Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Clin Invest ; 134(3)2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-38032734

RESUMEN

Pancreatic ß cells are specialized for coupling glucose metabolism to insulin peptide production and secretion. Acute glucose exposure robustly and coordinately increases translation of proinsulin and proteins required for secretion of mature insulin peptide. By contrast, chronically elevated glucose levels that occur during diabetes impair ß cell insulin secretion and have been shown experimentally to suppress insulin translation. Whether translation of other genes critical for insulin secretion is similarly downregulated by chronic high glucose is unknown. Here, we used high-throughput ribosome profiling and nascent proteomics in MIN6 insulinoma cells to elucidate the genome-wide impact of sustained high glucose on ß cell mRNA translation. Before induction of ER stress or suppression of global translation, sustained high glucose suppressed glucose-stimulated insulin secretion and downregulated translation of not only insulin, but also mRNAs related to insulin secretory granule formation, exocytosis, and metabolism-coupled insulin secretion. Translation of these mRNAs was also downregulated in primary rat and human islets following ex vivo incubation with sustained high glucose and in an in vivo model of chronic mild hyperglycemia. Furthermore, translational downregulation decreased cellular abundance of these proteins. Our study uncovered a translational regulatory circuit during ß cell glucose toxicity that impairs expression of proteins with critical roles in ß cell function.


Asunto(s)
Hiperglucemia , Células Secretoras de Insulina , Islotes Pancreáticos , Neoplasias Pancreáticas , Ratas , Humanos , Animales , Secreción de Insulina , ARN Mensajero/metabolismo , Insulina/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Péptidos/metabolismo , Neoplasias Pancreáticas/metabolismo , Islotes Pancreáticos/metabolismo
2.
EBioMedicine ; 92: 104627, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37267847

RESUMEN

BACKGROUND: GM1 gangliosidosis is a rare, fatal, neurodegenerative disease caused by mutations in the GLB1 gene and deficiency in ß-galactosidase. Delay of symptom onset and increase in lifespan in a GM1 gangliosidosis cat model after adeno-associated viral (AAV) gene therapy treatment provide the basis for AAV gene therapy trials. The availability of validated biomarkers would greatly improve assessment of therapeutic efficacy. METHODS: The liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to screen oligosaccharides as potential biomarkers for GM1 gangliosidosis. The structures of pentasaccharide biomarkers were determined with mass spectrometry, as well as chemical and enzymatic degradations. Comparison of LC-MS/MS data of endogenous and synthetic compounds confirmed the identification. The study samples were analyzed with fully validated LC-MS/MS methods. FINDINGS: We identified two pentasaccharide biomarkers, H3N2a and H3N2b, that were elevated more than 18-fold in patient plasma, cerebrospinal fluid (CSF), and urine. Only H3N2b was detectable in the cat model, and it was negatively correlated with ß-galactosidase activity. Following intravenous (IV) AAV9 gene therapy treatment, reduction of H3N2b was observed in central nervous system, urine, plasma, and CSF samples from the cat model and in urine, plasma, and CSF samples from a patient. Reduction of H3N2b accurately reflected normalization of neuropathology in the cat model and improvement of clinical outcomes in the patient. INTERPRETATIONS: These results demonstrate that H3N2b is a useful pharmacodynamic biomarker to evaluate the efficacy of gene therapy for GM1 gangliosidosis. H3N2b will facilitate the translation of gene therapy from animal models to patients. FUNDING: This work was supported by grants U01NS114156, R01HD060576, ZIAHG200409, and P30 DK020579 from the National Institutes of Health (NIH) and a grant from National Tay-Sachs and Allied Diseases Association Inc.


Asunto(s)
Gangliosidosis GM1 , Enfermedades Neurodegenerativas , Animales , Gangliosidosis GM1/genética , Gangliosidosis GM1/terapia , Gangliosidosis GM1/patología , Enfermedades Neurodegenerativas/terapia , Cromatografía Liquida , Espectrometría de Masas en Tándem , beta-Galactosidasa/genética , beta-Galactosidasa/química , beta-Galactosidasa/uso terapéutico , Biomarcadores/líquido cefalorraquídeo , Terapia Genética
3.
J Lipid Res ; 61(3): 403-412, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31988149

RESUMEN

Niemann-Pick disease type C (NPC) disease is a lipid-storage disorder that is caused by mutations in the genes encoding NPC proteins and results in lysosomal cholesterol accumulation. 2-Hydroxypropyl-ß-cyclodextrin (CD) has been shown to reduce lysosomal cholesterol levels and enhance sterol homeostatic responses, but CD's mechanism of action remains unknown. Recent work provides evidence that CD stimulates lysosomal exocytosis, raising the possibility that lysosomal cholesterol is released in exosomes. However, therapeutic concentrations of CD do not alter total cellular cholesterol, and cholesterol homeostatic responses at the ER are most consistent with increased ER membrane cholesterol. To address these disparate findings, here we used stable isotope labeling to track the movement of lipoprotein cholesterol cargo in response to CD in NPC1-deficient U2OS cells. Although released cholesterol was detectable, it was not associated with extracellular vesicles. Rather, we demonstrate that lysosomal cholesterol trafficks to the plasma membrane (PM), where it exchanges with lipoprotein-bound cholesterol in a CD-dependent manner. We found that in the absence of suitable extracellular cholesterol acceptors, cholesterol exchange is abrogated, cholesterol accumulates in the PM, and reesterification at the ER is increased. These results support a model in which CD promotes intracellular redistribution of lysosomal cholesterol, but not cholesterol exocytosis or efflux, during the restoration of cholesterol homeostatic responses.


Asunto(s)
Colesterol/metabolismo , Ciclodextrinas/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colesterol/análisis , Homeostasis/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Marcaje Isotópico , Lisosomas/química , Lisosomas/metabolismo , Proteína Niemann-Pick C1
4.
J Lipid Res ; 60(3): 707-716, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30617147

RESUMEN

Cholesterol is an essential structural component of cellular membranes and precursor molecule for oxysterol, bile acid, and hormone synthesis. The study of intracellular cholesterol trafficking pathways has been limited in part due to a lack of suitable cholesterol analogues. Herein, we developed three novel diazirine alkyne cholesterol probes: LKM38, KK174, and KK175. We evaluated these probes as well as a previously described diazirine alkyne cholesterol analogue, trans-sterol, for their fidelity as cholesterol mimics and for study of cholesterol trafficking. LKM38 emerged as a promising cholesterol mimic because it both sustained the growth of cholesterol-auxotrophic cells and appropriately regulated key cholesterol homeostatic pathways. When presented as an ester in lipoprotein particles, LKM38 initially localized to the lysosome and subsequently trafficked to the plasma membrane and endoplasmic reticulum. LKM38 bound to diverse, established cholesterol binding proteins. Through a detailed characterization of the cellular behavior of a panel of diazirine alkyne probes using cell biological, biochemical trafficking assays and immunofluorescence approaches, we conclude that LKM38 can serve as a powerful tool for the study of cholesterol protein interactions and trafficking.


Asunto(s)
Alquinos/química , Colesterol/metabolismo , Diazometano/síntesis química , Diazometano/metabolismo , Espacio Intracelular/metabolismo , Sondas Moleculares/síntesis química , Sondas Moleculares/metabolismo , Transporte Biológico , Línea Celular Tumoral , Técnicas de Química Sintética , Diazometano/química , Homeostasis , Humanos , Lipoproteínas/metabolismo , Lisosomas/metabolismo , Sondas Moleculares/química
5.
Obesity (Silver Spring) ; 26(2): 284-290, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29243396

