Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2869, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693144

ABSTRACT

Only ~20% of heavy drinkers develop alcohol cirrhosis (AC). While differences in metabolism, inflammation, signaling, microbiome signatures and genetic variations have been tied to the pathogenesis of AC, the key underlying mechanisms for this interindividual variability, remain to be fully elucidated. Induced pluripotent stem cell-derived hepatocytes (iHLCs) from patients with AC and healthy controls differ transcriptomically, bioenergetically and histologically. They include a greater number of lipid droplets (LDs) and LD-associated mitochondria compared to control cells. These pre-pathologic indicators are effectively reversed by Aramchol, an inhibitor of stearoyl-CoA desaturase. Bioenergetically, AC iHLCs have lower spare capacity, slower ATP production and their mitochondrial fuel flexibility towards fatty acids and glutamate is weakened. MARC1 and PNPLA3, genes implicated by GWAS in alcohol cirrhosis, show to correlate with lipid droplet-associated and mitochondria-mediated oxidative damage in AC iHLCs. Knockdown of PNPLA3 expression exacerbates mitochondrial deficits and leads to lipid droplets alterations. These findings suggest that differences in mitochondrial bioenergetics and lipid droplet formation are intrinsic to AC hepatocytes and can play a role in its pathogenesis.


Subject(s)
Acyltransferases , Energy Metabolism , Hepatocytes , Induced Pluripotent Stem Cells , Lipase , Lipid Droplets , Liver Cirrhosis, Alcoholic , Mitochondria , Phospholipases A2, Calcium-Independent , Humans , Hepatocytes/metabolism , Hepatocytes/pathology , Induced Pluripotent Stem Cells/metabolism , Lipid Droplets/metabolism , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/pathology , Liver Cirrhosis, Alcoholic/genetics , Lipase/metabolism , Lipase/genetics , Mitochondria/metabolism , Male , Membrane Proteins/metabolism , Membrane Proteins/genetics , Female , Middle Aged , Adult , Oxidative Stress
2.
Cell Death Discov ; 9(1): 104, 2023 Mar 25.
Article in English | MEDLINE | ID: mdl-36966147

ABSTRACT

The proliferation and differentiation of hepatic progenitor cells (HPCs) drive the homeostatic renewal of the liver under diverse conditions. Liver regeneration is associated with an increase in Axin2+Cnr1+ HPCs, along with a marked increase in the levels of the endocannabinoid anandamide (AEA). But the molecular mechanism linking AEA signaling to HPC proliferation and/or differentiation has not been explored. Here, we show that in vitro exposure of HPCs to AEA triggers both cell cycling and differentiation along with increased expression of Cnr1, Krt19, and Axin2. Mechanistically, we found that AEA promotes the nuclear localization of the transcription factor ß-catenin, with subsequent induction of its downstream targets. Systemic analyses of cells after CRISPR-mediated knockout of the ß-catenin-regulated transcriptome revealed that AEA modulates ß-catenin-dependent cell cycling and differentiation, as well as interleukin pathways. Further, we found that AEA promotes OXPHOS in HPCs when amino acids and glucose are readily available as substrates, but AEA inhibits it when the cells rely primarily on fatty acid oxidation. Thus, the endocannabinoid system promotes hepatocyte renewal and maturation by stimulating the proliferation of Axin2+Cnr1+ HPCs via the ß-catenin pathways while modulating the metabolic activity of their precursor cells.

3.
Cell Metab ; 29(6): 1320-1333.e8, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31105045

ABSTRACT

Endocannabinoids acting on the cannabinoid-1 receptor (CB1R) or ghrelin acting on its receptor (GHS-R1A) both promote alcohol-seeking behavior, but an interaction between the two signaling systems has not been explored. Here, we report that the peripheral CB1R inverse agonist JD5037 reduces ethanol drinking in wild-type mice but not in mice lacking CB1R, ghrelin peptide or GHS-R1A. JD5037 treatment of alcohol-drinking mice inhibits the formation of biologically active octanoyl-ghrelin without affecting its inactive precursor desacyl-ghrelin. In ghrelin-producing stomach cells, JD5037 reduced the level of the substrate octanoyl-carnitine generated from palmitoyl-carnitine by increasing fatty acid ß-oxidation. Blocking gastric vagal afferents abrogated the ability of either CB1R or GHS-R1A blockade to reduce ethanol drinking. We conclude that blocking CB1R in ghrelin-producing cells reduces alcohol drinking by inhibiting the formation of active ghrelin and its signaling via gastric vagal afferents. Thus, peripheral CB1R blockade may have therapeutic potential in the treatment of alcoholism.


Subject(s)
Alcohol Drinking/genetics , Brain/physiology , Intestines/physiology , Receptor, Cannabinoid, CB1/genetics , Acyltransferases/genetics , Acyltransferases/physiology , Alcohol Drinking/physiopathology , Alcoholism/genetics , Alcoholism/physiopathology , Animals , Brain/drug effects , Cells, Cultured , Gene Deletion , Ghrelin/metabolism , Ghrelin/physiology , Intestines/drug effects , Male , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptors, Ghrelin/genetics , Receptors, Ghrelin/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/pharmacology
4.
EBioMedicine ; 40: 471-487, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30606699

ABSTRACT

BACKGROUND: Redox metabolism is often considered a potential target for cancer treatment, but a systematic examination of redox responses in hepatocellular carcinoma (HCC) is missing. METHODS: Here, we employed systems biology and biological network analyses to reveal key roles of genes associated with redox metabolism in HCC by integrating multi-omics data. FINDINGS: We found that several redox genes, including 25 novel potential prognostic genes, are significantly co-expressed with liver-specific genes and genes associated with immunity and inflammation. Based on an integrative analysis, we found that HCC tumors display antagonistic behaviors in redox responses. The two HCC groups are associated with altered fatty acid, amino acid, drug and hormone metabolism, differentiation, proliferation, and NADPH-independent vs -dependent antioxidant defenses. Redox behavior varies with known tumor subtypes and progression, affecting patient survival. These antagonistic responses are also displayed at the protein and metabolite level and were validated in several independent cohorts. We finally showed the differential redox behavior using mice transcriptomics in HCC and noncancerous tissues and associated with hypoxic features of the two redox gene groups. INTERPRETATION: Our integrative approaches highlighted mechanistic differences among tumors and allowed the identification of a survival signature and several potential therapeutic targets for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Models, Biological , Oxidation-Reduction , Algorithms , Biomarkers, Tumor , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Computational Biology/methods , Disease Progression , Gene Expression Profiling , Gene Regulatory Networks , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Metabolic Networks and Pathways , Molecular Sequence Annotation , Prognosis , Reproducibility of Results , Signal Transduction , Transcriptome
5.
Mol Syst Biol ; 13(8): 938, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827398

ABSTRACT

We performed integrative network analyses to identify targets that can be used for effectively treating liver diseases with minimal side effects. We first generated co-expression networks (CNs) for 46 human tissues and liver cancer to explore the functional relationships between genes and examined the overlap between functional and physical interactions. Since increased de novo lipogenesis is a characteristic of nonalcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC), we investigated the liver-specific genes co-expressed with fatty acid synthase (FASN). CN analyses predicted that inhibition of these liver-specific genes decreases FASN expression. Experiments in human cancer cell lines, mouse liver samples, and primary human hepatocytes validated our predictions by demonstrating functional relationships between these liver genes, and showing that their inhibition decreases cell growth and liver fat content. In conclusion, we identified liver-specific genes linked to NAFLD pathogenesis, such as pyruvate kinase liver and red blood cell (PKLR), or to HCC pathogenesis, such as PKLR, patatin-like phospholipase domain containing 3 (PNPLA3), and proprotein convertase subtilisin/kexin type 9 (PCSK9), all of which are potential targets for drug development.


Subject(s)
Carcinoma, Hepatocellular/genetics , Fatty Acid Synthase, Type I/genetics , Gene Regulatory Networks , Liver Neoplasms/genetics , Non-alcoholic Fatty Liver Disease/genetics , Systems Biology/methods , Animals , Carcinoma, Hepatocellular/drug therapy , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks/drug effects , Hep G2 Cells , Humans , K562 Cells , Liver/chemistry , Liver/drug effects , Liver Neoplasms/drug therapy , Mice , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Organ Specificity , Protein Interaction Maps , Sequence Analysis, RNA
6.
JCI Insight ; 1(11)2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27525312

ABSTRACT

Liver fibrosis, a consequence of chronic liver injury and a way station to cirrhosis and hepatocellular carcinoma, lacks effective treatment. Endocannabinoids acting via cannabinoid-1 receptors (CB1R) induce profibrotic gene expression and promote pathologies that predispose to liver fibrosis. CB1R antagonists produce opposite effects, but their therapeutic development was halted due to neuropsychiatric side effects. Inducible nitric oxide synthase (iNOS) also promotes liver fibrosis and its underlying pathologies, but iNOS inhibitors tested to date showed limited therapeutic efficacy in inflammatory diseases. Here, we introduce a peripherally restricted, orally bioavailable CB1R antagonist, which accumulates in liver to release an iNOS inhibitory leaving group. In mouse models of fibrosis induced by CCl4 or bile duct ligation, the hybrid CB1R/iNOS antagonist surpassed the antifibrotic efficacy of the CB1R antagonist rimonabant or the iNOS inhibitor 1400W, without inducing anxiety-like behaviors or CB1R occupancy in the CNS. The hybrid inhibitor also targeted CB1R-independent, iNOS-mediated profibrotic pathways, including increased PDGF, Nlrp3/Asc3, and integrin αvß6 signaling, as judged by its ability to inhibit these pathways in cnr1-/- but not in nos2-/- mice. Additionally, it was able to slow fibrosis progression and to attenuate established fibrosis. Thus, dual-target peripheral CB1R/iNOS antagonists have therapeutic potential in liver fibrosis.

7.
Development ; 143(4): 609-22, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26884397

ABSTRACT

Endocannabinoid (EC) signaling mediates psychotropic effects and regulates appetite. By contrast, potential roles in organ development and embryonic energy consumption remain unknown. Here, we demonstrate that genetic or chemical inhibition of cannabinoid receptor (Cnr) activity disrupts liver development and metabolic function in zebrafish (Danio rerio), impacting hepatic differentiation, but not endodermal specification: loss of cannabinoid receptor 1 (cnr1) and cnr2 activity leads to smaller livers with fewer hepatocytes, reduced liver-specific gene expression and proliferation. Functional assays reveal abnormal biliary anatomy and lipid handling. Adult cnr2 mutants are susceptible to hepatic steatosis. Metabolomic analysis reveals reduced methionine content in Cnr mutants. Methionine supplementation rescues developmental and metabolic defects in Cnr mutant livers, suggesting a causal relationship between EC signaling, methionine deficiency and impaired liver development. The effect of Cnr on methionine metabolism is regulated by sterol regulatory element-binding transcription factors (Srebfs), as their overexpression rescues Cnr mutant liver phenotypes in a methionine-dependent manner. Our work describes a novel developmental role for EC signaling, whereby Cnr-mediated regulation of Srebfs and methionine metabolism impacts liver development and function.


Subject(s)
Liver/embryology , Liver/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Cannabinoids/metabolism , Cell Count , Cell Proliferation/drug effects , Cysteine/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Metabolomics , Methionine/metabolism , Mutation/genetics , Organ Size/drug effects , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
8.
Cell Rep ; 13(9): 2014-26, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655911

ABSTRACT

Hepatocellular carcinoma (HCC) is a deadly form of liver cancer that is increasingly prevalent. We analyzed global gene expression profiling of 361 HCC tumors and 49 adjacent noncancerous liver samples by means of combinatorial network-based analysis. We investigated the correlation between transcriptome and proteome of HCC and reconstructed a functional genome-scale metabolic model (GEM) for HCC. We identified fundamental metabolic processes required for cell proliferation using the network centric view provided by the GEM. Our analysis revealed tight regulation of fatty acid biosynthesis (FAB) and highly significant deregulation of fatty acid oxidation in HCC. We predicted mitochondrial acetate as an emerging substrate for FAB through upregulation of mitochondrial acetyl-CoA synthetase (ACSS1) in HCC. We analyzed heterogeneous expression of ACSS1 and ACSS2 between HCC patients stratified by high and low ACSS1 and ACSS2 expression and revealed that ACSS1 is associated with tumor growth and malignancy under hypoxic conditions in human HCC.


Subject(s)
Acetates/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Acetate-CoA Ligase/genetics , Acetate-CoA Ligase/metabolism , Aged , Algorithms , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Fatty Acids/biosynthesis , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/metabolism , Male , Mice , Middle Aged , Mitochondria/metabolism , RNA, Messenger/metabolism
9.
Sci Rep ; 5: 14953, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26455425

ABSTRACT

We report an unexpected link between aging, thermogenesis and weight gain via the orphan G protein-coupled receptor GPR3. Mice lacking GPR3 and maintained on normal chow had similar body weights during their first 5 months of life, but gained considerably more weight thereafter and displayed reduced total energy expenditure and lower core body temperature. By the age of 5 months GPR3 KO mice already had lower thermogenic gene expression and uncoupling protein 1 protein level and showed impaired glucose uptake into interscapular brown adipose tissue (iBAT) relative to WT littermates. These molecular deviations in iBAT of GPR3 KO mice preceded measurable differences in body weight and core body temperature at ambient conditions, but were coupled to a failure to maintain thermal homeostasis during acute cold challenge. At the same time, the same cold challenge caused a 17-fold increase in Gpr3 expression in iBAT of WT mice. Thus, GPR3 appears to have a key role in the thermogenic response of iBAT and may represent a new therapeutic target in age-related obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Aging/genetics , Energy Metabolism/genetics , Obesity/genetics , Receptors, G-Protein-Coupled/genetics , Thermogenesis/genetics , Adipose Tissue, Brown/pathology , Aging/metabolism , Aging/pathology , Animals , Biological Transport , Body Temperature , Cold Temperature , Female , Gene Expression Regulation , Glucose/metabolism , Homeostasis , Ion Channels/genetics , Ion Channels/metabolism , Male , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Obesity/metabolism , Obesity/pathology , Phenotype , Receptors, G-Protein-Coupled/deficiency , Uncoupling Protein 1 , Weight Gain
10.
Hepatology ; 61(5): 1615-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25580584

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) has high mortality and no adequate treatment. Endocannabinoids interact with hepatic cannabinoid 1 receptors (CB1Rs) to promote hepatocyte proliferation in liver regeneration by inducing cell cycle proteins involved in mitotic progression, including Forkhead Box M1. Because this protein is highly expressed in HCC and contributes to its genesis and progression, we analyzed the involvement of the endocannabinoid/CB1R system in murine and human HCC. Postnatal diethylnitrosamine treatment induced HCC within 8 months in wild-type mice but fewer and smaller tumors in CB1R(-/-) mice or in wild-type mice treated with the peripheral CB1R antagonist JD5037, as monitored in vivo by serial magnetic resonance imaging. Genome-wide transcriptome analysis revealed CB1R-dependent, tumor-induced up-regulation of the hepatic expression of CB1R, its endogenous ligand anandamide, and a number of tumor-promoting genes, including the GRB2 interactome as well as Forkhead Box M1 and its downstream target, the tryptophan-catalyzing enzyme indoleamine 2,3-dioxygenase. Increased indoleamine 2,3-dioxygenase activity and consequent induction of immunosuppressive T-regulatory cells in tumor tissue promote immune tolerance. CONCLUSION: The endocannabinoid/CB1R system is up-regulated in chemically induced HCC, resulting in the induction of various tumor-promoting genes, including indoleamine 2,3-dioxygenase; and attenuation of these changes by blockade or genetic ablation of CB1R suppresses the growth of HCC and highlights the therapeutic potential of peripheral CB1R blockade.


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Receptor, Cannabinoid, CB1/physiology , Animals , Carcinoma, Hepatocellular/chemically induced , Diethylnitrosamine , Disease Progression , Endocannabinoids/physiology , Forkhead Box Protein M1 , Forkhead Transcription Factors/physiology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase , Liver Neoplasms/chemically induced , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/chemically induced , Up-Regulation
11.
Hepatology ; 59(1): 143-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23832510

ABSTRACT

UNLABELLED: Obesity is associated with increased activity of two lipid signaling systems (endocannabinoids [ECs] and ceramides), with both being implicated in insulin resistance. Cannabinoid-1 receptor (CB1 R) antagonists reverse obesity and insulin resistance, but have psychiatric side effects. Here we analyzed the role of ceramide in CB1 R-mediated insulin resistance in C57Bl6/J mice with high-fat diet-induced obesity (DIO), using JD5037, a peripherally restricted CB1 R inverse agonist. Chronic JD5037 treatment of DIO mice reduced body weight and steatosis and improved glucose tolerance and insulin sensitivity. Peripheral CB1 R blockade also attenuated the diet-induced increase in C14:0, C16:0, C18:0, and C20:0 ceramide species with either C16 or C18 sphingosine-base in the liver. Decreased ceramide levels reflected their reduced de novo synthesis, due to inhibition of the activity of serine-palmitoyl transferase (SPT) and the expression of its SPTLC3 catalytic subunit, as well as reduced ceramide synthase (CerS) activity related to reduced expression of CerS1 and CerS6. JD5037 treatment also increased ceramide degradation due to increased expression of ceramidases. In primary cultured mouse hepatocytes and HepG2 cells, the EC anandamide increased ceramide synthesis in an eIF2α-dependent manner, and inhibited insulin-induced akt phosphorylation by increased serine phosphorylation of IRS1 and increased expression of the serine/threonine phosphatase Phlpp1. These effects were abrogated by JD5037 or the SPT inhibitor myriocin. Chronic treatment of DIO mice with myriocin or JD5037 similarly reversed hepatic insulin resistance, as verified using a euglycemic/hyperinsulinemic clamp. CONCLUSION: ECs induce CB1 R-mediated, endoplasmic reticulum stress-dependent synthesis of specific ceramide subspecies in the liver, which plays a key role in obesity-related hepatic insulin resistance.


Subject(s)
Ceramides/biosynthesis , Diet, High-Fat/adverse effects , Insulin Resistance , Liver/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Body Weight , Endoplasmic Reticulum Stress , Fatty Liver/prevention & control , Hep G2 Cells , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred C57BL , Obesity/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Serine C-Palmitoyltransferase/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Up-Regulation
12.
Nat Med ; 19(9): 1132-40, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23955712

ABSTRACT

Type 2 diabetes mellitus (T2DM) progresses from compensated insulin resistance to beta cell failure resulting in uncompensated hyperglycemia, a process replicated in the Zucker diabetic fatty (ZDF) rat. The Nlrp3 inflammasome has been implicated in obesity-induced insulin resistance and beta cell failure. Endocannabinoids contribute to insulin resistance through activation of peripheral CB1 receptors (CB1Rs) and also promote beta cell failure. Here we show that beta cell failure in adult ZDF rats is not associated with CB1R signaling in beta cells, but rather in M1 macrophages infiltrating into pancreatic islets, and that this leads to activation of the Nlrp3-ASC inflammasome in the macrophages. These effects are replicated in vitro by incubating wild-type human or rodent macrophages, but not macrophages from CB1R-deficient (Cnr1(-/-)) or Nlrp3(-/-) mice, with the endocannabinoid anandamide. Peripheral CB1R blockade, in vivo depletion of macrophages or macrophage-specific knockdown of CB1R reverses or prevents these changes and restores normoglycemia and glucose-induced insulin secretion. These findings implicate endocannabinoids and inflammasome activation in beta cell failure and identify macrophage-expressed CB1R as a therapeutic target in T2DM.


Subject(s)
Arachidonic Acids/pharmacology , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Endocannabinoids/pharmacology , Inflammasomes/metabolism , Insulin-Secreting Cells/metabolism , Macrophages/metabolism , Polyunsaturated Alkamides/pharmacology , Animals , Apoptosis , Cannabinoid Receptor Agonists/pharmacology , Cell Line , Cell Survival , Humans , Hyperglycemia/metabolism , Insulin Resistance , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Obesity/metabolism , RNA Interference , RNA, Small Interfering , Rats
13.
Cell Metab ; 16(2): 167-79, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22841573

ABSTRACT

Obesity-related leptin resistance manifests in loss of leptin's ability to reduce appetite and increase energy expenditure. Obesity is also associated with increased activity of the endocannabinoid system, and CB(1) receptor (CB(1)R) inverse agonists reduce body weight and the associated metabolic complications, although adverse neuropsychiatric effects halted their therapeutic development. Here we show that in mice with diet-induced obesity (DIO), the peripherally restricted CB(1)R inverse agonist JD5037 is equieffective with its brain-penetrant parent compound in reducing appetite, body weight, hepatic steatosis, and insulin resistance, even though it does not occupy central CB(1)R or induce related behaviors. Appetite and weight reduction by JD5037 are mediated by resensitizing DIO mice to endogenous leptin through reversing the hyperleptinemia by decreasing leptin expression and secretion by adipocytes and increasing leptin clearance via the kidney. Thus, inverse agonism at peripheral CB(1)R not only improves cardiometabolic risk in obesity but has antiobesity effects by reversing leptin resistance.


Subject(s)
Anti-Obesity Agents/pharmacology , Drug Resistance/drug effects , Fatty Liver/drug therapy , Leptin/metabolism , Obesity/drug therapy , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Sulfonamides/pharmacology , Adipocytes/metabolism , Animals , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , Body Weight/drug effects , Drug Inverse Agonism , Fatty Liver/etiology , Insulin Resistance , Mice , Molecular Structure , Obesity/complications , Obesity/metabolism , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Regression Analysis , Sulfonamides/chemistry , Sulfonamides/therapeutic use
14.
Gastroenterology ; 142(5): 1218-1228.e1, 2012 May.
Article in English | MEDLINE | ID: mdl-22307032

ABSTRACT

BACKGROUND & AIMS: Obesity-related insulin resistance contributes to cardiovascular disease. Cannabinoid receptor-1 (CB(1)) blockade improves insulin sensitivity in obese animals and people, suggesting endocannabinoid involvement. We explored the role of hepatic CB(1) in insulin resistance and inhibition of insulin signaling pathways. METHODS: Wild-type mice and mice with disruption of CB(1) (CB(1)(-/-) mice) or with hepatocyte-specific deletion or transgenic overexpression of CB(1) were maintained on regular chow or a high-fat diet (HFD) to induce obesity and insulin resistance. Hyperinsulinemic-euglycemic clamp analysis was used to analyze the role of the liver and hepatic CB(1) in HFD-induced insulin resistance. The cellular mechanisms of insulin resistance were analyzed in mouse and human isolated hepatocytes using small interfering or short hairpin RNAs and lentiviral knockdown of gene expression. RESULTS: The HFD induced hepatic insulin resistance in wild-type mice, but not in CB(1)(-/-) mice or mice with hepatocyte-specific deletion of CB(1). CB(1)(-/-) mice that overexpressed CB(1) specifically in hepatocytes became hyperinsulinemic as a result of reduced insulin clearance due to down-regulation of the insulin-degrading enzyme. However, they had increased hepatic glucose production due to increased glycogenolysis, indicating hepatic insulin resistance; this was further increased by the HFD. In mice with hepatocytes that express CB(1), the HFD or CB(1) activation induced the endoplasmic reticulum stress response via activation of the Bip-PERK-eIF2α protein translation pathway. In hepatocytes isolated from human or mouse liver, CB(1) activation caused endoplasmic reticulum stress-dependent suppression of insulin-induced phosphorylation of akt-2 via phosphorylation of IRS1 at serine-307 and by inducing the expression of the serine and threonine phosphatase Phlpp1. Expression of CB(1) was up-regulated in samples from patients with nonalcoholic fatty liver disease. CONCLUSIONS: Endocannabinoids contribute to diet-induced insulin resistance in mice via hepatic CB(1)-mediated inhibition of insulin signaling and clearance.


Subject(s)
Insulin Resistance , Insulin/metabolism , Liver/metabolism , Receptor, Cannabinoid, CB1/physiology , Signal Transduction , Animals , Arachidonic Acids/pharmacology , Diet, High-Fat , Endocannabinoids , Endoplasmic Reticulum Stress , Fatty Liver/metabolism , Glucose Intolerance/etiology , Humans , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Phosphorylation , Polyunsaturated Alkamides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism
15.
Proc Natl Acad Sci U S A ; 108(15): 6323-8, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21383171

ABSTRACT

The mammalian liver regenerates upon tissue loss, which induces quiescent hepatocytes to enter the cell cycle and undergo limited replication under the control of multiple hormones, growth factors, and cytokines. Endocannabinoids acting via cannabinoid type 1 receptors (CB(1)R) promote neural progenitor cell proliferation, and in the liver they promote lipogenesis. These findings suggest the involvement of CB(1)R in the control of liver regeneration. Here we report that mice lacking CB(1)R globally or in hepatocytes only and wild-type mice treated with a CB(1)R antagonist have a delayed proliferative response to two-thirds partial hepatectomy (PHX). In wild-type mice, PHX leads to increased hepatic expression of CB(1)R and hyperactivation of the biosynthesis of the endocannabinoid anandamide in the liver via an in vivo pathway involving conjugation of arachidonic acid and ethanolamine by fatty-acid amide hydrolase. In wild-type but not CB(1)R(-/-) mice, PHX induces robust up-regulation of key cell-cycle proteins involved in mitotic progression, including cyclin-dependent kinase 1 (Cdk1), cyclin B2, and their transcriptional regulator forkhead box protein M1 (FoxM1), as revealed by ultrahigh-throughput RNA sequencing and pathway analysis and confirmed by real-time PCR and Western blot analyses. Treatment of wild-type mice with anandamide induces similar changes mediated via activation of the PI3K/Akt pathway. We conclude that activation of hepatic CB(1)R by newly synthesized anandamide promotes liver regeneration by controlling the expression of cell-cycle regulators that drive M phase progression.


Subject(s)
Arachidonic Acids/biosynthesis , Liver Regeneration , Liver/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Cell Cycle , Endocannabinoids , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Liver/cytology , Male , Mice , Mice, Knockout , Mitosis , Phosphatidylinositol 3-Kinases/metabolism , Polyunsaturated Alkamides , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Signal Transduction , Up-Regulation
16.
Hepatology ; 53(1): 346-55, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21254182

ABSTRACT

Endocannabinoids are lipid mediators of the same cannabinoid (CB) receptors that mediate the effects of marijuana. The endocannabinoid system (ECS) consists of CB receptors, endocannabinoids, and the enzymes involved in their biosynthesis and degradation, and it is present in both brain and peripheral tissues, including the liver. The hepatic ECS is activated in various liver diseases and contributes to the underlying pathologies. In patients with cirrhosis of various etiologies, the activation of vascular and cardiac CB(1) receptors by macrophage-derived and platelet-derived endocannabinoids contributes to the vasodilated state and cardiomyopathy, which can be reversed by CB(1) blockade. In mouse models of liver fibrosis, the activation of CB(1) receptors on hepatic stellate cells is fibrogenic, and CB(1) blockade slows the progression of fibrosis. Fatty liver induced by a high-fat diet or chronic alcohol feeding depends on the activation of peripheral receptors, including hepatic CB(1) receptors, which also contribute to insulin resistance and dyslipidemias. Although the documented therapeutic potential of CB(1) blockade is limited by neuropsychiatric side effects, these may be mitigated by using novel, peripherally restricted CB(1) antagonists.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Liver Diseases/physiopathology , Receptors, Cannabinoid/physiology , Animals , Cannabidiol/therapeutic use , Cannabinoid Receptor Antagonists , Fatty Liver/etiology , Fatty Liver, Alcoholic/physiopathology , Hepatic Encephalopathy/physiopathology , Hepatitis, Autoimmune/drug therapy , Humans , Liver Cirrhosis/physiopathology , Metabolic Syndrome/physiopathology , Mice , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/physiology , Reperfusion Injury/physiopathology
17.
J Am Coll Cardiol ; 56(25): 2115-25, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21144973

ABSTRACT

OBJECTIVES: In this study, we have investigated the effects of cannabidiol (CBD) on myocardial dysfunction, inflammation, oxidative/nitrative stress, cell death, and interrelated signaling pathways, using a mouse model of type I diabetic cardiomyopathy and primary human cardiomyocytes exposed to high glucose. BACKGROUND: Cannabidiol, the most abundant nonpsychoactive constituent of Cannabis sativa (marijuana) plant, exerts anti-inflammatory effects in various disease models and alleviates pain and spasticity associated with multiple sclerosis in humans. METHODS: Left ventricular function was measured by the pressure-volume system. Oxidative stress, cell death, and fibrosis markers were evaluated by molecular biology/biochemical techniques, electron spin resonance spectroscopy, and flow cytometry. RESULTS: Diabetic cardiomyopathy was characterized by declined diastolic and systolic myocardial performance associated with increased oxidative-nitrative stress, nuclear factor-κB and mitogen-activated protein kinase (c-Jun N-terminal kinase, p-38, p38α) activation, enhanced expression of adhesion molecules (intercellular adhesion molecule-1, vascular cell adhesion molecule-1), tumor necrosis factor-α, markers of fibrosis (transforming growth factor-ß, connective tissue growth factor, fibronectin, collagen-1, matrix metalloproteinase-2 and -9), enhanced cell death (caspase 3/7 and poly[adenosine diphosphate-ribose] polymerase activity, chromatin fragmentation, and terminal deoxynucleotidyl transferase dUTP nick end labeling), and diminished Akt phosphorylation. Remarkably, CBD attenuated myocardial dysfunction, cardiac fibrosis, oxidative/nitrative stress, inflammation, cell death, and interrelated signaling pathways. Furthermore, CBD also attenuated the high glucose-induced increased reactive oxygen species generation, nuclear factor-κB activation, and cell death in primary human cardiomyocytes. CONCLUSIONS: Collectively, these results coupled with the excellent safety and tolerability profile of CBD in humans, strongly suggest that it may have great therapeutic potential in the treatment of diabetic complications, and perhaps other cardiovascular disorders, by attenuating oxidative/nitrative stress, inflammation, cell death and fibrosis.


Subject(s)
Cannabidiol/therapeutic use , Diabetic Cardiomyopathies/drug therapy , Myocytes, Cardiac/drug effects , Animals , Apoptosis/drug effects , Blood Glucose/drug effects , Body Weight/drug effects , Cannabidiol/pharmacology , Cells, Cultured , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Fibrosis , Glucose , Hemodynamics/drug effects , Humans , Insulin/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Myocardium/pathology , NF-kappa B/metabolism , Oxidative Stress/drug effects , Pancreas/drug effects , Pancreas/metabolism , Reactive Oxygen Species/metabolism
18.
Chem Biol ; 17(11): 1256-66, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21095576

ABSTRACT

The enzyme fatty acid amide hydrolase (FAAH) catalyzes the in vivo degradation of the endocannabinoid anandamide, thus controlling its action at receptors. A novel FAAH inhibitor, AM3506, normalizes the elevated blood pressure and cardiac contractility of spontaneously hypertensive rats (SHR) without affecting these parameters in normotensive rats. These effects are due to blockade of FAAH and a corresponding rise in brain anandamide levels, resulting in CB1 receptor-mediated decrease in sympathetic tone. The supersensitivity of SHR to CB1 receptor-mediated cardiovascular depression is related to increased G protein coupling of CB1 receptors. Importantly, AM3506 does not elicit hyperglycemia and insulin resistance seen with other FAAH inhibitors or in FAAH⁻/⁻ mice, which is related to its inability to inhibit FAAH in the liver due to rapid hepatic uptake and metabolism. This unique activity profile offers improved therapeutic value in hypertension.


Subject(s)
Alkanesulfonates/chemistry , Amidohydrolases/antagonists & inhibitors , Antihypertensive Agents/chemistry , Enzyme Inhibitors/chemistry , Phenols/chemistry , Alkanesulfonates/pharmacology , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Antihypertensive Agents/therapeutic use , Arachidonic Acids/metabolism , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Endocannabinoids , Enzyme Inhibitors/therapeutic use , Hypertension/drug therapy , Male , Mice , Mice, Knockout , Phenols/pharmacology , Polyunsaturated Alkamides/metabolism , Rats , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism
19.
Br J Pharmacol ; 160(3): 657-68, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20590569

ABSTRACT

BACKGROUND AND PURPOSE: Accumulating recent evidence suggests that cannabinoid-1 (CB(1)) receptor activation may promote inflammation and cell death and its pharmacological inhibition is associated with anti-inflammatory and tissue-protective effects in various preclinical disease models, as well as in humans. EXPERIMENTAL APPROACH: In this study, using molecular biology and biochemistry methods, we have investigated the effects of genetic deletion or pharmacological inhibition of CB(1) receptors on inflammation, oxidative/nitrosative stress and cell death pathways associated with a clinically relevant model of nephropathy, induced by an important chemotherapeutic drug cisplatin. RESULTS: Cisplatin significantly increased endocannabinoid anandamide content, activation of p38 and JNK mitogen-activated protein kinases (MAPKs), apoptotic and poly (ADP-ribose)polymerase-dependent cell death, enhanced inflammation (leucocyte infiltration, tumour necrosis factor-alpha and interleukin-1beta) and promoted oxidative/nitrosative stress [increased expressions of superoxide-generating enzymes (NOX2(gp91phox), NOX4), inducible nitric oxide synthase and tissue 4-hydroxynonenal and nitrotyrosine levels] in the kidneys of mice, accompanied by marked histopathological damage and impaired renal function (elevated creatinine and serum blood urea nitrogen) 3 days following its administration. Both genetic deletion and pharmacological inhibition of CB(1) receptors with AM281 or SR141716 markedly attenuated the cisplatin-induced renal dysfunction and interrelated oxidative/nitrosative stress, p38 and JNK MAPK activation, cell death and inflammatory response in the kidney. CONCLUSIONS AND IMPLICATIONS: The endocannabinoid system through CB(1) receptors promotes cisplatin-induced tissue injury by amplifying MAPK activation, cell death and interrelated inflammation and oxidative/nitrosative stress. These results also suggest that inhibition of CB(1) receptors may exert beneficial effects in renal (and most likely other) diseases associated with enhanced inflammation, oxidative/nitrosative stress and cell death.


Subject(s)
Cell Death/physiology , Inflammation/physiopathology , Nephritis/chemically induced , Oxidative Stress/physiology , Receptor, Cannabinoid, CB1/physiology , Animals , Arachidonic Acids/metabolism , Cell Death/genetics , Cisplatin , Disease Models, Animal , Endocannabinoids , Glycerides/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Mice , Mice, Knockout , Morpholines/pharmacology , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Rimonabant , Signal Transduction/drug effects , Signal Transduction/physiology
20.
J Clin Invest ; 120(8): 2953-66, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20664173

ABSTRACT

Obesity and its metabolic consequences are a major public health concern worldwide. Obesity is associated with overactivity of the endocannabinoid system, which is involved in the regulation of appetite, lipogenesis, and insulin resistance. Cannabinoid-1 receptor (CB1R) antagonists reduce body weight and improve cardiometabolic abnormalities in experimental and human obesity, but their therapeutic potential is limited by neuropsychiatric side effects. Here we have demonstrated that a CB1R neutral antagonist largely restricted to the periphery does not affect behavioral responses mediated by CB1R in the brains of mice with genetic or diet-induced obesity, but it does cause weight-independent improvements in glucose homeostasis, fatty liver, and plasma lipid profile. These effects were due to blockade of CB1R in peripheral tissues, including the liver, as verified through the use of CB1R-deficient mice with or without transgenic expression of CB1R in the liver. These results suggest that targeting peripheral CB1R has therapeutic potential for alleviating cardiometabolic risk in obese patients.


Subject(s)
Morpholines/pharmacology , Obesity/drug therapy , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Insulin Resistance , Leptin/physiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Morpholines/therapeutic use , Obesity/complications , Obesity/metabolism , Piperidines/pharmacology , Pyrazoles/therapeutic use , Receptor, Cannabinoid, CB1/physiology , Rimonabant , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...