha) and having no significant effect on IL-6 secretion. Increased macrophage lipid content did not block statin-induced IL-1beta and IL-8 secretion. Simvastatin-stimulated IL-1beta secretion from macrophages was inhibited by isoprenoids. We therefore hypothesized that simvastatin stimulated IL-1beta secretion by affecting isoprenylation-dependent signaling pathways. Another possible mechanism for affecting such signaling is to impair isoprenoid transfer protein activity with specific inhibitors such as GGTI-297 and FTInhI. This treatment resulted in strong stimulation of IL-1beta secretion that was further enhanced when exogenous IL-1beta was present at the beginning of treatment. These data suggest an isoprenylation-dependent negative-feedback loop for macrophage IL-1beta secretion that is inhibited by statin treatment.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Interleucina-1beta/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Prenilación de Proteína/efectos de los fármacos , Simvastatina/farmacología , Benzamidas/farmacología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/metabolismo , Retroalimentación Fisiológica/efectos de los fármacos , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Humanos , Interleucina-1beta/farmacología , Interleucina-8/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Lipids Health Dis ; 5: 17, 2006 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-16800873

RESUMEN

BACKGROUND: Macrophage foam cell formation is a prominent feature of human atherosclerotic plaques, usually considered to be correlated to uptake of and inflammatory response to oxidized low density lipoproteins (OxLDL). However, there are alternative pathways for formation of macrophage foam cells and the effect of such lipid loading on macrophage function remains to be fully characterized. In the present study we investigated basal and inducible cytokine expression in primary human macrophages either loaded with triglycerides through incubation with very low density lipoproteins (VLDL) or with cholesterol through incubation with aggregated LDL (AgLDL). We then analyzed how foam cell lipid content affected secretion of three pro-inflammatory cytokines: interleukin-1beta (IL-1beta), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha), and of one chemokine: interleukin-8 (IL-8), all of which are considered pro-inflammatory, pro-atherosclerotic, and are expressed by cells in atherosclerotic tissue. RESULTS: Formation of triglyceride-loaded foam cells resulted in a four-fold increase in basal IL-1beta secretion, whereas cholesterol loading lacked significant effect on IL-1beta secretion. In contrast, secretion of TNF-alpha and IL-6 decreased significantly following both cholesterol and triglyceride loading, with a similar trend for secretion of IL-8. Lipid loading did not affect cell viability or expression of caspase-3, and did not significantly affect macrophage ability to respond to stimulation with exogenous TNF-alpha. CONCLUSION: Lipid loading of primary human macrophages resulted in altered cytokine secretion from cells, where effects were similar regardless of neutral lipid composition of cells. The exception was IL-1beta, where triglyceride, but not cholesterol, lipid loading resulted in a stimulation of basal secretion of the cytokine. It is apparent that macrophage cytokine secretion is affected by lipid loading by lipoproteins other than OxLDL. As both VLDL and AgLDL have been found in the vessel wall, macrophage cytokine response to uptake of these lipoproteins may have a direct effect on atherosclerotic development in vivo. However, macrophage neutral lipid amount and composition did not affect cellular activation by exogenous TNF-alpha, making it likely that lipoprotein lipid loading can affect foam cell cytokine secretion during basal conditions but that the effects can be overruled by TNF-alpha during acute inflammation.


Asunto(s)
Citocinas/metabolismo , Metabolismo de los Lípidos , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Células Cultivadas , Colesterol/metabolismo , Células Espumosas/metabolismo , Humanos , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Macrófagos/citología , Monocitos/citología , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Biochem Biophys Res Commun ; 335(2): 603-8, 2005 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-16087165

RESUMEN

Elevated plasma level of very low-density lipoprotein (VLDL) is a risk factor for coronary heart disease. We investigated the effect of VLDL on expression of the pro-inflammatory cytokine interleukin-1beta (IL-1beta) in human peripheral blood monocyte-derived macrophages. IL-1beta mRNA and protein expression was analysed by PCR and ELISA, respectively. Caspase activation was assessed by immunoblotting. Apart from potentiating lipopolysaccharide-induced secretion of IL-1beta, VLDL alone induced secretion of IL-1beta from human monocyte-derived macrophages. This effect was suppressed by an inhibitor of caspase-1, the protease which cleaves pro-IL-1beta. VLDL treatment activated caspase-1, as indicated by increased levels of the caspase-1 p20 subunit. Furthermore, VLDL increased IL-1beta mRNA expression, which was associated with activation of transcription factor AP-1. Inhibition of caspase-1 did not influence IL-1beta mRNA expression. In conclusion, VLDL induces IL-1beta mRNA expression, caspase-1 activation, and IL-1beta release from macrophages, suggesting that VLDL can promote inflammation in atherosclerotic lesions.


Asunto(s)
Interleucina-1/biosíntesis , Lipoproteínas VLDL/fisiología , Macrófagos/metabolismo , Arteriosclerosis/metabolismo , Caspasa 1/metabolismo , Caspasas/metabolismo , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , ADN/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inflamación , Interleucina-1/metabolismo , Lipopolisacáridos/metabolismo , Lipoproteínas/metabolismo , Lipoproteínas VLDL/metabolismo , Monocitos/metabolismo , Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA