Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Dermatol ; 16(1): 9, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27401543

RESUMEN

BACKGROUND: Risk of cardiovascular disease is increased in patients with psoriasis, but molecular mechanisms linking the two conditions have not been clearly established. Lack of appropriate animal models has hampered generation of new knowledge in this area of research and we therefore sought to develop an animal model with combined atherosclerosis and psoriasis-like skin inflammation. METHODS: Topical 12-O-tetradecanoylphorbol-13-acetate (TPA) was applied to the ears twice per week for 8 weeks in atherosclerosis-prone apolipoprotein E deficient (ApoE(-/-)) mice. RESULTS: TPA led to localized skin inflammation with increased epidermal thickness, infiltration of inflammatory-like cells and augmented tissue interleukin-17F levels. Systemic effects of the topical application of TPA were demonstrated by increased plasma concentration of serum amyloid A and splenic immune modulation, respectively. However, atherosclerotic plaque area and composition, and mRNA levels of several inflammatory genes in the aortic wall were not significantly affected by TPA-induced skin inflammation. CONCLUSIONS: TPA-induced psoriasis-like skin inflammation in atherosclerosis-prone ApoE(-/-) mice evoked systemic immune-inflammatory effects, but did not affect atherogenesis. The results may question the role of psoriasis-induced inflammation in the pathogenesis of atherosclerosis in psoriasis patients.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/patología , Carcinógenos/farmacología , Hipercolesterolemia , Inflamación , Psoriasis/inducido químicamente , Acetato de Tetradecanoilforbol/farmacología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hipercolesterolemia/inmunología , Hipercolesterolemia/patología , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo , Proteína Amiloide A Sérica/metabolismo , Bazo/metabolismo
2.
J Lipid Res ; 56(3): 754-759, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25561460

RESUMEN

apoM is a member of the lipocalin superfamily and circulates in plasma attached to HDL particles. apoM plays a role in cholesterol metabolism and has recently been identified as transporter for the signaling lipid, sphingosine-1-phosphate (S1P), in plasma. S1P is implicated in several inflammatory diseases such as multiple sclerosis and rheumatoid arthritis. The ability to accurately measure apoM is crucial for investigating its biological functions and possible clinical implications. However, reliable commercial methods have been lacking so far. Therefore, we have developed an assay that specifically recognizes human apoM in plasma using commercially available reagents. Commercial apoM antibodies were screened for compatibility in a sandwich ELISA-based assay. One optimal pair of antibodies was chosen, and sample preparation, buffers, and incubation times were optimized to generate a simple and reproducible method. Validation and comparison to a previously described ELISA for apoM confirmed that the assay displays a high degree of sensitivity, specificity, and precision. Our results show that commercially available antibodies can be used to accurately measure human plasma apoM. This method can be implemented in every laboratory and will help promote high quality research.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Apolipoproteínas/sangre , Lipocalinas/sangre , Desplegamiento Proteico , Apolipoproteínas M , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos
3.
Cardiovasc Diabetol ; 14: 87, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26169175

RESUMEN

BACKGROUND: Type 2 diabetes is associated with abnormal electrical conduction and sudden cardiac death, but the pathogenic mechanism remains unknown. This study describes electrophysiological alterations in a diet-induced pre-diabetic rat model and examines the underlying mechanism. METHODS: Sprague-Dawley rats were fed either high-fat diet and fructose water or normal chow and water for 6 weeks. The electrophysiological properties of the whole heart was analyzed by in vivo surface ECG recordings, as wells as ex vivo in Langendorff perfused hearts during baseline, ischemia and reperfussion. Conduction velocity was examined in isolated tissue strips. Ion channel and gap junction conductances were analyzed by patch-clamp studies in isolated cardiomyocytes. Fibrosis was examined by Masson's Trichrome staining and thin-layer chromatography was used to analyze cardiac lipid content. Connexin43 (Cx43) expression and distribution was examined by western blotting and immunofluorescence respectively. RESULTS: Following 6 weeks of feeding, fructose-fat fed rats (FFFRs) showed QRS prolongation compared to controls (16.1 ± 0.51 (n = 6) vs. 14.7 ± 0.32 ms (n = 4), p < 0.05). Conduction velocity was slowed in FFFRs vs. controls (0.62 ± 0.02 (n = 13) vs. 0.79 ± 0.06 m/s (n = 11), p < 0.05) and Langendorff perfused FFFR hearts were more prone to ventricular fibrillation during reperfusion following ischemia (p < 0.05). The patch-clamp studies revealed no changes in Na(+) or K(+) currents, cell capacitance or gap junctional coupling. Cx43 expression was also unaltered in FFFRs, but immunofluorescence demonstrated an increased fraction of Cx43 localized at the intercalated discs in FFFRs compared to controls (78 ± 3.3 (n = 5) vs. 60 ± 4.2 % (n = 6), p < 0.01). No fibrosis was detected but FFFRs showed a significant increase in cardiac triglyceride content (1.93 ± 0.19 (n = 12) vs. 0.77 ± 0.13 nmol/mg (n = 12), p < 0.0001). CONCLUSION: Six weeks on a high fructose-fat diet cause electrophysiological changes, which leads to QRS prolongation, decreased conduction velocity and increased arrhythmogenesis during reperfusion. These alterations are not explained by altered gap junctional coupling, Na(+), or K(+) currents, differences in cell size or fibrosis.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Sistema de Conducción Cardíaco/fisiopatología , Isquemia Miocárdica/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Estado Prediabético/fisiopatología , Animales , Conexina 43/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Electrocardiografía , Fructosa , Uniones Comunicantes/metabolismo , Masculino , Contracción Miocárdica , Reperfusión Miocárdica , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Canales de Sodio/metabolismo , Triglicéridos/metabolismo
4.
Biochim Biophys Acta ; 1831(7): 1287-92, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24046869

RESUMEN

OBJECTIVE: The HDL associated apolipoprotein M (apoM) protects against experimental atherosclerosis but the mechanism is unknown. ApoM increases prebeta-HDL formation. We explored whether plasma apoM affects mobilization of cholesterol from peripheral cells in mice. METHODS AND RESULTS: ApoM-enriched HDL from apoM-transgenic mice increased the in vitro efflux of 3H-cholesterol from macrophages by 24 +/- 3% (p < 0.05) as compared with HDL from wild type (WT) mice, thus confirming previous findings. However, apoM-free HDL was not poorer than that of WT HDL to mobilize 3H-cholesterol. 3H-cholesterol-labeled foam cells were implanted in the peritoneal cavity of apoM-/-, WT and apoM-transgenic mice to assess the mobilization of cholesterol from foam cells in vivo and subsequent excretion into feces. The results showed a statistically non-significant trend towards increased mobilization of cellular cholesterol to plasma with increasing plasma apoM. However, the apoM-genotype did not affect the excretion of 3H-cholesterol in feces. Nevertheless, when apoM-/-, apoM-transgenic and WT mice received a constant intravenous infusion of 13C2-cholesterol/intralipid for 5 h, the rate of enrichment of blood free cholesterol with free 13C2-cholesterol was significantly lower (consistent with an increase in flux of unlabeled free cholesterol into the plasma) in the apoM-transgenic (3.0 +/- 0.9 per thousand/h) as compared to WT (5.7 +/- 0.9 per thousand/h, p < 0.05) and apoM-/- (6.5 +/- 0.6 per thousand/h, p < 0.01) mice. CONCLUSION: The present data indicate that the plasma apoM levels modulate the ability of plasma to mobilize cellular cholesterol, whereas apoM has no major effect on the excretion of cholesterol into feces.


Asunto(s)
Apolipoproteínas/metabolismo , Colesterol/sangre , Colesterol/metabolismo , Animales , Apolipoproteínas/sangre , Apolipoproteínas/genética , Apolipoproteínas M , Aterosclerosis/sangre , Aterosclerosis/metabolismo , Bilis/metabolismo , Línea Celular , Colesterol/administración & dosificación , Colesterol/análisis , Heces/química , Células Espumosas/metabolismo , Eliminación de Gen , Humanos , Metabolismo de los Lípidos , Lipoproteínas HDL/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
5.
Arch Environ Contam Toxicol ; 67(1): 9-20, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24435476

RESUMEN

Polybrominated diphenyl ethers (PBDEs), a group of flame retardants, and perfluoroalkyl substances (PFASs) were analysed in serum samples of pregnant women from Denmark to provide information about their exposure and to study indications of common exposure pathways. The main BDE congener was the fully brominated BDE-209 with a median value of 7.5 ng/g lipid (46 pg/mL; 9.8 pmol/g lipid). Other BDE congeners decreased in the order BDE-47 > BDE-99 > BDE-153. The summed concentration of tri- to hepta-BDEs was 7.7 ng/g lipid, i.e. in the higher end of previously reported concentrations from Europe, including plasma samples of pregnant Danish women. Total lipid contents were relatively low, on average 5.9 g/L (9.0 mmol/L). The main PFAS compound was perfluorooctane sulfonate with a median concentration of 8.4 ng/mL. Other PFASs decreased in the order perfluorooctanoic acid > perfluorononanoic acid > perfluorodecanoic acid > perfluorohexane sulfonate and resulted in a ΣPFAS of 12 ng/mL. Within each group, compounds were highly intercorrelated with the exception of BDE-209, which was not correlated with any of the other compounds. No correlations were found either between PFASs and PBDEs suggesting different sources of exposure and/or pharmacokinetic and metabolisation processes. PBDE and PFAS concentrations were in the range associated with adverse effects in some epidemiological studies.


Asunto(s)
Monitoreo del Ambiente , Contaminantes Ambientales/sangre , Fluorocarburos/sangre , Éteres Difenilos Halogenados/sangre , Exposición Materna/estadística & datos numéricos , Adulto , Dinamarca , Femenino , Humanos , Embarazo
6.
Curr Opin Lipidol ; 24(4): 295-300, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23652568

RESUMEN

PURPOSE OF REVIEW: The review will address the potential roles of apolipoprotein M (apoM) as a carrier protein and modulator of sphingosine-1-phosphate (S1P) bioactivity. RECENT FINDINGS: Recombinant apoM can bind small lipids such as retinoic acid, oxidized phospholipids, and S1P. Thus, the effects of apoM may be pleiotrophic. The S1P binding ability of apoM has biological impact. ApoM-bound S1P can activate S1P1 receptors on endothelial cells and deficiency of apoM abolishes the presence of S1P in HDL. In mice, the lack of apoM causes dysfunctional endothelial barrier function in the lungs. In humans, sepsis that is characterized by impaired endothelial function is associated with low plasma apoM. SUMMARY: Plasma apoM is mainly bound to HDL. The roles of apoM in atherosclerosis and lipoprotein metabolism have been given much attention. New in the field is the discovery of apoM as a chaperone for S1P. S1P is a bioactive lipid with effects on angiogenesis, lymphocyte trafficking, endothelial cell migration, and inflammation. A drug targeting the S1P-system (fingolimod) is now used for treatment of multiple sclerosis. It improves the blood-brain barrier and inhibits migration of lymphocytes into the brain. Further exploration of the apoM/S1P axis may uncover its potential as a biomarker and target for new treatments.


Asunto(s)
Apolipoproteínas/fisiología , Células Endoteliales/fisiología , Lipocalinas/fisiología , Lipoproteínas HDL/fisiología , Animales , Apolipoproteínas M , Aterosclerosis/sangre , Humanos , Metabolismo de los Lípidos , Lisofosfolípidos/sangre , Unión Proteica , Sepsis/sangre , Esfingosina/análogos & derivados , Esfingosina/sangre
7.
Front Pharmacol ; 15: 1328259, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38313311

RESUMEN

Aim: Apolipoprotein M (apoM) is mainly expressed in liver and in proximal tubular epithelial cells in the kidney. In plasma, apoM associates with HDL particles via a retained signal peptide and carries sphingosine-1-phosphate (S1P), a small bioactive lipid. ApoM is undetectable in urine from healthy individuals but lack of megalin receptors in proximal tubuli cells induces loss of apoM into the urine. Besides this, very little is known about kidney-derived apoM. The aim of this study was to address the role of apoM in kidney biology and in acute kidney injury. Methods: A novel kidney-specific human apoM transgenic mouse model (RPTEC-hapoMTG) was generated and subjected to either cisplatin or ischemia/reperfusion injury. Further, a stable transfection of HK-2 cells overexpressing human apoM (HK-2-hapoMTG) was developed to study the pattern of apoM secretion in proximal tubuli cells. Results: Human apoM was present in plasma from RPTEC-hapoMTG mice (mean 0.18 µM), with a significant increase in plasma S1P levels. In vitro apoM was secreted to both the apical (urine) and basolateral (blood) compartment from proximal tubular epithelial cells. However, no differences in kidney injury score was seen between RPTEC-hapoMTG and wild type (WT) mice upon kidney injury. Further, gene expression of inflammatory markers (i.e., IL6, MCP-1) was similar upon ischemia/reperfusion injury. Conclusion: Our study suggests that kidney-derived apoM is secreted to plasma, supporting a role for apoM in sequestering molecules from excretion in urine. However, overexpression of human apoM in the kidney did not protect against acute kidney injury.

8.
J Physiol ; 590(14): 3349-60, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22586215

RESUMEN

Skeletal muscle mitochondrial content varies extensively between human subjects. Biochemical measures of mitochondrial proteins, enzyme activities and lipids are often used as markers of mitochondrial content and muscle oxidative capacity (OXPHOS). The purpose of this study was to determine how closely associated these commonly used biochemical measures are to muscle mitochondrial content and OXPHOS. Sixteen young healthy male subjects were recruited for this study. Subjects completed a graded exercise test to determine maximal oxygen uptake (VO2peak) and muscle biopsies were obtained from the vastus lateralis. Mitochondrial content was determined using transmission electron microscopy imaging and OXPHOS was determined as the maximal coupled respiration in permeabilized fibres. Biomarkers of interest were citrate synthase (CS) activity, cardiolipin content, mitochondrial DNA content (mtDNA), complex I­V protein content, and complex I­IV activity. Spearman correlation coefficient tests and Lin's concordance tests were applied to assess the absolute and relative association between the markers and mitochondrial content or OXPHOS. Subjects had a large range of VO2peak (range 29.9­71.6ml min−1 kg−1) and mitochondrial content (4­15% of cell volume).Cardiolipin content showed the strongest association with mitochondrial content followed by CS and complex I activities. mtDNA was not related to mitochondrial content. Complex IV activity showed the strongest association with muscle oxidative capacity followed by complex II activity.We conclude that cardiolipin content, and CS and complex I activities are the biomarkers that exhibit the strongest association with mitochondrial content, while complex IV activity is strongly associated with OXPHOS capacity in human skeletal muscle.


Asunto(s)
Biomarcadores/análisis , Mitocondrias Musculares/química , Fibras Musculares Esqueléticas/química , Músculo Cuádriceps/química , Adenosina Trifosfatasas/análisis , Adulto , Cardiolipinas/análisis , Proteínas Portadoras/análisis , Citrato (si)-Sintasa/análisis , Complejo I de Transporte de Electrón/análisis , Prueba de Esfuerzo , Humanos , Masculino , Proteínas de la Membrana/análisis , Microscopía Electrónica de Transmisión , Mitocondrias Musculares/ultraestructura , ATPasas de Translocación de Protón Mitocondriales , Fibras Musculares Esqueléticas/ultraestructura , Fosforilación Oxidativa , Consumo de Oxígeno , Músculo Cuádriceps/citología
9.
FASEB J ; 25(11): 3803-14, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21784784

RESUMEN

GPR39, a constitutively active 7TM receptor important for glucose-induced insulin secretion and maturation of pancreatic ß-cell function, is up-regulated in adipose tissue on abstinence from food and chemically induced diabetes. In the present study, we investigated the effect of GPR39 deficiency on body weight and adipocyte metabolism. GPR39-deficient mice were subjected to a high-fat diet and body composition, glucose tolerance, insulin secretion, food intake, and energy expenditure were evaluated. The cell biology of adipocyte metabolism was studied on both mRNA and protein levels. A significant increase in body weight corresponding to a 2-fold selective increase in fat mass was observed in GPR39-deficient mice fed a high-fat diet as compared with wild-type littermate controls fed the same diet. The GPR39-deficient animals had similar food intake but displayed almost eliminated diet-induced thermogenesis, measured by the oxygen consumption rate (Vo(2)) on change from normal to high-fat diet. Analysis of the adipose tissue for lipolytic enzymes demonstrated decreased level of phosphorylated hormone-sensitive lipase (HSL) and a decreased level of adipose triglyceride lipase (ATGL) by 35 and 60%, respectively, after food withdrawal in the GPR39-deficient mice. Extracellular signal-regulated kinases (ERK1/2), a signaling pathway known to be important for lipolysis, was decreased by 56% in the GPR39-deficient mice. GPR39 deficiency is associated with increased fat accumulation on a high-fat diet, conceivably due to decreased energy expenditure and adipocyte lipolytic activity.


Asunto(s)
Adipocitos/metabolismo , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Animales , Dieta Alta en Grasa , Metabolismo Energético , Femenino , Perfilación de la Expresión Génica , Metabolismo de los Lípidos/genética , Masculino , Ratones , Consumo de Oxígeno
10.
Circ Res ; 106(10): 1624-34, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20360257

RESUMEN

RATIONALE: Plasma apolipoprotein (apo)M is mainly associated with high-density lipoprotein (HDL). HDL-bound apoM is antiatherogenic in vitro. However, plasma apoM is not associated with coronary heart disease in humans, perhaps because of a positive correlation with plasma low-density lipoprotein (LDL). OBJECTIVE: We explored putative links between apoM and very-low-density (VLDL)/LDL metabolism and the antiatherogenic potential of apoM in vivo. METHODS AND RESULTS: Plasma apoM was increased approximately 2.1 and approximately 1.5 fold in mice lacking LDL receptors (Ldlr(-/-)) and expressing dysfunctional LDL receptor-related protein 1 (Lrp1(n2/n2)), respectively, but was unaffected in apoE-deficient (ApoE(-/-)) mice. Thus, pathways controlling catabolism of VLDL and LDL affect plasma apoM. Overexpression (approximately 10-fold) of human apoM increased (50% to 70%) and apoM deficiency decreased ( approximately 25%) plasma VLDL/LDL cholesterol in Ldlr(-/-) mice, whereas apoM did not affect plasma VLDL/LDL in mice with intact LDL receptors. Moreover, plasma clearance of apoM-enriched VLDL/LDL was slower than that of control VLDL/LDL in mice lacking functional LDL receptors and LRP1, suggesting that apoM impairs the catabolism of VLDL/LDL that occurs independently of the LDL receptor and LRP1. ApoM overexpression decreased atherosclerosis in ApoE(-/-) (60%) and cholate/cholesterol-fed wild-type mice (70%). However, in Ldlr(-/-) mice the antiatherogenic effect of apoM was attenuated by its VLDL/LDL-raising effect. CONCLUSION: The data suggest that defect LDL receptor function leads to increased plasma apoM concentrations, which in turn, impairs the removal of VLDL/LDL from plasma. This mechanism opposes the otherwise antiatherogenic effect of apoM.


Asunto(s)
Apolipoproteínas B/metabolismo , Apolipoproteínas/farmacología , Aterosclerosis/metabolismo , Animales , Apolipoproteínas/deficiencia , Apolipoproteínas/genética , Apolipoproteínas B/farmacología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Colesterol/sangre , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Cruzamientos Genéticos , Ensayo de Inmunoadsorción Enzimática , Humanos , Lipoproteínas VLDL/metabolismo , Ratones , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética , Triglicéridos/sangre
11.
Crit Care ; 16(3): 126, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22587809

RESUMEN

Sepsis is one of the leading causes of mortality in non-cardiac intensive care units, and the need for markers of progression and severity are high. Also, treatment of sepsis is highly debated and potential new targets of treatment are of great interest. In the previous issue of Critical Care Kumaraswamy and colleagues have investigated whether plasma apolipoprotein M (apoM) is affected during different grades of sepsis, septic shock and systemic inflammatory response syndrome. Interestingly, plasma apoM was significantly decreased in all groups of patients with a relationship to severity of disease. This identifies apoM as a potential new biomarker in sepsis. It also underscores the possibility that altered high-density lipoprotein in sepsis patients can affect the course of disease. Thus, since apoM is the carrier of Sphingosine-1-P (S1P), a molecule with great influence on vascular barrier function, the study presented raises the interest and relevance for further studies of apoM and S1P in relation to sepsis and inflammation.


Asunto(s)
Apolipoproteínas/sangre , Lipocalinas/sangre , Sepsis/sangre , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Femenino , Humanos , Masculino
12.
J Biol Chem ; 285(14): 10583-90, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20103594

RESUMEN

The ability to produce apolipoprotein (apo) B-containing lipoproteins enables hepatocytes, enterocytes, and cardiomyocytes to export triglycerides. In this study, we examined secretion of apoB-containing lipoproteins from mouse kidney and its putative impact on triglyceride accumulation in the tubular epithelium. Mouse kidney expressed both the apoB and microsomal triglyceride transfer protein genes, which permit lipoprotein formation. To examine de novo lipoprotein secretion, kidneys from human apoB-transgenic mice were minced and placed in medium with (35)S-amino acids. Upon sucrose gradient ultracentrifugation of the labeled medium, fractions were analyzed by apoB immunoprecipitation. (35)S-Labeled apoB100 was recovered in approximately 1.03-1.04 g/ml lipoproteins (i.e. similar to the density of plasma low density lipoproteins). Immunohistochemistry of kidney sections suggested that apoB mainly is produced by tubular epithelial cells. ApoB expression in the kidney cortex was reduced approximately 90% in vivo by treating wild type mice with apoB-antisense locked nucleic acid oligonucleotide. Inhibition of apoB expression increased fasting-induced triglyceride accumulation in the kidney cortex by 20-25% (p = 0.008). Cholesterol stores were unaffected. Treatment with control oligonucleotides with 1 or 4 mismatching base pairs affected neither the triglyceride nor the cholesterol content of the kidney cortex. The results suggest that mammalian kidney secretes apoB100-containing lipoproteins. One biological effect may be to dampen excess storage of triglycerides in proximal tubule cells.


Asunto(s)
Apolipoproteínas B/fisiología , Colesterol/metabolismo , Riñón/metabolismo , Triglicéridos/metabolismo , Animales , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Western Blotting , Humanos , Riñón/citología , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Lipid Res ; 51(1): 103-11, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19546343

RESUMEN

Promoter polymorphisms in microsomal triglyceride transfer protein (MTTP) have been associated with decreased plasma lipids but an increased risk for ischemic heart disease (IHD), indicating that MTTP influences the susceptibility for IHD independent of plasma lipids. The objective of this study was to characterize the functional promoter polymorphism in MTTP predisposing to IHD and its underlying mechanism. Use of pyrosequencing technology revealed that presence of the minor alleles of the promoter polymorphisms -493G>T and -164T>C result in lower transcription of MTTP in vivo in the heart, liver, and macrophages. In vitro experiments indicated that the minor -164C allele mediates the lower gene expression and that C/EBP binds to the polymorphic region in an allele-specific manner. Furthermore, homozygous carriers of the -164C were found to have increased risk for IHD as shown in a case-control study including a total of 544 IHD patients and 544 healthy control subjects. We concluded that carriers of the minor -164C allele have lower expression of MTTP in the heart, mediated at least partly by the transcription factor CCAAT/enhancer binding protein, and that reduced concentration of MTTP in the myocardium may contribute to IHD upon ischemic damage.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Portadoras/genética , Hígado Graso/metabolismo , Regulación de la Expresión Génica , Isquemia Miocárdica/genética , Anciano , Alelos , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT/genética , Estudios de Casos y Controles , Hígado Graso/genética , Femenino , Células HeLa , Corazón/fisiología , Humanos , Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Elementos de Respuesta/genética
14.
Gynecol Endocrinol ; 25(3): 175-82, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19347707

RESUMEN

OBJECTIVE: To study the effect of 17beta-estradiol (E(2)) or conjugated equine estrogens (CEE) alone and in combination with norethisterone acetate (NETA) or medroxyprogesterone acetate (MPA) on the endothelin-1 (ET-1) system. METHODS: New Zealand White rabbits were treated with E(2), CEE, E(2) + NETA, CEE + MPA or placebo. The thoracic aorta and the epicardial coronary artery were used for mRNA expression and myograph analyses, respectively. RESULTS: E(2) and CEE alone significantly reduced ET-1 receptor subtype A (ET(A)) mRNA expression compared with placebo treatment. The E(2)-induced reduction in ET(A) mRNA expression persisted with the co-administration of NETA, but the CEE induced reduction in ET(A) mRNA expression was not maintained with the co-administration of MPA. Treatment with CEE alone significantly increased endotelin-1 converting enzyme (ECE) mRNA expression and CEE combined with MPA reduced prepro-endothelin-1 (ppET-1) mRNA expression when compared with placebo. ET-1 receptor subtype B mRNA expression and ET-1 induced vasocontraction was unaffected by treatment. CONCLUSIONS: E(2) and CEE treatment exert potentially beneficial vascular effects through regulation of the ET(A) receptor. The effect was maintained with the co-administration of NETA, but not MPA. The differential effects of specific hormone components may explain the variable effects of hormone therapy on the arterial wall.


Asunto(s)
Aorta/efectos de los fármacos , Endotelina-1/genética , Terapia de Reemplazo de Estrógeno , Estrógenos Conjugados (USP)/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Animales , Aorta/metabolismo , Combinación de Medicamentos , Endotelina-1/metabolismo , Estradiol/administración & dosificación , Estradiol/farmacología , Estrógenos Conjugados (USP)/administración & dosificación , Femenino , Noretindrona/administración & dosificación , Noretindrona/análogos & derivados , Noretindrona/farmacología , Acetato de Noretindrona , Ovariectomía , ARN Mensajero/metabolismo , Conejos
15.
J Appl Physiol (1985) ; 125(3): 901-908, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29543138

RESUMEN

Both exercise training and weight loss reduce cardiovascular risk, but the independent importance of the two strategies is unclear. We aimed to investigate independent and combined effects of exercise training and weight loss on lipoproteins and dyslipidemia in overweight sedentary men. Sixty individuals were randomized to 12 wk of endurance training (T), energy-reduced diet (D), training and energy increased diet (T-iD), or control (C). Equal energetic deficits (-600 kcal/day) were prescribed by exercise for T and caloric restriction for D. T-iD completed similar exercise but remained in energy balance due to the dietary replacement of calories expended during exercise. Total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), apolipoprotein (apo)B and A1, pre-ß-HDL, and susceptibility of LDL-C to oxidation were measured. Body weight was reduced similarly between T (-5.9 ± 0.7 kg) and D (-5.2 ± 0.8 kg), whereas T-iD (-1.0 ± 0.5 kg) and C (0.1 ± 0.6 kg) remained weight stable. Plasma TC, LDL-C, and apolipoprotein B were reduced in T compared with C ( P < 0.001 for both), but this was not observed for D ( P > 0.17). Changes in TC and LDL-C were associated with changes in body weight and body fat ( P < 0.01). In T-iD, increases in HDL-C and apolipoprotein A1 were observed ( P < 0.001). In conclusion, an exercise-induced decline in body weight reduces proatherogenic apoB-containing lipoproteins, whereas exercise compensated by energy intake increases the key component of reverse cholesterol transport, i.e., apoA1-containing HDL-C. NEW & NOTEWORTHY Exercise has additive effects in lowering plasma lipoprotein particles to diet-induced weight loss in individuals with increased cardiovascular risk. In the present study, we investigated whether training per se would have beneficial cardiovascular effects. We found that 3 mo of exercise-induced weight loss reduced proatherogenic lipoproteins, whereas endurance training without weight loss improved factors involved in reverse cholesterol transport in a group of overweight sedentary men.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Ejercicio Físico , Sobrepeso/complicaciones , Pérdida de Peso , Adulto , Apolipoproteínas B/sangre , Composición Corporal , Restricción Calórica , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/etiología , Colesterol/sangre , Dislipidemias/sangre , Dislipidemias/prevención & control , Humanos , Lipoproteínas/sangre , Masculino , Aptitud Física , Factores de Riesgo
16.
Atherosclerosis ; 275: 304-311, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29980057

RESUMEN

BACKGROUND AND AIMS: Plasma apolipoprotein M (APOM) is bound to HDL-particles and has anti-atherogenic effects. The present study explored whether plasma APOM is reduced in patients with chronic kidney disease (CKD), and associated with cardiovascular disease (CVD). In addition, we tested the hypothesis that the excretion of APOM into the urine is increased in patients with kidney disease. METHODS: Plasma samples were collected from a cohort of patients with CKD stages 1 to 5D (N = 409) and controls (N = 35). Urine was collected from 47 subjects. Plasma APOM was measured with sandwich ELISA and urine APOM with competitive ELISA. RESULTS: Plasma APOM levels were reduced in patients with CKD stages 3-5D as compared to patients with CKD stages 1 + 2 and controls (p < 0.01). CKD patients with known CVD displayed even further reduction in plasma APOM levels than CKD patients without known CVD (p < 0.001). Fast-phase liquid chromatography showed that plasma APOM was primarily associated with HDL-cholesterol (HDL-C) across CKD stages. Accordingly, when plasma APOM values were corrected for HDL-C, a significant difference only persisted between patients with CKD stage 3 and stages 1 + 2 (p < 0.05), and the difference between CKD patients with and without known CVD disappeared. Urine APOM/creatinine ratio was not significantly increased in patients with kidney disease. CONCLUSIONS: The results show that the difference in plasma APOM levels observed between patients with mild and advanced CKD may mainly be due to differences in plasma HDL-C. Whether APOM plays a role in human uremic atherogenesis warrants further experimental studies.


Asunto(s)
Apolipoproteínas M/sangre , Enfermedades Cardiovasculares/sangre , Insuficiencia Renal Crónica/sangre , Adulto , Anciano , Apolipoproteínas M/orina , Biomarcadores/sangre , Biomarcadores/orina , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/orina , Estudios de Casos y Controles , HDL-Colesterol/sangre , Cromatografía Líquida de Alta Presión , Dinamarca/epidemiología , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/epidemiología , Insuficiencia Renal Crónica/orina , Medición de Riesgo , Factores de Riesgo , Urinálisis
17.
J Diabetes Res ; 2018: 6823193, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29682581

RESUMEN

BACKGROUND: Obesity and metabolic syndrome (MetS) are major risk factors for atherosclerotic diseases; however, a causal link remains elusive. Animal models resembling human MetS and its complications, while important, are scarce. We aimed at developing a porcine model of human MetS. METHODS: Forty pigs with familial hypercholesterolemia were fed a high fat + fructose diet for 30 weeks. Metabolic assessments and subcutaneous fat biopsies were obtained at 18 and 30 weeks, and fat distribution was assessed by CT-scans. Postmortem, macrophage density, and phenotype in fat tissues were quantified along with atherosclerotic burden. RESULTS: During the experiment, we observed a >4-fold in body weight, a significant but small increase in fasting glucose (4.1 mmol/L), insulin (3.1 mU/L), triglycerides (0.5 mmol/L), and HDL cholesterol (2.6 mmol/L). Subcutaneous fat correlated with insulin resistance, but intra-abdominal fat correlated inversely with insulin resistance and LDL cholesterol. More inflammatory macrophages were found in visceral versus subcutaneous fat, and inflammation decreased in subcutaneous fat over time. CONCLUSIONS: MetS based on human criteria was not achieved. Surprisingly, visceral fat seemed part of a healthier metabolic and inflammatory profile. These results differ from human findings, and further research is needed to understand the relationship between obesity and MetS in porcine models.


Asunto(s)
Aterosclerosis/metabolismo , Dieta Alta en Grasa/efectos adversos , Hipercolesterolemia/metabolismo , Resistencia a la Insulina/fisiología , Síndrome Metabólico/metabolismo , Obesidad Abdominal/metabolismo , Animales , Aterosclerosis/etiología , Aterosclerosis/patología , Composición Corporal/fisiología , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Femenino , Hipercolesterolemia/etiología , Hipercolesterolemia/patología , Grasa Intraabdominal/metabolismo , Síndrome Metabólico/etiología , Síndrome Metabólico/patología , Obesidad Abdominal/etiología , Obesidad Abdominal/patología , Grasa Subcutánea/metabolismo , Porcinos , Porcinos Enanos , Triglicéridos/metabolismo
18.
Regul Pept ; 141(1-3): 135-9, 2007 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-17289171

RESUMEN

BACKGROUND: Maternal diabetes increases the risk of hypertrophic cardiomyopathy in the fetus. As signaling via the C-type natriuretic peptide (CNP) specific receptor protects against cardiac hypertrophy, we examined whether maternal type 1 diabetes affects the plasma concentrations of proCNP-derived peptides in newborn infants. METHODS: Plasma concentrations of proCNP-derived peptides were measured in umbilical cord plasma and human placental tissue extracts using sequence-specific radioimmunoassays raised against N-terminal and C-terminal proCNP regions, respectively. RESULTS: The median proCNP concentrations were similar in umbilical cord plasma from pregnant women with and without type 1 diabetes (17 pmol/L vs. 19 pmol/L, P not significant) and did not correlate with the proBNP concentrations in the same samples. However, the molar ratio between the proCNP and the CNP peptide was increased in umbilical cord plasma compared to adult plasma (4.6 vs. 1.1), which parallels our earlier findings for proBNP and BNP peptides. CONCLUSIONS: There is a discordant expression of CNP and BNP peptides in newborn infants of mothers with diabetes. Moreover, fetal metabolism of proCNP and CNP appears to differ from healthy adults. The precise mechanism underlying these differences warrants further investigation.


Asunto(s)
Diabetes Mellitus Tipo 1/sangre , Expresión Génica , Péptido Natriurético Encefálico/sangre , Péptido Natriurético Tipo-C/sangre , Precursores de Proteínas/sangre , Adulto , Femenino , Sangre Fetal/química , Hemoglobina Glucada/análisis , Humanos , Recién Nacido , Péptido Natriurético Encefálico/genética , Péptido Natriurético Tipo-C/genética , Placenta/química , Placenta/cirugía , Embarazo , Precursores de Proteínas/genética , Radioinmunoensayo , Población Blanca/estadística & datos numéricos
19.
Mol Endocrinol ; 20(1): 212-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16099815

RESUMEN

Apolipoprotein (apo) M is a novel apolipoprotein belonging to the lipocalin protein superfamily, i.e. proteins binding small lipophilic compounds. Like other apolipoproteins, it is expressed in hepatocytes and secreted into plasma where it associates with high-density lipoprotein particles. In addition, apoM is expressed at high levels in the kidney tubule cells. In this study, we show that the multiligand receptor megalin, which is expressed in kidney proximal tubule cells, is a receptor for apoM and mediates its uptake in the kidney. To examine apoM binding to megalin, a recombinant apoM was expressed in Escherichia coli and used in surface plasmon resonance and cell culture studies. The results showed apoM binding to immobilized megalin [dissociation constant (Kd) approximately 0.3-1 microm] and that the apoM was endocytosed by cultured rat yolk sac cells in a megalin-dependent manner. To examine the importance of apoM binding by megalin in vivo, we analyzed mice with a tissue-specific deficiency of megalin in the kidney. Megalin deficiency was associated with pronounced urinary excretion of apoM, whereas apoM was not detected in normal mouse, human, or rat urine. Gel filtration analysis showed that the urinary apoM-containing particles were small and devoid of apoA-I. The results suggest that apoM binds to megalin and that megalin-mediated endocytosis in kidney proximal tubules prevents apoM excretion in the urine.


Asunto(s)
Apolipoproteínas/metabolismo , Riñón/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Animales , Apolipoproteína A-I/metabolismo , Apolipoproteína A-I/orina , Apolipoproteínas/sangre , Apolipoproteínas/orina , Apolipoproteínas M , Células Cultivadas , Endocitosis , Humanos , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Ratones Transgénicos , Unión Proteica , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Saco Vitelino/citología
20.
Atherosclerosis ; 265: 93-101, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28866363

RESUMEN

BACKGROUND AND AIMS: Chronic kidney disease is characterized by uremia and causes premature death, partly due to accelerated atherosclerosis. Apolipoprotein (apo) M is a plasma carrier protein for the lipid sphingosine-1-phosphate (S1P). The Apom-S1P complex associates with HDL, and may contribute to its anti-atherosclerotic effects. The role of Apom/S1P in atherosclerosis is presently controversial and has not been explored in a uremic setting. We aimed to explore whether plasma concentrations of Apom/S1P are altered by uremia and whether Apom overexpression or deficiency affects classical and uremic atherosclerosis. METHODS: Mild to moderate uremia was induced by subtotal nephrectomy (NX) in 86-92 Apoe-deficient mice that were either Apom-wild type, Apom-deficient, or overexpressed Apom (∼10 fold). The effects of uremia on plasma Apom/S1P and atherosclerosis were evaluated and compared to non-nephrectomized controls. RESULTS: Uremia increased plasma Apom by ∼25%, but not S1P. Plasma S1P was elevated by ∼300% in mice overexpressing Apom, and decreased by ∼25% in Apom-deficient mice. Apom overexpression augmented aortic root atherosclerosis and plasma cholesterol. In contrast, aortic arch atherosclerosis was unaffected by the Apom genotype. There was no effect of Apom-deficiency or Apom overexpression on uremic atherosclerosis. CONCLUSIONS: This study highlights the complexity of Apom/S1P in atherosclerosis and challenges the notion that the Apom/S1P complex is anti-atherogenic, at least in Apoe-deficient mice.


Asunto(s)
Enfermedades de la Aorta/sangre , Apolipoproteínas M/sangre , Aterosclerosis/sangre , Lisofosfolípidos/sangre , Esfingosina/análogos & derivados , Uremia/sangre , Animales , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas M/deficiencia , Apolipoproteínas M/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Nefrectomía , Fenotipo , Placa Aterosclerótica , Esfingosina/sangre , Uremia/etiología , Uremia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA