Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Metab ; 79: 101850, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38065435

ABSTRACT

OBJECTIVE: The metabolic benefits of GLP-1 receptor (GLP-1R) agonists on glycemic and weight control are well established as therapy for type 2 diabetes and obesity. Glucagon's ability to increase energy expenditure is well described, and the combination of these mechanisms-of-actions has the potential to further lower hepatic steatosis in metabolic disorders and could therefore be attractive for the treatment for non-alcoholic steatohepatitis (NASH). Here, we have investigated the effects of a dual GLP-1/glucagon receptor agonist NN1177 on hepatic steatosis, fibrosis, and inflammation in a preclinical mouse model of NASH. Having observed strong effects on body weight loss in a pilot study with NN1177, we hypothesized that direct engagement of the hepatic glucagon receptor (GCGR) would result in a superior effect on steatosis and other liver related parameters as compared to the GLP-1R agonist semaglutide at equal body weight. METHODS: Male C57Bl/6 mice were fed a diet high in trans-fat, fructose, and cholesterol (Diet-Induced Obese (DIO)-NASH) for 36 weeks. Following randomization based on the degree of fibrosis at baseline, mice were treated once daily with subcutaneous administration of a vehicle or three different doses of NN1177 or semaglutide for 8 weeks. Hepatic steatosis, inflammation and fibrosis were assessed by immunohistochemistry and morphometric analyses. Plasma levels of lipids and liver enzymes were determined, and hepatic gene expression was analyzed by RNA sequencing. RESULTS: NN1177 dose-dependently reduced body weight up to 22% compared to vehicle treatment. Plasma levels of ALT, a measure of liver injury, were reduced in all treatment groups with body weight loss. The dual agonist reduced hepatic steatosis to a greater extent than semaglutide at equal body weight loss, as demonstrated by three independent methods. Both the co-agonist and semaglutide significantly decreased histological markers of inflammation such as CD11b and Galectin-3, in addition to markers of hepatic stellate activation (αSMA) and fibrosis (Collagen I). Interestingly, the maximal beneficial effects on above mentioned clinically relevant endpoints of NN1177 treatment on hepatic health appear to be achieved with the middle dose tested. Administering the highest dose resulted in a further reduction of liver fat and accompanied by a massive induction in genes involved in oxidative phosphorylation and resulted in exaggerated body weight loss and a downregulation of a module of co-expressed genes involved in steroid hormone biology, bile secretion, and retinol and linoleic acid metabolism that are also downregulated due to NASH itself. CONCLUSIONS: These results indicate that, in a setting of overnutrition, the liver health benefits of activating the fasting-related metabolic pathways controlled by the glucagon receptor displays a bell-shaped curve. This observation is of interest to the scientific community, due to the high number of ongoing clinical trials attempting to leverage the positive effects of glucagon biology to improve metabolic health.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Humans , Male , Animals , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Glucagon , Receptors, Glucagon/genetics , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Pilot Projects , Obesity/metabolism , Body Weight , Diet , Liver Cirrhosis/metabolism , Weight Loss , Glucagon-Like Peptide 1/agonists , Inflammation , Biopsy
2.
J Clin Med ; 10(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807699

ABSTRACT

The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing and approximately 25% of the global population may have NAFLD. NAFLD is associated with obesity and metabolic syndrome, but its pathophysiology is complex and only partly understood. The transsulfuration pathway (TSP) is a metabolic pathway regulating homocysteine and cysteine metabolism and is vital in controlling sulfur balance in the organism. Precise control of this pathway is critical for maintenance of optimal cellular function. The TSP is closely linked to other pathways such as the folate and methionine cycles, hydrogen sulfide (H2S) and glutathione (GSH) production. Impaired activity of the TSP will cause an increase in homocysteine and a decrease in cysteine levels. Homocysteine will also be increased due to impairment of the folate and methionine cycles. The key enzymes of the TSP, cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CSE), are highly expressed in the liver and deficient CBS and CSE expression causes hepatic steatosis, inflammation, and fibrosis in animal models. A causative link between the TSP and NAFLD has not been established. However, dysfunctions in the TSP and related pathways, in terms of enzyme expression and the plasma levels of the metabolites (e.g., homocysteine, cystathionine, and cysteine), have been reported in NAFLD and liver cirrhosis in both animal models and humans. Further investigation of the TSP in relation to NAFLD may reveal mechanisms involved in the development and progression of NAFLD.

3.
Forensic Sci Med Pathol ; 17(2): 322-326, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33847875

ABSTRACT

We report a case of sudden death in a 31-year-old male diagnosed at autopsy with clinical undiagnosed acromegaly. The purpose of this report is to underline the importance of health professionals reacting to phenotypic acromegaly, such as acral enlargement and/or unexplained hypertension, including a range of severe comorbidities, to avoid a fatal outcome. Recent studies have shown that the increased mortality seen in acromegaly patients can be reversed with modern treatment aimed at normalizing GH and IGF-I levels. One year before death, the presented case was diagnosed with hypertension, but was otherwise described as healthy. The forensic autopsy, including post-mortem CT, showed phenotypic facial and body characteristics for acromegaly, general visceromegaly, and a pituitary tumor. The cause of death was heart failure due to end-stage acromegalic cardiopathy. Because the disease is slowly progressive, the individual himself, and the people close to him, might not have considered the acromegaly-related facial changes as abnormal.


Subject(s)
Acromegaly , Heart Failure , Human Growth Hormone , Adult , Death, Sudden/etiology , Humans , Male
4.
Int J Legal Med ; 135(3): 861-870, 2021 May.
Article in English | MEDLINE | ID: mdl-33410922

ABSTRACT

Clinical forensic assessments of injuries' life-threatening danger may have an impact on the legal aftermath following a violent assault. The pursuit of evidence-based guidelines should ensure a user-independent and reproducible forensic practice. However, does it? The aim of this study was to evaluate the forensic life-threatening danger assessments after a protocol implementation in 2016. The evaluation concerned usability and reproducibility of the protocol, and its influence on assessment severity. We analyzed the level of inter- and intra-rater agreement using 169 blinded, prior-protocol cases that were reassessed by two forensic specialists. We compared assessment made the year before and after protocol implementation (n = 262), and the forensic specialists' reassessments with the prior-protocol cases' original assessments (n = 169). Whether to make an assessment, the levels of agreement varied between weak agreement (inter-rater, Κ = 0.43; assessor 1, Κ = 0.57) and strong agreement (assessor 2, Κ = 0.90). Regarding severity, the levels of agreement varied between strong agreement (inter-rater, Κ = 0.87; assessor 1: Κ = 0.90) and almost perfect agreement (assessor 2: Κ = 0.94). The assessments were statistically significant redistributed after the implementation (chi-square test: p < 0.0001). The proportion of cases assessed as having not been in life-threatening danger increased from 9 to 43%, and moderate severity assessments decreased from 55 to 23%. Of the moderate severity assessments, 55% were reassessed as having not been in life-threatening danger. The protocol ensured independent and reproducible assessments when the forensic specialists agreed on making one. The protocol resulted in less severe assessments. Future studies should examine the reliability of the protocol and its consequences for legal aftermaths.


Subject(s)
Forensic Medicine/standards , Practice Guidelines as Topic/standards , Trauma Severity Indices , Wounds, Penetrating/classification , Adolescent , Adult , Aged , Aged, 80 and over , Denmark , Female , Humans , Male , Middle Aged , Observer Variation , Reproducibility of Results , Young Adult
5.
Toxicol Appl Pharmacol ; 399: 115035, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32422327

ABSTRACT

BACKGROUND AND AIMS: Cholecystokinin (CCK) may potentially be used to treat obesity. However, it is well-known to induce acute pancreatitis and pancreas neoplasia in rodents, but not in primates. Here we report the nonclinical safety profile of a long-acting CCK-1 receptor (CCK-1R) agonist, NN9056, in rats and monkeys to support a First-in-Man clinical trial with NN9056. METHODS: Thirteen-week toxicological studies were conducted in rats and non-human primates followed by histopathological evaluation of affected tissues. NN9056 was characterised in vitro, and CCK-1R expression was assessed by in situ hybridization in cynomolgus monkey and human pancreas tissues. RESULTS: Affinity and potency of NN9056 was comparable to native sulphated CCK-8 (CCK-8) across species on the CCK-1R while it had no effect on the CCK-2 receptor (CCK-2R). In situ hybridization demonstrated abundant expression of CCK-1Rs in the exocrine pancreas of the rat. In contrast, it was only discreetly expressed on pancreatic acinar cells in the periphery of scattered lobules in monkeys. A similar expression pattern was observed in human pancreas. 13-weeks daily dosing with NN9056 produced the expected pancreatic pathological findings in rats. In monkeys, NN9056 increased pancreas weight and induced histopathological changes despite the low expression level of CCK-1Rs. CONCLUSION: Surprisingly, chronic CCK-1R activation constitutes a risk for pancreatitis and trophic actions on the exocrine pancreas in monkeys. Since similar CCK-1R expression patterns were found in pancreas of monkeys and humans this risk is likely translatable to humans and clinical development of NN9056 was therefore halted.


Subject(s)
Pancreas, Exocrine/drug effects , Pancreas, Exocrine/pathology , Pancreas/drug effects , Pancreas/pathology , Receptors, Cholecystokinin/agonists , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , COS Cells , Chlorocebus aethiops , Cholecystokinin/metabolism , Humans , Macaca fascicularis , Primates , Rats
6.
Int J Obes (Lond) ; 44(2): 447-456, 2020 02.
Article in English | MEDLINE | ID: mdl-31175319

ABSTRACT

BACKGROUND/OBJECTIVES: Cholecystokinin (CCK) is a regulator of appetite and energy intake in man. The aim of this study was to determine the effect of NN9056, a long-acting CCK-1 receptor-selective CCK analogue, on food intake and body weight (BW) in obese Göttingen Minipigs. SUBJECTS/METHODS: Tolerability of NN9056 and acute effects on food intake, pancreas histology, amylase and lipase levels were assessed in lean domestic pigs in doses up to 100 nmol/kg (n = 3-4). Subsequently, obese Göttingen Minipigs were treated subcutaneously (s.c.) once daily for 13 weeks with vehicle, NN9056 low dose (regulated from 5 to 2 nmol/kg) or NN9056 high dose (10 nmol/kg) (n = 7-8). Food intake was measured daily and BW twice weekly. At the end of the treatment period, an intravenous glucose tolerance test (IVGTT) and a 24-h exposure profile was obtained. Data are mean ± SD. RESULTS: The acute studies in domestic pigs showed significant and dose-dependent effect of NN9056 on food intake, acceptable tolerability and no histopathological signs of pancreatitis. Sub-chronic treatment in obese Göttingen Minipigs was also well tolerated and accumulated food intake was significantly lower in both treated groups compared to vehicle, with no significant difference between the dose levels of NN9056 (41.8 ± 12.6, 51.5 ± 13.8 and 86.5 ± 19.5 kg in high-dose, low-dose and vehicle groups, respectively, p = 0.012 and p < 0.0001 for low and high dose vs. vehicle, respectively). Accordingly, there was a weight loss in both treated groups vs. a weight gain in the vehicle group (-7.2 ± 4.6%, -2.3 ± 3.2% and 12.3 ± 3.9% in the high-dose, low-dose and vehicle groups, respectively, p < 0.0001 for both vs. vehicle). IVGTT data were not significantly different between groups. CONCLUSION: NN9056, a long-acting CCK-1 receptor-selective CCK analogue, significantly reduced food intake and BW in obese Göttingen Minipigs after once daily s.c. dosing for 13 weeks.


Subject(s)
Body Weight/drug effects , Cholecystokinin , Eating/drug effects , Energy Intake/drug effects , Obesity/metabolism , Animals , Cholecystokinin/adverse effects , Cholecystokinin/analogs & derivatives , Cholecystokinin/metabolism , Cholecystokinin/pharmacology , Disease Models, Animal , Female , Humans , Protein Binding , Swine , Swine, Miniature
7.
Forensic Sci Med Pathol ; 15(3): 470-473, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31011986

ABSTRACT

Total parenteral nutrition (TPN) using a central line is an invasive and widely used procedure associated with several complications. Pleural effusion secondary to the leakage of alimentation into the pleural cavity is a rare but encountered complication of central-line TPN administration. This case study focuses on a postmortem examination of an 84-year-old woman, hospitalized due to malnutrition and dysphagia. The left internal jugular vein was cannulated, with no post-procedural x-ray check-up of the catheter tip position. An autopsy revealed 800 ml of milky-white fluid in the left pleural cavity, raising the issue of whether the pleural effusion was of chylous or TPN origin. The aim of this case study was to describe the postmortem diagnostic approach of the fluid analysis. Suspecting chylothorax, we first analyzed the triglyceride (TG) levels in the fluid, which, according to the literature, is the best parameter to detect chyle when lipoprotein electrophoresis (chylomicron %) is unavailable. Biochemistry showed increased levels of TG, which can be found in both chylous and TPN fluid. We then added glucose and potassium to the biochemical analysis, again showing increased levels at 46 mmol/L and 22 mmol/L, respectively. We had no information about the given TPN, and thus, comparing the final chemical results with the TPN composition was impossible. The presence of increased levels of triglycerides, glucose, and potassium in the white fluid more strongly resembled a standard TPN than chyle. Hence, by using these three measurements, we concluded that the milky-white fluid was a leakage of TPN.


Subject(s)
Catheterization, Central Venous/adverse effects , Parenteral Nutrition, Total/adverse effects , Pleural Effusion/pathology , Aged, 80 and over , Chylothorax , Diagnosis, Differential , Fatal Outcome , Female , Humans , Jugular Veins , Pleural Effusion/diagnostic imaging , Tomography, X-Ray Computed
8.
Eur J Pharmacol ; 833: 357-363, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29920282

ABSTRACT

The gastrointestinal hormone cholecystokinin (CCK) regulates digestive processes and satiety in addition to centrally mediated effects on nociception and anxiety. CCK signals through two seven-trans-membrane receptors named the CCK-1 receptor and the CCK-2 receptor. The expression pattern and biological effects mediated by the CCK-1 and CCK-2 receptors are highly divergent. The pig is a widely used preclinical animal model in medical research, but up until recently, the porcine CCK-2 receptor was described as a pseudogene in the publicly available genomic sequence databases. Thus, it was challenging to interpret data from this animal model in studies of CCK biology and pharmacology. Here we describe an in silico prediction of the porcine CCK-2 receptor and the subsequent cloning, expression, and in vitro pharmacological characterization. We find a high degree of sequence homology with the human orthologue as well as CCK-2 receptors of other major species used in pre-clinical research. We also show that the endogenous ligands CCK-8 and Gastrin-17 bind and activate the porcine CCK-2 receptor with similar affinities and potencies as seen for the human CCK-2 receptor. We conclude that the pig has a functional CCK-2 receptor which is highly comparable to the human orthologue and therefore the pig qualifies as a valid preclinical model for the study of human CCK biology and pharmacology.


Subject(s)
Cholecystokinin/physiology , Models, Animal , Receptor, Cholecystokinin B/metabolism , Swine , Animals , COS Cells , Chlorocebus aethiops , Cholecystokinin/agonists , Computational Biology , Computer Simulation , Female , Gastrins/metabolism , Protein Structure, Secondary , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/isolation & purification , Sequence Homology, Amino Acid , Sincalide/metabolism
9.
Diabetol Metab Syndr ; 10: 36, 2018.
Article in English | MEDLINE | ID: mdl-29713389

ABSTRACT

BACKGROUND: Individuals who suffer from mental illness are more prone to obesity and related co-morbidities, including the metabolic syndrome. Autopsies provide an outstanding platform for the macroscopic, microscopic and molecular-biological investigation of diseases. Autopsy-based findings may assist in the investigation of the metabolic syndrome. To utilise the vast information that an autopsy encompasses to elucidate the pathophysiology behind the syndrome further, we aimed to both develop and evaluate a method for the post mortem definition of the metabolic syndrome. METHODS: Based on the nationwide Danish SURVIVE study of deceased mentally ill, we established a set of post mortem criteria for each of the harmonized criteria of the metabolic syndrome. We based the post mortem (PM) evaluation on information from the police reports and the data collected at autopsy, such as anthropometric measurements and biochemical and toxicological analyses (PM information). We compared our PM evaluation with the data from the Danish health registries [ante mortem (AM) information, considered the gold standard] from each individual. RESULTS: The study included 443 deceased individuals (272 male and 171 female) with a mean age of 50.4 (± 15.5) years and a median (interquartile range) post mortem interval of 114 (84-156) hours. We found no significant difference when defining the metabolic syndrome from the PM information in comparison to the AM information (P = 0.175). The PM evaluation yielded a high specificity (0.93) and a moderate sensitivity (0.63) with a moderate level of agreement compared to the AM evaluation (Cohen's κ = 0.51). Neither age nor post mortem interval affected the final results. CONCLUSIONS: Our model of a PM definition of the metabolic syndrome proved reliable when compared to the AM information. We believe that an appropriate estimate of the prevalence of the metabolic syndrome can be established post mortem. However, while neither the PM nor the AM information is exhaustive in terms of defining an individual's health status, a superlative estimate may be obtained by combining the PM and the AM information. With this model, we open up the possibility of utilising autopsy data for future studies of the metabolic syndrome.

10.
Nat Med ; 23(10): 1158-1166, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28846099

ABSTRACT

Growth differentiation factor 15 (GDF15; also known as MIC-1) is a divergent member of the TGF-ß superfamily and is associated with body-weight regulation in humans and rodents. However, the cognate receptor of GDF15 is unknown. Here we show that GDF15 binds specifically to GDNF family receptor α-like (GFRAL) with high affinity, and that GFRAL requires association with the coreceptor RET to elicit intracellular signaling in response to GDF15 stimulation. We also found that GDF15-mediated reductions in food intake and body weight of mice with obesity were abolished in GFRAL-knockout mice. We further found that GFRAL expression was limited to hindbrain neurons and not present in peripheral tissues, which suggests that GDF15-GFRAL-mediated regulation of food intake is by a central mechanism. Lastly, given that GDF15 did not increase energy expenditure in treated mice with obesity, the anti-obesity actions of the cytokine are likely driven primarily by a reduction in food intake.


Subject(s)
Eating/drug effects , Energy Metabolism/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/drug effects , Growth Differentiation Factor 15/pharmacology , Obesity/metabolism , Weight Loss/drug effects , Animals , Eating/genetics , Energy Metabolism/genetics , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Surface Plasmon Resonance , Weight Loss/genetics
11.
Biochimie ; 136: 100-104, 2017 May.
Article in English | MEDLINE | ID: mdl-27916640

ABSTRACT

Over the last couple of decades, the PPAR family of transcription factors has received much attention from the pharmaceutical industry due to their profound ability to improve glucose and lipid metabolism upon agonist activation. However, more recently the interest in these nuclear receptors has faded because several clinical trials have shown that it is difficult to develop a ligand that significantly ameliorates glucose and lipid metabolism disorders without concomitantly inducing unacceptable side-effects. Nevertheless, the data also suggests that tissue specific targeting could pave the way to renewed interest and clinical use of PPAR ligands. In this review we summarize the results and learnings from the clinical trials on PPAR agonism and discuss the possibilities for tissue targeting of PPAR ligands by using state of the art technology to fuse them to peptides homing selectively to tissues expressing the cognate surface receptor.


Subject(s)
Peroxisome Proliferator-Activated Receptors/agonists , Animals , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
12.
Methods Enzymol ; 538: 233-47, 2014.
Article in English | MEDLINE | ID: mdl-24529442

ABSTRACT

The realization that obesity and its associated diseases have become one of modern society's major challenges to the health of the world's population has fueled much effort to understand white adipocyte biology and elucidate pathways to increase energy expenditure. One strategy has been to increase the oxidative capacity and activity of the adipocytes themselves. This has the advantage that free fatty acids (FAs) would not be released into the circulation in copious amounts, which can have detrimental effects. This is particularly true for obese individuals, who often already display severe dyslipidemia, putting them at increased risk for cardiovascular diseases. It was recently discovered that adult humans, in addition to infants, possess active brown adipocytes, characterized by expression of the mitochondrial electron gradient dissipater uncoupling protein 1 (UCP1). This has generated renewed interest in finding ways to "convert" or "adapt" white adipocytes into a more brown adipocyte-like state by increasing mitochondrial content and expression of UCP1 and activating UCP1 via lipolysis-mediated free FAs. Another approach to consider is elevating the activity of the not insignificant amount of mitochondria found in white adipocytes. The invention of the XF Flux Analyzer by Seahorse Bioscience has revolutionized this line of research as it allows for real-time measurements of respiration in multiple samples simultaneously. In this chapter, we describe our approaches and experience with employing this technology to study the metabolism of mouse and human primary and immortalized cells and mouse white adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Oxygen Consumption , Animals , Cell Line , Cell Respiration , Cells, Cultured , Energy Metabolism , Humans , Ion Channels/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Uncoupling Protein 1
13.
J Lipid Res ; 55(2): 247-57, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24259533

ABSTRACT

Liver X receptors (LXRs) are transcription factors known for their role in hepatic cholesterol and lipid metabolism. Though highly expressed in fat, the role of LXR in this tissue is not well characterized. We generated adipose tissue LXRα knockout (ATaKO) mice and showed that these mice gain more weight and fat mass on a high-fat diet compared with wild-type controls. White adipose tissue (WAT) accretion in ATaKO mice results from both a decrease in WAT lipolytic and oxidative capacities. This was demonstrated by decreased expression of the ß2- and ß3-adrenergic receptors, reduced level of phosphorylated hormone-sensitive lipase, and lower oxygen consumption rates (OCRs) in WAT of ATaKO mice. Furthermore, LXR activation in vivo and in vitro led to decreased adipocyte size in WAT and increased glycerol release from primary adipocytes, respectively, with a concomitant increase in OCR in both models. Our findings show that absence of LXRα in adipose tissue results in elevated adiposity through a decrease in WAT oxidation, secondary to attenuated FA availability.


Subject(s)
Adipocytes, White/metabolism , Fatty Acids/metabolism , Lipolysis , Orphan Nuclear Receptors/metabolism , Oxygen Consumption , Adipocytes, White/cytology , Adipocytes, White/drug effects , Animals , Body Weight/drug effects , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Hydrocarbons, Fluorinated/pharmacology , Lipolysis/drug effects , Liver X Receptors , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/metabolism , Obesity/pathology , Orphan Nuclear Receptors/deficiency , Orphan Nuclear Receptors/genetics , Oxidation-Reduction , Oxygen Consumption/drug effects , Phenotype , Receptors, Adrenergic, beta/metabolism , Sulfonamides/pharmacology
14.
Nature ; 503(7476): 410-413, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24162845

ABSTRACT

Circadian oscillation of body temperature is a basic, evolutionarily conserved feature of mammalian biology. In addition, homeostatic pathways allow organisms to protect their core temperatures in response to cold exposure. However, the mechanism responsible for coordinating daily body temperature rhythm and adaptability to environmental challenges is unknown. Here we show that the nuclear receptor Rev-erbα (also known as Nr1d1), a powerful transcriptional repressor, links circadian and thermogenic networks through the regulation of brown adipose tissue (BAT) function. Mice exposed to cold fare considerably better at 05:00 (Zeitgeber time 22) when Rev-erbα is barely expressed than at 17:00 (Zeitgeber time 10) when Rev-erbα is abundant. Deletion of Rev-erbα markedly improves cold tolerance at 17:00, indicating that overcoming Rev-erbα-dependent repression is a fundamental feature of the thermogenic response to cold. Physiological induction of uncoupling protein 1 (Ucp1) by cold temperatures is preceded by rapid downregulation of Rev-erbα in BAT. Rev-erbα represses Ucp1 in a brown-adipose-cell-autonomous manner and BAT Ucp1 levels are high in Rev-erbα-null mice, even at thermoneutrality. Genetic loss of Rev-erbα also abolishes normal rhythms of body temperature and BAT activity. Thus, Rev-erbα acts as a thermogenic focal point required for establishing and maintaining body temperature rhythm in a manner that is adaptable to environmental demands.


Subject(s)
Body Temperature Regulation/physiology , Circadian Rhythm/physiology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Acclimatization/genetics , Acclimatization/physiology , Adipose Tissue, Brown/metabolism , Animals , Body Temperature Regulation/genetics , Circadian Rhythm/genetics , Cold Temperature , Down-Regulation , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Thermogenesis/genetics , Thermogenesis/physiology , Time Factors , Uncoupling Protein 1
15.
Genes Dev ; 26(7): 657-67, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22474260

ABSTRACT

The nuclear receptor Rev-erbα regulates circadian rhythm and metabolism, but its effects are modest and it has been considered to be a secondary regulator of the cell-autonomous clock. Here we report that depletion of Rev-erbα together with closely related Rev-erbß has dramatic effects on the cell-autonomous clock as well as hepatic lipid metabolism. Mouse embryonic fibroblasts were rendered arrhythmic by depletion of both Rev-erbs. In mouse livers, Rev-erbß mRNA and protein levels oscillate with a diurnal pattern similar to that of Rev-erbα, and both Rev-erbs are recruited to a remarkably similar set of binding sites across the genome, enriched near metabolic genes. Depletion of both Rev-erbs in liver synergistically derepresses several metabolic genes as well as genes that control the positive limb of the molecular clock. Moreover, deficiency of both Rev-erbs causes marked hepatic steatosis, in contrast to relatively subtle changes upon loss of either subtype alone. These findings establish the two Rev-erbs as major regulators of both clock function and metabolism, displaying a level of subtype collaboration that is unusual among nuclear receptors but common among core clock proteins, protecting the organism from major perturbations in circadian and metabolic physiology.


Subject(s)
Circadian Rhythm , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Animals , Cells, Cultured , Gene Expression Regulation , Genome , Histone Deacetylases/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/deficiency , Repressor Proteins/metabolism
16.
Science ; 331(6022): 1315-9, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21393543

ABSTRACT

Disruption of the circadian clock exacerbates metabolic diseases, including obesity and diabetes. We show that histone deacetylase 3 (HDAC3) recruitment to the genome displays a circadian rhythm in mouse liver. Histone acetylation is inversely related to HDAC3 binding, and this rhythm is lost when HDAC3 is absent. Although amounts of HDAC3 are constant, its genomic recruitment in liver corresponds to the expression pattern of the circadian nuclear receptor Rev-erbα. Rev-erbα colocalizes with HDAC3 near genes regulating lipid metabolism, and deletion of HDAC3 or Rev-erbα in mouse liver causes hepatic steatosis. Thus, genomic recruitment of HDAC3 by Rev-erbα directs a circadian rhythm of histone acetylation and gene expression required for normal hepatic lipid homeostasis.


Subject(s)
Circadian Clocks , Circadian Rhythm , Fatty Liver/metabolism , Genome , Histone Deacetylases/metabolism , Lipid Metabolism , Liver/metabolism , Animals , Binding Sites , Chromatin Immunoprecipitation , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , DNA/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Histones/metabolism , Homeostasis , Lipogenesis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , RNA Polymerase II/metabolism , Up-Regulation
17.
PPAR Res ; 20102010.
Article in English | MEDLINE | ID: mdl-20862367

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) are central regulators of fat metabolism, energy homeostasis, proliferation, and inflammation. The three PPAR subtypes, PPARα, ß/δ, and γ activate overlapping but also very different target gene programs. This review summarizes the insights into PPAR subtype-specific transactivation provided by genome-wide studies and discusses the recent advances in the understanding of the molecular mechanisms underlying PPAR subtype specificity with special focus on the regulatory role of AF-1.

18.
J Mol Endocrinol ; 45(4): 219-28, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20639404

ABSTRACT

Epigenetic deregulation contributes to diseases including cancer, neurodegeneration, osteodystrophy, cardiovascular defects, and obesity. For this reason, several inhibitors for histone deacetylases (HDACs) are being validated as novel anti-cancer drugs in clinical studies and display important anti-proliferative activities. While most inhibitors act on both class I, II, and IV HDACs, evidence is accumulating that class I is directly involved in regulation of cell growth and death, whereas class II members regulate differentiation processes, such as muscle and neuronal differentiation. Here, we show that the novel class II-selective inhibitor MC1568 interferes with the RAR- and peroxisome proliferator-activated receptor γ (PPARγ)-mediated differentiation-inducing signaling pathways. In F9 cells, this inhibitor specifically blocks endodermal differentiation despite not affecting retinoic acid-induced maturation of promyelocytic NB4 cells. In 3T3-L1 cells, MC1568 attenuates PPARγ-induced adipogenesis, while the class I-selective MS275 blocked adipogenesis completely thus revealing a different mode of action and/or target profile of the two classes of HDACs. Using in vivo reporting PPRE-Luc mice, we find that MC1568 impairs PPARγ signaling mostly in the heart and adipose tissues. These results illustrate how HDAC functions can be dissected by selective inhibitors.


Subject(s)
Cell Differentiation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Adipogenesis/drug effects , Animals , Biomarkers/metabolism , Cell Line , Humans , Mice , Organ Specificity/drug effects , PPAR gamma/metabolism , Signal Transduction/drug effects , Tretinoin/pharmacology
19.
J Biol Chem ; 285(23): 17310-7, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20360005

ABSTRACT

Uncoupling Proteins (UCPs) are integral ion channels residing in the inner mitochondrial membrane. UCP2 is ubiquitously expressed, while UCP3 is found primarily in muscles and adipose tissue. Although the exact molecular mechanism of action is controversial, it is generally agreed that both homologues function to facilitate mitochondrial fatty acid oxidation. UCP2 and -3 expression is activated by the peroxisome proliferator-activated receptors (PPARs), but so far no PPAR response element has been reported in the vicinity of the Ucp2 and Ucp3 genes. Using genome-wide profiling of PPARgamma occupancy in 3T3-L1 adipocytes we demonstrate that PPARgamma associates with three chromosomal regions in the vicinity of the Ucp3 locus and weakly with a site in intron 1 of the Ucp2 gene. These sites are isolated from the nearest neighboring sites by >900 kb. The most prominent PPARgamma binding site in the Ucp2 and Ucp3 loci is located in intron 1 of the Ucp3 gene and is the only site that facilitates PPARgamma transactivation of a heterologous promoter. This site furthermore transactivates the endogenous Ucp3 promoter, and using chromatin conformation capture we show that it loops out to specifically interact with the Ucp2 promoter and intron 1. Our data indicate that PPARgamma transactivation of both UCP2 and -3 is mediated through this novel enhancer in Ucp3 intron 1.


Subject(s)
Adipocytes/metabolism , Enhancer Elements, Genetic , Introns , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , PPAR gamma/genetics , Transcriptional Activation , 3T3-L1 Cells , Adipocytes/cytology , Animals , Chromatin/chemistry , Humans , Mice , PPAR gamma/metabolism , Promoter Regions, Genetic , Protein Binding , Retroviridae/genetics , Uncoupling Protein 2 , Uncoupling Protein 3
20.
J Biol Chem ; 284(39): 26385-93, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19633298

ABSTRACT

The ligand-inducible nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) plays a key role in the differentiation, maintenance, and function of adipocytes and is the molecular target for the insulin-sensitizing thiazoledinediones (TZDs). Although a number of PPARgamma target genes that may contribute to the reduction of circulating free fatty acids after TZD treatment have been identified, the relevant PPARgamma target genes that may exert the anti-lipolytic effect of TZDs are unknown. Here we identified the anti-lipolytic human G-protein-coupled receptor 81 (GPR81), GPR109A, and the (human-specific) GPR109B genes as well as the mouse Gpr81 and Gpr109A genes as novel TZD-induced genes in mature adipocytes. GPR81/Gpr81 is a direct PPARgamma target gene, because mRNA expression of GPR81/Gpr81 (and GPR109A/Gpr109A) increased in mature human and murine adipocytes as well as in vivo in epididymal fat pads of mice upon rosiglitazone stimulation, whereas small interfering RNA-mediated knockdown of PPARgamma in differentiated 3T3-L1 adipocytes showed a significant decrease in Gpr81 protein expression. In addition, chromatin immunoprecipitation sequencing analysis in differentiated 3T3-L1 cells revealed a conserved PPAR:retinoid X receptor-binding site in the proximal promoter of the Gpr81 gene, which was proven to be functional by electromobility shift assay and reporter assays. Importantly, small interfering RNA-mediated knockdown of Gpr81 partly reversed the inhibitory effect of TZDs on lipolysis in 3T3-L1 adipocytes. The coordinated PPARgamma-mediated regulation of the GPR81/Gpr81 and GPR109A/Gpr109A genes (and GPR109B in humans) presents a novel mechanism by which TZDs may reduce circulating free fatty acid levels and perhaps ameliorate insulin resistance in obese patients.


Subject(s)
Adipocytes/metabolism , Gene Expression Regulation , PPAR gamma/genetics , Receptors, G-Protein-Coupled/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Base Sequence , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Humans , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , PPAR gamma/agonists , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Sequence Homology, Nucleic Acid , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...