Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Diabetologia ; 65(10): 1721-1733, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35802167

RESUMEN

AIMS/HYPOTHESIS: The general population is ageing, involving an enhanced incidence of chronic diseases such as type 2 diabetes. With ageing, DNA methylation of FHL2 increases, as well as expression of the four and a half LIM domains 2 (FHL2) protein in human pancreatic islets. We hypothesised that FHL2 is actively involved in glucose metabolism. METHODS: Publicly available microarray datasets from human pancreatic islets were analysed for FHL2 expression. In FHL2-deficient mice, we studied glucose clearance and insulin secretion. Gene expression analysis and glucose-stimulated insulin secretion (GSIS) were determined in isolated murine FHL2-deficient islets to evaluate insulin-secretory capacity. Moreover, knockdown and overexpression of FHL2 were accomplished in MIN6 cells to delineate the underlying mechanism of FHL2 function. RESULTS: Transcriptomics of human pancreatic islets revealed that individuals with elevated levels of HbA1c displayed increased FHL2 expression, which correlated negatively with insulin secretion pathways. In line with this observation, FHL2-deficient mice cleared glucose more efficiently than wild-type littermates through increased plasma insulin levels. Insulin sensitivity was comparable between these genotypes. Interestingly, pancreatic islets isolated from FHL2-deficient mice secreted more insulin in GSIS assays than wild-type mouse islets even though insulin content and islet size was similar. To support this observation, we demonstrated increased expression of the transcription factor crucial in insulin secretion, MAF BZIP transcription factor A (MafA), higher expression of GLUT2 and reduced expression of the adverse factor c-Jun in FHL2-deficient islets. The underlying mechanism of FHL2 was further delineated in MIN6 cells. FHL2-knockdown led to enhanced activation of forkhead box protein O1 (FOXO1) and its downstream genes such as Mafa and Pdx1 (encoding pancreatic and duodenal homeobox 1), as well as increased glucose uptake. On the other hand, FHL2 overexpression in MIN6 cells blocked GSIS, increased the formation of reactive oxygen species and increased c-Jun activity. CONCLUSIONS/INTERPRETATION: Our data demonstrate that FHL2 deficiency improves insulin secretion from beta cells and improves glucose tolerance in mice. Given that FHL2 expression in humans increases with age and that high expression levels of FHL2 are associated with beta cell dysfunction, we propose that enhanced FHL2 expression in elderly individuals contributes to glucose intolerance and the development of type 2 diabetes. DATA AVAILABILITY: The human islet microarray datasets used are publicly available and can be found on https://www.ncbi.nlm.nih.gov/geo/ .


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Anciano , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteína Forkhead Box O1/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas con Homeodominio LIM/genética , Ratones , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/metabolismo
2.
Haematologica ; 105(6): 1677-1685, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31467128

RESUMEN

Bleeding disorders and thrombotic complications are major causes of morbidity and mortality with many cases being unexplained. Thrombus formation involves aberrant expression and activation of tissue factor (TF) in vascular endothelial and smooth muscle cells. Here, we sought to identify factors that modulate TF gene expression and activity in these vascular cells. The LIM-only protein FHL2 is a scaffolding protein that modulates signal transduction pathways with crucial functions in endothelial and smooth muscle cells. However, the role of FHL2 in TF regulation and thrombosis remains unexplored. Using a murine model of venous thrombosis in mesenteric vessels, we demonstrated that FHL2 deficiency results in exacerbated thrombus formation. Gain- and loss-of-function experiments revealed that FHL2 represses TF expression in endothelial and smooth muscle cells through inhibition of the transcription factors nuclear factor κB and activating protein-1. Furthermore, we observed that FHL2 interacts with the cytoplasmic tail of TF. In line with our in vivo observations, FHL2 decreases TF activity in endothelial and smooth muscle cells whereas FHL2 knockdown or deficiency results in enhanced TF activity. Finally, the FHL2 single nucleotide polymorphism rs4851770 was associated with the risk of venous thrombosis in a large population of venous thrombosis cases and control subjects from 12 studies (INVENT consortium). Altogether, our results highlight functional involvement of FHL2 in TF-mediated coagulation and identify FHL2 as a novel gene associated with venous thrombosis in humans.


Asunto(s)
Tromboplastina , Trombosis de la Vena , Animales , Variación Genética , Humanos , Proteínas con Homeodominio LIM/genética , Ratones , Proteínas Musculares/genética , Tromboplastina/genética , Factores de Transcripción/genética , Trombosis de la Vena/genética
3.
J Neurosci ; 38(6): 1335-1350, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29295823

RESUMEN

The energetic costs of behavioral chronic stress are unlikely to be sustainable without neuronal plasticity. Mitochondria have the capacity to handle synaptic activity up to a limit before energetic depletion occurs. Protective mechanisms driven by the induction of neuronal genes likely evolved to buffer the consequences of chronic stress on excitatory neurons in prefrontal cortex (PFC), as this circuitry is vulnerable to excitotoxic insults. Little is known about the genes involved in mitochondrial adaptation to the buildup of chronic stress. Using combinations of genetic manipulations and stress for analyzing structural, transcriptional, mitochondrial, and behavioral outcomes, we characterized NR4A1 as a stress-inducible modifier of mitochondrial energetic competence and dendritic spine number in PFC. NR4A1 acted as a transcription factor for changing the expression of target genes previously involved in mitochondrial uncoupling, AMP-activated protein kinase activation, and synaptic growth. Maintenance of NR4A1 activity by chronic stress played a critical role in the regressive synaptic organization in PFC of mouse models of stress (male only). Knockdown, dominant-negative approach, and knockout of Nr4a1 in mice and rats (male only) protected pyramidal neurons against the adverse effects of chronic stress. In human PFC tissues of men and women, high levels of the transcriptionally active NR4A1 correlated with measures of synaptic loss and cognitive impairment. In the context of chronic stress, prolonged expression and activity of NR4A1 may lead to responses of mitochondria and synaptic connectivity that do not match environmental demand, resulting in circuit malfunction between PFC and other brain regions, constituting a pathological feature across disorders.SIGNIFICANCE STATEMENT The bioenergetic cost of chronic stress is too high to be sustainable by pyramidal prefrontal neurons. Cellular checkpoints have evolved to adjust the responses of mitochondria and synapses to the buildup of chronic stress. NR4A1 plays such a role by controlling the energetic competence of mitochondria with respect to synapse number. As an immediate-early gene, Nr4a1 promotes neuronal plasticity, but sustained expression or activity can be detrimental. NR4A1 expression and activity is sustained by chronic stress in animal models and in human studies of neuropathologies sensitive to the buildup of chronic stress. Therefore, antagonism of NR4A1 is a promising avenue for preventing the regressive synaptic reorganization in cortical systems in the context of chronic stress.


Asunto(s)
Mitocondrias/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Corteza Prefrontal/fisiopatología , Estrés Psicológico/fisiopatología , Sinapsis/metabolismo , Anciano , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Recuento de Células , Enfermedad Crónica , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/psicología , Espinas Dendríticas , Femenino , Regulación de la Expresión Génica/genética , Suspensión Trasera , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/genética , Corteza Prefrontal/citología , Células Piramidales/fisiología , Ratas , Estrés Psicológico/psicología
4.
J Biol Chem ; 293(39): 15070-15083, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30111591

RESUMEN

Gene targeting via homologous recombination can occasionally result in incomplete disruption of the targeted gene. Here, we show that a widely used Nur77-deficient transgenic mouse model expresses a truncated protein encoding for part of the N-terminal domain of nuclear receptor Nur77. This truncated Nur77 protein is absent in a newly developed Nur77-deficient mouse strain generated using Cre-Lox recombination. Comparison of these two mouse strains using immunohistochemistry, flow cytometry, and colony-forming assays shows that homologous recombination-derived Nur77-deficient mice, but not WT or Cre-Lox-derived Nur77-deficient mice, suffer from liver immune cell infiltrates, loss of splenic architecture, and increased numbers of bone marrow hematopoietic stem cells and splenic colony-forming cells with age. Mechanistically, we demonstrate that the truncated Nur77 N-terminal domain protein maintains the stability and activity of hypoxia-inducible factor (HIF)-1, a transcription factor known to regulate bone marrow homeostasis. Additionally, a previously discovered, but uncharacterized, human Nur77 transcript variant that encodes solely for its N-terminal domain, designated TR3ß, can also stabilize and activate HIF-1α. Meta-analysis of publicly available microarray data sets shows that TR3ß is highly expressed in human bone marrow cells and acute myeloid leukemia samples. In conclusion, our study provides evidence that a transgenic mouse model commonly used to study the biological function of Nur77 has several major drawbacks, while simultaneously identifying the importance of nongenomic Nur77 activity in the regulation of bone marrow homeostasis.


Asunto(s)
Células de la Médula Ósea/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Dominios Proteicos/genética , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Citometría de Flujo , Regulación de la Expresión Génica/genética , Homeostasis/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Ratones , Ratones Transgénicos , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/química
5.
Haematologica ; 103(6): 1073-1082, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29545340

RESUMEN

Tissue Factor is a cell-surface glycoprotein expressed in various cells of the vasculature and is the principal regulator of the blood coagulation cascade and hemostasis. Notably, aberrant expression of Tissue Factor is associated with cardiovascular pathologies such as atherosclerosis and thrombosis. Here, we sought to identify factors that regulate Tissue Factor gene expression and activity. Tissue Factor gene expression is regulated by various transcription factors, including activating protein-1 and nuclear factor-κ B. The peptidyl-prolyl isomerase Pin1 is known to modulate the activity of these two transcription factors, and we now show that Pin1 augments Tissue Factor gene expression in both vascular smooth muscle cells and activated endothelial cells via activating protein-1 and nuclear factor-κ B signaling. Furthermore, the cytoplasmic domain of Tissue Factor contains a well-conserved phospho-Ser258-Pro259 amino-acid motif recognized by Pin1. Using co-immunoprecipitation and solution nuclear magnetic resonance spectroscopy, we show that the WW-domain of Pin1 directly binds the cytoplasmic domain of Tissue Factor. This interaction occurs via the phospho-Ser258-Pro259 sequence in the Tissue Factor cytoplasmic domain and results in increased protein half-life and pro-coagulant activity. Taken together, our results establish Pin1 as an upstream regulator of Tissue Factor-mediated coagulation, thereby opening up new avenues for research into the use of specific Pin1 inhibitors for the treatment of diseases characterized by pathological coagulation, such as thrombosis and atherosclerosis.


Asunto(s)
Coagulantes/metabolismo , Expresión Génica , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Tromboplastina/química , Factor de Transcripción AP-1/metabolismo
6.
J Pathol ; 243(3): 294-306, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28727149

RESUMEN

Marfan syndrome (MFS) is a connective tissue disorder in which aortic rupture is the major cause of death. MFS patients with an aortic diameter below the advised limit for prophylactic surgery (<5 cm) may unexpectedly experience an aortic dissection or rupture, despite yearly monitoring. Hence, there is a clear need for improved prognostic markers to predict such aortic events. We hypothesize that elastin fragments play a causal role in aortic calcification in MFS, and that microcalcification serves as a marker for aortic disease severity. To address this hypothesis, we analysed MFS patient and mouse aortas. MFS patient aortic tissue showed enhanced microcalcification in areas with extensive elastic lamina fragmentation in the media. A causal relationship between medial injury and microcalcification was revealed by studies in vascular smooth muscle cells (SMCs); elastin peptides were shown to increase the activity of the calcification marker alkaline phosphatase (ALP) and reduce the expression of the calcification inhibitor matrix GLA protein in human SMCs. In murine Fbn1C1039G/+ MFS aortic SMCs, Alpl mRNA and activity were upregulated as compared with wild-type SMCs. The elastin peptide-induced ALP activity was prevented by incubation with lactose or a neuraminidase inhibitor, which inhibit the elastin receptor complex, and a mitogen-activated protein kinase kinase-1/2 inhibitor, indicating downstream involvement of extracellular signal-regulated kinase-1/2 (ERK1/2) phosphorylation. Histological analyses in MFS mice revealed macrocalcification in the aortic root, whereas the ascending aorta contained microcalcification, as identified with the near-infrared fluorescent bisphosphonate probe OsteoSense-800. Significantly, microcalcification correlated strongly with aortic diameter, distensibility, elastin breaks, and phosphorylated ERK1/2. In conclusion, microcalcification co-localizes with aortic elastin degradation in MFS aortas of humans and mice, where elastin-derived peptides induce a calcification process in SMCs via the elastin receptor complex and ERK1/2 activation. We propose microcalcification as a novel imaging marker to monitor local elastin degradation and thus predict aortic events in MFS patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Elastina/metabolismo , Síndrome de Marfan/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Aorta/metabolismo , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Calcinosis/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Síndrome de Marfan/patología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología
7.
J Immunol ; 195(4): 1388-98, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26170382

RESUMEN

Allergic asthma is characterized by persistent chronic airway inflammation, which leads to mucus hypersecretion and airway hyperresponsiveness. Nuclear receptor Nur77 plays a pivotal role in distinct immune and inflammatory cells and is expressed in eosinophils and lung epithelium. However, the role of Nur77 in allergic airway inflammation has not been studied so far. In the present study, we determined the role of Nur77 in airway inflammation using a murine model of OVA-induced allergic airway inflammation. We found that OVA-challenged Nur77 knockout (KO) mice show significantly enhanced infiltration of inflammatory cells, including eosinophils and lymphocytes, and aggravated mucus production. The infiltration of macrophages is limited in this model and was similar in wild-type and Nur77 KO mice. Higher levels of Th2 cytokines were found in bronchoalveolar lavage fluid and draining lymph node cells of Nur77-KO mice, as well as increased serum IgG1 and IgG2a levels. Knockdown of Nur77 in human lung epithelial cells resulted in a marked increase in IκBα phosphorylation, corresponding with elevated NF-κB activity, whereas Nur77 overexpression decreased NF-κB activity. Consistently, Nur77 significantly decreased mRNA levels of inflammatory cytokines and Muc5ac expression and also attenuated mucus production in lung epithelial cells. To further corroborate these findings, we searched for association of single nucleotide polymorphisms in Nur77 gene with asthma and with the severity of bronchial hyperresponsiveness. We identified three Nur77 single nucleotide polymorphisms showing association with severity of bronchial hyperresponsiveness in asthma patients. Collectively, these findings support a protective role of Nur77 in OVA-induced airway inflammation and identify Nur77 as a novel therapeutic target for airway inflammation.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Inflamación/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Hipersensibilidad Respiratoria/metabolismo , Alelos , Animales , Asma/genética , Asma/inmunología , Asma/metabolismo , Asma/patología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Eosinófilos/patología , Expresión Génica , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Recuento de Leucocitos , Ganglios Linfáticos/inmunología , Ratones , Ratones Noqueados , Mucina 5AC/genética , Mucina 5AC/metabolismo , FN-kappa B/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Transducción de Señal
8.
Eur J Immunol ; 45(11): 3098-106, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26300344

RESUMEN

LIM-only protein FHL2 is associated with several immune and inflammatory diseases such as arthritis, influenza A virus infection, and lung inflammation. However, the role of FHL2 in macrophage differentiation and in the development of granuloma formation is unknown. Here, we show that expression of FHL2 is induced in mouse bone marrow derived macrophages (BMMs) following stimulation with M2 cytokines such as IL-4 and IL-10. FHL2-knockout (FHL2-KO) BMMs exhibit a proinflammatory M1 phenotype after LPS treatment and display a reduced anti-inflammatory M2 phenotype following IL-4 treatment. Furthermore, thioglycollate-induced migration of macrophages and B cells is enhanced in FHL2-KO mice. To evaluate the importance of FHL2 in the development of pulmonary granuloma formation, FHL2-KO mice were challenged with Schistosoma mansoni eggs. FHL2-KO mice show an enhanced number of granulomas and display decreased expression of Th2 markers and an exacerbated Th1 type of inflammation, characterized by enhanced expression of neutrophil markers and Th1 cytokines. Furthermore, the expression of barrier proteins is reduced in FHL2-KO lung compared to WT. Collectively, these data identify a previously unrecognized role for FHL2 in the pathogenesis of pulmonary granulomatous inflammation, partly through its effect on macrophage polarization, modulation of the Th1/Th2 balance and regulation of permeability in lung.


Asunto(s)
Granuloma del Sistema Respiratorio/inmunología , Proteínas con Homeodominio LIM/inmunología , Proteínas Musculares/inmunología , Esquistosomiasis mansoni/inmunología , Factores de Transcripción/inmunología , Animales , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Granuloma del Sistema Respiratorio/patología , Inmunohistoquímica , Inflamación/inmunología , Inflamación/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Esquistosomiasis mansoni/patología
9.
J Immunol ; 192(9): 4370-8, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24670805

RESUMEN

Azathioprine and its metabolite 6-mercaptopurine (6-MP) are well established immunosuppressive drugs. Common understanding of their immunosuppressive properties is largely limited to immune cells. However, in this study, the mechanism underlying the protective role of 6-MP in endothelial cell activation is investigated. Because 6-MP and its derivative 6-thioguanosine-5'-triphosphate (6-T-GTP) were shown to block activation of GTPase Rac1 in T lymphocytes, we focused on Rac1-mediated processes in endothelial cells. Indeed, 6-MP and 6-T-GTP decreased Rac1 activation in endothelial cells. As a result, the compounds inhibited TNF-α-induced downstream signaling via JNK and reduced activation of transcription factors c-Jun, activating transcription factor-2 and, in addition, NF κ-light-chain-enhancer of activated B cells (NF-κB), which led to decreased transcription of proinflammatory cytokines. Moreover, 6-MP and 6-T-GTP selectively decreased TNF-α-induced VCAM-1 but not ICAM-1 protein levels. Rac1-mediated generation of cell membrane protrusions, which form docking structures to capture leukocytes, also was reduced by 6-MP/6-T-GTP. Consequently, leukocyte transmigration was inhibited after 6-MP/6-T-GTP treatment. These data underscore the anti-inflammatory effect of 6-MP and 6-T-GTP on endothelial cells by blocking Rac1 activation. Our data provide mechanistic insight that supports development of novel Rac1-specific therapeutic approaches against chronic inflammatory diseases.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Inmunosupresores/farmacología , Mercaptopurina/farmacología , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , Western Blotting , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Humanos , Microscopía Confocal , Microscopía Electrónica de Rastreo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Transcriptoma , Migración Transendotelial y Transepitelial/efectos de los fármacos
10.
Circ Res ; 110(3): 428-38, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22194623

RESUMEN

RATIONALE: Nuclear receptor Nur77, also known as NR4A1, TR3, or NGFI-B, is expressed in human atherosclerotic lesions in macrophages, endothelial cells, T cells and smooth muscle cells. Macrophages play a critical role in atherosclerosis and the function of Nur77 in lesion macrophages has not yet been investigated. OBJECTIVE: This study aims to delineate the function of Nur77 in macrophages and to assess the effect of bone marrow-specific deficiency of Nur77 on atherosclerosis. METHODS AND RESULTS: We investigated Nur77 in macrophage polarization using bone marrow-derived macrophages (BMM) from wild-type and Nur77-knockout (Nur77(-/-)) mice. Nur77(-/-) BMM exhibit changed expression of M2-specific markers and an inflammatory M1-phenotype with enhanced expression of interleukin-12, IFNγ, and SDF-1α and increased NO synthesis in (non)-stimulated Nur77(-/-) BMM cells. SDF-1α expression in nonstimulated Nur77(-/-) BMM is repressed by Nur77 and the chemoattractive activity of Nur77(-/-) BMM is abolished by SDF-1α inhibiting antibodies. Furthermore, Nur77(-/-) mice show enhanced thioglycollate-elicited migration of macrophages and B cells. The effect of bone marrow-specific deficiency of Nur77 on atherosclerosis was studied in low density lipoprotein receptor-deficient (Ldlr(-/-)) mice. Ldlr(-/-) mice with a Nur77(-/-)-deficient bone marrow transplant developed 2.1-fold larger atherosclerotic lesions than wild-type bone marrow-transplanted mice. These lesions contain more macrophages, T cells, smooth muscle cells and larger necrotic cores. SDF-1α expression is higher in lesions of Nur77(-/-)-transplanted mice, which may explain the observed aggravation of lesion formation. CONCLUSIONS: In conclusion, in bone marrow-derived cells the nuclear receptor Nur77 has an anti-inflammatory function, represses SDF-1α expression and inhibits atherosclerosis.


Asunto(s)
Aterosclerosis/patología , Aterosclerosis/fisiopatología , Macrófagos/metabolismo , Macrófagos/patología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Animales , Aterosclerosis/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Quimiocina CXCL12/metabolismo , Colesterol/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/patología , Inflamación/fisiopatología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Fenotipo
11.
Circulation ; 121(18): 2023-32, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20421523

RESUMEN

BACKGROUND: Restenosis is the major drawback of percutaneous coronary interventions involving excessive activation and proliferation of vascular smooth muscle cells (SMCs). The nuclear receptor Nurr1 is an early response gene known mainly for its critical role in the development of dopamine neurons. In the present study, we investigated Nurr1 in human and experimental vascular restenosis. METHODS AND RESULTS: In a prospective cohort of 601 patients undergoing percutaneous coronary intervention, including stent placement, we found a strong association between Nurr1 haplotypes and in-stent restenosis risk. Furthermore, Nurr1 is specifically expressed in human in-stent restenosis and induced in cultured human SMCs in response to serum or tumor necrosis factor-alpha. Lentivirus-mediated gain- and loss-of-function experiments in SMCs demonstrated that overexpression of Nurr1 inhibited proliferation, consistent with increased expression of the key cell-cycle inhibitor p27(Kip1), whereas Nurr1 silencing enhanced SMC growth. The tumor necrosis factor-alpha-induced proinflammatory response of SMCs is inhibited by Nurr1, as reflected by reduced interleukin-1beta, tumor necrosis factor-alpha, and monocyte chemoattractant protein-1 expression. Consistent with our in vitro data, endogenous Nurr1 reduced wire injury-induced proliferation and vascular lesion formation in carotid arteries of ApoE(-/-) mice. CONCLUSIONS: Nurr1 haplotypes are associated with human restenosis risk, and Nurr1 is expressed in human in-stent restenosis. In SMCs, Nurr1 inhibits proliferation and inflammatory responses, which explains the inhibition of SMC-rich lesion formation in mice. The recently identified small-molecule drugs that enhance the activity of Nurr1 reveal this nuclear receptor as an attractive novel target for (local) intervention in restenosis.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Reestenosis Coronaria/genética , Músculo Liso Vascular/patología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Vasculitis/genética , Angioplastia Coronaria con Balón , Animales , Apolipoproteínas E/genética , División Celular/fisiología , Células Cultivadas , Enfermedad de la Arteria Coronaria/epidemiología , Enfermedad de la Arteria Coronaria/terapia , Reestenosis Coronaria/epidemiología , Reestenosis Coronaria/patología , Vasos Coronarios/inmunología , Vasos Coronarios/patología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Haplotipos , Humanos , Desequilibrio de Ligamiento , Ratones , Ratones Mutantes , Músculo Liso Vascular/fisiología , Neovascularización Fisiológica/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Factores de Riesgo , Stents , Vasculitis/epidemiología , Vasculitis/patología
12.
Arterioscler Thromb Vasc Biol ; 30(5): 931-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20167657

RESUMEN

OBJECTIVE: The iminosugar N-(5'-adamantane-1'-yl-methoxy)-pentyl-1-deoxynoijirimycin (AMP-DNM), an inhibitor of the enzyme glucosylceramide synthase catalyzing glycosphingolipid (GSL) biosynthesis, ameliorates diabetes and reduces liver steatosis in ob/ob mice. Because an accumulation of sphingolipids, including sphingomyelin and GSLs, has been reported in atherosclerotic lesions in animal models and in humans, the objective of this study was to determine whether AMP-DNM also exerts beneficial effects on the development of atherosclerosis. METHODS AND RESULTS: APOE*3 Leiden mice, maintained on a high-cholesterol diet, were treated for up to 18 weeks with AMP-DNM. The iminosugar prevented hyperlipidemia, generated a less atherogenic lipid profile, and induced a dramatic reduction in the development of atherosclerotic lesions. At the highest dose, no lesions were detectable. The effect of AMP-DNM was associated with a decrease in liver cholesterol, an increase in bile secretion, and enhanced excretion of cholesterol in the feces. Similar effects of AMP-DNM were observed in mice deficient for the low-density lipoprotein receptor. CONCLUSION: By lowering plasma cholesterol, the iminosugar AMP-DNM dramatically reduces the development of atherosclerosis in APOE*3 Leiden and low-density lipoprotein receptor -/- mice. Thus, targeting GSL synthesis may be a new treatment modality to prevent cardiovascular disease.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Adamantano/análogos & derivados , Apolipoproteínas E/genética , Aterosclerosis/prevención & control , Colesterol/sangre , Inhibidores Enzimáticos/farmacología , Glucosiltransferasas/antagonistas & inhibidores , Glicoesfingolípidos/biosíntesis , Receptores de LDL/deficiencia , 1-Desoxinojirimicina/farmacología , Adamantano/farmacología , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Bilis/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Heces/química , Femenino , Glucosiltransferasas/metabolismo , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Hiperlipidemias/prevención & control , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , Receptores de LDL/genética
13.
Arterioscler Thromb Vasc Biol ; 30(8): 1591-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20413732

RESUMEN

OBJECTIVE: 6-Mercaptopurine (6-MP), the active metabolite of the immunosuppressive prodrug azathioprine, is commonly used in autoimmune diseases and transplant recipients, who are at high risk for cardiovascular disease. Here, we aimed to gain knowledge on the action of 6-MP in atherosclerosis, with a focus on monocytes and macrophages. METHODS AND RESULTS: We demonstrate that 6-MP induces apoptosis of THP-1 monocytes, involving decreased expression of the intrinsic antiapoptotic factors B-cell CLL/Lymphoma-2 (Bcl-2) and Bcl2-like 1 (Bcl-x(L)). In addition, we show that 6-MP decreases expression of the monocyte adhesion molecules platelet endothelial adhesion molecule-1 (PECAM-1) and very late antigen-4 (VLA-4) and inhibits monocyte adhesion. Screening of a panel of cytokines relevant to atherosclerosis revealed that 6-MP robustly inhibits monocyte chemoattractant chemokine-1 (MCP-1) expression in macrophages stimulated with lipopolysaccharide (LPS). Finally, local delivery of 6-MP to the vessel wall, using a drug-eluting cuff, attenuates atherosclerosis in hypercholesterolemic apolipoprotein E*3-Leiden transgenic mice (P<0.05). In line with our in vitro data, this inhibition of atherosclerosis by 6-MP was accompanied with decreased lesion monocyte chemoattractant chemokine-1 levels, enhanced vascular apoptosis, and reduced macrophage content. CONCLUSIONS: We report novel, previously unrecognized atheroprotective actions of 6-MP in cultured monocytes/macrophages and in a mouse model of atherosclerosis, providing further insight into the effect of the immunosuppressive drug azathioprine in atherosclerosis.


Asunto(s)
Apolipoproteína E3/metabolismo , Aterosclerosis/prevención & control , Inmunosupresores/farmacología , Macrófagos/efectos de los fármacos , Mercaptopurina/farmacología , Monocitos/efectos de los fármacos , Animales , Apolipoproteína E3/genética , Apoptosis/efectos de los fármacos , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiotaxis/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Inmunosupresores/administración & dosificación , Mediadores de Inflamación/metabolismo , Integrina alfa4beta1/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Mercaptopurina/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factores de Tiempo , Proteína bcl-X/metabolismo
14.
Metabolism ; 121: 154815, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34119536

RESUMEN

OBJECTIVE: Four-and-a-Half-LIM-domain-protein 2 (FHL2) modulates multiple signal transduction pathways but has not been implicated in obesity or energy metabolism. In humans, methylation and expression of the FHL2 gene increases with age, and high FHL2 expression is associated with increased body weight in humans and mice. This led us to hypothesize that FHL2 is a determinant of diet-induced obesity. METHODS: FHL2-deficient (FHL2-/-) and wild type male mice were fed a high-fat diet. Metabolic phenotyping of these mice, as well as transcriptional analysis of key metabolic tissues was performed. Correlation of the expression of FHL2 and relevant genes was assessed in datasets from white adipose tissue of individuals with and without obesity. RESULTS: FHL2 Deficiency protects mice from high-fat diet-induced weight gain, whereas glucose handling is normal. We observed enhanced energy expenditure, which may be explained by a combination of changes in multiple tissues; mild activation of brown adipose tissue with increased fatty acid uptake, increased cardiac glucose uptake and browning of white adipose tissue. Corroborating our findings in mice, expression of FHL2 in human white adipose tissue positively correlates with obesity and negatively with expression of browning-associated genes. CONCLUSION: Our results position FHL2 as a novel regulator of obesity and energy expenditure in mice and human. Given that FHL2 expression increases during aging, we now show that low FHL2 expression associates with a healthy metabolic state.


Asunto(s)
Proteínas con Homeodominio LIM/genética , Proteínas Musculares/genética , Obesidad/genética , Factores de Transcripción/genética , Adipogénesis/genética , Tejido Adiposo Blanco/metabolismo , Adulto , Anciano , Animales , Biomarcadores/metabolismo , Dieta Alta en Grasa , Femenino , Predisposición Genética a la Enfermedad , Humanos , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteínas Musculares/metabolismo , Obesidad/diagnóstico , Obesidad/metabolismo , Factores de Transcripción/metabolismo , Aumento de Peso/genética
15.
Cardiovasc Pathol ; 38: 1-6, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30359839

RESUMEN

Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the fibrillin-1 gene (FBN1), resulting in aortic aneurysm formation and dissections. Interestingly, variable aortopathy is observed even within MFS families with the same mutation. Thus, additional risk factors determine disease severity. Here, we describe a case of a 2-month-old Fbn1C1039G/+ MFS mouse with extreme aortic dilatation and increased vascular inflammation, when compared to MFS siblings, which coincided with unilateral renal cystic disease. In addition, this mouse presented with increased serum levels of creatinine, angiotensin-converting enzyme, corticosterone, macrophage chemoattractant protein-1, and interleukin-6, which may have contributed to the vascular pathology. Possibly, cystic kidney disease is associated with aneurysm progression in MFS patients. Therefore, we propose that close monitoring of the presence of renal cysts in MFS patients, during regular vascular imaging of the whole aorta trajectory, may provide insight in the frequency of cystic kidney disease and its potential as a novel indicator of aneurysm progression in MFS patients.


Asunto(s)
Aorta/patología , Aneurisma de la Aorta/etiología , Fibrilina-1/genética , Enfermedades Renales Quísticas/etiología , Síndrome de Marfan/genética , Animales , Aorta/metabolismo , Aneurisma de la Aorta/sangre , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/patología , Aortitis/sangre , Aortitis/etiología , Aortitis/genética , Aortitis/patología , Biomarcadores/sangre , Dilatación Patológica , Modelos Animales de Enfermedad , Fibrilina-1/metabolismo , Predisposición Genética a la Enfermedad , Enfermedades Renales Quísticas/sangre , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Masculino , Síndrome de Marfan/sangre , Síndrome de Marfan/complicaciones , Síndrome de Marfan/diagnóstico , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo
16.
Circulation ; 115(4): 493-500, 2007 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17242285

RESUMEN

BACKGROUND: Restenosis is a common complication after percutaneous coronary interventions and is characterized by excessive proliferation of vascular smooth muscle cells (SMCs). We have shown that the nuclear receptor Nur77 protects against SMC-rich lesion formation, and it has been demonstrated that 6-mercaptopurine (6-MP) enhances Nur77 activity. We hypothesized that 6-MP inhibits neointima formation through activation of Nur77. METHODS AND RESULTS: It is demonstrated that 6-MP increases Nur77 activity in cultured SMCs, which results in reduced [3H]thymidine incorporation, whereas Nur77 small interfering RNA knockdown partially restores DNA synthesis. Furthermore, we studied the effect of 6-MP in a murine model of cuff-induced neointima formation. Nur77 mRNA is upregulated in cuffed arteries, with optimal expression after 6 hours and elevated expression up to 7 days after vascular injury. Local perivascular delivery of 6-MP with a drug-eluting cuff significantly inhibits neointima formation in wild-type mice. Locally applied 6-MP does not affect inflammatory responses or apoptosis but inhibits expression of proliferating cell nuclear antigen and enhances protein levels of the cell-cycle inhibitor p27(Kip1) in the vessel wall. An even stronger inhibition of neointima formation in response to local 6-MP delivery was observed in transgenic mice that overexpressed Nur77. In contrast, 6-MP does not alter lesion formation in transgenic mice that overexpress a dominant-negative variant of Nur77 in arterial SMCs, which provides evidence for the involvement of Nur77-like factors. CONCLUSIONS: Enhancement of the activity of Nur77 by 6-MP protects against excessive SMC proliferation and SMC-rich neointima formation. We propose that activation of the nuclear receptor Nur77 is a rational approach to treating in-stent restenosis.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Reestenosis Coronaria/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Mercaptopurina/farmacología , Músculo Liso Vascular/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Reestenosis Coronaria/metabolismo , Reestenosis Coronaria/patología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Implantes de Medicamentos , Arteria Femoral/patología , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Músculo Liso Vascular/patología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Esteroides/genética , Factores de Transcripción/genética , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Arterias Umbilicales/citología
17.
Biochem Biophys Res Commun ; 366(4): 910-6, 2008 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-18086558

RESUMEN

NR4A nuclear receptors are induced in the liver upon fasting and regulate hepatic gluconeogenesis. Here, we studied the role of nuclear receptor Nur77 (NR4A1) in hepatic lipid metabolism. We generated mice expressing hepatic Nur77 using adenoviral vectors, and demonstrate that these mice exhibit a modulation of the plasma lipid profile and a reduction in hepatic triglyceride. Expression analysis of >25 key genes involved in lipid metabolism revealed that Nur77 inhibits SREBP1c expression. This results in decreased SREBP1c activity as is illustrated by reduced expression of its target genes stearoyl-coA desaturase-1, mitochondrial glycerol-3-phosphate acyltransferase, fatty acid synthase and the LDL receptor, and provides a mechanism for the physiological changes observed in response to Nur77. Expression of LXR target genes Abcg5 and Abcg8 is reduced by Nur77, and may suggest involvement of LXR in the inhibitory action of Nur77 on SREBP1c expression. Taken together, our study demonstrates that Nur77 modulates hepatic lipid metabolism through suppression of SREBP1c activity.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Adenoviridae , Animales , Regulación de la Expresión Génica , Hígado/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , ARN Mensajero/genética , ARN Mensajero/metabolismo , Triglicéridos/sangre
18.
PLoS One ; 13(1): e0190962, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29346401

RESUMEN

An abdominal aortic aneurysm (AAA) is a dilatation of the abdominal aorta leading to serious complications and mostly to death. AAA development is associated with an accumulation of inflammatory cells in the aorta including NKT cells. An important factor in promoting the recruitment of these inflammatory cells into tissues and thereby contributing to the development of AAA is angiotensin II (Ang II). We demonstrate that a deficiency in CD1d dependent NKT cells under hyperlipidemic conditions (LDLr-/-CD1d-/- mice) results in a strong decline in the severity of angiotensin II induced aneurysm formation when compared with LDLr-/- mice. In addition, we show that Ang II amplifies the activation of NKT cells both in vivo and in vitro. We also provide evidence that type I NKT cells contribute to AAA development by inducing the expression of matrix degrading enzymes in vSMCs and macrophages, and by cytokine dependently decreasing vSMC viability. Altogether, these data prove that CD1d-dependent NKT cells contribute to AAA development in the Ang II-mediated aneurysm model by enhancing aortic degradation, establishing that therapeutic applications which target NKT cells can be a successful way to prevent AAA development.


Asunto(s)
Antígenos CD1d/genética , Aneurisma de la Aorta Abdominal/prevención & control , Receptores de LDL/genética , Angiotensina II/administración & dosificación , Animales , Apoptosis/inmunología , Citometría de Flujo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/patología , Células 3T3 NIH , Células T Asesinas Naturales/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Cell Rep ; 24(8): 2127-2140.e7, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30134173

RESUMEN

Activation of macrophages by inflammatory stimuli induces reprogramming of mitochondrial metabolism to support the production of pro-inflammatory cytokines and nitric oxide. Hallmarks of this metabolic rewiring are downregulation of α-ketoglutarate formation by isocitrate dehydrogenase (IDH) and accumulation of glutamine-derived succinate, which enhances the inflammatory response via the activity of succinate dehydrogenase (SDH). Here, we identify the nuclear receptor Nur77 (Nr4a1) as a key upstream transcriptional regulator of this pro-inflammatory metabolic switch in macrophages. Nur77-deficient macrophages fail to downregulate IDH expression and accumulate higher levels of succinate and other TCA cycle-derived metabolites in response to inflammatory stimulation in a glutamine-independent manner. Consequently, these macrophages produce more nitric oxide and pro-inflammatory cytokines in an SDH-dependent manner. In vivo, bone marrow Nur77 deficiency exacerbates atherosclerosis development and leads to increased circulating succinate levels. In summary, Nur77 induces an anti-inflammatory metabolic state in macrophages that protects against chronic inflammatory diseases such as atherosclerosis.


Asunto(s)
Regulación de la Expresión Génica/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Humanos
20.
Psychoneuroendocrinology ; 32(8-10): 1144-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17850983

RESUMEN

Saliva for measurement of cortisol is generally sampled by swabbing the mouth with a cotton roll, but this method has drawbacks. In the present study, we evaluated the use of an eye sponge as an oral collection device for saliva cortisol. The eye sponge was compared with commercial cotton rolls, and tested for use in infants as well as adults. Our results show that the eye sponge has adequate cortisol recoveries, even after samples have been kept at 4-8 degrees C for up to a week. In adults, volumes of 200-250 microl are obtained without problem; although smaller volumes are obtained in young infants, they are sufficient for assays requiring only 50-100 microl of saliva. In conclusion, the eye sponge is a valid and adequate collection device for saliva cortisol. Additional advantages as compared to cotton rolls are: more comfortable sampling, tastelessness, no need to manipulate the absorbing material, and the ease with which the untrained eye can determine that enough saliva has been collected.


Asunto(s)
Hidrocortisona/análisis , Saliva , Manejo de Especímenes/instrumentación , Adulto , Fibra de Algodón , Humanos , Lactante , Saliva/química , Manejo de Especímenes/métodos , Tapones Quirúrgicos de Gaza
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA