Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35205709

ABSTRACT

An elevated neutrophil-lymphocyte ratio negatively predicts the outcome of patients with cancer and is associated with cachexia, the terminal wasting syndrome. Here, using murine model systems of colorectal and pancreatic cancer we show that neutrophilia in the circulation and multiple organs, accompanied by extramedullary hematopoiesis, is an early event during cancer progression. Transcriptomic and metabolic assessment reveals that neutrophils in tumor-bearing animals utilize aerobic glycolysis, similar to cancer cells. Although pharmacological inhibition of aerobic glycolysis slows down tumor growth in C26 tumor-bearing mice, it precipitates cachexia, thereby shortening the overall survival. This negative effect may be explained by our observation that acute depletion of neutrophils in pre-cachectic mice impairs systemic glucose homeostasis secondary to altered hepatic lipid processing. Thus, changes in neutrophil number, distribution, and metabolism play an adaptive role in host metabolic homeostasis during cancer progression. Our findings provide insight into early events during cancer progression to cachexia, with implications for therapy.

2.
Annu Rev Biochem ; 88: 247-280, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30901264

ABSTRACT

The complexity of human cancer underlies its devastating clinical consequences. Drugs designed to target the genetic alterations that drive cancer have improved the outcome for many patients, but not the majority of them. Here, we review the genomic landscape of cancer, how genomic data can provide much more than a sum of its parts, and the approaches developed to identify and validate genomic alterations with potential therapeutic value. We highlight notable successes and pitfalls in predicting the value of potential therapeutic targets and discuss the use of multi-omic data to better understand cancer dependencies and drug sensitivity. We discuss how integrated approaches to collecting, curating, and sharing these large data sets might improve the identification and prioritization of cancer vulnerabilities as well as patient stratification within clinical trials. Finally, we outline how future approaches might improve the efficiency and speed of translating genomic data into clinically effective therapies and how the use of unbiased genome-wide information can identify novel predictive biomarkers that can be either simple or complex.


Subject(s)
Genomics , Mutation , Neoplasms/drug therapy , Humans , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine
3.
Sci Rep ; 6: 39278, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27995969

ABSTRACT

The proliferative-crypt compartment of the intestinal epithelium is enriched in phospholipids and accumulation of phospholipids has been described in colorectal tumors. Here we hypothesize that biliary phospholipid flow could directly contribute to the proliferative power of normal and dysplastic enterocytes. We used Abcb4-/- mice which lack biliary phospholipid secretion. We first show that Abcb4-/- mice are protected against intestinal tumorigenesis. At the molecular level, the transcriptional activity of the nuclear receptor Liver Receptor Homolog-1 (Lrh1) is reduced in Abcb4-/- mice and its re-activation re-establishes a tumor burden comparable to control mice. Feeding Abcb4-/- mice a diet supplemented with phospholipids completely overcomes the intestinal tumor protective phenotype, thus corroborating the hypothesis that the absence of biliary phospholipids and not lack of Abcb4 gene per se is responsible for the protection. In turn, phospholipids cannot re-establish intestinal tumorigenesis in Abcb4-/- mice crossed with mice with intestinal specific ablation of Lrh1, a nuclear hormone receptor that is activates by phospholipids. Our data identify the key role of biliary phospholipids in sustaining intestinal mucosa proliferation and tumor progression through the activation of nuclear receptor Lrh1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Carcinogenesis/pathology , Enterocytes/metabolism , Intestinal Neoplasms/pathology , Phospholipids/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Biliary Tract/metabolism , Cell Proliferation , Cyclin D1/biosynthesis , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Mice , Mice, Knockout , Proliferating Cell Nuclear Antigen/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , ATP-Binding Cassette Sub-Family B Member 4
5.
Genes Dev ; 30(5): 489-501, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26944676

ABSTRACT

Metabolic dysfunction contributes to the clinical deterioration observed in advanced cancer patients and is characterized by weight loss, skeletal muscle wasting, and atrophy of the adipose tissue. This systemic syndrome, termed cancer-associated cachexia (CAC), is a major cause of morbidity and mortality. While once attributed solely to decreased food intake, the present description of cancer cachexia is a disorder of multiorgan energy imbalance. Here we review the molecules and pathways responsible for metabolic dysfunction in CAC and the ideas that led to the current understanding.


Subject(s)
Cachexia/etiology , Cachexia/physiopathology , Neoplasms/complications , Adipose Tissue, White/physiopathology , Carbohydrate Metabolism/physiology , Endocrine System/physiopathology , Humans , Inflammation/complications , Lipid Metabolism , Liver/physiopathology , Muscular Atrophy/etiology
7.
Cell Metab ; 20(3): 433-47, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25043816

ABSTRACT

Cancer-associated cachexia (CAC) is a wasting syndrome characterized by systemic inflammation, body weight loss, atrophy of white adipose tissue (WAT) and skeletal muscle. Limited therapeutic options are available and the underlying mechanisms are poorly defined. Here we show that a phenotypic switch from WAT to brown fat, a phenomenon termed WAT browning, takes place in the initial stages of CAC, before skeletal muscle atrophy. WAT browning is associated with increased expression of uncoupling protein 1 (UCP1), which uncouples mitochondrial respiration toward thermogenesis instead of ATP synthesis, leading to increased lipid mobilization and energy expenditure in cachectic mice. Chronic inflammation and the cytokine interleukin-6 increase UCP1 expression in WAT, and treatments that reduce inflammation or ß-adrenergic blockade reduce WAT browning and ameliorate the severity of cachexia. Importantly, UCP1 staining is observed in WAT from CAC patients. Thus, inhibition of WAT browning represents a promising approach to ameliorate cachexia in cancer patients.


Subject(s)
Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Cachexia/complications , Neoplasms/complications , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Cachexia/immunology , Cachexia/metabolism , Cachexia/pathology , Energy Metabolism , Humans , Inflammation/complications , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Ion Channels/analysis , Mice , Mitochondrial Proteins/analysis , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Uncoupling Protein 1
8.
Gastroenterology ; 142(2): 355-65.e1-4, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22057115

ABSTRACT

BACKGROUND & AIMS: Cholestasis is a liver disorder characterized by impaired bile flow, reduction of bile acids (BAs) in the intestine, and retention of BAs in the liver. The farnesoid X receptor (FXR) is the transcriptional regulator of BA homeostasis. Activation of FXR by BAs reduces circulating BA levels in a feedback mechanism, repressing hepatic cholesterol 7α-hydroxylase (Cyp7a1), the rate-limiting enzyme for the conversion of cholesterol to BAs. This mechanism involves the hepatic nuclear receptor small heterodimer partner and the intestinal fibroblast growth factor (FGF) 19 and 15. We investigated the role of activation of intestine-specific FXR in reducing hepatic levels of BAs and protecting the liver from cholestasis in mice. METHODS: We generated transgenic mice that express a constitutively active FXR in the intestine. Using FXR gain- and loss-of-function models, we studied the roles of intestinal FXR in mice with intrahepatic and extrahepatic cholestasis. RESULTS: Selective activation of intestinal FXR induced FGF15 and repressed hepatic Cyp7a1, reducing the pool size of BAs and changing the BA pool composition. Activation of intestinal FXR protected mice from obstructive extrahepatic cholestasis after bile duct ligation or administration of α-naphthylisothiocyanate. In Mdr2(-/-) mice, transgenic expression of activated FXR in the intestine protected against liver damage, whereas absence of FXR promoted progression of liver disease. CONCLUSIONS: Activation of FXR transcription in the intestine protects the liver from cholestasis in mice by inducing FGF15 expression and reducing the hepatic pool of BA; this approach might be developed to reverse cholestasis in patients.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/prevention & control , Intestinal Mucosa/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcriptional Activation , Animals , Cholestasis/metabolism , Cholestasis/pathology , Cholesterol 7-alpha-Hydroxylase/metabolism , Fibroblast Growth Factors/metabolism , Liver/pathology , Male , Mice , Mice, Transgenic , Random Allocation , Receptors, Cytoplasmic and Nuclear/genetics
9.
Eur J Med Chem ; 46(10): 5218-24, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21889235

ABSTRACT

A new series of gemfibrozil analogues conjugated with α-asarone, trans-stilbene, chalcone, and their bioisosteric modifications were synthesized and evaluated to develop PPARα agonists. In this attempt, we have removed the methyls on the phenyl ring of gemfibrozil and introduced the above scaffolds in para position synthesizing two series of derivatives, keeping the dimethylpentanoic skeleton of gemfibrozil unaltered or demethylated. Four compounds exhibited good activation of the PPARα receptor and were also screened for their activity on PPARα-regulated gene CPT1A.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Fatty Acids/metabolism , Gemfibrozil/analogs & derivatives , Gemfibrozil/pharmacology , PPAR alpha/agonists , PPAR alpha/metabolism , Hep G2 Cells , Humans , Up-Regulation/drug effects
10.
Cell Metab ; 12(2): 187-93, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20674863

ABSTRACT

Several steps of the HDL-mediated reverse cholesterol transport (RCT) are transcriptionally regulated by the nuclear receptors LXRs in the macrophages, liver, and intestine. Systemic LXR activation via synthetic ligands induces RCT but also causes increased hepatic fatty acid synthesis and steatosis, limiting the potential therapeutic use of LXR agonists. During the last few years, the participation of the intestine in the control of RCT has appeared more evident. Here we show that while hepatic-specific LXR activation does not contribute to RCT, intestinal-specific LXR activation leads to decreased intestinal cholesterol absorption, improved lipoprotein profile, and increased RCT in vivo in the absence of hepatic steatosis. These events protect against atherosclerosis in the background of the LDLR-deficient mice. Our study fully characterizes the molecular and metabolic scenario that elects the intestine as a key player in the LXR-driven protective environment against cardiovascular disease.


Subject(s)
Atherosclerosis/prevention & control , Cholesterol/metabolism , Intestinal Mucosa/metabolism , Orphan Nuclear Receptors/metabolism , Animals , Biological Transport , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orphan Nuclear Receptors/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism
11.
Cell Metab ; 11(5): 345-6, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20444415

ABSTRACT

Nutritional and genetic factors alter the intestinal microflora, predisposing individuals toward metabolic syndrome. Gewirtz and coworkers (Vijay-Kumar et al., 2010), employing Toll-like receptor 5 null mice, present evidence for a direct relationship between malfunction of the innate immune system, changes in gut microbiota composition, and development of metabolic syndrome.

12.
J Med Chem ; 52(20): 6224-32, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19791744

ABSTRACT

A series of 2-heteroarylthioalkanoic acids were synthesized through systematic structural modifications of clofibric acid and evaluated for human peroxisome proliferator-activated receptor alpha (PPARalpha) transactivation activity, with the aim of obtaining new hypolipidemic compounds. Some thiophene and benzothiazole derivatives showing a good activation of the receptor alpha were screened for activity against the PPARgamma isoform. The gene induction of selected compounds was also investigated in the human hepatoma cell line.


Subject(s)
Clofibric Acid/analogs & derivatives , Clofibric Acid/pharmacology , PPAR alpha/agonists , Sulfur/chemistry , Cell Line, Tumor , Clofibric Acid/chemical synthesis , Clofibric Acid/chemistry , Humans , Oxygen/chemistry , PPAR alpha/genetics , Stereoisomerism , Transcriptional Activation
13.
FEBS Lett ; 583(8): 1274-80, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19303409

ABSTRACT

Intraluminal phospholipids affect micellar solubilization and absorption of cholesterol. We here study cholesterol transport from taurocholate-phospholipid-cholesterol micelles to CaCo2 cells, and associated effects on ABC-A1 mediated cholesterol efflux. Micellar incorporation of egg-yolk-phosphatidylcholine markedly increased apical retention of the sterol with decreased expression of ABC-A1, an effect that is prevented by synthetic liver X receptor (LXR) or retinoid X receptor (RXR) agonists. On the other hand, incorporation of lyso-phosphatidylcholine (LysoPC) increased ABC-A1-HDL-dependent basolateral cholesterol efflux, an effect that is abated when LXR is silenced. Thus, the modulation of cholesterol metabolism via intraluminal phospholipids is related to the activity of the oxysterol nuclear receptor LXR.


Subject(s)
Cholesterol/metabolism , DNA-Binding Proteins/metabolism , Lipid Metabolism , Micelles , Receptors, Cytoplasmic and Nuclear/metabolism , Biological Transport , Caco-2 Cells , DNA-Binding Proteins/genetics , Humans , Liver X Receptors , Orphan Nuclear Receptors , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/genetics
14.
Toxicol Lett ; 182(1-3): 36-41, 2008 Nov 10.
Article in English | MEDLINE | ID: mdl-18771718

ABSTRACT

Mechanisms by which hydrophobic bile salts cause tissue changes below their critical micellar concentration (CMC, 1-2mM) and above (4-8mM) remain poorly understood. In this study, rat colonic mucosa was exposed to different concentrations of taurodeoxycholate (TDC), t-butyl-hydroperoxide (t-BH) or glutathione ester with or without pre-incubation with 2mM TDC. Exposure to 2mM TDC was associated with 10% higher tissue levels of total glutathione (GSH, basal values: 33.7+/-3.3 nmol/mg prot). With TDC 8mM, GSH decreased to 16.4+/-2.3 nmol/mg prot (P<0.05), oxidized glutathione (GSSG) increased by 60% (P<0.05), glutathione peroxidase (GSH-Px) and reductase activities were threefold increased, protein carbonyls fourfold increased, protein sulfhydrils decreased by 78%, lactate dehydrogenase (LDH) and GSSG release in the incubation medium were sixfold higher. In 2mM TDC pre-treated tissues, the subsequent incubation with 8mM TDC induced a lower loss of tissue GSH, and a lower release of LDH and GSSG. Pre-incubation with 2mM TDC partly protected against t-BH toxicity, while glutathione ester protected against 8mM TDC toxicity. In conclusion, TDC exposure causes opposite effects depending on CMC: induction of antioxidant protective systems including glutathione system (pre-conditioning effect) was observed with TDC below CMC, oxidative damages pointing to decreased mucosal detoxification potential with above CMC.


Subject(s)
Cholagogues and Choleretics/toxicity , Intestinal Diseases/chemically induced , Ischemic Preconditioning , Oxidative Stress/drug effects , Taurodeoxycholic Acid/toxicity , Animals , Cholagogues and Choleretics/chemistry , Glutathione/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/pathology , L-Lactate Dehydrogenase/metabolism , Male , Micelles , Oxidation-Reduction , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Sulfhydryl Compounds/metabolism , Taurodeoxycholic Acid/chemistry
15.
Gastroenterology ; 134(7): 2091-100, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18466903

ABSTRACT

BACKGROUNDS & AIMS: ATP8B1 is a phosphatidylserine flippase in the canalicular membrane; patients with mutations in ATP8B1 develop severe chronic (PFIC1) or periodic (BRIC1) cholestatic liver disease. We have observed that Atp8b1 deficiency leads to enhanced biliary cholesterol excretion. It has been established that biliary cholesterol excretion depends on transport by the heterodimer Abcg5/Abcg8. We hypothesized that the increased cholesterol output was due to enhanced extraction from the altered canalicular membrane rather than to higher Abcg5/Abcg8 activity. We therefore studied the relation between Abcg5/Abcg8 expression and biliary cholesterol excretion in mice lacking Atp8b1, Abcg8, or both (GF mice). METHODS: Bile formation was studied in LXR agonist-fed wild-type mice as well as mice lacking Atp8b1 or Abcg8, or in GF mice upon infusion of taurocholate. Bile samples were analyzed for cholesterol, bile salt, phospholipids, and ectoenzyme content. RESULTS: LXR agonist increased Abcg5/8 expression, and this was accompanied by increased biliary cholesterol output in both wild-type and Atp8b1(G308V/G308V) mice. However, Atp8b1(G308V/G308V) mice maintained higher cholesterol output. Although in Abcg8(-/-) mice biliary cholesterol output was severely reduced, GF mice displayed high biliary cholesterol output, which was comparable with wild-type mice. Bile of both Atp8b1(G308V/G308V) and GF mice displayed elevated levels of phosphatidylserine and sphingomyelin, indicating membrane stress. CONCLUSIONS: Our data demonstrate that the increased biliary cholesterol excretion in Atp8b1-deficient mice is independent of Abcg5/8 activity. This implicates that Atp8b1 deficiency leads to a decrease in the detergent resistance and subsequent nonspecific extraction of cholesterol from the canalicular membrane by bile salts.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/metabolism , Bile/metabolism , Cholesterol/metabolism , Lipoproteins/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Bile/enzymology , Bile Acids and Salts/blood , Bile Canaliculi/metabolism , Cell Membrane/metabolism , DNA-Binding Proteins/agonists , DNA-Binding Proteins/metabolism , Hydrocarbons, Fluorinated , Lipoproteins/deficiency , Lipoproteins/genetics , Liver/drug effects , Liver/enzymology , Liver X Receptors , Male , Membrane Fluidity , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Phospholipid Transfer Proteins , Phospholipids/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfonamides/pharmacology , Time Factors , Up-Regulation
16.
Biochim Biophys Acta ; 1781(3): 79-96, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18194677

ABSTRACT

Bile formation springs from an extraordinary sophisticated secretory network, which combines the activity of transport proteins with the physico-chemical properties of small albeit powerful lipophilic compounds. This robust interplay is activated in response to dietary stimuli, circadian rhythms, and metabolic demands to regulate cholesterol disposal, lipid digestion and absorption in the enterohepatic system. As a result, bile flow is a complex multi-organ effort that requires an integrated flux of information between liver and intestine. A coordinate regulatory task is achieved by nuclear receptors, which are ligand activated transcription factors, responsible for the coherent activation of sets of genes involved in multiple physiological actions, including hepato-biliary homeostasis. Mastering the molecular pathways underlying functional and pharmacological modulation of bile flow has great translational value for potential future treatment of cholestasis and cholelithiasis. In this review, we focus on recent discoveries in the functional biology of bile formation with the explicit aim of underlining their putative clinical relevance.


Subject(s)
Biliary Tract/metabolism , Lipid Metabolism , Animals , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Bile Canaliculi/metabolism , Disease , Gene Expression Regulation , Humans
17.
Hepatology ; 46(6): 2040-2, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18046711
18.
Curr Pharm Des ; 13(21): 2199-207, 2007.
Article in English | MEDLINE | ID: mdl-17627553

ABSTRACT

The metabolic syndrome is an emerging global epidemic characterized by clustering of metabolic abnormalities leading to increased cardiovascular risk: glucose intolerance or type 2 diabetes, dyslipidemia, hypertension, and "central" obesity. Scientists are decoding and piecing together the molecular texture underlying the metabolic syndrome: insulin resistance and dyslipidemia stand out as central pathophysiological events. In this picture, the liver rises as the leading organ in the maintenance of metabolic fitness; it serves as the first relay station for processing dietary information, and encloses the whole biochemical machinery for both glucose and lipid storage and disposal. In addition, the liver is a target of the different endocrine molecules secreted by pancreatic beta-cells and adipose tissue. Evidence collected in animal models supports the central role of the liver in the metabolic syndrome. While specific bereft of insulin sensitivity in skeletal muscle and adipose tissue fails to induce diabetes at certain extent, this is constantly the outcome in case of hepatic insulin resistance. Also, dyslipidemia is currently interpreted as the result of increased flux of free fatty acids to the liver with ensuing misbalance of lipoprotein synthesis and removal. In this review we bring together recent advances in the field of lipid sensing nuclear receptors, adipokines and other molecules responsible for metabolic fitness, and provide a putative coherent frame to conceive the pathophysiology of the metabolic syndrome.


Subject(s)
Disease Models, Animal , Liver/physiology , Metabolic Syndrome/etiology , Adiponectin/physiology , Animals , Carbohydrate Metabolism , Dyslipidemias/etiology , Humans , Insulin Resistance , Leptin/physiology , Lipid Metabolism , PPAR alpha/physiology , PPAR gamma/physiology
19.
Clin Biochem ; 40(8): 503-10, 2007 May.
Article in English | MEDLINE | ID: mdl-17321514

ABSTRACT

The strong analgesic, anti-inflammatory effects of non-steroidal anti-inflammatory drugs (NSAIDs) are hampered by high occurrence of gastrointestinal side effects. Therapeutic actions of NSAIDs result from cyclooxygenase (COX) enzymes inhibition with reduced synthesis of prostaglandins, major modulators of inflammation. Since prostaglandins also regulate key events in gut homeostasis -mucosal secretion, blood flow, epithelial regeneration - COX inhibition has been accepted as the reason for NSAID gastrointestinal toxicity. Several findings challenge this theory: first, intestinal damage by NSAIDs occurs also in COX-1 knockout mice, demonstrating that topical (non-prostaglandin mediated) mechanisms are involved; second, no correlation is found in vivo between the extent of intestinal injury and the degree of inhibition of prostaglandin synthesis; third, bile flow interruption in animal models completely prevents intestinal damage by parenterally administered NSAIDs. What is in bile that could play a role in NSAID toxicity? This timely review will critically discuss the role of bile salts in NSAID-dependent gut damage.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bile Acids and Salts/metabolism , Intestinal Mucosa/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Bile Acids and Salts/chemistry , Bile Acids and Salts/physiology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Models, Biological , Phospholipids/chemistry , Phospholipids/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...