Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 167(3): 843-857.e14, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27720451

RESUMEN

Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.


Asunto(s)
Glucagón/uso terapéutico , Enfermedades Metabólicas/tratamiento farmacológico , Triyodotironina/efectos de los fármacos , Animales , Aterosclerosis/tratamiento farmacológico , Peso Corporal/efectos de los fármacos , Huesos/efectos de los fármacos , Ingeniería Química/métodos , Colesterol/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Combinación de Medicamentos , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Glucagón/efectos adversos , Glucagón/química , Glucagón/farmacología , Hiperglucemia/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Terapia Molecular Dirigida , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Triyodotironina/efectos adversos , Triyodotironina/química , Triyodotironina/farmacología
2.
Exp Cell Res ; 425(1): 113536, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36858342

RESUMEN

The neurotoxin MPP+ triggers cell death of dopamine neurons and induces Parkinson's disease symptoms in mice and men, but the immediate transcriptional response to this neurotoxin has not been studied. We therefore treated human SH-SY5Y cells with a low dose (0.1 mM) of MPP+ and measured the effect on nascent transcription by precision run-on sequencing (PRO-seq). We found that transcription of the mitochondrial genome was significantly reduced already after 30 min, whereas nuclear gene transcription was unaffected. Inhibition of respiratory complex I by MPP+ led to reduced ATP production, that may explain the diminished activity of mitochondrial RNA polymerase. Our results show that MPP+ has a direct effect on mitochondrial function and transcription, and that other gene expression or epigenetic changes induced by this neurotoxin are secondary effects that reflect a cellular adaptation program.


Asunto(s)
Neuroblastoma , Neurotoxinas , Humanos , Neurotoxinas/toxicidad , Neurotoxinas/metabolismo , 1-Metil-4-fenilpiridinio/toxicidad , Neuronas/metabolismo , Neuroblastoma/metabolismo , Transcripción Genética , Línea Celular Tumoral , Apoptosis
3.
FASEB J ; 28(2): 761-70, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24200885

RESUMEN

Mitochondrial dysfunction in white adipose tissue plays a key role in the pathogenesis of type 2 diabetes. Emerging evidence specifically suggests that altered oxidative phosphorylation in adipocytes may have a relevant effect on systemic glucose homeostasis, requiring understanding of adipocyte bioenergetics. We analyzed energetic flux of an intact human adipocyte cell model by plate-based respirometry and extracellular acidification. During differentiation, we discovered that glycolytic ATP production was increasingly replaced by mitochondrial oxidative metabolism (from 20 to 60%). This observation was corroborated by simultaneous up-regulation of canonical mitochondrial gene programs, such as peroxisome proliferator-activated receptor γ coactivator α (PGC1α; 150-fold) and cytochrome c-1 (CytC; 3-fold). Mimicking diabetic phenotypes by exposure to various glucose levels (0, 5, and 25 mM) resulted in immediate adjustments of glycolytic and mitochondrial activity that aimed to maintain intracellular ATP. We conclude that ATP deficits by mitochondrial failure are compensated by glycolytic ATP production, resulting in inefficient conversion of glucose to cellular ATP. Metabolic inefficiency may enhance glucose uptake, therefore improving systemic glucose homeostasis. Notably, mature adipocytes developed a high spare respiratory capacity (increased by 6-fold) permitting rapid adaptation to metabolic changes. Spare respiratory capacity may also allow additional metabolic scope for energy dissipation, potentially offering new therapeutic targets for the treatment of metabolic disease.


Asunto(s)
Adenosina Trifosfato/metabolismo , Adipocitos/metabolismo , Adipocitos/efectos de los fármacos , Células Cultivadas , Citocromos c1/metabolismo , Glucosa/farmacología , Glucólisis , Humanos , Fosforilación Oxidativa , PPAR gamma/metabolismo
4.
Circulation ; 128(22): 2364-71, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24170386

RESUMEN

BACKGROUND: Abnormal glucose metabolism is a central feature of disorders with increased rates of cardiovascular disease. Low levels of high-density lipoprotein (HDL) are a key predictor for cardiovascular disease. We used genetic mouse models with increased HDL levels (apolipoprotein A-I transgenic [apoA-I tg]) and reduced HDL levels (apoA-I-deficient [apoA-I ko]) to investigate whether HDL modulates mitochondrial bioenergetics in skeletal muscle. METHODS AND RESULTS: ApoA-I ko mice exhibited fasting hyperglycemia and impaired glucose tolerance test compared with wild-type mice. Mitochondria isolated from gastrocnemius muscle of apoA-I ko mice displayed markedly blunted ATP synthesis. Endurance capacity during exercise exhaustion test was impaired in apoA-I ko mice. HDL directly enhanced glucose oxidation by increasing glycolysis and mitochondrial respiration rate in C2C12 muscle cells. ApoA-I tg mice exhibited lower fasting glucose levels, improved glucose tolerance test, increased lactate levels, reduced fat mass, associated with protection against age-induced decline of endurance capacity compared with wild-type mice. Circulating levels of fibroblast growth factor 21, a novel biomarker for mitochondrial respiratory chain deficiencies and inhibitor of white adipose lipolysis, were significantly reduced in apoA-I tg mice. Consistent with an increase in glucose utilization of skeletal muscle, genetically increased HDL and apoA-I levels in mice prevented high-fat diet-induced impairment of glucose homeostasis. CONCLUSIONS: In view of impaired mitochondrial function and decreased HDL levels in type 2 diabetes mellitus, our findings indicate that HDL-raising therapies may preserve muscle mitochondrial function and address key aspects of type 2 diabetes mellitus beyond cardiovascular disease.


Asunto(s)
Glucemia/metabolismo , Intolerancia a la Glucosa/metabolismo , Hiperglucemia/metabolismo , Lipoproteínas HDL/metabolismo , Músculo Esquelético/metabolismo , Animales , Apolipoproteína A-I/genética , Respiración de la Célula/fisiología , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/sangre , Factores de Crecimiento de Fibroblastos/sangre , Homeostasis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Musculares/metabolismo , Resistencia Física/fisiología
5.
Science ; 384(6700): 1111-1117, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38843333

RESUMEN

Brown adipose tissue (BAT) is a heater organ that expresses thermogenic uncoupling protein 1 (UCP1) to maintain high body temperatures during cold stress. BAT thermogenesis is considered an overarching mammalian trait, but its evolutionary origin is unknown. We show that adipose tissue of marsupials, which diverged from eutherian mammals ~150 million years ago, expresses a nonthermogenic UCP1 variant governed by a partial transcriptomic BAT signature similar to that found in eutherian beige adipose tissue. We found that the reconstructed UCP1 sequence of the common eutherian ancestor displayed typical thermogenic activity, whereas therian ancestor UCP1 is nonthermogenic. Thus, mammalian adipose tissue thermogenesis may have evolved in two distinct stages, with a prethermogenic stage in the common therian ancestor linking UCP1 expression to adipose tissue and thermal stress. We propose that in a second stage, UCP1 acquired its thermogenic function specifically in eutherians, such that the onset of mammalian BAT thermogenesis occurred only after the divergence from marsupials.


Asunto(s)
Tejido Adiposo Pardo , Evolución Biológica , Marsupiales , Termogénesis , Proteína Desacopladora 1 , Animales , Humanos , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Euterios/genética , Euterios/fisiología , Evolución Molecular , Marsupiales/genética , Marsupiales/fisiología , Filogenia , Termogénesis/genética , Transcriptoma , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
6.
Redox Biol ; 66: 102874, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37683300

RESUMEN

OBJECTIVE: Enhancing energy turnover via uncoupled mitochondrial respiration in adipose tissue has great potential to improve human obesity and other metabolic complications. However, the amount of human brown adipose tissue and its uncoupling protein 1 (UCP1) is low in obese patients. Recently, a class of endogenous molecules, N-acyl amino acids (NAAs), was identified as mitochondrial uncouplers in murine adipocytes, presumably acting via the adenine nucleotide translocator (ANT). Given the translational potential, we investigated the bioenergetic effects of NAAs in human adipocytes, characterizing beneficial and adverse effects, dose ranges, amino acid derivatives and underlying mechanisms. METHOD: NAAs with neutral (phenylalanine, leucine, isoleucine) and polar (lysine) residues were synthetized and assessed in intact and permeabilized human adipocytes using plate-based respirometry. The Seahorse technology was applied to measure bioenergetic parameters, dose-dependency, interference with UCP1 and adenine nucleotide translocase (ANT) activity, as well as differences to the established chemical uncouplers niclosamide ethanolamine (NEN) and 2,4-dinitrophenol (DNP). RESULT: NAAs with neutral amino acid residues potently induce uncoupled respiration in human adipocytes in a dose-dependent manner, even in the presence of the UCP1-inhibitor guanosine diphosphate (GDP) and the ANT-inhibitor carboxyatractylate (CAT). However, neutral NAAs significantly reduce maximal oxidation rates, mitochondrial ATP-production, coupling efficiency and reduce adipocyte viability at concentrations above 25 µM. The in vitro therapeutic index (using induced proton leak and viability as determinants) of NAAs is lower than that of NEN and DNP. CONCLUSION: NAAs are potent mitochondrial uncouplers in human adipocytes, independent of UCP1 and ANT. However, previously unnoticed adverse effects harm adipocyte functionality, reduce the therapeutic index of NAAs in vitro and therefore question their suitability as anti-obesity agents without further chemical modifications.


Asunto(s)
Adipocitos , Aminoácidos , Humanos , Animales , Ratones , Etanolamina , Tejido Adiposo Pardo , Metabolismo Energético
7.
Exp Cell Res ; 317(15): 2200-9, 2011 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-21756905

RESUMEN

Adipocyte apoptosis is an important regulator of adipocyte number in fat depots. We have previously shown that an inhibition of protein synthesis sensitizes human adipocytes for apoptosis. In vivo, dramatic changes in the fat cell's protein expression should be anticipated under special conditions such as calorie restriction. Here, we studied the underlying mechanism by which human preadipocytes and adipocytes are sensitized for death receptor induced apoptosis in vitro. The protein synthesis blocker cycloheximide (CHX) sensitized human fat cells for CD95-induced apoptosis in a caspase-dependent manner. Treatment with CHX differentially changed expression of pro- and anti-apoptotic proteins. Most noticeably, FLICE-like inhibitory protein (FLIP) expression rapidly decreased during CHX treatment. Reduction of FLIP levels resulted in undetectable amounts of FLIP at the CD95 death-inducing signaling complex (DISC) upon CD95 stimulation, thereby enhancing recruitment and activation at caspase-8. Down-regulation of FLIP by shRNA sensitized preadipocytes for CD95-induced apoptosis. In mice, adipose tissue mRNA levels of Flip were down-regulated upon fasting. In conclusion, we identify FLIP as an important regulator of apoptosis sensitivity in fat cells. Modulating adipocyte homeostasis by apoptosis might provide a new therapeutic concept to get rid of excess adipose tissue, and FLIP might be a possible target molecule.


Asunto(s)
Adipocitos/metabolismo , Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Cicloheximida/farmacología , Regulación hacia Abajo , Receptor fas/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Células Cultivadas , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
8.
Mol Cell Endocrinol ; 533: 111337, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062167

RESUMEN

Thermogenic adipose tissue, which comprises classical brown and beige adipose tissue, has the ability to improve systemic metabolism. Its identification in adult humans has fostered extensive investigations on the therapeutic value to counteract obesity and metabolic disorders. Sex and gender differences of human thermogenic adipose tissue, however, are still understudied despite their importance for personalized treatment options. Here, we review studies reporting human sex differences of thermogenic adipose tissue and related potential improvements of systemic energy metabolism. An increasing body of evidence suggests higher prevalence, mass and activity of thermogenic adipose tissue in women, but the consequences for metabolic disease progression and mechanisms are largely unknown. Therefore, we also discuss observations on sex-specific adipose metabolism in experimental mouse and rat studies that may assist to establish molecular mechanisms and instruct future investigations in humans.


Asunto(s)
Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Enfermedades Metabólicas/metabolismo , Obesidad/metabolismo , Animales , Metabolismo Energético , Femenino , Humanos , Metabolismo de los Lípidos , Masculino , Caracteres Sexuales , Termogénesis
9.
Cells ; 10(12)2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34944072

RESUMEN

BACKGROUND: Tissue-resident macrophages have mixed developmental origins. They derive in variable extent from yolk sac (YS) hematopoiesis during embryonic development. Bone marrow (BM) hematopoietic progenitors give rise to tissue macrophages in postnatal life, and their contribution increases upon organ injury. Since the phenotype and functions of macrophages are modulated by the tissue of residence, the impact of their origin and developmental paths has remained incompletely understood. METHODS: In order to decipher cell-intrinsic macrophage programs, we immortalized hematopoietic progenitors from YS and BM using conditional HoxB8, and carried out an in-depth functional and molecular analysis of differentiated macrophages. RESULTS: While YS and BM macrophages demonstrate close similarities in terms of cellular growth, differentiation, cell death susceptibility and phagocytic properties, they display differences in cell metabolism, expression of inflammatory markers and inflammasome activation. Reduced abundance of PYCARD (ASC) and CASPASE-1 proteins in YS macrophages abrogated interleukin-1ß production in response to canonical and non-canonical inflammasome activation. CONCLUSIONS: Macrophage ontogeny is associated with distinct cellular programs and immune response. Our findings contribute to the understanding of the regulation and programming of macrophage functions.


Asunto(s)
Médula Ósea/patología , Inflamación/patología , Macrófagos/patología , Saco Vitelino/patología , Animales , Diferenciación Celular/genética , Proliferación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucólisis , Células HEK293 , Células Madre Hematopoyéticas/patología , Proteínas de Homeodominio/metabolismo , Humanos , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Fagocitosis , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transcriptoma/genética
10.
Exp Clin Endocrinol Diabetes ; 128(11): 752-770, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31108554

RESUMEN

Human fibroblast growth factor 21 (FGF21) is primarily produced and secreted by the liver as a hepatokine. This hormone circulates to its target tissues (e. g., brain, adipose tissue), which requires two components, one of the preferred FGF receptor isoforms (FGFR1c and FGFR3c) and the co-factor beta-Klotho (KLB) to trigger downstream signaling pathways. Although targeting FGF21 signaling in humans by analogues and receptor agonists results in beneficial effects, e. g., improvements in plasma lipids and decreased body weight, it failed to recapitulate the improvements in glucose handling shown for many mouse models. FGF21's role and metabolic effects in mice and its therapeutic potential have extensively been reviewed elsewhere. In this review we focus on circulating FGF21 levels in humans and their associations with disease and clinical parameters, focusing primarily on obesity and obesity-associated diseases such as type-2 diabetes. We provide a comprehensive overview on human circulating FGF21 levels under normal physiology and metabolic disease. We discuss the emerging field of inactivating FGF21 in human blood by fibroblast activation protein (FAP) and its potential clinical implications.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Endopeptidasas/metabolismo , Factores de Crecimiento de Fibroblastos/sangre , Proteínas de la Membrana/metabolismo , Enfermedades Metabólicas/sangre , Obesidad/sangre , Biomarcadores/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Humanos , Enfermedades Metabólicas/diagnóstico , Obesidad/diagnóstico
11.
Sci Rep ; 10(1): 12407, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32709986

RESUMEN

Among obese subjects, metabolically healthy (MHO) and unhealthy obese (MUHO) subjects exist, the latter being characterized by whole-body insulin resistance, hepatic steatosis, and subclinical inflammation. Insulin resistance and obesity are known to associate with alterations in mitochondrial density, morphology, and function. Therefore, we assessed mitochondrial function in human subcutaneous preadipocytes as well as in differentiated adipocytes derived from well-matched donors. Primary subcutaneous preadipocytes from 4 insulin-resistant (MUHO) versus 4 insulin-sensitive (MHO), non-diabetic, morbidly obese Caucasians (BMI > 40 kg/m2), matched for sex, age, BMI, and percentage of body fat, were differentiated in vitro to adipocytes. Real-time cellular respiration was measured using an XF24 Extracellular Flux Analyzer (Seahorse). Lipolysis was stimulated by forskolin (FSK) treatment. Mitochondrial respiration was fourfold higher in adipocytes versus preadipocytes (p = 1.6*10-9). In adipocytes, a negative correlation of mitochondrial respiration with donors' insulin sensitivity was shown (p = 0.0008). Correspondingly, in adipocytes of MUHO subjects, an increased basal respiration (p = 0.002), higher proton leak (p = 0.04), elevated ATP production (p = 0.01), increased maximal respiration (p = 0.02), and higher spare respiratory capacity (p = 0.03) were found, compared to MHO. After stimulation with FSK, the differences in ATP production, maximal respiration and spare respiratory capacity were blunted. The differences in mitochondrial respiration between MUHO/MHO were not due to altered mitochondrial content, fuel switch, or lipid metabolism. Thus, despite the insulin resistance of MUHO, we could clearly show an elevated mitochondrial respiration of MUHO adipocytes. We suggest that the higher mitochondrial respiration reflects a compensatory mechanism to cope with insulin resistance and its consequences. Preserving this state of compensation might be an attractive goal for preventing or delaying the transition from insulin resistance to overt diabetes.


Asunto(s)
Adipocitos/patología , Salud , Mitocondrias/metabolismo , Obesidad/metabolismo , Obesidad/patología , Adulto , Índice de Masa Corporal , Respiración de la Célula , Femenino , Glucólisis , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
12.
Endocr Connect ; 8(6): R105-R121, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31085768

RESUMEN

The crosstalk between macrophages (MΦ) and adipocytes within white adipose tissue (WAT) influences obesity-associated insulin resistance and other associated metabolic disorders, such as atherosclerosis, hypertension and type 2 diabetes. MΦ infiltration is increased in WAT during obesity, which is linked to decreased mitochondrial content and activity. The mechanistic interplay between MΦ and mitochondrial function of adipocytes is under intense investigation, as MΦ and inflammatory pathways exhibit a pivotal role in the reprogramming of WAT metabolism in physiological responses during cold, fasting and exercise. Thus, the underlying immunometabolic pathways may offer therapeutic targets to correct obesity and metabolic disease. Here, I review the current knowledge on the quantity and the quality of human adipose tissue macrophages (ATMΦ) and their impact on the bioenergetics of human adipocytes. The effects of ATMΦ and their secreted factors on mitochondrial function of white adipocytes are discussed, including recent research on MΦ as part of an immune signaling cascade involved in the 'browning' of WAT, which is defined as the conversion from white, energy-storing adipocytes into brown, energy-dissipating adipocytes.

13.
Mol Aspects Med ; 68: 6-17, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31238069

RESUMEN

In eutherian mammals, brown adipose tissue (BAT) permits non-shivering thermogenesis (NST) through high metabolic rates catalyzed by the unique mitochondrial uncoupling protein 1 (UCP1). The tissue has recently gained remarkable attention due to its discovery in adult humans. Approaching BAT and UCP1 as therapeutic targets to combust surplus energy bares high potential to combat the epidemic of the metabolic syndrome that has precipitated in our society as a result of our modern lifestyles. Our understanding of the physiological and molecular control of BAT may benefit tremendously from consideration of its evolution that basically outlines the blueprint of how to construct a fat burning tissue. Here, we discuss the evolutionary history of UCP1 and BAT, from its origins and emergence to its downfall in several mammalian lineages. Additionally, we delineate the annotation of UCPs in vertebrates by analyzing genomic organization and summarize the phylogeny of UCP1 within the closest relatives of humans, the great apes. Outlining whether the molecular networks controlling thermogenesis in adipose tissue (commonly known as the "browning potential") pre-dated the classical thermogenic function of BAT and UCP1, and whether the evolutionary inactivation of UCP1 enhanced compensatory thermogenic mechanisms, should be of major interest to those who aim to access adipose tissue thermogenesis in a biomedical context.


Asunto(s)
Enfermedad , Evolución Molecular , Termogénesis , Proteína Desacopladora 1/genética , Tejido Adiposo Pardo/metabolismo , Humanos , Especificidad de Órganos/genética
14.
Mol Metab ; 20: 28-37, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30528280

RESUMEN

BACKGROUND/OBJECTIVES: Although the prevalence of obesity and its associated metabolic disorders is increasing in both sexes, the clinical phenotype differs between men and women, highlighting the need for individual treatment options. Mitochondrial dysfunction in various tissues, including white adipose tissue (WAT), has been accepted as a key factor for obesity-associated comorbidities such as diabetes. Given higher expression of mitochondria-related genes in the WAT of women, we hypothesized that gender differences in the bioenergetic profile of white (pre-) adipocytes from obese (age- and BMI-matched) donors must exist. SUBJECTS/METHODS: Using Seahorse technology, we measured oxygen consumption rates (OCR) and extracellular acidification rates (ECAR) of (pre-)adipocytes from male (n = 10) and female (n = 10) deeply-phenotyped obese donors under hypo-, normo- and hyperglycemic (0, 5 and 25 mM glucose) and insulin-stimulated conditions. Additionally, expression levels (mRNA/protein) of mitochondria-related genes (e.g. UQCRC2) and glycolytic enzymes (e.g. PKM2) were determined. RESULTS: Dissecting cellular OCR and ECAR into different functional modules revealed that preadipocytes from female donors show significantly higher mitochondrial to glycolytic activity (higher OCR/ECAR ratio, p = 0.036), which is supported by a higher ratio of UQCRC2 to PKM2 mRNA levels (p = 0.021). However, no major gender differences are detectable in in vitro differentiated adipocytes (e.g. OCR/ECAR, p = 0.248). Importantly, glucose and insulin suppress mitochondrial activity (i.e. ATP-linked respiration) significantly only in preadipocytes of female donors, reflecting their trends towards higher insulin sensitivity. CONCLUSIONS: Collectively, we show that preadipocytes, but not in vitro differentiated adipocytes, represent a model system to reveal gender differences with clinical importance for metabolic disease status. In particular preadipocytes of females maintain enhanced mitochondrial flexibility, as demonstrated by pronounced responses of ATP-linked respiration to glucose.


Asunto(s)
Adipocitos Blancos/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Adulto , Proteínas Portadoras/metabolismo , Células Cultivadas , Complejo III de Transporte de Electrones/metabolismo , Femenino , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Consumo de Oxígeno , Factores Sexuales , Hormonas Tiroideas/metabolismo , Proteínas de Unión a Hormona Tiroide
15.
Endocr Rev ; 38(5): 468-488, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28938407

RESUMEN

Since its identification in 2000, the interest of scientists in the hepatokine fibroblast growth factor (FGF) 21 has tremendously grown, and still remains high, due to a wealth of very robust data documenting this factor's favorable effects on glucose and lipid metabolism in mice. For more than ten years now, intense in vivo and ex vivo experimentation addressed the physiological functions of FGF21 in humans as well as its pathophysiological role and pharmacological effects in human metabolic disease. This work produced a comprehensive collection of data revealing overlaps in FGF21 expression and function but also significant differences between mice and humans that have to be considered before translation from bench to bedside can be successful. This review summarizes what is known about FGF21 in mice and humans with a special focus on this factor's role in glucose and lipid metabolism and in metabolic diseases, such as obesity and type 2 diabetes mellitus. We highlight the discrepancies between mice and humans and try to decipher their underlying reasons.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Tejido Adiposo , Animales , Huesos , Encéfalo , Diabetes Mellitus Tipo 2 , Ejercicio Físico/fisiología , Hígado Graso , Factores de Crecimiento de Fibroblastos/genética , Glucosa/metabolismo , Humanos , Metabolismo de los Lípidos , Enfermedades Metabólicas/fisiopatología , Ratones , Fenómenos Fisiológicos de la Nutrición , Obesidad , Transducción de Señal
16.
Mol Metab ; 6(10): 1226-1239, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29031722

RESUMEN

OBJECTIVE: Obesity-associated WAT inflammation is characterized by the accumulation and local activation of macrophages (MΦs), and recent data from mouse studies suggest that macrophages are modifiers of adipocyte energy metabolism and mitochondrial function. As mitochondrial dysfunction has been associated with obesity and the metabolic syndrome in humans, herein we aimed to delineate how human macrophages may affect energy metabolism of white adipocytes. METHODS: Human adipose tissue gene expression analysis for markers of macrophage activation and tissue inflammation (CD11c, CD40, CD163, CD206, CD80, MCP1, TNFα) in relationship to mitochondrial complex I (NDUFB8) and complex III (UQCRC2) was performed on subcutaneous WAT of 24 women (BMI 20-61 kg/m2). Guided by these results, the impact of secreted factors of LPS/IFNγ- and IL10/TGFß-activated human macrophages (THP1, primary blood-derived) on mitochondrial function in human subcutaneous white adipocytes (SGBS, primary) was determined by extracellular flux analysis (Seahorse technology) and gene/protein expression. RESULTS: Stepwise regression analysis of human WAT gene expression data revealed that a linear combination of CD40 and CD163 was the strongest predictor for mitochondrial complex I (NDUFB8) and complex III (UQCRC2) levels, independent of BMI. IL10/TGFß-activated MΦs displayed high CD163 and low CD40 expression and secreted factors that decreased UQCRC2 gene/protein expression and ATP-linked respiration in human white adipocytes. In contrast, LPS/IFNγ-activated MΦs showed high CD40 and low CD163 expression and secreted factors that enhanced adipocyte mitochondrial activity resulting in a total difference of 37% in ATP-linked respiration of white adipocytes (p = 0.0024) when comparing the effect of LPS/IFNγ- vs IL10/TGFß-activated MΦs. CONCLUSION: Our data demonstrate that macrophages modulate human adipocyte energy metabolism via an activation-dependent paracrine mechanism.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Activación de Macrófagos/fisiología , Mitocondrias/metabolismo , Adipocitos Blancos/metabolismo , Tejido Adiposo Blanco/citología , Adulto , Anciano , Antígenos CD/metabolismo , Citocinas/metabolismo , Metabolismo Energético , Femenino , Humanos , Macrófagos/metabolismo , Persona de Mediana Edad , Obesidad/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
17.
Cell Death Dis ; 7(10): e2412, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27735943

RESUMEN

Tumor necrosis factor-α (TNFα) and other ligands of the TNF superfamily are potent regulators of adipose tissue metabolism and play a crucial role in the obesity-induced inflammation of adipose tissue. Adipose tissue expression levels of TRAIL (TNF-related apoptosis-inducing ligand) and its receptor were shown to be upregulated by overfeeding and decreased by fasting in mice. In the present study we aimed to elucidate the impact of TRAIL on adipogenesis. To this end, human Simpson-Golabi-Behmel syndrome (SGBS) preadipocytes as well as stromal-vascular cells isolated from human white adipose tissue were used as model systems. Human recombinant TRAIL inhibited adipogenic differentiation in a dose-dependent manner. It activated the cleavage of caspase-8 and -3, which in turn resulted in a downregulation of the key adipogenic transcription factors C/EBPα, C/EBPδ, and PPARγ. The effect was completely blocked by pharmacological or genetic inhibition of caspases. Taken together we discovered a so far unrecognized function of TRAIL in the regulation of adipogenesis. Targeting the TRAIL/TRAIL receptor system might provide a novel strategy to interfere with adipose tissue homeostasis.


Asunto(s)
Adipocitos/citología , Adipocitos/enzimología , Adipogénesis/efectos de los fármacos , Caspasas/metabolismo , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Factores de Transcripción/metabolismo , Adipocitos/efectos de los fármacos , Adulto , Arritmias Cardíacas/patología , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Gigantismo/patología , Cardiopatías Congénitas/patología , Humanos , Discapacidad Intelectual/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/patología
18.
Mol Cell Endocrinol ; 382(1): 368-376, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24397922

RESUMEN

Targeting apoptotic pathways in adipocytes has been suggested as a pharmacological approach to treat obesity. However, adipocyte apoptosis was identified as a cause for macrophage infiltration into adipose tissue. Previous studies suggest that mature adipocytes are less sensitive to apoptotic stimuli as compared to preadipocytes. Here, we aimed to identify proteins mediating apoptosis resistance in adipocytes. Our data revealed that the anti-apoptotic protein Bcl-2 (B-cell lymphoma 2) is up-regulated during adipogenic differentiation. Bcl-2 overexpression in preadipocytes lowers their apoptosis sensitivity to the level of mature adipocytes. Vice versa Bcl-2 knockdown in adipocytes sensitizes these cells to CD95-induced apoptosis. Taken together, our findings suggest a shift in the balance of pro-apoptotic and anti-apoptotic molecules during adipogenesis resulting in a higher apoptosis resistance. This study sheds new light on the apoptotic process in human fat cells and may constitute a new possible target for the specific regulation of adipose tissue mass.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis , Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Regulación hacia Arriba , Supervivencia Celular , Técnicas de Silenciamiento del Gen , Humanos , Receptor fas/metabolismo
19.
PLoS One ; 8(2): e57796, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23460908

RESUMEN

AIMS/HYPOTHESIS: The excessive accumulation of adipose tissue in the obese state is linked to an altered secretion profile of adipocytes, chronic low-grade inflammation and metabolic complications. RBP4 has been implicated in these alterations, especially insulin resistance. The aim of the present study was to determine if a local inflammatory micro-environment in adipose tissue regulates RBP4 expression and secretion. METHODS: Human SGBS and primary adipocytes cultured with conditioned media from human THP-1 macrophages were used as an in vitro model for adipose inflammation. Adipocytes were exposed to recombinant TNF-α, IL-1ß, IL-6 or IL-8. In addition, coexpression of IL-1ß and RBP4 was measured in adipose tissue samples from 18 healthy females. RBP4 expression was studied by quantitative PCR and ELISA. RESULTS: RBP4 mRNA expression and secretion was significantly reduced upon incubation with macrophage-conditioned media in SGBS adipocytes and human primary adipocytes. Out of several factors studied we identified IL-1ß as a new factor regulating RBP4. IL-1ß significantly downregulated RBP4 mRNA and secretion in a time- and dose-dependent manner. IL-1ß mediated its inhibitory effects on RBP4 expression via IL-1 receptor and NF-κB, as incubation with the IL-1 receptor blocking antibody and the NF-κB inhibitors CAPE and SC-514 reversed its effect. Most interestingly, RBP4 mRNA was negatively correlated with IL-1ß mRNA in subcutaneous adipose tissue. CONCLUSIONS: Adipose tissue inflammation as found in the obese state might lead to a downregulation in local RBP4 levels. IL-1ß was identified as a major factor contributing to the decrease in RBP4. The increase in circulating RBP4 that often precedes the development of systemic insulin resistance is most likely unrelated to inflammatory processes in adipose tissue.


Asunto(s)
Adipocitos/metabolismo , Regulación hacia Abajo/genética , Interleucina-1beta/metabolismo , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Adipocitos/efectos de los fármacos , Tejido Adiposo/metabolismo , Adulto , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Interleucina-1beta/genética , Interleucina-6/metabolismo , Interleucina-8/metabolismo , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-1/metabolismo , Proteínas Plasmáticas de Unión al Retinol/biosíntesis , Proteínas Plasmáticas de Unión al Retinol/genética , Factores de Tiempo
20.
Artículo en Inglés | MEDLINE | ID: mdl-22645513

RESUMEN

Obesity is associated with an accumulation of macrophages in adipose tissue. This inflammation of adipose tissue is a key event in the pathogenesis of several obesity-related disorders, particularly insulin resistance. Here, we summarized existing model systems that mimic the situation of inflamed adipose tissue in vitro, most of them being murine. Importantly, we introduce our newly established human model system which combines the THP-1 monocytic cell line and the preadipocyte cell strain Simpson-Golabi-Behmel syndrome (SGBS). THP-1 cells, which originate from an acute monocytic leukemia, differentiate easily into macrophages in vitro. The human preadipocyte cell strain SGBS was recently introduced as a unique tool to study human fat cell functions. SGBS cells are characterized by a high capacity for adipogenic differentiation. SGBS adipocytes are capable of fat cell-specific metabolic functions such as insulin-stimulated glucose uptake, insulin-stimulated de novo lipogenesis and ß-adrenergic-stimulated lipolysis and they secrete typical adipokines including leptin, adiponectin, and RBP4. Applying either macrophage-conditioned medium or a direct co-culture of macrophages and fat cells, our model system can be used to distinguish between paracrine and cell-contact dependent effects. In conclusion, we propose this model as a useful tool to study adipose inflammation in vitro. It represents an inexpensive, highly reproducible human system. The methods described here can be easily extended for usage of primary human macrophages and fat cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA