Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 110
1.
J Agric Food Chem ; 70(20): 6108-6122, 2022 May 25.
Article En | MEDLINE | ID: mdl-35536225

Dysregulated hepatic lipogenesis represents a promising druggable target for treating nonalcoholic steatohepatitis (NASH). This work aims to evaluate the therapeutic efficacy of caffeine in a NASH mouse model displaying increased hepatic lipogenesis driven by constitutive hepatic overexpression of the active v-akt murine thymoma viral oncogene homolog (AKT). Caffeine was administered in the AKT mice to study the efficacy in vivo. AKT-transfected and insulin-stimulated human hepatoma cells were used for in vitro experiments. The results demonstrated that caffeine ameliorated hepatic steatosis and inflammatory injury in vivo. Mechanistically, caffeine repressed the AKT/mTORC1 and SREBP-1/ACC/FASN signaling in mice and in vitro. Furthermore, caffeine impaired NF-κB activation by stabilizing IκBα, resulting in a reduction of proinflammatory mediators interleukin-6 (IL-6) and tumor necrosis factor α (TNF-α). Notably, caffeine abolished mTORC1/FASN-dependent MyD88 palmitoylation, which could be essential for its anti-inflammatory potential. Collectively, these results suggest that caffeine consumption could be advantageous in the prevention and therapy of NASH, especially in the subset accompanied by increased de novo lipogenesis.


Caffeine , Non-alcoholic Fatty Liver Disease , Adaptor Proteins, Signal Transducing/metabolism , Animals , Caffeine/metabolism , Caffeine/therapeutic use , Lipogenesis/drug effects , Lipoylation/drug effects , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Myeloid Differentiation Factor 88/drug effects , Myeloid Differentiation Factor 88/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
2.
J Invest Dermatol ; 142(2): 293-302.e1, 2022 02.
Article En | MEDLINE | ID: mdl-34362555

The G protein-coupled MC1R is expressed in melanocytes and has a pivotal role in human skin pigmentation, with reduced function in human genetic variants exhibiting a red hair phenotype and increased melanoma predisposition. Beyond its role in pigmentation, MC1R is increasingly recognized as promoting UV-induced DNA damage repair. Consequently, there is mounting interest in targeting MC1R for therapeutic benefit. However, whether MC1R expression is restricted to melanocytes or is more widely expressed remains a matter of debate. In this paper, we review MC1R function and highlight that unbiased analysis suggests that its expression is restricted to melanocytes, granulocytes, and the brain.


Melanoma/genetics , Receptor, Melanocortin, Type 1/metabolism , Skin Neoplasms/genetics , Animals , Brain/metabolism , DNA Damage/radiation effects , DNA Repair , Disease Models, Animal , Genetic Predisposition to Disease , Genetic Variation , Granulocytes/metabolism , Hair Color , Humans , Lipoylation/drug effects , Loss of Function Mutation , Melanins/metabolism , Melanocytes/metabolism , Melanoma/pathology , Mice , Molecular Targeted Therapy/methods , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Pigmentation , Ultraviolet Rays/adverse effects
3.
Brain Res ; 1774: 147709, 2022 01 01.
Article En | MEDLINE | ID: mdl-34758347

The increase in Aß1-42 is a neurotoxic effect induced by aluminum which can lead to impairment of learning and memory, but its mechanism has yet to be fully elucidated. Studies have shown that APP palmitoylation is appears to be involved in the production process of Aß1-42. Here, we investigated whether APP palmitoylation is related to the increase in Aß caused by aluminum and its specific mechanism of action. In this study, APP palmitoylation was studied in the setting of aluminum-induced increases in Aß1-42 from two perspectives: whole animal experiments and in vitro cell experiments. First, the learning and memory of rats were impaired and the number of rat cortical neurons was decreased after staining with aluminum. Second, the expression of palmitoyl APP, APP in lipid rafts and palmitoyl acyltransferase zDHHC7 both in rat cerebral cortex and PC12 cells increased with the production of Aß1-42 induced by aluminum in a dose-dependent manner. Finally, the intervention with the palmitoylation inhibitors 2-BP and siRNA zDHHC7 in PC12 cells reduced levels of palmitoyl APP, the expression of APP in lipid rafts and the content of Aß1-42 induced by aluminum to a certain extent. Our results indicate that increased APP palmitoylation levels may be related to the increase in Aß1-42 caused by aluminum, and the mechanism may involve APP palmitoylation promoting the accumulation of APP protein on lipid rafts and the cleavage of APP by BACE1 in amyloidogenic pathway. The increase in expression of zDHHC7 may be one of the reasons for the increase in levels of APP palmitoylation caused by aluminum.


Aluminum/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Neurons/drug effects , Peptide Fragments/metabolism , Acetyltransferases/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Learning/drug effects , Lipoylation/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Memory/drug effects , Neurons/metabolism , PC12 Cells , Rats
4.
Life Sci ; 291: 120263, 2022 Feb 15.
Article En | MEDLINE | ID: mdl-34971697

AIMS: Myocardial infarction (MI) is a major global cause of death. Massive cell death leads to inflammation, which is necessary for ensuing wound healing. Extensive inflammation, however, promotes infarct expansion and adverse remodeling. The DNA sensing receptor cyclic GMP-AMP synthase and its downstream signaling effector stimulator of interferon genes (cGAS-STING) is central in innate immune reactions in infections or autoimmunity. Cytosolic double-strand DNA activates the pathway and down-stream inflammatory responses. Recent papers demonstrated that this pathway is also active following MI and that its genetic targeting improves outcome. Thus, we investigated if pharmacologic pathway inhibition is protective after MI in order to test its translational potential. MAIN METHODS: We investigated novel and selective small-molecule STING inhibitors that inhibit STING palmitoylation and multimerization and thereby downstream pathway activation in a preclinical murine MI model. We assessed structural and functional cardiac remodeling, infarct expansion and fibrosis, as well as cardiomyocyte hypertrophy and the expression of inflammatory genes. KEY FINDINGS: Pharmacologic STING inhibition did not reduce mortality due to myocardial rupture in non-reperfused MI. Infarct size at day one was comparable. However, three weeks of pharmacologic STING inhibition after reperfused MI decreased infarct expansion and scarring, increased left ventricular systolic function to levels approaching normal values, and reduced myocardial hypertrophy. SIGNIFICANCE: Selective small-molecule STING inhibition after myocardial infarction has the potential to improve wound healing responses and pathological remodeling and thereby attenuate the development of ischemic heart failure.


Membrane Proteins/metabolism , Myocardial Infarction/metabolism , Nucleotidyltransferases/metabolism , Animals , Heart/physiopathology , Heart Failure/physiopathology , Inflammation/pathology , Lipoylation/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Nucleotidyltransferases/physiology , Signal Transduction , Systole , Ventricular Function, Left/physiology , Ventricular Remodeling/physiology
5.
Int J Biol Sci ; 17(15): 4223-4237, 2021.
Article En | MEDLINE | ID: mdl-34803494

Lipid metabolites are emerging as pivotal regulators of protein function and cell signaling. The availability of intracellular fatty acid is tightly regulated by glycolipid metabolism and may affect human body through many biological mechanisms. Recent studies have demonstrated palmitate, either from exogenous fatty acid uptake or de novo fatty acid synthesis, may serve as the substrate for protein palmitoylation and regulate protein function via palmitoylation. Palmitoylation, the most-studied protein lipidation, encompasses the reversible covalent attachment of palmitate moieties to protein cysteine residues. It controls various cellular physiological processes and alters protein stability, conformation, localization, membrane association and interaction with other effectors. Dysregulation of palmitoylation has been implicated in a plethora of diseases, such as metabolic syndrome, cancers, neurological disorders and infections. Accordingly, it could be one of the molecular mechanisms underlying the impact of palmitate metabolite on cellular homeostasis and human diseases. Herein, we explore the relationship between lipid metabolites and the regulation of protein function through palmitoylation. We review the current progress made on the putative role of palmitate in altering the palmitoylation of key proteins and thus contributing to the pathogenesis of various diseases, among which we focus on metabolic disorders, cancers, inflammation and infections, neurodegenerative diseases. We also highlight the opportunities and new therapeutics to target palmitoylation in disease development.


Cell Communication/drug effects , Cell Communication/physiology , Palmitates/pharmacology , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipoylation/drug effects
6.
PLoS Biol ; 19(10): e3001408, 2021 10.
Article En | MEDLINE | ID: mdl-34695132

We have combined chemical biology and genetic modification approaches to investigate the importance of protein myristoylation in the human malaria parasite, Plasmodium falciparum. Parasite treatment during schizogony in the last 10 to 15 hours of the erythrocytic cycle with IMP-1002, an inhibitor of N-myristoyl transferase (NMT), led to a significant blockade in parasite egress from the infected erythrocyte. Two rhoptry proteins were mislocalized in the cell, suggesting that rhoptry function is disrupted. We identified 16 NMT substrates for which myristoylation was significantly reduced by NMT inhibitor (NMTi) treatment, and, of these, 6 proteins were substantially reduced in abundance. In a viability screen, we showed that for 4 of these proteins replacement of the N-terminal glycine with alanine to prevent myristoylation had a substantial effect on parasite fitness. In detailed studies of one NMT substrate, glideosome-associated protein 45 (GAP45), loss of myristoylation had no impact on protein location or glideosome assembly, in contrast to the disruption caused by GAP45 gene deletion, but GAP45 myristoylation was essential for erythrocyte invasion. Therefore, there are at least 3 mechanisms by which inhibition of NMT can disrupt parasite development and growth: early in parasite development, leading to the inhibition of schizogony and formation of "pseudoschizonts," which has been described previously; at the end of schizogony, with disruption of rhoptry formation, merozoite development and egress from the infected erythrocyte; and at invasion, when impairment of motor complex function prevents invasion of new erythrocytes. These results underline the importance of P. falciparum NMT as a drug target because of the pleiotropic effect of its inhibition.


Erythrocytes/parasitology , Myristic Acid/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Lipoylation/drug effects , Merozoites/drug effects , Merozoites/metabolism , Parasites/drug effects , Parasites/growth & development , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium falciparum/ultrastructure , Solubility , Substrate Specificity/drug effects
7.
Cell Death Dis ; 12(10): 934, 2021 10 12.
Article En | MEDLINE | ID: mdl-34642298

Metformin, traditionally regarded as a hypoglycemic drug, has been studied in other various fields including inflammation. The specific mechanism of metformin's effect on immune cells remains unclear. Herein, it is verified that LPS-induced macrophages are characterized by enhanced endogenous fatty acid synthesis and the inhibition of fatty acid synthase (FASN) downregulates proinflammatory responses. We further show that metformin could suppress such elevation of FASN as well as proinflammatory activation in macrophages. In vivo, metformin treatment ameliorates dextran sulfate sodium (DSS)-induced colitis through impairing proinflammatory activation of colonic lamina propria mononuclear cells (LPMCs). The reduction of FASN by metformin hinders Akt palmitoylation, which further disturbs Akt membrane attachment and its phosphorylation. Metformin-mediated suppression of FASN/Akt pathway and its downstream MAPK signaling contributes to its anti-inflammatory role in macrophages. From the perspective of immunometabolism, our work points towards metformin utilization as an effective and potential intervention against macrophages-involved inflammatory diseases.


Fatty Acid Synthases/metabolism , Inflammation/pathology , Lipoylation , Metformin/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Down-Regulation/drug effects , Enzyme Activation/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lipopolysaccharides , Lipoylation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Palmitates/pharmacology , Phosphorylation/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism
8.
Cell Death Dis ; 12(9): 824, 2021 09 01.
Article En | MEDLINE | ID: mdl-34471096

Cyclic adenosine monophosphate (cAMP) is a master regulator of mitochondrial metabolism but its precise mechanism of action yet remains unclear. Here, we found that a dietary saturated fatty acid (FA), palmitate increased intracellular cAMP synthesis through the palmitoylation of soluble adenylyl cyclase in cardiomyocytes. cAMP further induced exchange protein directly activated by cyclic AMP 1 (Epac1) activation, which was upregulated in the myocardium of obese patients. Epac1 enhanced the activity of a key enzyme regulating mitochondrial FA uptake, carnitine palmitoyltransferase 1. Consistently, pharmacological or genetic Epac1 inhibition prevented lipid overload, increased FA oxidation (FAO), and protected against mitochondrial dysfunction in cardiomyocytes. In addition, analysis of Epac1 phosphoproteome led us to identify two key mitochondrial enzymes of the the ß-oxidation cycle as targets of Epac1, the long-chain FA acyl-CoA dehydrogenase (ACADL) and the 3-ketoacyl-CoA thiolase (3-KAT). Epac1 formed molecular complexes with the Ca2+/calmodulin-dependent protein kinase II (CaMKII), which phosphorylated ACADL and 3-KAT at specific amino acid residues to decrease lipid oxidation. The Epac1-CaMKII axis also interacted with the α subunit of ATP synthase, thereby further impairing mitochondrial energetics. Altogether, these findings indicate that Epac1 disrupts the balance between mitochondrial FA uptake and oxidation leading to lipid accumulation and mitochondrial dysfunction, and ultimately cardiomyocyte death.


Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Palmitic Acid/toxicity , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Catecholamines/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Lipoylation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Phosphoproteins/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Solubility , Stress, Physiological/drug effects
9.
Neurobiol Dis ; 158: 105479, 2021 10.
Article En | MEDLINE | ID: mdl-34390831

Huntington disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HTT gene that codes for an elongated polyglutamine tract in the huntingtin (HTT) protein. HTT is subject to multiple post-translational modifications (PTMs) that regulate its cellular function. Mutating specific PTM sites within mutant HTT (mHTT) in HD mouse models can modulate disease phenotypes, highlighting the key role of HTT PTMs in the pathogenesis of HD. These findings have led to increased interest in developing small molecules to modulate HTT PTMs in order to decrease mHTT toxicity. However, the therapeutic efficacy of pharmacological modulation of HTT PTMs in preclinical HD models remains largely unknown. HTT is palmitoylated at cysteine 214 by the huntingtin-interacting protein 14 (HIP14 or ZDHHC17) and 14-like (HIP14L or ZDHHC13) acyltransferases. Here, we assessed if HTT palmitoylation should be regarded as a therapeutic target to treat HD by (1) investigating palmitoylation dysregulation in rodent and human HD model systems, (2) measuring the impact of mHTT-lowering therapy on brain palmitoylation, and (3) evaluating if HTT palmitoylation can be pharmacologically modulated. We show that palmitoylation of mHTT and some HIP14/HIP14L-substrates is decreased early in multiple HD mouse models, and that mHTT palmitoylation decreases further with aging. Lowering mHTT in the brain of YAC128 mice is not sufficient to rescue aberrant palmitoylation. However, we demonstrate that mHTT palmitoylation can be normalized in COS-7 cells, in YAC128 cortico-striatal primary neurons and HD patient-derived lymphoblasts using an acyl-protein thioesterase (APT) inhibitor. Moreover, we show that modulating palmitoylation reduces mHTT aggregation and mHTT-induced cytotoxicity in COS-7 cells and YAC128 neurons.


Huntingtin Protein/genetics , Huntingtin Protein/toxicity , Lipoylation/drug effects , Lipoylation/genetics , Acyltransferases/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cysteine/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Mice , Mutation , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/metabolism , Rats
10.
ACS Chem Biol ; 16(8): 1546-1556, 2021 08 20.
Article En | MEDLINE | ID: mdl-34309372

Protein S-acylation is a dynamic lipid post-translational modification that can modulate the localization and activity of target proteins. In humans, the installation of the lipid onto target proteins is catalyzed by a family of 23 Asp-His-His-Cys domain-containing protein acyltransferases (DHHC-PATs). DHHCs are increasingly recognized as critical players in cellular signaling events and in human disease. However, progress elucidating the functions and mechanisms of DHHC "writers" has been hampered by a lack of chemical tools to perturb their activity in live cells. Herein, we report the synthesis and characterization of cyano-myracrylamide (CMA), a broad-spectrum DHHC family inhibitor with similar potency to 2-bromopalmitate (2BP), the most commonly used DHHC inhibitor in the field. Possessing an acrylamide warhead instead of 2BP's α-halo fatty acid, CMA inhibits DHHC family proteins in cellulo while demonstrating decreased toxicity and avoiding inhibition of the S-acylation eraser enzymes, two of the major weaknesses of 2BP. Our studies show that CMA engages with DHHC family proteins in cells, inhibits protein S-acylation, and disrupts DHHC-regulated cellular events. CMA represents an improved chemical scaffold for untangling the complexities of DHHC-mediated cell signaling by protein S-acylation.


Acrylamides/pharmacology , Acyltransferases/antagonists & inhibitors , CD36 Antigens/metabolism , Enzyme Inhibitors/pharmacology , Acrylamides/chemical synthesis , Acrylamides/toxicity , Acylation/drug effects , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/toxicity , ErbB Receptors/metabolism , Humans , Lipoylation/drug effects , Mice , Protein Processing, Post-Translational/drug effects
11.
Cell Rep ; 36(5): 109479, 2021 08 03.
Article En | MEDLINE | ID: mdl-34320401

Coronaviruses rely on host membranes for entry, establishment of replication centers, and egress. Compounds targeting cellular membrane biology and lipid biosynthetic pathways have previously shown promise as antivirals and are actively being pursued as treatments for other conditions. Here, we test small molecule inhibitors that target the PI3 kinase VPS34 or fatty acid metabolism for anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) activity. Our studies determine that compounds targeting VPS34 are potent SARS-CoV-2 inhibitors. Mechanistic studies with compounds targeting multiple steps up- and downstream of fatty acid synthase (FASN) identify the importance of triacylglycerol production and protein palmitoylation as requirements for efficient viral RNA synthesis and infectious virus production. Further, FASN knockout results in significantly impaired SARS-CoV-2 replication that can be rescued with fatty acid supplementation. Together, these studies clarify roles for VPS34 and fatty acid metabolism in SARS-CoV-2 replication and identify promising avenues for the development of countermeasures against SARS-CoV-2.


Antiviral Agents/pharmacology , COVID-19/virology , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Lipid Metabolism/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Virus Replication/drug effects , Aminopyridines/pharmacology , Animals , Caco-2 Cells , Cell Line , Chlorocebus aethiops , Class III Phosphatidylinositol 3-Kinases/metabolism , Fatty Acid Synthases/drug effects , Fatty Acid Synthases/genetics , Gene Knockout Techniques , Humans , Lipoylation/drug effects , Pyrimidines/pharmacology , RNA, Viral/metabolism , Triglycerides/metabolism , Vero Cells
12.
J Neurosci ; 41(10): 2119-2134, 2021 03 10.
Article En | MEDLINE | ID: mdl-33526476

NMDARs are ligand-gated ion channels that cause an influx of Na+ and Ca2+ into postsynaptic neurons. The resulting intracellular Ca2+ transient triggers synaptic plasticity. When prolonged, it may induce excitotoxicity, but it may also activate negative feedback to control the activity of NMDARs. Here, we report that a transient rise in intracellular Ca2+ (Ca2+ challenge) increases the sensitivity of NMDARs but not AMPARs/kainate receptors to the endogenous inhibitory neurosteroid 20-oxo-5ß-pregnan-3α-yl 3-sulfate and to its synthetic analogs, such as 20-oxo-5ß-pregnan-3α-yl 3-hemipimelate (PAhPim). In cultured hippocampal neurons, 30 µm PAhPim had virtually no effect on NMDAR responses; however, following the Ca2+ challenge, it inhibited the responses by 62%; similarly, the Ca2+ challenge induced a 3.7-fold decrease in the steroid IC50 on recombinant GluN1/GluN2B receptors. The increase in the NMDAR sensitivity to PAhPim was dependent on three cysteines (C849, C854, and C871) located in the carboxy-terminal domain of the GluN2B subunit, previously identified to be palmitoylated (Hayashi et al., 2009). Our experiments suggested that the Ca2+ challenge induced receptor depalmitoylation, and single-channel analysis revealed that this was accompanied by a 55% reduction in the probability of channel opening. Results of in silico modeling indicate that receptor palmitoylation promotes anchoring of the GluN2B subunit carboxy-terminal domain to the plasma membrane and facilitates channel opening. Depalmitoylation-induced changes in the NMDAR pharmacology explain the neuroprotective effect of PAhPim on NMDA-induced excitotoxicity. We propose that palmitoylation-dependent changes in the NMDAR sensitivity to steroids serve as an acute endogenous mechanism that controls NMDAR activity.SIGNIFICANCE STATEMENT There is considerable interest in negative allosteric modulators of NMDARs that could compensate for receptor overactivation by glutamate or de novo gain-of-function mutations in neurodevelopmental disorders. By a combination of electrophysiological, pharmacological, and computational techniques we describe a novel feedback mechanism regulating NMDAR activity. We find that a transient rise in intracellular Ca2+ increases NMDAR sensitivity to inhibitory neurosteroids in a process dependent on GluN2B subunit depalmitoylation. These results improve our understanding of the molecular mechanisms of steroid action at the NMDAR and indeed of the basic properties of this important glutamate-gated ion channel and may aid in the development of therapeutics for treating neurologic and psychiatric diseases related to overactivation of NMDARs without affecting normal physiological functions.


Lipoylation/physiology , Neuroprotection/physiology , Pregnanes/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , HEK293 Cells , Hippocampus/physiology , Humans , Lipoylation/drug effects , Male , Pregnanes/metabolism , Rats , Rats, Wistar
13.
Mol Med Rep ; 22(2): 939-947, 2020 08.
Article En | MEDLINE | ID: mdl-32468006

Although non­alcoholic fatty liver disease (NAFLD) is considered a benign disorder, hepatic steatosis has been proposed to be involved in the tumorigenesis of liver cancer. However, the underlying mechanism for carcinogenesis in fatty liver diseases remains unclear. Cancer stem cells (CSCs) have been hypothesized to serve a key role in tumorigenesis. Tumor formation begins with a subset of heterogeneous cells that share properties with stem cells, such as self­renewal and undifferentiated properties. Our previous study reported that the saturated fatty acid palmitate (PA) significantly enhanced the CSC properties of the HepG2 human liver cancer cell line; however, its underlying mechanisms are unknown. In the present study, a proteomic approach was used to investigate the palmitoylation of proteins in HepG2 CSCs. CSC behavior was induced in HepG2 cells via 200 µM PA. Proteomic analysis was performed to identify post­transcriptional modifications of proteins in HepG2 CSCs in response to PA treatment. The present study identified proteins modified by palmitoylation in HepG2 CSC spheres formed following PA treatment. It was therefore hypothesized that palmitoylation may be crucial for CSC sphere formation. Furthermore, the present study demonstrated that two palmitoylation inhibitors, tunicamycin (5, 10 and 25 µg/ml) and 2­bromohexadecanoic acid (25, 50 and 150 µM), significantly decreased CSC sphere formation without affecting cell viability. An association was identified between sphere formation capacity and tumor­initiating capacity of CSCs. The results of the present study demonstrated that protein palmitoylation may influence the PA­induced CSC tumor­initiating capacity, and that the inhibition of palmitoylation may be a suitable chemopreventive strategy for treating patients with NAFLD.


Lipoylation/drug effects , Liver Neoplasms/drug therapy , Protein Processing, Post-Translational/drug effects , Proteins/metabolism , Spheroids, Cellular/drug effects , Cell Survival/drug effects , Chromatography, Liquid , Hep G2 Cells/drug effects , Hep G2 Cells/metabolism , Hep G2 Cells/pathology , Humans , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Palmitates/pharmacology , Proteins/chemistry , Proteomics , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tandem Mass Spectrometry , Tunicamycin/pharmacology
15.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118620, 2020 02.
Article En | MEDLINE | ID: mdl-31812495

Protein S-palmitoylation, the covalent lipid modification of the side chain of Cys residues with the 16­carbon fatty acid palmitate, is the most common acylation, and it enhances the membrane stability of ion channels. This post-translational modification (PTM) determines a functional mechanism of ion channel life cycle from maturation and membrane trafficking to localization. Especially, neurodevelopment is regulated by balancing the level of synaptic protein palmitoylation/depalmitoylation. Recently, we revealed the pathological role of the transient receptor potential canonical type 5 (TRPC5) channel in striatal neuronal loss during Huntington's disease (HD), which is abnormally activated by oxidative stress. Here, we report a mechanism of TRPC5 palmitoylation at a conserved cysteine residue, that is critical for intrinsic channel activity. Furthermore, we identified the therapeutic effect of TRPC5 depalmitoylation by enhancing the TRPC5 membrane instability on HD striatal cells in order to lower TRPC5 toxicity. Collectively, these findings suggest that controlling S-palmitoylation of the TRPC5 channel as a potential risk factor can modulate TRPC5 channel expression and activity, providing new insights into a therapeutic strategy for neurodegenerative diseases.


Neurons/metabolism , Oxidative Stress , TRPC Cation Channels/metabolism , Amino Acid Motifs , Animals , Antineoplastic Agents, Alkylating/toxicity , Apoptosis/drug effects , Carmustine/toxicity , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Lipoylation/drug effects , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Oxidative Stress/drug effects , Palmitates/pharmacology , Protein Stability , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics
16.
Biochem Soc Trans ; 48(1): 281-290, 2020 02 28.
Article En | MEDLINE | ID: mdl-31872231

The post-translational modification protein S-acylation (commonly known as palmitoylation) plays a critical role in regulating a wide range of biological processes including cell growth, cardiac contractility, synaptic plasticity, endocytosis, vesicle trafficking, membrane transport and biased-receptor signalling. As a consequence, zDHHC-protein acyl transferases (zDHHC-PATs), enzymes that catalyse the addition of fatty acid groups to specific cysteine residues on target proteins, and acyl proteins thioesterases, proteins that hydrolyse thioester linkages, are important pharmaceutical targets. At present, no therapeutic drugs have been developed that act by changing the palmitoylation status of specific target proteins. Here, we consider the role that palmitoylation plays in the development of diseases such as cancer and detail possible strategies for selectively manipulating the palmitoylation status of specific target proteins, a necessary first step towards developing clinically useful molecules for the treatment of disease.


Acyltransferases/metabolism , B7-H1 Antigen/metabolism , Lipoylation/drug effects , Neoplasms/drug therapy , Receptor, Melanocortin, Type 1/metabolism , ras Proteins/metabolism , Animals , Cysteine/metabolism , Drug Discovery/methods , Humans , Lipoylation/physiology , Mice , Neoplasms/metabolism , Palmitoyl-CoA Hydrolase/metabolism , Protein Processing, Post-Translational
17.
Expert Opin Ther Targets ; 23(10): 883-891, 2019 10.
Article En | MEDLINE | ID: mdl-31637934

Introduction: In line with the monoamine hypothesis of major depressive disorder (MDD), the clinical efficacy of the selective serotonin reuptake inhibitor fluoxetine has classically been ascribed to central serotonin enhancing properties. Current research described disturbances in brain energy metabolism in MDD. Additionally, fluoxetine showed beneficial effects in neuropsychiatric disorders associated with central energy imbalance. Areas covered: The effect of in vitro fluoxetine exposure on cellular glucose uptake and cerebral glucose transporter function was assessed in human peripheral blood mononuclear cells (PBMC) and murine neuroblastoma N2a cells. Fluoxetine augmented glucose uptake, measured by utilizing the radionuclide-labled glucose analog [18]F-fluorodeoxyglucose, in PBMC without affecting glucose transporter protein content. Analysis of protein palmitoylation using the acyl-biotinyl exchange method revealed GLUT3 to be palmitoylated in PBMC and N2a cells, while palmitoylation of GLUT1 was detected only in N2a cells. Treatment with fluoxetine significantly increased palmitoylation of GLUT3 in PBMC and strongly induced palmitoylation of GLUT1 in PBMC and N2a cells. Expert opinion: Our findings suggest a novel mechanism exerted by fluoxetine targeting glucose metabolism by regulating glucose transporter palmitoylation. Thus, fluoxetine might evoke its therapeutic effects in neuropsychiatric diseases characterized by disturbances in central energy metabolism at least partly by improving cerebral glucose uptake.


Energy Metabolism/drug effects , Fluoxetine/pharmacology , Glucose/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Adult , Animals , Glucose Transport Proteins, Facilitative/drug effects , Glucose Transport Proteins, Facilitative/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipoylation/drug effects , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neuroblastoma/metabolism
18.
J Endocrinol ; 240(2): 229-241, 2019 02 01.
Article En | MEDLINE | ID: mdl-30400032

The molecular mechanisms underlying the ERα nuclear/cytoplasmic pool that modulates pituitary cell proliferation have been widely described, but it is still not clear how ERα is targeted to the plasma membrane. The aim of this study was to analyse ERα palmitoylation and the plasma membrane ERα (mERα) pool, and their participation in E2-triggered membrane-initiated signalling in normal and pituitary tumour cell growth. Cell cultures were prepared from anterior pituitaries of female Wistar rats and tumour GH3 cells, and treated with 10 nM of oestradiol (E2). The basal expression of ERα was higher in tumour GH3 than in normal pituitary cells. Full-length palmitoylated ERα was observed in normal and pituitary tumour cells, demonstrating that E2 stimulation increased both, ERα in plasma membrane and ERα and caveolin-1 interaction after short-term treatment. In addition, the Dhhc7 and Dhhc21 palmitoylases were negatively regulated after sustained stimulation of E2 for 3 h. Although the uptake of BrdU into the nucleus in normal pituitary cells was not modified by E2, a significant increase in the GH3 tumoural cell, as well as ERK1/2 activation, with this effect being mimicked by PPT, a selective antagonist of ERα. These proliferative effects were blocked by ICI 182780 and the global inhibitor of palmitoylation. These findings indicate that ERα palmitoylation modulated the mERα pool and consequently the ERK1/2 pathway, thereby contributing to pituitary tumour cell proliferation. These results suggest that the plasma membrane ERα pool might be related to the proliferative behaviour of prolactinoma and may be a marker of pituitary tumour growth.


Cell Membrane/metabolism , Cell Proliferation , Estrogen Receptor alpha/metabolism , Pituitary Neoplasms/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cells, Cultured , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Female , Fulvestrant/pharmacology , Gene Expression/drug effects , Lipoylation/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Rats, Wistar
19.
J Am Chem Soc ; 140(50): 17374-17378, 2018 12 19.
Article En | MEDLINE | ID: mdl-30516377

Post-translational S-palmitoylation plays a central role in protein localization, trafficking, stability, aggregation, and cell signaling. Dysregulation of palmitoylation pathways in cells can alter protein function and is the cause of several diseases. Considering the biological and clinical importance of S-palmitoylation, tools for direct, in vivo modulation of this lipid modification would be extremely valuable. Here, we describe a method for the cleavage of native S-palmitoyl groups from proteins in living cells. Using a cell permeable, cysteine-functionalized amphiphile, we demonstrate the direct depalmitoylation of cellular proteins. We show that amphiphile-mediated depalmitoylation (AMD) can effectively cleave S-palmitoyl groups from the native GTPase HRas and successfully depalmitoylate mislocalized proteins in an infantile neuronal ceroid lipofuscinosis (INCL) disease model. AMD enables direct and facile depalmitoylation of proteins in live cells and has potential therapeutic applications for diseases such as INCL, where native protein thioesterase activity is deficient.


Lipoylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cysteine/analogs & derivatives , Cysteine/pharmacology , GAP-43 Protein/chemistry , GAP-43 Protein/metabolism , Humans , Lipopeptides/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins p21(ras)/chemistry
20.
Nature ; 559(7713): 269-273, 2018 07.
Article En | MEDLINE | ID: mdl-29973723

Aberrant activation of innate immune pathways is associated with a variety of diseases. Progress in understanding the molecular mechanisms of innate immune pathways has led to the promise of targeted therapeutic approaches, but the development of drugs that act specifically on molecules of interest remains challenging. Here we report the discovery and characterization of highly potent and selective small-molecule antagonists of the stimulator of interferon genes (STING) protein, which is a central signalling component of the intracellular DNA sensing pathway1,2. Mechanistically, the identified compounds covalently target the predicted transmembrane cysteine residue 91 and thereby block the activation-induced palmitoylation of STING. Using these inhibitors, we show that the palmitoylation of STING is essential for its assembly into multimeric complexes at the Golgi apparatus and, in turn, for the recruitment of downstream signalling factors. The identified compounds and their derivatives reduce STING-mediated inflammatory cytokine production in both human and mouse cells. Furthermore, we show that these small-molecule antagonists attenuate pathological features of autoinflammatory disease in mice. In summary, our work uncovers a mechanism by which STING can be inhibited pharmacologically and demonstrates the potential of therapies that target STING for the treatment of autoinflammatory disease.


Membrane Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Binding Sites , Cell Line , Cysteine/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Hereditary Autoinflammatory Diseases/drug therapy , Hereditary Autoinflammatory Diseases/metabolism , Humans , Lipoylation/drug effects , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Signal Transduction/drug effects , Small Molecule Libraries/analysis , Small Molecule Libraries/metabolism
...