Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Publication year range
1.
Psychiatry Clin Neurosci ; 72(5): 349-361, 2018 May.
Article in English | MEDLINE | ID: mdl-29356314

ABSTRACT

AIM: This study sought to characterize the plasma metabolite profiling of patients with major depressive disorder (MDD). METHODS: Psychiatric assessments were made with the Structured Clinical Interview for DSM-IV Axis I Disorders. In the exploratory cohort, plasma metabolite profiles of 34 MDD patients and 31 mentally healthy controls were compared using capillary electrophoresis-mass spectrometry. Among the candidate metabolites, we focused on a metabolite showing the largest difference. The absolute concentrations were measured in two cohorts from a psychiatric primary care clinic to characterize the accuracy of the metabolite biomarker. RESULTS: Among 23 metabolites significantly lower in the MDD group than in healthy controls, we focused on phosphoethanolamine (PEA) as a candidate. The reduction of PEA levels in MDD was checked in independent clinical sample sets. An ion-chromatography-fluorescence detection method was developed to measure plasma PEA levels. In the preliminary cohort, we examined 34 MDD and 43 non-MDD subjects. The area under the receiver-operator curve (AUC) was 0.92, with sensitivity/specificity greater than 88%, at a cut-off of 1.46 µM. In the checking cohort, with 10 MDD and 13 non-MDD subjects, AUC was 0.89, with sensitivity/specificity of 86% and 100%, respectively, at a cut-off of 1.48 µM. Plasma PEA inversely correlated with MDD severity, depressed mood, loss of interest, and psychomotor retardation. CONCLUSION: These results suggest that plasma PEA level could be a candidate biomarker of MDD in the clinical setting. Further studies comparing MDD and mentally healthy controls are needed to confirm the utility of PEA as a biomarker for depression.


Subject(s)
Biomarkers/blood , Depressive Disorder, Major/blood , Depressive Disorder, Major/physiopathology , Ethanolamines/blood , Metabolome/physiology , Adult , Cohort Studies , Female , Humans , Male , Middle Aged , Sensitivity and Specificity
2.
Cell Metab ; 7(3): 227-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18316028

ABSTRACT

Alcohol-induced fatty liver, a major cause of morbidity, has been attributed to enhanced hepatic lipogenesis and decreased fat clearance of unknown mechanism. Here we report that the steatosis induced in mice by a low-fat, liquid ethanol diet is attenuated by concurrent blockade of cannabinoid CB1 receptors. Global or hepatocyte-specific CB1 knockout mice are resistant to ethanol-induced steatosis and increases in lipogenic gene expression and have increased carnitine palmitoyltransferase 1 activity, which, unlike in controls, is not reduced by ethanol treatment. Ethanol feeding increases the hepatic expression of CB1 receptors and upregulates the endocannabinoid 2-arachidonoylglycerol (2-AG) and its biosynthetic enzyme diacylglycerol lipase beta selectively in hepatic stellate cells. In control but not CB1 receptor-deficient hepatocytes, coculture with stellate cells from ethanol-fed mice results in upregulation of CB1 receptors and lipogenic gene expression. We conclude that paracrine activation of hepatic CB1 receptors by stellate cell-derived 2-AG mediates ethanol-induced steatosis through increasing lipogenesis and decreasing fatty acid oxidation.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Fatty Liver, Alcoholic/metabolism , Lipogenesis , Liver/metabolism , Paracrine Communication , Receptor, Cannabinoid, CB1/metabolism , Animals , Arachidonic Acids/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Coculture Techniques , Diet, Fat-Restricted , Disease Models, Animal , Ethanol , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/pathology , Fatty Liver, Alcoholic/prevention & control , Glycerides/metabolism , Hepatocytes/metabolism , Lipogenesis/drug effects , Lipogenesis/genetics , Lipoprotein Lipase/metabolism , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Paracrine Communication/drug effects , Paracrine Communication/genetics , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Rimonabant , Sterol Regulatory Element Binding Protein 1/metabolism , Up-Regulation
3.
J Biol Chem ; 285(25): 19002-11, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20410309

