Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 905
Filtrar
1.
Stem Cell Res Ther ; 14(1): 70, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37024989

RESUMEN

BACKGROUND: Adipose tissue-derived stromal vascular fraction (SVF) harbors multipotent cells with potential therapeutic relevance. We developed a method to form adipose spheroids (AS) from the SVF with complex organoid structure and enhanced leptin secretion upon insulin stimulation. METHODS: SVF was generated from the interscapular brown adipose tissue of newborn mice. Immunophenotype and stemness of cultured SVF were determined by flow cytometry and in vitro differentiation, respectively. Spheroids were generated in hanging drops and non-adherent plates and compared by morphometric methods. The adipogenic potential was compared between preadipocyte monolayers and spheroids. Extracellular leptin was quantified by immunoassay. Lipolysis was stimulated with isoprenaline and quantified by colorimetric methods. AS viability and ultrastructure were determined by confocal and transmission electron microscopy analyses. RESULTS: Cultured SVF contained Sca1 + CD29 + CD44 + CD11b- CD45- CD90- cells with adipogenic and chondrogenic but no osteogenic potential. Culture on non-adherent plates yielded the highest quantity and biggest size of spheroids. Differentiation of AS for 15 days in a culture medium supplemented with insulin and rosiglitazone resulted in greater Pparg, Plin1, and Lep expression compared to differentiated adipocytes monolayers. AS were viable and maintained leptin secretion even in the absence of adipogenic stimulation. Glycerol release after isoprenaline stimulation was higher in AS compared to adipocytes in monolayers. AS were composed of outer layers of unilocular mature adipocytes and an inner structure composed of preadipocytes, immature adipocytes and an abundant loose extracellular matrix. CONCLUSION: Newborn mice adipose SVF can be efficiently differentiated into leptin-secreting AS. Prolonged stimulation with insulin and rosiglitazone allows the formation of structurally complex adipose organoids able to respond to adrenergic lipolytic stimulation.


Asunto(s)
Adipocitos , Tejido Adiposo Pardo , Diferenciación Celular , Leptina , Leptina/metabolismo , Organoides , Insulina/farmacología , Animales , Ratones , Tejido Adiposo Pardo/citología , Rosiglitazona/farmacología , Células Cultivadas , Animales Recién Nacidos , Inmunofenotipificación , Osteogénesis , Condrogénesis , Adipocitos/ultraestructura , Lipólisis , Cultivo Primario de Células
2.
Nature ; 613(7942): 160-168, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36477540

RESUMEN

Multilocular adipocytes are a hallmark of thermogenic adipose tissue1,2, but the factors that enforce this cellular phenotype are largely unknown. Here, we show that an adipocyte-selective product of the Clstn3 locus (CLSTN3ß) present in only placental mammals facilitates the efficient use of stored triglyceride by limiting lipid droplet (LD) expansion. CLSTN3ß is an integral endoplasmic reticulum (ER) membrane protein that localizes to ER-LD contact sites through a conserved hairpin-like domain. Mice lacking CLSTN3ß have abnormal LD morphology and altered substrate use in brown adipose tissue, and are more susceptible to cold-induced hypothermia despite having no defect in adrenergic signalling. Conversely, forced expression of CLSTN3ß is sufficient to enforce a multilocular LD phenotype in cultured cells and adipose tissue. CLSTN3ß associates with cell death-inducing DFFA-like effector proteins and impairs their ability to transfer lipid between LDs, thereby restricting LD fusion and expansion. Functionally, increased LD surface area in CLSTN3ß-expressing adipocytes promotes engagement of the lipolytic machinery and facilitates fatty acid oxidation. In human fat, CLSTN3B is a selective marker of multilocular adipocytes. These findings define a molecular mechanism that regulates LD form and function to facilitate lipid utilization in thermogenic adipocytes.


Asunto(s)
Adipocitos , Proteínas de Unión al Calcio , Metabolismo de los Lípidos , Proteínas de la Membrana , Animales , Femenino , Humanos , Ratones , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Placenta , Triglicéridos/metabolismo , Retículo Endoplásmico/metabolismo , Gotas Lipídicas/metabolismo , Ácidos Grasos/metabolismo , Hipotermia/metabolismo , Termogénesis
3.
Proc Natl Acad Sci U S A ; 119(40): e2203307119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161914

RESUMEN

Brown adipose tissue (BAT) is a highly specialized adipose tissue in its immobile location and size during the entire adulthood. In response to cold exposure and other ß3-adrenoreceptor stimuli, BAT commits energy consumption by nonshivering thermogenesis (NST). However, the molecular machinery in controlling the BAT mass in adults is unknown. Here, we show our surprising findings that the BAT mass and functions can be manipulated in adult animals by controlling BAT adipocyte differentiation in vivo. Platelet-derived growth factor receptor α (PDGFα) expressed in BAT progenitor cells served a signaling function to avert adipose progenitor differentiation. Genetic and pharmacological loss-of-function of PDGFRα eliminated the differentiation barrier and permitted progenitor cell differentiation to mature and functional BAT adipocytes. Consequently, an enlarged BAT mass (megaBAT) was created by PDGFRα inhibition owing to increases of brown adipocyte numbers. Under cold exposure, a microRNA-485 (miR-485) was identified as a master suppressor of the PDGFRα signaling, and delivery of miR-485 also produced megaBAT in adult animals. Noticeably, megaBAT markedly improved global metabolism, insulin sensitivity, high-fat-diet (HFD)-induced obesity, and diabetes by enhancing NST. Together, our findings demonstrate that the adult BAT mass can be increased by blocking the previously unprecedented inhibitory signaling for BAT progenitor cell differentiation. Thus, blocking the PDGFRα for the generation of megaBAT provides an attractive strategy for treating obesity and type 2 diabetes mellitus (T2DM).


Asunto(s)
Adipocitos Marrones , Adipocitos , Adipogénesis , Tejido Adiposo Pardo , MicroARNs , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Adipocitos/citología , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Animales , Diabetes Mellitus Tipo 2/terapia , Metabolismo Energético , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/terapia , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Termogénesis/genética
4.
Mol Med ; 28(1): 6, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35062859

RESUMEN

BACKGROUND: Activation of brown adipose tissue (BAT) increases energy expenditure, which makes it an attractive therapeutic strategy for obesity. LncRNAs play an important role in adipocyte differentiation and regulation. Here we assessed the effect of lncRNA XIST on brown preadipocytes differentiation and metabolic regulation. METHODS: XIST expression levels were detected in human perirenal (peri-N) and subcutaneous adipose tissues (sub-Q), brown preadipocytes and 3T3-L1 preadipocytes. XIST overexpression and knockdown experiments were performed in brown preadipocytes. XIST overexpression mouse model was established by plasmid injection through tail vein. RESULTS: In human adipose tissues, XIST expression was significantly higher in female than in male individuals. In vitro, XIST expression was significantly up-regulated during brown adipocyte differentiation. XIST knockdown inhibited differentiation of brown preadipocytes, while overexpression of XIST promotes brown preadipocytes to fully differentiation. RNA Binding Protein Immunoprecipitation (RIP) experiment revealed that XIST could directly bind to C/EBPα. In vivo, XIST overexpression prevents high-fat diet induced obesity and improves metabolic dysorder in male mice. CONCLUSION: Our results suggest that XIST combats obesity through BAT activation at least partly by combination with transcription factor C/EBPα.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Obesidad/etiología , Obesidad/metabolismo , ARN Largo no Codificante/genética , Células 3T3-L1 , Animales , Biomarcadores , Diferenciación Celular , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Inmunofenotipificación , Masculino , Ratones , Obesidad/patología , Interferencia de ARN
5.
Sci Rep ; 11(1): 23290, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857816

RESUMEN

The Fuegians, ancient inhabitants of Tierra del Fuego, are an exemplary case of a cold-adapted population, since they were capable of living in extreme climatic conditions without any adequate clothing. However, the mechanisms of their extraordinary resistance to cold remain enigmatic. Brown adipose tissue (BAT) plays a crucial role in this kind of adaptation, besides having a protective role on the detrimental effect of low temperatures on bone structure. Skeletal remains of 12 adult Fuegians, collected in the second half of XIX century, were analyzed for bone mineral density and structure. We show that, despite the unfavorable climate, bone mineral density of Fuegians was close to that seen in modern humans living in temperate zones. Furthermore, we report significant differences between Fuegians and other cold-adapted populations in the frequency of the Homeobox protein Hox-C4 (HOXC4) rs190771160 variant, a gene involved in BAT differentiation, whose identified variant is predicted to upregulate HOXC4 expression. Greater BAT accumulation might therefore explain the Fuegians extreme cold-resistance and the protection against major cold-related damage. These results increase our understanding of how ecological challenges have been important drivers of human-environment interactions during Humankind history.


Asunto(s)
Aclimatación/genética , Adaptación Fisiológica/genética , Densidad Ósea/genética , Frío , Ecología , Interacción Gen-Ambiente , Genómica , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/fisiología , Restos Mortales , Diferenciación Celular/genética , Chile , Expresión Génica/genética , Variación Genética , Proteínas de Homeodominio/genética , Humanos , Regulación hacia Arriba/genética
6.
Nat Commun ; 12(1): 6845, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34824246

RESUMEN

Maternal obesity (MO) predisposes offspring to obesity and metabolic disorders but little is known about the contribution of offspring brown adipose tissue (BAT). We find that MO impairs fetal BAT development, which persistently suppresses BAT thermogenesis and primes female offspring to metabolic dysfunction. In fetal BAT, MO enhances expression of Dio3, which encodes deiodinase 3 (D3) to catabolize triiodothyronine (T3), while a maternally imprinted long noncoding RNA, Dio3 antisense RNA (Dio3os), is inhibited, leading to intracellular T3 deficiency and suppression of BAT development. Gain and loss of function shows Dio3os reduces D3 content and enhances BAT thermogenesis, rendering female offspring resistant to high fat diet-induced obesity. Attributing to Dio3os inactivation, its promoter has higher DNA methylation in obese dam oocytes which persists in fetal and adult BAT, uncovering an oocyte origin of intergenerational obesity. Overall, our data uncover key features of Dio3os activation in BAT to prevent intergenerational obesity and metabolic dysfunctions.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Obesidad/genética , ARN Largo no Codificante/genética , Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Adipogénesis , Tejido Adiposo Pardo/citología , Animales , Diferenciación Celular , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dieta Occidental/efectos adversos , Metabolismo Energético , Femenino , Impresión Genómica , Yoduro Peroxidasa/metabolismo , Ratones , Obesidad/etiología , Obesidad/metabolismo , Obesidad Materna/genética , Obesidad Materna/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Embarazo , ARN Largo no Codificante/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Triyodotironina/metabolismo
7.
Food Funct ; 12(19): 9151-9164, 2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34606532

RESUMEN

Metabolic syndrome caused obesity has long been recognized as a risk of health. Celery and celery extracts have various medicinal properties, such as anti-diabetes and anti-inflammatory properties and blood glucose and serum lipid reduction. However, the effect of probiotic fermentation on celery juice and the association between fermented celery juice (FCJ) and obesity were unclear. This study aimed to evaluate the beneficial effects of FCJ on high-fat diet (HFD) induced obesity and related metabolic syndromes. C57BL/6 mice were randomly divided into six groups (n = 15 per group) fed either a normal diet (ND) or HFD with or without CJ/FCJ (10 g kg-1 day-1) by oral gavage for 12 weeks. Here we demonstrated that the probiotic fermentation of celery juice (CJ) could enhance the active ingredients in celery, such as total polyphenols, flavonoids, vitamin C and SOD. Compared to the slight improvement induced by CJ ingestion, FCJ intake significantly inhibited body weight gain, prevented dyslipidemia and hyperglycemia, and suppressed visceral fat accumulation. Furthermore, 16S rRNA sequencing analysis revealed that FCJ intake altered the composition of gut microbiota, increasing the ratio of Firmicutes/Bacteroidetes and the relative abundance of beneficial bacteria (Lactobacillus, Ruminococcaceae_UCG-014, Faecalibaculum and Blautia), and decreasing the relative abundance of harmful bacteria (Alloprevotella and Helicobacter). These findings suggest that FCJ can prevent HFD-induced obesity and become a novel gut microbiota modulator to prevent HFD-induced gut dysbiosis and obesity-related metabolic disorders.


Asunto(s)
Apium , Dieta Alta en Grasa , Suplementos Dietéticos , Bebidas Fermentadas , Microbioma Gastrointestinal , Obesidad/prevención & control , Adipocitos/citología , Adipocitos/fisiología , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Diabetes Mellitus Tipo 2/prevención & control , Dislipidemias/prevención & control , Bebidas Fermentadas/análisis , Hiperglucemia/prevención & control , Grasa Intraabdominal/anatomía & histología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/prevención & control
8.
FASEB J ; 35(11): e21965, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34669999

RESUMEN

Obesity and metabolic disorders caused by energy surplus pose an increasing concern within the global population. Brown adipose tissue (BAT) dissipates energy through mitochondrial non-shivering thermogenesis, thus representing a powerful agent against obesity. Here we explore the novel role of a mitochondrial outer membrane protein, LETM1-domain containing 1 (LETMD1), in BAT. We generated a knockout (Letmd1KO ) mouse model and analyzed BAT morphology, function and gene expression under various physiological conditions. While the Letmd1KO mice are born normally and have normal morphology and body weight, they lose multilocular brown adipocytes completely and have diminished mitochondrial abundance, DNA copy number, cristae structure, and thermogenic gene expression in the intrascapular BAT, associated with elevated reactive oxidative stress. In consequence, the Letmd1KO mice fail to maintain body temperature in response to acute cold exposure without food and become hypothermic within 4 h. Although the cold-exposed Letmd1KO mice can maintain body temperature in the presence of food, they cannot upregulate expression of uncoupling protein 1 (UCP1) and convert white to beige adipocytes, nor can they respond to adrenergic stimulation. These results demonstrate that LETMD1 is essential for mitochondrial structure and function, and thermogenesis of brown adipocytes.


Asunto(s)
Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Mitocondrias/metabolismo , Proteínas Oncogénicas/fisiología , Receptores de Superficie Celular/fisiología , Termogénesis , Adipocitos Marrones/citología , Tejido Adiposo Pardo/citología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo
9.
FASEB J ; 35(11): e21966, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34624148

RESUMEN

Adipose tissue is central to the regulation of energy balance. While white adipose tissue (WAT) is responsible for triglyceride storage, brown adipose tissue specializes in energy expenditure. Deterioration of brown adipocyte function contributes to the development of metabolic complications like obesity and diabetes. These disorders are also leading symptoms of the Bardet-Biedl syndrome (BBS), a hereditary disorder in humans which is caused by dysfunctions of the primary cilium and which therefore belongs to the group of ciliopathies. The cilium is a hair-like organelle involved in cellular signal transduction. The BBSome, a supercomplex of several Bbs gene products, localizes to the basal body of cilia and is thought to be involved in protein sorting to and from the ciliary membrane. The effects of a functional BBSome on energy metabolism and lipid mobilization in brown and white adipocytes were tested in whole-body Bbs4 knockout mice that were subjected to metabolic challenges. Chronic cold exposure reveals cold-intolerance of knockout mice but also ameliorates the markers of metabolic pathology detected in knockouts prior to cold. Hepatic triglyceride content is markedly reduced in knockout mice while circulating lipids are elevated, altogether suggesting that defective lipid metabolism in adipose tissue creates increased demand for systemic lipid mobilization to meet energetic demands of reduced body temperatures. These findings taken together suggest that Bbs4 is essential for the regulation of adipose tissue lipid metabolism, representing a potential target to treat metabolic disorders.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo de los Lípidos , Proteínas Asociadas a Microtúbulos/fisiología , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Metabolismo Energético , Masculino , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Termogénesis
10.
Bull Exp Biol Med ; 171(6): 722-726, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34705171

RESUMEN

We studied the effect of bacterial pathogen-associated molecular patterns and myokines on the secretion of adipokines by mesenchymal stem cells (MSC) and products of their adipogenic differentiation. The secretion of adiponectin, adipsin, leptin, and insulin by adipogenically differentiated cell cultures was quantitatively determined using multiplex ELISA. MSC obtained from the stromal vascular fraction of human subcutaneous adipose tissue were shown to secrete a known adipokine adipsin. The ability of white adipocytes to secrete significant amounts of insulin (in vitro) has been shown for the first time. Control cultures of white adipocytes secreted much higher levels of adiponectin, leptin, and insulin when compared to other adipocytes cultures. On the other hand, beige and brown adipocyte cultures secreted more adipsin than white adipocyte cultures. The influence of myokine ß-aminoisobutyric acid on the secretion of adipsin in MSC, white, beige, and brown adipocytes was also studied.


Asunto(s)
Adipocitos Beige/efectos de los fármacos , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Adipoquinas/farmacología , Ácidos Aminoisobutíricos/farmacología , Flagelina/farmacología , Lipopolisacáridos/farmacología , Adipocitos Beige/citología , Adipocitos Beige/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/cirugía , Diferenciación Celular/efectos de los fármacos , Factor D del Complemento/genética , Factor D del Complemento/metabolismo , Regulación de la Expresión Génica , Humanos , Insulina/genética , Insulina/metabolismo , Leptina/genética , Leptina/metabolismo , Lipectomía/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Especificidad de Órganos , Cultivo Primario de Células
11.
PLoS One ; 16(9): e0249438, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34473703

RESUMEN

Muscle derived stem cells (MDSCs) and myoblast play an important role in myotube regeneration when muscle tissue is injured. However, these cells can be induced to differentiate into adipocytes once exposed to PPARγ activator like EPA and DHA that are highly suggested during pregnancy. The objective of this study aims at determining the identity of trans-differentiated cells by exploring the effect of EPA and DHA on C2C12 undergoing differentiation into brown and white adipocytes. DHA but not EPA committed C2C12 cells reprograming into white like adipocyte phenotype. Also, DHA promoted the expression of lipolysis regulating genes but had no effect on genes regulating ß-oxidation referring to its implication in lipid re-esterification. Furthermore, DHA impaired C2C12 cells differentiation into brown adipocytes through reducing the thermogenic capacity and mitochondrial biogenesis of derived cells independent of UCP1. Accordingly, DHA treated groups showed an increased accumulation of lipid droplets and suppressed mitochondrial maximal respiration and spare respiratory capacity. EPA, on the other hand, reduced myogenesis regulating genes, but no significant differences were observed in the expression of adipogenesis key genes. Likewise, EPA suppressed the expression of WAT signature genes indicating that EPA and DHA have an independent role on white adipogensis. Unlike DHA treatment, EPA supplementation had no effect on the differential of C2C12 cells into brown adipocytes. In conclusion, DHA is a potent adipogenic and lipogenic factor that can change the metabolic profile of muscle cells by increasing myocellular fat.


Asunto(s)
Adipocitos Blancos/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/citología , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/efectos de los fármacos , Animales , Línea Celular , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , ADN Mitocondrial , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lipólisis/efectos de los fármacos , Ratones , Mioblastos/citología , Mioblastos/efectos de los fármacos
12.
Nat Commun ; 12(1): 5255, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34489438

RESUMEN

Monocytes are part of the mononuclear phagocytic system. Monocytes play a central role during inflammatory conditions and a better understanding of their dynamics might open therapeutic opportunities. In the present study, we focused on the characterization and impact of monocytes on brown adipose tissue (BAT) functions during tissue remodeling. Single-cell RNA sequencing analysis of BAT immune cells uncovered a large diversity in monocyte and macrophage populations. Fate-mapping experiments demonstrated that the BAT macrophage pool requires constant replenishment from monocytes. Using a genetic model of BAT expansion, we found that brown fat monocyte numbers were selectively increased in this scenario. This observation was confirmed using a CCR2-binding radiotracer and positron emission tomography. Importantly, in line with their tissue recruitment, blood monocyte counts were decreased while bone marrow hematopoiesis was not affected. Monocyte depletion prevented brown adipose tissue expansion and altered its architecture. Podoplanin engagement is strictly required for BAT expansion. Together, these data redefine the diversity of immune cells in the BAT and emphasize the role of monocyte recruitment for tissue remodeling.


Asunto(s)
Tejido Adiposo Pardo/citología , Monocitos/fisiología , Adiponectina/genética , Tejido Adiposo Pardo/fisiología , Animales , Diferenciación Celular/genética , Recuento de Leucocitos , Macrófagos/citología , Macrófagos/fisiología , Glicoproteínas de Membrana/metabolismo , Ratones Transgénicos , Monocitos/citología , Tomografía de Emisión de Positrones , Receptores CCR2/genética , Receptores CCR2/metabolismo
13.
Int J Mol Sci ; 22(17)2021 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-34502211

RESUMEN

Obesity is a condition characterized by uncontrolled expansion of adipose tissue mass resulting in pathological weight gain. Histone deacetylases (HDACs) have emerged as crucial players in epigenetic regulation of adipocyte metabolism. Previously, we demonstrated that selective inhibition of class I HDACs improves white adipocyte functionality and promotes the browning phenotype of murine mesenchymal stem cells (MSCs) C3H/10T1/2 differentiated to adipocytes. These effects were also observed in db/db and diet induced obesity mouse models and in mice with adipose-selective inactivation of HDAC3, a member of class I HDACs. The molecular basis of class I HDACs action in adipose tissue is not deeply characterized and it is not known whether the effects of their inhibition are exerted on adipocyte precursors or mature adipocytes. Therefore, the aim of the present work was to explore the molecular mechanism of class I HDAC action in adipocytes by evaluating the effects of HDAC3-specific silencing at different stages of differentiation. HDAC3 was silenced in C3H/10T1/2 MSCs at different stages of differentiation to adipocytes. shRNA targeting HDAC3 was used to generate the knock-down model. Proper HDAC3 silencing was assessed by measuring both mRNA and protein levels of mouse HDAC3 via qPCR and western blot, respectively. Mitochondrial DNA content and gene expression were quantified via qPCR. HDAC3 silencing at the beginning of differentiation enhanced adipocyte functionality by amplifying the expression of genes regulating differentiation, oxidative metabolism, browning and mitochondrial activity, starting from 72 h after induction of differentiation and silencing. Insulin signaling was enhanced as demonstrated by increased AKT phosphorylation following HDAC3 silencing. Mitochondrial content/density did not change, while the increased expression of the transcriptional co-activator Ppargc1b suggests the observed phenotype was related to enhanced mitochondrial activity, which was confirmed by increased maximal respiration and proton leak linked to reduced coupling efficiency. Moreover, the expression of pro-inflammatory markers increased with HDAC3 early silencing. To the contrary, no differences in terms of gene expression were found when HDAC3 silencing occurred in terminally differentiated adipocyte. Our data demonstrated that early epigenetic events mediated by class I HDAC inhibition/silencing are crucial to commit adipocyte precursors towards the above-mentioned metabolic phenotype. Moreover, our data suggest that these effects are exerted on adipocyte precursors.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Diferenciación Celular , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Mitocondrias/metabolismo , Fenotipo , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Histona Desacetilasas/genética , Ratones , Ratones Endogámicos C3H
14.
Mol Nutr Food Res ; 65(19): e2100315, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34363644

RESUMEN

INTRODUCTION: Obesity causes many life-threatening diseases. It is important to develop effective approaches for obesity treatment. Oral supplementation with spermidine retards age-related processes, but its influences on obesity and various metabolic tissues remain largely unknow. This study aims to investigate the effects of oral spermidine on brown adipose tissue (BAT) and skeletal muscle as well as its roles in counteracting obesity and metabolic disorders. METHODS AND RESULTS: Spermidine is orally administrated into high-fat diet (HFD)-fed mice. The weight gain, insulin resistance, and hepatic steatosis are attenuated by oral spermidine in HFD-fed mice, accompanied by an alleviation of white adipose tissue inflammation. Oral spermidine promotes BAT activation and metabolic adaptation of skeletal muscle in HFD-fed mice, evidenced by UCP-1 induction and CREB activation in both tissues. Notably, oral spermidine upregulates tyrosine hydroxylase in hypothalamus of HFD-fed mice; spermidine treatment increases tyrosine hydroxylase expression and norepinephrine production in neurocytes, which leads to CREB activation and UCP-1 induction in brown adipocytes and myotubes. Spermidine also directly promotes UCP-1 and PGC-1α expression in brown adipocytes and myotubes. CONCLUSION: Spermidine serves as an oral supplement to attenuate obesity and metabolic disorders through hypothalamus-dependent or -independent BAT activation and skeletal muscle adaptation.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Espermidina/administración & dosificación , Espermidina/farmacología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Administración Oral , Animales , Dieta Alta en Grasa/efectos adversos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/etiología , Paniculitis/tratamiento farmacológico , Paniculitis/etiología , Tirosina 3-Monooxigenasa/metabolismo
15.
Int J Biochem Cell Biol ; 138: 106053, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34371171

RESUMEN

Increased browning of white adipocytes (beiging) is considered a promising therapeutic strategy to fight obesity and its associated metabolic complications. However, the molecular mechanism modulating brown and beige fat-mediated thermogenesis is not fully elucidated. Here, we identified the lymphocyte cytosolic protein 1 (LCP1) as a factor that obstructs fat browning in white adipocytes. LCP1 plays a vital role in non-hematopoietic malignancies, and is also a well-known tumor biomarker; however, evidence regarding its function in adipocytes remains to be elucidated. The current study explores the physiological role of LCP1 in cultured 3T3-L1 white adipocytes, by applying the loss-of-function study using siRNA. Induction of fat browning by LCP1 depletion was evidenced by evaluating the gene and protein expression levels of brown fat-associated markers through real-time qRT-PCR and immunoblot analysis, respectively. We observed that deficiency of LCP1 promotes mitochondrial biogenesis, and significantly enhances expressions of the core brown fat-specific genes (Cd137, Cidea, Cited1, Tbx1, and Tmem26) and proteins (PGC-1α, PRDM16, and UCP1). In addition, deficiency of LCP1 promotes lipid catabolism as well as suppresses adipogenesis and lipogenesis. Loss of LCP1 also ameliorates cellular stress by downregulating JNK and c-JUN in adipocytes, and stimulates apoptosis. A mechanistic study revealed that deficiency of LCP1 induces browning in white adipocytes, independently via ß3-AR and the ERK signaling pathway. The current data reveals a previously unknown mechanism of LCP1 in browning of white adipocytes, and highlights the potential of LCP1 as a pharmacotherapeutic target for treating obesity and other metabolic disorders.


Asunto(s)
Adipocitos Blancos/citología , Tejido Adiposo Pardo/citología , Proteínas de Microfilamentos/deficiencia , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Estrés Fisiológico , Células 3T3-L1 , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Lipogénesis , Lipólisis , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Receptores Adrenérgicos beta 3/genética , Transducción de Señal , Termogénesis
16.
Int J Mol Sci ; 22(15)2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34361032

RESUMEN

17,18-Epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP) are bioactive epoxides produced from n-3 polyunsaturated fatty acid eicosapentaenoic acid and docosahexaenoic acid, respectively. However, these epoxides are quickly metabolized into less active diols by soluble epoxide hydrolase (sEH). We have previously demonstrated that an sEH inhibitor, t-TUCB, decreased serum triglycerides (TG) and increased lipid metabolic protein expression in the brown adipose tissue (BAT) of diet-induced obese mice. This study investigates the preventive effects of t-TUCB (T) alone or combined with 19,20-EDP (T + EDP) or 17,18-EEQ (T + EEQ) on BAT activation in the development of diet-induced obesity and metabolic disorders via osmotic minipump delivery in mice. Both T + EDP and T + EEQ groups showed significant improvement in fasting glucose, serum triglycerides, and higher core body temperature, whereas heat production was only significantly increased in the T + EEQ group. Moreover, both the T + EDP and T + EEQ groups showed less lipid accumulation in the BAT. Although UCP1 expression was not changed, PGC1α expression was increased in all three treated groups. In contrast, the expression of CPT1A and CPT1B, which are responsible for the rate-limiting step for fatty acid oxidation, was only increased in the T + EDP and T + EEQ groups. Interestingly, as a fatty acid transporter, CD36 expression was only increased in the T + EEQ group. Furthermore, both the T + EDP and T + EEQ groups showed decreased inflammatory NFκB signaling in the BAT. Our results suggest that 17,18-EEQ or 19,20-EDP combined with t-TUCB may prevent high-fat diet-induced metabolic disorders, in part through increased thermogenesis, upregulating lipid metabolic protein expression, and decreasing inflammation in the BAT.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Ácidos Araquidónicos/uso terapéutico , Benzoatos/uso terapéutico , Obesidad/tratamiento farmacológico , Compuestos de Fenilurea/uso terapéutico , Adipogénesis , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Fármacos Antiobesidad/administración & dosificación , Fármacos Antiobesidad/farmacología , Ácidos Araquidónicos/administración & dosificación , Ácidos Araquidónicos/farmacología , Benzoatos/administración & dosificación , Benzoatos/farmacología , Glucemia/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Dieta Alta en Grasa , Epóxido Hidrolasas/antagonistas & inhibidores , Ácidos Grasos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología
17.
Cells ; 10(5)2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-34066631

RESUMEN

Obesity is becoming a pandemic, and its prevalence is still increasing. Considering that obesity increases the risk of developing cardiometabolic diseases, research efforts are focusing on new ways to combat obesity. Brown adipose tissue (BAT) has emerged as a possible target to achieve this for its functional role in energy expenditure by means of increasing thermogenesis. An important metabolic sensor and regulator of whole-body energy balance is AMP-activated protein kinase (AMPK), and its role in energy metabolism is evident. This review highlights the mechanisms of BAT activation and investigates how AMPK can be used as a target for BAT activation. We review compounds and other factors that are able to activate AMPK and further discuss the therapeutic use of AMPK in BAT activation. Extensive research shows that AMPK can be activated by a number of different kinases, such as LKB1, CaMKK, but also small molecules, hormones, and metabolic stresses. AMPK is able to activate BAT by inducing adipogenesis, maintaining mitochondrial homeostasis and inducing browning in white adipose tissue. We conclude that, despite encouraging results, many uncertainties should be clarified before AMPK can be posed as a target for anti-obesity treatment via BAT activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Tejido Adiposo Pardo/metabolismo , Obesidad/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/patología , Animales , Técnicas de Cultivo de Célula , Metabolismo Energético , Humanos , Mitocondrias/metabolismo , Termogénesis
18.
Cells ; 10(5)2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-34066961

RESUMEN

Brown adipose tissue (BAT) activity plays a key role in regulating systemic energy. The activation of BAT results in increased energy expenditure, making this tissue an attractive pharmacological target for therapies against obesity and type 2 diabetes. Sirtuin 5 (SIRT5) affects BAT function by regulating adipogenic transcription factor expression and mitochondrial respiration. We analyzed the expression of SIRT5 in the different adipose depots of mice. We treated 3T3-L1 preadipocytes and mouse primary preadipocyte cultures with the SIRT5 inhibitor MC3482 and investigated the effects of this compound on adipose differentiation and function. The administration of MC3482 during the early stages of differentiation promoted the expression of brown adipocyte and mitochondrial biogenesis markers. Upon treatment with MC3482, 3T3-L1 adipocytes showed an increased activation of the AMP-activated protein kinase (AMPK), which is known to stimulate brown adipocyte differentiation. This effect was paralleled by an increase in autophagic/mitophagic flux and a reduction in lipid droplet size, mediated by a higher lipolytic rate. Of note, MC3482 increased the expression and the activity of adipose triglyceride lipase, without modulating hormone-sensitive lipase. Our findings reveal that SIRT5 inhibition stimulates brown adipogenesis in vitro, supporting this approach as a strategy to stimulate BAT and counteract obesity.


Asunto(s)
Adipogénesis , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Regulación de la Expresión Génica , Sirtuinas/antagonistas & inhibidores , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Diferenciación Celular , Metabolismo Energético , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Consumo de Oxígeno , Fenotipo
19.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(10): 158992, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34147658

RESUMEN

Brown adipose tissue (BAT) is specialized for uncoupled heat production through mitochondrion fueled majorly from fatty acids (FAs) of lipid droplets (LDs). How the interaction between the two organelles contributes the generation of heat remains elusive. Here, we report that LD-anchored mitochondria (LDAM) were observed in the BAT of mice raised at three different temperatures, 30 °C, 23 °C, and 6 °C. The biochemical analyses including Western blotting of electron transport chain subunits showed that LDAM were functional. Comparative proteomics analysis was conducted, which revealed differential expressions of proteins between LDAM and cytoplasmic mitochondria (CM) at different temperatures. Higher expressions of proteins at low temperature were observed for i) FA ß-oxidation in LDAM including FA synthesis and uncoupling, ii) pseudo-futile cycle in CM, and iii) two shuttle systems: glycerol 3-phosphate in both CM and LDAM and citrate malate in CM. Together, these results suggest that LDs and LDAM form a preorganized and functional organelle complex that permits the rapid response to cold.


Asunto(s)
Adipocitos Marrones/metabolismo , Frío/efectos adversos , Metabolismo Energético/genética , Gotas Lipídicas/metabolismo , Mitocondrias/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/ultraestructura , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/ultraestructura , Animales , Antígenos CD36/metabolismo , Fraccionamiento Celular , Separación Celular , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Gotas Lipídicas/ultraestructura , Metabolismo de los Lípidos/genética , Masculino , Ratones , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Oxidación-Reducción , Proteómica , Proteínas de Transporte Vesicular/metabolismo
20.
Cryo Letters ; 42(2): 96-105, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33970986

RESUMEN

BACKGROUND: Local fat accumulation is a health risk and this has raised interest in the development of aesthetic treatments, such as cryo-radiofrequency (CRF). OBJECTIVE: To evaluate the consequences of CRF in adipose tissue remodeling in a model system. MATERIALS AND METHODS: Lean and high-fat diet-induced obese mice were assessed 7 days after one CRF application; and lean mice were assessed 0, 3, 6 and 12 h after one application of CRF. Assessments included histology, DNA degradation, gene expression, ELISA of cytokines, serum analysis and neutrophil presence. RESULTS: Unchanged fat mass was found 7 days after CRF in obese and lean mice. However, lean mice showed smaller adipocyte size with areas resembling a browning process. TNF levels, apoptotic cells, and UCP-1 expression increased 7 days after CRF in inguinal adipose tissue of lean mice. Although no differences were found in fat mass, adipocyte size decreased just after CRF and this changed was maintained until 12 h, with cells resembling beige adipocytes. CONCLUSION: We suggest that CRF therapy is capable of inducing thermogenic adipocytes in lean mice.


Asunto(s)
Tejido Adiposo Pardo , Tejido Adiposo Blanco , Crioterapia , Obesidad/terapia , Terapia por Radiofrecuencia , Adipocitos , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Ratones , Ratones Endogámicos C57BL , Termogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA