Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Elife ; 122024 Feb 02.
Article in English | MEDLINE | ID: mdl-38305778

ABSTRACT

One primary metabolic manifestation of inflammation is the diversion of cis-aconitate within the tricarboxylic acid (TCA) cycle to synthesize the immunometabolite itaconate. Itaconate is well established to possess immunomodulatory and metabolic effects within myeloid cells and lymphocytes, however, its effects in other organ systems during sepsis remain less clear. Utilizing Acod1 knockout mice that are deficient in synthesizing itaconate, we aimed to understand the metabolic role of itaconate in the liver and systemically during sepsis. We find itaconate aids in lipid metabolism during sepsis. Specifically, Acod1 KO mice develop a heightened level of hepatic steatosis when induced with polymicrobial sepsis. Proteomics analysis reveals enhanced expression of enzymes involved in fatty acid oxidation in following 4-octyl itaconate (4-OI) treatment in vitro. Downstream analysis reveals itaconate stabilizes the expression of the mitochondrial fatty acid uptake enzyme CPT1a, mediated by its hypoubiquitination. Chemoproteomic analysis revealed itaconate interacts with proteins involved in protein ubiquitination as a potential mechanism underlying its stabilizing effect on CPT1a. From a systemic perspective, we find itaconate deficiency triggers a hypothermic response following endotoxin stimulation, potentially mediated by brown adipose tissue (BAT) dysfunction. Finally, by use of metabolic cage studies, we demonstrate Acod1 KO mice rely more heavily on carbohydrates versus fatty acid sources for systemic fuel utilization in response to endotoxin treatment. Our data reveal a novel metabolic role of itaconate in modulating fatty acid oxidation during polymicrobial sepsis.


Subject(s)
Inflammation , Sepsis , Succinates , Mice , Animals , Inflammation/metabolism , Endotoxins , Fatty Acids/metabolism , Lipids
2.
Cell Rep ; 42(1): 111941, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640341

ABSTRACT

Activating the macrophage NLRP3 inflammasome can promote excessive inflammation with severe cell and tissue damage and organ dysfunction. Here, we show that pharmacological or genetic inhibition of pyruvate dehydrogenase kinase (PDHK) significantly attenuates NLRP3 inflammasome activation in murine and human macrophages and septic mice by lowering caspase-1 cleavage and interleukin-1ß (IL-1ß) secretion. Inhibiting PDHK reverses NLRP3 inflammasome-induced metabolic reprogramming, enhances autophagy, promotes mitochondrial fusion over fission, preserves crista ultrastructure, and attenuates mitochondrial reactive oxygen species (ROS) production. The suppressive effect of PDHK inhibition on the NLRP3 inflammasome is independent of its canonical role as a pyruvate dehydrogenase regulator. Our study suggests a non-canonical role of mitochondrial PDHK in promoting mitochondrial stress and supporting NLRP3 inflammasome activation during acute inflammation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Mice , Animals , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Macrophages/metabolism , Inflammation/metabolism , Reactive Oxygen Species/metabolism , Interleukin-1beta/metabolism , Mice, Inbred C57BL
3.
J Leukoc Biol ; 112(6): 1509-1514, 2022 12.
Article in English | MEDLINE | ID: mdl-35866365

ABSTRACT

Balancing high energy-consuming danger resistance and low energy supply of disease tolerance is a universal survival principle that often fails during sepsis. Our research supports the concept that sepsis phosphorylates and deactivates mitochondrial pyruvate dehydrogenase complex control over the tricarboxylic cycle and the electron transport chain. StimulatIng mitochondrial energetics in septic mice and human sepsis cell models can be achieved by inhibiting pyruvate dehydrogenase kinases with the pyruvate structural analog dichloroacetate. Stimulating the pyruvate dehydrogenase complex by dichloroacetate reverses a disruption in the tricarboxylic cycle that induces itaconate, a key mediator of the disease tolerance pathway. Dichloroacetate treatment increases mitochondrial respiration and ATP synthesis, decreases oxidant stress, overcomes metabolic paralysis, regenerates tissue, organ, and innate and adaptive immune cells, and doubles the survival rate in a murine model of sepsis.


Subject(s)
Pyruvic Acid , Sepsis , Mice , Humans , Animals , Pyruvic Acid/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Mitochondria/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Acetates/pharmacology
4.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35730570

ABSTRACT

Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to an infection. The metabolic aberrations associated with sepsis underly an acute and organism-wide hyperinflammatory response and multiple organ dysfunction; however, crosstalk between systemic metabolomic alterations and metabolic reprogramming at organ levels remains unknown. We analyzed substrate utilization by the respiratory exchange ratio, energy expenditure, metabolomic screening, and transcriptional profiling in a cecal ligation and puncture model to show that sepsis increases circulating free fatty acids and acylcarnitines but decreases levels of amino acids and carbohydrates, leading to a drastic shift in systemic fuel preference. Comparative analysis of previously published metabolomics from septic liver indicated a positive correlation with hepatic and plasma metabolites during sepsis. In particular, glycine deficiency was a common abnormality of the plasma and liver during sepsis. Interrogation of the hepatic transcriptome in septic mice suggested that the septic liver may contribute to systemic glycine deficiency by downregulating genes involved in glycine synthesis. Interestingly, intraperitoneal injection of the pyruvate dehydrogenase kinase (PDK) inhibitor dichloroacetate reversed sepsis-induced anorexia, energy imbalance, inflammation, dyslipidemia, hypoglycemia, and glycine deficiency. Collectively, our data indicated that PDK inhibition rescued systemic energy imbalance and metabolic dysfunction in sepsis partly through restoration of hepatic fuel metabolism.


Subject(s)
Sepsis , Acetates/pharmacology , Animals , Energy Metabolism , Feeding Behavior , Glycine , Inflammation/metabolism , Mice , Sepsis/complications
5.
Front Immunol ; 13: 748375, 2022.
Article in English | MEDLINE | ID: mdl-35265064

ABSTRACT

A Krebs cycle intermediate metabolite, itaconate, has gained attention as a potential antimicrobial and autoimmune disease treatment due to its anti-inflammatory effects. While itaconate and its derivatives pose an attractive therapeutic option for the treatment of inflammatory diseases, the effects outside the immune system still remain limited, particularly in the muscle. Therefore, we endeavored to determine if itaconate signaling impacts muscle differentiation. Utilizing the well-established C2C12 model of in vitro myogenesis, we evaluated the effects of itaconate and its derivatives on transcriptional and protein markers of muscle differentiation as well as mitochondrial function. We found itaconate and the derivatives dimethyl itaconate and 4-octyl itaconate disrupt differentiation media-induced myogenesis. A primary biological effect of itaconate is a succinate dehydrogenase (SDH) inhibitor. We find the SDH inhibitors dimethyl malonate and harzianopyridone phenocopie the anti-myogenic effects of itaconate. Furthermore, we find treatment with exogenous succinate results in blunted myogenesis. Together our data indicate itaconate and its derivatives interfere with in vitro myogenesis, potentially through inhibition of SDH and subsequent succinate accumulation. We also show 4-octyl itaconate suppresses injury-induced MYOG expression in vivo. More importantly, our findings suggest the therapeutic potential of itaconate, and its derivatives could be limited due to deleterious effects on myogenesis.


Subject(s)
Succinates , Succinic Acid , Muscle Development , Signal Transduction , Succinates/metabolism , Succinates/pharmacology , Succinates/therapeutic use , Succinic Acid/metabolism , Succinic Acid/pharmacology
6.
Elife ; 102021 02 22.
Article in English | MEDLINE | ID: mdl-33616039

ABSTRACT

Metabolic reprogramming between resistance and tolerance occurs within the immune system in response to sepsis. While metabolic tissues such as the liver are subjected to damage during sepsis, how their metabolic and energy reprogramming ensures survival is unclear. Employing comprehensive metabolomic, lipidomic, and transcriptional profiling in a mouse model of sepsis, we show that hepatocyte lipid metabolism, mitochondrial tricarboxylic acid (TCA) energetics, and redox balance are significantly reprogrammed after cecal ligation and puncture (CLP). We identify increases in TCA cycle metabolites citrate, cis-aconitate, and itaconate with reduced fumarate and triglyceride accumulation in septic hepatocytes. Transcriptomic analysis of liver tissue supports and extends the hepatocyte findings. Strikingly, the administration of the pyruvate dehydrogenase kinase (PDK) inhibitor dichloroacetate reverses dysregulated hepatocyte metabolism and mitochondrial dysfunction. In summary, our data indicate that sepsis promotes hepatic metabolic dysfunction and that targeting the mitochondrial PDC/PDK energy homeostat rebalances transcriptional and metabolic manifestations of sepsis within the liver.


Subject(s)
Dichloroacetic Acid/pharmacology , Hepatocytes/metabolism , Mitochondria/metabolism , Sepsis/drug therapy , Sepsis/metabolism , Animals , Citric Acid Cycle/drug effects , Disease Models, Animal , Energy Metabolism/drug effects , Hepatocytes/pathology , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors
7.
Proc Natl Acad Sci U S A ; 117(46): 28971-28979, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33127753

ABSTRACT

Blocking the action of FSH genetically or pharmacologically in mice reduces body fat, lowers serum cholesterol, and increases bone mass, making an anti-FSH agent a potential therapeutic for three global epidemics: obesity, osteoporosis, and hypercholesterolemia. Here, we report the generation, structure, and function of a first-in-class, fully humanized, epitope-specific FSH blocking antibody with a KD of 7 nM. Protein thermal shift, molecular dynamics, and fine mapping of the FSH-FSH receptor interface confirm stable binding of the Fab domain to two of five receptor-interacting residues of the FSHß subunit, which is sufficient to block its interaction with the FSH receptor. In doing so, the humanized antibody profoundly inhibited FSH action in cell-based assays, a prelude to further preclinical and clinical testing.


Subject(s)
Adipose Tissue/metabolism , Antibodies, Blocking/immunology , Bone and Bones/metabolism , Epitopes , Follicle Stimulating Hormone/immunology , Animals , Antibodies, Blocking/chemistry , Antibodies, Monoclonal , Bone Density , Female , Follicle Stimulating Hormone/chemistry , Follicle Stimulating Hormone, beta Subunit/immunology , Humans , Hypercholesterolemia , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Obesity , Osteoporosis , Receptors, FSH/metabolism
8.
Geroscience ; 42(6): 1649-1661, 2020 12.
Article in English | MEDLINE | ID: mdl-32588342

ABSTRACT

The trend to delay pregnancy in the USA has resulted in the number of advanced maternal age (AMA) pregnancies to also increase. In humans, AMA is associated with a variety of pregnancy-related pathologies such as preeclampsia (PE). While AMA is known to be a factor which contributes to the development of pregnancy-induced diseases, the molecular and cellular mechanisms giving rise to this phenomenon are still very limited. This is due in part to lack of a preclinical model which has physiologic relevance to human pregnancy while also allowing control of environmental and genetic variability inherent in human studies. To determine potential physiologic relevance of the vervet/African green monkey (Chlorocebus aethiops sabaeus) as a preclinical model to study the effects of AMA on adaptations to pregnancy, thirteen age-diverse pregnant vervet monkeys (3-16 years old) were utilized to measure third trimester blood pressure (BP), complete blood count, iron measurements, and hormone levels. Significant associations were observed between third trimester diastolic BP and maternal age. Furthermore, the presence of leukocytosis with enhanced circulating neutrophils was observed in AMA mothers compared to younger mothers. Moreover, we observed a negative relationship between maternal age and estradiol, progesterone, and cortisol levels. Finally, offspring born to AMA mothers displayed a postnatal growth retardation phenotype. These studies demonstrate physiologic impairment in the adaptation to pregnancy in AMA vervet/African green monkeys. Our data indicate that the vervet/African green monkey may serve as a useful preclinical model and tool for deciphering pathological mediators of maternal disease in AMA pregnancy.


Subject(s)
Adaptation, Physiological , Animals , Blood Pressure , Chlorocebus aethiops , Female , Maternal Age , Phenotype , Pregnancy
9.
Cell ; 181(7): 1445-1449, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32533917

ABSTRACT

The COVID19 crisis has magnified the issues plaguing academic science, but it has also provided the scientific establishment with an unprecedented opportunity to reset. Shoring up the foundation of academic science will require a concerted effort between funding agencies, universities, and the public to rethink how we support scientists, with a special emphasis on early career researchers.


Subject(s)
Career Mobility , Research Personnel/trends , Research/trends , Achievement , Biomedical Research , Humans , Research Personnel/education , Science/education , Science/trends , Universities
10.
iScience ; 23(5): 101125, 2020 May 22.
Article in English | MEDLINE | ID: mdl-32428862

ABSTRACT

Increased flux of glucose through glycolysis is a hallmark of inflammatory macrophages and is essential for optimal effector functions. Solute carrier (SLC) 37A2 is an endoplasmic reticulum-anchored phosphate-linked glucose-6-phosphate transporter that is highly expressed in macrophages and neutrophils. We demonstrate that SLC37A2 plays a pivotal role in murine macrophage inflammatory activation and cellular metabolic rewiring. Toll-like receptor (TLR) 4 stimulation by lipopolysaccharide (LPS) rapidly increases macrophage SLC37A2 protein expression. SLC37A2 deletion reprograms macrophages to a hyper-glycolytic process and accelerates LPS-induced inflammatory cytokine production, which partially depends on nicotinamide adenine dinucleotide (NAD+) biosynthesis. Blockade of glycolysis normalizes the differential expression of pro-inflammatory cytokines between control and SLC37A2 deficient macrophages. Conversely, overexpression of SLC37A2 lowers macrophage glycolysis and significantly reduces LPS-induced pro-inflammatory cytokine expression. In conclusion, our study suggests that SLC37A2 dampens murine macrophage inflammation by down-regulating glycolytic reprogramming as a part of macrophage negative feedback system to curtail acute innate activation.

11.
J Lipid Res ; 61(7): 1075-1086, 2020 07.
Article in English | MEDLINE | ID: mdl-32430316

ABSTRACT

The glycerol phosphate pathway produces more than 90% of the liver triacylglycerol (TAG). LysoPA, an intermediate in this pathway, is produced by glycerol-3-phosphate acyltransferase. Glycerophosphodiester phosphodiesterase domain containing 3 (GDPD3), whose gene was recently cloned, contains lysophospholipase D activity, which produces LysoPA from lysophospholipids. Whether human GDPD3 plays a role in hepatic TAG homeostasis is unknown. We hypothesized that human GDPD3 increases LysoPA production and availability in the glycerol phosphate pathway, promoting TAG biosynthesis. To test our hypothesis, we infected C57BL/6J mice with adeno-associated virus encoding a hepatocyte-specific albumin promoter that drives GFP (control) or FLAG-tagged human GDPD3 overexpression and fed the mice chow or a Western diet to induce hepatosteatosis. Hepatic human GDPD3 overexpression induced LysoPA production and increased FA uptake and incorporation into TAG in mouse hepatocytes and livers, ultimately exacerbating Western diet-induced liver steatosis. Our results also showed that individuals with hepatic steatosis have increased GDPD3 mRNA levels compared with individuals without steatosis. Collectively, these findings indicate that upregulation of GDPD3 expression may play a key role in hepatic TAG accumulation and may represent a molecular target for managing hepatic steatosis.


