87 results on '"Tailleux, Anne"'
Search Results
2. Endothelial Dysfunction and Pre-Existing Cognitive Disorders in Stroke Patients.
- Author
-
Mendyk-Bordet AM, Ouk T, Muhr-Tailleux A, Pétrault M, Vallez E, Gelé P, Dondaine T, Labreuche J, Deplanque D, and Bordet R
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Prospective Studies, Cognitive Dysfunction physiopathology, Biomarkers blood, Magnetic Resonance Imaging, Risk Factors, Stroke complications, Stroke physiopathology, Endothelium, Vascular physiopathology
- Abstract
Background: The origin of pre-existing cognitive impairment in stroke patients remains controversial, with a vascular or a degenerative hypothesis., Objective: To determine whether endothelial dysfunction is associated with pre-existing cognitive problems, lesion load and biological anomalies in stroke patients., Methods: Patients originated from the prospective STROKDEM study. The baseline cognitive state, assessed using the IQ-CODE, and risk factors for stroke were recorded at inclusion. Patients with an IQ-CODE score >64 were excluded. Endothelial function was determined 72 h after stroke symptom onset by non-invasive digital measurement of endothelium-dependent flow-mediated dilation and calculation of the reactive hyperemia index (RHI). RHI ≤ 1.67 indicated endothelial dysfunction. Different biomarkers of endothelial dysfunction were analysed in blood or plasma. All patients underwent MRI 72 h after stroke symptom onset., Results: A total of 86 patients were included (52 males; mean age 63.5 ± 11.5 years). Patients with abnormal RHI have hypertension or antihypertensive treatment more often. The baseline IQ-CODE was abnormal in 33 (38.4%) patients, indicating a pre-existing cognitive problem. Baseline IQ-CODE > 48 was observed in 15 patients (28.3%) with normal RHI and in 18 patients (54.6%) with abnormal RHI ( p = 0.016). The RHI median was significantly lower in patients with abnormal IQ-CODE. Abnormal RHI was associated with a significantly higher median FAZEKAS score (2.5 vs. 2; p = 0.008), a significantly higher frequency of periventricular lesions ( p = 0.015), more white matter lesions ( p = 0.007) and a significantly higher cerebral atrophy score ( p < 0.001) on MRI. Vascular biomarkers significantly associated with abnormal RHI were MCP-1 ( p = 0.009), MIP_1a ( p = 0.042), and homocysteinemia ( p < 0.05)., Conclusions: A vascular mechanism may be responsible for cognitive problems pre-existing stroke. The measurement of endothelial dysfunction after stroke could become an important element of follow-up, providing an indication of the functional and cognitive prognosis of stroke patients.
- Published
- 2024
- Full Text
- View/download PDF
3. Multi-Omics Data Integration Reveals Sex-Dependent Hippocampal Programming by Maternal High-Fat Diet during Lactation in Adult Mouse Offspring.
- Author
-
Gauvrit T, Benderradji H, Pelletier A, Aboulouard S, Faivre E, Carvalho K, Deleau A, Vallez E, Launay A, Bogdanova A, Besegher M, Le Gras S, Tailleux A, Salzet M, Buée L, Delahaye F, Blum D, and Vieau D
- Subjects
- Animals, Mice, Female, Male, Humans, Obesity etiology, Obesity metabolism, Multiomics, Proteomics, Lactation, Hippocampus metabolism, Maternal Nutritional Physiological Phenomena physiology, Diet, High-Fat adverse effects, Prenatal Exposure Delayed Effects metabolism
- Abstract
Early-life exposure to high-fat diets (HF) can program metabolic and cognitive alterations in adult offspring. Although the hippocampus plays a crucial role in memory and metabolic homeostasis, few studies have reported the impact of maternal HF on this structure. We assessed the effects of maternal HF during lactation on physiological, metabolic, and cognitive parameters in young adult offspring mice. To identify early-programming mechanisms in the hippocampus, we developed a multi-omics strategy in male and female offspring. Maternal HF induced a transient increased body weight at weaning, and a mild glucose intolerance only in 3-month-old male mice with no change in plasma metabolic parameters in adult male and female offspring. Behavioral alterations revealed by a Barnes maze test were observed both in 6-month-old male and female mice. The multi-omics strategy unveiled sex-specific transcriptomic and proteomic modifications in the hippocampus of adult offspring. These studies that were confirmed by regulon analysis show that, although genes whose expression was modified by maternal HF were different between sexes, the main pathways affected were similar with mitochondria and synapses as main hippocampal targets of maternal HF. The effects of maternal HF reported here may help to better characterize sex-dependent molecular pathways involved in cognitive disorders and neurodegenerative diseases.
- Published
- 2023
- Full Text
- View/download PDF
4. Roux-en-Y gastric bypass induces hepatic transcriptomic signatures and plasma metabolite changes indicative of improved cholesterol homeostasis.
- Author
-
Lalloyer F, Mogilenko DA, Verrijken A, Haas JT, Lamazière A, Kouach M, Descat A, Caron S, Vallez E, Derudas B, Gheeraert C, Baugé E, Despres G, Dirinck E, Tailleux A, Dombrowicz D, Van Gaal L, Eeckhoute J, Lefebvre P, Goossens JF, Francque S, and Staels B
- Subjects
- Humans, Transcriptome, Obesity complications, Cholesterol, Homeostasis, Inflammation complications, Gastric Bypass methods, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease surgery, Obesity, Morbid complications
- Abstract
Background & Aims: Roux-en-Y gastric bypass (RYGB), the most effective surgical procedure for weight loss, decreases obesity and ameliorates comorbidities, such as non-alcoholic fatty liver (NAFLD) and cardiovascular (CVD) diseases. Cholesterol is a major CVD risk factor and modulator of NAFLD development, and the liver tightly controls its metabolism. How RYGB surgery modulates systemic and hepatic cholesterol metabolism is still unclear., Methods: We studied the hepatic transcriptome of 26 patients with obesity but not diabetes before and 1 year after undergoing RYGB. In parallel, we measured quantitative changes in plasma cholesterol metabolites and bile acids (BAs)., Results: RYGB surgery improved systemic cholesterol metabolism and increased plasma total and primary BA levels. Transcriptomic analysis revealed specific alterations in the liver after RYGB, with the downregulation of a module of genes implicated in inflammation and the upregulation of three modules, one associated with BA metabolism. A dedicated analysis of hepatic genes related to cholesterol homeostasis pointed towards increased biliary cholesterol elimination after RYGB, associated with enhancement of the alternate, but not the classical, BA synthesis pathway. In parallel, alterations in the expression of genes involved in cholesterol uptake and intracellular trafficking indicate improved hepatic free cholesterol handling. Finally, RYGB decreased plasma markers of cholesterol synthesis, which correlated with an improvement in liver disease status after surgery., Conclusions: Our results identify specific regulatory effects of RYGB on inflammation and cholesterol metabolism. RYGB alters the hepatic transcriptome signature, likely improving liver cholesterol homeostasis. These gene regulatory effects are reflected by systemic post-surgery changes of cholesterol-related metabolites, corroborating the beneficial effects of RYGB on both hepatic and systemic cholesterol homeostasis., Impact and Implications: Roux-en-Y gastric bypass (RYGB) is a widely used bariatric surgery procedure with proven efficacy in body weight management, combatting cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). RYGB exerts many beneficial metabolic effects, by lowering plasma cholesterol and improving atherogenic dyslipidemia. Using a cohort of patients undergoing RYGB, studied before and 1 year after surgery, we analyzed how RYGB modulates hepatic and systemic cholesterol and bile acid metabolism. The results of our study provide important insights on the regulation of cholesterol homeostasis after RYGB and open avenues that could guide future monitoring and treatment strategies targeting CVD and NAFLD in obesity., (Copyright © 2023 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. Discovery, Structure-Activity Relationships, and In Vivo Activity of Dihydropyridone Agonists of the Bile Acid Receptor TGR5.
- Author
-
Picon S, Boulahjar R, Hoguet V, Baron M, Duplan I, Vallez E, Hennuyer N, Dumont J, Touche V, Dorchies E, Lasalle M, Descat A, Piveteau C, Biela A, Chaput L, Villoutreix BO, Lipka E, Sevin E, Culot M, Gosselet F, Lestavel S, Roussel P, Deprez-Poulain R, Leroux F, Staels B, Deprez B, Tailleux A, and Charton J
- Subjects
- Animals, Mice, Glucagon-Like Peptide 1, Bile Acids and Salts, Quantitative Structure-Activity Relationship, Transcription Factors
- Abstract
A novel series of potent agonists of the bile acid receptor TGR5 bearing a dihydropyridone scaffold was developed from a high-throughput screen. Starting from a micromolar hit compound, we implemented an extensive structure-activity-relationship (SAR) study with the synthesis and biological evaluation of 83 analogues. The project culminated with the identification of the potent nanomolar TGR5 agonist 77A . We report the GLP-1 secretagogue effect of our lead compound ex vivo in mouse colonoids and in vivo. In addition, to identify specific features favorable for TGR5 activation, we generated and optimized a three-dimensional quantitative SAR model that contributed to our understanding of our activity profile and could guide further development of this dihydropyridone series.
- Published
- 2023
- Full Text
- View/download PDF
6. Apolipoprotein F is reduced in humans with steatosis and controls plasma triglyceride-rich lipoprotein metabolism.
- Author
-
Deprince A, Hennuyer N, Kooijman S, Pronk ACM, Baugé E, Lienard V, Verrijken A, Dirinck E, Vonghia L, Woitrain E, Kloosterhuis NJ, Marez E, Jacquemain P, Wolters JC, Lalloyer F, Eberlé D, Quemener S, Vallez E, Tailleux A, Kouach M, Goossens JF, Raverdy V, Derudas B, Kuivenhoven JA, Croyal M, van de Sluis B, Francque S, Pattou F, Rensen PCN, Staels B, and Haas JT
- Subjects
- Mice, Animals, Humans, Lipoproteins metabolism, Apolipoproteins metabolism, Apolipoproteins pharmacology, Triglycerides metabolism, Liver metabolism, Lipoproteins, VLDL metabolism, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Background: NAFLD affects nearly 25% of the global population. Cardiovascular disease (CVD) is the most common cause of death among patients with NAFLD, in line with highly prevalent dyslipidemia in this population. Increased plasma triglyceride (TG)-rich lipoprotein (TRL) concentrations, an important risk factor for CVD, are closely linked with hepatic TG content. Therefore, it is of great interest to identify regulatory mechanisms of hepatic TRL production and remnant uptake in the setting of hepatic steatosis., Approach and Results: To identify liver-regulated pathways linking intrahepatic and plasma TG metabolism, we performed transcriptomic analysis of liver biopsies from two independent cohorts of obese patients. Hepatic encoding apolipoprotein F ( APOF ) expression showed the fourth-strongest negatively correlation with hepatic steatosis and the strongest negative correlation with plasma TG levels. The effects of adenoviral-mediated human ApoF (hApoF) overexpression on plasma and hepatic TG were assessed in C57BL6/J mice. Surprisingly, hApoF overexpression increased both hepatic very low density lipoprotein (VLDL)-TG secretion and hepatic lipoprotein remnant clearance, associated a ~25% reduction in plasma TG levels. Conversely, reducing endogenous ApoF expression reduced VLDL secretion in vivo , and reduced hepatocyte VLDL uptake by ~15% in vitro . Transcriptomic analysis of APOF -overexpressing mouse livers revealed a gene signature related to enhanced ApoB-lipoprotein clearance, including increased expression of Ldlr and Lrp1 , among others., Conclusion: These data reveal a previously undescribed role for ApoF in the control of plasma and hepatic lipoprotein metabolism by favoring VLDL-TG secretion and hepatic lipoprotein remnant particle clearance., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
7. Adipocyte-specific FXR-deficiency protects adipose tissue from oxidative stress and insulin resistance and improves glucose homeostasis.
- Author
-
Dehondt H, Marino A, Butruille L, Mogilenko DA, Nzoussi Loubota AC, Chávez-Talavera O, Dorchies E, Vallez E, Haas J, Derudas B, Bongiovanni A, Tardivel M, Kuipers F, Lefebvre P, Lestavel S, Tailleux A, Dombrowicz D, Caron S, and Staels B
- Subjects
- Animals, Mice, Adipocytes metabolism, Adipose Tissue metabolism, Glucose metabolism, Homeostasis, Inflammation metabolism, Oxidative Stress, Receptors, Cytoplasmic and Nuclear metabolism, Insulin Resistance physiology
- Abstract
Objective: Obesity is associated with metabolic dysfunction of white adipose tissue (WAT). Activated adipocytes secrete pro-inflammatory cytokines resulting in the recruitment of pro-inflammatory macrophages, which contribute to WAT insulin resistance. The bile acid (BA)-activated nuclear Farnesoid X Receptor (FXR) controls systemic glucose and lipid metabolism. Here, we studied the role of FXR in adipose tissue function., Methods: We first investigated the immune phenotype of epididymal WAT (eWAT) from high fat diet (HFD)-fed whole-body FXR-deficient (FXR
-/- ) mice by flow cytometry and gene expression analysis. We then generated adipocyte-specific FXR-deficient (Ad-FXR-/- ) mice and analyzed systemic and eWAT metabolism and immune phenotype upon HFD feeding. Transcriptomic analysis was done on mature eWAT adipocytes from HFD-fed Ad-FXR-/- mice., Results: eWAT from HFD-fed whole-body FXR-/- and Ad-FXR-/- mice displayed decreased pro-inflammatory macrophage infiltration and inflammation. Ad-FXR-/- mice showed lower blood glucose concentrations, improved systemic glucose tolerance and WAT insulin sensitivity and oxidative stress. Transcriptomic analysis identified Gsta4, a modulator of oxidative stress in WAT, as the most upregulated gene in Ad-FXR-/- mouse adipocytes. Finally, chromatin immunoprecipitation analysis showed that FXR binds the Gsta4 gene promoter., Conclusions: These results indicate a role for the adipocyte FXR-GSTA4 axis in controlling HFD-induced inflammation and systemic glucose homeostasis., (Copyright © 2023 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
8. Enterohepatic Takeda G-Protein Coupled Receptor 5 Agonism in Metabolic Dysfunction-Associated Fatty Liver Disease and Related Glucose Dysmetabolism.
- Author
-
Gillard J, Picalausa C, Ullmer C, Adorini L, Staels B, Tailleux A, and Leclercq IA
- Subjects
- Animals, Glucose therapeutic use, Mice, Mice, Inbred C57BL, Receptors, G-Protein-Coupled metabolism, Bile Acids and Salts, Non-alcoholic Fatty Liver Disease drug therapy, Non-alcoholic Fatty Liver Disease etiology, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a major health concern with no approved pharmacological therapies. Molecules developed to activate the bile acid-receptor TGR5 regulate pathways involved in MALFD pathogenesis, but the therapeutic value of TGR5 activation on the active form of MAFLD, non-alcoholic steatohepatitis (NASH), still needs to be evaluated. As TGR5 agonism is low in MAFLD, we used strategies to promote the production of endogenous TGR5 ligands or administered pharmacological TGR5 agonists, INT-777 and RO5527239, to study the effect of TGR5 activation on liver and metabolic diseases in high-fat diet-fed foz/foz mice. Although described in the literature, treatment with fexaramine, an intestine-restricted FXR agonist, did not raise the concentrations of TGR5 ligands nor modulate TGR5 signaling and, accordingly, did not improve dysmetabolic status. INT-777 and RO5527239 directly activated TGR5. INT-777 only increased the TGR5 activation capacity of the portal blood; RO5527239 also amplified the TGR5 activation capacity of systemic blood. Both molecules improved glucose tolerance. In spite of the TGR5 activation capacity, INT-777, but not RO5527239, reduced liver disease severity. In conclusion, TGR5 activation in enterohepatic, rather than in peripheral, tissues has beneficial effects on glucose tolerance and MAFLD.
- Published
- 2022
- Full Text
- View/download PDF
9. Enterohepatic, Gluco-metabolic, and Gut Microbial Characterization of Individuals With Bile Acid Malabsorption.
- Author
-
Kårhus ML, Sonne DP, Thomasen M, Ellegaard AM, Holst JJ, Rehfeld JF, Chávez-Talavera O, Tailleux A, Staels B, Nielsen DS, Krych L, Dragsted LO, Vilsbøll T, Brønden A, and Knop FK
- Abstract
Background and Aims: Bile acid malabsorption (BAM) is a debilitating disease characterized by loose stools and high stool frequency. The pathophysiology of BAM is not well-understood. We investigated postprandial enterohepatic and gluco-metabolic physiology, as well as gut microbiome composition and fecal bile acid content in patients with BAM., Methods: Twelve participants with selenium-75 homocholic acid taurine test-verified BAM and 12 healthy controls, individually matched on sex, age, and body mass index, were included. Each participant underwent 2 mixed meal tests (with and without administration of the bile acid sequestrant colesevelam) with blood sampling and evaluation of gallbladder motility; bile acid content and microbiota composition were evaluated in fecal specimens., Results: Patients with BAM were characterized by increased bile acid synthesis as assessed by circulating 7-alpha-hydroxy-4-cholesten-3-one, fecal bile acid content, and postprandial concentrations of glucose, insulin, C-peptide, and glucagon. The McAuley index of insulin sensitivity was lower in patients with BAM than that in healthy controls. In patients with BAM, colesevelam co-administered with the meal reduced postprandial concentrations of bile acids and fibroblast growth factor 19 and increased 7-alpha-hydroxy-4-cholesten-3-one concentrations but did not affect postprandial glucagon-like peptide 1 responses or other gluco-metabolic parameters. Patients with BAM were characterized by a gut microbiome with low relative abundance of bifidobacteria and high relative abundance of Blautia , Streptococcus , Ruminococcus gnavus , and Akkermansia muciniphila ., Conclusion: Patients with BAM are characterized by an overproduction of bile acids, greater fecal bile acid content, and a gluco-metabolic profile indicative of a dysmetabolic prediabetic-like state, with changes in their gut microbiome composition potentially linking their enterohepatic pathophysiology and their dysmetabolic phenotype. ClinicalTrials.gov number NCT03009916., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
10. Impaired Glucose Homeostasis in a Tau Knock-In Mouse Model.
- Author
-
Benderradji H, Kraiem S, Courty E, Eddarkaoui S, Bourouh C, Faivre E, Rolland L, Caron E, Besegher M, Oger F, Boschetti T, Carvalho K, Thiroux B, Gauvrit T, Nicolas E, Gomez-Murcia V, Bogdanova A, Bongiovanni A, Muhr-Tailleux A, Lancel S, Bantubungi K, Sergeant N, Annicotte JS, Buée L, Vieau D, Blum D, and Buée-Scherrer V
- Abstract
Alzheimer's disease (AD) is the leading cause of dementia. While impaired glucose homeostasis has been shown to increase AD risk and pathological loss of tau function, the latter has been suggested to contribute to the emergence of the glucose homeostasis alterations observed in AD patients. However, the links between tau impairments and glucose homeostasis, remain unclear. In this context, the present study aimed at investigating the metabolic phenotype of a new tau knock-in (KI) mouse model, expressing, at a physiological level, a human tau protein bearing the P301L mutation under the control of the endogenous mouse Mapt promoter. Metabolic investigations revealed that, while under chow diet tau KI mice do not exhibit significant metabolic impairments, male but not female tau KI animals under High-Fat Diet (HFD) exhibited higher insulinemia as well as glucose intolerance as compared to control littermates. Using immunofluorescence, tau protein was found colocalized with insulin in the β cells of pancreatic islets in both mouse (WT, KI) and human pancreas. Isolated islets from tau KI and tau knock-out mice exhibited impaired glucose-stimulated insulin secretion (GSIS), an effect recapitulated in the mouse pancreatic β-cell line (MIN6) following tau knock-down. Altogether, our data indicate that loss of tau function in tau KI mice and, particularly, dysfunction of pancreatic β cells might promote glucose homeostasis impairments and contribute to metabolic changes observed in AD., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Benderradji, Kraiem, Courty, Eddarkaoui, Bourouh, Faivre, Rolland, Caron, Besegher, Oger, Boschetti, Carvalho, Thiroux, Gauvrit, Nicolas, Gomez-Murcia, Bogdanova, Bongiovanni, Muhr-Tailleux, Lancel, Bantubungi, Sergeant, Annicotte, Buée, Vieau, Blum and Buée-Scherrer.)
- Published
- 2022
- Full Text
- View/download PDF
11. Farnesoid X Receptor Activation in Brain Alters Brown Adipose Tissue Function via the Sympathetic System.
- Author
-
Deckmyn B, Domenger D, Blondel C, Ducastel S, Nicolas E, Dorchies E, Caron E, Charton J, Vallez E, Deprez B, Annicotte JS, Lestavel S, Tailleux A, Magnan C, Staels B, and Bantubungi K
- Abstract
The nuclear bile acid (BA) receptor farnesoid X receptor (FXR) is a major regulator of metabolic/energy homeostasis in peripheral organs. Indeed, enterohepatic-expressed FXR controls metabolic processes (BA, glucose and lipid metabolism, fat mass, body weight). The central nervous system (CNS) regulates energy homeostasis in close interaction with peripheral organs. While FXR has been reported to be expressed in the brain, its function has not been studied so far. We studied the role of FXR in brain control of energy homeostasis by treating wild-type and FXR-deficient mice by intracerebroventricular (ICV) injection with the reference FXR agonist GW4064. Here we show that pharmacological activation of brain FXR modifies energy homeostasis by affecting brown adipose tissue (BAT) function. Brain FXR activation decreases the rate-limiting enzyme in catecholamine synthesis, tyrosine hydroxylase (TH), and consequently the sympathetic tone. FXR activation acts by inhibiting hypothalamic PKA-CREB induction of TH expression. These findings identify a function of brain FXR in the control of energy homeostasis and shed new light on the complex control of energy homeostasis by BA through FXR., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The reviewer DV declared a shared affiliation, though no other collaboration, with several of the authors, KB, BDc, DD, CB, SD, EN, ED, EC, JC, EV, J-SA, AT, BS, BDp, SL., (Copyright © 2022 Deckmyn, Domenger, Blondel, Ducastel, Nicolas, Dorchies, Caron, Charton, Vallez, Deprez, Annicotte, Lestavel, Tailleux, Magnan, Staels and Bantubungi.)
- Published
- 2022
- Full Text
- View/download PDF
12. The hepatic compensatory response to elevated systemic sulfide promotes diabetes.
- Author
-
Carter RN, Gibbins MTG, Barrios-Llerena ME, Wilkie SE, Freddolino PL, Libiad M, Vitvitsky V, Emerson B, Le Bihan T, Brice M, Su H, Denham SG, Homer NZM, Mc Fadden C, Tailleux A, Faresse N, Sulpice T, Briand F, Gillingwater T, Ahn KH, Singha S, McMaster C, Hartley RC, Staels B, Gray GA, Finch AJ, Selman C, Banerjee R, and Morton NM
- Subjects
- Animals, Diabetes Mellitus etiology, Diabetes Mellitus metabolism, Dyslipidemias etiology, Dyslipidemias metabolism, Glucose metabolism, Lipid Metabolism, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-E2-Related Factor 2 metabolism, Proteome metabolism, Diabetes Mellitus pathology, Dyslipidemias pathology, Gluconeogenesis, Liver pathology, Sulfides metabolism, Thiosulfate Sulfurtransferase physiology
- Abstract
Impaired hepatic glucose and lipid metabolism are hallmarks of type 2 diabetes. Increased sulfide production or sulfide donor compounds may beneficially regulate hepatic metabolism. Disposal of sulfide through the sulfide oxidation pathway (SOP) is critical for maintaining sulfide within a safe physiological range. We show that mice lacking the liver- enriched mitochondrial SOP enzyme thiosulfate sulfurtransferase (Tst
-/- mice) exhibit high circulating sulfide, increased gluconeogenesis, hypertriglyceridemia, and fatty liver. Unexpectedly, hepatic sulfide levels are normal in Tst-/- mice because of exaggerated induction of sulfide disposal, with associated suppression of global protein persulfidation and nuclear respiratory factor 2 target protein levels. Hepatic proteomic and persulfidomic profiles converge on gluconeogenesis and lipid metabolism, revealing a selective deficit in medium-chain fatty acid oxidation in Tst-/- mice. We reveal a critical role of TST in hepatic metabolism that has implications for sulfide donor strategies in the context of metabolic disease., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
13. Bile acids contribute to the development of non-alcoholic steatohepatitis in mice.
- Author
-
Gillard J, Clerbaux LA, Nachit M, Sempoux C, Staels B, Bindels LB, Tailleux A, and Leclercq IA
- Abstract
Background & Aims: Through FXR and TGR5 signaling, bile acids (BAs) modulate lipid and glucose metabolism, inflammation and fibrosis. Hence, BAs returning to the liver after enteric secretion, modification and reabsorption may contribute to the pathogenesis of non-alcoholic steatohepatitis (NASH). Herein, we characterized the enterohepatic profile and signaling of BAs in preclinical models of NASH, and explored the consequences of experimental manipulation of BA composition., Methods: We used high-fat diet (HFD)-fed foz/foz and high-fructose western diet-fed C57BL/6J mice, and compared them to their respective controls. Mice received a diet supplemented with deoxycholic acid (DCA) to modulate BA composition., Results: Compared to controls, mice with NASH had lower concentrations of BAs in their portal blood and bile, while systemic BA concentrations were not significantly altered. Notably, the concentrations of secondary BAs, and especially of DCA, and the ratio of secondary to primary BAs were strikingly lower in bile and portal blood of mice with NASH. Hence, portal blood was poor in FXR and TGR5 ligands, and conferred poor anti-inflammatory protection in mice with NASH. Enhanced primary BAs synthesis and conversion of secondary to primary BAs in NASH livers contributed to the depletion in secondary BAs. Dietary DCA supplementation in HFD-fed foz/foz mice restored the BA concentrations in portal blood, increased TGR5 and FXR signaling, improved the dysmetabolic status, protected from steatosis and hepatocellular ballooning, and reduced macrophage infiltration., Conclusions: BA composition in the enterohepatic cycle, but not in systemic circulation, is profoundly altered in preclinical models of NASH, with specific depletion in secondary BAs. Dietary correction of the BA profile protected from NASH, supporting a role for enterohepatic BAs in the pathogenesis of NASH., Lay Summary: This study clearly demonstrates that the alterations of enterohepatic bile acids significantly contribute to the development of non-alcoholic steatohepatitis in relevant preclinical models. Indeed, experimental modulation of bile acid composition restored perturbed FXR and TGR5 signaling and prevented non-alcoholic steatohepatitis and associated metabolic disorders., Competing Interests: The authors declare that they have no conflict of interest in relation to this work to disclose. Please refer to the accompanying ICMJE disclosure forms for further details., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
14. The ALGOVUE Clinical Trial: Effects of the Daily Consumption of Eggs Enriched with Lutein and Docosahexaenoic Acid on Plasma Composition and Macular Pigment Optical Density.
- Author
-
Schnebelen-Berthier C, Acar N, Simon E, Thabuis C, Bourdillon A, Mathiaud A, Dauchet L, Delcourt C, Benlian P, Crochet M, Defoort S, Tailleux A, Staels B, Bretillon L, and Lecerf JM
- Subjects
- Adult, Erythrocytes metabolism, Female, Humans, Lipoproteins blood, Male, Optical Phenomena, Patient Compliance, Xanthophylls blood, Young Adult, Zeaxanthins blood, Docosahexaenoic Acids blood, Food, Fortified, Lutein blood, Macular Pigment blood
- Abstract
Background: Carotenoids and docosahexaenoic acid (DHA) were identified as essential components for eye health and are both naturally present in eggs., Objective: We aimed to evaluate the effect of the daily consumption of two eggs enriched with lutein/zeaxanthin and DHA on macular pigment optical density (MPOD) and on circulating xanthophyll and fatty acid concentrations in healthy participants., Methods: Ninety-nine healthy volunteers consumed either two standard eggs or two enriched eggs per day for 4 months. MPOD was measured at baseline (V0) and at follow-up (V4) using a modified confocal scanning laser ophthalmoscope (primary outcome). Blood samples were collected to determine total plasma and lipoprotein fatty acids and lutein/zeaxanthin compositions at V0 and V4 (secondary outcomes)., Results: A slight but significant increase in MPOD was observed for all study participants consuming two eggs per day for 4 months at all eccentricities (0.5°, 1°, 2°, and 4°). Plasma and lipoprotein lutein, zeaxanthin, and DHA concentrations significantly increased in both groups but were greater in the enriched group (for the enriched group (V0 vs. V4): lutein, 167 vs. 369 ng/mL; zeaxanthin, 17.7 vs. 29.2 ng/mL; DHA, 1.89 vs. 2.56% of total fatty acids). Interestingly, lutein from high-density lipoprotein (HDL) was strongly correlated with MPOD at 0.5 and 1° eccentricities (rho = 0.385, p = 0.008, and rho = 0.461, p = 0.001, respectively)., Conclusions: MPOD was slightly increased in both groups. Lutein, zeaxanthin, and DHA plasma concentrations were strongly enhanced in the enriched group compared with the standard group. A significant correlation was found between MPOD level and lutein concentration in HDL.
- Published
- 2021
- Full Text
- View/download PDF
15. Hypothalamic bile acid-TGR5 signaling protects from obesity.
- Author
-
Castellanos-Jankiewicz A, Guzmán-Quevedo O, Fénelon VS, Zizzari P, Quarta C, Bellocchio L, Tailleux A, Charton J, Fernandois D, Henricsson M, Piveteau C, Simon V, Allard C, Quemener S, Guinot V, Hennuyer N, Perino A, Duveau A, Maitre M, Leste-Lasserre T, Clark S, Dupuy N, Cannich A, Gonzales D, Deprez B, Mithieux G, Dombrowicz D, Bäckhed F, Prevot V, Marsicano G, Staels B, Schoonjans K, and Cota D
- Subjects
- Animals, Body Weight genetics, Energy Metabolism genetics, HEK293 Cells, Humans, Hypothalamus metabolism, Mice, Mice, Inbred C57BL, Mice, Obese, Mice, Transgenic, Obesity genetics, Obesity prevention & control, Receptors, G-Protein-Coupled genetics, Signal Transduction physiology, Bile Acids and Salts metabolism, Obesity metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Bile acids (BAs) improve metabolism and exert anti-obesity effects through the activation of the Takeda G protein-coupled receptor 5 (TGR5) in peripheral tissues. TGR5 is also found in the brain hypothalamus, but whether hypothalamic BA signaling is implicated in body weight control and obesity pathophysiology remains unknown. Here we show that hypothalamic BA content is reduced in diet-induced obese mice. Central administration of BAs or a specific TGR5 agonist in these animals decreases body weight and fat mass by activating the sympathetic nervous system, thereby promoting negative energy balance. Conversely, genetic downregulation of hypothalamic TGR5 expression in the mediobasal hypothalamus favors the development of obesity and worsens established obesity by blunting sympathetic activity. Lastly, hypothalamic TGR5 signaling is required for the anti-obesity action of dietary BA supplementation. Together, these findings identify hypothalamic TGR5 signaling as a key mediator of a top-down neural mechanism that counteracts diet-induced obesity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
16. A randomized placebo-controlled trial of elafibranor in patients with primary biliary cholangitis and incomplete response to UDCA.
- Author
-
Schattenberg JM, Pares A, Kowdley KV, Heneghan MA, Caldwell S, Pratt D, Bonder A, Hirschfield GM, Levy C, Vierling J, Jones D, Tailleux A, Staels B, Megnien S, Hanf R, Magrez D, Birman P, and Luketic V
- Subjects
- Adolescent, Adult, Aged, Alkaline Phosphatase blood, Bilirubin blood, Double-Blind Method, Female, Humans, Liver Cirrhosis, Biliary blood, Liver Cirrhosis, Biliary complications, Male, Middle Aged, Pruritus complications, Pruritus drug therapy, Quality of Life, Treatment Outcome, Young Adult, Chalcones adverse effects, Liver Cirrhosis, Biliary drug therapy, PPAR alpha agonists, PPAR delta agonists, Propionates adverse effects, Ursodeoxycholic Acid therapeutic use
- Abstract
Background & Aims: Patients with primary biliary cholangitis (PBC) who have an incomplete response to ursodeoxycholic acid remain at risk of disease progression. We investigated the safety and efficacy of elafibranor, a dual PPARα/δ agonist, in patients with PBC., Methods: This 12-week, double-blind phase II trial enrolled 45 adults with PBC who had incomplete response to ursodeoxycholic acid (alkaline phosphatase levels ≥1.67-fold the upper limit of normal (ULN). Patients were randomly assigned to elafibranor 80 mg, elafibranor 120 mg or placebo. The primary endpoint was the relative change of ALP at 12 weeks (NCT03124108)., Results: At 12 weeks, ALP was reduced by -48.3±14.8% in the elafibranor 80 mg group (p <0.001 vs. placebo) and by -40.6±17.4% in the elafibranor 120 mg group (p <0.001) compared to a +3.2±14.8% increase in the placebo group. The composite endpoint of ALP ≤1.67-fold the ULN, decrease of ALP >15% and total bilirubin below the ULN was achieved in 67% patients in the elafibranor 80 mg group and 79% patients in the elafibranor 120 mg group, vs. 6.7% patients in the placebo group. Levels of gamma-glutamyltransferase decreased by 37.0±25.5% in the elafibranor 80 mg group (p <0.001) and 40.0±24.1% in the elafibranor 120 mg group (p <0.01) compared to no change (+0.2±26.0%) in the placebo group. Levels of disease markers such as IgM, 5'-nucleotidase or high-sensitivity C-reactive protein were likewise reduced by elafibranor. Pruritus was not induced or exacerbated by elafibranor and patients with pruritus at baseline reported less pruritic symptoms at the end of treatment. All possibly drug-related non-serious adverse events were mild to moderate., Conclusion: In this randomized phase II trial, elafibranor was generally safe and well tolerated and significantly reduced levels of ALP, composite endpoints of bilirubin and ALP, as well as other markers of disease activity in patients with PBC and an incomplete response to ursodeoxycholic acid., Lay Summary: Patients with primary biliary cholangitis (a rare chronic liver disease) that do not respond to standard therapy remain at risk of disease progression toward cirrhosis and impaired quality of life. Elafibranor is a nuclear receptor agonist that we tested in a randomized clinical trial over 12 weeks. It successfully decreased levels of disease activity markers, including alkaline phosphatase. Thus, this study is the foundation for a larger prospective study that will determine the efficacy and safety of this drug as a second-line therapy., Clinical Trial Registration Number: Clinical Trials.gov NCT03124108., Competing Interests: Conflict of interest JMS reports consultancy: BMS, Boehringer Ingelheim, Echosens, Galmed, Genfit, Gilead Sciences, Intercept Pharmaceuticals, Madrigal, Nordic Bioscience, Novartis, Pfizer, Roche, Sanofi, Zydus. Research Funding: Gilead Sciences. AP has received grant funding, personal fees, and advisory board fees from Intercept Pharmaceuticals; advisory board fees and fees for teaching from Novartis; and personal fees from CymaBay Therapeutics and Inova Diagnostics. KVK: serves as consultant to or an advisory boards for Conatus, CymaBay, Gilead, Intercept, La Jolla, Merck and Novartis. He receives research support from Genfit, Gilead, High Tide, Intercept, NGM Biopharma and Novartis and serves as a speaker for Abbvie, Gilead Sciences and Intercept. MAH: Consultancy for Roche, Novartis, Falk and Intercept. SC received research support from Genfit, Gilead and Zydus. DP: nothing to disclose. AB: nothing to disclose. GMH has consulted for Intercept, Genfit, Novartis, GSK, Cymabay and Gilead. CL reports research grants: Gilead, Intercept, CymaBay, Genfit, Genkyotex, Enanta, GSK, Novartis, NGM, High Tide, Durect, Alnylam, Zydus, Cara Therapeutics, Target PharmaSolutions; Consulting fees/Advisory boards: CymaBay, GSK, Shire, Pliant, Target PharmaSolutions, Flashlight Therapeutics, Cara Therapeutics; Royalties: Up-to-date; other: Editorial board Liver Transplantation. JV Research Grants: Allergan, Arena, CymaBay, Enanta, Genkyotex, Intercept, Lilly, NGM Pharmaceuticals, Novartis, TaiwanJ, Scientific Advisor: Arena, BioIncept, Blade, CymaBay, Enanta, Genkyotex, Glaxo-Smith-Kline, Intercept, Lilly, Novartis, TaiwanJ, Authorship: Up-to-Date Immunosuppression in Liver Transplantation; AASLD Writing Committee AIH Guidance In Press, 2019. DJ reports consultancy and grant funding from Intercept and Consultancy from Novartis. AT has nothing to disclose. BS is consultant and president of the SAB of Genfit SA. SM was a former Genfit employee and has currently no COI. RH, DM and PB are Genfit employees. VL reports consultancy for Genfit., (Copyright © 2021 European Association for the Study of the Liver. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
17. Characterization of one anastomosis gastric bypass and impact of biliary and common limbs on bile acid and postprandial glucose metabolism in a minipig model.
- Author
-
Marciniak C, Chávez-Talavera O, Caiazzo R, Hubert T, Zubiaga L, Baud G, Quenon A, Descat A, Vallez E, Goossens JF, Kouach M, Vangelder V, Gobert M, Daoudi M, Derudas B, Pigny P, Klein A, Gmyr V, Raverdy V, Lestavel S, Laferrère B, Staels B, Tailleux A, and Pattou F
- Subjects
- Anastomosis, Surgical methods, Animals, Bile Acids and Salts blood, Biliary Tract metabolism, Biliary Tract Surgical Procedures methods, Blood Glucose metabolism, Common Bile Duct metabolism, Common Bile Duct surgery, Female, Models, Animal, Obesity, Morbid metabolism, Obesity, Morbid surgery, Postprandial Period, Random Allocation, Swine, Swine, Miniature, Weight Loss physiology, Bile Acids and Salts metabolism, Biliary Tract pathology, Common Bile Duct pathology, Gastric Bypass methods, Glucose metabolism
- Abstract
The alimentary limb has been proposed to be a key driver of the weight-loss-independent metabolic improvements that occur upon bariatric surgery. However, the one anastomosis gastric bypass (OAGB) procedure, consisting of one long biliary limb and a short common limb, induces similar beneficial metabolic effects compared to Roux-en-Y Gastric Bypass (RYGB) in humans, despite the lack of an alimentary limb. The aim of this study was to assess the role of the length of biliary and common limbs in the weight loss and metabolic effects that occur upon OAGB. OAGB and sham surgery, with or without modifications of the length of either the biliary limb or the common limb, were performed in Gottingen minipigs. Weight loss, metabolic changes, and the effects on plasma and intestinal bile acids (BAs) were assessed 15 days after surgery. OAGB significantly decreased body weight, improved glucose homeostasis, increased postprandial GLP-1 and fasting plasma BAs, and qualitatively changed the intestinal BA species composition. Resection of the biliary limb prevented the body weight loss effects of OAGB and attenuated the postprandial GLP-1 increase. Improvements in glucose homeostasis along with changes in plasma and intestinal BAs occurred after OAGB regardless of the biliary limb length. Resection of only the common limb reproduced the glucose homeostasis effects and the changes in intestinal BAs. Our results suggest that the changes in glucose metabolism and BAs after OAGB are mainly mediated by the length of the common limb, whereas the length of the biliary limb contributes to body weight loss. NEW & NOTEWORTHY Common limb mediates postprandial glucose metabolism change after gastric bypass whereas biliary limb contributes to weight loss.
- Published
- 2021
- Full Text
- View/download PDF
18. Deletion of fibroblast activation protein provides atheroprotection.
- Author
-
Stein S, Weber J, Nusser-Stein S, Pahla J, Zhang HE, Mohammed SA, Oppi S, Gaul DS, Paneni F, Tailleux A, Staels B, von Meyenn F, Ruschitzka F, Gorrell MD, Lüscher TF, and Matter CM
- Subjects
- Animals, Aorta pathology, Aortic Diseases enzymology, Aortic Diseases genetics, Aortic Diseases pathology, Atherosclerosis enzymology, Atherosclerosis genetics, Atherosclerosis pathology, Case-Control Studies, Collagen genetics, Collagen metabolism, Disease Models, Animal, Endopeptidases genetics, Fibrosis, Gene Deletion, Humans, Lipids blood, Male, Membrane Proteins genetics, Mice, Inbred C57BL, Mice, Knockout, ApoE, Muscle, Smooth, Vascular enzymology, Muscle, Smooth, Vascular pathology, Plaque, Atherosclerotic, Proteome, Receptors, LDL deficiency, Receptors, LDL genetics, Transcriptome, Mice, Aorta enzymology, Aortic Diseases prevention & control, Atherosclerosis prevention & control, Endopeptidases deficiency, Membrane Proteins deficiency, Vascular Remodeling
- Abstract
Aims: Fibroblast activation protein (FAP) is upregulated at sites of tissue remodelling including chronic arthritis, solid tumours, and fibrotic hearts. It has also been associated with human coronary atherosclerotic plaques. Yet, the causal role of FAP in atherosclerosis remains unknown. To investigate the cause-effect relationship of endogenous FAP in atherogenesis, we assessed the effects of constitutive Fap deletion on plaque formation in atherosclerosis-prone apolipoprotein E (Apoe) or low-density lipoprotein receptor (Ldlr) knockout mice., Methods and Results: Using en face analyses of thoraco-abdominal aortae and aortic sinus cross-sections, we demonstrate that Fap deficiency decreased plaque formation in two atherosclerotic mouse models (-46% in Apoe and -34% in Ldlr knockout mice). As a surrogate of plaque vulnerability fibrous cap thickness was used; it was increased in Fap-deficient mice, whereas Sirius red staining demonstrated that total collagen content remained unchanged. Using polarized light, atherosclerotic lesions from Fap-deficient mice displayed increased FAP targets in terms of enhanced collagen birefringence in plaques and increased pre-COL3A1 expression in aortic lysates. Analyses of the Stockholm Atherosclerosis Gene Expression data revealed that FAP expression was increased in human atherosclerotic compared to non-atherosclerotic arteries., Conclusions: Our data provide causal evidence that constitutive Fap deletion decreases progression of experimental atherosclerosis and increases features of plaque stability with decreased collagen breakdown. Thus, inhibition of FAP expression or activity may not only represent a promising therapeutic target in atherosclerosis but appears safe at the experimental level for FAP-targeted cancer therapies., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2020. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
19. Multiple Selection Criteria for Probiotic Strains with High Potential for Obesity Management.
- Author
-
Alard J, Cudennec B, Boutillier D, Peucelle V, Descat A, Decoin R, Kuylle S, Jablaoui A, Rhimi M, Wolowczuk I, Pot B, Tailleux A, Maguin E, Holowacz S, and Grangette C
- Subjects
- Animals, Bile Acids and Salts metabolism, Diet adverse effects, Disease Models, Animal, Gastrointestinal Hormones metabolism, Hypothalamus metabolism, Leptin metabolism, Mice, Obesity etiology, Obesity Management methods, Receptors, Leptin metabolism, Weight Gain physiology, Adipocytes microbiology, Enteroendocrine Cells microbiology, Gastrointestinal Microbiome physiology, Obesity microbiology, Probiotics pharmacology
- Abstract
Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We evaluated in vitro the ability of 23 strains to limit lipid accumulation in adipocytes and to enhance the secretion of satiety-promoting gut peptide in enteroendocrine cells. Following the in vitro screening, selected strains were further investigated in vivo, single, or as mixtures, using a murine model of diet-induced obesity. Strain Bifidobacterium longum PI10 administrated alone and the mixture of B. animalis subsp. lactis LA804 and Lactobacillus gasseri LA806 limited body weight gain and reduced obesity-associated metabolic dysfunction and inflammation. These protective effects were associated with changes in the hypothalamic gene expression of leptin and leptin receptor as well as with changes in the composition of gut microbiota and the profile of bile acids. This study provides crucial clues to identify new potential probiotics as effective therapeutic approaches in the management of obesity, while also providing some insights into their mechanisms of action.
- Published
- 2021
- Full Text
- View/download PDF
20. Beyond the Rule of 5: Impact of PEGylation with Various Polymer Sizes on Pharmacokinetic Properties, Structure-Properties Relationships of mPEGylated Small Agonists of TGR5 Receptor.
- Author
-
Hoguet V, Lasalle M, Maingot M, Dequirez G, Boulahjar R, Leroux F, Piveteau C, Herledan A, Biela A, Dumont J, Chávez-Talavera O, Belloy L, Duplan I, Hennuyer N, Butruille L, Lestavel S, Sevin E, Culot M, Gosselet F, Staels B, Deprez B, Tailleux A, and Charton J
- Subjects
- Animals, Blood-Brain Barrier metabolism, Caco-2 Cells, HEK293 Cells, Humans, Hypoglycemic Agents pharmacokinetics, Male, Mice, Mice, Inbred C57BL, Microsomes, Liver metabolism, Polyethylene Glycols chemistry, Receptors, G-Protein-Coupled chemistry, Structure-Activity Relationship, Hypoglycemic Agents chemical synthesis, Hypoglycemic Agents pharmacology, Receptors, G-Protein-Coupled agonists
- Abstract
PEGylation of therapeutic agents is known to improve the pharmacokinetic behavior of macromolecular drugs and nanoparticles. In this work, we performed the conjugation of polyethylene glycols (220-5000 Da) to a series of non-steroidal small agonists of the bile acids receptor TGR5. A suitable anchoring position on the agonist was identified to retain full agonistic potency with the conjugates. We describe herein an extensive structure-properties relationships study allowing us to finely describe the non-linear effects of the PEG length on the physicochemical as well as the in vitro and in vivo pharmacokinetic properties of these compounds. When appending a PEG of suitable length to the TGR5 pharmacophore, we were able to identify either systemic or gut lumen-restricted TGR5 agonists.
- Published
- 2021
- Full Text
- View/download PDF
21. Inflammation-induced cholestasis in cancer cachexia.
- Author
-
Thibaut MM, Sboarina M, Roumain M, Pötgens SA, Neyrinck AM, Destrée F, Gillard J, Leclercq IA, Dachy G, Demoulin JB, Tailleux A, Lestavel S, Rastelli M, Everard A, Cani PD, Porporato PE, Loumaye A, Thissen JP, Muccioli GG, Delzenne NM, and Bindels LB
- Subjects
- Animals, Cytokines, Humans, Mice, Cachexia etiology, Cholestasis etiology, Inflammation complications, Neoplasms complications
- Abstract
Background: Cancer cachexia is a debilitating metabolic syndrome contributing to cancer death. Organs other than the muscle may contribute to the pathogenesis of cancer cachexia. This work explores new mechanisms underlying hepatic alterations in cancer cachexia., Methods: We used transcriptomics to reveal the hepatic gene expression profile in the colon carcinoma 26 cachectic mouse model. We performed bile acid, tissue mRNA, histological, biochemical, and western blot analyses. Two interventional studies were performed using a neutralizing interleukin 6 antibody and a bile acid sequestrant, cholestyramine. Our findings were evaluated in a cohort of 94 colorectal cancer patients with or without cachexia (43/51)., Results: In colon carcinoma 26 cachectic mice, we discovered alterations in five inflammatory pathways as well as in other pathways, including bile acid metabolism, fatty acid metabolism, and xenobiotic metabolism (normalized enrichment scores of -1.97, -2.16, and -1.34, respectively; all Padj < 0.05). The hepatobiliary transport system was deeply impaired in cachectic mice, leading to increased systemic and hepatic bile acid levels (+1512 ± 511.6 pmol/mg, P = 0.01) and increased hepatic inflammatory cytokines and neutrophil recruitment to the liver of cachectic mice (+43.36 ± 16.01 neutrophils per square millimetre, P = 0.001). Adaptive mechanisms were set up to counteract this bile acid accumulation by repressing bile acid synthesis and by enhancing alternative routes of basolateral bile acid efflux. Targeting bile acids using cholestyramine reduced hepatic inflammation, without affecting the hepatobiliary transporters (e.g. tumour necrosis factor α signalling via NFκB and inflammatory response pathways, normalized enrichment scores of -1.44 and -1.36, all Padj < 0.05). Reducing interleukin 6 levels counteracted the change in expression of genes involved in the hepatobiliary transport, bile acid synthesis, and inflammation. Serum bile acid levels were increased in cachectic vs. non-cachectic cancer patients (e.g. total bile acids, +5.409 ± 1.834 μM, P = 0.026) and were strongly correlated to systemic inflammation (taurochenodeoxycholic acid and C-reactive protein: ρ = 0.36, Padj = 0.017)., Conclusions: We show alterations in bile acid metabolism and hepatobiliary secretion in cancer cachexia. In this context, we demonstrate the contribution of systemic inflammation to the impairment of the hepatobiliary transport system and the role played by bile acids in the hepatic inflammation. This work paves the way to a better understanding of the role of the liver in cancer cachexia., (© 2020 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)
- Published
- 2021
- Full Text
- View/download PDF
22. NASH-related increases in plasma bile acid levels depend on insulin resistance.
- Author
-
Grzych G, Chávez-Talavera O, Descat A, Thuillier D, Verrijken A, Kouach M, Legry V, Verkindt H, Raverdy V, Legendre B, Caiazzo R, Van Gaal L, Goossens JF, Paumelle R, Francque S, Pattou F, Haas JT, Tailleux A, and Staels B
- Abstract
Background & Aims: Plasma bile acids (BAs) have been extensively studied as pathophysiological actors in non-alcoholic steatohepatitis (NASH). However, results from clinical studies are often complicated by the association of NASH with type 2 diabetes (T2D), obesity, and insulin resistance (IR). Here, we sought to dissect the relationship between NASH, T2D, and plasma BA levels in a large patient cohort., Methods: Four groups of patients from the Biological Atlas of Severe Obesity (ABOS) cohort (Clinical Trials number NCT01129297) were included based on the presence or absence of histologically evaluated NASH with or without coincident T2D. Patients were matched for BMI, homeostatic model assessment 2 (HOMA2)-assessed IR, glycated haemoglobin, age, and gender. To study the effect of IR and BMI on the association of plasma BA and NASH, patients from the HEPADIP study were included. In both cohorts, fasting plasma BA concentrations were measured., Results: Plasma BA concentrations were higher in NASH compared with No-NASH patients both in T2D and NoT2D patients from the ABOS cohort. As we previously reported that plasma BA levels were unaltered in NASH patients of the HEPADIP cohort, we assessed the impact of BMI and IR on the association of NASH and BA on the combined BA datasets. Our results revealed that NASH-associated increases in plasma total cholic acid (CA) concentrations depend on the degree of HOMA2-assessed systemic IR, but not on β-cell function nor on BMI., Conclusions: Plasma BA concentrations are elevated only in those NASH patients exhibiting pronounced IR., Lay Summary: Non-alcoholic steatohepatitis (NASH) is a progressive liver disease that frequently occurs in patients with obesity and type 2 diabetes. Reliable markers for the diagnosis of NASH are needed. Plasma bile acids have been proposed as NASH biomarkers. Herein, we found that plasma bile acids are only elevated in patients with NASH when significant insulin resistance is present, limiting their utility as NASH markers., Competing Interests: The authors declare that they have no conflicts of interest that pertain to this work. Please refer to the accompanying ICMJE disclosure forms for further details., (© 2021 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
23. Deletion of the nuclear receptor RORα in macrophages does not modify the development of obesity, insulin resistance and NASH.
- Author
-
L'homme L, Sermikli BP, Molendi-Coste O, Fleury S, Quemener S, Le Maître M, Joseph ML, Pineau L, Duhem C, Gross B, Vallez E, Tailleux A, Staels B, and Dombrowicz D
- Subjects
- Animals, Cells, Cultured, Diet, High-Fat adverse effects, Gene Deletion, Kupffer Cells metabolism, Male, Mice, Mice, Inbred C57BL, Non-alcoholic Fatty Liver Disease etiology, Nuclear Receptor Subfamily 1, Group F, Member 1 genetics, Obesity etiology, Insulin Resistance, Macrophages metabolism, Non-alcoholic Fatty Liver Disease metabolism, Nuclear Receptor Subfamily 1, Group F, Member 1 metabolism, Obesity metabolism
- Abstract
Retinoic acid receptor-related orphan receptor-alpha (RORα) is a transcription factor from the nuclear receptor family expressed by immune cells and involved in the development of obesity, insulin resistance (IR) and non-alcoholic steatohepatitis (NASH). It was recently reported that mice deficient for RORα in macrophages develop more severe NASH upon high fat diet (HFD) feeding due to altered Kupffer cell function. To better understand the role of RORα in obesity and IR, we independently generated a macrophage RORα-deficient mouse line. We report that RORα deletion in macrophages does not impact on HFD-induced obesity and IR. Surprisingly, we did not confirm an effect on NASH development upon HFD feeding nor in the more severe and obesity-independent choline-deficient, L-amino acid-defined diet model. Our results therefore show that RORα deletion in macrophages does not alter the development of obesity and IR and question its role in NASH.
- Published
- 2020
- Full Text
- View/download PDF
24. Analysis of the association of MPO and MMP-9 with stroke severity and outcome: Cohort study.
- Author
-
Maestrini I, Tagzirt M, Gautier S, Dupont A, Mendyk AM, Susen S, Tailleux A, Vallez E, Staels B, Cordonnier C, Leys D, and Bordet R
- Subjects
- Aged, Cohort Studies, Female, Humans, Lymphocyte Count, Male, Middle Aged, Neutrophils, Biomarkers blood, Matrix Metalloproteinase 9 blood, Peroxidase blood, Recovery of Function physiology, Stroke blood
- Abstract
Objective: In acute cerebral ischemia, circulating neutrophil count and neutrophil-to-lymphocyte ratio (NLR) are positively associated with stroke severity and worse outcomes. Mediators of this effect are unknown. We aimed to investigate (1) the relationship between plasma matrix metalloproteinase-9 (MMP-9) and myeloperoxidase (MPO) concentrations with stroke severity and outcome and (2) MMP-9 and MPO release after ex vivo stimulation of neutrophils by recombinant tissue plasminogen activator (rtPA)., Methods: We analyzed data collected in 255 patients with supratentorial cerebral infarcts recruited within 48 hours of symptoms onset irrespective of rtPA treatment. The endpoints were excellent outcome (modified Rankin Scale score 0-1), symptomatic intracerebral hemorrhage (European Cooperative Acute Stroke Study-II definition), and death at 3 months. The role of rtPA treatment on peripheral neutrophil degranulation was investigated in 18 patients within 4.5 hours and after 72 hours., Results: Neutrophil counts, NLR, and MPO plasma concentrations, but not MMP-9, were positively correlated with stroke severity. Higher neutrophil counts and NLR were independently associated with worse outcomes and higher mortality rates at month 3. Higher MPO plasma concentrations, but not MMP-9, were associated with worse outcome. Neutrophil-derived MMP-9, after ex vivo rtPA stimulation, but not MPO, were higher after 72 hours in patients treated by IV rtPA but not associated with hemorrhagic transformation., Conclusions: Neutrophil counts, NLR, and MPO plasma concentrations are associated with worse outcome in patients with acute cerebral ischemia, in contrast to MMP-9. Further investigations are needed to deepen our knowledge on MPO's role in the deleterious effect of neutrophils because it could represent a potential therapeutic target., (© 2020 American Academy of Neurology.)
- Published
- 2020
- Full Text
- View/download PDF
25. Plasma BCAA Changes in Patients With NAFLD Are Sex Dependent.
- Author
-
Grzych G, Vonghia L, Bout MA, Weyler J, Verrijken A, Dirinck E, Chevalier Curt MJ, Van Gaal L, Paumelle R, Francque S, Tailleux A, Haas JT, and Staels B
- Subjects
- Adult, Blood Glucose metabolism, Body Mass Index, Cross-Sectional Studies, Female, Humans, Insulin blood, Male, Middle Aged, Sex Factors, Tandem Mass Spectrometry, Amino Acids, Branched-Chain blood, Insulin Resistance physiology, Non-alcoholic Fatty Liver Disease blood, Obesity blood
- Abstract
Context: Plasma branched chain amino acid (BCAA) concentrations correlate positively with body mass index (BMI), measures of insulin resistance (IR), and severity of nonalcoholic fatty liver disease (NAFLD). Moreover, plasma BCAA concentrations also differ between the sexes, which display different susceptibilities to cardio-metabolic diseases., Objective: Assess whether plasma BCAA concentrations associate with NAFLD severity independently of BMI, IR, and sex., Patients: Patients visiting the obesity clinic of the Antwerp University Hospital were consecutively recruited from 2006 to 2014., Design and Setting: A cross-sectional study cohort of 112 obese patients (59 women and 53 men) was divided into 4 groups according to NAFLD severity. Groups were matched for sex, age, BMI, homeostatic model assessment of IR, and hemoglobin A1c., Main Outcome Measures: Fasting plasma BCAA concentrations were measured by tandem mass spectrometry using the aTRAQ™ method., Results: In the study cohort, a modest positive correlation was observed between plasma BCAA concentrations and NAFLD severity, as well as a strong effect of sex on plasma BCAA levels. Subgroup analysis by sex revealed that while plasma BCAA concentrations increased with severity of NAFLD in women, they tended to decrease in men. Additionally, only women displayed significantly increased plasma BCAAs with increasing fibrosis., Conclusion: Plasma BCAA concentrations display sex-dimorphic changes with increasing severity of NAFLD, independently of BMI, IR, and age. Additionally, plasma BCAA are associated with significant fibrosis in women, but not in men. These results highlight the importance of a careful consideration of sex as a major confounding factor in cross-sectional studies of NAFLD., (© Endocrine Society 2020. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
26. Sirt6 deletion in bone marrow-derived cells increases atherosclerosis - Central role of macrophage scavenger receptor 1.
