Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 19588, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37949969

RESUMEN

Arterial macrophage foam cells are filled with cholesterol ester (CE) stored in cytosolic lipid droplets (LDs). Foam cells are central players in progression of atherosclerosis as regulators of lipid metabolism and inflammation, two major driving forces of atherosclerosis development. Thus, foam cells are considered plausible targets for intervention in atherosclerosis. However, a compound that directly regulates the lipid metabolism of LDs in the arterial foam cells has not yet been identified. In this study, we screened compounds that inhibit macrophage foam cell formation using a library of 2697 FDA-approved drugs. From the foam cells generated via loading of human oxidized low-density lipoprotein (oxLDL), we found 21 and 6 compounds that reduced and enhanced accumulations of lipids respectively. Among them, verteporfin most significantly reduced oxLDL-induced foam cell formation whereas it did not display a significant impact on foam cell formation induced by fatty acid. Mechanistically our data demonstrate that verteporfin acts via inhibition of oxLDL association with macrophages, reducing accumulation of CE. Interestingly, while other drugs that reduced foam cell formation did not have impact on pre-existing foam cells, verteporfin treatment significantly reduced their total lipids, CE, and pro-inflammatory gene expression. Together, our study identifies verteporfin as a novel regulator of foam cell lipid metabolism and inflammation and a potential compound for intervention in atherosclerosis.


Asunto(s)
Aterosclerosis , Células Espumosas , Humanos , Células Espumosas/metabolismo , Verteporfina/farmacología , Verteporfina/metabolismo , Metabolismo de los Lípidos , Macrófagos/metabolismo , Aterosclerosis/metabolismo , Lipoproteínas LDL/metabolismo , Ésteres del Colesterol/metabolismo , Inflamación/metabolismo
2.
J Am Heart Assoc ; 10(2): e018151, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33401929

RESUMEN

Background Despite compelling epidemiological evidence that circadian disruption inherent to long-term shift work enhances atherosclerosis progression and vascular events, the underlying mechanisms remain poorly understood. A challenge to the use of mouse models for mechanistic and interventional studies involving light-dark patterns is that the spectral and absolute sensitivities of the murine and human circadian systems are very different, and light stimuli in nocturnal mice should be scaled to represent the sensitivities of the human circadian system. Methods and Results We used calibrated devices to deliver to low-density lipoprotein receptor knockout mice light-dark patterns representative of that experienced by humans working day shifts or rotating shift schedules. Mice under day shifts were maintained under regular 12 hours of light and 12 hours of dark cycles. Mice under rotating shift schedules were subjected for 11 weeks to reversed light-dark patterns 4 days in a row per week, followed by 3 days of regular light-dark patterns. In both protocols the light phases consisted of monochromatic green light at an irradiance of 4 µW/cm2. We found that the shift work paradigm disrupts the foam cell's molecular clock and increases endoplasmic reticulum stress and apoptosis. Lesions of mice under rotating shift schedules were larger and contained less prostabilizing fibrillar collagen and significantly increased areas of necrosis. Conclusions Low-density lipoprotein receptor knockout mice under light-dark patterns analogous to that experienced by rotating shift workers develop larger and more vulnerable plaques and may represent a valuable model for further mechanistic and/or interventional studies against the deleterious vascular effects of rotating shift work.


Asunto(s)
Apoptosis/fisiología , Aterosclerosis , Relojes Circadianos/fisiología , Estrés del Retículo Endoplásmico/fisiología , Células Espumosas , Placa Aterosclerótica , Horario de Trabajo por Turnos , Animales , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Ritmo Circadiano/fisiología , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Lipoproteínas LDL/genética , Ratones , Ratones Noqueados , Modelos Animales , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
3.
Nat Chem Biol ; 16(2): 206-213, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932720

RESUMEN

Genetic screens in cultured human cells represent a powerful unbiased strategy to identify cellular pathways that determine drug efficacy, providing critical information for clinical development. We used insertional mutagenesis-based screens in haploid cells to identify genes required for the sensitivity to lasonolide A (LasA), a macrolide derived from a marine sponge that kills certain types of cancer cells at low nanomolar concentrations. Our screens converged on a single gene, LDAH, encoding a member of the metabolite serine hydrolase family that is localized on the surface of lipid droplets. Mechanistic studies revealed that LasA accumulates in lipid droplets, where it is cleaved into a toxic metabolite by LDAH. We suggest that selective partitioning of hydrophobic drugs into the oil phase of lipid droplets can influence their activation and eventual toxicity to cells.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Gotas Lipídicas/metabolismo , Macrólidos/farmacocinética , Macrólidos/toxicidad , Proteínas/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Haploidia , Humanos , Inactivación Metabólica , Gotas Lipídicas/efectos de los fármacos , Macrólidos/metabolismo , Proteínas/genética
4.
Int J Mol Sci ; 20(15)2019 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-31382484

RESUMEN

Arterial foam cells are central players of atherogenesis. Cholesterol acceptors, apolipoprotein A-I (apoA-I) and high-density lipoprotein (HDL), take up cholesterol and phospholipids effluxed from foam cells into the circulation. Due to the high abundance of cholesterol in foam cells, most previous studies focused on apoA-I/HDL-mediated free cholesterol (FC) transport. However, recent lipidomics of human atherosclerotic plaques also identified that oxidized sterols (oxysterols) and non-sterol lipid species accumulate as atherogenesis progresses. While it is known that these lipids regulate expression of pro-inflammatory genes linked to plaque instability, how cholesterol acceptors impact the foam cell lipidome, particularly oxysterols and non-sterol lipids, remains unexplored. Using lipidomics analyses, we found cholesterol acceptors remodel foam cell lipidomes. Lipid subclass analyses revealed various oxysterols, sphingomyelins, and ceramides, species uniquely enriched in human plaques were significantly reduced by cholesterol acceptors, especially by apoA-I. These results indicate that the function of lipid-poor apoA-I is not limited to the efflux of cholesterol and phospholipids but suggest that apoA-I serves as a major regulator of the foam cell lipidome and might play an important role in reducing multiple lipid species involved in the pathogenesis of atherosclerosis.


Asunto(s)
Colesterol/metabolismo , Células Espumosas/metabolismo , Placa Aterosclerótica/metabolismo , Animales , Apolipoproteína A-I/metabolismo , Aterosclerosis/metabolismo , Células Cultivadas , Humanos , Lipidómica , Lipoproteínas LDL/metabolismo , Masculino , Ratones Endogámicos C57BL , Oxiesteroles/metabolismo
5.
Sci Rep ; 7(1): 2743, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28578400

RESUMEN

Lipid droplet (LD)-associated hydrolase (LDAH) is a newly identified LD protein abundantly expressed in tissues that predominantly store triacylglycerol (TAG). However, how LDAH regulates TAG metabolism remains unknown. We found that upon oleic acid loading LDAH translocalizes from the ER to newly formed LDs, and induces LD coalescence in a tubulin-dependent manner. LDAH overexpression and downregulation in HEK293 cells increase and decrease, respectively, TAG levels. Pulse and chase experiments show that LDAH enhances TAG biogenesis, but also decreases TAG turnover and fatty acid release from cells. Mutations in predicted catalytic and acyltransferase motifs do not influence TAG levels, suggesting that the effect is independent of LDAH's enzymatic activity. However, a LDAH alternative-splicing variant missing 90 amino acids at C-terminus does not promote LD fusion or TAG accumulation, while it still localizes to LDs. Interestingly, LDAH enhances polyubiquitination and proteasomal degradation of adipose triglyceride lipase (ATGL), a rate limiting enzyme of TAG hydrolysis. Co-expression of ATGL reverses the changes in LD phenotype induced by LDAH, and both proteins counterbalance their effects on TAG stores. Together, these studies support that under conditions of TAG storage in LDs LDAH plays a primarily lipogenic role, inducing LD growth and enhancing degradation of ATGL.


Asunto(s)
Lipasa/metabolismo , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/genética , Triglicéridos/metabolismo , Empalme Alternativo/genética , Ácidos Grasos/metabolismo , Células HEK293 , Células HeLa , Humanos , Hidrólisis , Lipasa/genética , Mutación , Ácido Oléico/metabolismo , Triglicéridos/genética
6.
Genom Data ; 9: 37-9, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27408807

RESUMEN

Hypercholesterolemia is a main risk factor for atherosclerosis development. Arterial macrophages, or foam cells, take-up and process lipoprotein particles deposited in arteries, and store much of the cholesterol carried by these particles in their cytoplasm. However, the effects of exposure to different cholesterol levels on foam cells remain poorly understood. Given the remarkable plasticity of macrophages in response to environmental variables, studies on macrophage biology should ideally be performed in the environment where they exert their physiological functions, namely atherosclerotic lesions in the case of foam cells. We used a mouse model of atherosclerosis, the apolipoprotein E-deficient mouse, to study in vivo the transcriptional response of foam cells to short- and long-term elevations in plasma cholesterol, induced by feeding mice a western type diet. The microarray data sets from this study have been deposited in NCBI's Gene Expression Omnibus under the accession number GSE70619. Here we provide detailed information on the experimental set-up, on the isolation of RNA by laser capture microdissection, and on the methodology used for RNA amplification and analysis by microarray and quantitative real-time PCR.

7.
Cell Rep ; 15(1): 158-170, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-27052172

RESUMEN

The cardiac trabeculae are sheet-like structures extending from the myocardium that function to increase surface area. A lack of trabeculation causes embryonic lethality due to compromised cardiac function. To understand the cellular and molecular mechanisms of trabecular formation, we genetically labeled individual cardiomyocytes prior to trabeculation via the brainbow multicolor system and traced and analyzed the labeled cells during trabeculation by whole-embryo clearing and imaging. The clones derived from labeled single cells displayed four different geometric patterns that are derived from different patterns of oriented cell division (OCD) and migration. Of the four types of clones, the inner, transmural, and mixed clones contributed to trabecular cardiomyocytes. Further studies showed that perpendicular OCD is an extrinsic asymmetric cell division that putatively contributes to trabecular regional specification. Furthermore, N-Cadherin deletion in labeled clones disrupted the clonal patterns. In summary, our data demonstrate that OCD contributes to trabecular morphogenesis and specification.


Asunto(s)
División Celular Asimétrica , Linaje de la Célula , Corazón/embriología , Morfogénesis , Miocitos Cardíacos/citología , Animales , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Movimiento Celular , Femenino , Masculino , Ratones , Miocitos Cardíacos/metabolismo
8.
Mol Med Rep ; 13(6): 4527-34, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27082419

RESUMEN

Accumulation of atherosclerotic plaques in arterial walls leads to major cardiovascular diseases and stroke. Macrophages/foam cells are central components of atherosclerotic plaques, which populate the arterial wall in order to remove harmful modified low­density lipoprotein (LDL) particles, resulting in the accumulation of lipids, mostly LDL­derived cholesterol ester, in cytosolic lipid droplets (LDs). At present, LDs are recognized as dynamic organelles that govern cellular metabolic processes. LDs consist of an inner core of neutral lipids surrounded by a monolayer of phospholipids and free cholesterol, and contain LD­associated proteins (LDAPs) that regulate LD functions. Foam cells are characterized by an aberrant accumulation of cytosolic LDs, and are considered a hallmark of atherosclerotic lesions through all stages of development. Previous studies have investigated the mechanisms underlying foam cell formation, aiming to discover therapeutic strategies that target foam cells and intervene against atherosclerosis. It is well established that LDAPs have a major role in the pathogenesis of metabolic diseases caused by dysfunction of lipid metabolism, and several studies have linked LDAPs to the development of atherosclerosis. In this review, several foam cell­targeting pathways have been described, with an emphasis on the role of LDAPs in cholesterol mobilization from macrophages. In addition, the potential of LDAPs as therapeutic targets to prevent the progression and/or facilitate the regression of the disease has been discussed.


Asunto(s)
Aterosclerosis/etiología , Aterosclerosis/metabolismo , Proteínas Asociadas a Gotas Lipídicas/metabolismo , Animales , Aterosclerosis/patología , Transporte Biológico , Colesterol/metabolismo , Ésteres del Colesterol/metabolismo , Modelos Animales de Enfermedad , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Hidrólisis , Proteínas Asociadas a Gotas Lipídicas/genética , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
9.
J Am Heart Assoc ; 5(4): e002663, 2016 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-27091181

RESUMEN

BACKGROUND: Foam cells are central to two major pathogenic processes in atherogenesis: cholesterol buildup in arteries and inflammation. The main underlying cause of cholesterol deposition in arteries is hypercholesterolemia. This study aimed to assess, in vivo, whether elevated plasma cholesterol also alters the inflammatory balance of foam cells. METHODS AND RESULTS: Apolipoprotein E-deficient mice were fed regular mouse chow through the study or were switched to a Western-type diet (WD) 2 or 14 weeks before death. Consecutive sections of the aortic sinus were used for lesion quantification or to isolate RNA from foam cells by laser-capture microdissection (LCM) for microarray and quantitative polymerase chain reaction analyses. WD feeding for 2 or 14 weeks significantly increased plasma cholesterol, but the size of atherosclerotic lesions increased only in the 14-week WD group. Expression of more genes was affected in foam cells of mice under prolonged hypercholesterolemia than in mice fed WD for 2 weeks. However, most transcripts coding for inflammatory mediators remained unchanged in both WD groups. Among the main players in inflammatory or immune responses, chemokine (C-X-C motif) ligand 13 was induced in foam cells of mice under WD for 2 weeks. The interferon-inducible GTPases, guanylate-binding proteins (GBP)3 and GBP6, were induced in the 14-week WD group, and other GBP family members were moderately increased. CONCLUSIONS: Our results indicate that acceleration of atherosclerosis by hypercholesterolemia is not linked to global changes in the inflammatory balance of foam cells. However, induction of GBPs uncovers a novel family of immune modulators with a potential role in atherogenesis.


Asunto(s)
Aterosclerosis/etiología , Dieta Occidental/efectos adversos , Células Espumosas/química , Proteínas de Unión al GTP/análisis , Inflamación/fisiopatología , Animales , Aterosclerosis/inducido químicamente , Femenino , Células Espumosas/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Captura por Microdisección con Láser , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Seno Aórtico/química
10.
Cardiovasc Res ; 109(2): 294-304, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26487692

RESUMEN

AIMS: Atherosclerosis development can be ameliorated by promoting reverse cholesterol transport (RCT) from arteries. The process involves cholesterol efflux from foam cells to extracellular acceptors such as apolipoprotein A-I (apoA-I) and high-density lipoprotein (HDL) that mediate transport to the liver. Perilipin-2 (PLIN2) is a lipid droplet (LD)-associated protein that in macrophages facilitates cholesterol storage and prevents efflux. We hypothesized that atheroprotection would be enhanced by concurrently targeting PLIN2 to increase the efflux capacity of foam cells and increasing plasma apoA-I and HDL. METHODS AND RESULTS: PLIN2-knockout and wild-type mice lacking apolipoprotein E (PLIN2(-/-)/apoE(-/-) and PLIN2(+/+)/apoE(-/-)) were treated with a helper-dependent adenoviral vector encoding human apoA-I (HDAd-AI) or with control empty vector. Treatment with HDAd-AI increased hepatic apoA-I production, plasma apoA-I and HDL-cholesterol (HDL-C), and apoA-I deposition in lesions to a similar extent in PLIN2(-/-)/apoE(-/-) and PLIN2(+/+)/apoE(-/-) mice. However, atherosclerosis development at the aortic sinus was considerably lower in HDAd-AI-treated PLIN2(-/-)/apoE(-/-) mice. A more stable lesion phenotype, with increased collagen content, was primarily associated to treatment with HDAd-AI, but was enhanced under PLIN2 deficiency. PLIN2 deficiency and apoA-I cumulatively reduced LDs and cholesterol ester content in cultured macrophages. Neutral lipid in atheroma was significantly reduced in HDAd-AI-treated PLIN2(-/-)/apoE(-/-) mice, and RCT from macrophages to feces was enhanced in PLIN2(-/-) macrophages. CONCLUSION: These studies demonstrate a mutually beneficial relationship between PLIN2 deficiency and elevated apoA-I/HDL-C in preventing atherosclerosis development. The data support that targeting foam cell components to mobilize cholesterol may be a promising strategy to enhance the atheroprotection of plasma cholesterol acceptors.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Células Espumosas/metabolismo , Perilipina-2/metabolismo , Animales , Apolipoproteína A-I/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/genética , Transporte Biológico/genética , Transporte Biológico/fisiología , Ésteres del Colesterol/genética , Ésteres del Colesterol/metabolismo , Lipoproteínas HDL/metabolismo , Macrófagos/metabolismo , Ratones Noqueados , Perilipina-2/genética
11.
Arterioscler Thromb Vasc Biol ; 34(2): 386-96, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24357060

