Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 64(16): 12261-12272, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34382796

RESUMEN

Modern adjuvants for vaccine formulations are immunostimulating agents whose action is based on the activation of pattern recognition receptors (PRRs) by well-defined ligands to boost innate and adaptive immune responses. Monophosphoryl lipid A (MPLA), a detoxified analogue of lipid A, is a clinically approved adjuvant that stimulates toll-like receptor 4 (TLR4). The synthesis of MPLA poses manufacturing and quality assessment challenges. Bridging this gap, we report here the development and preclinical testing of chemically simplified TLR4 agonists that could sustainably be produced in high purity and on a large scale. Underpinned by computational and biological experiments, we show that synthetic monosaccharide-based molecules (FP compounds) bind to the TLR4/MD-2 dimer with submicromolar affinities stabilizing the active receptor conformation. This results in the activation of MyD88- and TRIF-dependent TLR4 signaling and the NLRP3 inflammasome. FP compounds lack in vivo toxicity and exhibit adjuvant activity by stimulating antibody responses with a potency comparable to MPLA.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Glucosamina/farmacología , Glucolípidos/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Adyuvantes Inmunológicos/síntesis química , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/toxicidad , Animales , Femenino , Glucosamina/síntesis química , Glucosamina/metabolismo , Glucosamina/toxicidad , Glucolípidos/síntesis química , Glucolípidos/metabolismo , Glucolípidos/toxicidad , Humanos , Inflamasomas/metabolismo , Interleucina-1/metabolismo , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
2.
Innate Immun ; 27(3): 275-284, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33858242

RESUMEN

TLRs, including TLR4, play a crucial role in inflammatory-based diseases, and TLR4 has been identified as a therapeutic target for pharmacological intervention. In previous studies, we investigated the potential of FP7, a novel synthetic glycolipid active as a TLR4 antagonist, to inhibit haematopoietic and non-haematopoietic MyD88-dependent TLR4 pro-inflammatory signalling. The main aim of this study was to investigate the action of FP7 and its derivative FP12 on MyD88-independent TLR4 signalling in THP-1 derived macrophages. Western blotting, Ab array and ELISA approaches were used to explore the effect of FP7 and FP12 on TRIF-dependent TLR4 functional activity in response to LPS and other endogenous TLR4 ligands in THP-1 macrophages. A different kinetic in the inhibition of endotoxin-driven TBK1, IRF3 and STAT1 phosphorylation was observed using different LPS chemotypes. Following activation of TLR4 by LPS, data revealed that FP7 and FP12 inhibited TBK1, IRF3 and STAT1 phosphorylation which was associated with down-regulation IFN-ß and IP-10. Specific blockage of the IFN type one receptor showed that these novel molecules inhibited TRIF-dependent TLR4 signalling via IFN-ß pathways. These results add novel information on the mechanism of action of monosaccharide FP derivatives. The inhibition of the TRIF-dependent pathway in human macrophages suggests potential therapeutic uses for these novel TLR4 antagonists in pharmacological interventions on inflammatory diseases.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Antiinflamatorios/uso terapéutico , Glucolípidos/uso terapéutico , Inflamación/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Receptor Toll-Like 4/metabolismo , Antiinflamatorios/farmacología , Quimiocina CXCL10/metabolismo , Descubrimiento de Drogas , Glucolípidos/farmacología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Fosforilación , Transducción de Señal , Células THP-1 , Receptor Toll-Like 4/antagonistas & inhibidores
3.
Innate Immun ; 24(7): 411-421, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30208782

RESUMEN

TLRs, including TLR4, have been shown to play a crucial role in cardiovascular inflammatory-based diseases. The main goal of this study was to determine the potential of FP7, a synthetic glycolipid active as a TLR4 antagonist, to modulate haematopoietic and non-haematopoietic vascular TLR4 pro-inflammatory signalling. HUVEC, human THP-1 monocytes, THP-1-derived macrophages, mouse RAW-264.7 macrophages and Angiotensin II-infused apolipoprotein E-deficient mice were in vitro and in vivo models, respectively. Western blotting, Ab array and ELISA approaches were used to explore the effect of FP7 on TLR4 functional activity in response to bacterial LPS ( in vitro) and endogenous ligands of sterile inflammation ( in vitro and in vivo). Following activation of TLR4, in vitro and in vivo data revealed that FP7 inhibited p38 MAPK and p65 NF-kB phosphorylation associated with down-regulation of a number of TLR4-dependent pro-inflammatory proteins. In addition to inhibition of LPS-induced TLR4 signalling, FP7 negatively regulated TLR4 activation in response to ligands of sterile inflammation (hydroperoxide-rich oxidised LDL, in vitro and Angiotensin II infusion, in vivo). These results demonstrate the ability of FP7 to negatively regulate in vitro and in vivo haematopoietic and non-haematopoietic vascular TLR4 signalling both in humans and mice, suggesting the potential therapeutic use of this TLR4 antagonist for pharmacological intervention of vascular inflammatory diseases.


Asunto(s)
Células Sanguíneas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Glucolípidos/uso terapéutico , Receptor Toll-Like 4/antagonistas & inhibidores , Vasculitis/tratamiento farmacológico , Angiotensina II/metabolismo , Animales , Células Sanguíneas/inmunología , Células Endoteliales/inmunología , Glucolípidos/síntesis química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Lipoproteínas LDL/metabolismo , Ratones , Ratones Noqueados para ApoE , FN-kappa B/metabolismo , Fosforilación , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Células THP-1 , Receptor Toll-Like 4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
J Med Chem ; 61(7): 2895-2909, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29494148

RESUMEN

The structure-activity relationship was investigated in a series of synthetic TLR4 antagonists formed by a glucosamine core linked to two phosphate esters and two linear carbon chains. Molecular modeling showed that the compounds with 10, 12, and 14 carbons chains are associated with higher stabilization of the MD-2/TLR4 antagonist conformation than in the case of the C16 variant. Binding experiments with human MD-2 showed that the C12 and C14 variants have higher affinity than C10, while the C16 variant did not interact with the protein. The molecules, with the exception of the C16 variant, inhibited the LPS-stimulated TLR4 signal in human and murine cells, and the antagonist potency mirrored the MD-2 affinity calculated from in vitro binding experiments. Fourier-transform infrared, nuclear magnetic resonance, and small angle X-ray scattering measurements suggested that the aggregation state in aqueous solution depends on fatty acid chain lengths and that this property can influence TLR4 activity in this series of compounds.


Asunto(s)
Monosacáridos/química , Monosacáridos/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Unión Competitiva/efectos de los fármacos , Línea Celular , Ácidos Grasos/química , Células HEK293 , Humanos , Interleucina-8/biosíntesis , Ligandos , Lipopolisacáridos/metabolismo , Ratones , Modelos Moleculares , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
5.
Atherosclerosis ; 242(2): 563-70, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26318106

RESUMEN

OBJECTIVES: The toll-like receptors (TLRs), including TLR4, have been shown to play a crucial role in vascular inflammatory diseases, such as atherosclerosis and aneurysm. The main goal of this study was to determine the potential of IAXO-102 (Innaxon, Tewkesbury), a novel small molecule TLR4 antagonist, to modulate non-hematopoietic TLR4 proinflammatory signalling and inhibit experimental abdominal aortic aneurysm (AAA) development. METHODS: Human umbilical vein endothelial cells (HUVEC) and Angiotensin II-induced experimental AAA development were our in vitro and in vivo models respectively. Western blotting, antibody array and ELISA approaches were used to explore the effect of IAXO-102 on TLR4 functional activity on two levels: modulation of TLR4-induced mitogen activated protein kinases (MAPK) and p65 NF-kB phosphorylation and expression of TLR4 dependent proinflammatory proteins. RESULTS: Following activation of TLR4, in vitro/in vivo data revealed that IAXO-102 inhibited MAPK and p65 NF-kB phosphorylation associated with down regulation of the expression of TLR4 and TLR4 dependent proinflammatory proteins. Furthermore, IAXO-102 decreased Angiotensin II-induced aortic expansion, rupture and incidence of AAA. CONCLUSIONS: These results demonstrate the ability of IAXO-102 to negatively regulate TLR4 signalling and to inhibit experimental AAA development, suggesting the potential therapeutic use of this TLR4 antagonist for pharmacological intervention of AAA.


Asunto(s)
Amino Azúcares/farmacología , Aorta/metabolismo , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Aneurisma de la Aorta Abdominal/prevención & control , Glucolípidos/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo , Animales , Apolipoproteínas E/genética , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Incidencia , Inflamación , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal , Factor de Transcripción ReIA/metabolismo
6.
Reproduction ; 150(4): 269-77, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26183892

RESUMEN

Preterm labour (PTL) is commonly associated with infection and/or inflammation. Lipopolysaccharide (LPS) from different bacteria can be used to independently or mutually activate Jun N-terminal kinase (JNK)/AP1- or NF-κB-driven inflammatory pathways that lead to PTL. Previous studies using Salmonella abortus LPS, which activates both JNK/AP-1 and NF-κB, showed that selective inhibition of NF-κB delays labour and improves pup outcome. Where labour is induced using Escherichia coli LPS (O111), which upregulates JNK/AP-1 but not NF-κB, inhibition of JNK/AP-1 activation also delays labour. In this study, to determine the potential role of JNK as a therapeutic target in PTL, we investigated the specific contribution of JNK signalling to S. Abortus LPS-induced PTL in mice. Intrauterine administration of S. Abortus LPS to pregnant mice resulted in the activation of JNK in the maternal uterus and fetal brain, upregulation of pro-inflammatory proteins COX-2, CXCL1, and CCL2, phosphorylation of cPLA2 in myometrium, and induction of PTL. Specific inhibition of JNK by co-administration of specific D-JNK inhibitory peptide (D-JNKI) delayed LPS-induced preterm delivery and reduced fetal mortality. This is associated with inhibition of myometrial cPLA2 phosphorylation and proinflammatory proteins synthesis. In addition, we report that D-JNKI inhibits the activation of JNK/JNK3 and caspase-3, which are important mediators of neural cell death in the neonatal brain. Our data demonstrate that specific inhibition of TLR4-activated JNK signalling pathways has potential as a therapeutic approach in the management of infection/inflammation-associated PTL and prevention of the associated detrimental effects to the neonatal brain.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Muerte Fetal/prevención & control , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Trabajo de Parto Prematuro/prevención & control , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Brucella abortus/química , Caspasa 3/biosíntesis , Caspasa 3/efectos de los fármacos , Femenino , Fosfolipasas A2 Grupo II/biosíntesis , Fosfolipasas A2 Grupo II/genética , Inflamación/enzimología , Lipopolisacáridos , Ratones , Proteína Quinasa 10 Activada por Mitógenos/biosíntesis , Proteína Quinasa 10 Activada por Mitógenos/genética , Miometrio/efectos de los fármacos , Miometrio/enzimología , Trabajo de Parto Prematuro/inducido químicamente , Embarazo , Transducción de Señal/efectos de los fármacos , Útero/efectos de los fármacos , Útero/enzimología
7.
J Vasc Res ; 52(2): 81-93, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26113112

RESUMEN

Peroxisome proliferator-activated receptor x03B3; agonists have been shown to inhibit angiotensin II (AngII)-induced experimental abdominal aortic aneurysms. Macrophage infiltration to the vascular wall is an early event in this pathology, and therefore we explored the effects of the peroxisome proliferator-activated receptor x03B3; agonist pioglitazone on AngII-treated macrophages. Using microarray-based expression profiling of phorbol ester-stimulated THP-1 cells, we found that a number of aneurysm-related gene changes effected by AngII were modulated following the addition of pioglitazone. Among those genes, polycystic kidney disease 1 (PKD1) was significantly up-regulated (multiple testing corrected p < 0.05). The analysis of the PKD1 proximal promoter revealed a putative early growth response 1 (EGR1) binding site, which was confirmed by chromatin immunoprecipitation (ChIP) and quantitative PCR. Further analysis of publicly available ChIP-sequencing data revealed that this putative binding site overlapped with a conserved EGR1 binding peak present in 5 other cell lines. Quantitative real-time PCR showed that EGR1 suppressed PKD1, while AngII significantly up-regulated PKD1, an effect counteracted by pioglitazone. Conversely, in EGR1 short hairpin RNA lentivirally transduced THP-1 cells, reduced EGR1 led to a significant up-regulation of PKD1, especially after treatment with pioglitazone. In vivo, deficiency of Egr1 in the haematopoietic compartment of mice completely abolished the incidence of CaCl2-induced aneurysm formation.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Macrófagos/efectos de los fármacos , Tiazolidinedionas/farmacología , Angiotensina II/farmacología , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Secuencia de Bases , Sitios de Unión , Cloruro de Calcio , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/deficiencia , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Femenino , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Masculino , Ratones Noqueados , Datos de Secuencia Molecular , PPAR gamma/agonistas , PPAR gamma/metabolismo , Pioglitazona , Regiones Promotoras Genéticas , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factores de Tiempo , Transfección
8.
Aorta (Stamford) ; 3(5): 152-66, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27175366

RESUMEN

BACKGROUND: Since aortic diameter is the most -significant risk factor for rupture, we sought to identify stress-dependent changes in gene expression to illuminate novel molecular processes in aneurysm rupture. MATERIALS AND METHODS: We constructed finite element maps of abdominal computerized tomography scans (CTs) of seven abdominal aortic aneurysm (AAA) patients to map wall stress. Paired biopsies from high- and low-stress areas were collected at surgery using vascular landmarks as coordinates. Differential gene expression was evaluated by Illumina Array analysis, using the whole genome DNA-mediated, annealing, selection, extension, and ligation (DASL) gene chip (n = 3 paired samples). RESULTS: The sole significant candidate from this analysis, Lamin A/C, was validated at the protein level, using western blotting. Lamin A/C expression in the inferior mesenteric vein (IMV) of AAA patients was compared to a control group and in aortic smooth muscle cells in culture in response to physiological pulsatile stretch. -Areas of high wall stress (n = 7) correlate to those -regions which have the thinnest walls [778 µm (585-1120 µm)] in comparison to areas of lowest wall stress [1620 µm (962-2919 µm)]. Induced expression of Lamin A/C -correlated with areas of high wall stress from AAAs but was not significantly induced in the IMV from AAA patients compared to controls (n = 16). Stress-induced expression of Lamin A/C was mimicked by exposing aortic smooth muscle cells to prolonged pulsatile stretch. CONCLUSION: Lamin A/C protein is specifically increased in areas of high wall stress in AAA from patients, but is not increased on other vascular beds of aneurysm patients, suggesting that its elevation may be a compensatory response to the pathobiology leading to aneurysms.

9.
Anticancer Agents Med Chem ; 14(8): 1169-78, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24954859

RESUMEN

Lupulone, a ß-acid derived from hop extracts has been shown to exhibit cytotoxic activity against cancer cells. In this study we investigated the functional role of different modes of cell death that mediate anticancer effect of lupulone derivatives in prostate cancer cells. ELISA, immunoblotting and siRNA approaches were utilised to study cell death, expression of proteins of interest and their functional activities. We found that the anticancer effect of lupulone derivatives on prostate cancer cells is associated with induction of apoptosis and autophagy as determined by increases of DNA fragmentation and LC3I/ LC3II conversion respectively. Inhibition of apoptosis using a pan-caspase inhibitor resulted in increased levels of autophagy. Following screening of proteins associated with autophagy we found that Atg4ß expression was increased in prostate cancer cells after treatment with lupulone. Transfection of cells with siRNA against Atg4ß resulted in increased levels of apoptosis in prostate cancer cells. Treatment of prostate cancer cells with lupulone derivatives initiated two modes of cell death: apoptosis as a killing pathway and autophagy as a protection against cell death. Further studies are required to investigate the regulation of Atg4ß activity in lupulone derivatives-induced negative crosstalk between apoptosis and autophagy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias de la Próstata/patología , Terpenos/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Antineoplásicos/química , Proteínas Relacionadas con la Autofagia , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Línea Celular Tumoral , Cisteína Endopeptidasas/metabolismo , Fragmentación del ADN/efectos de los fármacos , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Terpenos/química
10.
Nutr Cancer ; 65(7): 1086-92, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24168111

RESUMEN

Lupulone, a ß-acid derived from hop extracts has been shown to exhibit antibacterial and anticancer activity. In this study we investigated the anticancer potency of lupulone and its novel derivatives and their mechanism of action on prostate cancer cells. Cell viability was determined using the MTT assay, and the ELISA approach was used to investigate induction of apoptosis. Immunoblot analysis was carried out to determine activation and regulation of proteins associated with cell death. Screening of natural and new lupulone derivatives for their anticancer activity demonstrated that one (lupulone derivative 1h) displayed stronger anticancer activity than lupulone itself on PC3 and DU145 prostate cancer cells. We further found that lupulone derivatives induced caspase-dependent apoptosis that is associated with activation of caspases 8, 9, and 3. Furthermore, caspase 8 inhibitor Z-IETD-fmk reduced cell death induced by lupulone derivatives, suggesting that apoptosis is mediated by caspase 8. Finally, we found that lupulone and its synthetic derivatives also increased formation of LC3II suggesting that autophagy is also implicated in prostate cancer cell death. The new lupulone derivatives induce caspase-dependent apoptosis and autophagy in prostate cancer cells and appear to be good candidates for further preclinical studies of prostate cancer treatment.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Humulus/química , Extractos Vegetales/farmacología , Neoplasias de la Próstata/metabolismo , Terpenos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Masculino , Oligopéptidos/farmacología
11.
Biol Cell ; 105(9): 414-29, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23734772