Subject(s)
Fatty Acids/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Liver/metabolism , Lysophospholipids/biosynthesis , Phosphoric Diester Hydrolases/genetics , Animals , Biological Transport/genetics , Gene Expression , Humans , Mice
12.
Commun Biol ; 2: 104, 2019.
Article in English | MEDLINE | ID: mdl-30911679

ABSTRACT

Excessive or chronic stress can lead to a variety of diseases due to aberrant activation of the glucocorticoid receptor (GR), a ligand activated transcription factor. Pregnancy represents a particular window of sensitivity in which excessive stress can have adverse outcomes, particularly on the developing fetus. Here we show maternal hepatic stress hormone responsiveness is diminished via epigenetic silencing of the glucocorticoid receptor during pregnancy. Provocatively, reinstallation of GR to hepatocytes during pregnancy by adeno-associated viral transduction dysregulates genes involved in proliferation, resulting in impaired pregnancy-induced hepatomegaly. Disruption of the maternal hepatic adaptation to pregnancy results in in utero growth restriction (IUGR). These data demonstrate pregnancy antagonizes the liver-specific effects of stress hormone signaling in the maternal compartment to ultimately support the healthy development of embryos.


Subject(s)
Fetal Development/genetics , Gene Silencing , Hepatocytes/metabolism , Maternal Inheritance , Receptors, Glucocorticoid/genetics , Animals , Female , Glucocorticoids/pharmacology , Hepatomegaly/etiology , Hepatomegaly/metabolism , Liver/metabolism , Male , Mice , Pregnancy , Receptors, Glucocorticoid/metabolism
13.
Cell Rep ; 22(10): 2690-2701, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29514097

ABSTRACT

Glucocorticoids (GCs) are master regulators of systemic metabolism. Intriguingly, Cushing's syndrome, a disorder of excessive GCs, phenocopies several menopause-induced metabolic pathologies. Here, we show that the glucocorticoid receptor (GR) drives steatosis in hypogonadal female mice because hepatocyte-specific GR knockout mice are refractory to developing ovariectomy-induced steatosis. Intriguingly, transcriptional profiling revealed that ovariectomy elicits hepatic GC hypersensitivity globally. Hypogonadism-induced GC hypersensitivity results from a loss of systemic but not hepatic estrogen (E2) signaling, given that hepatocyte-specific E2 receptor deletion does not confer GC hypersensitivity. Mechanistically, enhanced chromatin recruitment and ligand-dependent hyperphosphorylation of GR underlie ovariectomy-induced glucocorticoid hypersensitivity. The dysregulated glucocorticoid-mediated signaling present in hypogonadal females is a product of increased follicle-stimulating hormone (FSH) production because FSH treatment in ovary-intact mice recapitulates glucocorticoid hypersensitivity similar to hypogonadal female mice. Our findings uncover a regulatory axis between estradiol, FSH, and hepatic glucocorticoid receptor signaling that, when disrupted, as in menopause, promotes hepatic steatosis.


Subject(s)
Estrogens/deficiency , Fatty Liver/metabolism , Fatty Liver/pathology , Receptors, Glucocorticoid/metabolism , Animals , Chromatin/metabolism , Cushing Syndrome/metabolism , Estradiol/deficiency , Estradiol/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Ligands , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Metabolic Networks and Pathways/drug effects , Mice, Inbred C57BL , Ovariectomy , Ovary/metabolism , Ovary/pathology , Phosphorylation/drug effects , Phosphoserine/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects
14.
FASEB J ; 30(2): 971-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26581598

ABSTRACT

An individual's sex affects gene expression and many inflammatory diseases present in a sex-biased manner. Glucocorticoid receptors (GRs) are regulators of inflammatory genes, but their role in sex-specific responses is unclear. Our goal was to evaluate whether GR differentially regulates inflammatory gene expression in male and female mouse liver. Twenty-five percent of the 251 genes assayed by nanostring analysis were influenced by sex. Of these baseline sexually dimorphic inflammatory genes, 82% was expressed higher in female liver. Pathway analyses defined pattern-recognition receptors as the most sexually dimorphic pathway. We next exposed male and female mice to the proinflammatory stimulus LPS. Female mice had 177 genes regulated by treatment with LPS, whereas males had 149, with only 66% of LPS-regulated genes common between the sexes. To determine the contribution of GR to sexually dimorphic inflammatory genes we performed nanostring analysis on liver-specific GR knockout (LGRKO) mice in the presence or absence of LPS. Comparing LGRKO to GR(flox/flox) revealed that 36 genes required GR for sexually dimorphic expression, whereas 24 genes became sexually dimorphic in LGRKO. Fifteen percent of LPS-regulated genes in GR(flox/flox) were not regulated in male and female LGRKO mice treated with LPS. Thus, GR action is influenced by sex to regulate inflammatory gene expression.


Subject(s)
Gene Expression Regulation , Liver/metabolism , Receptors, Glucocorticoid/biosynthesis , Sex Characteristics , Signal Transduction , Animals , Female , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Liver/pathology , Male , Mice , Mice, Knockout , Receptors, Glucocorticoid/genetics
15.
Endocrinology ; 155(2): 463-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24196358

ABSTRACT

Glucocorticoids have an important role in the resolution of inflammation and clinically they are routinely used to treat allergies, asthma, sepsis, and autoimmune diseases. In addition, glucocorticoids are well recognized to negatively impact the development and function of T cells in the immune system by inducing apoptosis. Less is known however about glucocorticoid function in B lymphocytes. Herein, we demonstrate that the glucocorticoid receptor (GR) is present in B-cell populations isolated from both the spleen and the bone marrow. B-cell populations were found to express more GR than non-B-cell populations from both the spleen and the bone marrow. GR protein was found in all B-cell (B220+) developmental subsets (Mature IgM+IgD+, Immature IgM+IgD-, and Pro/Pre IgM-IgD-) isolated from spleen. GR staining intensity was varied among the B-cell developmental subsets and was found to be higher in B cells isolated from the spleen (secondary lymphoid organ) versus the bone marrow (primary lymphoid organ). Ex vivo cell culture of murine splenocytes and bone marrow lymphocytes indicated that dexamethasone stimulated apoptosis in all B-cell developmental subsets demonstrating glucocorticoid responsiveness. Furthermore, in vivo administration of dexamethasone to adrenalectomized mice reduced B-cell numbers in both spleen and bone marrow. These data suggest that glucocorticoid signaling has an important understudied role in B-cell life-or-death decisions.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Dexamethasone/pharmacology , Receptors, Glucocorticoid/metabolism , Adrenalectomy , Animals , B-Lymphocytes/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Male , Mice , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
16.
Lab Invest ; 91(7): 1007-17, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21464819

ABSTRACT

Cholangiocarcinomas are devastating cancers of biliary origin with limited treatment options. It has previously been shown that the endocannabinoid anandamide exerts antiproliferative effects on cholangiocarcinoma independent of any known cannabinoid receptors, and by the stabilization of lipid rafts, thereby allowing the recruitment and activation of the Fas death receptor complex. Recently, GPR55 was identified as a putative cannabinoid receptor; therefore, the role of GPR55 in the antiproliferative effects of anandamide was evaluated. GPR55 is expressed in all cholangiocarcinoma cells and liver biopsy samples to a similar level as in non-malignant cholangiocytes. Treatment with either anandamide or the GPR55 agonist, O-1602, reduced cholangiocarcinoma cell proliferation in vitro and in vivo. Furthermore, knocking down the expression of GPR55 prevented the antiproliferative effects of anandamide. Coupled to these effects was an increase in JNK activity. The antiproliferative effects of anandamide could be blocked by pretreatment with a JNK inhibitor and the lipid raft disruptors ß-methylcyclodextrin and fillipin III. Activation of GPR55 by anandamide or O-1602 increased the amount of Fas in the lipid raft fractions, which could be blocked by pretreatment with the JNK inhibitor. These data represent the first evidence that GPR55 activation by anandamide can lead to the recruitment and activation of the Fas death receptor complex and that targeting GPR55 activation may be a viable option for the development of therapeutic strategies to treat cholangiocarcinoma.


Subject(s)
Arachidonic Acids/pharmacology , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cannabinoid Receptor Modulators/pharmacology , Cell Proliferation/drug effects , Cholangiocarcinoma/pathology , Polyunsaturated Alkamides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line, Tumor , Endocannabinoids , Humans , Mice , Mice, Nude , Receptors, Cannabinoid
17.
J Pharmacol Exp Ther ; 337(3): 718-23, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21383022

ABSTRACT

FK506 [tacrolimus; hexadecahydro-5,19-dihydroxy-3-[2-(4-hydroxy-3-methoxycyclohexyl)-1-methylethenyl]-14,16-dimethoxy-4,10,12,18-tetramethyl-8-(2-propenyl)-15,19-epoxy-3H-pyrido[2,1-c][1,4]oxa-azacyclotricosine-1,7,20,21(4H,23H)-tetrone] is used clinically to reduce the incidence of allograft rejection; however, chronic administration leads to endothelial dysfunction and hypertension. We have previously shown that FK506 activates Ca(2+)/diacylglycerol-dependent conventional protein kinase C (cPKC), which phosphorylates endothelial nitric oxide synthase (eNOS) at one of its inhibitory sites, Thr495. However, which cPKC isoform is responsible for phosphorylating eNOS Thr495 is unknown. The aim of the current study was to determine the cPKC isoform that is activated by FK506, leading to decreased endothelial function. FK506 reduced endothelium-dependent relaxation responses, yet had no effect on endothelium-independent relaxation responses in aortas from control mice. Of the various cPKC isoforms, only the administration of a PKCß(II) isoform-specific peptide inhibitor restored aortic relaxation responses to that of controls. In aortic endothelial cells, FK506 significantly increased PKCß(II) activation compared with vehicle-treated controls, and this was prevented by a PKCß(II) isoform-specific peptide inhibitor. In addition, a PKCß(II) isoform-specific peptide inhibitor prevented the increase in eNOS Thr495 phosphorylation induced by FK506. Taken together, our results indicate that ß(II) is the cPKC isoform responsible for phosphorylating eNOS at the inhibitory site Thr495 in response to FK506. PKCß(II) inhibition could prove beneficial in ameliorating the endothelial dysfunction and hypertension in patients treated with FK506.


Subject(s)
Aorta/drug effects , Endothelial Cells/drug effects , Immunosuppressive Agents/toxicity , Nitric Oxide Synthase Type III/metabolism , Protein Kinase C/metabolism , Tacrolimus/toxicity , Threonine/metabolism , Animals , Cells, Cultured , Immunosuppressive Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Rats , Tacrolimus/metabolism , Tacrolimus/pharmacology , Vascular Diseases/drug therapy
18.
Dig Liver Dis ; 43(3): 188-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20934397

ABSTRACT

Endocannabinoids are ubiquitous signalling molecules that exert their effects through a number of specific cannabinoid receptors. Recent studies have indicated that this endocannabinoid system is involved in the pathophysiological processes associated with both acute and chronic liver diseases as well as in the complications that arise from these diseases such as hepatic encephalopathy and cardiac problems. Targeting this signalling system has been useful in ameliorating some of the symptoms and consequences in experimental models of these liver diseases. This review summarises the recent advances into our knowledge and understanding of endocannabinoids in liver diseases and highlights potential novel therapeutic strategies that may prove useful to treat these diseases.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Liver Diseases/physiopathology , Receptors, Cannabinoid/metabolism , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/physiopathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/physiopathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/physiopathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/physiopathology , End Stage Liver Disease/complications , End Stage Liver Disease/metabolism , End Stage Liver Disease/physiopathology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/physiopathology , Liver Diseases/metabolism , Liver Diseases/therapy , Liver Failure, Acute/metabolism , Liver Failure, Acute/physiopathology , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...