RESUMEN

OBJECTIVE: Lipid-laden macrophages or foam cells are characterized by massive cytosolic lipid droplet (LD) deposition containing mostly cholesterol ester (CE) derived from the lipoproteins cleared from the arterial wall. Cholesterol efflux from foam cells is considered to be atheroprotective. Because cholesterol is effluxed as free cholesterol, CE accumulation in LDs may limit free cholesterol efflux. Our objective was to identify proteins that regulate cholesterol trafficking through LDs. APPROACH AND RESULTS: In a proteomic analysis of the LD fraction of RAW 264.7 macrophages, we identified an evolutionarily conserved protein with a canonical GXSXG lipase catalytic motif and a predicted α/ß-hydrolase fold, the RIKEN cDNA 1110057K04 gene, which we named LD-associated hydrolase (LDAH). LDAH association with LDs was confirmed by immunoblotting and immunocytochemistry. LDAH was labeled with a probe specific for active serine hydrolases. LDAH showed relatively weak in vitro CE hydrolase activity. However, cholesterol measurements in intact cells supported a significant role of LDAH in CE homeostasis because LDAH upregulation and downregulation decreased and increased, respectively, intracellular cholesterol and CE in human embryonic kidney-293 cells and RAW 264.7 macrophages. Mutation of the putative nucleophilic serine impaired active hydrolase probe binding, in vitro CE hydrolase activity, and cholesterol-lowering effect in cells, whereas this mutant still localized to the LD. LDAH upregulation increased CE hydrolysis and cholesterol efflux from macrophages, and, interestingly, LDAH is highly expressed in macrophage-rich areas within mouse and human atherosclerotic lesions. CONCLUSIONS: The data identify a candidate target to promote reverse cholesterol transport from atherosclerotic lesions.


Asunto(s)
Colesterol/metabolismo , Células Espumosas/enzimología , Serina Proteasas/metabolismo , Secuencia de Aminoácidos , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Transporte Biológico , Ésteres del Colesterol/metabolismo , Modelos Animales de Enfermedad , Células Espumosas/patología , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Hidrólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Placa Aterosclerótica , Proteómica/métodos , Interferencia de ARN , Serina Proteasas/química , Serina Proteasas/genética , Especificidad por Sustrato , Factores de Tiempo , Transfección
12.
PLoS One ; 7(3): e33063, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427949

RESUMEN

Interventions on macrophages/foam cells to redirect intracellular cholesterol towards efflux pathways could become a very valuable addition to our therapeutic arsenal against atherosclerosis. However, certain manipulations of the cholesteryl ester cycle, such as the inhibition of ACAT1, an ER-resident enzyme that re-esterifies cholesterol, are not well tolerated. Previously we showed that targeting perilipin-2 (PLIN2), a major lipid droplet (LD)-associated protein in macrophages, prevents foam cell formation and protects against atherosclerosis. Here we have assessed the tolerance of PLIN2-deficient bone marrow derived macrophages (BMM) to several lipid loading conditions similar to the found during atherosclerosis development, including exposure to modified low-density lipoprotein (mLDL) and 7-ketocholesterol (7-KC), a free cholesterol (FC) metabolite, in media with or without cholesterol acceptors. BMM isolated from mice that do or do not express PLIN2 were tested for apoptosis (TUNEL and cleaved caspase-3), ER stress (CHOP induction and XBP-1 splicing), and inflammation (TNF-α and IL-6 mRNA levels). Like in other cell types, PLIN2 deficiency impairs LD buildup in BMM. However, while most stress parameters were elevated in macrophages under ACAT inhibition and 7-KC loading, PLIN2 inactivation was well tolerated. The data support the safety of targeting PLIN2 to prevent foam cell formation and atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , LDL-Colesterol/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/deficiencia , Análisis de Varianza , Animales , Apoptosis/fisiología , Aterosclerosis/prevención & control , Compuestos Azo , Western Blotting , Técnicas de Cultivo de Célula , LDL-Colesterol/toxicidad , Cartilla de ADN/genética , Estrés del Retículo Endoplásmico/fisiología , Células Espumosas/metabolismo , Etiquetado Corte-Fin in Situ , Interleucina-6/metabolismo , Cetocolesteroles/metabolismo , Ratones , Ratones Endogámicos C57BL , Perilipina-2 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo
13.
Eur Heart J ; 33(24): 3114-23, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21804111

RESUMEN

AIMS: Interleukin-7 (IL-7) is a master regulator of T-cell development and homoeostasis. Increased IL-7 levels are associated with inflammatory diseases. The aims of this study were to determine whether IL-7 is a biomarker for inflammatory conditions or an active participant in atherogenesis. METHODS AND RESULTS: Advanced atherosclerotic lesions in Apoe(-/-) mice were regressed by long-term cholesterol lowering through treatment with a helper-dependent adenovirus expressing apolipoprotein E (n= 6-10). Using this model, gene expression patterns in the aorta were analysed at an early phase of regression by microarray. After stringent statistical analysis, we found that IL-7 expression is significantly reduced in response to lowering of cholesterol (n= 6). To understand the importance of IL-7 down-regulation for atherosclerotic regression, we studied the effects and mechanisms of action of IL-7 on endothelial cells (ECs) in vitro as well as in vivo. Our major findings are: (i) IL-7 up-regulates cell adhesion molecules and monocyte chemoattractant protein-1 in ECs and promotes monocyte adhesion to ECs; (ii) this regulation is mediated by phosphatidylinositol 3-kinase (PI3K)/AKT-dependent and -independent activation of NF-κB; (iii) elevation of plasma IL-7 induces recruitment of monocytes/macrophages to endothelium without affecting plasma cholesterol (n= 5, 6); and (4) lack of IL-7 in bone marrow-derived cells reduces migration of monocytes/macrophages to the lesions (n= 5, 6). CONCLUSION: These results suggest that IL-7 inflames endothelium via PI3K/AKT-dependent and -independent activation of NF-κB and recruits monocytes/macrophages to the endothelium, thus playing an active role in atherogenesis.


Asunto(s)
Aterosclerosis/etiología , Endotelio Vascular/efectos de los fármacos , Interleucina-7/farmacología , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Adenoviridae , Animales , Aorta Torácica , Apolipoproteína E3/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/metabolismo , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Vectores Genéticos , Interleucina-7/fisiología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Regulación hacia Arriba
14.
Endocrinology ; 150(11): 4863-73, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19819964

RESUMEN

Insulin deficiency in type 1 diabetes leads to disruptions in glucose, lipid, and ketone metabolism with resultant hyperglycemia, hyperlipidemia, and ketonemia. Exogenous insulin and hepatic insulin gene therapy cannot mimic the robust glucose-stimulated insulin secretion (GSIS) from native pancreatic islets. Gene therapy of streptozotocin-diabetic mice with neurogenin 3 (Ngn3) and betacellulin (Btc) leads to the induction of periportal oval cell-derived neo-islets that exhibit GSIS. We hence hypothesized that this gene therapy regimen may lead to a complete correction of the glucose and lipid metabolic abnormalities associated with insulin deficiency; we further hypothesized that the neo-islets formed in response to Ngn3-Btc gene delivery may display an ultrastructure and transcription profile similar to that of pancreatic islets. We injected streptozotocin-diabetic mice with helper-dependent adenoviral vectors carrying Ngn3 and Btc, which restored GSIS and reversed hyperglycemia in these animals. The treatment also normalized hepatic glucose secretion and reversed ketonemia. Furthermore, it restored hepatic glycogen content and reinstated hepatic lipogenesis-related gene transcripts back to nondiabetic levels. By transmission electron microscopy, the neo-islets displayed electron-dense granules that were similar in appearance to those in pancreatic islets. Finally, using RNA obtained by laser capture microdissection of the periportal neo-islets and normal pancreatic islets, we found that the neo-islets and pancreatic islets exhibited a very similar transcription profile on microarray-based transcriptome analysis. Taken together, this indicates that Ngn3-Btc gene therapy corrects the underlying dysregulated glucose and lipid metabolism in insulin-deficient diabetic mice by inducing neo-islets in the liver that are similar to pancreatic islets in structure and gene expression profile.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/uso terapéutico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Terapia Genética , Insulina/deficiencia , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Proteínas del Tejido Nervioso/uso terapéutico , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Betacelulina , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
15.
Circulation ; 119(20): 2708-17, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19433759

RESUMEN

BACKGROUND: Monocyte activation and migration into the arterial wall are key events in atherogenesis associated with hypercholesterolemia. CD11c/CD18, a beta2 integrin expressed on human monocytes and a subset of mouse monocytes, has been shown to play a distinct role in human monocyte adhesion on endothelial cells, but the regulation of CD11c in hypercholesterolemia and its role in atherogenesis are unknown. METHODS AND RESULTS: Mice genetically deficient in CD11c were generated and crossbred with apolipoprotein E (apoE)-/- mice to generate CD11c-/-/apoE-/- mice. Using flow cytometry, we examined CD11c on blood leukocytes in apoE-/- hypercholesterolemic mice and found that compared with wild-type and apoE-/- mice on a normal diet, apoE-/- mice on a Western high-fat diet had increased CD11c+ monocytes. Circulating CD11c+ monocytes from apoE-/- mice fed a high-fat diet exhibited cytoplasmic lipid vacuoles and expressed higher levels of CD11b and CD29. Deficiency of CD11c decreased firm arrest of mouse monocytes on vascular cell adhesion molecule-1 and E-selectin in a shear flow assay, reduced monocyte/macrophage accumulation in atherosclerotic lesions, and decreased atherosclerosis development in apoE-/- mice on a high-fat diet. CONCLUSIONS: CD11c, which increases on blood monocytes during hypercholesterolemia, plays an important role in monocyte recruitment and atherosclerosis development in an apoE-/- mouse model of hypercholesterolemia.


Asunto(s)
Aterosclerosis/etiología , Antígeno CD11c/fisiología , Hipercolesterolemia/complicaciones , Monocitos/fisiología , Animales , Apolipoproteínas E/deficiencia , Antígeno CD11c/análisis , Antígeno CD11c/genética , Quimiotaxis de Leucocito , Selectina E/metabolismo , Ratones , Ratones Noqueados , Monocitos/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
Biochem Biophys Res Commun ; 384(1): 93-9, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19393222

RESUMEN

There is much speculation whether extravascular inflammation accelerates atherosclerosis. We tested this hypothesis in apoE(-/-) mice using three well-characterized models of non-autoimmune chronic inflammation: croton oil-induced skin inflammation, Aspergillus fumigatus antigen-induced allergic lung disease, and A. fumigatus antigen-induced peritonitis. The croton oil model produced recurrent inflammatory skin ulceration, and marked increases in plasma levels of IL-6 and serum amyloid A (SAA). The allergic lung disease model showed strong local inflammation with eosinophilic infiltration and serum IgE induction. The recurrent peritonitis model was accompanied by mild elevation in plasma SAA levels. Aortic atherosclerosis was quantified by computer-assisted morphometry of en face arteries in apoE(-/-) mice at 34 weeks for the croton oil model, 26 and 42 weeks for the allergic lung disease model, and 26 weeks for the peritonitis model. We found that all three forms of chronic extravascular inflammation had no effect on the rate of atherosclerosis development.


Asunto(s)
Aspergilosis Broncopulmonar Alérgica/complicaciones , Aspergillus fumigatus , Aterosclerosis/etiología , Dermatitis/complicaciones , Peritonitis/complicaciones , Animales , Antígenos Fúngicos/inmunología , Apolipoproteínas E/genética , Aspergilosis Broncopulmonar Alérgica/inmunología , Aspergillus fumigatus/inmunología , Aterosclerosis/genética , Aterosclerosis/inmunología , Ratones , Ratones Noqueados
17.
Dev Cell ; 16(3): 358-73, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19289082

RESUMEN

The transcription factor Neurogenin3 (Ngn3) is required for islet-cell type specification. Here, we show that hepatic gene transfer of Ngn3 transiently induces insulin in terminally differentiated hepatocytes but fails to transdifferentiate them, i.e., switch their lineage into islet cells. However, Ngn3 leads to long-term diabetes reversal in mice due to the emergence of periportal islet-like cell clusters. These neo-islets display glycemia-regulated insulin, beta-cell-specific transcripts, and an islet-specific transcription cascade, and they produce all four major islet hormones. They appear to arise from hepatic progenitor cells, most likely endoderm-derived oval cells. Thus, transfer of a single lineage-defining transcription factor, Ngn3, is sufficient to induce cell-lineage switching from a hepatic to an islet lineage in these progenitor cells, a process consistent with transdetermination, i.e, lineage switching in lineage-determined, but not terminally differentiated, cells. This paradigm of induced transdetermination of receptive progenitor cells in vivo may be generally applicable to therapeutic organogenesis for multiple diseases, including diabetes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Hepatocitos/citología , Islotes Pancreáticos/citología , Proteínas del Tejido Nervioso/fisiología , Células Madre/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Betacelulina , Diferenciación Celular/fisiología , Transdiferenciación Celular/fisiología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/terapia , Técnicas de Transferencia de Gen , Hepatocitos/fisiología , Insulina/biosíntesis , Insulina/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/fisiología , Islotes Pancreáticos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/fisiología , Transcripción Genética
18.
Curr Hypertens Rep ; 10(6): 461-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18959832

RESUMEN

Cytoplasmic lipid droplets (LDs) are organelles in which cells store neutral lipids for use as an energy source in times of need, but they also play important roles in the regulation of key metabolic processes. Although LDs are essential for normal cell function, excess accumulation of intracellular lipid is associated with several metabolic diseases, including obesity, type 2 diabetes, and atherosclerosis. The function of LDs is regulated by their associated proteins, including the members of the PAT family: perilipin, adipophilin/adipose differentiation-related protein, tail-interacting protein 47, S3-12, and OXPAT/myocardial LD protein/lipid-storage droplet protein 5. In this review we discuss the PAT proteins in two cardiovascular contexts: 1) in the atherosclerotic vessel wall, where LDs within macrophage foam cells store cholesteryl esters derived from modified lipoproteins, and 2) in the myocardium, where LDs store fatty acids, the major energy substrate for normal heart function, as triglyceride.


Asunto(s)
Endotelio Vascular/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Proteínas Portadoras , Células Cultivadas , Endotelio Vascular/citología , Humanos , Metabolismo de los Lípidos/fisiología , Lipólisis/fisiología , Miocitos Cardíacos/citología , Péptidos/metabolismo , Perilipina-1 , Perilipina-2 , Sensibilidad y Especificidad
19.
Circ Res ; 102(12): 1492-501, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18483409

RESUMEN

Foam cells are a hallmark of atherosclerosis. However, it is unclear whether foam cell formation per se protects against atherosclerosis or fuels it. In this study, we investigated the role of adipose differentiation-related protein (ADFP), a major lipid droplet protein (LDP), in the regulation of foam cell formation and atherosclerosis. We show that ADFP expression facilitates foam cell formation induced by modified lipoproteins in mouse macrophages in vitro. We show further that Adfp gene inactivation in apolipoprotein E-deficient (ApoE(-/-)) mice reduces the number of lipid droplets in foam cells in atherosclerotic lesions and protects the mice against atherosclerosis. Moreover, transplantation of ADFP-null bone marrow-derived cells effectively attenuated atherosclerosis in ApoE(-/-) mice. Deficiency of ADFP did not cause a detectable compensatory increase in the other PAT domain proteins in macrophages in vitro or in vivo. Mechanistically, ADFP enables the macrophage to maintain its lipid content by hindering lipid efflux. We detected no significant difference in lesion composition or in multiple parameters of inflammation in macrophages or in their phagocytic activity between mice with and without ADFP. In conclusion, Adfp inactivation in ApoE(-/-) background protects against atherosclerosis and appears to be a relatively pure model of impaired foam cell formation.


Asunto(s)
Aterosclerosis/prevención & control , Células Espumosas/metabolismo , Metabolismo de los Lípidos/fisiología , Proteínas de la Membrana/fisiología , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Aterosclerosis/embriología , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Transporte Biológico , Antígenos CD36/fisiología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Colesterol/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Células Espumosas/efectos de los fármacos , Células Espumosas/patología , Lipoproteínas LDL/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Perilipina-2 , Perilipina-3 , Estructura Terciaria de Proteína , ARN Mensajero/biosíntesis , Receptores Depuradores de Clase A/fisiología
20.
Atherosclerosis ; 200(2): 257-63, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18289551

RESUMEN

Genetically modified mice susceptible to atherosclerosis are widely used in atherosclerosis research. Although the atherosclerotic lesions in these animals show similarities to those in humans, comprehensive expression profile analysis of these lesions is limited by their very small size. In this communication, we have developed an approach to analyze global gene expression in mouse lesions by a combination of (a) laser capture microdissection (LCM) to isolate RNA from specific lesions, and (b) an efficient RNA amplification method that reliably yields sufficient quantities of high quality cRNA for quantitative real-time PCR (qPCR), as well as for microarray analysis. The RNA passed multiple quality controls and the expression profile observed in lesional cells compared with the whole artery encompasses genes that are characteristic of a macrophage/foam cell population. We believe that this method represents a useful new tool for the unbiased analysis of global gene expression of specific sub-regions in atherosclerotic lesions in different rodent models.


Asunto(s)
Aterosclerosis/diagnóstico , Aterosclerosis/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Aterosclerosis/patología , Células Espumosas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Rayos Láser , Macrófagos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Control de Calidad , ARN/metabolismo , ARN Complementario/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...