Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 135(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34698355

RESUMEN

The bone marrow has emerged as a potentially important target in cardiovascular disease as it generates all leukocytes involved in atherogenesis. In the current study, we evaluated whether a change in bone marrow functionality underlies the increased atherosclerosis susceptibility associated with high-density lipoprotein (HDL) deficiency. We found that HDL deficiency in mice due to the genetic lack of hepatocyte-derived apolipoprotein A1 (APOA1) was associated with an increase in the Lin-Sca-1+Kit+ (LSK) bone marrow stem cell population and lymphoid-primed multipotent progenitor numbers, which translated into a higher production and systemic flux of T cell subsets. In accordance with APOA1 deficiency-associated priming of stem cells to increase T lymphocyte production, atherogenic diet-fed low-density lipoprotein receptor knockout mice transplanted with bone marrow from APOA1-knockout mice displayed marked lymphocytosis as compared to wild-type bone marrow recipients. However, atherosclerotic lesion sizes and collagen contents were similar in the two groups of bone marrow recipients. In conclusion, systemic lack of APOA1 primes bone marrow stem cells for T cell lymphopoiesis. Our data provide novel evidence for a regulatory role of HDL in bone marrow functioning in normolipidemic mice.


Asunto(s)
Apolipoproteína A-I , Linfopoyesis , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Células de la Médula Ósea , Trasplante de Médula Ósea , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL , Linfocitos T
2.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34716267

RESUMEN

The rapid development of nanotechnology has greatly benefited modern science and engineering and also led to an increased environmental exposure to nanoparticles (NPs). While recent research has established a correlation between the exposure of NPs and cardiovascular diseases, the intrinsic mechanisms of such a connection remain unclear. Inhaled NPs can penetrate the air-blood barrier from the lung to systemic circulation, thereby intruding the cardiovascular system and generating cardiotoxic effects. In this study, on-site cardiovascular damage was observed in mice upon respiratory exposure of silica nanoparticles (SiNPs), and the corresponding mechanism was investigated by focusing on the interaction of SiNPs and their encountered biomacromolecules en route. SiNPs were found to collect a significant amount of apolipoprotein A-I (Apo A-I) from the blood, in particular when the SiNPs were preadsorbed with pulmonary surfactants. While the adsorbed Apo A-I ameliorated the cytotoxic and proinflammatory effects of SiNPs, the protein was eliminated from the blood upon clearance of the NPs. However, supplementation of Apo A-I mimic peptide mitigated the atherosclerotic lesion induced by SiNPs. In addition, we found a further declined plasma Apo A-I level in clinical silicosis patients than coronary heart disease patients, suggesting clearance of SiNPs sequestered Apo A-I to compromise the coronal protein's regular biological functions. Together, this study has provided evidence that the protein corona of SiNPs acquired in the blood depletes Apo A-I, a biomarker for prediction of cardiovascular diseases, which gives rise to unexpected toxic effects of the nanoparticles.


Asunto(s)
Apolipoproteína A-I/deficiencia , Enfermedades Cardiovasculares/etiología , Nanopartículas/efectos adversos , Adsorción/efectos de los fármacos , Animales , Apolipoproteína A-I/sangre , Sistema Cardiovascular , Pulmón , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/química , Nanotecnología , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Transducción de Señal/efectos de los fármacos , Dióxido de Silicio/efectos adversos , Dióxido de Silicio/química
3.
J Lipid Res ; 61(3): 328-337, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31915139

RESUMEN

Liver-derived serum amyloid A (SAA) is present in plasma where it is mainly associated with HDL and from which it is cleared more rapidly than are the other major HDL-associated apolipoproteins. Although evidence suggests that lipid-free and HDL-associated forms of SAA have different activities, the pathways by which SAA associates and disassociates with HDL are poorly understood. In this study, we investigated SAA lipidation by hepatocytes and how this lipidation relates to the formation of nascent HDL particles. We also examined hepatocyte-mediated clearance of lipid-free and HDL-associated SAA. We prepared hepatocytes from mice injected with lipopolysaccharide or an SAA-expressing adenoviral vector. Alternatively, we incubated primary hepatocytes from SAA-deficient mice with purified SAA. We analyzed conditioned media to determine the lipidation status of endogenously produced and exogenously added SAA. Examining the migration of lipidated species, we found that SAA is lipidated and forms nascent particles that are distinct from apoA-I-containing particles and that apoA-I lipidation is unaltered when SAA is overexpressed or added to the cells, indicating that SAA is not incorporated into apoA-I-containing HDL during HDL biogenesis. Like apoA-I formation, generation of SAA-containing particles was dependent on ABCA1, but not on scavenger receptor class B type I. Hepatocytes degraded significantly more SAA than apoA-I. Taken together, our results indicate that SAA's lipidation and metabolism by the liver is independent of apoA-I and that SAA is not incorporated into HDL during HDL biogenesis.


Asunto(s)
Lipoproteínas HDL/metabolismo , Proteína Amiloide A Sérica/metabolismo , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/metabolismo , Hepatocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Amiloide A Sérica/deficiencia , Proteína Amiloide A Sérica/genética
4.
Respir Res ; 21(1): 293, 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33148285

RESUMEN

BACKGROUND: Septic-acute respiratory distress syndrome (ARDS), characterized by the acute lung injury (ALI) secondary to aberrant systemic inflammatory response, has high morbidity and mortality. Despite increased understanding of ALI pathogenesis, the therapies to prevent lung dysfunction underlying systemic inflammatory disorder remain elusive. The high density lipoprotein (HDL) has critical protective effects in sepsis and its dysfunction has a manifested contribution to septic organ failure. However, the adverse changes in HDL composition and function in septic-ARDS patients are large unknown. METHODS: To investigate HDL remodeling in septic-ARDS, we analyzed the changes of HDL composition from 40 patients with septic-ARDS (A-HDL) and 40 matched normal controls (N-HDL). To determine the deleterious functional remodeling of HDL, A-HDL or N-HDL was administrated to C57BL/6 and apoA-I knock-out (KO) mice after cecal ligation and puncture (CLP) procedure. Mouse lung microvascular endothelial cells (MLECs) were further treated by these HDLs to investigate whether the adverse effects of A-HDL were associated with endothelial dysfunction. RESULTS: Septic-ARDS patients showed significant changes of HDL composition, accompanied with significantly decreased HDL-C. We further indicated that A-HDL treatment aggravated CLP induced ALI. Intriguingly, these deleterious effects of A-HDL were associated with pulmonary endothelial dysfunction, rather than the increased plasma lipopolysaccharide (LPS). Further in vitro results demonstrated the direct effects of A-HDL on MLECs, including increased endothelial permeability, enhanced expressions of adhesion proteins and pro-inflammatory cytokines via activating NF-κB signaling and decreased junction protein expression. CONCLUSIONS: Our results depicted the remodeling of HDL composition in sepsis, which predisposes lung to ARDS via inducing ECs dysfunction. These results also demonstrated the importance of circulating HDL in regulating alveolar homeostasis.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Células Endoteliales/metabolismo , Lipoproteínas HDL/toxicidad , Pulmón/irrigación sanguínea , Microvasos/metabolismo , Síndrome de Dificultad Respiratoria/etiología , Sepsis/complicaciones , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Permeabilidad Capilar , Estudios de Casos y Controles , Ciego/microbiología , Ciego/cirugía , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Ligadura , Lipoproteínas HDL/sangre , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Punciones , Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/patología , Sepsis/microbiología , Proteínas de Uniones Estrechas/metabolismo , Adulto Joven
5.
J Proteome Res ; 18(1): 48-56, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30543107

RESUMEN

Type 2 diabetes mellitus (T2DM) has become a tremendous problem in public health nowadays. High-density lipoprotein (HDL) refers to a group of heterogeneous particles that circulate in blood, and a recent research finds that HDL acts a pivotal part of glucose metabolism. To understand systemic metabolic changes correlated with HDL in glucose metabolism, we applied LC-MS-based metabolomics and lipidomics to detect metabolomic and lipidomic profiles of plasma from apoA-I knockout mice fed a high-fat diet. Multivariate analysis was applied to differentiate apoA-I knockout mice and controls, and potential biomarkers were found. Pathway analysis demonstrated that several metabolic pathways such as aminoacyl-tRNA biosynthesis, arginine and proline metabolism, and phenylalanine, tyrosine, and tryptophan biosynthesis were dysregulated in apoA-I knockout mice. This study may provide a new insight into the underlying pathogenesis in T2DM and prove that LC-MS-based metabolomics and lipidomics are powerful approaches in finding potential biomarkers and disturbed pathways.


Asunto(s)
Glucosa/metabolismo , Lipidómica/métodos , Lipoproteínas HDL , Metabolómica/métodos , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Cromatografía Liquida , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Espectrometría de Masas en Tándem
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 991-997, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29852278

RESUMEN

Proprotein convertase subtilisin/kexin 9 (PCSK9), a protein regulating the number of cell-surface LDL receptors (LDLR), circulates partially associated to plasma lipoproteins. How this interaction alters PCSK9 plasma levels is still unclear. In the present study, we took advantage of the availability of a large cohort of carriers of genetic HDL disorders to evaluate how HDL defects affect plasma PCSK9 levels and its distribution among lipoproteins. Plasma PCSK9 concentrations were determined by ELISA in carriers of mutations in LCAT, ABCA1, or APOAI genes, and lipoprotein distribution was analyzed by FPLC. Carriers of one or two mutations in the LCAT gene show plasma PCSK9 levels comparable to that of unaffected family controls (homozygotes, 159.4 ng/mL (124.9;243.3); heterozygotes, 180.3 ng/mL (127.6;251.5) and controls, 190.4 ng/mL (146.7;264.4); P for trend = 0.33). Measurement of PCSK9 in plasma of subjects carrying mutations in ABCA1 or APOAI genes confirmed normal values. When fractionated by FPLC, PCSK9 peaked in a region between LDL and HDL in control subjects. In carriers of all HDL defects, lipoprotein profile shows a strong reduction of HDL, but the distribution of PCSK9 was superimposable to that of controls. In conclusion, the present study demonstrates that in genetically determined low HDL states plasma PCSK9 concentrations and lipoprotein distribution are preserved, thus suggesting that HDL may not be involved in PCSK9 transport in plasma.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/sangre , Apolipoproteína A-I/sangre , Hipolipoproteinemias/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Proproteína Convertasa 9/sangre , Transportador 1 de Casete de Unión a ATP/deficiencia , Transportador 1 de Casete de Unión a ATP/genética , Adulto , Anciano , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Heterocigoto , Homocigoto , Humanos , Hipolipoproteinemias/genética , Hipolipoproteinemias/patología , Lipoproteínas HDL/sangre , Lipoproteínas HDL/genética , Lipoproteínas LDL/sangre , Lipoproteínas LDL/genética , Masculino , Persona de Mediana Edad , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Proproteína Convertasa 9/genética
7.
J Lipid Res ; 58(5): 994-1001, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28351888

RESUMEN

The aim of this study was to evaluate the vasoprotective effects of HDL isolated from carriers of LCAT deficiency, which are characterized by a selective depletion of LpA-I:A-II particles and predominance of preß migrating HDL. HDLs were isolated from LCAT-deficient carriers and tested in vitro for their capacity to promote NO production and to inhibit vascular cell adhesion molecule-1 (VCAM-1) expression in cultured endothelial cells. HDLs from carriers were more effective than control HDLs in promoting eNOS activation with a gene-dose-dependent effect (PTrend = 0.048). As a consequence, NO production induced by HDL from carriers was significantly higher than that promoted by control HDL (1.63 ± 0.24-fold vs. 1.34 ± 0.07-fold, P = 0.031). HDLs from carriers were also more effective than control HDLs in inhibiting the expression of VCAM-1 (homozygotes, 65.0 ± 8.6%; heterozygotes, 53.1 ± 7.2%; controls, 44.4 ± 4.1%; PTrend = 0.0003). The increased efficiency of carrier HDL was likely due to the depletion in LpA-I:A-II particles. The in vitro findings might explain why carriers of LCAT deficiency showed flow-mediated vasodilation and plasma-soluble cell adhesion molecule concentrations comparable to controls, despite low HDL-cholesterol levels. These results indicate that selective depletion of apoA-II-containing HDL, as observed in carriers of LCAT deficiency, leads to an increased capacity of HDL to stimulate endothelial NO production, suggesting that changes in HDL apolipoprotein composition may be the target of therapeutic interventions designed to improve HDL functionality.


Asunto(s)
Apolipoproteína A-II/deficiencia , Apolipoproteína A-I/deficiencia , Células Endoteliales/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteínas HDL/metabolismo , Adulto , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 36(7): 1356-66, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27150392

RESUMEN

OBJECTIVE: Plasma levels of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (ApoA-I) are reduced in individuals with defective insulin signaling. Initial studies using liver-specific insulin receptor (InsR) knockout mice identified reduced expression of type 1 deiodinase (Dio1) as a potentially novel link between defective hepatic insulin signaling and reduced expression of the ApoA-I gene. Our objective was to examine the regulation of ApoA-I expression by Dio1. APPROACH AND RESULTS: Acute inactivation of InsR by adenoviral delivery of Cre recombinase to InsR floxed mice reduced HDL-C and expression of both ApoA-I and Dio1. Overexpression of Dio1 in InsR knockout mice restored HDL-C and ApoA-I levels and increased the expression of ApoA-I. Dio1 knockout mice had low expression of ApoA-I and reduced serum levels of HDL-C and ApoA-I. Treatment of C57BL/6J mice with antisense to Dio1 reduced ApoA-I mRNA, HDL-C, and serum ApoA-I. Hepatic 3,5,3'-triiodothyronine content was normal or elevated in InsR knockout mice or Dio1 knockout mice. Knockdown of either InsR or Dio1 by siRNA in HepG2 cells decreased the expression of ApoA-I and ApoA-I synthesis and secretion. siRNA knockdown of InsR or Dio1 decreased activity of a region of the ApoA-I promoter lacking thyroid hormone response elements (region B). Electrophoretic mobility shift assay demonstrated that reduced Dio1 expression decreased the binding of nuclear proteins to region B. CONCLUSIONS: Reductions in Dio1 expression reduce the expression of ApoA-I in a 3,5,3'-triiodothyronine-/thyroid hormone response element-independent manner.


Asunto(s)
Apolipoproteína A-I/metabolismo , Yoduro Peroxidasa/metabolismo , Hígado/enzimología , Transducción de Señal , Triyodotironina/metabolismo , Animales , Apolipoproteína A-I/sangre , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , HDL-Colesterol/sangre , Regulación de la Expresión Génica , Genotipo , Células Hep G2 , Humanos , Yoduro Peroxidasa/deficiencia , Yoduro Peroxidasa/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Insulina/deficiencia , Receptor de Insulina/genética , Elementos de Respuesta , Transfección
9.
BMC Ophthalmol ; 17(1): 11, 2017 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-28178939

RESUMEN

BACKGROUND: Apo A-I deficiency clinically shows low serum levels of HDL cholesterol and corneal opacity at a young age. Histopathological evaluations of affected corneas are not enough, and the mechanism of corneal opacity is still unclear. CASE PRESENTATION: A 61-year-old woman suffered from blurred vision with a corneal opacity. She had significantly reduced serum levels of high-density lipoprotein cholesterol and Apo A-I, stenosis of the coronary arteries, and ischemic heart failure. On genetic examination, a homozygous mutation of Apo A-ITsukuba was identified. Histopathological examination of the corneal button after PKP showed numerous vesicles in the corneal stroma, which were more prominent in the deep stroma than in the shallow stroma. Collagen VI was observed in some of those vesicles. CONCLUSION: We experienced a rare case of corneal opacity due to Apo A-I deficiency. Our histopathological findings indicated that structural changes in corneal collagen fibrils contribute to the formation of stromal vesicles.


Asunto(s)
Apolipoproteína A-I/deficiencia , Colágeno Tipo VI/metabolismo , Córnea/patología , Opacidad de la Córnea/etiología , Dislipidemias/complicaciones , Apolipoproteína A-I/sangre , Córnea/metabolismo , Opacidad de la Córnea/diagnóstico , Opacidad de la Córnea/metabolismo , Dislipidemias/diagnóstico , Dislipidemias/metabolismo , Femenino , Humanos , Inmunohistoquímica , Microscopía Confocal , Persona de Mediana Edad
10.
Biochim Biophys Acta ; 1851(9): 1254-61, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26037829

RESUMEN

BACKGROUND: Low plasma levels of high-density lipoprotein-cholesterol (HDL-C) are typical of acute myocardial infarction (MI) and predict risk of recurrent cardiovascular events. The potential relationships between modifications in the molecular composition and the functionality of HDL subpopulations in acute MI however remain indeterminate. METHODS AND RESULTS: ST segment elevation MI (STEMI) patients were recruited within 24h after diagnosis (n=16) and featured low HDL-C (-31%, p<0.05) and acute-phase inflammation (determined as marked elevations in C-reactive protein, serum amyloid A (SAA) and interleukin-6) as compared to age- and sex-matched controls (n=10). STEMI plasma HDL and its subpopulations (HDL2b, 2a, 3a, 3b, 3c) displayed attenuated cholesterol efflux capacity from THP-1 cells (up to -32%, p<0.01, on a unit phospholipid mass basis) vs. CONTROLS: Plasma HDL and small, dense HDL3b and 3c subpopulations from STEMI patients exhibited reduced anti-oxidative activity (up to -68%, p<0.05, on a unit HDL mass basis). HDL subpopulations in STEMI were enriched in two proinflammatory bioactive lipids, lysophosphatidylcholine (up to 3.0-fold, p<0.05) and phosphatidic acid (up to 8.4-fold, p<0.05), depleted in apolipoprotein A-I (up to -23%, p<0.05) and enriched in SAA (up to +10.2-fold, p<0.05); such changes were most marked in the HDL3b subfraction. In vitro HDL enrichment in both lysophosphatidylcholine and phosphatidic acid exerted deleterious effects on HDL functionality. CONCLUSIONS: In the early phase of STEMI, HDL particle subpopulations display marked, concomitant alterations in both lipidome and proteome which are implicated in impaired HDL functionality. Such modifications may act synergistically to confer novel deleterious biological activities to STEMI HDL. SIGNIFICANCE: Our present data highlight complex changes in the molecular composition and functionality of HDL particle subpopulations in the acute phase of STEMI, and for the first time, reveal that concomitant modifications in both the lipidome and proteome contribute to functional deficiencies in cholesterol efflux and antioxidative activities of HDL particles. These findings may provide new biomarkers and new insights in therapeutic strategy to reduce cardiovascular risk in this clinical setting where such net deficiency in HDL function, multiplied by low circulating HDL concentrations, can be expected to contribute to accelerated atherogenesis.


Asunto(s)
Lipoproteínas HDL3/sangre , Lisofosfatidilcolinas/sangre , Infarto del Miocardio/sangre , Ácidos Fosfatidicos/sangre , Proteína Amiloide A Sérica/metabolismo , Adulto , Anciano , Apolipoproteína A-I/química , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/metabolismo , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Línea Celular , Femenino , Humanos , Interleucina-6/sangre , Lipoproteínas HDL3/química , Lisofosfatidilcolinas/química , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Infarto del Miocardio/patología , Ácidos Fosfatidicos/química , Proteoma/química , Proteoma/metabolismo
11.
Lab Invest ; 96(7): 763-72, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27088511

RESUMEN

Imbalances in lipid metabolism affect bone homeostasis, altering bone mass and quality. A link between bone mass and high-density lipoprotein (HDL) has been proposed. Indeed, it has been recently shown that absence of the HDL receptor scavenger receptor class B type I (SR-B1) causes dense bone mediated by increased adrenocorticotropic hormone (ACTH). In the present study we aimed at further expanding the current knowledge as regards the fascinating bone-HDL connection studying bone turnover in apoA-1-deficient mice. Interestingly, we found that bone mass was greatly reduced in the apoA-1-deficient mice compared with their wild-type counterparts. More specifically, static and dynamic histomorphometry showed that the reduced bone mass in apoA-1(-/-) mice reflect decreased bone formation. Biochemical composition and biomechanical properties of ApoA-1(-/-) femora were significantly impaired. Mesenchymal stem cell (MSC) differentiation from the apoA-1(-/-) mice showed reduced osteoblasts, and increased adipocytes, relative to wild type, in identical differentiation conditions. This suggests a shift in MSC subtypes toward adipocyte precursors, a result that is in line with our finding of increased bone marrow adiposity in apoA-1(-/-) mouse femora. Notably, osteoclast differentiation in vitro and osteoclast surface in vivo were unaffected in the knock-out mice. In whole bone marrow, PPARγ was greatly increased, consistent with increased adipocytes and committed precursors. Further, in the apoA-1(-/-) mice marrow, CXCL12 and ANXA2 levels were significantly decreased, whereas CXCR4 were increased, consistent with reduced signaling in a pathway that supports MSC homing and osteoblast generation. In keeping, in the apoA-1(-/-) animals the osteoblast-related factors Runx2, osterix, and Col1a1 were also decreased. The apoA-1(-/-) phenotype also included augmented CEPBa levels, suggesting complex changes in growth and differentiation that deserve further investigation. We conclude that the apoA-1 deficiency generates changes in the bone cell precursor population that increase adipoblast, and decrease osteoblast production resulting in reduced bone mass and impaired bone quality in mice.


Asunto(s)
Adipocitos/metabolismo , Apolipoproteína A-I/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Adipocitos/citología , Adipogénesis , Hormona Adrenocorticotrópica/metabolismo , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Densidad Ósea , Diferenciación Celular , Quimiocina CXCL12/genética , Hidrocortisona/biosíntesis , Lipoproteínas HDL/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Receptores de Lipoproteína/metabolismo , Receptores Depuradores de Clase B/genética
12.
Arterioscler Thromb Vasc Biol ; 35(8): 1826-34, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26044581

RESUMEN

OBJECTIVE: Dyslipidemia is implicated in abdominal aortic aneurysms (AAAs) in humans and angiotensin (Ang) II-infused mice. This study determined effects of major lipoprotein classes on AngII-induced AAAs using multiple mouse strains with dietary and pharmacological manipulations. APPROACH AND RESULTS: Western diet had minor effects on plasma cholesterol concentrations and the low incidence of AngII-induced AAAs in C57BL/6J mice. Low incidence of AAAs in this strain was not attributed to protection from high-density lipoprotein, because apolipoprotein (apo) AI deficiency did not increase AngII-induced AAAs. ApoAI deletion also failed to alter AAA occurrence in hypercholesterolemic mice. Low-density lipoprotein receptor-/- mice fed normal diet had low incidence of AngII-induced AAAs. Western diet feeding of this strain provoked pronounced hypercholesterolemia because of increased apoB-containing lipoproteins with attendant increases of atherosclerosis in both sexes, but AAAs only in male mice. ApoE-deficient mice fed normal diet were modestly hypercholesterolemic, whereas this strain fed Western diet was severely hypercholesterolemic because of increased apoB-containing lipoprotein concentrations. The latter augmented atherosclerosis, but did not change the high incidence of AAAs in this strain. To determine whether reductions in apoB-containing lipoproteins influenced AngII-induced AAAs, ezetimibe was administered at a dose that partially reduced plasma cholesterol concentrations to ApoE-deficient mice fed Western diet. This decreased atherosclerosis, but not AAAs. This ezetimibe dose in ApoE-deficient mice fed normal diet significantly decreased plasma apoB-containing lipoprotein concentrations and reduced AngII-induced AAAs. CONCLUSIONS: ApoB-containing lipoproteins contribute to augmentation of AngII-induced AAA in male mice. However, unlike atherosclerosis, AAA occurrence was not correlated with increases in plasma apoB-containing lipoprotein concentrations.


Asunto(s)
Angiotensina II , Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/inducido químicamente , Apolipoproteína A-I/sangre , Apolipoproteínas B/sangre , Hipercolesterolemia/complicaciones , Animales , Anticolesterolemiantes/farmacología , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/prevención & control , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Apolipoproteína B-100 , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Azetidinas/farmacología , Dieta Occidental , Modelos Animales de Enfermedad , Ezetimiba , Femenino , Hipercolesterolemia/sangre , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores Sexuales
13.
Brain ; 138(Pt 12): 3699-715, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26510953

RESUMEN

UNLABELLED: ATP binding cassette transporter A1 (encoded by ABCA1) regulates cholesterol efflux from cells to apolipoproteins A-I and E (ApoA-I and APOE; encoded by APOA1 and APOE, respectively) and the generation of high density lipoproteins. In Abca1 knockout mice (Abca1(ko)), high density lipoproteins and ApoA-I are virtually lacking, and total APOE and APOE-containing lipoproteins in brain substantially decreased. As the ε4 allele of APOE is the major genetic risk factor for late-onset Alzheimer's disease, ABCA1 role as a modifier of APOE lipidation is of significance for this disease. Reportedly, Abca1 deficiency in mice expressing human APP accelerates amyloid deposition and behaviour deficits. We used APP/PS1dE9 mice crossed to Apoe and Apoa1 knockout mice to generate Apoe/Apoa1 double-knockout mice. We hypothesized that Apoe/Apoa1 double-knockout mice would mimic the phenotype of APP/Abca1(ko) mice in regards to amyloid plaques and cognitive deficits. Amyloid pathology, peripheral lipoprotein metabolism, cognitive deficits and dendritic morphology of Apoe/Apoa1 double-knockout mice were compared to APP/Abca1(ko), APP/PS1dE9, and single Apoa1 and Apoe knockouts. Contrary to our prediction, the results demonstrate that double deletion of Apoe and Apoa1 ameliorated the amyloid pathology, including amyloid plaques and soluble amyloid. In double knockout mice we show that (125)I-amyloid-ß microinjected into the central nervous system cleared at a rate twice faster compared to Abca1 knockout mice. We tested the effect of Apoe, Apoa1 or Abca1 deficiency on spreading of exogenous amyloid-ß seeds injected into the brain of young pre-depositing APP mice. The results show that lack of Abca1 augments dissemination of exogenous amyloid significantly more than the lack of Apoe. In the periphery, Apoe/Apoa1 double-knockout mice exhibited substantial atherosclerosis and very high levels of low density lipoproteins compared to APP/PS1dE9 and APP/Abca1(ko). Plasma level of amyloid-ß42 measured at several time points for each mouse was significantly higher in Apoe/Apoa1 double-knockout then in APP/Abca1(ko) mice. This result demonstrates that mice with the lowest level of plasma lipoproteins, APP/Abca1(ko), have the lowest level of peripheral amyloid-ß. Unexpectedly, and independent of amyloid pathology, the deletion of both apolipoproteins worsened behaviour deficits of double knockout mice and their performance was undistinguishable from those of Abca1 knockout mice. Finally we observed that the dendritic complexity in the CA1 region of hippocampus but not in CA2 is significantly impaired by Apoe/Apoa1 double deletion as well as by lack of ABCA1. IN CONCLUSION: (i) plasma lipoproteins may affect amyloid-ß clearance from the brain by the 'peripheral sink' mechanism; and (ii) deficiency of brain APOE-containing lipoproteins is of significance for dendritic complexity and cognition.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Apolipoproteína A-I/deficiencia , Apolipoproteínas E/deficiencia , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/psicología , Eliminación de Gen , Placa Amiloide/genética , Transportador 1 de Casete de Unión a ATP/genética , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/sangre , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacocinética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Apolipoproteína A-I/genética , Apolipoproteínas E/genética , Encéfalo/metabolismo , Encéfalo/patología , Trastornos del Conocimiento/patología , Femenino , Hipocampo/metabolismo , Lipoproteínas/sangre , Masculino , Ratones , Ratones Noqueados , Microinyecciones , Neuritas/patología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacocinética , Placa Amiloide/patología , Placa Amiloide/psicología
14.
Arterioscler Thromb Vasc Biol ; 34(2): 262-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24334873

RESUMEN

OBJECTIVE: Inbred mouse strains have different susceptibilities to experimental atherosclerosis. The C57BL/6 strain is among the most sensitive and has, therefore, been the most widely used in atherosclerosis studies, whereas many strains are resistant. The FVB/N strain is highly resistant to atherosclerosis on the apolipoprotein E (apoE)- and low-density lipoprotein (LDL) receptor-deficient backgrounds. High-density lipoprotein and its major apoprotein, apoA-I, have been shown to be protective against atherogenesis on the C57BL/6 background. We here examine the influence of genetic background on the atheroprotective nature of apoA-I. APPROACH AND RESULTS: ApoE-deficient/apoA-I-deficient mice were generated in the C57BL/6 and FVB/N strains from apoE-deficient mice. After 6 to 10 weeks on a Western-type diet, plasma lipids and atherosclerotic lesion size were assessed. Macrophage recruitment, cholesterol regulation, and blood monocyte levels were examined as potential mechanisms driving lesion size differences. FVB/N knockout mice had higher plasma very-LDL/LDL cholesterol than their C57BL/6 counterparts. ApoA-I deficiency decreased very-LDL/LDL cholesterol in C57BL/6 mice but not in FVB/N mice. FVB/N single and double knockout mice had less lesion than C57BL/6 6 to 10 weeks on diet. ApoA-I deficiency augmented lesion development only in C57BL/6 mice. Macrophage recruitment to thioglycollate-treated peritoneum and diet-induced blood monocyte levels reflected the pattern of lesion development among the 4 genotypes. ApoA-I deficiency increased macrophage cholesterol content only in C57BL/6. FVB/N plasma was a better acceptor for macrophage cholesterol efflux than C57BL/6. CONCLUSIONS: ApoA-I is atheroprotective only in certain genetic contexts. In the C57BL/6 context, but not FVB/N, apoA-I decreases inflammatory macrophage recruitment and monocytosis, contributors to lesion formation.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Apolipoproteína A-I/metabolismo , Aterosclerosis/prevención & control , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Línea Celular , Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Especificidad de la Especie , Factores de Tiempo , Triglicéridos/sangre
15.
Arterioscler Thromb Vasc Biol ; 34(4): 779-89, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24407029

RESUMEN

OBJECTIVE: Preclinical and clinical studies have shown beneficial effects of infusions of apolipoprotein A-I (ApoA-I) on atherosclerosis. ApoA-I is also a target for myeloperoxidase-mediated oxidation, leading in vitro to a loss of its ability to promote ATP-binding cassette transporter A1-dependent macrophage cholesterol efflux. Therefore, we hypothesized that myeloperoxidase-mediated ApoA-I oxidation would impair its promotion of reverse cholesterol transport in vivo and the beneficial effects on atherosclerotic plaques. APPROACH AND RESULTS: ApoA-I(-/-) or apolipoprotein E-deficient mice were subcutaneously injected with native human ApoA-I, oxidized human ApoA-I (myeloperoxidase/hydrogen peroxide/chloride treated), or carrier. Although early postinjection (8 hours) levels of total ApoA-I in plasma were similar for native versus oxidized human ApoA-I, native ApoA-I primarily resided within the high-density lipoprotein fraction, whereas the majority of oxidized human ApoA-I was highly cross-linked and not high-density lipoprotein particle associated, consistent with impaired ATP-binding cassette transporter A1 interaction. In ApoA-I(-/-) mice, ApoA-I oxidation significantly impaired reverse cholesterol transport in vivo. In advanced aortic root atherosclerotic plaques of apolipoprotein E-deficient mice, native ApoA-I injections led to significant decreases in lipid content, macrophage number, and an increase in collagen content; in contrast, oxidized human ApoA-I failed to mediate these changes. The decrease in plaque macrophages with native ApoA-I was accompanied by significant induction of their chemokine receptor CCR7. Furthermore, only native ApoA-I injections led to a significant reduction of inflammatory M1 and increase in anti-inflammatory M2 macrophage markers in the plaques. CONCLUSIONS: Myeloperoxidase-mediated oxidation renders ApoA-I dysfunctional and unable to (1) promote reverse cholesterol transport, (2) mediate beneficial changes in the composition of atherosclerotic plaques, and (3) pacify the inflammatory status of plaque macrophages.


Asunto(s)
Apolipoproteína A-I/sangre , Aterosclerosis/enzimología , Colesterol/sangre , Inflamación/enzimología , Macrófagos/enzimología , Peroxidasa/metabolismo , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteína A-I/administración & dosificación , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Transporte Biológico , Línea Celular , HDL-Colesterol/sangre , Colágeno/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación/sangre , Inflamación/genética , Inflamación/patología , Inflamación/prevención & control , Inyecciones Subcutáneas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Placa Aterosclerótica , Receptores CCR7/metabolismo
16.
J Lipid Res ; 55(12): 2509-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25341944

RESUMEN

To evaluate functional and compositional properties of HDL in subjects from a kindred of genetic apoA-I deficiency, two homozygotes and six heterozygotes, with a nonsense mutation at APOA1 codon -2, Q[-2]X, were recruited together with age- and sex-matched healthy controls (n = 11). Homozygotes displayed undetectable plasma levels of apoA-I and reduced levels of HDL-cholesterol (HDL-C) and apoC-III (5.4% and 42.6% of controls, respectively). Heterozygotes displayed low HDL-C (21 ± 9 mg/dl), low apoA-I (79 ± 24 mg/dl), normal LDL-cholesterol (132 ± 25 mg/dl), and elevated TG (130 ± 45 mg/dl) levels. Cholesterol efflux capacity of ultracentrifugally isolated HDL subpopulations was reduced (up to -25%, P < 0.01, on a glycerophospholipid [GP] basis) in heterozygotes versus controls. Small, dense HDL3 and total HDL from heterozygotes exhibited diminished antioxidative activity (up to -48%, P < 0.001 on a total mass basis) versus controls. HDL subpopulations from both homozygotes and heterozygotes displayed altered chemical composition, with depletion in apoA-I, GP, and cholesteryl ester; enrichment in apoA-II, free cholesterol, and TG; and altered phosphosphingolipidome. The defective atheroprotective activities of HDL were correlated with altered lipid and apo composition. These data reveal that atheroprotective activities of HDL particles are impaired in homozygous and heterozygous apoA-I deficiency and are intimately related to marked alterations in protein and lipid composition.


Asunto(s)
Apolipoproteína A-I/deficiencia , Apolipoproteína C-III/sangre , Proteínas de Transferencia de Ésteres de Colesterol/sangre , HDL-Colesterol/sangre , Hipoalfalipoproteinemias/sangre , Lipoproteínas HDL/sangre , Adulto , Apolipoproteína A-I/sangre , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Apolipoproteína C-III/metabolismo , Brasil , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Ésteres del Colesterol/sangre , Ésteres del Colesterol/metabolismo , HDL-Colesterol/metabolismo , LDL-Colesterol/sangre , LDL-Colesterol/metabolismo , Codón sin Sentido , Salud de la Familia , Femenino , Heterocigoto , Homocigoto , Humanos , Hipoalfalipoproteinemias/genética , Hipoalfalipoproteinemias/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL3/sangre , Lipoproteínas HDL3/metabolismo , Masculino , Fosfolípidos/sangre , Fosfolípidos/metabolismo , Esfingolípidos/sangre , Esfingolípidos/metabolismo , Triglicéridos/sangre , Triglicéridos/metabolismo
17.
J Biol Chem ; 288(29): 21237-21252, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23720750

RESUMEN

Here, we show that apolipoprotein A1 (apoA1), the major protein component of high density lipoprotein (HDL), through both innate and adaptive immune processes, potently suppresses tumor growth and metastasis in multiple animal tumor models, including the aggressive B16F10L murine malignant melanoma model. Mice expressing the human apoA1 transgene (A1Tg) exhibited increased infiltration of CD11b(+) F4/80(+) macrophages with M1, anti-tumor phenotype, reduced tumor burden and metastasis, and enhanced survival. In contrast, apoA1-deficient (A1KO) mice showed markedly heightened tumor growth and reduced survival. Injection of human apoA1 into A1KO mice inoculated with tumor cells remarkably reduced both tumor growth and metastasis, enhanced survival, and promoted regression of both tumor and metastasis burden when administered following palpable tumor formation and metastasis development. Studies with apolipoprotein A2 revealed the anti-cancer therapeutic effect was specific to apoA1. In vitro studies ruled out substantial direct suppressive effects by apoA1 or HDL on tumor cells. Animal models defective in different aspects of immunity revealed both innate and adaptive arms of immunity contribute to complete apoA1 anti-tumor activity. This study reveals a potent immunomodulatory role for apoA1 in the tumor microenvironment, altering tumor-associated macrophages from a pro-tumor M2 to an anti-tumor M1 phenotype. Use of apoA1 to redirect in vivo elicited tumor-infiltrating macrophages toward tumor rejection may hold benefit as a potential cancer therapeutic.


Asunto(s)
Antineoplásicos/farmacología , Apolipoproteína A-I/metabolismo , Cardiotónicos/farmacología , Animales , Presentación de Antígeno/efectos de los fármacos , Antineoplásicos/uso terapéutico , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/farmacología , Apolipoproteína A-II/farmacología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunidad/efectos de los fármacos , Inmunocompetencia/efectos de los fármacos , Lipoproteínas HDL/metabolismo , Lisofosfolípidos/sangre , Lisofosfolípidos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Inducción de Remisión , Análisis de Supervivencia , Microambiente Tumoral/efectos de los fármacos
18.
Osteoarthritis Cartilage ; 21(2): 322-30, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23151457

RESUMEN

OBJECTIVE: Recent data suggest that obesity and related metabolic aberrations are associated with osteoarthritis (OA) development, a phenomenon that is attributed at least in part to the consumption of lipid-rich diets. To date, the molecular mechanisms that govern the lipid-OA connection remain largely unknown. Given the important role of high-density lipoprotein (HDL) in plasma and tissue lipid metabolism, the main purpose of the present study was to investigate the role of HDL metabolism in the pathobiology of OA. METHODS: We used apolipoprotein A-I (apoA-I)(-/-) mice that lack classical apoA-I containing HDL, LCAT(-/-) mice that have only immature HDL and relatively reduced HDL-cholesterol levels and control C57BL/6 mice. Mice were placed on chow or western-type (WTD) and monitored for 24 weeks. Knee joints were removed and articular cartilage was isolated for further analyses. RESULTS: The LCAT(-/-) mice were significantly more sensitive to the development of diet-induced obesity compared to the C57BL/6 and apoA-I(-/-) mice. Morphological, biochemical and molecular analyses revealed that the LCAT(-/-) obese mice developed OA, while the C57BL/6 mice that were fed WTD did not. Notably, apoA-I(-/-) mice that received WTD also developed OA although their body-weight gain was similar to their wild-type counterparts. Interestingly, bone marrow from LCAT(-/-) and apoA-I(-/-) mice contained significantly increased number of adipocytes, compared to the other groups. CONCLUSIONS: Our findings suggest that perturbations in HDL metabolism predispose to OA following chronic insult with WTD and raise the challenging possibility that HDL has a causative relation to OA in patients with metabolic syndrome.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Lipoproteínas HDL/metabolismo , Redes y Vías Metabólicas/fisiología , Obesidad/metabolismo , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/fisiopatología , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Peso Corporal/fisiología , Causalidad , Modelos Animales de Enfermedad , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/fisiopatología , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/fisiopatología , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Factores de Tiempo , Mundo Occidental
19.
Arterioscler Thromb Vasc Biol ; 32(6): 1460-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22499994

RESUMEN

OBJECTIVE: Reverse cholesterol transport (RCT) involves the removal of cholesterol from peripheral tissue for excretion in the feces. Here, we determined whether red blood cells (RBCs) can contribute to RCT. METHODS AND RESULTS: We performed a series of studies in apolipoprotein AI-deficient mice where the high-density lipoprotein-mediated pathway of RCT is greatly diminished. RBCs carried a higher fraction of whole blood cholesterol than plasma in apolipoprotein AI-deficient mice, and as least as much of the labeled cholesterol derived from injected foam cells appeared in RBCs compared with plasma. To determine whether RBCs mediate RCT to the fecal compartment, we measured RCT in anemic and control apolipoprotein AI-deficient mice and found that anemia decreased RCT to the feces by over 35% after correcting for fecal mass. Transfusion of [(3)H]cholesterol-labeled RBCs led to robust delivery of the labeled cholesterol to the feces in apolipoprotein AI-deficient hosts. In wild-type mice, the majority of the blood cholesterol mass, as well as [(3)H]cholesterol derived from the injected foam cells, was found in plasma, and anemia did not significantly alter RCT to the feces after correction for fecal mass. CONCLUSIONS: The RBC cholesterol pool is dynamic and facilitates RCT of peripheral cholesterol to the feces, particularly in the low high-density lipoprotein state.


Asunto(s)
Colesterol/sangre , Eritrocitos/metabolismo , Anemia/sangre , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Transporte Biológico , Células Cultivadas , HDL-Colesterol/sangre , Modelos Animales de Enfermedad , Transfusión de Eritrocitos , Heces/química , Células Espumosas/metabolismo , Hematócrito , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Tiempo , Tritio
20.
Nephrology (Carlton) ; 18(8): 549-54, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23730806

RESUMEN

AIM: Apolipoprotein A-I amyloidosis is a rare, autosomal dominant disorder characterized by progressive accumulation of amyloid fibrils in tissues, leading to renal and hepatic disease. We describe the clinical manifestations and pathologic features of kidney disease in three Irish families. METHODS: This observational study examines all known cases of chronic kidney disease due to hereditary apolipoprotein A-I amyloidosis in Ireland. Patients were identified by physician interview. In all of the affected individuals the disease was caused by the Gly26Arg heterozygous mutation. Immunohistochemistry confirmed that amyloid deposits were composed of apolipoprotein A-I fibrils. Family trees and clinical data were obtained via analysis of patient medical records. RESULTS: The vast majority of affected cases had demonstrable kidney disease, with variable liver disease. Renal disease most commonly manifested as slowly progressive renal impairment with mild proteinuria. In one kindred, a severe, debilitating peripheral neuropathy was common among affected family members. Histology demonstrated tubulointerstitial fibrosis with amyloid deposition in the medulla. There was very high penetrance within affected families. Of five patients who were transplanted, one transplant was lost after 5 years due to recurrent disease. One patient died from sepsis shortly after transplant. CONCLUSION: Hereditary apolipoprotein A-I amyloidosis is characterized by slowly progressive renal disease. Amyloid is deposited in the renal medulla highlighting the need to examine the medulla on renal biopsy. Overall, kidney transplantation conferred a survival advantage.


Asunto(s)
Amiloide/genética , Amiloidosis Familiar/genética , Apolipoproteína A-I/genética , Riñón/metabolismo , Mutación , Insuficiencia Renal Crónica/genética , Adulto , Anciano , Amiloide/metabolismo , Amiloidosis Familiar/complicaciones , Amiloidosis Familiar/diagnóstico , Amiloidosis Familiar/metabolismo , Amiloidosis Familiar/mortalidad , Apolipoproteína A-I/deficiencia , Biopsia , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Herencia , Heterocigoto , Humanos , Irlanda , Riñón/patología , Riñón/cirugía , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Proteinuria/genética , Proteinuria/metabolismo , Recurrencia , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/mortalidad , Insuficiencia Renal Crónica/cirugía , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA