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.
Mol Metab ; 24: 108-119, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30940487

RESUMEN

OBJECTIVE: Impaired expansion of peripheral fat contributes to the pathogenesis of insulin resistance and Type 2 Diabetes (T2D). We aimed to identify novel disease-gene interactions during adipocyte differentiation. METHODS: Genes in disease-associated loci for T2D, adiposity and insulin resistance were ranked according to expression in human adipocytes. The top 125 genes were ablated in human pre-adipocytes via CRISPR/CAS9 and the resulting cellular phenotypes quantified during adipocyte differentiation with high-content microscopy and automated image analysis. Morphometric measurements were extracted from all images and used to construct morphologic profiles for each gene. RESULTS: Over 107 morphometric measurements were obtained. Clustering of the morphologic profiles accross all genes revealed a group of 14 genes characterized by decreased lipid accumulation, and enriched for known lipodystrophy genes. For two lipodystrophy genes, BSCL2 and AGPAT2, sub-clusters with PLIN1 and CEBPA identifed by morphological similarity were validated by independent experiments as novel protein-protein and gene regulatory interactions. CONCLUSIONS: A morphometric approach in adipocytes can resolve multiple cellular mechanisms for metabolic disease loci; this approach enables mechanistic interrogation of the hundreds of metabolic disease loci whose function still remains unknown.


Asunto(s)
Adipocitos/citología , Adipogénesis , Diabetes Mellitus/genética , Redes Reguladoras de Genes , Mapas de Interacción de Proteínas , Aciltransferasas/genética , Aciltransferasas/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Diabetes Mellitus/patología , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Células HEK293 , Humanos , Resistencia a la Insulina , Perilipina-1/genética , Perilipina-1/metabolismo , Fenotipo , Transcriptoma
2.
Cell Metab ; 27(6): 1236-1248.e6, 2018 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-29706567

RESUMEN

Diacylglycerol acyltransferase 2 (DGAT2) catalyzes the final step in triglyceride (TG) synthesis and has been shown to play a role in regulating hepatic very-low-density lipoprotein (VLDL) production in rodents. To explore the potential of DGAT2 as a therapeutic target for the treatment of dyslipidemia, we tested the effects of small-molecule inhibitors and gene silencing both in vitro and in vivo. Consistent with prior reports, chronic inhibition of DGAT2 in a murine model of obesity led to correction of multiple lipid parameters. In contrast, experiments in primary human, rhesus, and cynomolgus hepatocytes demonstrated that selective inhibition of DGAT2 has only a modest effect. Acute and chronic inhibition of DGAT2 in rhesus primates recapitulated the in vitro data yielding no significant effects on production of plasma TG or VLDL apolipoprotein B. These results call into question whether selective inhibition of DGAT2 is sufficient for remediation of dyslipidemia.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/antagonistas & inhibidores , Dislipidemias/metabolismo , Hepatocitos/metabolismo , Obesidad/metabolismo , Triglicéridos/metabolismo , Animales , Apolipoproteínas B/metabolismo , Células Cultivadas , Diacilglicerol O-Acetiltransferasa/genética , Modelos Animales de Enfermedad , Silenciador del Gen , Humanos , Lipoproteínas VLDL/metabolismo , Macaca fascicularis , Macaca mulatta , Ratones , Ratones Endogámicos C57BL
3.
Metabolism ; 71: 202-212, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28521874

RESUMEN

BACKGROUND: SREBP cleavage-activating protein (SCAP) is a cholesterol binding endoplasmic reticulum (ER) membrane protein that is required to activate SREBP transcription factors. SREBPs regulate genes involved in lipid biosynthesis. They also influence lipid clearance by modulating the expression of LDL receptor (LDLR) and proprotein convertase subtilisin/kexin type 9 (PCSK9) genes. Inhibiting SCAP decreases circulating PCSK9, triglycerides (TG), and LDL-cholesterol (LDL-C), both in vitro and in vivo. Type 2 diabetics with dyslipidemia are at high risk for cardiovascular diseases. These patients present a unique pathophysiological lipid profile characterized by moderately elevated LDL-C, elevated TG and reduced HDL-cholesterol (HDL-C). The spontaneous dysmetabolic rhesus monkey model (DysMet RhM) recapitulates this human dyslipidemia and therefore is an attractive preclinical model to evaluate SCAP inhibition as a therapy for this disease population. The objective to of this study was to assess the effect of SCAP inhibition on the lipid profile of DysMet RhM. METHOD: We assessed the effect of inhibiting hepatic SCAP on the lipid profile of DysMet RhM using an siRNA encapsulated lipid nanoparticle (siRNA-LNP). RESULTS: The SCAP siRNA-LNP significantly reduced LDL-C, PCSK9 and TG in DysMet RhM; LDL-C was reduced by ≥20%, circulating PCSK9 by 30-40% and TG by >25%. These changes by the SCAP siRNA-LNP agree with the predicted effect of SCAP inhibition and reduced SREBP tone on these endpoints. CONCLUSION: These data demonstrate that a SCAP siRNA-LNP improved the lipid profile in a clinically relevant preclinical disease model and provide evidence for SCAP inhibition as a therapy for diabetic dyslipidemic patients.


Asunto(s)
Dislipidemias/prevención & control , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , ARN Interferente Pequeño/uso terapéutico , Animales , Biomarcadores/sangre , Colesterol/sangre , Femenino , Lípidos/sangre , Hígado/metabolismo , Macaca mulatta , Masculino , Nanopartículas , Ácido Palmítico/sangre , Proproteína Convertasa 9/sangre , ARN Interferente Pequeño/administración & dosificación , Triglicéridos/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(9): 2389-2394, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28193887

RESUMEN

Parkinson disease (PD) is a neurodegenerative disorder pathologically characterized by nigrostriatal dopamine neuron loss and the postmortem presence of Lewy bodies, depositions of insoluble α-synuclein, and other proteins that likely contribute to cellular toxicity and death during the disease. Genetic and biochemical studies have implicated impaired lysosomal and mitochondrial function in the pathogenesis of PD. Transmembrane protein 175 (TMEM175), the lysosomal K+ channel, is centered under a major genome-wide association studies peak for PD, making it a potential candidate risk factor for the disease. To address the possibility that variation in TMEM175 could play a role in PD pathogenesis, TMEM175 function was investigated in a neuronal model system. Studies confirmed that TMEM175 deficiency results in unstable lysosomal pH, which led to decreased lysosomal catalytic activity, decreased glucocerebrosidase activity, impaired autophagosome clearance by the lysosome, and decreased mitochondrial respiration. Moreover, TMEM175 deficiency in rat primary neurons resulted in increased susceptibility to exogenous α-synuclein fibrils. Following α-synuclein fibril treatment, neurons deficient in TMEM175 were found to have increased phosphorylated and detergent-insoluble α-synuclein deposits. Taken together, data from these studies suggest that TMEM175 plays a direct and critical role in lysosomal and mitochondrial function and PD pathogenesis and highlight this ion channel as a potential therapeutic target for treating PD.


Asunto(s)
Autofagosomas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Lisosomas/metabolismo , Mitocondrias/metabolismo , Canales de Potasio/genética , alfa-Sinucleína/química , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/patología , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Regulación de la Expresión Génica , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/efectos de los fármacos , Lisosomas/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Modelos Biológicos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Canales de Potasio/deficiencia , Cultivo Primario de Células , Agregado de Proteínas/efectos de los fármacos , Ratas , alfa-Sinucleína/farmacología
5.
J Lipid Res ; 57(12): 2150-2162, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27707816

RESUMEN

SREBP cleavage-activating protein (SCAP) is a key protein in the regulation of lipid metabolism and a potential target for treatment of dyslipidemia. SCAP is required for activation of the transcription factors SREBP-1 and -2. SREBPs regulate the expression of genes involved in fatty acid and cholesterol biosynthesis, and LDL-C clearance through the regulation of LDL receptor (LDLR) and PCSK9 expression. To further test the potential of SCAP as a novel target for treatment of dyslipidemia, we used siRNAs to inhibit hepatic SCAP expression and assess the effect on PCSK9, LDLR, and lipids in mice and rhesus monkeys. In mice, robust liver Scap mRNA knockdown (KD) was achieved, accompanied by dose-dependent reduction in SREBP-regulated gene expression, de novo lipogenesis, and plasma PCSK9 and lipids. In rhesus monkeys, over 90% SCAP mRNA KD was achieved resulting in approximately 75, 50, and 50% reduction of plasma PCSK9, TG, and LDL-C, respectively. Inhibition of SCAP function was demonstrated by reduced expression of SREBP-regulated genes and de novo lipogenesis. In conclusion, siRNA-mediated inhibition of SCAP resulted in a significant reduction in circulating PCSK9 and LDL-C in rodent and primate models supporting SCAP as a novel target for the treatment of dyslipidemia.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Lípidos/sangre , Proteínas de la Membrana/genética , Proproteína Convertasa 9/genética , ARN Interferente Pequeño/genética , Receptores de LDL/genética , Animales , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Hipolipemiantes/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipogénesis , Hígado/enzimología , Macaca mulatta , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proproteína Convertasa 9/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de LDL/metabolismo , Transducción de Señal , Simvastatina/farmacología , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
6.
Mol Ther Nucleic Acids ; 5(8): e342, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27483025

RESUMEN

Short interfering RNAs (siRNAs) are a valuable tool for gene silencing with applications in both target validation and therapeutics. Many advances have recently been made to improve potency and specificity, and reduce toxicity and immunostimulation. However, siRNA delivery to a variety of tissues remains an obstacle for this technology. To date, siRNA delivery to muscle has only been achieved by local administration or by methods with limited potential use in the clinic. We report systemic delivery of a highly chemically modified cholesterol-conjugated siRNA targeting muscle-specific gene myostatin (Mstn) to a full range of muscles in mice. Following a single intravenous injection, we observe 85-95% knockdown of Mstn mRNA in skeletal muscle and >65% reduction in circulating Mstn protein sustained for >21 days. This level of Mstn knockdown is also accompanied by a functional effect on skeletal muscle, with animals showing an increase in muscle mass, size, and strength. The cholesterol-conjugated siRNA platform described here could have major implications for treatment of a variety of muscle disorders, including muscular atrophic diseases, muscular dystrophy, and type II diabetes.

7.
Pharmacol Res Perspect ; 4(1): e00207, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26977298

RESUMEN

The benefits of novel oral anticoagulants are hampered by bleeding. Since coagulation factor IX (fIX) lies upstream of fX in the coagulation cascade, and intermediate levels have been associated with reduced incidence of thrombotic events, we evaluated the viability of fIXa as an antithrombotic target. We applied translational pharmacokinetics/pharmacodynamics (PK/PD) principles to predict the therapeutic window (TW) associated with a selective small molecule inhibitor (SMi) of fIXa, compound 1 (CPD1, rat fIXa inhibition constant (Ki, 21 nmol/L) relative to clinically relevant exposures of apixaban (rat fXa Ki 4.3 nmol/L). Concentrations encompassing the minimal clinical plasma concentration (C min) of the 5 mg twice daily (BID) dose of apixaban were tested in rat arteriovenous shunt (AVS/thrombosis) and cuticle bleeding time (CBT) models. An I max and a linear model were used to fit clot weight (CW) and CBT. The following differences in biology were observed: (1) antithrombotic activity and bleeding increased in parallel for apixaban, but to a lesser extent for CPD1 and (2) antithrombotic activity occurred at high (>99%) enzyme occupancy (EO) for fXa or moderate (>65% EO) for fIXa. translational PK/PD analysis indicated that noninferiority was observed for concentrations of CPD1 that provided between 86% and 96% EO and that superior TW existed between 86% and 90% EO. These findings were confirmed in a study comparing short interfering (si)RNA-mediated knockdown (KD) modulation of fIX and fX mRNA. In summary, using principles of translational biology to relate preclinical markers of efficacy and safety to clinical doses of apixaban, we found that modulation of fIXa can be superior to apixaban.

8.
Atherosclerosis ; 242(2): 383-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26275376

RESUMEN

The greater genomic conservation between humans and non-human primates (NHP) enables target validation studies for developing of therapeutic strategies for human diseases. Together with predicting activity and potential adverse clinical signs, the inclusion of NHP testing bequeaths to efficacy models for dose titration and pharmacodynamic effects. We have used lipid nanoparticle encapsulated siRNA to silence ApoB in the liver and assessed the phenotypic effects on serum lipids with various levels of hepatic ApoB mRNA knockdown in healthy lean cynomolgus monkeys. ApoB siRNA dosed animals demonstrated significant reductions of hepatic ApoB mRNA and serum APOB protein, with a substantial lowering of plasma lipid levels without obvious signs of toxicity. Microarray based assessment of ApoB siRNA mediated effects revealed a number of differentially expressed genes which mapped onto biological pathways and processes related to lipid and cholesterol metabolism. Furthermore, we identified potential targets and cellular effects that could be studied for therapeutic benchmarking of APOB mediated effects. The network of ApoB regulated genes should be of significance for the understanding and development of novel hypercholesterolemia therapies.


Asunto(s)
Apolipoproteínas B/genética , Regulación de la Expresión Génica , Hígado/metabolismo , Interferencia de ARN , Animales , Biopsia , Colesterol/metabolismo , Genoma , Genómica , Humanos , Metabolismo de los Lípidos , Lípidos/sangre , Lípidos/química , Hígado/patología , Macaca fascicularis , Masculino , Nanopartículas/química , Análisis de Secuencia por Matrices de Oligonucleótidos , Biosíntesis de Proteínas , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Blood Coagul Fibrinolysis ; 26(8): 893-902, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26192114

RESUMEN

This report aims at exploring quantitatively the relationship between FXII inhibition and thromboprotection. FXII full and partial null in rats were established via zinc finger nuclease-mediated knockout and siRNA-mediated knockdown, respectively. The rats were subsequently characterized in thrombosis and hemostasis models. Knockout rats exhibited complete thromboprotection in both the arteriovenous shunt model (∼100% clot weight reduction) and the FeCl3-induced arterial thrombosis model (no reduction in blood flow), without any increase in cuticle bleeding time compared with wild-type control rats. Ex-vivo aPTT and the ellagic acid-triggered thrombin generation assay (TGA) exhibited anticoagulant changes. In contrast, ex-vivo PT or high tissue factor-triggered TGA was indistinguishable from control. Rats receiving single doses (0, 0.01, 0.03, 0.1, 0.3, 1 mg/kg) of FXII siRNA exhibited dose-dependent knockdown in liver FXII mRNA and plasma FXII protein (95 and 99%, respectively, at 1 mg/kg) at day 7 post dosing. FXII knockdown was associated with dose-dependent thromboprotection (maximal efficacy achieved with 1 mg/kg in both models) and negligible change in cuticle bleeding times. Ex-vivo TGA triggered with low-level (0.5 µmol/l) ellagic acid tracked best with the knockdown levels and efficacy. Our findings confirm and extend literature reports of an attractive benefit-to-risk profile of targeting FXII for antithrombotic therapies. Titrating of FXII is instructive for its pharmacological inhibition. The knockout rat is valuable for evaluating both mechanism-based safety concerns and off-target effects of FXII(a) inhibitors. Detailed TGA analyses will inform on optimal trigger conditions in studying pharmacodynamic effects of FXII(a) inhibition.


Asunto(s)
Endodesoxirribonucleasas/genética , Factor XII/antagonistas & inhibidores , ARN Interferente Pequeño/administración & dosificación , Terapia Trombolítica/métodos , Trombosis/terapia , Animales , Derivación Arteriovenosa Quirúrgica , Cloruros/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ácido Elágico/farmacología , Endodesoxirribonucleasas/metabolismo , Factor XII/genética , Factor XII/metabolismo , Compuestos Férricos/farmacología , Técnicas de Inactivación de Genes , Hemorragia/prevención & control , Humanos , Hígado/metabolismo , Masculino , Tiempo de Tromboplastina Parcial , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Trombina/metabolismo , Trombosis/inducido químicamente , Trombosis/genética , Trombosis/patología , Dedos de Zinc/genética
10.
Thromb Haemost ; 113(6): 1300-11, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25790442

RESUMEN

Haemophilia A and B are characterised by a life-long bleeding predisposition, and several lines of evidence suggest that risks of atherothrombotic events may also be reduced. Establishing a direct correlation between coagulation factor levels, thrombotic risks and bleeding propensity has long been hampered by an inability to selectively and specifically inhibit coagulation factor levels. Here, the exquisite selectivity of gene silencing combined with a gene knockout (KO) approach was used to define the relative contribution of factor IX (fIX) to thrombosis and primary haemostasis in the rat. Using a lipid nanoparticle (LNP) formulation, we successfully delivered fIX siRNAs to the liver by intravenous administration. The knockdown (KD) of target gene mRNA was achieved rapidly (within 24 hour post-siRNA dosing), sustained (maintained for at least 7 days post dosing) and not associated with changes in mRNA expression levels of other coagulation factors. We found that intermediate levels of liver fIX mRNA silencing (60-95 %) translating into a 50-99 % reduction of plasma fIX activity provided protection from thrombosis without prolonging the cuticle bleeding time. Over 99 % inhibition of fIX activity was required to observe increase in bleeding, a phenotype confirmed in fIX KO rats. These data provide substantial evidence of a participation of fIX in the mechanisms regulating thrombosis prior to those regulating primary haemostasis, therefore highlighting the potential of fIX as a therapeutic target. In addition, hepatic mRNA silencing using LNP-encapsulated siRNAs may represent a promising novel approach for the chronic treatment and prevention of coagulation-dependent thrombotic disorders in humans.


Asunto(s)
Factor IX/genética , Hemofilia B/genética , Hemorragia/genética , Hígado/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia , Trombosis/prevención & control , Animales , Línea Celular , Cloruros , Modelos Animales de Enfermedad , Factor IX/metabolismo , Compuestos Férricos , Regulación de la Expresión Génica , Genotipo , Hemofilia B/sangre , Hemorragia/sangre , Hemostasis/genética , Masculino , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Trombosis/sangre , Trombosis/inducido químicamente , Trombosis/genética , Factores de Tiempo , Transfección
11.
J Cardiovasc Transl Res ; 8(1): 44-53, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25604958

RESUMEN

Lipoprotein(a) [Lp(a)] has recently been recognized as an independent risk factor for coronary heart disease. While plasma Lp(a) levels are correlated with cardiovascular risk, the mechanism by which this particle contributes to atherosclerosis is largely unknown. Although humanized transgenic mouse model has recently been described to study Lp(a) biology, non-human primates (NHP) are the only preclinical model available that allow study of the role of Lp(a) in atherosclerosis in an innate setting. We describe targeting of LPA using lipid nanoparticle formulated short interfering RNAs (siRNAs) in lean rhesus macaque monkeys. We show >90 % LPA mRNA lowering in the liver and >95 % Lp(a) plasma reduction for over 3 weeks after a single siRNA dose. Given the potency of LPA siRNAs, siRNA approach may enable chronic reduction of Lp(a) in atherosclerotic NHP and help to unmask the role for Lp(a) in the genesis and progression of atherosclerosis in man.


Asunto(s)
Aterosclerosis/terapia , Lipoproteína(a)/genética , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia , Animales , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Humanos , Lípidos/química , Lipoproteína(a)/sangre , Hígado/metabolismo , Macaca mulatta , Masculino , Nanomedicina , Nanopartículas , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Transfección
12.
Mol Ther Nucleic Acids ; 4: e224, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25625614

RESUMEN

The present study aimed at establishing feasibility of delivering short interfering RNA (siRNA) to target the coagulation cascade in rat and rabbit, two commonly used species for studying thrombosis and hemostasis. siRNAs that produced over 90% mRNA knockdown of rat plasma prekallikrein and rabbit Factor X (FX) were identified from in vitro screens. An ionizable amino lipid based lipid nanoparticle (LNP) formulation for siRNA in vivo delivery was characterized as tolerable and exerting no appreciable effect on coagulability at day 7 postdosing in both species. Both prekallikrein siRNA-LNP and FX siRNA-LNP resulted in dose-dependent and selective knockdown of target gene mRNA in the liver with maximum reduction of over 90% on day 7 following a single dose of siRNA-LNP. Knockdown of plasma prekallikrein was associated with modest clot weight reduction in the rat arteriovenous shunt thrombosis model and no increase in the cuticle bleeding time. Knockdown of FX in the rabbit was accompanied with prolongation in ex vivo clotting times. Results fit the expectations with both targets and demonstrate for the first time, the feasibility of targeting coagulation factors in rat, and, more broadly, targeting a gene of interest in rabbit, via systemic delivery of ionizable LNP formulated siRNA.

13.
PLoS Genet ; 10(5): e1004333, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24831815

RESUMEN

Inherited hypertrichoses are rare syndromes characterized by excessive hair growth that does not result from androgen stimulation, and are often associated with additional congenital abnormalities. In this study, we investigated the genetic defect in a case of autosomal recessive congenital generalized hypertrichosis terminalis (CGHT) (OMIM135400) using whole-exome sequencing. We identified a single base pair substitution in the 5' donor splice site of intron 32 in the ABC lipid transporter gene ABCA5 that leads to aberrant splicing of the transcript and a decrease in protein levels throughout patient hair follicles. The homozygous recessive disruption of ABCA5 leads to reduced lysosome function, which results in an accumulation of autophagosomes, autophagosomal cargos as well as increased endolysosomal cholesterol in CGHT keratinocytes. In an unrelated sporadic case of CGHT, we identified a 1.3 Mb cryptic deletion of chr17q24.2-q24.3 encompassing ABCA5 and found that ABCA5 levels are dramatically reduced throughout patient hair follicles. Collectively, our findings support ABCA5 as a gene underlying the CGHT phenotype and suggest a novel, previously unrecognized role for this gene in regulating hair growth.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Colesterol/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Cabello/crecimiento & desarrollo , Hipertricosis/congénito , Preescolar , Colesterol/genética , Deleción Cromosómica , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Cabello/patología , Humanos , Hipertricosis/genética , Hipertricosis/patología , Lactante , Queratinocitos/metabolismo , Queratinocitos/patología , Mutación , Linaje , Fenotipo , Empalme del ARN/genética , Eliminación de Secuencia
14.
Hypertens Res ; 37(5): 405-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24335718

RESUMEN

Angiotensinogen (AGT) is the precursor of active vasoconstrictive octapeptide angiotensin II (Ang II) in the renin-angiotensin-aldosterone system. Blocking the AGT-converting enzymes in the pathway and the Ang II receptor through pharmacological agents has been proven to be effective in lowering blood pressure (BP) in hypertensive patients. In this study, we developed chemically modified small interfering RNAs (siRNA) to target hepatic AGT mRNA in rats. Lipid nanoparticle encapsulated siRNAs were efficiently delivered to rat liver and resulted in significant reduction in hepatic Agt mRNA levels and plasma AGT concentration without impairing liver function. Single intravenous injection of Agt siRNA led to significant and sustained BP lowering in spontaneous hypertensive rats and in Sprague-Dawley rats, and the effect was maintained by weekly siRNA dosing. Data presented here provide proof-of-feasibility for the use of siRNA technology for inhibition of peripheral AGT levels via hepatic mRNA silencing with beneficial effects on BP in preclinical rat models. Similar approach could be used for validation of novel hypertension hepatic and extrahepatic targets.


Asunto(s)
Angiotensinógeno/metabolismo , Presión Sanguínea/genética , Hipertensión/metabolismo , Hígado/metabolismo , Sistema Renina-Angiotensina/fisiología , Angiotensinógeno/genética , Animales , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Hipertensión/genética , Hipertensión/fisiopatología , Hígado/fisiopatología , Masculino , Nanopartículas , ARN Interferente Pequeño , Ratas , Ratas Endogámicas SHR , Ratas Sprague-Dawley
15.
Proc Natl Acad Sci U S A ; 110(19): 7790-5, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23603273

RESUMEN

X-linked congenital generalized hypertrichosis (Online Mendelian Inheritance in Man 307150) is an extremely rare condition of hair overgrowth on different body sites. We previously reported linkage in a large Mexican family with X-linked congenital generalized hypertrichosis cosegregating with deafness and with dental and palate anomalies to Xq24-27. Using SNP oligonucleotide microarray analysis and whole-genome sequencing, we identified a 389-kb interchromosomal insertion at an extragenic palindrome site at Xq27.1 that completely cosegregates with the disease. Among the genes surrounding the insertion, we found that Fibroblast Growth Factor 13 (FGF13) mRNA levels were significantly reduced in affected individuals, and immunofluorescence staining revealed a striking decrease in FGF13 localization throughout the outer root sheath of affected hair follicles. Taken together, our findings suggest a role for FGF13 in hair follicle growth and in the hair cycle.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/fisiología , Regulación de la Expresión Génica , Hipertricosis/congénito , Empalme Alternativo , Animales , Mapeo Cromosómico , Femenino , Ligamiento Genético , Genoma Humano , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/fisiología , Heterocigoto , Humanos , Hipertricosis/genética , Queratinocitos/metabolismo , Masculino , Ratones , Mutagénesis Insercional , Linaje , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN
16.
J Lipid Res ; 53(5): 859-867, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22355095

RESUMEN

Microsomal triglyceride transfer protein (Mtp) inhibitors represent a novel therapeutic approach to lower circulating LDL cholesterol, although therapeutic development has been hindered by the observed increase in hepatic triglycerides and liver steatosis following treatment. Here, we used small interfering RNAs (siRNA) targeting Mtp to achieve target-specific silencing to study this phenomenon and to determine to what extent liver steatosis is induced by changes in Mtp expression. We observed that Mtp silencing led to a decrease in many genes involved in hepatic triglyceride synthesis. Given the role of diacylglycerol O-acyltransferase 2 (Dgat2) in regulating hepatic triglyceride synthesis, we then evaluated whether target-specific silencing of both Dgat2 and Mtp were sufficient to attenuate Mtp silencing-induced liver steatosis. We showed that the simultaneous inhibition of Dgat2 and Mtp led to a decrease in plasma cholesterol and a reduction in the accumulation of hepatic triglycerides caused by the inhibition of Mtp. Collectively, these findings provide a proof-of-principle for a triglyceride synthesis/Mtp inhibitor combination and represent a potentially novel approach for therapeutic development in which targeting multiple pathways can achieve the desired response.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diacilglicerol O-Acetiltransferasa/deficiencia , Diacilglicerol O-Acetiltransferasa/genética , Hígado Graso/genética , Silenciador del Gen , ARN Interferente Pequeño/genética , Animales , Apolipoproteínas B/deficiencia , Apolipoproteínas B/genética , Colesterol/sangre , Hígado Graso/sangre , Hígado Graso/enzimología , Hígado Graso/metabolismo , Hígado/metabolismo , Masculino , Ratones , Triglicéridos/metabolismo
17.
J Proteome Res ; 10(10): 4683-91, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21819150

RESUMEN

The purpose of this study was to evaluate the use of high resolution LC-MS together with metabolomics and D(4)-cholic acid (D(4)-CA) as a metabolic tracer to measure the metabolism and reconjugation of bile acids (BAs) in vitro and in vivo. Metabolic tracers are very important because they allow for the direct detection (substrate-to-product) of small and significant biological perturbations that may not be apparent when monitoring "static" endogenous levels of particular metabolites. Slc27a5, also known as fatty acid transport protein 5 (FATP5), is the hepatic BA-CoA ligase involved in reconjugating BAs during enterohepatic BA recycling. Using Slc27a5-cKD mice, silencing of ∼90% gene expression was achieved followed by reduction in the reconjugation of D(4)-CA to D(4)-taurocholic acid (D(4)-TCA), as well as other conjugated BA metabolites in plasma (p = 0.0031). The method described allowed a rapid measure of many D(4) and endogenous BA. Analysis of bile resulted in the detection of 39 BA metabolites from a 13 min analytical run. Finally, the utilization of a novel high resolution mass spectrometry method in combination with metabolomics and a stable isotope metabolic tracer allowed for the detection of targeted and untargeted BAs following silencing of the Slc27a5 gene in primary hepatocytes and in mice.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Cromatografía Liquida/métodos , Proteínas de Transporte de Ácidos Grasos/metabolismo , Hígado/patología , Espectrometría de Masas/métodos , Metabolómica/métodos , Animales , Silenciador del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Espectrometría de Masa por Ionización de Electrospray
18.
Lipids ; 46(11): 991-1003, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21826528

RESUMEN

The association between hypercholesterolemia and elevated serum apolipoprotein B (APOB) has generated interest in APOB as a therapeutic target for patients at risk of developing cardiovascular disease. In the clinic, mipomersen, an antisense oligonucleotide (ASO) APOB inhibitor, was associated with a trend toward increased hepatic triglycerides, and liver steatosis remains a concern. We found that siRNA-mediated knockdown of ApoB led to elevated hepatic triglycerides and liver steatosis in mice engineered to exhibit a human-like lipid profile. Many genes required for fatty acid synthesis were reduced, suggesting that the observed elevation in hepatic triglycerides is maintained by the cell through fatty acid uptake as opposed to fatty acid synthesis. Fatty acid transport protein 5 (Fatp5/Slc27a5) is required for long chain fatty acid (LCFA) uptake and bile acid reconjugation by the liver. Fatp5 knockout mice exhibited lower levels of hepatic triglycerides due to decreased fatty acid uptake, and shRNA-mediated knockdown of Fatp5 protected mice from diet-induced liver steatosis. Here, we evaluated if siRNA-mediated knockdown of Fatp5 was sufficient to alleviate ApoB knockdown-induced steatosis. We determined that, although Fatp5 siRNA treatment was sufficient to increase the proportion of unconjugated bile acids 100-fold, consistent with FATP5's role in bile acid reconjugation, Fatp5 knockdown failed to influence the degree, zonal distribution, or composition of the hepatic triglycerides that accumulated following ApoB siRNA treatment.


Asunto(s)
Apolipoproteínas B/genética , Proteínas de Transporte de Ácidos Grasos/genética , Hígado Graso/genética , Animales , Apolipoproteínas B/sangre , Ácidos y Sales Biliares/metabolismo , Colesterol/sangre , HDL-Colesterol/sangre , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño , Triglicéridos/sangre
19.
J Lipid Res ; 52(6): 1084-1097, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21398511

RESUMEN

Increased serum apolipoprotein (apo)B and associated LDL levels are well-correlated with an increased risk of coronary disease. ApoE⁻/⁻ and low density lipoprotein receptor (LDLr)⁻/⁻ mice have been extensively used for studies of coronary atherosclerosis. These animals show atherosclerotic lesions similar to those in humans, but their serum lipids are low in apoB-containing LDL particles. We describe the development of a new mouse model with a human-like lipid profile. Ldlr CETP⁺/⁻ hemizygous mice carry a single copy of the human CETP transgene and a single copy of a LDL receptor mutation. To evaluate the apoB pathways in this mouse model, we used novel short-interfering RNAs (siRNA) formulated in lipid nanoparticles (LNP). ApoB siRNAs induced up to 95% reduction of liver ApoB mRNA and serum apoB protein, and a significant lowering of serum LDL in Ldlr CETP⁺/⁻ mice. ApoB targeting is specific and dose-dependent, and it shows lipid-lowering effects for over three weeks. Although specific triglycerides (TG) were affected by ApoB mRNA knockdown (KD) and the total plasma lipid levels were decreased by 70%, the overall lipid distribution did not change. Results presented here demonstrate a new mouse model for investigating additional targets within the ApoB pathways using the siRNA modality.


Asunto(s)
Apolipoproteínas B/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/genética , LDL-Colesterol/sangre , Modelos Animales de Enfermedad , Receptores de LDL/genética , Animales , Apolipoproteínas B/sangre , Apolipoproteínas E/sangre , Apolipoproteínas E/genética , Aterosclerosis/patología , Línea Celular Tumoral , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Efecto Fundador , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hemicigoto , Humanos , Metabolismo de los Lípidos/genética , Liposomas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nanopartículas/administración & dosificación , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Receptores de LDL/metabolismo , Triglicéridos/sangre
20.
J Lipid Res ; 52(4): 679-87, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21262787

RESUMEN

Reducing circulating LDL-cholesterol (LDL-c) reduces the risk of cardiovascular disease in people with hypercholesterolemia. Current approaches to reduce circulating LDL-c include statins, which inhibit cholesterol synthesis, and ezetimibe, which blocks cholesterol absorption. Both elevate serum PCSK9 protein levels in patients, which could attenuate their efficacy by reducing the amount of cholesterol cleared from circulation. To determine whether PCSK9 inhibition could enhance LDL-c lowering of both statins and ezetimibe, we utilized small interfering RNAs (siRNAs) to knock down Pcsk9, together with ezetimibe, rosuvastatin, and an ezetimibe/rosuvastatin combination in a mouse model with a human-like lipid profile. We found that ezetimibe, rosuvastatin, and ezetimibe/rosuvastatin combined lower serum cholesterol but induce the expression of Pcsk9 as well as the Srebp-2 hepatic cholesterol biosynthesis pathway. Pcsk9 knockdown in combination with either treatment led to greater reductions in serum non-HDL with a near-uniform reduction of all LDL-c subfractions. In addition to reducing serum cholesterol, the combined rosuvastatin/ezetimibe/Pcsk9 siRNA treatment exhibited a significant reduction in serum APOB protein and triglyceride levels. Taken together, these data provide evidence that PCSK9 inhibitors, in combination with current therapies, have the potential to achieve greater reductions in both serum cholesterol and triglycerides.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Azetidinas/uso terapéutico , Fluorobencenos/uso terapéutico , Pirimidinas/uso terapéutico , Serina Endopeptidasas/metabolismo , Sulfonamidas/uso terapéutico , Animales , Apolipoproteínas B/sangre , Colesterol/sangre , LDL-Colesterol/sangre , Ensayo de Inmunoadsorción Enzimática , Ezetimiba , Hipercolesterolemia/sangre , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/terapia , Ratones , Ratones Endogámicos C57BL , Proproteína Convertasa 9 , Proproteína Convertasas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosuvastatina Cálcica , Serina Endopeptidasas/genética , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...