Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Lipid Res ; 60(8): 1425-1431, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31133557

RESUMEN

apoM is a minor HDL apolipoprotein and carrier for sphingosine-1-phosphate (S1P). HDL apoM and S1P concentrations are inversely associated with atherosclerosis progression in rodents. We evaluated associations between plasma concentrations of S1P, plasma concentrations of apoM, and HDL apoM levels with prevalent subclinical atherosclerosis and mortality in the African American-Diabetes Heart Study participants (N = 545). Associations between plasma S1P, plasma apoM, and HDL apoM with subclinical atherosclerosis and mortality were assessed using multivariate parametric, nonparametric, and Cox proportional hazards models. At baseline, participants' median (25th percentile, 75th percentile) age was 55 (49, 62) years old and their coronary artery calcium (CAC) mass score was 26.5 (0.0, 346.5). Plasma S1P, plasma apoM, and HDL apoM were not associated with CAC. After 64 (57.6, 70.3) months of follow-up, 81 deaths were recorded. Higher concentrations of plasma S1P [odds ratio (OR) = 0.14, P = 0.01] and plasma apoM (OR = 0.10, P = 0.02), but not HDL apoM (P = 0.89), were associated with lower mortality after adjusting for age, sex, statin use, CAC, kidney function, and albuminuria. We conclude that plasma S1P and apoM concentrations are inversely and independently associated with mortality, but not CAC, in African Americans with type 2 diabetes after accounting for conventional risk factors.


Asunto(s)
Apolipoproteínas M/sangre , Negro o Afroamericano , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/mortalidad , Lisofosfolípidos/sangre , Esfingosina/análogos & derivados , Biomarcadores/sangre , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esfingosina/sangre , Tasa de Supervivencia
2.
Physiol Genomics ; 50(8): 605-614, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29750602

RESUMEN

Type 2 diabetes is a complex disorder affected by multiple genes and the environment. Our laboratory has shown that in response to a glucose challenge, two-pore channel 2 ( Tpcn2) knockout mice exhibit a decreased insulin response but normal glucose clearance, suggesting they have improved insulin sensitivity compared with wild-type mice. We tested the hypothesis that improved insulin sensitivity in Tpcn2 knockout mice would protect against the negative effects of a high fat diet. Male and female Tpcn2 knockout (KO), heterozygous (Het), and wild-type (WT) mice were fed a low-fat (LF) or high-fat (HF) diet for 24 wk. HF diet significantly increases body weight in WT mice relative to those on the LF diet; this HF diet-induced increase in body weight is blunted in the Het and KO mice. Despite the protection against diet-induced weight gain, however, Tpcn2 KO mice are not protected against HF-diet-induced changes in glucose or insulin area under the curve during glucose tolerance tests in female mice, while HF diet has no significant effect on glucose tolerance in the male mice, regardless of genotype. Glucose disappearance during an insulin tolerance test is augmented in male KO mice, consistent with our previous findings suggesting enhanced insulin sensitivity in these mice. Male KO mice exhibit increased fasting plasma total cholesterol and triglyceride concentrations relative to WT mice on the LF diet, but this difference disappears in HF diet-fed mice where there is increased cholesterol and triglycerides across all genotypes. These data demonstrate that knockout of Tpcn2 may increase insulin action in male, but not female, mice. In addition, both male and female KO mice are protected against diet-induced weight gain, but this protection is likely independent from glucose tolerance, insulin sensitivity, and plasma lipid levels.


Asunto(s)
Canales de Calcio/genética , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/genética , Obesidad/genética , Aumento de Peso/genética , Animales , Canales de Calcio/metabolismo , Colesterol/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Factores Sexuales , Triglicéridos/sangre
3.
Food Chem Toxicol ; 113: 287-295, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29421645

RESUMEN

Although epidemiological data and results from rodent studies support an inverse relationship between nicotine consumption and body weight, the molecular mechanisms are poorly understood. CD-1 mice were fed a basal diet or a basal diet containing low or high dose smokeless tobacco blend or high dose nicotine tartrate for 14 weeks. High dose tobacco blend and nicotine tartrate diets vs. basal diet reduced mouse body weight (16.3% and 19.7%, respectively), epididymal (67.6% and 72.5%, respectively) and brown adipose weight (42% and 38%, respectively), epididymal adipocyte size (46.4% and 41.4%, respectively), and brown adipose tissue lipid droplet abundance, with no elevation of adipose tissue inflammation. High dose tobacco blend and nicotine diets also increased mouse physical activity and decreased respiratory exchange ratio, suggesting that high dose nicotine intake induces adipose tissue triglyceride lipolysis to provide fatty acids as an energy source. Both low and high dose tobacco blend and nicotine diet feeding vs. basal diet increased plasma insulin levels (2.9, 3.6 and 4.3-fold, respectively) and improved blood glucose disposal without affecting insulin sensitivity. Feeding of the high dose tobacco blend or nicotine feeding in mice induces body weight loss likely by increasing physical activity and stimulating adipose tissue triglyceride lipolysis.


Asunto(s)
Adipocitos/efectos de los fármacos , Nicotiana , Nicotina/farmacología , Condicionamiento Físico Animal , Pérdida de Peso/efectos de los fármacos , Adipocitos/citología , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Tamaño de la Célula , Conducta Alimentaria , Insulina/sangre , Resistencia a la Insulina , Lipólisis , Masculino , Ratones , Triglicéridos/metabolismo
4.
Immunology ; 152(3): 484-493, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28672048

RESUMEN

Acquisition of a 'quiescence programme' by naive T cells is important to provide a stress-free environment and resistance to apoptosis while preserving their responsiveness to activating stimuli. Therefore, the survival and proper function of naive T cells depends on their ability to maintain quiescence. Recently we demonstrated that by preventing chronic unresolved endoplasmic reticulum (ER) stress, Schlafen2 (Slfn2) maintains a stress-free environment to conserve a pool of naive T cells ready to respond to a microbial invasion. These findings strongly suggest an intimate association between quiescence and stress signalling. However, the connection between ER stress conditions and loss of T-cell quiescence is unknown. Here we demonstrate that homeostasis of cholesterol and lipids, is disrupted in T cells and monocytes from Slfn2-mutant, elektra, mice with higher levels of lipid rafts and lipid droplets found in these cells. Moreover, elektra T cells had elevated levels of free cholesterol and cholesteryl ester due to increased de novo synthesis and higher levels of the enzyme HMG-CoA reductase. As cholesterol plays an important role in the transition of T cells from resting to active state, and ER regulates cholesterol and lipid synthesis, we suggest that regulation of cholesterol levels through the prevention of ER stress is an essential component of the mechanism by which Slfn2 regulates quiescence.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Senescencia Celular , Colesterol/biosíntesis , Activación de Linfocitos , Mutación , Linfocitos T/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Ésteres del Colesterol/biosíntesis , Estrés del Retículo Endoplásmico , Genotipo , Hidroximetilglutaril-CoA Reductasas/metabolismo , Gotas Lipídicas/metabolismo , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Mutantes , Monocitos/inmunología , Monocitos/metabolismo , Fenotipo , Linfocitos T/inmunología , Regulación hacia Arriba
5.
J Lipid Res ; 57(1): 120-30, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26586272

RESUMEN

APOL1 gene renal-risk variants are associated with nephropathy and CVD in African Americans; however, little is known about the circulating APOL1 variant proteins which reportedly bind to HDL. We examined whether APOL1 G1 and G2 renal-risk variant serum concentrations or lipoprotein distributions differed from nonrisk G0 APOL1 in African Americans without nephropathy. Serum APOL1 protein concentrations were similar regardless of APOL1 genotype. In addition, serum APOL1 protein was bound to protein complexes in two nonoverlapping peaks, herein referred to as APOL1 complex A (12.2 nm diameter) and complex B (20.0 nm diameter). Neither of these protein complexes associated with HDL or LDL. Proteomic analysis revealed that complex A was composed of APOA1, haptoglobin-related protein (HPR), and complement C3, whereas complex B contained APOA1, HPR, IgM, and fibronectin. Serum HPR was less abundant on complex B in individuals with G1 and G2 renal-risk variant genotypes, relative to G0 (P = 0.0002-0.037). These circulating complexes may play roles in HDL metabolism and susceptibility to CVD.


Asunto(s)
Apolipoproteínas/sangre , Negro o Afroamericano , Lipoproteínas HDL/sangre , Adulto , Apolipoproteína L1 , Apolipoproteínas/genética , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/genética , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Enfermedades Renales/sangre , Enfermedades Renales/genética , Lipoproteínas HDL/genética , Masculino , Persona de Mediana Edad , Proteómica , Factores de Riesgo
6.
J Biol Chem ; 290(12): 7861-70, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25627684

RESUMEN

Apolipoprotein M (apoM), a plasma sphingosine 1-phosphate (S1P) carrier, associates with plasma HDL via its uncleaved signal peptide. Hepatocyte-specific apoM overexpression in mice stimulates formation of both larger nascent HDL in hepatocytes and larger mature apoM/S1P-enriched HDL particles in plasma by enhancing hepatic S1P synthesis and secretion. Mutagenesis of apoM glutamine 22 to alanine (apoM(Q22A)) introduces a functional signal peptidase cleavage site. Expression of apoM(Q22A) in ABCA1-expressing HEK293 cells resulted in the formation of smaller nascent HDL particles compared with wild type apoM (apoM(WT)). When apoM(Q22A) was expressed in vivo, using recombinant adenoviruses, smaller plasma HDL particles and decreased plasma S1P and apoM were observed relative to expression of apoM(WT). Hepatocytes isolated from both apoM(WT)- and apoM(Q22A)-expressing mice displayed an equivalent increase in cellular levels of S1P, relative to LacZ controls; however, relative to apoM(WT), apoM(Q22A) hepatocytes displayed more rapid apoM and S1P secretion but minimal apoM(Q22A) bound to nascent lipoproteins. Pharmacologic inhibition of ceramide synthesis increased cellular sphingosine and S1P but not medium S1P in both apoM(WT) and apoM(Q22A) hepatocytes. We conclude that apoM secretion is rate-limiting for hepatocyte S1P secretion and that its uncleaved signal peptide delays apoM trafficking out of the cell, promoting formation of larger nascent apoM- and S1P-enriched HDL particles that are probably precursors of larger apoM/S1P-enriched plasma HDL.


Asunto(s)
Apolipoproteínas/metabolismo , Lipoproteínas HDL/química , Lisofosfolípidos/química , Señales de Clasificación de Proteína , Esfingosina/análogos & derivados , Animales , Apolipoproteínas/química , Apolipoproteínas M , Masculino , Ratones , Ratones Endogámicos C57BL , Esfingosina/química
7.
Arterioscler Thromb Vasc Biol ; 34(9): 1888-99, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24833800

RESUMEN

OBJECTIVE: Transplantation studies suggest that bone marrow cell ATP-binding cassette transporter A1 protects against atherosclerosis development. However, the in vivo effect of macrophage ATP-binding cassette transporter A1 expression on atherogenesis is not fully understood because bone marrow contains other leukocytes and hematopoietic stem and progenitor cells. Myeloid-specific ATP-binding cassette transporter A1 knockout mice in the low-density lipoprotein (LDL) receptor knockout C57BL/6 background were developed to address this question. APPROACH AND RESULTS: Chow-fed myeloid-specific ATP-binding cassette transporter A1 knockout/LDL receptor knockout (double knockout [DKO]) versus LDL receptor knockout (single knockout [SKO]) mice had similar plasma lipid concentrations, but atherogenic diet (AD)-fed DKO mice had reduced plasma very-LDL (VLDL)/LDL concentrations resulting from decreased hepatic VLDL triglyceride secretion. Resident peritoneal macrophages from AD-fed DKO versus SKO mice had significantly higher cholesterol content but similar proinflammatory gene expression. Atherosclerosis extent was similar between genotypes after 10 to 16 weeks of AD but increased modestly in DKO mice by 24 weeks of AD. Lesional macrophage content was similar, likely because of the higher monocyte flux through aortic root lesions in DKO versus SKO mice. After transplantation of DKO or SKO bone marrow into SKO mice and 16 weeks of AD feeding, atherosclerosis extent was similar and plasma apolipoprotein B lipoproteins were reduced in mice receiving DKO bone marrow. When differences in plasma VLDL/LDL concentrations were minimized by maintaining mice on chow for 24 weeks, DKO mice had modest, but significantly more, atherosclerosis compared with SKO mice. CONCLUSIONS: Myeloid cell ATP-binding cassette transporter A1 increases hepatic VLDL triglyceride secretion and plasma VLDL/LDL concentrations in AD-fed LDL receptor knockout mice, offsetting its atheroprotective role in decreasing macrophage cholesterol content, resulting in a minimal increase in atherosclerosis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/fisiología , Aterosclerosis/metabolismo , Colesterol/metabolismo , Dieta Aterogénica/efectos adversos , Macrófagos Peritoneales/metabolismo , Células Mieloides/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aterosclerosis/genética , Aterosclerosis/prevención & control , Trasplante de Médula Ósea , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética , Triglicéridos/sangre
8.
J Biol Chem ; 289(5): 2801-14, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24318881

RESUMEN

Apolipoprotein M (apoM), a lipocalin family member, preferentially associates with plasma HDL and binds plasma sphingosine 1-phosphate (S1P), a signaling molecule active in immune homeostasis and endothelial barrier function. ApoM overexpression in ABCA1-expressing HEK293 cells stimulated larger nascent HDL formation, compared with cells that did not express apoM; however, the in vivo role of apoM in HDL metabolism remains poorly understood. To test whether hepatic apoM overexpression increases plasma HDL size, we generated hepatocyte-specific apoM transgenic (APOM Tg) mice, which had an ∼3-5-fold increase in plasma apoM levels compared with wild-type mice. Although HDL cholesterol concentrations were similar to wild-type mice, APOM Tg mice had larger plasma HDLs enriched in apoM, cholesteryl ester, lecithin:cholesterol acyltransferase, and S1P. Despite the presence of larger plasma HDLs in APOM Tg mice, in vivo macrophage reverse cholesterol transport capacity was similar to that in wild-type mice. APOM Tg mice had an ∼5-fold increase in plasma S1P, which was predominantly associated with larger plasma HDLs. Primary hepatocytes from APOM Tg mice generated larger nascent HDLs and displayed increased sphingolipid synthesis and S1P secretion. Inhibition of ceramide synthases in hepatocytes increased cellular S1P levels but not S1P secretion, suggesting that apoM is rate-limiting in the export of hepatocyte S1P. Our data indicate that hepatocyte-specific apoM overexpression generates larger nascent HDLs and larger plasma HDLs, which preferentially bind apoM and S1P, and stimulates S1P biosynthesis for secretion. The unique apoM/S1P-enriched plasma HDL may serve to deliver S1P to extrahepatic tissues for atheroprotection and may have other as yet unidentified functions.


Asunto(s)
Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Hepatocitos/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Lipoproteínas HDL/metabolismo , Hígado/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Apolipoproteínas E/sangre , Apolipoproteínas M , Femenino , Humanos , Lipoproteínas HDL/sangre , Lipoproteínas HDL/química , Hígado/citología , Lisofosfolípidos/biosíntesis , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Tamaño de la Partícula , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Esfingosina/biosíntesis , Esfingosina/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 33(10): 2288-96, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23814116

RESUMEN

OBJECTIVE: Hepatic ATP binding cassette transporter A1 (ABCA1) expression is critical for maintaining plasma high-density lipoprotein (HDL) concentrations, but its role in macrophage reverse cholesterol transport and atherosclerosis is not fully understood. We investigated atherosclerosis development and reverse cholesterol transport in hepatocyte-specific ABCA1 knockout (HSKO) mice in the low-density lipoprotein (LDL) receptor KO (LDLrKO) C57BL/6 background. APPROACH AND RESULTS: Male and female LDLrKO and HSKO/LDLrKO mice were switched from chow at 8 weeks of age to an atherogenic diet (10% palm oil, 0.2% cholesterol) for 16 weeks. Chow-fed HSKO/LDLrKO mice had HDL concentrations 10% to 20% of LDLrKO mice, but similar very low-density lipoprotein and LDL concentrations. Surprisingly, HSKO/LDLrKO mice fed the atherogenic diet had significantly lower (40% to 60%) very low-density lipoprotein, LDL, and HDL concentrations (50%) compared with LDLrKO mice. Aortic surface lesion area and cholesterol content were similar for both genotypes of mice, but aortic root intimal area was significantly lower (20% to 40%) in HSKO/LDLrKO mice. Although macrophage (3)H-cholesterol efflux to apoB lipoprotein-depleted plasma was 24% lower for atherogenic diet-fed HSKO/LDLrKO versus LDLrKO mice, variation in percentage efflux among individual mice was <2-fold compared with a 10-fold variation in plasma HDL concentrations, suggesting that HDL levels, per se, were not the primary determinant of plasma efflux capacity. In vivo reverse cholesterol transport, resident peritoneal macrophage sterol content, biliary lipid composition, and fecal cholesterol mass were similar between both genotypes of mice. CONCLUSIONS: The markedly reduced plasma HDL pool in HSKO/LDLrKO mice is sufficient to maintain macrophage reverse cholesterol transport, which, along with reduced plasma very low-density lipoprotein and LDL concentrations, prevented the expected increase in atherosclerosis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/deficiencia , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Colesterol/metabolismo , Hígado/metabolismo , Macrófagos Peritoneales/metabolismo , Receptores de LDL/deficiencia , Transportador 1 de Casete de Unión a ATP/genética , Animales , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteína B-100 , Apolipoproteínas B/sangre , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Bilis/metabolismo , Transporte Biológico , Línea Celular , Colesterol/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Dieta Aterogénica , Modelos Animales de Enfermedad , Heces/química , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/genética , Factores de Tiempo
10.
J Lipid Res ; 54(10): 2708-17, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23894207

RESUMEN

Obesity-associated low-grade chronic inflammation plays an important role in the development of insulin resistance. The membrane lipid transporter ATP-binding cassette transporter A1 (ABCA1) promotes formation of nascent HDL particles. ABCA1 also dampens macrophage inflammation by reducing cellular membrane cholesterol and lipid raft content. We tested the hypothesis that myeloid-specific ABCA1 deletion may exacerbate insulin resistance by increasing the obesity-associated chronic low-grade inflammation. Myeloid cell-specific ABCA1 knockout (MSKO) and wild-type (WT) mice developed obesity, insulin resistance, mild hypercholesterolemia, and hepatic steatosis to a similar extent with a 45% high-fat (HF) diet feeding or after crossing into the ob/ob background. Resident peritoneal macrophages and stromal vascular cells from obese MSKO mice accumulated significantly more cholesterol. Relative to chow, HF diet markedly induced macrophage infiltration and inflammatory cytokine expression to a similar extent in adipose tissue of WT and MSKO mice. Among pro-inflammatory cytokines examined, only IL-6 was highly upregulated in MSKO-ob/ob versus ob/ob mouse peritoneal macrophages, indicating a nonsignificant effect of myeloid ABCA1 deficiency on obesity-associated chronic inflammation. In conclusion, myeloid-specific ABCA1 deficiency does not exacerbate obesity-associated low-grade chronic inflammation and has minimal impact on the pathogenesis of insulin resistance in both HF diet-induced and genetically obese mouse models.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/metabolismo , Transportador 1 de Casete de Unión a ATP/deficiencia , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Colesterol/sangre , Epidídimo/inmunología , Epidídimo/metabolismo , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Obesidad/etiología , Obesidad/inmunología
11.
Nutrients ; 5(7): 2629-45, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23857172

RESUMEN

Echium oil (EO), which is enriched in SDA (18:4 n-3), reduces plasma triglyceride (TG) concentrations in humans and mice. We compared mechanisms by which EO and fish oil (FO) reduce plasma TG concentrations in mildly hypertriglyceridemic male apoB100-only LDLrKO mice. Mice were fed one of three atherogenic diets containing 0.2% cholesterol and palm oil (PO; 20%), EO (10% EO + 10% PO), or FO (10% FO + 10% PO). Livers from PO- and EO-fed mice had similar TG and cholesteryl ester (CE) content, which was significantly higher than in FO-fed mice. Plasma TG secretion was reduced in FO vs. EO-fed mice. Plasma very low density lipoprotein (VLDL) particle size was ordered: PO (63 ± 4 nm) > EO (55 ± 3 nm) > FO (40 ± 2 nm). Post-heparin lipolytic activity was similar among groups, but TG hydrolysis by purified lipoprotein lipase was significantly greater for EO and FO VLDL compared to PO VLDL. Removal of VLDL tracer from plasma was marginally faster in EO vs. PO fed mice. Our results suggest that EO reduces plasma TG primarily through increased intravascular lipolysis of TG and VLDL clearance. Finally, EO may substitute for FO to reduce plasma TG concentrations, but not hepatic steatosis in this mouse model.


Asunto(s)
Apolipoproteína B-100/sangre , Echium/química , Lipólisis/efectos de los fármacos , Aceites de Plantas/farmacología , Receptores de LDL/sangre , Triglicéridos/sangre , Animales , Ésteres del Colesterol/sangre , VLDL-Colesterol/sangre , Aceites de Pescado/farmacología , Expresión Génica , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Aceite de Palma , ARN/genética , ARN/aislamiento & purificación
12.
PLoS One ; 8(5): e63746, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23704936

RESUMEN

Gammadelta (γδ) T lymphocytes respond quickly upon antigen encounter to produce a cytokine response. In this study, we sought to understand how functions of γδ T cells are differentially regulated compared to αß T cells. We found that cholesterol, an integral component of the plasma membrane and a regulator of TCR signaling, is increased in γδ T cells compared to αß T cells, and modulates their function. Higher levels of activation markers, and increased lipid raft content in γδ cells suggest that γδ T cells are more activated. Cholesterol depletion effectively decreased lipid raft formation and activation of γδ T cells, indicating that increased cholesterol content contributes to the hyper-activated phenotype of γδ T cells, possibly through enhanced clustering of TCR signals in lipid rafts. TCR stimulation assays and western blotting revealed that instead of a lower TCR threshold, enhanced TCR signaling through ERK1/2 activation is likely the cause for high cholesterol-induced rapid activation and proliferation in γδ T cells. Our data indicate that cholesterol metabolism is differentially regulated in γδ T cells. The high intracellular cholesterol content leads to enhanced TCR signaling and increases activation and proliferation of γδ T cells.


Asunto(s)
Colesterol/metabolismo , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Animales , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Espacio Intracelular/metabolismo , Activación de Linfocitos/genética , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Fosforilación , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/citología , Linfocitos T/enzimología
13.
J Immunol ; 189(11): 5129-38, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23100511

RESUMEN

ATP-binding cassette transporter G1 (ABCG1) plays a role in the intracellular transport of cholesterol. Invariant NKT (iNKT) cells are a subpopulation of T lymphocytes that recognize glycolipid Ags. In this study, we demonstrate that ABCG1 regulates iNKT cell development and functions in a cell-intrinsic manner. Abcg1(-/-) mice displayed reduced frequencies of iNKT cells in thymus and periphery. Thymic iNKT cells deficient in ABCG1 had reduced membrane lipid raft content, and showed impaired proliferation and defective maturation during the early stages of development. Moreover, we found that Abcg1(-/-) mice possess a higher frequency of Vß7(+) iNKT cells, suggesting alterations in iNKT cell thymic selection. Furthermore, in response to CD3ε/CD28 stimulation, Abcg1(-/-) thymic iNKT cells showed reduced production of IL-4 but increased production of IFN-γ. Our results demonstrate that changes in intracellular cholesterol homeostasis by ABCG1 profoundly impact iNKT cell development and function.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/inmunología , Colesterol/inmunología , Regulación de la Expresión Génica/inmunología , Lipoproteínas/inmunología , Células T Asesinas Naturales/inmunología , Transducción de Señal , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Anticuerpos/farmacología , Transporte Biológico/genética , Transporte Biológico/inmunología , Antígenos CD28/agonistas , Antígenos CD28/inmunología , Complejo CD3/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular , Colesterol/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-4/biosíntesis , Interleucina-4/inmunología , Lipoproteínas/genética , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/citología , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timo
14.
Circ Res ; 111(11): 1398-409, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22955730

RESUMEN

RATIONALE: ATP-binding cassette transporter A1 (ABCA1) plays a critical role in eliminating excess free cholesterol from tissues by effluxing cellular free cholesterol and phospholipids to lipid-poor apolipoprotein AI. Macrophage ABCA1 also dampens proinflammatory myeloid differentiation primary-response protein 88-dependent toll-like receptor signaling by reducing cellular membrane free cholesterol and lipid raft content, indicating a role of ABCA1 in innate immunity. However, whether ABCA1 expression has a role in regulating macrophage function in vivo is unknown. OBJECTIVE: We investigated whether macrophage ABCA1 expression impacts host defense function, including microbial killing and chemotaxis. METHODS AND RESULTS: Myeloid cell-specific ABCA1 knockout (MSKO) vs wild-type mice were infected with Listeria monocytogenes (Lm) for 36 hours or 72 hours before euthanasia. Lm-induced monocytosis was similar for wild-type and MSKO mice; however, MSKO mice were more resistant to Lm infection, with significantly less body weight loss, less Lm burden in liver and spleen, and less hepatic damage 3 days postinfection. In addition, Lm-infected MSKO mouse livers had: (1) greater monocyte chemoattractant protein-1 and macrophage inflammatory protein-2 expression; (2) more monocyte/macrophage infiltration; (3) less neutral lipid accumulation; and (4) diminished expression of lipogenic genes. MSKO macrophages showed enhanced chemotaxis toward chemokines in vitro and increased migration from peritoneum in response to lipopolysaccharide in vivo. Lm infection of wild-type macrophages markedly reduced expression of ABCA1 protein, as well as other cholesterol export proteins (such as ATP-binding cassette transporter G1 and apolipoprotein E). CONCLUSIONS: Myeloid-specific ABCA1 deletion favors host response to and clearance of Lm. Macrophage Lm infection reduces expression of cholesterol export proteins, suggesting that diminished cholesterol efflux enhances innate immune function of macrophages.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Células Mieloides/inmunología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/inmunología , Apolipoproteínas E/metabolismo , Apoptosis/inmunología , Western Blotting , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Quimiocinas/sangre , Quimiocinas/genética , Quimiocinas/inmunología , Citocinas/sangre , Citocinas/genética , Citocinas/inmunología , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Femenino , Interacciones Huésped-Patógeno/inmunología , Listeria monocytogenes/fisiología , Listeriosis/genética , Listeriosis/microbiología , Hígado/inmunología , Hígado/microbiología , Hígado/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Células Mieloides/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Arterioscler Thromb Vasc Biol ; 32(9): 2122-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22814747

RESUMEN

OBJECTIVE: Fish oil, containing omega-3 fatty acids, attenuates atherosclerosis. We hypothesized that omega-3 fatty acid-enriched oils are atheroprotective through alteration of monocyte subsets and their trafficking into atherosclerotic lesions. METHODS AND RESULTS: Low-density lipoprotein receptor knockout and apolipoprotein E(-/-) mice were fed diets containing 10% (calories) palm oil and 0.2% cholesterol, supplemented with an additional 10% palm oil, echium oil (containing 18:4 n-3), or fish oil. Compared with palm oil-fed low-density lipoprotein receptor knockout mice, echium oil and fish oil significantly reduced plasma cholesterol, splenic Ly6C(hi) monocytosis by ≈50%, atherosclerosis by 40% to 70%, monocyte trafficking into the aortic root by ≈50%, and atherosclerotic lesion macrophage content by 30% to 44%. In contrast, atherosclerosis and monocyte trafficking into the artery wall was not altered by omega-3 fatty acids in apolipoprotein E(-/-) mice; however, Ly6C(hi) splenic monocytes positively correlated with aortic root intimal area across all diet groups. In apolipoprotein E(-/-) mice, fish oil reduced the percentage of blood Ly6C(hi) monocytes, despite an average 2-fold higher plasma cholesterol relative to palm oil. CONCLUSIONS: The presence of splenic Ly6C(hi) monocytes parallels the appearance of atherosclerotic disease in both low-density lipoprotein receptor knockout and apolipoprotein E(-/-) mice. Furthermore, omega-3 fatty acids favorably alter monocyte subsets independently from effects on plasma cholesterol and reduce monocyte recruitment into atherosclerotic lesions.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Quimiotaxis/efectos de los fármacos , Suplementos Dietéticos , Ácidos Grasos Omega-3/farmacología , Monocitos/efectos de los fármacos , Aceites de Plantas/farmacología , Bazo/efectos de los fármacos , Animales , Antígenos Ly/sangre , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Biomarcadores/sangre , Colesterol en la Dieta/sangre , Modelos Animales de Enfermedad , Echium , Femenino , Mediadores de Inflamación/sangre , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Aceite de Palma , Receptores de LDL/deficiencia , Receptores de LDL/genética , Bazo/inmunología , Bazo/metabolismo , Factores de Tiempo
16.
J Lipid Res ; 53(4): 686-95, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22279185

RESUMEN

12/15 lipoxygenase (12/15LO) oxidizes polyunsaturated fatty acids (PUFAs) to form bioactive lipid mediators. The role of 12/15LO in atherosclerosis development remains controversial. We evaluated atherosclerosis development and lipid metabolism in 12/15LO-LDL receptor (LDLr) double knockout (DK) vs. LDLr knockout (SK) mice fed a PUFA-enriched diet to enhance production of 12/15LO products. Compared with SK controls, DK mice fed a PUFA-enriched diet had decreased plasma and liver lipid levels, hepatic lipogenic gene expression, VLDL secretion, and aortic atherosclerosis and increased VLDL turnover. Bone marrow transplantation and Kupffer cell ablation studies suggested both circulating leukocytes and Kupffer cells contributed to the lipid phenotype in 12/15LO-deficient mice. Conditioned medium from in vitro incubation of DK vs. SK macrophages reduced triglyceride secretion in McArdle 7777 hepatoma cells. Our results suggest that, in the context of dietary PUFA enrichment, macrophage 12/15LO expression adversely affects plasma and hepatic lipid metabolism, resulting in exacerbated atherosclerosis.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Aterosclerosis/patología , Metabolismo de los Lípidos , Macrófagos/enzimología , Animales , Trasplante de Médula Ósea , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Medios de Cultivo Condicionados , Dieta Aterogénica/efectos adversos , Grasas Insaturadas en la Dieta/administración & dosificación , Ácidos Grasos Insaturados/administración & dosificación , Ácidos Grasos Insaturados/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Hepatocitos/metabolismo , Macrófagos del Hígado/metabolismo , Leucocitos/metabolismo , Lipoproteínas VLDL/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Placa Aterosclerótica/patología
17.
Circulation ; 124(15): 1663-72, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21931081

RESUMEN

BACKGROUND: Adipose tissue (AT) is the body's largest free cholesterol reservoir and abundantly expresses ATP binding cassette transporter A1 (ABCA1), a key cholesterol transporter for high-density lipoprotein (HDL) biogenesis. However, the extent to which AT ABCA1 expression contributes to HDL biogenesis in vivo is unknown. METHODS AND RESULTS: Adipocyte-specific ABCA1 knockout mice (ABCA1(-A/-A)) were generated by crossing ABCA1(floxed) mice with aP2Cre transgenic mice. AT from ABCA1(-A/-A) mice had <10% of wild-type ABCA1 protein expression but normal hepatic and intestinal expression. Deletion of adipocyte ABCA1 resulted in a significant decrease in plasma HDL cholesterol (approximately 15%) and apolipoprotein A-I (approximately 13%) concentrations. AT from ABCA1(-A/-A) mice had a 2-fold increase in free cholesterol content compared with wild-type mice and failed to efflux cholesterol to apolipoprotein A-I. However, cholesterol efflux from AT to plasma HDL was similar for both genotypes of mice. Incubation of wild-type AT explants with apolipoprotein A-I resulted in the formation of multiple discrete-sized nascent HDL particles ranging in diameter from 7.1 to 12 nm; similar incubations with ABCA1(-A/-A) AT explants resulted in nascent HDL <8 nm. Plasma decay and tissue uptake of wild-type (125)I-HDL tracer were similar in both genotypes of recipient mice, suggesting that adipocyte ABCA1 deficiency reduces plasma HDL concentrations solely by reducing nascent HDL particle formation. CONCLUSIONS: We provide in vivo evidence that AT ABCA1-dependent cholesterol efflux and nascent HDL particle formation contribute to systemic HDL biogenesis and that AT ABCA1 expression plays an important role in adipocyte cholesterol homeostasis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Tejido Adiposo/metabolismo , Lipoproteínas HDL/biosíntesis , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Apolipoproteína A-I/sangre , Colesterol/metabolismo , Homeostasis , Lipoproteínas HDL/sangre , Lipoproteínas HDL/química , Ratones , Ratones Noqueados , Ratones Transgénicos , Tamaño de la Partícula
18.
J Lipid Res ; 51(4): 729-42, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20215580

RESUMEN

In Tangier disease, absence of ATP binding cassette transporter A1 (ABCA1) results in reduced plasma HDL and elevated triglyceride (TG) levels. We hypothesized that hepatocyte ABCA1 regulates VLDL TG secretion through nascent HDL production. Silencing of ABCA1 expression in oleate-stimulated rat hepatoma cells resulted in: 1) decreased large nascent HDL (>10 nm diameter) and increased small nascent HDL (<10 nm) formation, 2) increased large buoyant VLDL1 particle secretion, and 3) decreased phosphatidylinositol-3 (PI3) kinase activation. Nascent HDL-containing conditioned medium from rat hepatoma cells or HEK293 cells transfected with ABCA1 was effective in increasing PI3 kinase activation and reducing VLDL TG secretion in ABCA1-silenced hepatoma cells. Addition of isolated large nascent HDL particles to ABCA1-silenced hepatoma cells inhibited VLDL TG secretion to a greater extent than small nascent HDL. Similarly, addition of recombinant HDL, but not human plasma HDL, was effective in attenuating TG secretion and increasing PI3 kinase activation in ABCA1-silenced cells. Collectively, these data suggest that large nascent HDL particles, assembled by hepatic ABCA1, generate a PI3 kinase-mediated autocrine signal that attenuates VLDL maturation and TG secretion. This pathway may explain the elevated plasma TG concentration that occurs in most Tangier subjects and may also account, in part, for the inverse relationship between plasma HDL and TG concentrations in individuals with compromised ABCA1 function.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Lipoproteínas de Alta Densidad Pre-beta/fisiología , Lipoproteínas VLDL/metabolismo , Hígado/metabolismo , Triglicéridos/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apolipoproteína A-I/análisis , Apolipoproteína A-I/metabolismo , Apolipoproteínas B/análisis , Electroforesis de las Proteínas Sanguíneas , Línea Celular , Línea Celular Tumoral , Centrifugación por Gradiente de Densidad , Activación Enzimática , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/aislamiento & purificación , Lipoproteínas HDL/fisiología , Lipoproteínas VLDL/química , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Enfermedad de Tangier/fisiopatología
19.
Arterioscler Thromb Vasc Biol ; 30(4): 809-17, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20110576

RESUMEN

OBJECTIVE: Activated endothelium and increased monocyte-endothelial interactions in the vessel wall are key early events in atherogenesis. ATP binding cassette (ABC) transporters play important roles in regulating sterol homeostasis in many cell types. Endothelial cells (EC) have a high capacity to efflux sterols and express the ABC transporter, ABCG1. Here, we define the role of ABCG1 in the regulation of lipid homeostasis and inflammation in aortic EC. METHODS AND RESULTS: Using EC isolated from ABCG1-deficient mice (ABCG1 KO), we observed reduced cholesterol efflux to high-density lipoprotein compared to C57BL/6 (B6) EC. However, total cholesteryl ester levels were not changed in ABCG1 KO EC. Secretions of KC, MCP-1, and IL-6 by ABCG1 KO EC were significantly increased, and surface expressions of intercellular adhesion molecule-1 and E-selectin were increased several-fold on ABCG1 KO EC. Concomitant with these findings, we observed a 4-fold increase in monocyte adhesion to the intact aortic endothelium of ABCG1 KO mice ex vivo and to isolated aortic EC from these mice in vitro. In a gain-of-function study in vitro, restoration of ABCG1 expression in ABCG1 KO EC reduced monocyte-endothelial interactions. Utilizing pharmacological inhibitors for STAT3 and the IL-6 receptor, we found that blockade of STAT3 and IL-6 receptor signaling in ABCG1 KO EC completely abrogated monocyte adhesion to ABCG1 KO endothelium. CONCLUSIONS: ABCG1 deficiency in aortic endothelial cells activates endothelial IL-6-IL-6 receptor-STAT3 signaling, thereby increasing monocyte-endothelial interactions and vascular inflammation.


Asunto(s)
Adhesión Celular , Células Endoteliales/metabolismo , Inflamación/metabolismo , Lipoproteínas/deficiencia , Monocitos/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Quimiotaxis , Colesterol/metabolismo , HDL-Colesterol/metabolismo , Selectina E/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Lipoproteínas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/inmunología , Receptores de Interleucina-6/antagonistas & inhibidores , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
20.
J Immunol ; 184(1): 173-83, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19949102

RESUMEN

Cholesterol is a key component of cell membranes and is essential for cell growth and proliferation. How the accumulation of cellular cholesterol affects lymphocyte development and function is not well understood. We demonstrate that ATP-binding cassette transporter G1 (ABCG1) regulates cholesterol homeostasis in thymocytes and peripheral CD4 T cells. Our work is the first to describe a cell type in Abcg1-deficient mice with such a robust change in cholesterol content and the expression of cholesterol metabolism genes. Abcg1-deficient mice display increased thymocyte cellularity and enhanced proliferation of thymocytes and peripheral T lymphocytes in vivo. The absence of ABCG1 in CD4 T cells results in hyperproliferation in vitro, but only when cells are stimulated through the TCR. We hypothesize that cholesterol accumulation in Abcg1(-/-) T cells alters the plasma membrane structure, resulting in enhanced TCR signaling for proliferation. Supporting this idea, we demonstrate that B6 T cells pretreated with soluble cholesterol have a significant increase in proliferation. Cholesterol accumulation in Abcg1(-/-) CD4 T cells results in enhanced basal phosphorylation levels of ZAP70 and ERK1/2. Furthermore, inhibition of ERK phosphorylation in TCR-stimulated Abcg1(-/-) T cells rescues the hyperproliferative phenotype. We describe a novel mechanism by which cholesterol can alter signaling from the plasma membrane to affect downstream signaling pathways and proliferation. These results implicate ABCG1 as an important negative regulator of lymphocyte proliferation through the maintenance of cellular cholesterol homeostasis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proliferación Celular , Colesterol/metabolismo , Lipoproteínas/metabolismo , Transducción de Señal/fisiología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Animales , Western Blotting , Recuento de Células , Membrana Celular/metabolismo , Citometría de Flujo , Homeostasis/fisiología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...