ABSTRACT

Alcoholism can result in fatty liver that can progress to steatohepatitis, cirrhosis, and liver cancer. Mice fed alcohol develop fatty liver through endocannabinoid activation of hepatic CB(1) cannabinoid receptors (CB(1)R), which increases lipogenesis and decreases fatty acid oxidation. Chronic alcohol feeding also up-regulates CB(1)R in hepatocytes in vivo, which could be replicated in vitro by co-culturing control hepatocytes with hepatic stellate cells (HSC) isolated from ethanol-fed mice, implicating HSC-derived mediator(s) in the regulation of hepatic CB(1)R (Jeong, W. I., Osei-Hyiaman, D., Park, O., Liu, J., Bátkai, S., Mukhopadhyay, P., Horiguchi, N., Harvey-White, J., Marsicano, G., Lutz, B., Gao, B., and Kunos, G. (2008) Cell Metab. 7, 227-235). HSC being a rich source of retinoic acid (RA), we tested whether RA and its receptors may regulate CB(1)R expression in cultured mouse hepatocytes. Incubation of hepatocytes with RA or RA receptor (RAR) agonists increased CB(1)R mRNA and protein, the most efficacious being the RARgamma agonist CD437 and the pan-RAR agonist TTNPB. The endocannabinoid 2-arachidonoylglycerol (2-AG) also increased hepatic CB(1)R expression, which was mediated indirectly via RA, because it was absent in hepatocytes from mice lacking retinaldehyde dehydrogenase 1, the enzyme catalyzing the generation of RA from retinaldehyde. The binding of RARgamma to the CB(1)R gene 5' upstream domain in hepatocytes treated with RAR agonists or 2-AG was confirmed by chromatin immunoprecipitation and electrophoretic mobility shift and antibody supershift assays. Finally, TTNPB-induced CB(1)R expression was attenuated by small interfering RNA knockdown of RARgamma in hepatocytes. We conclude that RARgamma regulates CB(1)R expression and is thus involved in the control of hepatic fat metabolism by endocannabinoids.


Subject(s)
Liver/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Retinoic Acid/metabolism , Transcription, Genetic , Tretinoin/metabolism , Animals , Arachidonic Acids/chemistry , Catalysis , Chromatin Immunoprecipitation , Endocannabinoids , Fatty Liver/metabolism , Glycerides/chemistry , Hepatocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Retinoids/chemistry , Retinoic Acid Receptor gamma
4.
J Clin Invest ; 118(9): 3160-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18677409

ABSTRACT

Diet-induced obesity is associated with fatty liver, insulin resistance, leptin resistance, and changes in plasma lipid profile. Endocannabinoids have been implicated in the development of these associated phenotypes, because mice deficient for the cannabinoid receptor CB1 (CB1-/-) do not display these changes in association with diet-induced obesity. The target tissues that mediate these effects, however, remain unknown. We therefore investigated the relative role of hepatic versus extrahepatic CB1 receptors in the metabolic consequences of a high-fat diet, using liver-specific CB1 knockout (LCB1-/-) mice. LCB1(-/-) mice fed a high-fat diet developed a similar degree of obesity as that of wild-type mice, but, similar to CB1(-/-) mice, had less steatosis, hyperglycemia, dyslipidemia, and insulin and leptin resistance than did wild-type mice fed a high-fat diet. CB1 agonist-induced increase in de novo hepatic lipogenesis and decrease in the activity of carnitine palmitoyltransferase-1 and total energy expenditure were absent in both CB1(-/-) and LCB1(-/-) mice. We conclude that endocannabinoid activation of hepatic CB1 receptors contributes to the diet-induced steatosis and associated hormonal and metabolic changes, but not to the increase in adiposity, observed with high-fat diet feeding. Theses studies suggest that peripheral CB1 receptors could be selectively targeted for the treatment of fatty liver, impaired glucose homeostasis, and dyslipidemia in order to minimize the neuropsychiatric side effects of nonselective CB1 blockade during treatment of obesity-associated conditions.


Subject(s)
Animal Feed , Dyslipidemias/metabolism , Fatty Liver/metabolism , Insulin/metabolism , Leptin/metabolism , Liver/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Dyslipidemias/genetics , Fatty Liver/genetics , Female , Insulin/genetics , Insulin Resistance/genetics , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/complications , Receptor, Cannabinoid, CB1/genetics
5.
Trends Pharmacol Sci ; 30(1): 1-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19042036

ABSTRACT

Endocannabinoids, endogenous lipid ligands of cannabinoid receptors, mediate a variety of effects similar to those of marijuana. Cannabinoid CB(1) receptors are highly abundant in the brain and mediate psychotropic effects, which limits their value as a potential therapeutic target. There is growing evidence for CB(1) receptors in peripheral tissues that modulate a variety of functions, including pain sensitivity and obesity-related hormonal and metabolic abnormalities. In this review we propose that selective targeting of peripheral CB(1) receptors has potential therapeutic value because it would help to minimize addictive, psychoactive effects in the case of CB(1) agonists used as analgesics, or depression and anxiety in the case of CB(1) antagonists used in the management of cardiometabolic risk factors associated with the metabolic syndrome.


Subject(s)
Metabolic Diseases/drug therapy , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Drug Design , Humans , Inflammation/physiopathology , Inflammation/prevention & control , Metabolic Diseases/physiopathology , Metabolic Diseases/prevention & control , Pain/drug therapy , Pain/physiopathology , Pain/prevention & control , Receptor, Cannabinoid, CB1/physiology , Terminology as Topic
6.
PLoS Negl Trop Dis ; 14(12): e0008866, 2020 12.
Article in English | MEDLINE | ID: mdl-33315875

ABSTRACT

Helminths are parasitic worms that infect over a billion people worldwide. The pathological consequences from infection are due in part, to parasite-induced changes in host metabolic pathways. Here, we analyse the changes in host metabolic profiles, in response to the first Schistosoma haematobium infection and treatment in Zimbabwean children. A cohort of 83 schistosome-negative children (2-5 years old) as determined by parasitological examination, guardian interviews and examination of medical records, was recruited at baseline. Children were followed up after three months for parasitological diagnosis of their first S. haematobium infection, by detection of parasite eggs excreted in urine. Children positive for infection were treated with the antihelminthic drug praziquantel, and treatment efficacy checked three months after treatment. Blood samples were taken at each time point, and capillary electrophoresis mass spectrometry in conjunction with multivariate analysis were used to compare the change in serum metabolite profiles in schistosome-infected versus uninfected children. Following baseline at the three-month follow up, 11 children had become infected with S. haematobium (incidence = 13.3%). Our results showed that infection with S. haematobium was associated with significant increases (>2-fold) in discriminatory metabolites, linked primarily with energy (G6P, 3-PG, AMP, ADP) and purine (AMP, ADP) metabolism. These observed changes were commensurate with schistosome infection intensity, and levels of the affected metabolites were reduced following treatment, albeit not significantly. This study demonstrates that early infection with S. haematobium is associated with alterations in host energy and purine metabolism. Taken together, these changes are consistent with parasite-related clinical manifestations of malnutrition, poor growth and poor physical and cognitive performance observed in schistosome-infected children.


Subject(s)
Energy Metabolism , Purines/metabolism , Schistosoma haematobium , Schistosomiasis haematobia/drug therapy , Schistosomiasis haematobia/metabolism , Animals , Anthelmintics/therapeutic use , Child, Preschool , Female , Humans , Male , Praziquantel/therapeutic use
7.
Gastroenterology ; 134(4): 1148-58, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18395093

ABSTRACT

BACKGROUND & AIMS: Signal transducer and activator of transcription 3 (STAT3) is known to be activated in human alcoholic liver disease, but its role in the pathogenesis of alcoholic liver injury remains obscure. METHODS: The role of STAT3 in alcoholic liver injury was investigated in hepatocyte-specific STAT3 knockout (H-STAT3KO) mice and macrophage/neutrophil-specific STAT3 KO (M/N-STAT3KO) mice. Alcoholic liver injury was achieved by feeding mice a liquid diet containing 5% ethanol for up to 8 weeks. RESULTS: Compared with wild-type mice, feeding H-STAT3KO mice with an ethanol-containing diet induced greater hepatic steatosis, hypertriglyceridemia, and hepatic expression of lipogenic genes (sterol regulatory element-binding protein, fatty acid synthase, acetyl-CoA carboxylase-1, and stearoyl-CoA desaturase 1), but less inflammation and lower expression of hepatic proinflammatory cytokines. In contrast, ethanol-fed M/N-STAT3KO mice showed more hepatic inflammation, worse injury, and increased hepatic expression of proinflammatory cytokines compared with wild-type mice. Kupffer cells isolated from ethanol-fed H-STAT3KO mice produced similar amounts of reactive oxygen species and tumor necrosis factor alpha, whereas Kupffer cells from M/N-STAT3KO mice produced more reactive oxygen species and tumor necrosis factor alpha compared with wild-type controls. CONCLUSIONS: These findings suggest that STAT3 regulates hepatic inflammation in a cell type-dependent manner during alcoholic liver injury: STAT3 in hepatocytes promotes whereas STAT3 in macrophages/Kupffer cells suppresses inflammation. In addition, activation of hepatocellular STAT3 ameliorates alcoholic fatty liver via inhibition of sterol regulatory element-binding protein 1c expression.


Subject(s)
Hepatocytes/pathology , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Experimental/metabolism , STAT3 Transcription Factor/genetics , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Blotting, Western , Cell Proliferation , Central Nervous System Depressants/toxicity , Chronic Disease , Disease Models, Animal , Ethanol/toxicity , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Immunohistochemistry , Liver Cirrhosis, Alcoholic/genetics , Liver Cirrhosis, Alcoholic/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Mice , Mice, Knockout , RNA, Messenger/genetics , Receptors, CCR2/biosynthesis , Receptors, CCR2/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Sterol Regulatory Element Binding Protein 1/biosynthesis , Sterol Regulatory Element Binding Protein 1/genetics
8.
J Pharmacol Exp Ther ; 328(1): 351-61, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18923087

ABSTRACT

The novel endocannabinoid-like lipid N-arachidonoyl L-serine (ARA-S) causes vasodilation through both endothelium-dependent and -independent mechanisms. We have analyzed the vasorelaxant effect of ARA-S in isolated vascular preparations and its effects on Ca(2+)-activated K(+) currents in human embryonic kidney cells stably transfected with the alpha-subunit of the human, large conductance Ca(+)-activated K(+) (BK(Ca)) channel [human embryonic kidney (HEK) 293hSlo cells]. ARA-S caused relaxation of rat isolated, intact and denuded, small mesenteric arteries preconstricted with (R)-(-)-1-(3-hydroxyphenyl)-2-methylaminoethanol hydrochloride (pEC(50), 5.49 and 5.14, respectively), whereas it caused further contraction of vessels preconstricted with KCl (pEC(50), 5.48 and 4.82, respectively). Vasorelaxation by ARA-S was inhibited by 100 nM iberiotoxin. In human embryonic kidney cells stably transfected with the alpha-subunit of the human BK(Ca) channel cells, ARA-S and its enantiomer, N-arachidonoyl-D-serine, enhanced the whole-cell outward K(+) current with similar potency (pEC(50), 5.63 and 5.32, respectively). The potentiation was not altered by the beta(1) subunit or mediated by ARA-S metabolites, stimulation of known cannabinoid receptors, G proteins, protein kinases, or Ca(2+)-dependent processes; it was lost after patch excision or after membrane cholesterol depletion but was restored after cholesterol reconstitution. BK(Ca) currents were also enhanced by N-arachidonoyl ethanolamide (pEC(50), 5.27) but inhibited by another endocannabinoid, O-arachidonoyl ethanolamine (pIC(50), 6.35), or by the synthetic cannabinoid O-1918 [(-)-1,3-dimethoxy-2-(3-3,4-trans-p-menthadien-(1,8)-yl)-orcinol] (pIC(50), 6.59), which blocks ARA-S-induced vasodilation. We conclude the following. 1) ARA-S directly activates BK(Ca) channels. 2) This interaction does not involve cannabinoid receptors or cytosolic factors but is dependent on the presence of membrane cholesterol. 3) Direct BK(Ca) channel activation probably contributes to the endothelium-independent component of ARA-S-induced mesenteric vasorelaxation. 4) O-1918 is a BK(Ca) channel inhibitor.


Subject(s)
Arachidonic Acids/physiology , Brain/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Serine/analogs & derivatives , Alternative Splicing , Animals , Cell Line , Genetic Variation , Humans , Kidney/enzymology , Large-Conductance Calcium-Activated Potassium Channels/genetics , Male , Membrane Potentials/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Serine/physiology
9.
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
10.
J Clin Invest ; 115(5): 1298-305, 2005 May.
Article in English | MEDLINE | ID: mdl-15864349

ABSTRACT

Endogenous cannabinoids acting at CB(1) receptors stimulate appetite, and CB(1) antagonists show promise in the treatment of obesity. CB(1) (-/-) mice are resistant to diet-induced obesity even though their caloric intake is similar to that of wild-type mice, suggesting that endocannabinoids also regulate fat metabolism. Here, we investigated the possible role of endocannabinoids in the regulation of hepatic lipogenesis. Activation of CB(1) in mice increases the hepatic gene expression of the lipogenic transcription factor SREBP-1c and its targets acetyl-CoA carboxylase-1 and fatty acid synthase (FAS). Treatment with a CB(1) agonist also increases de novo fatty acid synthesis in the liver or in isolated hepatocytes, which express CB(1). High-fat diet increases hepatic levels of the endocannabinoid anandamide (arachidonoyl ethanolamide), CB(1) density, and basal rates of fatty acid synthesis, and the latter is reduced by CB(1) blockade. In the hypothalamus, where FAS inhibitors elicit anorexia, SREBP-1c and FAS expression are similarly affected by CB(1) ligands. We conclude that anandamide acting at hepatic CB(1) contributes to diet-induced obesity and that the FAS pathway may be a common molecular target for central appetitive and peripheral metabolic regulation.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Diet , Endocannabinoids , Fatty Acids/biosynthesis , Liver/metabolism , Obesity/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Arachidonic Acids/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Cannabinoid Receptor Modulators/pharmacology , DNA-Binding Proteins/metabolism , Dietary Fats/metabolism , Fatty Liver/etiology , Fatty Liver/metabolism , Hypothalamus/metabolism , Mice , Obesity/etiology , Polyunsaturated Alkamides , Receptor, Cannabinoid, CB1/genetics , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/metabolism
11.
Ann Surg ; 247(5): 779-90, 2008 May.
Article in English | MEDLINE | ID: mdl-18438115

ABSTRACT

OBJECTIVE: To determine the physiologic importance of endocannabinoids and mitochondrial function in the long-term outcome using a rat model of Roux-en-Y gastric bypass (RYGB) surgery. BACKGROUND: Sixteen million people are morbidly obese and RYGB surgery is the most effective treatment. Endocannabinoids are implicated in appetite stimulation and regulation of peripheral energy metabolism. We hypothesize that down-regulation of endocannabinoids and alterations in mitochondrial function and hormones favoring catabolism contribute to sustained RYGB-induced weight loss. METHODS: Diet-induced obese Sprague-Dawley rats were randomized to sham-operated obese controls, RYGB, and sham-operated obese pair-fed rats. Body weight and food intake were recorded, and food efficiency was calculated. Endocannabinoid levels in skeletal muscle and liver, muscle mitochondrial respiratory complex I-V content, and hormones concentrations were determined 14 and 28 days postsurgery, reflecting rapid and sustained weight loss periods after RYGB, respectively. RESULTS: Compared with pair-fed controls, RYGB rats had significant reduction in body weight and food efficiency (P < 0.001). Increased cholecystokinin, reduced insulin, leptin, adiponectin, T3, and down-regulation of mitochondrial complex I were evident on day 14 postsurgery. On day 28, leptin, insulin, and T3 remained low, whereas adiponectin and cholecystokinin were normal. Along with complex I, the endocannabinoids anandamide in muscle (P = 0.003) and 2-arachidonoylglycerol in liver were significantly down-regulated (P < 0.001). CONCLUSIONS: The attenuated caloric intake, reduced food efficiency, and normalization of hormonal levels on day 28 post-RYGB were associated with significant down-regulation of endocannabinoids anandamide and 2-arachidonoylglycerol in muscle and liver, respectively. These results suggest a role for endocannabinoids in the mechanism of sustained weight loss and RYGB success, and may have implications for treatment of morbid obesity.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Gastric Bypass , Mitochondrial Proteins/metabolism , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Weight Loss/physiology , Animals , Disease Models, Animal , Energy Metabolism/physiology , Male , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley
12.
FASEB J ; 21(8): 1788-800, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17327359

ABSTRACT

Hepatic ischemia-reperfusion (I/R) injury continues to be a fatal complication that can follow liver surgery or transplantation. We have investigated the involvement of the endocannabinoid system in hepatic I/R injury using an in vivo mouse model. Here we report that I/R triggers several-fold increases in the hepatic levels of the endocannabinoids anandamide and 2-arachidonoylglycerol, which originate from hepatocytes, Kupffer, and endothelial cells. The I/R-induced increased tissue endocannabinoid levels positively correlate with the degree of hepatic damage and serum TNF-alpha, MIP-1alpha, and MIP-2 levels. Furthermore, a brief exposure of hepatocytes to various oxidants (H2O2 and peroxynitrite) or inflammatory stimuli (endotoxin and TNF-alpha) also increases endocannabinoid levels. Activation of CB2 cannabinoid receptors by JWH133 protects against I/R damage by decreasing inflammatory cell infiltration, tissue and serum TNF-alpha, MIP-1alpha and MIP-2 levels, tissue lipid peroxidation, and expression of adhesion molecule ICAM-1 in vivo. JWH133 also attenuates the TNF-alpha-induced ICAM-1 and VCAM-1 expression in human liver sinusoidal endothelial cells (HLSECs) and the adhesion of human neutrophils to HLSECs in vitro. Consistent with the protective role of CB2 receptor activation, CB2-/- mice develop increased I/R-induced tissue damage and proinflammatory phenotype. These findings suggest that oxidative/nitrosative stress and inflammatory stimuli may trigger endocannabinoid production, and indicate that targeting CB2 cannabinoid receptors may represent a novel protective strategy against I/R injury. We also demonstrate that CB2-/- mice have a normal hemodynamic profile.


Subject(s)
Liver Diseases/pathology , Receptor, Cannabinoid, CB2/physiology , Reperfusion Injury , Animals , Arachidonic Acids/analysis , Cannabinoid Receptor Modulators/analysis , Cannabinoid Receptor Modulators/biosynthesis , Disease Models, Animal , Endocannabinoids , Glycerides/analysis , Humans , Inflammation , Liver/chemistry , Liver Diseases/metabolism , Mice , Mice, Knockout , Oxidative Stress , Polyunsaturated Alkamides/analysis , Reperfusion Injury/etiology , Up-Regulation
14.
J Leukoc Biol ; 82(6): 1382-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17652447

ABSTRACT

In this study, we have investigated the role of the cannabinoid CB(2) (CB(2)) receptor in an in vivo mouse model of hepatic ischemia/reperfusion (I/R) injury. In addition, we have assessed the role of the CB(2) receptor in TNF-alpha-induced ICAM-1 and VCAM-1 expression in human liver sinusoidal endothelial cells (HLSECs) and in the adhesion of human neutrophils to HLSECs in vitro. The potent CB(2) receptor agonist HU-308, given prior to the induction of I/R, significantly attenuated the extent of liver damage (measured by serum alanine aminotransferase and lactate dehydrogenase) and decreased serum and tissue TNF-alpha, MIP-1alpha, and MIP-2 levels, tissue lipid peroxidation, neutrophil infiltration, DNA fragmentation, and caspase 3 activity. The protective effect of HU-308 against liver damage was also preserved when given right after the ischemic episode. HU-308 also attenuated the TNF-alpha-induced ICAM-1 and VCAM-1 expression in HLSECs, which expressed CB(2) receptors, and the adhesion of human neutrophils to HLSECs in vitro. These findings suggest that selective CB(2) receptor agonists may represent a novel, protective strategy against I/R injury by attenuating oxidative stress, inflammatory response, and apoptosis.