- Author
-
Arsiwala T, Pahla J, van Tits LJ, Bisceglie L, Gaul DS, Costantino S, Miranda MX, Nussbaum K, Stivala S, Blyszczuk P, Weber J, Tailleux A, Stein S, Paneni F, Beer JH, Greter M, Becher B, Mostoslavsky R, Eriksson U, Staels B, Auwerx J, Hottiger MO, Lüscher TF, and Matter CM
- Subjects
- Animals, Aorta pathology, Apolipoproteins E deficiency, Apolipoproteins E metabolism, Bone Marrow Transplantation, Down-Regulation, Gene Knockdown Techniques, Hematopoiesis, Homozygote, Humans, Lipoproteins, LDL metabolism, Macrophages metabolism, Macrophages pathology, Mice, Models, Biological, Plaque, Atherosclerotic metabolism, Plaque, Atherosclerotic pathology, Proto-Oncogene Proteins c-myc metabolism, RAW 264.7 Cells, Atherosclerosis metabolism, Atherosclerosis pathology, Bone Marrow pathology, Gene Deletion, Scavenger Receptors, Class A metabolism, Sirtuins genetics, Sirtuins metabolism
- Abstract
Aims: Sirtuin 6 (Sirt6) is a NAD
+ -dependent deacetylase that plays a key role in DNA repair, inflammation and lipid regulation. Sirt6-null mice show severe metabolic defects and accelerated aging. Macrophage-foam cell formation via scavenger receptors is a key step in atherogenesis. We determined the effects of bone marrow-restricted Sirt6 deletion on foam cell formation and atherogenesis using a mouse model., Methods and Results: Sirt6 deletion in bone marrow-derived cells increased aortic plaques, lipid content and macrophage numbers in recipient Apoe- / - mice fed a high-cholesterol diet for 12 weeks (n = 12-14, p < .001). In RAW macrophages, Sirt6 overexpression reduced oxidized low-density lipoprotein (oxLDL) uptake, Sirt6 knockdown enhanced it and increased mRNA and protein levels of macrophage scavenger receptor 1 (Msr1), whereas levels of other oxLDL uptake and efflux transporters remained unchanged. Similarly, in human primary macrophages, Sirt6 knockdown increased MSR1 protein levels and oxLDL uptake. Double knockdown of Sirt6 and Msr1 abolished the increase in oxLDL uptake observed upon Sirt6 single knockdown. FACS analyses of macrophages from aortic plaques of Sirt6-deficient bone marrow-transplanted mice showed increased MSR1 protein expression. Double knockdown of Sirt6 and the transcription factor c-Myc in RAW cells abolished the increase in Msr1 mRNA and protein levels; c-Myc overexpression increased Msr1 mRNA and protein levels., Conclusions: Loss of Sirt6 in bone marrow-derived cells is proatherogenic; hereby macrophages play an important role given a c-Myc-dependent increase in MSR1 protein expression and an enhanced oxLDL uptake in human and murine macrophages. These findings assign endogenous SIRT6 in macrophages an important atheroprotective role., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
27. Bile acids associate with glucose metabolism, but do not predict conversion from impaired fasting glucose to diabetes.
- Author
-
Chávez-Talavera O, Wargny M, Pichelin M, Descat A, Vallez E, Kouach M, Bigot-Corbel E, Joliveau M, Goossens JF, Le May C, Hadjadj S, Hanf R, Tailleux A, Staels B, and Cariou B
- Subjects
- Adult, Aged, Biomarkers blood, Cohort Studies, Diabetes Mellitus, Type 2 metabolism, Disease Progression, Female, Follow-Up Studies, Glucose Intolerance metabolism, Humans, Insulin Resistance physiology, Male, Middle Aged, Prediabetic State metabolism, Predictive Value of Tests, Prospective Studies, Bile Acids and Salts blood, Diabetes Mellitus, Type 2 diagnosis, Glucose metabolism, Glucose Intolerance diagnosis, Prediabetic State diagnosis, Prediabetic State pathology
- Abstract
Objective: Bile acids (BAs) are signaling molecules controlling lipid and glucose metabolism. Since BA alterations are associated with obesity and insulin resistance, plasma BAs have been considered candidates to predict type 2 diabetes (T2D) risk. We aimed to determine (1) the association of BAs with glucose homeostasis parameters and (2) their predictive association with the risk of conversion from prediabetes to new-onset diabetes (NOD) in a prospective cohort study., Design: 205 patients with impaired fasting glucose (IFG) were followed each year during 5 years in the IT-DIAB cohort study. Twenty-one BA species and 7α-hydroxy-4-cholesten-3-one (C4), a marker of BA synthesis, were quantified by LC/MS-MS in plasma from fasted patients at baseline. Correlations between plasma BA species and metabolic parameters at baseline were assessed by Spearman's coefficients and the association between BAs and NOD was determined using Cox proportional-hazards models., Results: Among the analyzed BA species, total hyocholic acid (HCA) and the total HCA/total chenodeoxycholic acid (CDCA) ratio, reflecting hepatic BA 6α-hydroxylation activity, negatively correlated with BMI and HOMA-IR. The total HCA/total CDCA ratio also correlated negatively with HbA
1C . Conversion from IFG to NOD occurred in 33.7% of the participants during the follow-up. Plasma BA species were not independently associated with the conversion to NOD after adjustment with classical T2D risk factors., Conclusions: Fasting plasma BAs are not useful clinical biomarkers for predicting NOD in patients with IFG. However, an unexpected association between 6α-hydroxylated BAs and glucose parameters was found, suggesting a role for this specific BA pathway in metabolic homeostasis. IT-DIAB study registry number: NCT01218061., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
28. The nuclear receptor FXR inhibits Glucagon-Like Peptide-1 secretion in response to microbiota-derived Short-Chain Fatty Acids.
- Author
-
Ducastel S, Touche V, Trabelsi MS, Boulinguiez A, Butruille L, Nawrot M, Peschard S, Chávez-Talavera O, Dorchies E, Vallez E, Annicotte JS, Lancel S, Briand O, Bantubungi K, Caron S, Bindels LB, Delzenne NM, Tailleux A, Staels B, and Lestavel S
- Subjects
- Animals, Colon drug effects, Glucagon-Like Peptide 1 metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Colon metabolism, Fatty Acids, Volatile pharmacology, Glucagon-Like Peptide 1 antagonists & inhibitors, Microbiota, Receptors, Cytoplasmic and Nuclear physiology
- Abstract
The gut microbiota participates in the control of energy homeostasis partly through fermentation of dietary fibers hence producing short-chain fatty acids (SCFAs), which in turn promote the secretion of the incretin Glucagon-Like Peptide-1 (GLP-1) by binding to the SCFA receptors FFAR2 and FFAR3 on enteroendocrine L-cells. We have previously shown that activation of the nuclear Farnesoid X Receptor (FXR) decreases the L-cell response to glucose. Here, we investigated whether FXR also regulates the SCFA-induced GLP-1 secretion. GLP-1 secretion in response to SCFAs was evaluated ex vivo in murine colonic biopsies and in colonoids of wild-type (WT) and FXR knock-out (KO) mice, in vitro in GLUTag and NCI-H716 L-cells activated with the synthetic FXR agonist GW4064 and in vivo in WT and FXR KO mice after prebiotic supplementation. SCFA-induced GLP-1 secretion was blunted in colonic biopsies from GW4064-treated mice and enhanced in FXR KO colonoids. In vitro FXR activation inhibited GLP-1 secretion in response to SCFAs and FFAR2 synthetic ligands, mainly by decreasing FFAR2 expression and downstream Gαq-signaling. FXR KO mice displayed elevated colonic FFAR2 mRNA levels and increased plasma GLP-1 levels upon local supply of SCFAs with prebiotic supplementation. Our results demonstrate that FXR activation decreases L-cell GLP-1 secretion in response to inulin-derived SCFA by reducing FFAR2 expression and signaling. Inactivation of intestinal FXR using bile acid sequestrants or synthetic antagonists in combination with prebiotic supplementation may be a promising therapeutic approach to boost the incretin axis in type 2 diabetes.
- Published
- 2020
- Full Text
- View/download PDF
29. Transcription profiling in the liver of undernourished male rat offspring reveals altered lipid metabolism pathways and predisposition to hepatic steatosis.
- Author
-
Lecoutre S, Montel V, Vallez E, Pourpe C, Delmont A, Eury E, Verbanck M, Dickes-Coopman A, Daubersies P, Lesage J, Laborie C, Tailleux A, Staels B, Froguel P, Breton C, and Vieau D
- Subjects
- Animals, Bile Acids and Salts metabolism, Cholesterol metabolism, Fatty Acids metabolism, Fatty Liver metabolism, Fatty Liver pathology, Female, Gene Expression Profiling, Hepatocytes metabolism, Hepatocytes pathology, Lipid Droplets pathology, Liver pathology, Male, Oleic Acids metabolism, Pregnancy, Prenatal Exposure Delayed Effects metabolism, Prenatal Exposure Delayed Effects pathology, Prenatal Nutritional Physiological Phenomena genetics, Rats, Triglycerides metabolism, Fatty Liver genetics, Lipid Metabolism genetics, Liver metabolism, Malnutrition, Pregnancy Complications, Prenatal Exposure Delayed Effects genetics
- Abstract
Clinical and animal studies have reported an association between low birth weight and the development of nonalcoholic fatty liver disease (NAFLD) in offspring. Using a model of prenatal maternal 70% food restriction diet (FR30) in the rat, we previously showed that maternal undernutrition predisposes offspring to altered lipid metabolism in adipose tissue, especially on a high-fat (HF) diet. Here, using microarray-based expression profiling combined with metabolic, endocrine, biochemical, histological, and lipidomic approaches, we assessed whether FR30 procedure sensitizes adult male offspring to impaired lipid metabolism in the liver. No obvious differences were noted in the concentrations of triglycerides, cholesterol, and bile acids in the liver of 4-mo-old FR30 rats whichever postweaning diet was used. However, several clues suggest that offspring's lipid metabolism and steatosis are modified by maternal undernutrition. First, lipid composition was changed (i.e., higher total saturated fatty acids and lower elaidic acid) in the liver, whereas larger triglyceride droplets were observed in hepatocytes of undernourished rats. Second, FR30 offspring exhibited long - term impact on hepatic gene expression and lipid metabolism pathways on a chow diet. Although the transcriptome profile was globally modified by maternal undernutrition, cholesterol and bile acid biosynthesis pathways appear to be key targets, indicating that FR30 animals were predisposed to impaired hepatic cholesterol metabolism. Third, the FR30 protocol markedly modifies hepatic gene transcription profiles in undernourished offspring in response to postweaning HF. Overall, FR30 offspring may exhibit impaired metabolic flexibility, which does not enable them to properly cope with postweaning nutritional challenges influencing the development of nonalcoholic fatty liver.
- Published
- 2019
- Full Text
- View/download PDF
30. Bile acid alterations in nonalcoholic fatty liver disease, obesity, insulin resistance and type 2 diabetes: what do the human studies tell?
- Author
-
Chávez-Talavera O, Haas J, Grzych G, Tailleux A, and Staels B
- Subjects
- Humans, Bile Acids and Salts metabolism, Diabetes Mellitus, Type 2 metabolism, Insulin Resistance, Non-alcoholic Fatty Liver Disease metabolism, Obesity metabolism
- Abstract
Purpose of Review: The purpose of this review is to discuss the influence of obesity, insulin resistance, type 2 diabetes (T2D), and nonalcoholic fatty liver disease (NAFLD) on bile acid metabolism and to analyze whether these findings reinforce current beliefs about the role of bile acids in the pathophysiology of these diseases., Recent Findings: Discordant results on plasma bile acid alterations in NAFLD patients have been reported. Obesity, insulin resistance, and T2D, common comorbidities of NAFLD, have been associated with bile acid changes, but the individual bile acid species variations differ between studies (summarized in this review), perhaps because of clinicobiological differences between the studied patient populations and the heterogeneity of statistical analyses applied., Summary: The regulatory role of bile acids in metabolic and cellular homeostasis renders bile acids attractive candidates as players in the pathophysiology of NAFLD. However, considering the complex relationship between NAFLD, obesity, insulin resistance and T2D, it is difficult to establish clear and independent associations between bile acid alterations and these individual diseases. Though bile acid alterations may not drive NAFLD progression, signaling pathways activated by bile acids remain potent therapeutic targets for its treatment. Further studies with appropriate matching or adjustment for potential confounding factors are necessary to determine which pathophysiological conditions drive the alterations in bile acid metabolism.
- Published
- 2019
- Full Text
- View/download PDF
31. Brain insulin response and peripheral metabolic changes in a Tau transgenic mouse model.
- Author
-
Leboucher A, Ahmed T, Caron E, Tailleux A, Raison S, Joly-Amado A, Marciniak E, Carvalho K, Hamdane M, Bantubungi K, Lancel S, Eddarkaoui S, Caillierez R, Vallez E, Staels B, Vieau D, Balschun D, Buee L, and Blum D
- Subjects
- Animals, Insulin Resistance physiology, Mice, Mice, Inbred C57BL, Mice, Transgenic, tau Proteins genetics, Brain metabolism, Insulin metabolism, Tauopathies metabolism, tau Proteins metabolism
- Abstract
Accumulation of hyper-phosphorylated and aggregated Tau proteins is a neuropathological hallmark of Alzheimer's Disease (AD) and Tauopathies. AD patient brains also exhibit insulin resistance. Whereas, under normal physiological conditions insulin signaling in the brain mediates plasticity and memory formation, it can also regulate peripheral energy homeostasis. Thus, in AD, brain insulin resistance affects both cognitive and metabolic changes described in these patients. While a role of Aβ oligomers and APOE4 towards the development of brain insulin resistance emerged, contribution of Tau pathology has been largely overlooked. Our recent data demonstrated that one of the physiological function of Tau is to sustain brain insulin signaling. We postulated that under pathological conditions, hyper-phosphorylated/aggregated Tau is likely to lose this function and to favor the development of brain insulin resistance. This hypothesis was substantiated by observations from patient brains with pure Tauopathies. To address the potential link between Tau pathology and brain insulin resistance, we have evaluated the brain response to insulin in a transgenic mouse model of AD-like Tau pathology (THY-Tau22). Using electrophysiological and biochemical evaluations, we surprisingly observed that, at a time when Tau pathology and cognitive deficits are overt and obvious, the hippocampus of THY-Tau22 mice exhibits enhanced response to insulin. In addition, we demonstrated that the ability of i.c.v. insulin to promote body weight loss is enhanced in THY-Tau22 mice. In line with this, THY-Tau22 mice exhibited a lower body weight gain, hypoleptinemia and hypoinsulinemia and finally a metabolic resistance to high-fat diet. The present data highlight that the brain of transgenic Tau mice exhibit enhanced brain response to insulin. Whether these observations are ascribed to the development of Tau pathology, and therefore relevant to human Tauopathies, or unexpectedly results from the Tau transgene overexpression is debatable and discussed., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
32. Hepatic PPARα is critical in the metabolic adaptation to sepsis.
- Author
-
Paumelle R, Haas JT, Hennuyer N, Baugé E, Deleye Y, Mesotten D, Langouche L, Vanhoutte J, Cudejko C, Wouters K, Hannou SA, Legry V, Lancel S, Lalloyer F, Polizzi A, Smati S, Gourdy P, Vallez E, Bouchaert E, Derudas B, Dehondt H, Gheeraert C, Fleury S, Tailleux A, Montagner A, Wahli W, Van Den Berghe G, Guillou H, Dombrowicz D, and Staels B
- Subjects
- Animals, Bacterial Infections metabolism, Fatty Acids metabolism, Glucose metabolism, Humans, Inflammation etiology, Mice, Mice, Inbred C57BL, Adaptation, Physiological, Liver metabolism, PPAR alpha physiology, Sepsis metabolism
- Abstract
Background & Aims: Although the role of inflammation to combat infection is known, the contribution of metabolic changes in response to sepsis is poorly understood. Sepsis induces the release of lipid mediators, many of which activate nuclear receptors such as the peroxisome proliferator-activated receptor (PPAR)α, which controls both lipid metabolism and inflammation. We aimed to elucidate the previously unknown role of hepatic PPARα in the response to sepsis., Methods: Sepsis was induced by intraperitoneal injection of Escherichia coli in different models of cell-specific Ppara-deficiency and their controls. The systemic and hepatic metabolic response was analyzed using biochemical, transcriptomic and functional assays. PPARα expression was analyzed in livers from elective surgery and critically ill patients and correlated with hepatic gene expression and blood parameters., Results: Both whole body and non-hematopoietic Ppara-deficiency in mice decreased survival upon bacterial infection. Livers of septic Ppara-deficient mice displayed an impaired metabolic shift from glucose to lipid utilization resulting in more severe hypoglycemia, impaired induction of hyperketonemia and increased steatosis due to lower expression of genes involved in fatty acid catabolism and ketogenesis. Hepatocyte-specific deletion of PPARα impaired the metabolic response to sepsis and was sufficient to decrease survival upon bacterial infection. Hepatic PPARA expression was lower in critically ill patients and correlated positively with expression of lipid metabolism genes, but not with systemic inflammatory markers., Conclusion: During sepsis, Ppara-deficiency in hepatocytes is deleterious as it impairs the adaptive metabolic shift from glucose to FA utilization. Metabolic control by PPARα in hepatocytes plays a key role in the host defense against infection., Lay Summary: As the main cause of death in critically ill patients, sepsis remains a major health issue lacking efficacious therapies. While current clinical literature suggests an important role for inflammation, metabolic aspects of sepsis have mostly been overlooked. Here, we show that mice with an impaired metabolic response, due to deficiency of the nuclear receptor PPARα in the liver, exhibit enhanced mortality upon bacterial infection despite a similar inflammatory response, suggesting that metabolic interventions may be a viable strategy for improving sepsis outcomes., (Copyright © 2019 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
33. Targeting the gut microbiota with inulin-type fructans: preclinical demonstration of a novel approach in the management of endothelial dysfunction.
- Author
-
Catry E, Bindels LB, Tailleux A, Lestavel S, Neyrinck AM, Goossens JF, Lobysheva I, Plovier H, Essaghir A, Demoulin JB, Bouzin C, Pachikian BD, Cani PD, Staels B, Dessy C, and Delzenne NM
- Subjects
- Aminopeptidases genetics, Animals, Antimicrobial Cationic Peptides genetics, Bacteria drug effects, Bile Acids and Salts biosynthesis, Bile Acids and Salts blood, Carotid Arteries physiology, Cecum microbiology, Dietary Supplements, Disease Models, Animal, Fatty Acids, Omega-3 administration & dosage, Fatty Acids, Omega-3 deficiency, Gene Expression drug effects, Glucagon-Like Peptide 1 biosynthesis, Male, Mesenteric Arteries physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, ApoE, Neurotensin genetics, Nitric Oxide metabolism, Nitric Oxide Synthase metabolism, Organic Anion Transporters, Sodium-Dependent genetics, Proglucagon genetics, Symporters genetics, Vasodilation, Endothelium, Vascular drug effects, Endothelium, Vascular physiopathology, Fructans pharmacology, Gastrointestinal Microbiome drug effects, Prebiotics
- Abstract
Objective: To investigate the beneficial role of prebiotics on endothelial dysfunction, an early key marker of cardiovascular diseases, in an original mouse model linking steatosis and endothelial dysfunction., Design: We examined the contribution of the gut microbiota to vascular dysfunction observed in apolipoprotein E knockout (Apoe
-/- ) mice fed an n-3 polyunsaturated fatty acid (PUFA)-depleted diet for 12 weeks with or without inulin-type fructans (ITFs) supplementation for the last 15 days. Mesenteric and carotid arteries were isolated to evaluate endothelium-dependent relaxation ex vivo. Caecal microbiota composition (Illumina Sequencing of the 16S rRNA gene) and key pathways/mediators involved in the control of vascular function, including bile acid (BA) profiling, gut and liver key gene expression, nitric oxide and gut hormones production were also assessed., Results: ITF supplementation totally reverses endothelial dysfunction in mesenteric and carotid arteries of n-3 PUFA-depleted Apoe-/- mice via activation of the nitric oxide (NO) synthase/NO pathway. Gut microbiota changes induced by prebiotic treatment consist in increased NO-producing bacteria, replenishment of abundance in Akkermansia and decreased abundance in bacterial taxa involved in secondary BA synthesis. Changes in gut and liver gene expression also occur upon ITFs suggesting increased glucagon-like peptide 1 production and BA turnover as drivers of endothelium function preservation., Conclusions: We demonstrate for the first time that ITF improve endothelial dysfunction, implicating a short-term adaptation of both gut microbiota and key gut peptides. If confirmed in humans, prebiotics could be proposed as a novel approach in the prevention of metabolic disorders-related cardiovascular diseases., Competing Interests: Competing interests: None declared., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)- Published
- 2018
- Full Text
- View/download PDF
34. Bile Acid Alterations Are Associated With Insulin Resistance, but Not With NASH, in Obese Subjects.
- Author
-
Legry V, Francque S, Haas JT, Verrijken A, Caron S, Chávez-Talavera O, Vallez E, Vonghia L, Dirinck E, Verhaegen A, Kouach M, Lestavel S, Lefebvre P, Van Gaal L, Tailleux A, Paumelle R, and Staels B
- Subjects
- Adult, Case-Control Studies, Female, Gene Expression Regulation, Humans, Liver metabolism, Male, Middle Aged, Non-alcoholic Fatty Liver Disease complications, Non-alcoholic Fatty Liver Disease epidemiology, Obesity complications, Obesity epidemiology, Bile Acids and Salts metabolism, Insulin Resistance, Non-alcoholic Fatty Liver Disease metabolism, Obesity metabolism
- Abstract
Context: Bile acids (BAs) are signaling molecules controlling energy homeostasis that can be both toxic and protective for the liver. BA alterations have been reported in obesity, insulin resistance (IR), and nonalcoholic steatohepatitis (NASH). However, whether BA alterations contribute to NASH independently of the metabolic status is unclear., Objective: To assess BA alterations associated with NASH independently of body mass index and IR., Design and Setting: Patients visiting the obesity clinic of the Antwerp University Hospital (a tertiary referral facility) were recruited from 2006 to 2014., Patients: Obese patients with biopsy-proven NASH (n = 32) and healthy livers (n = 26) were matched on body mass index and homeostasis model assessment of IR., Main Outcome Measures: Transcriptomic analyses were performed on liver biopsies. Plasma concentrations of 21 BA species and 7α-hydroxy-4-cholesten-3-one, a marker of BA synthesis, were determined by liquid chromatography-tandem mass spectrometry. Plasma fibroblast growth factor 19 was measured by enzyme-linked immunosorbent assay., Results: Plasma BA concentrations did not correlate with any hepatic lesions, whereas, as previously reported, primary BA strongly correlated with IR. Transcriptomic analyses showed unaltered hepatic BA metabolism in NASH patients. In line, plasma 7α-hydroxy-4-cholesten-3-one was unchanged in NASH. Moreover, no sign of hepatic BA accumulation or activation of BA receptors-farnesoid X, pregnane X, and vitamin D receptors-was found. Finally, plasma fibroblast growth factor 19, secondary-to-primary BA, and free-to-conjugated BA ratios were similar, suggesting unaltered intestinal BA metabolism and signaling., Conclusions: In obese patients, BA alterations are related to the metabolic phenotype associated with NASH, especially IR, but not liver necroinflammation., (Copyright © 2017 Endocrine Society)
- Published
- 2017
- Full Text
- View/download PDF
35. Tau deletion promotes brain insulin resistance.
- Author
-
Marciniak E, Leboucher A, Caron E, Ahmed T, Tailleux A, Dumont J, Issad T, Gerhardt E, Pagesy P, Vileno M, Bournonville C, Hamdane M, Bantubungi K, Lancel S, Demeyer D, Eddarkaoui S, Vallez E, Vieau D, Humez S, Faivre E, Grenier-Boley B, Outeiro TF, Staels B, Amouyel P, Balschun D, Buee L, and Blum D
- Subjects
- Animals, Brain physiology, Cognitive Dysfunction etiology, Haplotypes, Hippocampus physiology, Humans, Insulin physiology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Brain metabolism, Insulin Resistance, tau Proteins physiology
- Abstract
The molecular pathways underlying tau pathology-induced synaptic/cognitive deficits and neurodegeneration are poorly understood. One prevalent hypothesis is that hyperphosphorylation, misfolding, and fibrillization of tau impair synaptic plasticity and cause degeneration. However, tau pathology may also result in the loss of specific physiological tau functions, which are largely unknown but could contribute to neuronal dysfunction. In the present study, we uncovered a novel function of tau in its ability to regulate brain insulin signaling. We found that tau deletion leads to an impaired hippocampal response to insulin, caused by altered IRS-1 and PTEN (phosphatase and tensin homologue on chromosome 10) activities. Our data also demonstrate that tau knockout mice exhibit an impaired hypothalamic anorexigenic effect of insulin that is associated with energy metabolism alterations. Consistently, we found that tau haplotypes are associated with glycemic traits in humans. The present data have far-reaching clinical implications and raise the hypothesis that pathophysiological tau loss-of-function favors brain insulin resistance, which is instrumental for cognitive and metabolic impairments in Alzheimer's disease patients., (© 2017 Marciniak et al.)
- Published
- 2017
- Full Text
- View/download PDF
36. Topical Intestinal Aminoimidazole Agonists of G-Protein-Coupled Bile Acid Receptor 1 Promote Glucagon Like Peptide-1 Secretion and Improve Glucose Tolerance.
- Author
-
Lasalle M, Hoguet V, Hennuyer N, Leroux F, Piveteau C, Belloy L, Lestavel S, Vallez E, Dorchies E, Duplan I, Sevin E, Culot M, Gosselet F, Boulahjar R, Herledan A, Staels B, Deprez B, Tailleux A, and Charton J
- Subjects
- Amination, Animals, Caco-2 Cells, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 metabolism, Glucose Tolerance Test, Humans, Hypoglycemic Agents pharmacokinetics, Imidazoles pharmacokinetics, Intestinal Absorption, Intestinal Mucosa metabolism, Mice, Inbred C57BL, Receptors, G-Protein-Coupled metabolism, Glucagon-Like Peptide 1 metabolism, Glucose metabolism, Hypoglycemic Agents chemistry, Hypoglycemic Agents pharmacology, Imidazoles chemistry, Imidazoles pharmacology, Receptors, G-Protein-Coupled agonists
- Abstract
The role of the G-protein-coupled bile acid receptor TGR5 in various organs, tissues, and cell types, specifically in intestinal endocrine L-cells and brown adipose tissue, has made it a promising therapeutical target in several diseases, especially type-2 diabetes and metabolic syndrome. However, recent studies have shown deleterious on-target effects of systemic TGR5 agonists. To avoid these systemic effects while stimulating glucagon-like peptide-1 (GLP-1) secreting enteroendocrine L-cells, we have designed TGR5 agonists with low intestinal permeability. In this article, we describe their synthesis, characterization, and biological evaluation. Among them, compound 24 is a potent GLP-1 secretagogue, has low effect on gallbladder volume, and improves glucose homeostasis in a preclinical murine model of diet-induced obesity and insulin resistance, making the proof of concept of the potential of topical intestinal TGR5 agonists as therapeutic agents in type-2 diabetes.
- Published
- 2017
- Full Text
- View/download PDF
37. Bile Acid Control of Metabolism and Inflammation in Obesity, Type 2 Diabetes, Dyslipidemia, and Nonalcoholic Fatty Liver Disease.
- Author
-
Chávez-Talavera O, Tailleux A, Lefebvre P, and Staels B
- Subjects
- Animals, Energy Metabolism, Gastrointestinal Microbiome, Glucose metabolism, Humans, Lipid Metabolism, Molecular Targeted Therapy, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Bile Acids and Salts metabolism, Diabetes Mellitus, Type 2 metabolism, Dyslipidemias metabolism, Inflammation metabolism, Non-alcoholic Fatty Liver Disease metabolism, Obesity metabolism
- Abstract
Bile acids are signaling molecules that coordinately regulate metabolism and inflammation via the nuclear farnesoid X receptor (FXR) and the Takeda G protein-coupled receptor 5 (TGR5). These receptors activate transcriptional networks and signaling cascades controlling the expression and activity of genes involved in bile acid, lipid and carbohydrate metabolism, energy expenditure, and inflammation by acting predominantly in enterohepatic tissues, but also in peripheral organs. In this review, we discuss the most recent findings on the inter-organ signaling and interplay with the gut microbiota of bile acids and their receptors in meta-inflammation, with a focus on their pathophysiologic roles in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic steatohepatitis, and their potential therapeutic applications., (Copyright © 2017 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
38. Anacetrapib, but not evacetrapib, impairs endothelial function in CETP-transgenic mice in spite of marked HDL-C increase.
- Author
-
Simic B, Mocharla P, Crucet M, Osto E, Kratzer A, Stivala S, Kühnast S, Speer T, Doycheva P, Princen HM, van der Hoorn JW, Jukema JW, Giral H, Tailleux A, Landmesser U, Staels B, and Lüscher TF
- Subjects
- Animals, Anticholesteremic Agents toxicity, Apolipoprotein E3 genetics, Aryldialkylphosphatase blood, Benzodiazepines toxicity, Biomarkers blood, Cholesterol Ester Transfer Proteins genetics, Cholesterol Ester Transfer Proteins metabolism, Diet, High-Fat, Disease Models, Animal, Dyslipidemias blood, Dyslipidemias genetics, Dyslipidemias physiopathology, Endothelium, Vascular metabolism, Endothelium, Vascular physiopathology, Female, Genetic Predisposition to Disease, Humans, Mice, Transgenic, Oxazolidinones toxicity, Phenotype, Reactive Oxygen Species metabolism, Triglycerides blood, Up-Regulation, Anticholesteremic Agents pharmacology, Benzodiazepines pharmacology, Cholesterol Ester Transfer Proteins antagonists & inhibitors, Cholesterol, HDL blood, Dyslipidemias drug therapy, Endothelium, Vascular drug effects, Oxazolidinones pharmacology, Vasodilation drug effects
- Abstract
Background and Aims: High-density lipoprotein cholesterol (HDL-C) is inversely related to cardiovascular risk. HDL-C raising ester transfer protein (CETP) inhibitors, are novel therapeutics. We studied the effects of CETP inhibitors anacetrapib and evacetrapib on triglycerides, cholesterol and lipoproteins, cholesterol efflux, paraoxonase activity (PON-1), reactive oxygen species (ROS), and endothelial function in E3L and E3L.CETP mice., Methods: Triglycerides and cholesterol were measured at weeks 5, 14 and 21 in E3L.CETP mice on high cholesterol diet and treated with anacetrapib (3 mg/kg/day), evacetrapib (3 mg/kg/day) or placebo. Cholesterol efflux was assessed ex-vivo in mice treated with CETP inhibitors for 3 weeks on a normal chow diet. Endothelial function was analyzed at week 21 in isolated aortic rings, and serum lipoproteins assessed by fast-performance liquid chromatography., Results: Anacetrapib and evacetrapib increased HDL-C levels (5- and 3.4-fold, resp.) and reduced triglycerides (-39% vs. placebo, p = 0.0174). Total cholesterol levels were reduced only in anacetrapib-treated mice (-32%, p = 0.0386). Cholesterol efflux and PON-1 activity (+45% and +35% vs. control, p < 0.005, resp.) were increased, while aortic ROS production was reduced with evacetrapib (-49% vs. control, p = 0.020). Anacetrapib, but not evacetrapib, impaired endothelium dependent vasorelaxation (p < 0.05). In contrast, no such effects were observed in E3L mice for all parameters tested., Conclusions: Notwithstanding a marked rise in HDL-C, evacetrapib did not improve endothelial function, while anacetrapib impaired it, suggesting that CETP inhibition does not provide vascular protection. Anacetrapib exerts unfavorable endothelial effects beyond CETP inhibition, which may explain the neutral results of large clinical trials in spite of increased HDL-C., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
39. Aβ1-40 and Aβ1-42 Plasmatic Levels In Stroke: Influence of Pre-Existing Cognitive Status and Stroke Characteristics.
- Author
-
Moulin S, Leys D, Schraen-Maschke S, Bombois S, Mendyk AM, Muhr-Tailleux A, Cordonnier C, Buee L, Pasquier F, and Bordet R
- Subjects
- Aged, Aged, 80 and over, Cognition Disorders blood, Cohort Studies, Female, Humans, Male, Statistics, Nonparametric, Amyloid beta-Peptides blood, Cognition Disorders etiology, Peptide Fragments blood, Stroke blood, Stroke complications
- Abstract
Many stroke patients have pre-existing cognitive impairment. Plasma amyloid β peptides (Aβ) - possible biomarkers of Alzheimer's pathology - induce vascular dysfunction. Our objective was to evaluate factors influencing plasma Aβ1-40 and Aβ1-42 peptides in a cohort of stroke patients. In the Biostroke study (ClinicalTrials.gov Identifier: NCT00763217), we collected vascular risk factors, neuroimaging features and biological tests including Aβ1-40 and Aβ1-42. We used the Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE) to systematically assess the pre-existing cognitive status. Of 403 patients (371 ischemia), 25 met criteria for pre-existing dementia, 142 for pre-existing cognitive decline-no-dementia, and 236 had no PCoI. Aβ1-42 was independently associated with PCoI (odds ratio 0.973; 95% confidence interval: 0.950-0.996; p=0.024). Factors associated with plasma Aβ1- 40 were age, smoking and diabetes mellitus. After exclusion of hemorrhagic strokes, the results remained unchanged, but blood samples taken less than 12 hours after onset were associated with lower plasma Aβ1-40. Our results support a dissociated response of the 2 plasma Aβ peptides in stroke patients, plasma Aβ1-40 being involved in vascular aspects whereas Aβ1-42 might be involved in neurodegenerative processes., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.)
- Published
- 2017
- Full Text
- View/download PDF
40. Lipid-lowering drugs prevent neurovascular and cognitive consequences of cardiopulmonary bypass.
- Author
-
Ouk T, Amr G, Azzaoui R, Delassus L, Fossaert E, Tailleux A, Bordet R, and Modine T
- Subjects
- Animals, Avoidance Learning drug effects, Cerebrovascular Disorders immunology, Cerebrovascular Disorders metabolism, Cerebrovascular Disorders pathology, Cognition Disorders immunology, Cognition Disorders metabolism, Cognition Disorders pathology, Endothelium, Vascular drug effects, Fenofibrate administration & dosage, Fenofibrate therapeutic use, Hemodynamics drug effects, Hypolipidemic Agents administration & dosage, Lipids blood, Male, Maze Learning drug effects, Neurons drug effects, Neurons immunology, Neurons pathology, Neuroprotective Agents administration & dosage, Rats, Sprague-Dawley, Simvastatin administration & dosage, Simvastatin therapeutic use, Tumor Necrosis Factor-alpha blood, Vasodilation drug effects, Cardiopulmonary Bypass adverse effects, Cerebrovascular Disorders prevention & control, Cognition Disorders prevention & control, Hypolipidemic Agents therapeutic use, Neuroprotective Agents therapeutic use
- Abstract
Inflammatory injury and hypoperfusion following cardiopulmonary bypass (CPB) are associated with potential brain injury in relationship between CPB, memory impairment, changes in cerebral vascular reactivity and both systemic and cerebral inflammatory reaction. The objective of this study was to assess the preventive effect of a pretreatment with simvastatin or fenofibrate on neurovascular and cognitive consequences of CPB. Male Sprague-Dawley rats were treated by control diet, simvastatin 10 mg/kg/day or fenofibrate 200 mg/kg/day for 14 days before CPB surgery and were sacrificed immediately after surgery or 24h later. Cognitive function, vascular reactivity, neuronal counts in CA1 and CA3 hippocampal regions, and inflammatory markers were assessed. CPB induced memory impairment and endothelial dysfunction 24h after surgery associated with neuronal loss. Neuronal loss was reduced by simvastatin or fenofibrate treatment in parallel to memory alteration prevention. Pretreatment by simvastatin and fenofibrate prevented CPB-induced endothelial dysfunction. CPB led to early and marked release of TNFα and overexpression of ICAM-1. Both inflammatory marker expression was decreased in the pretreated groups by lipid-lowering drugs. In a rat model of CPB, we demonstrated that simvastatin and fenofibrate protected against CPB-induced endothelial dysfunction, cerebral and systemic inflammation in parallel to memory impairment prevention., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
41. Metabolic effects of bile acid sequestration: impact on cardiovascular risk factors.
- Author
-
Spinelli V, Chávez-Talavera O, Tailleux A, and Staels B
- Subjects
- Animals, Blood Glucose drug effects, Blood Glucose metabolism, Cardiovascular Diseases etiology, Cardiovascular Diseases metabolism, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 diagnosis, Dyslipidemias complications, Dyslipidemias metabolism, Humans, Lipid Metabolism drug effects, Receptors, Cytoplasmic and Nuclear drug effects, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, G-Protein-Coupled drug effects, Receptors, G-Protein-Coupled metabolism, Risk Assessment, Risk Factors, Signal Transduction drug effects, Treatment Outcome, Bile Acids and Salts metabolism, Cardiovascular Diseases prevention & control, Diabetes Mellitus, Type 2 drug therapy, Dyslipidemias drug therapy, Sequestering Agents therapeutic use
- Abstract
Purpose of Review: This article discusses the impact of bile acid sequestrants (BAS) on cardiovascular risk factors (CVRFs), on the basis of recent (pre)clinical studies assessing the metabolic impact of modulation of enterohepatic bile acid signaling via the bile acid receptors farnesoid X receptor (FXR) and Takeda G-protein-coupled receptor 5 (TGR5)., Recent Findings: BAS decrease low-density lipoprotein-cholesterol by stimulating de novo hepatic bile acid synthesis and lowering intestinal lipid absorption, and improve glucose homeostasis in type 2 diabetes mellitus, at least in part by increasing GLP-1 production, via intestinal TGR5- and FXR-dependent mechanisms. Intestinal and peripheral FXR and TGR5 modulation also affects peripheral tissues, which can contribute to the reduction of CVRFs., Summary: Bile acids are regulators of metabolism acting in an integrated interorgan manner via FXR and TGR5. Modulation of the bile acid pool size and composition, and selective interference with their receptors could, therefore, be a therapeutic approach to decrease CVRFs. Even though clinical cardiovascular outcome studies using BAS are still lacking, the existing data point to BAS as an efficacious pharmacological approach to reduce CVRFs.
- Published
- 2016
- Full Text
- View/download PDF
42. Liver X Receptor Regulates Triglyceride Absorption Through Intestinal Down-regulation of Scavenger Receptor Class B, Type 1.
- Author
-
Briand O, Touche V, Colin S, Brufau G, Davalos A, Schonewille M, Bovenga F, Carrière V, de Boer JF, Dugardin C, Riveau B, Clavey V, Tailleux A, Moschetta A, Lasunción MA, Groen AK, Staels B, and Lestavel S
- Subjects
- Animals, Apolipoprotein B-100 metabolism, Apolipoproteins B metabolism, Benzoates pharmacology, Benzylamines pharmacology, Caco-2 Cells, Cholesterol, Dietary metabolism, Chylomicrons metabolism, DEAD-box RNA Helicases deficiency, DEAD-box RNA Helicases genetics, Down-Regulation, Humans, Hydrocarbons, Fluorinated pharmacology, Intestinal Mucosa drug effects, Jejunum drug effects, Liver X Receptors, Male, Mice, Inbred C57BL, Mice, Knockout, MicroRNAs genetics, MicroRNAs metabolism, Orphan Nuclear Receptors agonists, Protein Transport, RNA Interference, Ribonuclease III deficiency, Ribonuclease III genetics, Scavenger Receptors, Class B deficiency, Scavenger Receptors, Class B genetics, Signal Transduction, Sulfonamides pharmacology, Transcription, Genetic, Transfection, Intestinal Absorption drug effects, Intestinal Mucosa metabolism, Jejunum metabolism, Orphan Nuclear Receptors metabolism, Scavenger Receptors, Class B metabolism, Triglycerides metabolism
- Abstract
Background & Aims: Reducing postprandial triglyceridemia may be a promising strategy to lower the risk of cardiovascular disorders associated with obesity and type 2 diabetes. In enterocytes, scavenger receptor class B, type 1 (SR-B1, encoded by SCARB1) mediates lipid-micelle sensing to promote assembly and secretion of chylomicrons. The nuclear receptor subfamily 1, group H, members 2 and 3 (also known as liver X receptors [LXRs]) regulate genes involved in cholesterol and fatty acid metabolism. We aimed to determine whether intestinal LXRs regulate triglyceride absorption., Methods: C57BL/6J mice were either fed a cholesterol-enriched diet or given synthetic LXR agonists (GW3965 or T0901317). We measured the production of chylomicrons and localized SR-B1 by immunohistochemistry. Mechanisms of postprandial triglyceridemia and SR-B1 regulation were studied in Caco-2/TC7 cells incubated with LXR agonists., Results: In mice and in the Caco-2/TC7 cell line, LXR agonists caused localization of intestinal SR-B1 from apical membranes to intracellular organelles and reduced chylomicron secretion. In Caco-2/TC7 cells, LXR agonists reduced SR-B1-dependent lipidic-micelle-induced Erk phosphorylation. LXR agonists also reduced intracellular trafficking of the apical apolipoprotein B pool toward secretory compartments. LXR reduced levels of SR-B1 in Caco-2/TC7 cells via a post-transcriptional mechanism that involves microRNAs., Conclusion: In Caco-2/TC7 cells and mice, intestinal activation of LXR reduces the production of chylomicrons by a mechanism dependent on the apical localization of SR-B1., (Copyright © 2016 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
43. Screening strategy to generate cell specific recombination: a case report with the RIP-Cre mice.
- Author
-
Spinelli V, Martin C, Dorchies E, Vallez E, Dehondt H, Trabelsi MS, Tailleux A, Caron S, and Staels B
- Subjects
- Alleles, Animals, Germ Cells, Islets of Langerhans metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Polymerase Chain Reaction, Integrases genetics, Recombination, Genetic
- Abstract
Conditional gene knockout technology is a powerful tool to study the function of a gene in a specific tissue, organ or cell lineage. The most commonly used procedure applies the Cre-LoxP strategy, where the choice of the Cre driver promoter is critical to determine the efficiency and specificity of the system. However, a considered choice of an appropriate promoter does not always protect against the risk of unwanted recombination and the consequent deletion of the gene in other tissues than the desired one(s), due to phenomena of non-specific activation of the Cre transgene. Furthermore, the causes of these phenomena are not completely understood and this can potentially affect every strain of Cre-mice. In our study on the deletion of a same gene in two different tissues, we show that the incidence rate of non-specific recombination in unwanted tissues depends on the Cre driver strain, ranging from 100%, rendering it useless (aP2-Cre strain), to ~5%, which is still compatible with their use (RIP-Cre strain). The use of a simple PCR strategy conceived to detect this occurrence is indispensable when producing a tissue-specific knockout mouse. Therefore, when choosing the Cre-driver promoter, researchers not only have to be careful about its tissue-specificity and timing of activation, but should also include a systematical screening in order to exclude mice in which atypical recombination has occurred and to limit the unnecessary use of laboratory animals in uninterpretable experiments.
- Published
- 2015
- Full Text
- View/download PDF
44. The Bile Acid Chenodeoxycholic Acid Increases Human Brown Adipose Tissue Activity.
- Author
-
Broeders EP, Nascimento EB, Havekes B, Brans B, Roumans KH, Tailleux A, Schaart G, Kouach M, Charton J, Deprez B, Bouvy ND, Mottaghy F, Staels B, van Marken Lichtenbelt WD, and Schrauwen P
- Subjects
- Adipocytes, Brown drug effects, Adipocytes, Brown metabolism, Administration, Oral, Adult, Cells, Cultured, Chenodeoxycholic Acid administration & dosage, Chenodeoxycholic Acid blood, Female, Humans, Receptors, G-Protein-Coupled metabolism, Signal Transduction drug effects, Young Adult, Adipose Tissue, Brown drug effects, Adipose Tissue, Brown metabolism, Chenodeoxycholic Acid pharmacology, Energy Metabolism drug effects
- Abstract
The interest in brown adipose tissue (BAT) as a target to combat metabolic disease has recently been renewed with the discovery of functional BAT in humans. In rodents, BAT can be activated by bile acids, which activate type 2 iodothyronine deiodinase (D2) in BAT via the G-coupled protein receptor TGR5, resulting in increased oxygen consumption and energy expenditure. Here we examined the effects of oral supplementation of the bile acid chenodeoxycholic acid (CDCA) on human BAT activity. Treatment of 12 healthy female subjects with CDCA for 2 days resulted in increased BAT activity. Whole-body energy expenditure was also increased upon CDCA treatment. In vitro treatment of primary human brown adipocytes derived with CDCA or specific TGR5 agonists increased mitochondrial uncoupling and D2 expression, an effect that was absent in human primary white adipocytes. These findings identify bile acids as a target to activate BAT in humans., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
45. Bariatric surgery, lipoprotein metabolism and cardiovascular risk.
- Author
-
Tailleux A, Rouskas K, Pattou F, and Staels B
- Subjects
- Animals, Cardiovascular Diseases complications, Cardiovascular Diseases prevention & control, Humans, Risk Factors, Bariatric Surgery, Cardiovascular Diseases epidemiology, Cardiovascular Diseases metabolism, Lipoproteins metabolism
- Abstract
Purpose of Review: To summarize recent epidemiological, preclinical and clinical studies on the effects of Roux-en-Y-gastric bypass (RYGBP) surgery on cardiovascular risk factors and the underlying mechanisms., Recent Findings: Although RYGBP has mechanical effects on the gastrointestinal tract, the reduced gastric pouch and intestinal calorie absorption cannot fully explain the metabolic improvements., Summary: Obesity predisposes to cardiovascular risk factors such as dyslipidemia, type 2 diabetes, nonalcoholic fatty liver disease and hypertension. In contrast to the limited success of pharmacological and lifestyle interventions, RYGBP induces sustained weight loss, metabolic improvements and decreases morbidity/mortality. In line, RYGBP reduces cardiovascular risk factors. Although the mechanisms are not entirely understood, RYGBP induces complex changes in the gut affecting other organs through endocrine and metabolic signals from the intestine to all key metabolic organs, which can link RYGBP and decreased cardiovascular risk. Here, we discuss the roles of changes in lipid absorption and metabolism, bile acid metabolism, gut hormones and the microbiote as potential mechanisms in the decreased cardiovascular risk and metabolic improvement after RYGBP.
- Published
- 2015
- Full Text
- View/download PDF
46. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells.
- Author
-
Trabelsi MS, Daoudi M, Prawitt J, Ducastel S, Touche V, Sayin SI, Perino A, Brighton CA, Sebti Y, Kluza J, Briand O, Dehondt H, Vallez E, Dorchies E, Baud G, Spinelli V, Hennuyer N, Caron S, Bantubungi K, Caiazzo R, Reimann F, Marchetti P, Lefebvre P, Bäckhed F, Gribble FM, Schoonjans K, Pattou F, Tailleux A, Staels B, and Lestavel S
- Subjects
- Animals, Anticholesteremic Agents pharmacology, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Bile Acids and Salts metabolism, Blood Glucose metabolism, Colesevelam Hydrochloride pharmacology, Colon cytology, Colon metabolism, Diet, High-Fat, Glucagon-Like Peptide 1 metabolism, Glycolysis, Humans, Ileum cytology, Ileum metabolism, Insulin metabolism, Insulin Secretion, Insulin-Secreting Cells metabolism, Intestines cytology, Jejunum cytology, Jejunum metabolism, Mice, Mice, Knockout, Mice, Obese, Nuclear Proteins metabolism, Obesity genetics, Obesity metabolism, Proglucagon drug effects, Proglucagon genetics, Proglucagon metabolism, Receptors, G-Protein-Coupled genetics, Sequestering Agents pharmacology, Signal Transduction, Transcription Factors metabolism, Enteroendocrine Cells metabolism, Glucagon-Like Peptide 1 genetics, Intestinal Mucosa metabolism, RNA, Messenger metabolism, Receptors, Cytoplasmic and Nuclear genetics
- Abstract
Bile acids are signalling molecules, which activate the transmembrane receptor TGR5 and the nuclear receptor FXR. BA sequestrants (BAS) complex bile acids in the intestinal lumen and decrease intestinal FXR activity. The BAS-BA complex also induces glucagon-like peptide-1 (GLP-1) production by L cells which potentiates β-cell glucose-induced insulin secretion. Whether FXR is expressed in L cells and controls GLP-1 production is unknown. Here, we show that FXR activation in L cells decreases proglucagon expression by interfering with the glucose-responsive factor Carbohydrate-Responsive Element Binding Protein (ChREBP) and GLP-1 secretion by inhibiting glycolysis. In vivo, FXR deficiency increases GLP-1 gene expression and secretion in response to glucose hence improving glucose metabolism. Moreover, treatment of ob/ob mice with the BAS colesevelam increases intestinal proglucagon gene expression and improves glycaemia in a FXR-dependent manner. These findings identify the FXR/GLP-1 pathway as a new mechanism of BA control of glucose metabolism and a pharmacological target for type 2 diabetes.
- Published
- 2015
- Full Text
- View/download PDF
47. Rapid and body weight-independent improvement of endothelial and high-density lipoprotein function after Roux-en-Y gastric bypass: role of glucagon-like peptide-1.
- Author
-
Osto E, Doytcheva P, Corteville C, Bueter M, Dörig C, Stivala S, Buhmann H, Colin S, Rohrer L, Hasballa R, Tailleux A, Wolfrum C, Tona F, Manz J, Vetter D, Spliethoff K, Vanhoutte PM, Landmesser U, Pattou F, Staels B, Matter CM, Lutz TA, and Lüscher TF
- Subjects
- Adult, Animals, Antioxidants physiology, Case-Control Studies, Cells, Cultured, Diet, High-Fat adverse effects, Disease Models, Animal, Endothelium, Vascular pathology, Female, Gastric Bypass, Humans, Male, Nitric Oxide physiology, Obesity physiopathology, Oxidative Stress physiology, Proto-Oncogene Proteins c-akt physiology, Rats, Rats, Wistar, Signal Transduction, Treatment Outcome, Body Weight physiology, Endothelium, Vascular physiology, Glucagon-Like Peptide 1 physiology, Lipoproteins, HDL physiology, Obesity surgery, Weight Loss physiology
- Abstract
Background: Roux-en-Y gastric bypass (RYGB) reduces body weight and cardiovascular mortality in morbidly obese patients. Glucagon-like peptide-1 (GLP-1) seems to mediate the metabolic benefits of RYGB partly in a weight loss-independent manner. The present study investigated in rats and patients whether obesity-induced endothelial and high-density lipoprotein (HDL) dysfunction is rapidly improved after RYGB via a GLP-1-dependent mechanism., Methods and Results: Eight days after RYGB in diet-induced obese rats, higher plasma levels of bile acids and GLP-1 were associated with improved endothelium-dependent relaxation compared with sham-operated controls fed ad libitum and sham-operated rats that were weight matched to those undergoing RYGB. Compared with the sham-operated rats, RYGB improved nitric oxide (NO) bioavailability resulting from higher endothelial Akt/NO synthase activation, reduced c-Jun amino terminal kinase phosphorylation, and decreased oxidative stress. The protective effects of RYGB were prevented by the GLP-1 receptor antagonist exendin9-39 (10 μg·kg(-1)·h(-1)). Furthermore, in patients and rats, RYGB rapidly reversed HDL dysfunction and restored the endothelium-protective properties of the lipoprotein, including endothelial NO synthase activation, NO production, and anti-inflammatory, antiapoptotic, and antioxidant effects. Finally, RYGB restored HDL-mediated cholesterol efflux capacity. To demonstrate the role of increased GLP-1 signaling, sham-operated control rats were treated for 8 days with the GLP-1 analog liraglutide (0.2 mg/kg twice daily), which restored NO bioavailability and improved endothelium-dependent relaxations and HDL endothelium-protective properties, mimicking the effects of RYGB., Conclusions: RYGB rapidly reverses obesity-induced endothelial dysfunction and restores the endothelium-protective properties of HDL via a GLP-1-mediated mechanism. The present translational findings in rats and patients unmask novel, weight-independent mechanisms of cardiovascular protection in morbid obesity., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
48. Cholesterol uptake disruption, in association with chemotherapy, is a promising combined metabolic therapy for pancreatic adenocarcinoma.
- Author
-
Guillaumond F, Bidaut G, Ouaissi M, Servais S, Gouirand V, Olivares O, Lac S, Borge L, Roques J, Gayet O, Pinault M, Guimaraes C, Nigri J, Loncle C, Lavaut MN, Garcia S, Tailleux A, Staels B, Calvo E, Tomasini R, Iovanna JL, and Vasseur S
- Subjects
- Adenocarcinoma enzymology, Adenocarcinoma pathology, Animals, Cell Compartmentation drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Clone Cells, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Epithelial Cells drug effects, Epithelial Cells metabolism, Epithelial Cells pathology, Extracellular Signal-Regulated MAP Kinases metabolism, Gene Expression Regulation, Neoplastic drug effects, Gene Silencing drug effects, Humans, Lipoproteins metabolism, MAP Kinase Signaling System drug effects, Metabolic Networks and Pathways drug effects, Metabolic Networks and Pathways genetics, Mice, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms pathology, Phenotype, Prognosis, Receptors, LDL genetics, Receptors, LDL metabolism, Up-Regulation drug effects, Up-Regulation genetics, Gemcitabine, Pancreatic Neoplasms, Adenocarcinoma drug therapy, Adenocarcinoma metabolism, Cholesterol metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism
- Abstract
The malignant progression of pancreatic ductal adenocarcinoma (PDAC) is accompanied by a profound desmoplasia, which forces proliferating tumor cells to metabolically adapt to this new microenvironment. We established the PDAC metabolic signature to highlight the main activated tumor metabolic pathways. Comparative transcriptomic analysis identified lipid-related metabolic pathways as being the most highly enriched in PDAC, compared with a normal pancreas. Our study revealed that lipoprotein metabolic processes, in particular cholesterol uptake, are drastically activated in the tumor. This process results in an increase in the amount of cholesterol and an overexpression of the low-density lipoprotein receptor (LDLR) in pancreatic tumor cells. These findings identify LDLR as a novel metabolic target to limit PDAC progression. Here, we demonstrate that shRNA silencing of LDLR, in pancreatic tumor cells, profoundly reduces uptake of cholesterol and alters its distribution, decreases tumor cell proliferation, and limits activation of ERK1/2 survival pathway. Moreover, blocking cholesterol uptake sensitizes cells to chemotherapeutic drugs and potentiates the effect of chemotherapy on PDAC regression. Clinically, high PDAC Ldlr expression is not restricted to a specific tumor stage but is correlated to a higher risk of disease recurrence. This study provides a precise overview of lipid metabolic pathways that are disturbed in PDAC. We also highlight the high dependence of pancreatic cancer cells upon cholesterol uptake, and identify LDLR as a promising metabolic target for combined therapy, to limit PDAC progression and disease patient relapse.
- Published
- 2015
- Full Text
- View/download PDF
49. The Sirt1 activator SRT3025 provides atheroprotection in Apoe-/- mice by reducing hepatic Pcsk9 secretion and enhancing Ldlr expression.
- Author
-
Miranda MX, van Tits LJ, Lohmann C, Arsiwala T, Winnik S, Tailleux A, Stein S, Gomes AP, Suri V, Ellis JL, Lutz TA, Hottiger MO, Sinclair DA, Auwerx J, Schoonjans K, Staels B, Lüscher TF, and Matter CM
- Subjects
- Anilides pharmacology, Animals, Anticholesteremic Agents pharmacology, Apolipoproteins E deficiency, Cells, Cultured, Cholesterol, LDL metabolism, Enzyme Inhibitors pharmacology, Hepatocytes drug effects, In Vitro Techniques, Male, Mice, Inbred C57BL, Proprotein Convertase 9, RNA, Messenger metabolism, Receptors, LDL drug effects, Thiazoles pharmacology, Arteriosclerosis prevention & control, Hepatocytes metabolism, Proprotein Convertases metabolism, Receptors, LDL metabolism, Serine Endopeptidases metabolism, Sirtuin 1 metabolism
- Abstract
Aims: The deacetylase sirtuin 1 (Sirt1) exerts beneficial effects on lipid metabolism, but its roles in plasma LDL-cholesterol regulation and atherosclerosis are controversial. Thus, we applied the pharmacological Sirt1 activator SRT3025 in a mouse model of atherosclerosis and in hepatocyte culture., Methods and Results: Apolipoprotein E-deficient (Apoe(-/-)) mice were fed a high-cholesterol diet (1.25% w/w) supplemented with SRT3025 (3.18 g kg(-1) diet) for 12 weeks. In vitro, the drug activated wild-type Sirt1 protein, but not the activation-resistant Sirt1 mutant; in vivo, it increased deacetylation of hepatic p65 and skeletal muscle Foxo1. SRT3025 treatment decreased plasma levels of LDL-cholesterol and total cholesterol and reduced atherosclerosis. Drug treatment did not change mRNA expression of hepatic LDL receptor (Ldlr) and proprotein convertase subtilisin/kexin type 9 (Pcsk9), but increased their protein expression indicating post-translational effects. Consistent with hepatocyte Ldlr and Pcsk9 accumulation, we found reduced plasma levels of Pcsk9 after pharmacological Sirt1 activation. In vitro administration of SRT3025 to cultured AML12 hepatocytes attenuated Pcsk9 secretion and its binding to Ldlr, thereby reducing Pcsk9-mediated Ldlr degradation and increasing Ldlr expression and LDL uptake. Co-administration of exogenous Pcsk9 with SRT3025 blunted these effects. Sirt1 activation with SRT3025 in Ldlr(-/-) mice reduced neither plasma Pcsk9, nor LDL-cholesterol levels, nor atherosclerosis., Conclusion: We identify reduction in Pcsk9 secretion as a novel effect of Sirt1 activity and uncover Ldlr as a prerequisite for Sirt1-mediated atheroprotection in mice. Pharmacological activation of Sirt1 appears promising to be tested in patients for its effects on plasma Pcsk9, LDL-cholesterol, and atherosclerosis., (© The Author 2014. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2015
- Full Text
- View/download PDF
50. The transrepressive activity of peroxisome proliferator-activated receptor alpha is necessary and sufficient to prevent liver fibrosis in mice.
- Author
-
Pawlak M, Baugé E, Bourguet W, De Bosscher K, Lalloyer F, Tailleux A, Lebherz C, Lefebvre P, and Staels B
- Subjects
- Animals, Gene Expression, Lipopolysaccharides, Liver Cirrhosis metabolism, Liver Cirrhosis prevention & control, Mice, Inbred C57BL, Mutation, PPAR alpha agonists, PPAR alpha genetics, Signal Transduction, Fatty Liver complications, Liver Cirrhosis etiology, PPAR alpha metabolism
- Abstract
Unlabelled: Nonalcoholic fatty liver disease (NAFLD) is increasingly prevalent and strongly associated with central obesity, dyslipidemia, and insulin resistance. According to the multiple-hit model of NAFLD pathogenesis, lipid accumulation drives nonalcoholic steatohepatitis (NASH) initiation by triggering oxidative stress, lipotoxicity, and subsequent activation of hepatic inflammatory responses that may progress, in predisposed individuals, to fibrosis and cirrhosis. While there is an unmet therapeutical need for NASH and fibrosis, recent preclinical studies showed that peroxisome proliferator-activated receptor (PPAR)-α agonism can efficiently oppose these symptoms. To dissect the relative contribution of antisteatotic versus anti-inflammatory PPAR-α activities in counteracting dietary-induced liver fibrosis, we used a PPAR-α mutant lacking its DNA-binding-dependent activity on fatty acid metabolism. Liver-specific expression of wild-type or a DNA-binding-deficient PPAR-α in acute and chronic models of inflammation were used to study PPAR-α's anti-inflammatory versus metabolic activities in NASH and fibrosis. Pharmacologically activated PPAR-α inhibited hepatic inflammatory responses and the transition from steatosis toward NASH and fibrosis through a direct, anti-inflammatory mechanism independent of its lipid handling properties., Conclusion: The transrepression activity of PPAR-α on chronic liver inflammation is sufficient to prevent progression of NASH to liver fibrosis. Dissociated PPAR-α agonists, selectively modulating PPAR-α transrepression activity, could thus be an option to prevent NASH and fibrosis progression., (© 2014 by the American Association for the Study of Liver Diseases.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.