Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 403
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.
Am J Physiol Endocrinol Metab ; 321(5): E581-E591, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34459218

RESUMEN

This study determined if a perturbation in in utero adipogenesis leading to later life adipose tissue (AT) dysfunction underlies programming of cardiometabolic risk in offspring born to dams with metabolic dysfunction. Female mice heterozygous for the leptin receptor deficiency (Hetdb) had 2.4-fold higher prepregnancy fat mass and in late gestation had higher plasma insulin and triglycerides compared with wild-type (Wt) females (P < 0.05). To isolate the role of the intrauterine milieu, wild-type (Wt) offspring from each pregnancy were studied. Differentiation potential in isolated progenitors and cell size distribution analysis revealed accelerated adipogenesis in Wt pups born to Hetdb dams, accompanied by a higher accumulation of neonatal fat mass. In adulthood, whole body fat mass by NMR was higher in male (69%) and female (20%) Wt offspring born to Hetdb versus Wt pregnancies, along with adipocyte hypertrophy and hyperlipidemia (all P < 0.05). Lipidomic analyses by gas chromatography revealed an increased lipogenic index (16:0/18:2n6) after high-fat/fructose diet (HFFD). Postprandial insulin, ADIPO-IR, and ex vivo AT lipolytic responses to isoproterenol were all higher in Wt offspring born to Hetdb dams (P < 0.05). Intrauterine metabolic stimuli may direct a greater proportion of progenitors toward terminal differentiation, thereby predisposing to hypertrophy-induced adipocyte dysfunction.NEW & NOTEWORTHY This study reveals that accelerated adipogenesis during the perinatal window of adipose tissue development predisposes to later life hypertrophic adipocyte dysfunction, thereby compromising the buffering function of the subcutaneous depot.


Asunto(s)
Adipogénesis , Tejido Adiposo/metabolismo , Factores de Riesgo Cardiometabólico , Enfermedades Metabólicas/metabolismo , Adipocitos/ultraestructura , Tejido Adiposo/embriología , Tejido Adiposo/crecimiento & desarrollo , Animales , Tamaño de la Célula , Dieta Alta en Grasa , Femenino , Fructosa/farmacología , Hiperlipidemias/genética , Insulina/sangre , Lipidómica , Masculino , Enfermedades Metabólicas/patología , Ratones , Embarazo , Receptores de Leptina/genética , Células Madre , Triglicéridos/sangre
3.
Dermatol Surg ; 47(8): 1059-1064, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34115683

RESUMEN

BACKGROUND: Radiofrequency (RF) and high-intensity focused electromagnetic (HIFEM) technologies are used for noninvasive body shaping as standalone modalities. OBJECTIVE: To examine the effects of novel synchronized RF and HIFEM on subcutaneous adipose tissue in a porcine animal model. MATERIALS AND METHODS: Seven large white pigs aged 6 months received 3 abdominal treatments of simultaneous application of synchronized RF and HIFEM (30 minutes, once per week). Punch biopsies of treated and control subcutaneous tissue were collected at the baseline, 4 days, 2 weeks, 1 month, and 2 months. Specimens were examined by light and scanning electron microscopy. Adipocyte volume was analyzed. Fat tissue temperature was measured in situ (fiber optic probes) and superficially (thermal imager). RESULTS: Fat layer was heated to temperatures of 42 to 45°C. Signs of fat apoptosis (shape alternations and pyknotic nuclei) appeared at day 4 and peaked between 2 weeks and 1 month. Adipocyte volume decreased significantly (p < .001) by 31.1% at 2 weeks, 1 month (-23.6%), and 2 months (-22.0%). Control samples showed healthy adipocytes. Scanning electron microscopy micrographs corroborated histology findings, showing flattened, volume-depleted and disrupted adipocytes. CONCLUSION: Synchronized RF with HIFEM procedure resulted in a significant and sustained fat reduction with no adverse events.


Asunto(s)
Contorneado Corporal/métodos , Magnetoterapia/métodos , Terapia por Radiofrecuencia/métodos , Grasa Subcutánea/efectos de la radiación , Adipocitos/efectos de la radiación , Adipocitos/ultraestructura , Animales , Contorneado Corporal/efectos adversos , Contorneado Corporal/instrumentación , Terapia Combinada/instrumentación , Terapia Combinada/métodos , Femenino , Calor/efectos adversos , Magnetoterapia/efectos adversos , Magnetoterapia/instrumentación , Microscopía Electrónica , Modelos Animales , Terapia por Radiofrecuencia/efectos adversos , Terapia por Radiofrecuencia/instrumentación , Grasa Subcutánea/citología , Grasa Subcutánea/ultraestructura , Porcinos
4.
J Exp Med ; 218(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33315085

RESUMEN

White adipose tissues (WAT) play crucial roles in maintaining whole-body energy homeostasis, and their dysfunction can contribute to hepatic insulin resistance and type 2 diabetes mellitus (T2DM). However, the mechanisms underlying these alterations remain unknown. By analyzing the transcriptome landscape in human adipocytes based on available RNA-seq datasets from lean, obese, and T2DM patients, we reveal elevated mitochondrial reactive oxygen species (ROS) pathway and NF-κB signaling with altered fatty acid metabolism in T2DM adipocytes. Mice with adipose-specific deletion of mitochondrial redox Trx2 develop hyperglycemia, hepatic insulin resistance, and hepatic steatosis. Trx2-deficient WAT exhibited excessive mitophagy, increased inflammation, and lipolysis. Mechanistically, mitophagy was induced through increasing ROS generation and NF-κB-dependent accumulation of autophagy receptor p62/SQSTM1, which recruits damaged mitochondria with polyubiquitin chains. Importantly, administration of ROS scavenger or NF-κB inhibitor ameliorates glucose and lipid metabolic disorders and T2DM progression in mice. Taken together, this study reveals a previously unrecognized mechanism linking mitophagy-mediated adipose inflammation to T2DM with hepatic insulin resistance.


Asunto(s)
Tejido Adiposo/patología , Diabetes Mellitus Tipo 2/patología , Inflamación/patología , Resistencia a la Insulina , Hígado/patología , Mitofagia , Adipocitos/metabolismo , Adipocitos/ultraestructura , Animales , Dieta Alta en Grasa , Metabolismo Energético , Hígado Graso/patología , Eliminación de Gen , Marcación de Gen , Gluconeogénesis , Homeostasis , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/patología , Lipogénesis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , FN-kappa B/metabolismo , Estrés Oxidativo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Proteína Sequestosoma-1 , Transducción de Señal , Tiorredoxinas/metabolismo
5.
Biochimie ; 177: 98-107, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32822725

RESUMEN

The dietary intake of elaidate (elaidic acid), a trans-fatty acid, is associated with the development of various diseases. Since elaidate is a C18 unsaturated fatty acid with a steric structure similar to that of a C18 saturated fatty acid (stearate), we previously revealed that insulin-dependent glucose uptake was impaired in adipocytes exposed to elaidate prior to and during differentiation similar to stearate. However, it is still unknown whether the mechanism of impairment of insulin-dependent glucose uptake due to elaidate is similar to that of stearate. Here, we indicate that persistent exposure to elaidate has particular effects on insulin signaling and GLUT4 dynamics. Insulin-induced accumulation of Akt at the plasma membrane (PM) and elevations of phosphorylated Akt and AS160 levels in whole cells were suppressed in adipocytes persistently exposed to 50 µM elaidate. Interestingly, persistent exposure to the same concentration of stearate has no effect on the phosphorylated Akt and AS160 levels. When cells were exposed to these fatty acids, elaidate suppressed insulin-induced fusion, but not translocation, of GLUT4 storage vesicles in the PM, whereas stearate did not suppress the fusion and translocation of GLUT4 storage, indicating that elaidate has suppressive effects on the accumulation of Akt and fusion of GLUT4 storage vesicles and that both elaidate and stearate vary in the mechanisms by which they impair insulin-dependent glucose uptake.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Ácidos Oléicos/farmacología , Transducción de Señal/efectos de los fármacos , Estearatos/farmacología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/ultraestructura , Animales , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Membrana Celular/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Ratones , Ácidos Oléicos/química , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estearatos/química , Vesículas Transportadoras/efectos de los fármacos
6.
Cells ; 9(5)2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32455814

RESUMEN

Reduced neo-adipogenesis and dysfunctional lipid-overloaded adipocytes are hallmarks of hypertrophic obesity linked to insulin resistance. Identifying molecular features of hypertrophic adipocytes requires appropriate in vitro models. We describe the generation of a model of human hypertrophic-like adipocytes directly comparable to normal adipose cells and the pathologic evolution toward hypertrophic state. We generate in vitro hypertrophic cells from mature adipocytes, differentiated from human mesenchymal stem cells. Combining optical, confocal, and transmission electron microscopy with mRNA/protein quantification, we characterize this cellular model, confirming specific alterations also in subcutaneous adipose tissue. Specifically, we report the generation and morphological/molecular characterization of human normal and hypertrophic-like adipocytes. The latter displays altered morphology and unbalance between canonical and dominant negative (PPARGΔ5) transcripts of PPARG, paralleled by reduced expression of PPARγ targets, including GLUT4. Furthermore, the unbalance of PPARγ isoforms associates with GLUT4 down-regulation in subcutaneous adipose tissue of individuals with overweight/obesity or impaired glucose tolerance/type 2 diabetes, but not with normal weight or glucose tolerance. In conclusion, the hypertrophic-like cells described herein are an innovative tool for studying molecular dysfunctions in hypertrophic obesity and the unbalance between PPARγ isoforms associates with down-regulation of GLUT4 and other PPARγ targets, representing a new hallmark of hypertrophic adipocytes.


Asunto(s)
Adipocitos/metabolismo , Adipocitos/patología , PPAR gamma/metabolismo , Adipocitos/ultraestructura , Tejido Adiposo/patología , Diferenciación Celular , Línea Celular , Forma de la Célula , Tamaño de la Célula , Femenino , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Hipertrofia , Gotas Lipídicas/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Modelos Biológicos , Obesidad/metabolismo , Obesidad/patología , Isoformas de Proteínas/metabolismo
7.
Mol Metab ; 39: 101005, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32344065

RESUMEN

OBJECTIVE: Beclin1 is a core molecule of the macroautophagy machinery. Although dysregulation of macroautophagy is known to be involved in metabolic disorders, the function of Beclin1 in adipocyte metabolism has not been investigated. In the present study, we aimed to study the role of Beclin1 in lipolysis and mitochondrial homeostasis of adipocytes. METHODS: Autophagic flux during lipolysis was examined in adipocytes cultured in vitro and in the adipose tissue of mice. Adipocyte-specific Beclin1 knockout (KO) mice were used to investigate the activities of Beclin1 in adipose tissues. RESULTS: cAMP/PKA signaling increased the autophagic flux in adipocytes differentiated from C3H10T1/2 cells. In vivo autophagic flux was higher in the brown adipose tissue (BAT) than that in the white adipose tissue and was further increased by the ß3 adrenergic receptor agonist CL316243. In addition, surgical denervation of BAT greatly reduced autophagic flux, indicating that sympathetic nerve activity is a major regulator of tissue autophagy. Adipocyte-specific KO of Beclin1 led to a hypertrophic enlargement of lipid droplets in BAT and impaired CL316243-induced lipolysis/lipid mobilization and energy expenditure. While short-term effects of Beclin1 deletion were characterized by an increase in mitochondrial proteins, long-term Beclin1 deletion led to severe disruption of autophagy, resulting in mitochondrial loss, and dramatically reduced the expression of genes involved in lipid metabolism. Consequently, adipose tissue underwent increased activation of cell death signaling pathways, macrophage recruitment, and inflammation, particularly in BAT. CONCLUSIONS: The present study demonstrates the critical roles of Beclin1 in the maintenance of lipid metabolism and mitochondrial homeostasis in adipose tissues.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Beclina-1/genética , Eliminación de Gen , Lipólisis/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Adipocitos/ultraestructura , Tejido Adiposo Pardo/metabolismo , Animales , Autofagia/genética , Beclina-1/metabolismo , Línea Celular , AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Variaciones en el Número de Copia de ADN , Inmunidad , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Transducción de Señal , Termogénesis/genética
8.
Acta Histochem ; 122(4): 151537, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32197756

RESUMEN

The understanding of fat tissue plays an eminent role in plastic surgery as well as in metabolic research. Histopathological analysis of tissue samples provides insight in free fat graft survival and culture experiments help to better understand fat tissue derived stem cells (ASCs). To facilitate such experiments, modern image-based histology could provide an automatized approach to a large amount of data to gain not only qualitative but also quantitative data. This study was designed to critically evaluate image-based analysis of fat tissue samples in cell culture or in tissue probes and to identify critical parameters to avoid bias in further studies. In the first part of the study, ASCs were harvested and differentiated into adipocytes in cell culture. Histology was performed with the fluorescent dye BODIPY and the obtained digital images were analyzed using Image J software. In the second part of the study, digitalized histology of a previous in vivo study was subjected to automatized fat vacuole quantification using Image J. Both approaches were critically reviewed, and different software parameter settings were tested. Results showed that automatized digital image analysis allows the quantification of fat tissue probes with enough precision giving significant results. But the testing of different software parameters revealed a significant influence of parameters themselves on calculated results. Therefore, we recommend the use of image-based analysis to quantify fat tissue probes to improve the comparability of studies. But we also emphasize to calibrate software using internal controls in every single experimental approach.


Asunto(s)
Tejido Adiposo/anatomía & histología , Procesamiento de Imagen Asistido por Computador/métodos , Programas Informáticos , Adipocitos/ultraestructura , Tejido Adiposo/ultraestructura , Automatización , Células Cultivadas , Humanos , Reproducibilidad de los Resultados , Vacuolas/ultraestructura
9.
Cell Rep ; 30(4): 949-958.e6, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995765

RESUMEN

Under caloric restriction, bone marrow adipocytes (BM-Ads) do not decrease in size compared to white adipocytes, suggesting they harbor unique metabolic properties. We compare human primary BM-Ads with paired subcutaneous adipocytes (SC-Ads) using proteomic and lipidomic approaches. We find that, although SC-Ads and BM-Ads share similar morphological features, they possess distinct lipid metabolism. Although BM-Ad shows enrichment in proteins involved in cholesterol metabolism, correlating with increased free cholesterol content, proteins involved in lipolysis were downregulated. In particular, monoacylglycerol lipase expression is strongly reduced in BM-Ads, leading to monoacylglycerol accumulation. Consequently, basal and induced lipolytic responses are absent in BM-Ads, affirming their differences in metabolic fitness upon caloric restriction. These specific metabolic features are not recapitulated in vitro using common protocols to differentiate bone marrow mesenchymal stem cells. Thus, contrary to classical SC-Ads, BM-Ads display a specific lipid metabolism, as they are devoid of lipolytic activity and exhibit a cholesterol-orientated metabolism.


Asunto(s)
Adipocitos/metabolismo , Médula Ósea/metabolismo , Metabolismo de los Lípidos , Proteoma/metabolismo , Adipocitos/citología , Adipocitos/enzimología , Adipocitos/ultraestructura , Animales , Médula Ósea/enzimología , Restricción Calórica , Línea Celular , Células Cultivadas , Colesterol/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Lipólisis/fisiología , Ratones , Microscopía Electrónica de Transmisión , Monoacilglicerol Lipasas/genética , Monoacilglicerol Lipasas/metabolismo , Mapas de Interacción de Proteínas/genética , Mapas de Interacción de Proteínas/fisiología , Proteoma/genética , Proteómica
10.
Am J Physiol Endocrinol Metab ; 318(3): E371-E380, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31910029

RESUMEN

Excess androgen-induced obesity has become a public health problem, and its prevalence has increased substantially in recent years. Chemokine-like receptor 1 (CMKLR1), a receptor of chemerin secreted by adipose tissue, is linked to adipocyte differentiation, adipose tissue development, and obesity. However, the effect of CMKLR1 signaling on androgen-mediated adiposity in vivo remains unclear. Using CMKLR1-knockout mice, we constructed an androgen-excess female mouse model through 5α-dihydrotestosterone (DHT) treatment and an androgen-deficient male mouse model by orchidectomy (ORX). For mechanism investigation, we used 2-(α-Naphthoyl) ethyltrimethylammonium iodide (α-NETA), an antagonist of CMKLR1, to suppress CMKLR1 in vivo and wortmannin, a PI3K signaling antagonist, to treat brown adipose tissue (BAT) explant cultures in vitro. Furthermore, we used histological examination and quantitative PCR, as well as Western blot analysis, glucose tolerance tests, and biochemical analysis of serum, to describe the phenotypes and the changes in gene expression. We demonstrated that excess androgen in the female mice resulted in larger cells in the white adipose tissue (WAT) and the BAT, whereas androgen deprivation in the male mice induced a reduction in cell size. Both of these adipocyte size effects could be attenuated in the CMKLR1-knockout mice. CMKLR1 deficiency influenced the effect of androgen treatment on adipose tissue by regulating the mRNA expression of the androgen receptor (AR) and adipocyte markers (such as Fabp4 and Cidea). Moreover, suppression of CMKLR1 by α-NETA could also reduce the extent of the adipocyte cell enlargement caused by DHT. Furthermore, we found that DHT could reduce the levels of phosphorylated ERK (pERK) in the BAT, while CMKLR1 inactivation inhibited this effect, which had been induced by DHT, through the PI3K signaling pathway. These findings reveal an antiobesity role of CMKLR1 deficiency in regulating lipid accumulation, highlighting the scientific importance for the further development of small-molecule CMKLR1 antagonists as fundamental research tools and/or as potential drugs for use in the treatment of adiposity.


Asunto(s)
Andrógenos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Receptores de Quimiocina/deficiencia , Adipocitos/metabolismo , Adipocitos/ultraestructura , Tejido Adiposo Pardo/efectos de los fármacos , Andrógenos/deficiencia , Animales , Peso Corporal , Tamaño de la Célula , Dihidrotestosterona/farmacología , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Naftalenos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Compuestos de Amonio Cuaternario/farmacología , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/genética , Wortmanina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...