Subject(s)
Apoptosis/drug effects , Cannabinoid Receptor Agonists , Cannabinoids/pharmacology , Ischemia/prevention & control , Liver/pathology , Oxidative Stress/drug effects , Reperfusion Injury/prevention & control , Alanine Transaminase/metabolism , Animals , Cell Adhesion/drug effects , Chemokines/blood , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Inflammation , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/metabolism , L-Lactate Dehydrogenase/metabolism , Leukocytes/cytology , Leukocytes/drug effects , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/enzymology , Mice , Peroxidase/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism
15.
Pharmacol Ther ; 106(2): 133-45, 2005 May.
Article in English | MEDLINE | ID: mdl-15866316

ABSTRACT

Cannabinoids, including the bioactive constituents of the marijuana plant, their synthetic analogs, and endogenous lipids with cannabinoid-like activity, produce their biological effects by interacting with specific receptors. To date, two G protein-coupled cannabinoid receptors have been identified by molecular cloning, CB1 receptors mainly expressed in the brain and mediating most of the neurobehavioral effects of cannabinoids and CB2 receptors expressed by immune and hematopoietic tissues. Recent findings indicate that some cannabinoid effects are not mediated by either CB1 or CB2 receptors, and in some cases there is compelling evidence to implicate additional receptors in these actions. These include transient receptor potential vanilloid 1 (TRPV1) receptors and as-yet-unidentified receptors implicated in the endothelium-dependent vasodilator effect of certain cannabinoids and in the presynaptic inhibition of glutamatergic neurotransmission in the hippocampus. The case for these additional receptors is being reviewed here.


Subject(s)
Brain/drug effects , Cannabinoids/pharmacology , Receptors, Cannabinoid , Animals , Brain/metabolism , Cannabinoid Receptor Antagonists , Cannabinoids/antagonists & inhibitors , Drug Interactions , Humans , Ion Channels/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Cannabinoid/drug effects , Receptors, Cannabinoid/physiology , Rimonabant , TRPV Cation Channels
16.
Circulation ; 110(14): 1996-2002, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15451779

ABSTRACT

BACKGROUND: Endocannabinoids are novel lipid mediators with hypotensive and cardiodepressor activity. Here, we examined the possible role of the endocannabinergic system in cardiovascular regulation in hypertension. METHODS AND RESULTS: In spontaneously hypertensive rats (SHR), cannabinoid-1 receptor (CB1) antagonists increase blood pressure and left ventricular contractile performance. Conversely, preventing the degradation of the endocannabinoid anandamide by an inhibitor of fatty acid amidohydrolase reduces blood pressure, cardiac contractility, and vascular resistance to levels in normotensive rats, and these effects are prevented by CB1 antagonists. Similar changes are observed in 2 additional models of hypertension, whereas in normotensive control rats, the same parameters remain unaffected by any of these treatments. CB1 agonists lower blood pressure much more in SHR than in normotensive Wistar-Kyoto rats, and the expression of CB1 is increased in heart and aortic endothelium of SHR compared with Wistar-Kyoto rats. CONCLUSIONS: We conclude that endocannabinoids tonically suppress cardiac contractility in hypertension and that enhancing the CB1-mediated cardiodepressor and vasodilator effects of endogenous anandamide by blocking its hydrolysis can normalize blood pressure. Targeting the endocannabinoid system offers novel therapeutic strategies in the treatment of hypertension.


Subject(s)
Arachidonic Acids/pharmacology , Blood Pressure/drug effects , Dronabinol/analogs & derivatives , Hypertension/physiopathology , Myocardial Contraction/drug effects , Receptor, Cannabinoid, CB1/drug effects , Vascular Resistance/drug effects , Amidohydrolases/antagonists & inhibitors , Angiotensin II/toxicity , Animals , Arachidonic Acids/metabolism , Benzamides/pharmacology , Benzyl Compounds/pharmacology , Blood Pressure/physiology , Camphanes/pharmacology , Carbamates/pharmacology , Dronabinol/pharmacology , Endocannabinoids , Endothelium, Vascular/metabolism , Hypertension/chemically induced , Hypertension/genetics , Male , Models, Cardiovascular , Myocardial Contraction/physiology , Myocardium/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides , Pyrazoles/pharmacology , Rats , Rats, Inbred Dahl , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/physiology , Rimonabant , Up-Regulation , Vascular Resistance/physiology , Vasodilation/drug effects , Vasodilation/physiology , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
17.
Cell Biosci ; 5: 25, 2015.
Article in English | MEDLINE | ID: mdl-26064446

ABSTRACT

BACKGROUND: Interleukin-22 (IL-22), a cytokine with important functions in anti-microbial defense and tissue repair, has been recently suggested to have beneficial effects in obesity and metabolic syndrome in some but not in other studies. Here, we re-examined the effects of IL-22 on obesity, insulin resistance, and hepatic glucose metabolism. RESULTS: Genetic deletion of IL-22 did not affect high-fat-diet (HFD)-induced obesity and insulin resistance. IL-22 transgenic mice with relatively high levels of circulating IL-22 (~600 pg/ml) were completely resistant to Concanavalin A-induced liver injury but developed the same degree of high fat diet (HFD)-induced obesity, insulin resistance, and fatty liver as the wild-type littermate controls. Similarly, chronic treatment with recombinant mouse IL-22 (rmIL-22) protein did not affect HFD-induced obesity and the associated metabolic syndrome. In vivo treatment with a single dose of rmIL-22 downregulated the hepatic expression of gluconeogenic genes and subsequently inhibited hepatic gluconeogenesis and reduced blood glucose levels both in HFD-fed and streptozotocin (STZ)-treated mice without affecting insulin production. In vitro exposure of mouse primary hepatocytes to IL-22 suppressed glucose production and the expression of gluconeogenic genes. These inhibitory effects were partially reversed by blocking STAT3 or the AMPK signaling pathway. CONCLUSION: Biologically active, high levels of IL-22 do not affect obesity and the associated metabolic syndrome. Acute treatment with IL-22 inhibits hepatic gluconeogenesis, which is mediated via the activation of STAT3 and AMPK in hepatocytes.

18.
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
19.
Obesity (Silver Spring) ; 20(10): 1984-94, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22334255

ABSTRACT

Suppressing hyperactive endocannabinoid tone is a critical target for reducing obesity. The backbone of both endocannabinoids 2-arachidonoylglycerol (2-AG) and anandamide (AEA) is the ω-6 fatty acid arachidonic acid (AA). Here we posited that excessive dietary intake of linoleic acid (LA), the precursor of AA, would induce endocannabinoid hyperactivity and promote obesity. LA was isolated as an independent variable to reflect the dietary increase in LA from 1 percent of energy (en%) to 8 en% occurring in the United States during the 20th century. Mice were fed diets containing 1 en% LA, 8 en% LA, and 8 en% LA + 1 en% eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA) in medium-fat diets (35 en% fat) and high-fat diets (60 en%) for 14 weeks from weaning. Increasing LA from 1 en% to 8 en% elevated AA-phospholipids (PL) in liver and erythrocytes, tripled 2-AG + 1-AG and AEA associated with increased food intake, feed efficiency, and adiposity in mice. Reducing AA-PL by adding 1 en% long-chain ω-3 fats to 8 en% LA diets resulted in metabolic patterns resembling 1 en% LA diets. Selectively reducing LA to 1 en% reversed the obesogenic properties of a 60 en% fat diet. These animal diets modeled 20th century increases of human LA consumption, changes that closely correlate with increasing prevalence rates of obesity. In summary, dietary LA increased tissue AA, and subsequently elevated 2-AG + 1-AG and AEA resulting in the development of diet-induced obesity. The adipogenic effect of LA can be prevented by consuming sufficient EPA and DHA to reduce the AA-PL pool and normalize endocannabinoid tone.


Subject(s)
Arachidonic Acid/pharmacology , Arachidonic Acids/metabolism , Cannabinoid Receptor Agonists/pharmacology , Endocannabinoids/pharmacology , Linoleic Acid/pharmacology , Obesity/metabolism , Polyunsaturated Alkamides/metabolism , Animals , Animals, Newborn , Cannabinoid Receptor Modulators/pharmacology , Diet, High-Fat , Endocannabinoids/metabolism , Male , Mice , Obesity/etiology
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