Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Biol Ther ; 22(2): 164-174, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33612070

RESUMEN

Stearoyl-CoA-desaturase 1 (SCD1) deficiency mediates apoptosis in colorectal cancer cells by promoting ceramide de novo synthesis. The mechanisms underlying the cross-talk between SCD1 and ceramide synthesis have not been explored. We treated colorectal cancer cells with an SCD1 inhibitor and examined the effects on gene expression, cell growth, and cellular lipid contents. The main effect of SCD1 inhibition on the fatty acid contents of colorectal cancer cells was a decrease in monounsaturated fatty acids (MUFAs). RNA sequencing (RNA-seq) showed that the most intense alteration of gene expression after SCD1 inhibition occurred in the NF-κB signaling pathway. Further experiments revealed that SCD1 inhibition resulted in increased levels of phosphorylated NF-κB p65 and increased nuclear translocation of NF-κB p65. Treatment with an NF-κB inhibitor eliminated several effects of SCD1 inhibition, mainly including overexpression of serine palmitoyltransferase1 (SPT1), elevation of dihydroceramide contents, and suppression of cell growth. Furthermore, treatment with supplemental oleate counteracted the SCD1-induced NF-κB activation and downstream effects. In summary, our data demonstrate that the NF-κB pathway plays a role in SCD1 deficiency-induced ceramide de novo synthesis in colorectal cancer cells, and that reduced MUFA levels contribute to the course.


Asunto(s)
Ceramidas/metabolismo , FN-kappa B/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Humanos
2.
Biochem Biophys Res Commun ; 527(3): 589-595, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32423819

RESUMEN

In mouse, there are four stearoyl-CoA desaturase isoforms (SCD1-4) that catalyze the synthesis of monounsaturated fatty acids. Previously, we have shown that mice harboring a whole body deletion of the SCD1 isoform (SCD1KO) are protected from diet and genetically induced adiposity. Here, we report that global deletion of the SCD2 isoform (SCD2KO) provides a similar protective effect against the onset of both high-fat diet (HFD) and high-carbohydrate diet (HCD) induced adiposity. After 10 weeks of HFD feeding or 6 weeks of HCD feeding, SCD2KO mice failed to gain weight and had decreased fat mass. On HFD, SCD2KO mice remained glucose and insulin tolerant. Lastly, the markers for energy expenditure, UCP1 and PGC-1α, were increased in the brown adipose tissue of HFD fed SCD2KO mice.


Asunto(s)
Adiposidad , Dieta de Carga de Carbohidratos/efectos adversos , Dieta Alta en Grasa/efectos adversos , Eliminación de Gen , Obesidad/genética , Estearoil-CoA Desaturasa/genética , Animales , Metabolismo Energético , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Factores Protectores , Estearoil-CoA Desaturasa/deficiencia , Estearoil-CoA Desaturasa/metabolismo
3.
J Exp Med ; 217(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32097464

RESUMEN

Failure of remyelination underlies the progressive nature of demyelinating diseases such as multiple sclerosis. Macrophages and microglia are crucially involved in the formation and repair of demyelinated lesions. Here we show that myelin uptake temporarily skewed these phagocytes toward a disease-resolving phenotype, while sustained intracellular accumulation of myelin induced a lesion-promoting phenotype. This phenotypic shift was controlled by stearoyl-CoA desaturase-1 (SCD1), an enzyme responsible for the desaturation of saturated fatty acids. Monounsaturated fatty acids generated by SCD1 reduced the surface abundance of the cholesterol efflux transporter ABCA1, which in turn promoted lipid accumulation and induced an inflammatory phagocyte phenotype. Pharmacological inhibition or phagocyte-specific deficiency of Scd1 accelerated remyelination ex vivo and in vivo. These findings identify SCD1 as a novel therapeutic target to promote remyelination.


Asunto(s)
Encéfalo/patología , Macrófagos/enzimología , Microglía/enzimología , Estearoil-CoA Desaturasa/metabolismo , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Línea Celular , Colesterol/metabolismo , Endocitosis , Ácidos Grasos/metabolismo , Células Espumosas/metabolismo , Humanos , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/ultraestructura , Ratones , Microglía/metabolismo , Vaina de Mielina/metabolismo , Fagocitos/patología , Fagocitos/ultraestructura , Fenotipo , Proteína Quinasa C-delta/metabolismo , Estearoil-CoA Desaturasa/deficiencia
4.
Sci Rep ; 9(1): 15761, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31673045

RESUMEN

Stearoyl CoA desaturase 1 (SCD1) is a key enzyme in lipogenesis as it catalyzes the synthesis of monounsaturated fatty acids (MUFAs), mainly oleate (18:1n9) and palmitoleate (16:1n7) from saturated fatty acids (SFA), stearate (18:0) and palmitate (16:0), respectively. Studies on SCD1 deficiency in mouse models demonstrated beneficial metabolic phenotypes such as reduced adiposity and improved glucose tolerance. Even though, SCD1 represents a potential target to resolve obesity related metabolic diseases; SCD1 deficiency causes endoplasmic reticulum (ER) stress and activates unfolded protein response (UPR). The induction of ER stress in response to SCD1 deficiency is governed by the cofactor, PGC-1α. However, the mechanism by which SCD1 deficiency increases PGC-1α and subsequently induces ER stress still remains elusive. The present study demonstrates that despite reduced lipogenesis, liver specific SCD1 deficiency activates the mechanistic target of rapamycin complex 1 (mTORC1) along with induction of PGC-1α and ER stress. Further, mTORC1 inhibition attenuates SCD1 deficiency-mediated induction of both PGC-1α and ER stress. Similar observations were seen by restoring endogenously synthesized oleate, but not palmitoleate, suggesting a clear mTORC1-mediated regulation of ER stress during SCD1 deficiency. Overall, our results suggest a model whereby maintaining adequate levels of hepatic oleate is required to suppress mTORC1-mediated ER stress. In addition, the activation of mTORC1 by SCD1 deficiency reveals an important function of fatty acids in regulating different cellular processes through mTORC1 signaling.


Asunto(s)
Carbohidratos de la Dieta/farmacología , Estrés del Retículo Endoplásmico , Hígado/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Transducción de Señal , Estearoil-CoA Desaturasa/deficiencia , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Estearoil-CoA Desaturasa/metabolismo
5.
Biochem Biophys Res Commun ; 508(1): 87-91, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30470572

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1), a lipogenic enzyme that adds a double bond at the delta 9 position of stearate (C18: 0) and palmitate (C16: 0), has been proven to be important in the development of obesity. Mice with skin-specific deficiency of SCD1 (SKO) display increased whole-body energy expenditure, which is protective against adiposity from a high-fat diet because it improves glucose clearance, insulin sensitivity, and hepatic steatosis. Of note, these mice also display elevated levels of the "pro-inflammatory" plasma interleukin-6 (IL-6). In whole skin of SKO mice, IL-6 mRNA levels are increased, and protein expression is evident in hair follicle cells and in keratinocytes. Recently, the well-known role of IL-6 in causing white adipose tissue lipolysis has been linked to indirectly activating the gluconeogenic enzyme pyruvate carboxylase 1 in the liver, thereby increasing hepatic glucose production. In this study, we suggest that skin-derived IL-6 leads to white adipose tissue lipolysis, which contributes to the lean phenotype of SKO mice without the incidence of meta-inflammation that is associated with IL-6 signaling.


Asunto(s)
Interleucina-6/metabolismo , Piel/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Tejido Adiposo Blanco/metabolismo , Adiposidad , Animales , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos/metabolismo , Gluconeogénesis , Folículo Piloso/citología , Folículo Piloso/metabolismo , Interleucina-6/genética , Queratinocitos/metabolismo , Lipólisis , Hígado/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Piel/citología , Estearoil-CoA Desaturasa/genética , Delgadez/genética , Delgadez/metabolismo , Distribución Tisular
6.
J Agric Food Chem ; 66(38): 10041-10052, 2018 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-30180552

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1) is a fatty acid desaturase catalyzing cis-double-bond formation in the Δ9 position to produce monounsaturated fatty acids essential for the synthesis of milk fat. Previous studies using RNAi methods have provided support for a role of SCD1 in goat mammary epithelial cells (GMEC); however, RNAi presents several limitations that might preclude a truthful understanding of the biological function of SCD1. To explore the function of SCD1 on fatty acid metabolism in GMEC, we used CRISPR-Cas9-mediated SCD1 knockout through non-homologous end-joining (NHEJ) and homology-directed repair (HDR) pathways in GMEC. We successfully introduced nucleotide deletions and mutations in the SCD1 gene locus through the NHEJ pathway and disrupted its second exon via insertion of an EGFP-PuroR segment using the HDR pathway. In clones derived from the latter, gene- and protein-expression data indicated that we obtained a monoallelic SCD1 knockout. A T7EN1-mediated assay revealed no off-targets in the surveyed sites. The contents of triacylglycerol and cholesterol and the desaturase index were significantly decreased as a consequence of SCD1 knockout. The deletion of SCD1 decreased the expression of other genes involved in de novo fatty acid synthesis, including SREBF1 and FASN, as well the fatty acid transporters FABP3 and FABP4. The downregulation of these genes partly explains the decrease of intracellular triacylglycerols. Our results indicate a successful SCD1 knockout in goat mammary cells using CRISPR-Cas9. The demonstration of the successful use of CRISPR-Cas9 in GMEC is an important step to producing transgenic goats to study mammary biology in vivo.


Asunto(s)
Animales Modificados Genéticamente/genética , Células Epiteliales/enzimología , Ácidos Grasos/metabolismo , Técnicas de Inactivación de Genes/métodos , Cabras/genética , Glándulas Mamarias Animales/enzimología , Estearoil-CoA Desaturasa/deficiencia , Animales , Animales Modificados Genéticamente/metabolismo , Sistemas CRISPR-Cas , Células Epiteliales/metabolismo , Femenino , Eliminación de Gen , Cabras/metabolismo , Lipogénesis , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Leche/química , Leche/metabolismo , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
7.
J Lipid Res ; 59(10): 1818-1840, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30139760

RESUMEN

After crossing floxed stearoyl-CoA desaturase-1 (Scd1fl/fl) mice with LDL receptor-null (ldlr-/-) mice, and then Villin Cre (VilCre) mice, enterocyte Scd1 expression in Scd1fl/fl/ldlr-/-/VilCre mice was reduced 70%. On Western diet (WD), Scd1fl/fl/ldlr-/- mice gained more weight than Scd1fl/fl/ldlr-/-/VilCre mice (P < 0.0023). On WD, jejunum levels of lysophosphatidylcholine (LysoPC) 18:1 and lysophosphatidic acid (LPA) 18:1 were significantly less in Scd1fl/fl/ldlr-/-/VilCre compared with Scd1fl/fl/ldlr-/- mice (P < 0.0004 and P < 0.026, respectively). On WD, Scd1fl/fl/ldlr-/-/VilCre mice compared with Scd1fl/fl/ldlr-/- mice had lower protein levels of lipopolysaccharide-binding protein (LBP), cluster of differentiation 14 (CD14), toll-like receptor 4 (TLR4), and myeloid differentiation factor-88 (MyD88) in enterocytes and plasma, and less dyslipidemia and systemic inflammation. Adding a concentrate of tomatoes transgenic for the apoA-I mimetic peptide 6F (Tg6F) to WD resulted in reduced enterocyte protein levels of LBP, CD14, TLR4, and MyD88 in Scd1fl/fl/ldlr-/- mice similar to that seen in Scd1fl/fl/ldlr-/-/VilCre mice. Adding LysoPC 18:1 to WD did not reverse the effects of enterocyte Scd1 knockdown. Adding LysoPC 18:1 (but not LysoPC 18:0) to chow induced jejunum Scd1 expression and increased dyslipidemia and plasma serum amyloid A and interleukin 6 levels in Scd1fl/fl/ldlr-/- mice, but not in Scd1fl/fl/ldlr-/-/VilCre mice. We conclude that enterocyte Scd1 is partially responsible for LysoPC 18:1- and WD-induced dyslipidemia and inflammation in ldlr-/- mice.


Asunto(s)
Enterocitos/enzimología , Eliminación de Gen , Receptores de LDL/deficiencia , Receptores de LDL/genética , Estearoil-CoA Desaturasa/metabolismo , Proteínas de Fase Aguda/metabolismo , Animales , Peso Corporal , Proteínas Portadoras/metabolismo , HDL-Colesterol/sangre , Dislipidemias/enzimología , Dislipidemias/genética , Dislipidemias/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Yeyuno/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Lisofosfatidilcolinas/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Estearoil-CoA Desaturasa/genética , Receptor Toll-Like 4/metabolismo
8.
Sci Rep ; 8(1): 3358, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29463801

RESUMEN

The stearoyl-CoA desaturase (SCD) family of enzymes catalyzes monounsaturated fatty acid synthesis by inserting a cis double bond at the Δ9 position of saturated fatty acids. Disruption of these enzymes has been reported to induce a severe dry skin phenotype. Since lipid abnormalities in the meibomian glands have been associated with dry eye, we analyzed selected eye tissues contributing to tear volume and composition in genetically SCD-1-deficient mice (SCD-1 KO), including the lacrimal glands and conjunctiva. Previous histopathological analysis had revealed atrophy and loss of meibomian glands; taken together with the increased goblet cell and MUC5AC expression in the conjunctiva reported here, these findings suggest that the tear volume and mucin levels secreted are enhanced in the absence of lipid secretion as a compensatory mechanism. The expression of lipid metabolism genes in lacrimal glands was decreased in SCD1 KO mice. Thus, these results provide new pathophysiological mechanisms to pursue with regard to meibomian gland dysfunction. In addition, lack of SCD-1 causes a compensatory increase in the tear volume and mucin levels associated with changes in expression of lipid metabolism genes. These results may be useful as a new concept for dry eye treatment strategies.


Asunto(s)
Enfermedades de los Párpados/patología , Glándulas Tarsales/patología , Mucinas/análisis , Estearoil-CoA Desaturasa/deficiencia , Lágrimas/química , Lágrimas/metabolismo , Animales , Conjuntiva/patología , Modelos Animales de Enfermedad , Enfermedades de los Párpados/genética , Perfilación de la Expresión Génica , Células Caliciformes/patología , Histocitoquímica , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Mucina 5AC/biosíntesis
9.
Biochem Biophys Res Commun ; 492(1): 82-88, 2017 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-28803985

RESUMEN

Body fat mass is closely associated to diseases related to obesity. MicroRNAs (miRNAs, miR) are important regulatory molecules that function as post-transcriptional gene regulators of adipocyte development. In the current study, we revealed that reduced expression of miR-199a-3p in adipose tissue resulting from high fat diet (HFD)-induced obesity in mice. Overexpression of miR-199a-3p promoted adipocyte proliferation by regulating the expression of regulating factors of the cell cycle. Furthermore, miR-199a-3p blunted lipid accumulation in 3T3-L1 adipocytes. This was accompanied by a marked decrease in the expression of adipocyte-specific genes involved in lipogenic transcription, fatty acid synthesis, and fatty acid transportation. Furthermore, the fatty acid oxidation process was enhanced. Luciferase activity assays confirmed that miR-199a-3p regulates adipocyte differentiation by directly targeting the 3'-untranslated region (3'-UTR) of stearoyl-CoA desaturase (SCD). Moreover, miR-199a-3p regulates fatty acid composition by decreasing the ratio of unsaturated fatty acids (UFAs) in adipocytes transfected with miR-199a-3p mimics. These results suggest that miR-199a-3p may promote adipocyte proliferation, while also repressing adipocyte differentiation by down-regulating SCD and changing fatty acid composition during adipogenesis.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Diferenciación Celular/genética , Ácidos Grasos/metabolismo , MicroARNs/genética , Estearoil-CoA Desaturasa/deficiencia , Estearoil-CoA Desaturasa/genética , Células 3T3-L1 , Animales , Dieta Alta en Grasa , Masculino , Ratones , Ratones Endogámicos , Ratones Obesos
10.
J Invest Dermatol ; 137(7): 1424-1433, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28259688

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1) is the dominant member of the SCD-isozyme family, regarded as a major regulator of lipid and energy metabolism in liver and adipose tissue. SCD1 deficiency impairs the desaturation of de novo-synthesized palmitoyl- and stearoyl-CoA to palmitoleoyl- and oleoyl-CoA. Scd1-/- mice develop metabolic waste syndrome and skin lesions: epidermal barrier disruption, alopecia, and degeneration of sebaceous glands. The unifying molecular link between the two divergent traits remains incompletely understood. Here we show the absence of palmitoleic acid (9Z-16:1) in the lipidome of the scd1-null mouse, which prohibits posttranslational O-palmitoleoylation of Wnt3a protein, essential for Wnt3a/ß-catenin signaling in stem cell lineage decision in development of the epidermal barrier, hair growth cycle, and sebaceous glands. Substitution of the disrupted epidermal lipid barrier by an inert hydrocarbon coat prevents excessive transepidermal water loss, normalizes thermogenesis and metabolic parameters, and surprisingly leads to the activation of hair bulge progenitor cells and reprograming of a regular hair growth cycle and development of a regular fur in scd1-/- mice. Progenitor sebocytes are not activated. Independent of age, application or removal of the artificial lipid barrier allows the reversible telogen-anagen reentry and exit of the hair growth cycle.


Asunto(s)
Metabolismo Energético , Folículo Piloso/crecimiento & desarrollo , Metabolismo de los Lípidos , Glándulas Sebáceas/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Proteína Wnt3A/metabolismo , Tejido Adiposo/metabolismo , Animales , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Folículo Piloso/metabolismo , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Noqueados , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Estearoil-CoA Desaturasa/biosíntesis , Estearoil-CoA Desaturasa/genética
11.
Biochim Biophys Acta ; 1861(12 Pt A): 2029-2037, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27751891

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1) has recently been shown to be a critical control point in the regulation of cardiac metabolism and function. Peroxisome proliferator-activated receptor α (PPARα) is an important regulator of myocardial fatty acid uptake and utilization. The present study used SCD1 and PPARα double knockout (SCD1-/-/PPARα-/-) mice to test the hypothesis that PPARα is involved in metabolic changes in the heart that are caused by SCD1 downregulation/inhibition. SCD1 deficiency decreased the intracellular content of free fatty acids, triglycerides, and ceramide in the heart of SCD1-/- and SCD1-/-/PPARα-/- mice. SCD1 ablation in PPARα-/- mice decreased diacylglycerol content in cardiomyocytes. These results indicate that the reduction of fat accumulation in the heart associated with SCD1 deficiency occurs independently of the PPARα pathway. To elucidate the mechanism of the observed changes, we treated HL-1 cardiomyocytes with the SCD1 inhibitor A939572 and/or PPARα inhibitor GW6471. SCD1 inhibition decreased the level of lipogenic proteins and increased lipolysis, reflected by a decrease in the content of adipose triglyceride lipase inhibitor G0S2 and a decrease in the ratio of phosphorylated hormone-sensitive lipase (HSL) at Ser565 to HSL (pHSL[Ser565]/HSL). PPARα inhibition alone did not affect the aforementioned protein levels. Finally, PPARα inhibition decreased the phosphorylation level of 5'-adenosine monophosphate-activated protein kinase, indicating lower mitochondrial fatty acid oxidation. In summary, SCD1 ablation/inhibition decreased cardiac lipid content independently of the action of PPARα by reducing lipogenesis and activating lipolysis. The present data suggest that SCD1 is an important component in maintaining proper cardiac lipid metabolism.


Asunto(s)
Corazón/fisiología , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Lipólisis/fisiología , PPAR gamma/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Adenosina Monofosfato/metabolismo , Animales , Línea Celular , Ceramidas/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Lipogénesis/fisiología , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Fosforilación/fisiología , Triglicéridos/metabolismo
12.
Biochim Biophys Acta ; 1861(11): 1662-1670, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27477676

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1) is a delta-9 fatty acid desaturase that catalyzes the synthesis of mono-unsaturated fatty acids (MUFA). SCD1 is a critical control point regulating hepatic lipid synthesis and ß-oxidation. Scd1 KO mice are resistant to the development of diet-induced non-alcoholic fatty liver disease (NAFLD). Using a chronic-binge protocol of ethanol-mediated liver injury, we aimed to determine if these KO mice are also resistant to the development of alcoholic fatty liver disease (AFLD). Mice fed a low-fat diet (especially low in MUFA) containing 5% ethanol for 10days, followed by a single ethanol (5g/kg) gavage, developed severe liver injury manifesting as hepatic steatosis. This was associated with an increase in de novo lipogenesis and inflammation. Using this model, we show that Scd1 KO mice are resistant to the development of AFLD. Scd1 KO mice do not show accumulation of hepatic triglycerides, activation of de novo lipogenesis nor elevation of cytokines or other pro-inflammatory markers. Incubating HepG2 cells with a SCD1 inhibitor induced a similar resistance to the effect of ethanol, confirming a role for SCD1 activity in mediating ethanol-induced hepatic injury. Taken together, our study shows that SCD1 is a key player in the development of AFLD and associated deleterious effects, and suggests SCD1 inhibition as a therapeutic option for the treatment of this hepatic disease.


Asunto(s)
Hígado/enzimología , Hígado/lesiones , Sustancias Protectoras/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Animales , Composición Corporal , Dieta , Etanol , Ácidos Grasos/análisis , Hígado Graso Alcohólico/complicaciones , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Conducta Alimentaria , Eliminación de Gen , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Inflamación/complicaciones , Inflamación/genética , Inflamación/patología , Lipogénesis/genética , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Oxidación-Reducción , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Estearoil-CoA Desaturasa/metabolismo
13.
Cell Metab ; 22(6): 983-96, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26525535

RESUMEN

The gut microbiota plays a key role in host metabolism. Toll-like receptor 5 (TLR5), a flagellin receptor, is required for gut microbiota homeostasis. Accordingly, TLR5-deficient (T5KO) mice are prone to develop microbiota-dependent metabolic syndrome. Here we observed that T5KO mice display elevated neutral lipids with a compositional increase of oleate [C18:1 (n9)] relative to wild-type littermates. Increased oleate contribution to hepatic lipids and liver SCD1 expression were both microbiota dependent. Analysis of short-chain fatty acids (SCFAs) and (13)C-acetate label incorporation revealed elevated SCFA in ceca and hepatic portal blood and increased liver de novo lipogenesis in T5KO mice. Dietary SCFAs further aggravated metabolic syndrome in T5KO mice. Deletion of hepatic SCD1 not only prevented hepatic neutral lipid oleate enrichment but also ameliorated metabolic syndrome in T5KO mice. Collectively, these results underscore the key role of the gut microbiota-liver axis in the pathogenesis of metabolic diseases.


Asunto(s)
Hígado/metabolismo , Síndrome Metabólico/patología , Estearoil-CoA Desaturasa/metabolismo , Receptor Toll-Like 5/genética , Animales , Peso Corporal , Restricción Calórica , Dieta Alta en Grasa , Ácidos Grasos Volátiles/sangre , Heces/química , Femenino , Resistencia a la Insulina , Intestinos/microbiología , Lipogénesis , Espectroscopía de Resonancia Magnética , Masculino , Síndrome Metabólico/metabolismo , Ratones , Ratones Noqueados , Microbiota , Ácido Oléico/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Estearoil-CoA Desaturasa/genética , Receptor Toll-Like 5/deficiencia , Regulación hacia Arriba
14.
PLoS One ; 8(6): e64525, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785402

RESUMEN

The progressive deterioration of the neuromuscular axis is typically observed in degenerative conditions of the lower motor neurons, such as amyotrophic lateral sclerosis (ALS). Neurodegeneration in this disease is associated with systemic metabolic perturbations, including hypermetabolism and dyslipidemia. Our previous gene profiling studies on ALS muscle revealed down-regulation of delta-9 desaturase, or SCD1, which is the rate-limiting enzyme in the synthesis of monounsaturated fatty acids. Interestingly, knocking out SCD1 gene is known to induce hypermetabolism and stimulate fatty acid beta-oxidation. Here we investigated whether SCD1 deficiency can affect muscle function and its restoration in response to injury. The genetic ablation of SCD1 was not detrimental per se to muscle function. On the contrary, muscles in SCD1 knockout mice shifted toward a more oxidative metabolism, and enhanced the expression of synaptic genes. Repressing SCD1 expression or reducing SCD-dependent enzymatic activity accelerated the recovery of muscle function after inducing sciatic nerve crush. Overall, these findings provide evidence for a new role of SCD1 in modulating the restorative potential of skeletal muscles.


Asunto(s)
Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Estearoil-CoA Desaturasa/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/rehabilitación , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Oxidación-Reducción , Fenotipo , Recuperación de la Función , Estearoil-CoA Desaturasa/deficiencia , Estearoil-CoA Desaturasa/genética
15.
Postepy Biochem ; 58(2): 166-74, 2012.
Artículo en Polaco | MEDLINE | ID: mdl-23214140

RESUMEN

Stearoyl-CoA desaturase (SCD) is a regulatory enzyme in lipogenesis, catalyzing the rate-limiting step in the overall de novo synthesis of monounsaturated fatty acids, mainly oleate and palmitoleate from staroyl- and palmitoyl-CoA, respectively. These products are the most abundant monounsaturated fatty acids in various kinds of lipids, including phospholipids, triglicerides, cholesteryl esters, wax esters, and diacyloglycerols. SCD1 deficiency results in reduced body adiposity, increased insulin sensitivity, and resistance to diet-induced obesity. Recent studies have shown that SCD1 plays also a significant role, directly or indirectly, in the regulation of diverse metabolic processes, including lipogenesis, fatty acid oxidation, insulin signaling, thermogenesis, carcinogenesis, and inflammation. This review summarize the recent advances concerning the important role of SCD in the maintenance of metabolic homeostasis.


Asunto(s)
Homeostasis/fisiología , Estearoil-CoA Desaturasa/metabolismo , Animales , Parálisis Cerebral/metabolismo , Ácidos Grasos/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Humanos , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Lipogénesis/fisiología , Miocardio/metabolismo , Obesidad/metabolismo , Ácido Oléico/biosíntesis , Estearoil-CoA Desaturasa/química , Estearoil-CoA Desaturasa/deficiencia , Termogénesis/fisiología
16.
Comp Med ; 62(4): 257-63, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23043777

RESUMEN

Ulcerative dermatitis (UD) is a common cause of morbidity and euthanasia in mice with a C57BL/6 (B6) background. The purposes of the current study were to determine whether UD lesions could be reliably produced in B6 mice lacking stearoyl-CoA desaturase 1 (SCD1(-/-) mice), to ascertain whether the UD lesions in SCD1(-/-) mice were similar to those found in other B6 mice, and to characterize the cell invasion phenotype of Staphlococcus xylosus cultured from the lesions. S. xylosus isolates from the environment and human skin were used as controls. SCD1(-/-) (n = 8 per group) and nontransgenic B6 control mice (n = 22 mice pooled from 3 groups that received different concentrations of conjugated linoleic acid) were fed standard rodent chow or a semipurified diet (NIH AIN76A) for 4 wk. Samples from other B6 mice with UD (field cases; n = 7) also were submitted for histology and culture. All of the SCD1(-/-) mice developed UD lesions by 4 wk on NIH AIN76A. None of SCD1(-/-) fed standard rodent chow and none of the wildtype B6 mice fed NIH AIN76A developed UD. Supplementation with conjugated linoleic acid did not affect ulcerogenesis. UD lesions in SCD1(-/-) mice and field cases were grossly and histologically similar. S. xylosus was isolated from SCD1(-/-) mice with UD (71%) and field cases of UD (43%). These isolates were the most cell-invasive, followed by the environmental isolate, and finally the human skin isolate. Our results provide a basis for further pathologic and clinical study of UD.


Asunto(s)
Dermatitis/veterinaria , Ratones Endogámicos C57BL , Enfermedades de los Roedores/microbiología , Enfermedades de los Roedores/patología , Staphylococcus/fisiología , Estearoil-CoA Desaturasa/deficiencia , Alimentación Animal , Animales , Dermatitis/enzimología , Dermatitis/microbiología , Dermatitis/patología , Femenino , Humanos , Ácido Linoleico , Ratones , Ratones Noqueados , Enfermedades de los Roedores/enzimología , Estadísticas no Paramétricas , Estearoil-CoA Desaturasa/genética
17.
J Lipid Res ; 53(8): 1646-53, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22669918

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1) catalyzes the synthesis of monounsaturated fatty acids (MUFA) from saturated FA. Mice with whole-body or skin-specific deletion of SCD1 are resistant to obesity. Here, we show that mice lacking SCD1 in adipose and/or liver are not protected from either genetic- (agouti; A(y)/a) or diet-induced obesity (DIO) despite a robust reduction in SCD1 MUFA products in both subcutaneous and epididymal white adipose tissue. Adipose SCD1 deletion had no effect on glucose or insulin tolerance or on hepatic triglyceride (TG) accumulation. Interestingly, lack of SCD1 from liver lowered the MUFA levels of adipose tissue and vice versa, as reflected by the changes in FA composition. Simultaneous deletion of SCD1 from liver and adipose resulted in a synergistic lowering of tissue MUFA levels, especially in the A(y)/a model in which glucose tolerance was also improved. Lastly, we found that liver and plasma TG show nearly identical genotype-dependent differences in FA composition, indicating that FA composition of plasma TG is predictive for hepatic SCD1 activity and TG FA composition. The current study suggests that SCD1 deletion from adipose and/or liver is insufficient to elicit protection from obesity, but it supports the existence of extensive lipid cross-talk between liver and adipose tissue.


Asunto(s)
Tejido Adiposo/metabolismo , Eliminación de Gen , Hígado/metabolismo , Obesidad/enzimología , Obesidad/genética , Estearoil-CoA Desaturasa/deficiencia , Estearoil-CoA Desaturasa/genética , Animales , Ácidos Grasos Monoinsaturados/análisis , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Obesidad/sangre , Triglicéridos/sangre , Triglicéridos/química
18.
Mol Cancer Res ; 9(11): 1551-61, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21954435

RESUMEN

Emerging literature suggests that metabolic pathways play an important role in the maintenance and progression of human cancers. In particular, recent studies have implicated lipid biosynthesis and desaturation as a requirement for tumor cell survival. In the studies reported here, we aimed to understand whether tumor cells require the activity of either human isoform of stearoyl-CoA-desaturase (SCD1 or SCD5) for survival. Inhibition of SCD1 by siRNA or a small molecule antagonist results in strong induction of apoptosis and growth inhibition, when tumor cells are cultured in reduced (2%) serum conditions, but has little impact on cells cultured in 10% serum. Depletion of SCD5 had minimal effects on cell growth or apoptosis. Consistent with the observed dependence on SCD1, but not SCD5, levels of SCD1 protein increased in response to decreasing serum levels. Both induction of SCD1 protein and sensitivity to growth inhibition by SCD1 inhibition could be reversed by supplementing growth media with unsaturated fatty acids, the product of the enzymatic reaction catalyzed by SCD1. Transcription profiling of cells treated with an SCD inhibitor revealed strong induction of markers of endoplasmic reticulum stress. Underscoring its importance in cancer, SCD1 protein was found to be highly expressed in a large percentage of human cancer specimens. SCD inhibition resulted in tumor growth delay in a human gastric cancer xenograft model. Altogether, these results suggest that desaturated fatty acids are required for tumor cell survival and that SCD may represent a viable target for the development of novel agents for cancer therapy.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/patología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Estearoil-CoA Desaturasa/biosíntesis , Estearoil-CoA Desaturasa/deficiencia , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , Transfección
19.
PLoS One ; 6(5): e19734, 2011 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-21573029

RESUMEN

We previously reported that mice with skin-specific deletion of stearoyl-CoA desaturase-1 (Scd1) recapitulated the skin phenotype and hypermetabolism observed in mice with a whole-body deletion of Scd1. In this study, we first performed a diet-induced obesity experiment at thermoneutral temperature (33°C) and found that skin-specific Scd1 knockout (SKO) mice still remain resistant to obesity. To elucidate the metabolic changes in the skin that contribute to the obesity resistance and skin phenotype, we performed microarray analysis of skin gene expression in male SKO and control mice fed a standard rodent diet. We identified an extraordinary number of differentially expressed genes that support the previously documented histological observations of sebaceous gland hypoplasia, inflammation and epidermal hyperplasia in SKO mice. Additionally, transcript levels were reduced in skin of SKO mice for genes involved in fatty acid synthesis, elongation and desaturation, which may be attributed to decreased abundance of key transcription factors including SREBP1c, ChREBP and LXRα. Conversely, genes involved in cholesterol synthesis were increased, suggesting an imbalance between skin fatty acid and cholesterol synthesis. Unexpectedly, we observed a robust elevation in skin retinol, retinoic acid and retinoic acid-induced genes in SKO mice. Furthermore, SEB-1 sebocytes treated with retinol and SCD inhibitor also display an elevation in retinoic acid-induced genes. These results highlight the importance of monounsaturated fatty acid synthesis for maintaining retinol homeostasis and point to disturbed retinol metabolism as a novel contributor to the Scd1 deficiency-induced skin phenotype.


Asunto(s)
Piel/metabolismo , Estearoil-CoA Desaturasa/deficiencia , Vitamina A/metabolismo , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animales , Epidermis/efectos de los fármacos , Epidermis/patología , Ácidos Grasos/biosíntesis , Perfilación de la Expresión Génica , Cabello/efectos de los fármacos , Cabello/metabolismo , Cabello/patología , Hiperplasia , Inflamación/complicaciones , Inflamación/genética , Inflamación/patología , Lipocalina 2 , Lipocalinas/genética , Lipocalinas/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , PPAR delta/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Glándulas Sebáceas/anomalías , Glándulas Sebáceas/metabolismo , Glándulas Sebáceas/patología , Estearoil-CoA Desaturasa/metabolismo , Esteroles/metabolismo , Temperatura , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Tretinoina/farmacología
20.
Adv Nutr ; 2(1): 15-22, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22211186

RESUMEN

Stearoyl CoA desaturase 1 (SCD1) catalyzes the rate-limiting step in the production of MUFA that are major components of tissue lipids. Alteration in SCD1 expression changes the fatty acid profile of these lipids and produces diverse effects on cellular function. High SCD1 expression is correlated with metabolic diseases such as obesity and insulin resistance, whereas low levels are protective against these metabolic disturbances. However, SCD1 is also involved in the regulation of inflammation and stress in distinct cell types, including ß-cells, adipocytes, macrophages, endothelial cells, and myocytes. Furthermore, complete loss of SCD1 expression has been implicated in liver dysfunction and several inflammatory diseases such as dermatitis, atherosclerosis, and intestinal colitis. Thus, normal cellular function requires the expression of SCD1 to be tightly controlled. This review summarizes the current understanding of the role of SCD1 in modulating inflammation and stress.


Asunto(s)
Inflamación , Estearoil-CoA Desaturasa/fisiología , Estrés Fisiológico/fisiología , Adipocitos , Animales , Aterosclerosis , Fenómenos Fisiológicos Celulares , Células Endoteliales , Humanos , Resistencia a la Insulina , Células Secretoras de Insulina , Macrófagos , Ratones , Ratones Noqueados , Células Musculares , Obesidad , Estearoil-CoA Desaturasa/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...