RESUMEN

OBJECTIVE: To determine the effects of gastric bypass on myocardial lipid deposition and function and the plasma lipidome in women with obesity and heart failure with preserved ejection fraction (HFpEF). METHODS: A primary cohort (N = 12) with HFpEF and obesity underwent echocardiography and magnetic resonance spectroscopy both before and 3 months and 6 months after bariatric surgery. Plasma lipidomic analysis was performed before surgery and 3 months after surgery in the primary cohort and were confirmed in a validation cohort (N = 22). RESULTS: After surgery-induced weight loss, Minnesota Living with Heart Failure questionnaire scores, cardiac mass, and liver fat decreased (P < 0.02, P < 0.001, and P = 0.007, respectively); echo-derived e' increased (P = 0.03), but cardiac fat was unchanged. Although weight loss was associated with decreases in many plasma ceramide and sphingolipid species, plasma lipid and cardiac function changes did not correlate. CONCLUSIONS: Surgery-induced weight loss in women with HFpEF and obesity was associated with improved symptoms, reverse cardiac remodeling, and improved relaxation. Although weight loss was associated with plasma sphingolipidome changes, cardiac function improvement was not associated with lipidomic or myocardial triglyceride changes. The results of this study suggest that gastric bypass ameliorates obesity-related HFpEF and that cardiac fat deposition and lipidomic changes may not be critical to its pathogenesis.


Asunto(s)
Cirugía Bariátrica/efectos adversos , Insuficiencia Cardíaca/etiología , Corazón/fisiopatología , Obesidad/complicaciones , Volumen Sistólico/fisiología , Femenino , Insuficiencia Cardíaca/patología , Humanos , Masculino , Persona de Mediana Edad , Obesidad/patología
6.
Biophys J ; 113(6): 1342-1352, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28629620

RESUMEN

Extracellular vesicles (EVs) are proposed to play important roles in intercellular communication. Two classes of EVs can be distinguished based on their intracellular origin. Exosomes are generated within endosomes and released when these fuse with the plasma membrane, whereas ectosomes bud directly from the plasma membrane. Studies of EV function have been hindered by limited understanding of their biogenesis. Components of the endosomal sorting complex required for transport (ESCRT) machinery play essential roles in topologically equivalent processes at both the endosome and the plasma membrane and are consistently recovered in EVs, but whether they are generally required to produce EVs is still debated. Here, we study the effects of inhibiting the ESCRT-associated AAA+ ATPase VPS4 on EV release from cultured cells using two methods for EV recovery, differential centrifugation and polyethylene glycol precipitation followed by lectin affinity chromatography. We find that inhibiting VPS4 in HEK293 cells decreases release of EV-associated proteins and miRNA as well as the overall number of EV particles. The tetraspanins CD63 and CD9 are among the most frequently monitored EV proteins, but they differ in their subcellular localization, with CD63 primarily in endosomes and CD9 on the plasma membrane. We find that CD63 and CD9 are enriched in separable populations of EVs that are both sensitive to VPS4 inhibition. Serum stimulation increases release of both types of EVs and is also reduced by inhibiting VPS4. Taken together, our data indicate that VPS4 activity is important for generating exosomes and ectosomes, thereby generally implicating the ESCRT machinery in EV biogenesis.


Asunto(s)
Adenosina Trifosfatasas/química , Endosomas , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Células HEK293 , Humanos , Transporte de Proteínas
7.
Am J Respir Crit Care Med ; 191(3): 275-84, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25494452

RESUMEN

RATIONALE: Chronic obstructive pulmonary disease (COPD) occurs in a minority of smokers and is characterized by intermittent exacerbations and clinical subphenotypes such as emphysema and chronic bronchitis. Although sphingolipids as a class are implicated in the pathogenesis of COPD, the particular sphingolipid species associated with COPD subphenotypes remain unknown. OBJECTIVES: To use mass spectrometry to determine which plasma sphingolipids are associated with subphenotypes of COPD. METHODS: One hundred twenty-nine current and former smokers from the COPDGene cohort had 69 distinct sphingolipid species detected in plasma by targeted mass spectrometry. Of these, 23 were also measured in 131 plasma samples (117 independent subjects) using an untargeted platform in an independent laboratory. Regression analysis with adjustment for clinical covariates, correction for false discovery rate, and metaanalysis were used to test associations between COPD subphenotypes and sphingolipids. Peripheral blood mononuclear cells were used to test associations between sphingolipid gene expression and plasma sphingolipids. MEASUREMENTS AND MAIN RESULTS: Of the measured plasma sphingolipids, five sphingomyelins were associated with emphysema; four trihexosylceramides and three dihexosylceramides were associated with COPD exacerbations. Three sphingolipids were strongly associated with sphingolipid gene expression, and 15 sphingolipid gene/metabolite pairs were differentially regulated between COPD cases and control subjects. CONCLUSIONS: There is evidence of systemic dysregulation of sphingolipid metabolism in patients with COPD. Subphenotyping suggests that sphingomyelins are strongly associated with emphysema and glycosphingolipids are associated with COPD exacerbations.


Asunto(s)
Glicoesfingolípidos/sangre , Espectrometría de Masas , Enfermedad Pulmonar Obstructiva Crónica/sangre , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Biomarcadores/sangre , Regulación de la Expresión Génica , Humanos , Leucocitos Mononucleares/metabolismo , Fenotipo , Valor Predictivo de las Pruebas , Enfisema Pulmonar/sangre , Enfisema Pulmonar/diagnóstico , Factores de Riesgo , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Fumar/efectos adversos , Esfingomielinas/sangre , Trihexosilceramidas/sangre
8.
J Mol Cell Cardiol ; 79: 275-83, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25497302

RESUMEN

BACKGROUND: Obesity leads to metabolic heart disease (MHD) that is associated with a pathologic increase in myocardial fatty acid (FA) uptake and impairment of mitochondrial function. The mechanism of mitochondrial dysfunction in MHD, which results in oxidant production and decreased energetics, is poorly understood but may be related to excess FAs. Determining the effects of cardiac FA excess on mitochondria can be hindered by the systemic sequelae of obesity. Mice with cardiomyocyte-specific overexpression of the fatty acid transport protein FATP1 have increased cardiomyocyte FA uptake and develop MHD in the absence of systemic lipotoxicity, obesity or diabetes. We utilized this model to assess 1) the effect of cardiomyocyte lipid accumulation on mitochondrial structure and energetic function and 2) the role of lipid-driven transcriptional regulation, signaling, toxic metabolite accumulation, and mitochondrial oxidative stress in lipid-induced MHD. METHODS: Cardiac lipid species, lipid-dependent signaling, and mitochondrial structure/function were examined from FATP1 mice. Cardiac structure and function were assessed in mice overexpressing both FATP1 and mitochondrial-targeted catalase. RESULTS: FATP1 hearts exhibited a net increase (+12%) in diacylglycerol, with increases in several very long-chain diacylglycerol species (+160-212%, p<0.001) and no change in ceramide, sphingomyelin, or acylcarnitine content. This was associated with an increase in phosphorylation of PKCα and PKCδ, and a decrease in phosphorylation of AKT and expression of CREB, PGC1α, PPARα and the mitochondrial fusion genes MFN1, MFN2 and OPA1. FATP1 overexpression also led to marked decreases in mitochondrial size (-49%, p<0.01), complex II-driven respiration (-28.6%, p<0.05), activity of isolated complex II (-62%, p=0.05), and expression of complex II subunit B (SDHB) (-60% and -31%, p<0.01) in the absence of change in ATP synthesis. Hydrogen peroxide production was not increased in FATP1 mitochondria, and cardiac hypertrophy and diastolic dysfunction were not attenuated by overexpression of catalase in mitochondria in FATP1 mice. CONCLUSIONS: Excessive delivery of FAs to the cardiac myocyte in the absence of systemic disorders leads to activation of lipid-driven signaling and remodeling of mitochondrial structure and function.


Asunto(s)
Lípidos/efectos adversos , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Carnitina/análogos & derivados , Carnitina/metabolismo , Catalasa/metabolismo , Ceramidas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Diglicéridos/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Proteínas de Transporte de Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Peróxido de Hidrógeno/metabolismo , Ratones , Mitocondrias Cardíacas/ultraestructura , Modelos Biológicos , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Especificidad de Órganos , Consumo de Oxígeno , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esfingomielinas/metabolismo , Factores de Transcripción/metabolismo
9.
J Biol Chem ; 288(5): 2923-32, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23250746

RESUMEN

Macrophages play a key role in host defense and in tissue repair after injury. Emerging evidence suggests that macrophage dysfunction in states of lipid excess can contribute to the development of insulin resistance and may underlie inflammatory complications of diabetes. Ceramides are sphingolipids that modulate a variety of cellular responses including cell death, autophagy, insulin signaling, and inflammation. In this study we investigated the intersection between TLR4-mediated inflammatory signaling and saturated fatty acids with regard to ceramide generation. Primary macrophages treated with lipopolysaccharide (LPS) did not produce C16 ceramide, whereas palmitate exposure led to a modest increase in this sphingolipid. Strikingly, the combination of LPS and palmitate led to a synergistic increase in C16 ceramide. This response occurred via cross-talk at the level of de novo ceramide synthesis in the ER. The synergistic response required TLR4 signaling via MyD88 and TIR-domain-containing adaptor-inducing interferon beta (TRIF), whereas palmitate-induced ceramide production occurred independent of these inflammatory molecules. This ceramide response augmented IL-1ß and TNFα release, a process that may contribute to the enhanced inflammatory response in metabolic diseases characterized by dyslipidemia.


Asunto(s)
Ceramidas/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Palmitatos/farmacología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Vías Biosintéticas/efectos de los fármacos , Extractos Celulares , Células Cultivadas , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Esfingolípidos/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 33(2): 232-40, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23241406

RESUMEN

OBJECTIVE: Saturated fatty acids, such as palmitic and stearic acid, cause detrimental effects in endothelial cells and have been suggested to contribute to macrophage accumulation in adipose tissue and the vascular wall, in states of obesity and insulin resistance. Long-chain fatty acids are believed to require conversion into acyl-CoA derivatives to exert most of their detrimental effects, a reaction catalyzed by acyl-CoA synthetases (ACSLs). The objective of this study was to investigate the role of ACSL1, an ACSL isoform previously shown to mediate inflammatory effects in myeloid cells, in regulating endothelial cell responses to a saturated fatty acid-rich environment in vitro and in vivo. METHODS AND RESULTS: Saturated fatty acids caused increased inflammatory activation, endoplasmic reticulum stress, and apoptosis in mouse microvascular endothelial cells. Forced ACSL1 overexpression exacerbated the effects of saturated fatty acids on apoptosis and endoplasmic reticulum stress. However, endothelial ACSL1 deficiency did not protect against the effects of saturated fatty acids in vitro, nor did it protect insulin-resistant mice fed a saturated fatty acid-rich diet from macrophage adipose tissue accumulation or increased aortic adhesion molecule expression. CONCLUSIONS: Endothelial ACSL1 is not required for inflammatory and apoptotic effects of a saturated fatty acid-rich environment.


Asunto(s)
Apoptosis , Coenzima A Ligasas/metabolismo , Células Endoteliales/enzimología , Ácidos Grasos/metabolismo , Inflamación/enzimología , Obesidad/enzimología , Acilcoenzima A/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Aorta/metabolismo , Bovinos , Células Cultivadas , Coenzima A Ligasas/deficiencia , Coenzima A Ligasas/genética , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Células Endoteliales/inmunología , Células Endoteliales/patología , Activación Enzimática , Inflamación/inmunología , Inflamación/patología , Resistencia a la Insulina , Molécula 1 de Adhesión Intercelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/inmunología , Obesidad/patología , Palmitoil Coenzima A/metabolismo , Interferencia de ARN , Factores de Tiempo , Transfección , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Am J Physiol Heart Circ Physiol ; 303(11): H1366-73, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23042950

RESUMEN

Diabetes is associated with myocardial lipid accumulation and an increased risk of heart failure. Although cardiac myocyte lipid overload is thought to contribute to the pathogenesis of cardiomyopathy in the setting of diabetes, the mechanism(s) through which this occurs is not well understood. Increasingly, inflammation has been recognized as a key pathogenic feature of lipid excess and diabetes. In this study, we sought to investigate the role of inflammatory activation in the pathogenesis of lipotoxic cardiomyopathy using the α-myosin heavy chain promoter-driven long-chain acylCoA synthetase 1 (MHC-ACS) transgenic mouse model. We found that several inflammatory cytokines were upregulated in the myocardium of MHC-ACS mice before the onset of cardiac dysfunction, and this was accompanied by macrophage infiltration. Depletion of macrophages with liposomal clodrolip reduced the cardiac inflammatory response and improved cardiac function. Thus, in this model of lipotoxic cardiac injury, early induction of inflammation and macrophage recruitment contribute to adverse cardiac remodeling. These findings have implications for our understanding of heart failure in the setting of obesity and diabetes.


Asunto(s)
Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Corazón/fisiopatología , Lípidos/fisiología , Macrófagos/fisiología , Animales , Cardiomiopatías/metabolismo , Movimiento Celular/fisiología , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Metabolismo de los Lípidos/fisiología , Macrófagos/patología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo
12.
J Clin Invest ; 118(6): 2281-90, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18483620

RESUMEN

Niemann-Pick C1 (NPC1) is a key participant in cellular cholesterol trafficking. Loss of NPC1 function leads to defective suppression of SREBP-dependent gene expression and failure to appropriately activate liver X receptor-mediated (LXR-mediated) pathways, ultimately resulting in intracellular cholesterol accumulation. To determine whether NPC1 contributes to regulation of macrophage sterol homeostasis in vivo, we examined the effect of NPC1 deletion in BM-derived cells on atherosclerotic lesion development in the Ldlr-/- mouse model of atherosclerosis. High-fat diet-fed chimeric Npc1-/- mice reconstituted with Ldlr-/-Npc1-/- macrophages exhibited accelerated atherosclerosis despite lower serum cholesterol compared with mice reconstituted with wild-type macrophages. The discordance between the low serum lipoprotein levels and the presence of aortic atherosclerosis suggested that intrinsic alterations in macrophage sterol metabolism in the chimeric Npc1-/- mice played a greater role in atherosclerotic lesion formation than did serum lipoprotein levels. Macrophages from chimeric Npc1-/- mice showed decreased synthesis of 27-hydroxycholesterol (27-HC), an endogenous LXR ligand; decreased expression of LXR-regulated cholesterol transporters; and impaired cholesterol efflux. Lower 27-HC levels were associated with elevated cholesterol oxidation products in macrophages and plasma of chimeric Npc1-/- mice and with increased oxidative stress. Our results demonstrate that NPC1 serves an atheroprotective role in mice through regulation of LXR-dependent cholesterol efflux and mitigation of cholesterol-induced oxidative stress in macrophages.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Macrófagos/metabolismo , Proteínas/genética , Proteínas/fisiología , Alimentación Animal , Animales , Aorta/patología , Transporte Biológico , Colesterol/sangre , Hidroxicolesteroles/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Transgénicos , Modelos Biológicos , Proteína Niemann-Pick C1 , Estrés Oxidativo , Esteroles/metabolismo , Factores de Tiempo
13.
J Biol Chem ; 283(13): 8229-36, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18216017

RESUMEN

Over 200 disease-causing mutations have been identified in the NPC1 gene. The most prevalent mutation, NPC1(I1061T), is predicted to lie within the cysteine-rich luminal domain and is associated with the classic juvenile-onset phenotype of Niemann-Pick type C disease. To gain insight into the molecular mechanism by which the NPC1(I1061T) mutation causes disease, we examined expression of the mutant protein in human fibroblasts homozygous for the NPC1(I1061T) mutation. Despite similar NPC1 mRNA levels between wild type and NPC1(I1061T) fibroblasts, NPC1 protein levels are decreased by 85% in NPC1(I1061T) cells. Metabolic labeling studies demonstrate that unlike wild type protein, which undergoes a glycosylation pattern shift from Endo H-sensitive to Endo H-resistant species, NPC1(I1061T) protein remains almost exclusively Endo H-sensitive and exhibits a reduced half-life (t((1/2)) 6.5 h) versus wild type Endo H-resistant species (t((1/2)) 42 h). Treatment with chemical chaperones, growth at permissive temperature, or inhibition of proteasomal degradation increases NPC1(I1061T) protein levels, indicating that the mutant protein is likely targeted for endoplasmic reticulum-associated degradation (ERAD) due to protein misfolding. Overexpression of NPC1(I1061T) in NPC1-deficient cells results in late endosomal localization of the mutant protein and complementation of the NPC mutant phenotype, likely due to a small proportion of the nascent NPC1(I1061T) protein that is able to fold correctly and escape the endoplasmic reticulum quality control checkpoints. Our findings provide the first description of an endoplasmic reticulum trafficking defect as a mechanism for human NPC disease, shedding light on the mechanism by which the NPC1(I1061T) mutation causes disease and suggesting novel approaches to treat NPC disease caused by the NPC1(I1061T) mutation.


Asunto(s)
Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Pliegue de Proteína , Animales , Proteínas Portadoras/genética , Células Cultivadas , Colesterol/metabolismo , Esterificación , Fibroblastos , Regulación Enzimológica de la Expresión Génica , Glicósido Hidrolasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Isoleucina/genética , Isoleucina/metabolismo , Glicoproteínas de Membrana/genética , Mutación/genética , Proteína Niemann-Pick C1 , Complejo de la Endopetidasa Proteasomal , Treonina/genética , Treonina/metabolismo , Factores de Tiempo
14.
Proc Natl Acad Sci U S A ; 101(37): 13624-9, 2004 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-15347805

RESUMEN

The physiologic function of the progressive hyperleptinemia of diet-induced obesity is unknown. However, that lipotoxicity in nonadipose tissues of congenitally unleptinized obese rodents is far greater than in hyperleptinemic diet-induced obesity rodents has suggested an antilipotoxic role. To test this hypothesis, mice with severe lipotoxic cardiomyopathy, induced transgenically by cardiomyocyte-specific overexpression of the acyl CoA synthase (ACS) gene, were made hyperleptinemic by treatment with recombinant adenovirus containing the leptin cDNA. Normoleptinemic control ACS-transgenic mice developed severe dilated cardiomyopathy with thickened left ventricular walls and profound impairment of systolic function on echocardiogram; histologically, there was severe myofiber disorganization and interstitial fibrosis, with intracytoplasmic lipid vacuoles identifiable by electron microscope. By contrast, the hearts of hyperleptinemic ACS-transgenic mice appeared normal, with normal echocardiograms and cardiac triglyceride (TG) contents. Their lower myocardial TG content was ascribed primarily to profound lowering of plasma TG and free fatty acids; free fatty acids were 17% of normal at 8 weeks. Additionally, enhanced myocardial AMP-activated protein kinase phosphorylation may have increased fatty acid oxidation, thereby contributing to the lowering of lipid stores. We conclude that obesity-level hyperleptinemia protects the heart from lipotoxicity.


Asunto(s)
Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Coenzima A Ligasas/metabolismo , Leptina/sangre , Lípidos/sangre , Animales , Restricción Calórica , Cardiomiopatías/sangre , Cardiomiopatías/enzimología , Coenzima A Ligasas/genética , Ratones , Ratones Transgénicos , Miocardio/patología , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Triglicéridos/sangre
15.
J Biol Chem ; 279(10): 8938-45, 2004 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-14688288

RESUMEN

Lipid accumulation in pancreatic beta-cells is thought to cause its dysfunction and/or destruction via apoptosis. Our studies show that incubation of the beta-cell line RINm5F with the saturated free fatty acids (FFA) palmitate caused apoptosis based on increases in caspase 3 activity, Annexin V staining, and cell death. Furthermore, exposure of RINm5F cells to cAMP-increasing agents, 3-isobutyl-1-methylxanthine (IBMX), and forskolin completely abolished palmitate-mediated caspase 3 activity and significantly inhibited Annexin V staining and cell death. The cyclic AMP analogs cpt-cAMP and dibutyryl-cAMP mimicked the protective effects of IBMX and forskolin, suggesting that cAMP is the mediator of the anti-apoptotic effects. The protective action of IBMX and forskolin was rapid and did not appear to require gene transcription or new protein synthesis. However, these protective effects were clearly independent of protein kinase A (PKA) because of the lack of inhibition by the PKA inhibitors H-89 and KT5720. In attempts to identify this PKA-independent mechanism, we found that the newly developed cAMP analog 8CPT-2Me-cAMP, which selectively activates the cAMP-dependent guanine nucleotide exchange factor (cAMP-GEF) pathway, mimicked the protective effects of IBMX and forskolin, suggesting that the cAMP-GEF pathway is involved. In addition, both glucagon-like peptide (GLP-1) and its receptor agonist, Exenatide, inhibited palmitate-mediated caspase 3 activation in a dose-dependent manner. Unexpectedly, H-89 partially reversed the protective effects of GLP-1 and Exenatide, suggesting that PKA may play a role in the protective effects of these incretins. To explain these seemingly conflicting results, we demonstrated that low concentrations of cAMP produced by GLP-1 and Exenatide preferentially activate the PKA pathway, whereas higher cAMP concentrations produced by IBMX and forskolin activate the more dominant cAMP-GEF pathway. Taken together, these results indicate that intracellular concentrations of cAMP may play a key role in determining divergent signaling pathways that lead to antiapoptotic responses.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Islotes Pancreáticos/metabolismo , Sulfonamidas , 1-Metil-3-Isobutilxantina/farmacología , Animales , Apoptosis/fisiología , Carbazoles/farmacología , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Islotes Pancreáticos/patología , Isoquinolinas/farmacología , Ratones , Palmitatos/farmacología , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos
16.
J Biol Chem ; 277(10): 8267-72, 2002 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11751901

RESUMEN

Perilipin (Peri) A is a phosphoprotein located at the surface of intracellular lipid droplets in adipocytes. Activation of cyclic AMP-dependent protein kinase (PKA) results in the phosphorylation of Peri A and hormone-sensitive lipase (HSL), the predominant lipase in adipocytes, with concurrent stimulation of adipocyte lipolysis. To investigate the relative contributions of Peri A and HSL in basal and PKA-mediated lipolysis, we utilized NIH 3T3 fibroblasts lacking Peri A and HSL but stably overexpressing acyl-CoA synthetase 1 (ACS1) and fatty acid transport protein 1 (FATP1). When incubated with exogenous fatty acids, ACS1/FATP1 cells accumulated 5 times more triacylglycerol (TG) as compared with NIH 3T3 fibroblasts. Adenoviral-mediated expression of Peri A in ACS1/FATP1 cells enhanced TG accumulation and inhibited lipolysis, whereas expression of HSL fused to green fluorescent protein (GFPHSL) reduced TG accumulation and enhanced lipolysis. Forskolin treatment induced Peri A hyperphosphorylation and abrogated the inhibitory effect of Peri A on lipolysis. Expression of a mutated Peri A Delta 3 (Ser to Ala substitutions at PKA consensus sites Ser-81, Ser-222, and Ser-276) reduced Peri A hyperphosphorylation and blocked constitutive and forskolin-stimulated lipolysis. Thus, perilipin expression and phosphorylation state are critical regulators of lipid storage and hydrolysis in ACS1/FATP1 cells.


Asunto(s)
Adenoviridae/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfoproteínas/metabolismo , Esterol Esterasa/metabolismo , Células 3T3 , Alanina/química , Animales , Proteínas de la Membrana Bacteriana Externa/metabolismo , Western Blotting , Proteínas Portadoras , Línea Celular , Colforsina/farmacología , Hidrólisis , Metabolismo de los Lípidos , Ratones , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , Perilipina-1 , Fosforilación , Plásmidos/metabolismo , Unión Proteica , Conformación Proteica , Serina/química , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA