Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 4(16)2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31434800

RESUMEN

The common patatin-like phospholipase domain-containing protein 3 (PNPLA3) variant I148M predisposes to nonalcoholic liver disease but not its metabolic sequelae. We compared the handling of labeled polyunsaturated fatty acids (PUFAs) and saturated fatty acids (SFA) in vivo in humans and in cells harboring different PNPLA3 genotypes. In 148M homozygous individuals, triglycerides (TGs) in very low-density lipoproteins (VLDL) were depleted of PUFAs both under fasting and postprandial conditions compared with 148I homozygotes, and the PUFA/SFA ratio in VLDL-TGs was lower relative to the chylomicron precursor pool. In human PNPLA3-148M and PNPLA3-KO cells, PUFA but not SFA incorporation into TGs was increased at the expense of phosphatidylcholines, and under lipolytic conditions, PUFA-containing diacylglycerols (DAGs) accumulated compared with PNPLA3-148I cells. Polyunsaturated TGs were increased, while phosphatidylcholines (PCs) were decreased in the human liver in 148M homozygous individuals as compared with 148I homozygotes. We conclude that human PNPLA3-I148M is a loss-of-function allele that remodels liver TGs in a polyunsaturated direction by impairing hydrolysis/transacylation of PUFAs from DAGs to feed phosphatidylcholine synthesis.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Lipasa/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Adolescente , Adulto , Línea Celular , Línea Celular Tumoral , VLDL-Colesterol/metabolismo , Ácidos Grasos/metabolismo , Femenino , Heterocigoto , Humanos , Lipasa/genética , Mutación con Pérdida de Función , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Triglicéridos/metabolismo , Adulto Joven
2.
Mol Metab ; 28: 135-143, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31327756

RESUMEN

OBJECTIVE: Heat Shock Proteins (HSPs) maintain cellular homeostasis under stress. HSP70 represents a major stress-inducible family member and has been identified as a druggable target in inherited cholesterol-sphingolipid storage diseases. We investigated if HSP70 modulates cholesterol accumulation in more common conditions related to atherogenesis. METHODS: We studied the effects of recombinant HSP70 in cholesterol-laden primary macrophages from human blood donors and pharmacological HSP70 upregulation in high-cholesterol diet fed zebrafish. RESULTS: Recombinant HSP70 facilitated cholesterol removal from primary human macrophage foam cells. RNA sequencing revealed that HSP70 induced a robust transcriptional re-programming, including upregulation of key targets of liver X receptors (LXR), master regulators of whole-body cholesterol removal. Mechanistically, HSP70 interacted with the macrophage LXRalpha promoter, increased LXRalpha and its target mRNAs, and led to elevated levels of key proteins facilitating cholesterol efflux, including ATP-binding cassette transporters A1 and G1. Pharmacological augmentation of endogenous HSP70 in high-cholesterol diet fed zebrafish activated LXR and its target mRNAs and reduced cholesterol storage at the whole organism level. CONCLUSION: These data demonstrate that HSP70 exerts a cholesterol lowering effect in primary human cells and animals and uncover a nuclear action of HSP70 in mediating cross-talk between HSP and LXR transcriptional regulation.


Asunto(s)
Colesterol/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Receptores X del Hígado/metabolismo , Animales , Colesterol/administración & dosificación , Dieta , Humanos , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Proteínas Recombinantes/metabolismo , Pez Cebra
3.
Dev Cell ; 50(4): 478-493.e9, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31178403

RESUMEN

Seipin is an oligomeric integral endoplasmic reticulum (ER) protein involved in lipid droplet (LD) biogenesis. To study the role of seipin in LD formation, we relocalized it to the nuclear envelope and found that LDs formed at these new seipin-defined sites. The sites were characterized by uniform seipin-mediated ER-LD necks. At low seipin content, LDs only grew at seipin sites, and tiny, growth-incompetent LDs appeared in a Rab18-dependent manner. When seipin was removed from ER-LD contacts within 1 h, no lipid metabolic defects were observed, but LDs became heterogeneous in size. Studies in seipin-ablated cells and model membranes revealed that this heterogeneity arises via a biophysical ripening process, with triglycerides partitioning from smaller to larger LDs through droplet-bilayer contacts. These results suggest that seipin supports the formation of structurally uniform ER-LD contacts and facilitates the delivery of triglycerides from ER to LDs. This counteracts ripening-induced shrinkage of small LDs.


Asunto(s)
Retículo Endoplásmico/genética , Subunidades gamma de la Proteína de Unión al GTP/genética , Gotas Lipídicas/metabolismo , Fibroblastos/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Cultivo Primario de Células , Triglicéridos/genética , Triglicéridos/metabolismo
4.
EMBO J ; 36(12): 1736-1754, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28495678

RESUMEN

Sphingolipids are membrane lipids globally required for eukaryotic life. The sphingolipid content varies among endomembranes with pre- and post-Golgi compartments being poor and rich in sphingolipids, respectively. Due to this different sphingolipid content, pre- and post-Golgi membranes serve different cellular functions. The basis for maintaining distinct subcellular sphingolipid levels in the presence of membrane trafficking and metabolic fluxes is only partially understood. Here, we describe a homeostatic regulatory circuit that controls sphingolipid levels at the trans-Golgi network (TGN). Specifically, we show that sphingomyelin production at the TGN triggers a signalling pathway leading to PtdIns(4)P dephosphorylation. Since PtdIns(4)P is required for cholesterol and sphingolipid transport to the trans-Golgi network, PtdIns(4)P consumption interrupts this transport in response to excessive sphingomyelin production. Based on this evidence, we envisage a model where this homeostatic circuit maintains a constant lipid composition in the trans-Golgi network and post-Golgi compartments, thus counteracting fluctuations in the sphingolipid biosynthetic flow.


Asunto(s)
Fosfatidilinositoles/metabolismo , Esfingolípidos/metabolismo , Red trans-Golgi/metabolismo , Células HeLa , Homeostasis , Humanos , Modelos Biológicos
5.
Eur J Hum Genet ; 25(3): 315-323, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28051070

RESUMEN

In two brothers born to consanguineous parents, we identified an unusual neurological disease that manifested with ataxia, psychomotor retardation, cerebellar and cerebral atrophy, and leukodystrophy. Via linkage analysis and exome sequencing, we identified homozygous c.2801C>T (p.(Ser934Leu)) in POLR1A (encoding RPA194, largest subunit of RNA polymerase I) and c.511C>T (p.(Arg171Trp)) in OSBPL11 (encoding oxysterol-binding protein-like protein 11). Although in silico analysis, histopathologic evidence and functional verification indicated that both variants were deleterious, segregation with the patient phenotype established that the POLR1A defect underlies the disease, as a clinically unaffected sister also was homozygous for the OSBPL11 variant. Decreased nucleolar RPA194 was observed in the skin fibroblasts of only the affected brothers, whereas intracellular cholesterol accumulation was observed in the skin biopsies of the patients and the sister homozygous for the OSBPL11 variant. Our findings provide the first report showing a complex leukodystrophy associated with POLR1A. Variants in three other RNA polymerase subunits, POLR1C, POLR3A and POLR3B, are known to cause recessive leukodystrophy similar to the disease afflicting the present family but with a later onset. Of those, POLR1C is also implicated in a mandibulofacial dysostosis syndrome without leukodystrophy as POLR1A is. This syndrome is absent in the family we present.


Asunto(s)
Ataxia/genética , ARN Polimerasas Dirigidas por ADN/genética , Discapacidades del Desarrollo/genética , Leucoencefalopatías/genética , Mutación Missense , Adulto , Ataxia/diagnóstico , Células Cultivadas , Niño , Colesterol/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Discapacidades del Desarrollo/diagnóstico , Femenino , Fibroblastos/metabolismo , Homocigoto , Humanos , Leucoencefalopatías/diagnóstico , Masculino , Linaje , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Hermanos , Síndrome
6.
EMBO J ; 35(24): 2699-2716, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27879284

RESUMEN

Seipin is an endoplasmic reticulum (ER) membrane protein implicated in lipid droplet (LD) biogenesis and mutated in severe congenital lipodystrophy (BSCL2). Here, we show that seipin is stably associated with nascent ER-LD contacts in human cells, typically via one mobile focal point per LD Seipin appears critical for such contacts since ER-LD contacts were completely missing or morphologically aberrant in seipin knockout and BSCL2 patient cells. In parallel, LD mobility was increased and protein delivery from the ER to LDs to promote LD growth was decreased. Moreover, while growing LDs normally acquire lipid and protein constituents from the ER, this process was compromised in seipin-deficient cells. In the absence of seipin, the initial synthesis of neutral lipids from exogenous fatty acid was normal, but fatty acid incorporation into neutral lipids in cells with pre-existing LDs was impaired. Together, our data suggest that seipin helps to connect newly formed LDs to the ER and that by stabilizing ER-LD contacts seipin facilitates the incorporation of protein and lipid cargo into growing LDs in human cells.


Asunto(s)
Retículo Endoplásmico/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Gotas Lipídicas/metabolismo , Células Cultivadas , Subunidades gamma de la Proteína de Unión al GTP/genética , Técnicas de Inactivación de Genes , Humanos , Modelos Biológicos
7.
Traffic ; 17(9): 1054-7, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27187581

RESUMEN

Dipyrromethene difluoride-cholesterol (TopFluor-Cholesterol, TF-Chol) is a widely used cholesterol analogue due to its excellent fluorescence properties and considerable similarity with natural cholesterol in terms of membrane partitioning. However, the suitability of TF-Chol for detecting lysosomal cholesterol deposition has recently been questioned. Here, we highlight the fact that the method of lipid delivery and the analysis of time-point both affect the membrane distribution and labeling pattern of TF-Chol, similarly as with radiolabeled cholesterol. Lysosomal sterol accumulation characteristic to a lysosomal storage disease is most readily detected when the probe is introduced via the physiological route, i.e. as a sterol fatty acid ester in low-density lipoprotein particles. When administered to cells from solvent, lysosomal sterol sequestration becomes evident after an overnight equilibration between membranes.


Asunto(s)
Compuestos de Boro/química , Membrana Celular/metabolismo , Colesterol/metabolismo , Fibroblastos/metabolismo , Membranas Intracelulares/metabolismo , Lisosomas/metabolismo , Transporte Biológico , Proteínas Portadoras/genética , Línea Celular , Colesterol/química , Esterificación , Humanos , Péptidos y Proteínas de Señalización Intracelular , Enfermedades por Almacenamiento Lisosomal/diagnóstico , Enfermedades por Almacenamiento Lisosomal/metabolismo , Glicoproteínas de Membrana/genética , Microscopía Fluorescente , Proteína Niemann-Pick C1
8.
Biol Open ; 5(5): 584-95, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-27044324

RESUMEN

Mulibrey nanism (MUL) is a rare autosomal recessive multi-organ disorder characterized by severe prenatal-onset growth failure, infertility, cardiopathy, risk for tumors, fatty liver, and type 2 diabetes. MUL is caused by loss-of-function mutations in TRIM37, which encodes an E3 ubiquitin ligase belonging to the tripartite motif (TRIM) protein family and having both peroxisomal and nuclear localization. We describe a congenic Trim37 knock-out mouse (Trim37(-/-)) model for MUL. Trim37(-/-) mice were viable and had normal weight development until approximately 12 months of age, after which they started to manifest increasing problems in wellbeing and weight loss. Assessment of skeletal parameters with computer tomography revealed significantly smaller skull size, but no difference in the lengths of long bones in Trim37(-/-) mice as compared with wild-type. Both male and female Trim37(-/-) mice were infertile, the gonads showing germ cell aplasia, hilus and Leydig cell hyperplasia and accumulation of lipids in and around Leydig cells. Male Trim37(-/-) mice had elevated levels of follicle-stimulating and luteinizing hormones, but maintained normal levels of testosterone. Six-month-old Trim37(-/-) mice had elevated fasting blood glucose and low fasting serum insulin levels. At 1.5 years Trim37(-/-) mice showed non-compaction cardiomyopathy, hepatomegaly, fatty liver and various tumors. The amount and morphology of liver peroxisomes seemed normal in Trim37(-/-) mice. The most consistently seen phenotypes in Trim37(-/-) mice were infertility and the associated hormonal findings, whereas there was more variability in the other phenotypes observed. Trim37(-/-) mice recapitulate several features of the human MUL disease and thus provide a good model to study disease pathogenesis related to TRIM37 deficiency, including infertility, non-alcoholic fatty liver disease, cardiomyopathy and tumorigenesis.

9.
Am J Pathol ; 185(4): 987-1000, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25681734

RESUMEN

The STARD3 gene belongs to the minimal amplicon in HER2-positive breast cancers and encodes a cholesterol-binding membrane protein. To study how elevated StAR-related lipid transfer protein 3 (StARD3) expression affects breast cancer cells, we generated MCF-7 cells stably overexpressing StARD3-green fluorescent protein. We found that StARD3-overexpressing cells exhibited nonadherent morphological features, had increased Src levels, and had altered cholesterol balance, as evidenced by elevated mRNA levels of the cholesterol biosynthesis rate-limiting enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase, and increased plasma membrane cholesterol content. On removal of serum and insulin from the culture medium, the morphological characteristics of the StARD3-overexpressing cells changed, the cells became adherent, and they developed enlarged focal adhesions. Under these conditions, the StARD3-overexpressing cells maintained elevated Src and plasma membrane cholesterol content and showed increased phosphorylation of focal adhesion kinase. In two Finnish nationwide patient cohorts, approximately 10% (212/2220) breast cancers exhibited high StARD3 protein levels, which was strongly associated with HER2 amplification; several factors related to poor disease outcome and poor breast cancer-specific survival. In addition, high StARD3 levels in breast cancers were associated with elevated 3-hydroxy-3-methylglutaryl-coenzyme A reductase mRNA levels and anti-Src-Tyr416 immunoreactivity. These results provide evidence that StARD3 overexpression results in increased cholesterol biosynthesis and Src kinase activity in breast cancer cells and suggest that elevated StARD3 expression may contribute to breast cancer aggressiveness by increasing membrane cholesterol and enhancing oncogenic signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Portadoras/metabolismo , Colesterol/metabolismo , Progresión de la Enfermedad , Proteínas de la Membrana/metabolismo , Receptor ErbB-2/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Forma de la Célula/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Amplificación de Genes/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Insulina/farmacología , Células MCF-7 , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Suero/metabolismo , Familia-src Quinasas/metabolismo
10.
J Lipid Res ; 55(4): 739-46, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24511104

RESUMEN

The I148M substitution in patatin-like phospholipase domain containing 3 (PNPLA3(I148M)) determines a genetic form of nonalcoholic fatty liver disease. To elucidate the mode of PNPLA3 action in human hepatocytes, we studied effects of WT PNPLA3 (PNPLA3(WT)) and PNPLA3(I148M) on HuH7 cell lipidome after [(13)C]glycerol labeling, cellular turnover of oleic acid labeled with 17 deuterium atoms ([D17]oleic acid) in triacylglycerols (TAGs), and subcellular distribution of the protein variants. PNPLA3(I148M) induced a net accumulation of unlabeled TAGs, but not newly synthesized total [(13)C]TAGs. Principal component analysis (PCA) revealed that both PNPLA3(WT) and PNPLA3(I148M) induced a relative enrichment of TAGs with saturated FAs or MUFAs, with concurrent enrichment of polyunsaturated phosphatidylcholines. PNPLA3(WT) associated in PCA with newly synthesized [(13)C]TAGs, particularly 52:1 and 50:1, while PNPLA3(I148M) associated with similar preexisting TAGs. PNPLA3(WT) overexpression resulted in increased [D17]oleic acid labeling of TAGs during 24 h, and after longer incubations their turnover was accelerated, effects not detected with PNPLA3(I148M). PNPLA3(I148M) localized more extensively to lipid droplets (LDs) than PNPLA3(WT), suggesting that the substitution alters distribution of PNPLA3 between LDs and endoplasmic reticulum/cytosol. This study reveals a function of PNPLA3 in FA-selective TAG remodeling, resulting in increased TAG saturation. A defect in TAG remodeling activity likely contributes to the TAG accumulation observed in cells expressing PNPLA3(I148M).


Asunto(s)
Hepatocitos/enzimología , Lipasa/fisiología , Proteínas de la Membrana/fisiología , Triglicéridos/metabolismo , Línea Celular Tumoral , Ácidos Grasos/metabolismo , Humanos , Gotas Lipídicas/enzimología , Metabolismo de los Lípidos , Mutación Missense , Fosfolípidos/metabolismo , Transporte de Proteínas
11.
Hum Mol Genet ; 22(6): 1157-66, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23250914

RESUMEN

Mutations affecting the N-glycosylation site in Berardinelli-Seip lipodystrophy (BSCL)-associated gene BSCL2/seipin lead to a dominantly inherited spastic paraplegia termed seipinopathy. While the loss of function of seipin leads to severe congenital lipodystrophy, the effects of seipin N-glycosylation mutations on lipid balance in the nervous system are unknown. In this study, we show that expression of seipin N-glycosylation mutant N88S led to decreased triglyceride (TG) content in astrocytoma and motor neuron cell lines. This was corrected by supplementation with exogenous oleic acid. Upon oleic acid loading, seipin N88S protein was relocated from the endoplasmic reticulum (ER) to the surface of lipid droplets and this was paralleled by alleviation of ER stress induced by the mutant protein. This effect was not limited to seipin N88S, as oleic acid loading also reduced tunicamycin-induced ER stress in motor neuron cells. Furthermore, both seipin N88S and tunicamycin-induced ER stress were decreased by inhibiting lipolysis, suggesting that lipid droplets protected neuronal cells from ER stress. In developing zebrafish larvae, seipin N88S expression led to TG imbalance and reduced spontaneous free swimming. Importantly, supplementation with exogenous oleic acid reduced ER stress in the zebrafish head and increased fish motility. We propose that the decreased TG content contributes to the pathology induced by seipin N88S, and that rescuing TG levels may provide a novel therapeutic strategy in seipinopathy.


Asunto(s)
Estrés del Retículo Endoplásmico , Lipodistrofia/metabolismo , Triglicéridos/metabolismo , Animales , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Humanos , Lipodistrofia/genética , Pez Cebra
12.
Circ Res ; 110(3): 450-5, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22223354

RESUMEN

RATIONALE: The actin cytoskeleton has been implicated in the processing of atherogenic lipoproteins in macrophages. However, the functional role of actin and the regulatory proteins involved are unknown. OBJECTIVE: Coronin-1A (Coro1A) was identified as a differentially expressed transcript in wild-type versus Niemann-Pick type C1 deficient macrophages exposed to acetylated low-density lipoproteins (AcLDL). We investigated whether Coro1A plays a role in the uptake or processing of modified lipoproteins in macrophages and if this is related to its actin regulatory functions. METHODS AND RESULTS: In wild-type primary macrophages, filamentous actin transiently decorated AcLDL containing endosomes that also recruited Coro1A. This dynamic association of F-actin with endosomes was disturbed in Coro1A deficient macrophages. In Coro1A knockout macrophages the uptake of AcLDL was increased, rate of AcLDL delivery to lysosomes enhanced, and lipoprotein-derived cholesteryl ester hydrolysis accelerated. Overexpression of wild-type Coro1A normalized AcLDL uptake in Coro1A knockout macrophages while a Coro1A actin binding mutant did not. Furthermore, the effects of macrophage Coro1A silencing on endosomal actin association and AcLDL delivery to lysosomes resembled those of cofilin silencing. CONCLUSIONS: Coro1A controls actin association with endocytic organelles, thereby negatively regulating endo-lysosomal delivery, degradation of modified lipoproteins and cholesterol deposition in macrophages.


Asunto(s)
Actinas/metabolismo , Endosomas/metabolismo , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteolisis , Animales , Transporte Biológico , Células Cultivadas , Colesterol/metabolismo , Lisosomas/metabolismo , Macrófagos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Modelos Animales , Orgánulos/metabolismo
13.
Neuron ; 72(2): 257-68, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-21944779

RESUMEN

The chromosome 9p21 amyotrophic lateral sclerosis-frontotemporal dementia (ALS-FTD) locus contains one of the last major unidentified autosomal-dominant genes underlying these common neurodegenerative diseases. We have previously shown that a founder haplotype, covering the MOBKL2b, IFNK, and C9ORF72 genes, is present in the majority of cases linked to this region. Here we show that there is a large hexanucleotide (GGGGCC) repeat expansion in the first intron of C9ORF72 on the affected haplotype. This repeat expansion segregates perfectly with disease in the Finnish population, underlying 46.0% of familial ALS and 21.1% of sporadic ALS in that population. Taken together with the D90A SOD1 mutation, 87% of familial ALS in Finland is now explained by a simple monogenic cause. The repeat expansion is also present in one-third of familial ALS cases of outbred European descent, making it the most common genetic cause of these fatal neurodegenerative diseases identified to date.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Cromosomas Humanos Par 9 , Demencia Frontotemporal/genética , Repeticiones de Microsatélite , Alelos , Femenino , Finlandia , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Humanos , Masculino , Linaje , Polimorfismo de Nucleótido Simple
14.
J Cell Mol Med ; 15(2): 280-95, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19929948

RESUMEN

The mechanisms of endosomal and lysosomal cholesterol traffic are still poorly understood. We showed previously that unesterified cholesterol accumulates in the late endosomes and lysosomes of fibroblasts deficient in both lysosome associated membrane protein-2 (LAMP-2) and LAMP-1, two abundant membrane proteins of late endosomes and lysosomes. In this study we show that in cells deficient in both LAMP-1 and LAMP-2 (LAMP(-/-)), low-density lipoprotein (LDL) receptor levels and LDL uptake are increased as compared to wild-type cells. However, there is a defect in esterification of both endogenous and LDL cholesterol. These results suggest that LAMP(-/-) cells have a defect in cholesterol transport to the site of esterification in the endoplasmic reticulum, likely due to defective export of cholesterol out of late endosomes or lysosomes. We also show that cholesterol accumulates in LAMP-2 deficient liver and that overexpression of LAMP-2 retards the lysosomal cholesterol accumulation induced by U18666A. These results point to a critical role for LAMP-2 in endosomal/lysosomal cholesterol export. Moreover, the late endosomal/lysosomal cholesterol accumulation in LAMP(-/-) cells was diminished by overexpression of any of the three isoforms of LAMP-2, but not by LAMP-1. The LAMP-2 luminal domain, the membrane-proximal half in particular, was necessary and sufficient for the rescue effect. Taken together, our results suggest that LAMP-2, its luminal domain in particular, plays a critical role in endosomal cholesterol transport and that this is distinct from the chaperone-mediated autophagy function of LAMP-2.


Asunto(s)
Colesterol/metabolismo , Endosomas/metabolismo , Lipoproteínas LDL/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Androstenos/farmacología , Animales , Transporte Biológico , Línea Celular , Retículo Endoplásmico/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/química , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/deficiencia , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Estructura Terciaria de Proteína , Receptores de LDL/metabolismo
15.
Biochem J ; 429(2): 235-42, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20578994

RESUMEN

Zebrafish are an increasingly popular vertebrate model organism in which to study biological phenomena. It has been widely used, especially in developmental biology and neurobiology, and many aspects of its development and physiology are similar to those of mammals. The popularity of zebrafish relies on its relatively low cost, rapid development and ease of genetic manipulation. Moreover, the optical transparency of the developing fish together with novel imaging techniques enable the direct visualization of complex phenomena at the level of the entire organism. This potential is now also being increasingly appreciated by the lipid research community. In the present review we summarize basic information on the lipid composition and distribution in zebrafish tissues, including lipoprotein metabolism, intestinal lipid absorption, the yolk lipids and their mobilization, as well as lipids in the nervous system. We also discuss studies in which zebrafish have been employed for the visualization of whole-body lipid distribution and trafficking. Finally, recent advances in using zebrafish as a model for lipid-related diseases, including atherosclerosis, obesity, diabetes and hepatic steatosis are highlighted. As the insights into zebrafish lipid metabolism increase, it is likely that zebrafish as a model organism will become an increasingly powerful tool in lipid research.


Asunto(s)
Metabolismo de los Lípidos , Pez Cebra/metabolismo , Animales , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Colesterol/metabolismo , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/metabolismo , Lipoproteínas/metabolismo , Modelos Animales , Sistema Nervioso/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Saco Vitelino/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
Traffic ; 9(11): 1839-49, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18647169

RESUMEN

Analysis of sterol distribution and transport in living cells has been hampered by the lack of bright, photostable fluorescent sterol derivatives that closely resemble cholesterol. In this study, we employed atomistic simulations and experiments to characterize a cholesterol compound with fluorescent boron dipyrromethene difluoride linked to sterol carbon-24 (BODIPY-cholesterol). This probe packed in the membrane and behaved similarly to cholesterol both in normal and in cholesterol-storage disease cells and with trace amounts allowed the visualization of sterol movement in living systems. Upon injection into the yolk sac, BODIPY-cholesterol did not disturb zebrafish development and was targeted to sterol-enriched brain regions in live fish. We conclude that this new probe closely mimics the membrane partitioning and trafficking of cholesterol and, because of its excellent fluorescent properties, enables the direct monitoring of sterol movement by time-lapse imaging using trace amounts of the probe. This is, to our knowledge, the first cholesterol probe that fulfills these prerequisites.


Asunto(s)
Compuestos de Boro/química , Colesterol/química , Esteroles/metabolismo , Animales , Transporte Biológico , Células CHO , Cricetinae , Cricetulus , Colorantes Fluorescentes , Inmunohistoquímica , Pez Cebra
17.
J Mol Med (Berl) ; 85(6): 589-601, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17221218

RESUMEN

The HCR gene, officially called Coiled-Coil alpha-Helical Rod protein 1 (CCHCR1), located within the major psoriasis susceptibility locus PSORS1, is a plausible candidate gene for the risk effect. Recently, CCHCR1 was shown to promote steroidogenesis by interacting with the steroidogenic acute regulator protein (StAR). Here, we examined the role of CCHCR1 in psoriasis and cutaneous steroid metabolism. We found that CCHCR1 and StAR are expressed in basal keratinocytes in overlapping areas of the human skin, and CCHCR1 stimulated pregnenolone production in steroidogenesis assay. Overexpression of either the CCHCR1*WWCC risk allele or the non-risk allele enhanced steroid synthesis in vitro. Furthermore, the cytochrome P450scc enzyme was expressed in human keratinocytes and was induced by forskolin, a known activator of steroidogenesis, and forskolin also upregulated CCHCR1. CCHCR1 has an altered expression pattern in lesional psoriatic skin compared to normal healthy skin, suggesting its dysregulation in psoriasis. We found that the expression of CCHCR1 is downregulated twofold at the mRNA level in cultured non-lesional psoriatic keratinocytes when compared to non-psoriatic healthy cells. Our results also suggest a connection between CCHCR1 and vitamin D metabolism in keratinocytes. The expression of the vitamin D receptor (VDR) gene was lower in non-lesional psoriatic keratinocytes than in healthy cells. Furthermore, Vdr expression was downregulated in the keratinocytes of mice overexpressing the CCHCR1*WWCC risk allele when compared to keratinocytes from mice with the non-risk allele of CCHCR1. Finally, we demonstrate that other agents relevant for psoriasis and/or the regulation of steroidogenesis influence CCHCR1 expression in keratinocytes, including insulin, EGF, cholesterol, estrogen, and cyclosporin A. Taken the role of steroid hormones, including vitamin D and estrogen, in cell proliferation, epidermal barrier homeostasis, differentiation, and immune response, our results suggest a role for CCHCR1 in the pathogenesis of psoriasis via the regulation of skin steroid metabolism.


Asunto(s)
Regulación hacia Abajo/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Queratinocitos/metabolismo , Queratinocitos/patología , Psoriasis/genética , Piel/metabolismo , Esteroides/biosíntesis , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Especificidad de Órganos , Fosfoproteínas/genética , Transporte de Proteínas , Receptores de Calcitriol/genética , Transfección
18.
Mol Biol Cell ; 18(1): 47-56, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17050734

RESUMEN

The mechanisms by which low-density lipoprotein (LDL)-cholesterol exits the endocytic circuits are not well understood. The process is defective in Niemann-Pick type C (NPC) disease in which cholesterol and sphingolipids accumulate in late endosomal compartments. This is accompanied by defective cholesterol esterification in the endoplasmic reticulum and impaired ATP-binding cassette transporter A1 (ABCA1)-dependent cholesterol efflux. We show here that overexpression of the recycling/exocytic Rab GTPase Rab8 rescued the late endosomal cholesterol deposition and sphingolipid mistrafficking in NPC fibroblasts. Rab8 redistributed cholesterol from late endosomes to the cell periphery and stimulated cholesterol efflux to the ABCA1-ligand apolipoprotein A-I (apoA-I) without increasing cholesterol esterification. Depletion of Rab8 from wild-type fibroblasts resulted in cholesterol deposition within late endosomal compartments. This cholesterol accumulation was accompanied by impaired clearance of LDL-cholesterol from endocytic circuits to apoA-I and could not be bypassed by liver X receptor activation. Our findings establish Rab8 as a key component of the regulatory machinery that leads to ABCA1-dependent removal of cholesterol from endocytic circuits.


Asunto(s)
Colesterol/aislamiento & purificación , Colesterol/metabolismo , Endosomas/metabolismo , Fibroblastos/citología , Fibroblastos/patología , Enfermedades de Niemann-Pick/patología , Proteínas de Unión al GTP rab/metabolismo , Animales , Apolipoproteína A-I/metabolismo , Transporte Biológico , Línea Celular , Proteínas de Unión al ADN/agonistas , Expresión Génica , Humanos , Receptores X del Hígado , Proteínas de la Membrana/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/agonistas , Proteínas Recombinantes de Fusión/metabolismo , Esfingolípidos/metabolismo , Regulación hacia Arriba , Proteínas de Unión al GTP rab/deficiencia , Proteínas de Unión al GTP rab/aislamiento & purificación , Proteínas de Unión al GTP rab4/metabolismo , Proteínas de Unión a GTP rab7
19.
Mol Biol Cell ; 16(8): 3873-86, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15930133

RESUMEN

MLN64 is a late endosomal cholesterol-binding membrane protein of an unknown function. Here, we show that MLN64 depletion results in the dispersion of late endocytic organelles to the cell periphery similarly as upon pharmacological actin disruption. The dispersed organelles in MLN64 knockdown cells exhibited decreased association with actin and the Arp2/3 complex subunit p34-Arc. MLN64 depletion was accompanied by impaired fusion of late endocytic organelles and delayed cargo degradation. MLN64 overexpression increased the number of actin and p34-Arc-positive patches on late endosomes, enhanced the fusion of late endocytic organelles in an actin-dependent manner, and stimulated the deposition of sterol in late endosomes harboring the protein. Overexpression of wild-type MLN64 was capable of rescuing the endosome dispersion in MLN64-depleted cells, whereas mutants of MLN64 defective in cholesterol binding were not, suggesting a functional connection between MLN64-mediated sterol transfer and actin-dependent late endosome dynamics. We propose that local sterol enrichment by MLN64 in the late endosomal membranes facilitates their association with actin, thereby governing actin-dependent fusion and degradative activity of late endocytic organelles.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Endocitosis , Proteínas de la Membrana/metabolismo , Orgánulos/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Chlorocebus aethiops , Factor de Crecimiento Epidérmico/farmacología , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Orgánulos/efectos de los fármacos , Orgánulos/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Tiempo
20.
Semin Cell Dev Biol ; 15(4): 445-54, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15207834

RESUMEN

Niemann-Pick type C (NPC) is a lysosomal storage disorder that results in the accumulation of cholesterol and sphingolipids. Mutations in the NPC1 or NPC2 gene are responsible for the disease but the precise functions of the encoded proteins remain unresolved. Recent observations have challenged the traditional concept of NPC as a primary cholesterol transport defect. This review updates the recent NPC literature, summarizing the increasing insight into the cholesterol trafficking circuits and also addressing the contribution of other lipids in the cellular pathogenesis. The importance of NPC as a model for subcellular lipid imbalance in studying more common diseases, such as Alzheimer's and cardiovascular diseases, is discussed.


Asunto(s)
Enfermedades de Niemann-Pick/patología , Proteínas Portadoras/genética , Colesterol/metabolismo , Endocitosis , Glicoproteínas/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Metabolismo de los Lípidos , Glicoproteínas de Membrana/genética , Proteína Niemann-Pick C1 , Enfermedades de Niemann-Pick/etiología , Enfermedades de Niemann-Pick/metabolismo , Proteínas de Transporte Vesicular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...