RESUMEN

BACKGROUND INFORMATION: Tumour cells can be induced to undergo apoptosis after treatment with the tumour necrosis factor α-related death-inducing ligand (TRAIL). Although human pancreatic cancer cells show varying degrees of response they can be sensitised to the pro-apoptotic effects of TRAIL in the presence of celastrol, a natural compound extracted from the plant Tripterygium wilfordii Hook F. One important aspect of the cellular response to TRAIL is the control of protein synthesis, a key regulator of which is the eukaryotic initiation factor 4E-binding protein, 4E-BP1. RESULTS: We examined the effects of celastrol and TRAIL in several pancreatic cancer cell lines. In cells that are normally resistant to TRAIL, synergistic effects of TRAIL plus celastrol on commitment to apoptosis and inhibition of protein synthesis were observed. These were associated with a strong up-regulation and dephosphorylation of 4E-BP1. The enhancement of 4E-BP1 expression, which correlated with a threefold increase in the level of the 4E-BP1 transcript, was blocked by inhibitors of reactive oxygen species and the JNK protein kinase. When the expression of 4E-BP1 was reduced by an inducible micro-RNA, TRAIL-mediated apoptosis was inhibited. CONCLUSION: These results suggest that 4E-BP1 plays a critical role in the mechanism by which TRAIL and celastrol together cause apoptotic cell death in human pancreatic tumour cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fosfoproteínas/genética , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Triterpenos/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Triterpenos Pentacíclicos , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Biosíntesis de Proteínas , Proteínas Recombinantes/farmacología , Transducción de Señal , Neoplasias Pancreáticas
12.
Curr Vasc Pharmacol ; 11(3): 293-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22724482

RESUMEN

There is strong epidemiological evidence that patients with diabetes have a lower incidence of abdominal aortic aneurysm. The precise mechanism of this negative association is unknown. Whilst a number of studies have supported the hypothesis that protection is a function of diabetes-mediated changes in the vascular extracellular matrix biology, there is also support for the idea that the treatment regimens used in diabetes may afford protection against AAA. In particular the pleiotropic drug family, the thiazolidinediones have been examined as candidates to ameliorate aneurysm formation. Both the thiazolidinediones, and the structurally related family, fibrates, have been shown to have anti-inflammatory and antioxidative effects, in addition to ability to modulatate glucose and lipid homeostasis. In this brief review we present the current data exploring the use of thiazolidinediones in experimental aneurysm development. Despite the fact that both thiazolidinediones Rosiglitazone and Pioglitazone are no longer prescribed in Europe and the US, they have provided important insights into the mechanism of action, and the application of other pleiotropic drugs in the treatment of AAA. One such pleiotropic drug is high-density lipoproteins (HDLs), which have been shown to have a broad spectrum of effects, including activation of PPARs, which may favour their use as a new drug target for protection against AAA development.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Diabetes Mellitus/epidemiología , Tiazolidinedionas/farmacología , Animales , Aneurisma de la Aorta Abdominal/epidemiología , Diabetes Mellitus/tratamiento farmacológico , Humanos , Hipoglucemiantes/farmacología , Lipoproteínas HDL/metabolismo , Terapia Molecular Dirigida , Receptores Activados del Proliferador del Peroxisoma/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Pioglitazona , Factores de Riesgo , Rosiglitazona
13.
Arterioscler Thromb Vasc Biol ; 32(11): 2678-86, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023368

RESUMEN

OBJECTIVE: Patients with abdominal aortic aneurysms have lower concentrations of high-density lipoproteins (HDLs), leading us to investigate whether increasing plasma HDLs could influence aneurysm formation. METHODS AND RESULTS: Using the angiotensin II-induced hypercholesterolemic and the CaCl(2)-induced normocholesterolemic mouse model of AAA, we investigated the hypothesis that elevation of HDLs inhibits AAA. HDLs elevated before or at the time of AAA induction reduced AAA formation in both models but had no effect on early ruptures. Analysis of protein lysates from specific aortic segments demonstrated site-specific effects of HDLs on early signal transduction and cellular attrition. We found that HDLs reduced extracellular signal related kinases 1/2 activation in the suprarenal segment, while having no effect on p38 mitogen-associated protein kinase activation in any aortic segment and inhibiting c-Jun N-terminal kinase activation in all aortic segments. In addition, HDL elevation inhibited angiotensin II-induced apoptosis while inducing autophagy in the suprarenal segment of the aorta. Using Illumina gene array profiling we investigated the ability of HDL to modulate basal suprarenal aortic gene expression. CONCLUSIONS: Increasing plasma HDLs inhibit experimental AAA formation, independent of hypercholesterolemia via reduced extracellular signal related kinases 1/2 activation and alteration of the balance of cellular attrition. HDLs modulate genes involved in matrix remodelling, cell migration, and proliferation.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Lipoproteínas HDL/sangre , Angiotensina II , Animales , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/etiología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Rotura de la Aorta/sangre , Rotura de la Aorta/etiología , Rotura de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Autofagia , Cloruro de Calcio , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Inyecciones Subcutáneas , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipoproteínas HDL/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Atherosclerosis ; 225(1): 69-75, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22999334

RESUMEN

OBJECTIVE: Development and rupture of aortic aneurysms (AA) is a complex process involving inflammation, cell death, tissue and matrix remodelling. The thiazolidinediones (TZDs) including Rosiglitazone (RGZ) are a family of drugs which act as agonists of the nuclear peroxisome proliferator-activated receptors and have a broad spectrum of effects on a number of biological processes in the cardiovascular system. In our previous study we have demonstrated that RGZ has a marked effect on both aneurysm rupture and development, however, the precise mechanism of this is unknown. METHODS AND RESULTS: In the present study, we examined possible targets of RGZ action in the early stages of Angiotensin II-induced AA in apolipoprotein E-deficient mice. For this purpose we employed immunoblotting, ELISA and antibody array approaches. We found that RGZ significantly inhibited c-Jun N-terminal kinase (JNK) phosphorylation and down-regulated toll-like receptor 4 (TLR4) expression at the site of lesion formation in response to Angiotensin II infusion in the initiation stage (6-72 h) of experimental AA development. Importantly, this effect was also associated with a decrease of CD4 antigen and reduction in production of TLR4/JNK-dependant proinflammatory chemokines MCP-1 and MIP-1α. CONCLUSION: These data suggest that RGZ can modulate inflammatory processes by blocking TLR4/JNK signalling in initiation stages of AA development.


Asunto(s)
Aneurisma de la Aorta/inducido químicamente , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Angiotensina II , Animales , Aneurisma de la Aorta/prevención & control , Rotura de la Aorta/prevención & control , Regulación hacia Abajo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Fosforilación/efectos de los fármacos , Rosiglitazona , Receptor Toll-Like 4/metabolismo
15.
J Endocrinol ; 212(2): 187-97, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22083217

RESUMEN

This study investigated the role of urocortin (UCN), a member of the corticotrophin-releasing factor (CRF) family of peptides, in osteoclast maturation and function. We found that 10(-7) M UCN significantly (P<0.05) suppressed osteoclast differentiation from bone marrow precursor cells in culture and reduced the expression of several osteoclastic markers. Furthermore, UCN potently suppressed osteoclast bone resorption, by significantly inhibiting both the plan area of bone resorbed by osteoclasts and actin ring formation within osteoclasts at 10(-9) M (P<0.05), with complete inhibition at 10(-7) M (P<0.001). UCN also inhibited osteoclast motility (10(-7) M) but had no effect on osteoclast survival. Osteoclasts expressed mRNA encoding both UCN and the CRF receptor 2ß subtype. Pre-osteoclasts however, expressed CRF receptor 2ß alone. Unstimulated osteoclasts contained constitutively active cation channel currents with a unitary conductance of 3-4 pS, which were inhibited by over 70% with UCN (10(-7) M). Compounds that regulate calcium signalling and energy status of the cell, both crucial for osteoclast activity were investigated. The non-selective cation channel blockers, lanthanum (La(3)(+)) and gadolinium (Gd(3)(+)), inhibited actin ring formation in osteoclasts, whereas modulators of voltage-dependent Ca(2)(+) channels and K(ATP) channels had no effect. These findings show for the first time that UCN is a novel anti-resorptive molecule that acts through a direct effect on osteoclasts and their precursor cells.


Asunto(s)
Osteoclastos/citología , Osteoclastos/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Canales Catiónicos TRPC/metabolismo , Urocortinas/metabolismo , Células 3T3 , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Animales , Animales Recién Nacidos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/ultraestructura , Resorción Ósea/patología , Resorción Ósea/fisiopatología , Resorción Ósea/prevención & control , Huesos/citología , Huesos/ultraestructura , Señalización del Calcio , Bovinos , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Regulación de la Expresión Génica , Moduladores del Transporte de Membrana/farmacología , Ratones , Osteoclastos/efectos de los fármacos , Osteoclastos/ultraestructura , ARN Mensajero/metabolismo , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/genética , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Urocortinas/genética
16.
J Vasc Surg ; 54(4): 1100-1108.e6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21741794

RESUMEN

INTRODUCTION: Abdominal aortic aneurysms (AAA) are associated with inflammation, apoptosis, and matrix degradation. AAA tissue represents the end stage of disease, limiting its utility in identification of factors culpable for initiation of aneurysm development. Recent evidence suggests that AAAs are a local representation of a systemic disease of the vasculature. Morphologic and molecular changes, comparable to those found in the aneurysm wall, have been demonstrated in veins from patients with AAAs. Changes in the vascular tissue proteome of patients with AAAs were investigated, using inferior mesenteric vein (IMV), to gain insight into early molecular changes contributing to AAA development. METHODS: IMV was harvested from 16 patients with AAA and 16 matched controls. Whole IMV lysates were subjected to 2-D difference in gel electrophoresis (2D-DIGE) with quantitative densitometry. Protein spots differentially expressed in AAA were identified using mass spectrometry. Differential protein expression was validated by Western blotting and localized to cell type by immunohistochemistry (IHC). RESULTS: Decreased levels of prohibitin (AAA, 2.00 ± 1.37; controls, 3.81 ± 1.39; 1.9-fold change; P = .02) AAA (7.33 ± 3.9; controls, 14.5 ± 5.6; 2-fold change; P = .001), along with relative increases in a cleaved fragment of vimentin (AAA, 12.9 ± 9; controls, 6.9 ± 4.7; 2-fold change; P = .11) were identified in AAA patients. All proteins were localized to the vascular smooth muscle cells. CONCLUSIONS: Proteins important in combating the injurious effects of oxidative stress and modulating the response to inflammation appear reduced in the vasculature of patients with AAA. These changes may represent early events in AAA formation. Enhancing expression of these proteins might offer a novel therapeutic avenue to inhibit AAA development.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Músculo Liso Vascular/química , Miocitos del Músculo Liso/química , Proteínas/análisis , Proteómica , Anciano , Anexina A1/análisis , Aorta Abdominal/química , Western Blotting , Estudios de Casos y Controles , Densitometría , Electroforesis en Gel Bidimensional , Femenino , Humanos , Inmunohistoquímica , Londres , Masculino , Espectrometría de Masas , Venas Mesentéricas/química , Persona de Mediana Edad , Prohibitinas , Proteómica/métodos , Proteínas Represoras/análisis , Reproducibilidad de los Resultados , Vimentina/análisis
17.
J Neurosci Res ; 88(8): 1632-44, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20091773

RESUMEN

Activated microglia can influence the survival of neural cells through the release of cytotoxic factors. Here, we investigated the interaction between Toll-like receptor 4 (TLR4)-activated microglia and oligodendrocytes or their precursor cells (OPC). Primary rat or N9 microglial cells were activated by exposure to TLR4-specifc lipopolysaccharide (LPS), resulting in mitogen-activated protein kinase activation, increased CD68 and inducible nitric oxide synthase expression, and release of the proinflammatory cytokines tumor necrosis factor (TNF) and interleukin-6 (IL-6). Microglial conditioned medium (MGCM) from LPS-activated microglia attenuated primary OPC proliferation without inducing cell death. The microglial-induced inhibition of OPC proliferation was reversed by stimulating group III metabotropic glutamate receptors in microglia with the agonist L-AP4. In contrast to OPC, LPS-activated MGCM enhanced the survival of mature oligodendrocytes. Further investigation suggested that TNF and IL-6 released from TLR4-activated microglia might contribute to the effect of MGCM on OPC proliferation, insofar as TNF depletion of LPS-activated MGCM reduced the inhibition of OPC proliferation, and direct addition of TNF or IL-6 attenuated or increased proliferation, respectively. OPC themselves were also found to express proteins involved in TLR4 signalling, including TLR4, MyD88, and MAL. Although LPS stimulation of OPC did not induce proinflammatory cytokine release or affect their survival, it did trigger JNK phosphorylation, suggesting that TLR4 signalling in these cells is active. These findings suggest that OPC survival may be influenced not only by factors released from endotoxin-activated microglia but also through a direct response to endotoxins. This may have consequences for myelination under conditions in which microglial activation and cerebral infection are both implicated. , Inc.


Asunto(s)
Proliferación Celular , Microglía/fisiología , Oligodendroglía/fisiología , Células Madre/fisiología , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Gangliósidos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Antígeno Ki-67/metabolismo , Lipopolisacáridos/farmacología , Microglía/química , Microglía/efectos de los fármacos , Proteína Básica de Mielina/metabolismo , Neuroblastoma , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
18.
Cell Cycle ; 8(7): 1069-79, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19270512

RESUMEN

The potential of mesenchymal stem cells (MSC) to differentiate into neural lineages has raised the possibility of autologous cell transplantation as a therapy for neurodegenerative diseases. We have identified a population of circulating human fetal mesenchymal stem cells (hfMSC) that are highly proliferative and can readily differentiate into mesodermal lineages such as bone, cartilage, fat and muscle. Here, we demonstrate for the first time that primary hfMSC can differentiate into cells with an oligodendrocyte phenotype both in vitro and in vivo. By exposing hfMSC to neuronal conditioned medium or by introducing the pro-oligodendrocyte gene, Olig-2, hfMSC adopted an oligodendrocyte-like morphology, expressed oligodendrocyte markers and appeared to mature appropriately in culture. Importantly we also demonstrate the differentiation of a clonal population of hfMSC into both mesodermal (bone) and ectodermal (oligodendrocyte) lineages. In the developing murine brain transplanted hfMSC integrated into the parenchyma but oligodendrocyte differentiation of these naïve hfMSC was very low. However, the proportion of cells expressing oligodendrocyte markers increased significantly (from 0.2% to 4%) by preexposing the cells to differentiation medium in vitro prior to transplantation. Importantly, the process of in vivo differentiation occurred without cell fusion. These findings suggest that hfMSC may provide a potential source of oligodendrocytes for study and potential therapy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Madre Mesenquimatosas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Oligodendroglía/fisiología , Animales , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Medios de Cultivo Condicionados/farmacología , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/citología , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra
19.
Endocrinology ; 150(2): 699-706, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18845626

RESUMEN

Intrauterine infection is a common trigger for preterm birth and is also a risk factor for the subsequent development of neurodevelopmental abnormalities in the neonate. Bacterial lipopolysaccharide (LPS) binds to toll-like receptor-4 (TLR-4) to activate proinflammatory signaling pathways, which are implicated in both preterm delivery and antenatal brain injury. The transcription factor nuclear factor-kappaB (NF-kappaB) is a key player in the orchestration of the inflammatory response and has a central role in parturition. Here we show that intrauterine administration of TLR-4-specific LPS to pregnant mice results in the activation of NF-kappaB in the maternal uterus and the fetal brain, up-regulation of proinflammatory proteins cyclooxygenase-2, chemokine ligand 1, ChemoKine (C-C motif) ligand 2, and cytosolic phospholipase A(2) in myometrium, and induction of preterm delivery. 15-Deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) is an antiinflammatory prostaglandin that plays a role in promoting the resolution of inflammation. We report that coadministration of 15d-PGJ(2) and LPS to pregnant mice delays LPS-induced preterm delivery and confers protection from LPS-induced fetal mortality. This is associated with inhibition of myometrial NF-kappaB, cytosolic phospholipase A(2), and c-Jun N-terminal kinase activation, and of inflammatory protein synthesis. Therefore 15d-PGJ(2) has anti-inflammatory effects via inhibition of multiple aspects of inflammation-driven TRL-4 signaling pathway. Thus, 15d-PGJ(2) or compounds with similar antiinflammatory functions may have potential as therapeutic agents in the management of preterm labor with the added advantage of preventing detrimental effects to the fetus that may result from infection/inflammation.


Asunto(s)
Lipopolisacáridos/efectos adversos , Trabajo de Parto Prematuro/mortalidad , Trabajo de Parto Prematuro/prevención & control , Prostaglandina D2/análogos & derivados , Animales , Animales Recién Nacidos , Animales no Consanguíneos , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Ciclopentanos/farmacología , Evaluación Preclínica de Medicamentos , Femenino , Inflamación/complicaciones , Ratones , Miometrio/efectos de los fármacos , Miometrio/metabolismo , Miometrio/patología , FN-kappa B/metabolismo , Trabajo de Parto Prematuro/inducido químicamente , Fosfolipasas A2/metabolismo , Fosforilación/efectos de los fármacos , Embarazo , Prostaglandina D2/farmacología , Prostaglandina D2/uso terapéutico , Factores de Tiempo , Enfermedades Uterinas/tratamiento farmacológico , Enfermedades Uterinas/etiología , Útero/metabolismo , Útero/patología
20.
J Cereb Blood Flow Metab ; 27(5): 1022-32, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17063149

RESUMEN

c-Jun N-terminal kinase 3 (JNK3) is a member of the stress-activated group of mitogen-activated protein kinases. c-Jun N-terminal kinase 3 is a potent mediator of apoptosis and the use of JNK inhibitors or jnk3 gene deletion each protect against brain injury in adults. However, little is known about the role of JNK3 or its mechanism of action in neonatal brain injury. The aim of the present study was to compare the vulnerability of neonatal JNK3 knockout (JNK3 KO) mice and wild-type (WT) mice to cerebral hypoxic-ischaemic injury (HII) using unilateral-carotid occlusion combined with transient hypoxia. The degree of neural tissue loss in JNK3 KO mice was substantially reduced compared with WT mice (JNK3 KO 27.8%+/-2.8% versus WT 48.3%+/-2.0%, P

Asunto(s)
Animales Recién Nacidos/fisiología , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Proteína Quinasa 10 Activada por Mitógenos/fisiología , Factor de Transcripción Activador 2/genética , Animales , Apoptosis/genética , Apoptosis/fisiología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 10 Activada por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/patología , Fosforilación , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...