Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 13: 902016, 2022.
Article in English | MEDLINE | ID: mdl-36324684

ABSTRACT

Background: Tumor-associated macrophages (TAMs) are one of the most abundant immune cells in the pancreatic cancer stroma and are related to the poor prognosis of pancreatic ductal adenocarcinoma (PDAC) patients. Therefore, targeting tumor-associated macrophages is a possible strategy for the treatment of pancreatic cancer. Purpose: We would like to investigate the role of sphingomyelin synthase 2 (SMS2) and the effect of the synthase 2 selective inhibitor YE2 in TAMs and the pancreatic tumor microenvironment. In addition, we also would like to investigate the mechanism by which YE2 attenuates macrophage M2 polarization. Methods: YE2 was utilized to treat macrophages (in vitro) and mice (in vivo). Western blotting and real-time PCR were used to detect the protein levels and mRNA levels of macrophage M2 polarization markers and their downstream signaling pathways. Sphingomyelin synthase 2 gene knockout (KO) mice and their controls were used to establish a PANC-02 orthotopic pancreatic cancer model, and immune cell infiltration in the tumor tissue was analyzed by immunohistochemistry (IHC). Results: We found that sphingomyelin synthase 2 mRNA expression is positively correlated with tumor-associated macrophages, the immunosuppressive microenvironment, and poor prognosis in pancreatic ductal adenocarcinoma patients. Sphingomyelin synthase 2 deficiency was confirmed to have an inhibitory effect on the growth of orthotopic PANC-02 tumors in vivo. The deficiency not only reduced the infiltration of tumor-associated macrophages but also regulated other immune components in the tumor microenvironment. In tissue culture, YE2 inhibited M2 polarization in both bone marrow-derived macrophages (BMDMs) and THP-1 macrophages and eliminated the protumor effect of M2 macrophages. In the mouse model, YE2 treatment reduced the infiltration of TAMs and regulated other immune components in the tumor microenvironment, slowing the progression of PANC-02 tumors. In terms of mechanism, we found that the inhibition of sphingomyelin synthase 2 could downregulate the expression of IL4Rα and CSF1R, thereby attenuating M2 polarization. Conclusion: The sphingomyelin synthase 2 inhibitor YE2 or sphingomyelin synthase 2 deficiency can prevent macrophage M2 polarization in pancreatic cancer, and sphingomyelin synthase 2 could be a new potential target for the treatment of pancreatic cancer.

2.
Acta Biochim Biophys Sin (Shanghai) ; 55(1): 117-130, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36331295

ABSTRACT

Phosphatidylcholines (PCs) are major phospholipids in the mammalian cell membrane. Structural remodeling of PCs is associated with many biological processes. Lysophosphatidylcholine acyltransferase 3 (Lpcat3), which catalyzes the incorporation of polyunsaturated fatty acyl chains into the sn-2 site of PCs, plays an important role in maintaining plasma membrane fluidity. Adipose tissue is one of the main distribution organs of Lpcat3, while the relationship between Lpcat3 and adipose tissue dysfunction during overexpansion remains unknown. In this study, we reveal that both polyunsaturated PC content and Lpcat3 expression are increased in abdominal adipose tissues of high-fat diet-fed mice when compared with chow-diet-fed mice, indicating that Lpcat3 is involved in adipose tissue overexpansion and dysfunction. Our experiments in 3T3-L1 adipocytes show that inhibition of Lpcat3 does not change triglyceride accumulation but increases palmitic acid-induced inflammation and lipolysis. Conversely, Lpcat3 overexpression exhibits anti-inflammatory and anti-lipolytic effects. Furthermore, mechanistic studies demonstrate that Lpcat3 deficiency promotes reactive oxygen species (ROS) generation by increasing NOX enzyme activity by facilitating the translocation of NOX4 to lipid rafts, thereby aggregating 3T3-L1 adipocyte inflammation induced by palmitic acid. Moreover, overexpression of Lpcat3 exhibits the opposite effects. These findings suggest that Lpcat3 protects adipocytes from inflammation during adipose tissue overexpansion by reducing ROS generation. In conclusion, our study demonstrates that Lpcat3 deficiency promotes palmitic acid-induced inflammation in 3T3-L1 adipocytes by enhancing ROS generation.


Subject(s)
Adipocytes , Palmitic Acid , Animals , Mice , Palmitic Acid/pharmacology , Reactive Oxygen Species/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Inflammation/metabolism , Mammals/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism
3.
Acta Pharmacol Sin ; 42(1): 149-159, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32451413

ABSTRACT

High infiltration of M2-polarized macrophages in the primary tumor indicates unfavorable prognosis and poor overall survival in the patients with triple-negative breast cancer (TNBC). Thus, reversing M2-polarized tumor-associated macrophages in the tumors has been considered as a potential therapeutic strategy for TNBC. Sphingomyelin synthase 2 (SMS2) is the key enzyme for sphingomyelin production, which plays an important role in plasma membrane integrity and function. In this study we investigated whether SMS2 inhibitor or SMS2 gene knockout could reduce macrophages M2 polarization and tumor progression in a mouse model of TNBC. We showed that SMS2 mRNA expression was linked to immunosuppressive tumor microenvironment and poor prognosis in TNBC patients. The knockout of SMS2 or application of 15w (a specific SMS2 inhibitor) markedly decreased the generation of M2-type macrophages in vitro, and reduced the tumor weight and lung metastatic niche formation in a 4T1-TNBC mouse model. We further demonstrated that the in vivo antitumor efficacy of 15w was accompanied by a multifaceted remodeling of tumor immune environment reflecting not only the suppression of M2-type macrophages but also diminished levels of regulatory T cells and myeloid-derived suppressor cells leading to a dramatically improved infiltration of antitumor CD8+ T lymphocytes. Collectively, our results reveal a novel and important role of SMS2 in the protumorigenic function and may offer a new strategy for macrophage-targeted anticancer therapy.


Subject(s)
Macrophages/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism , Triple Negative Breast Neoplasms/physiopathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Progression , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Gene Knockout Techniques , Humans , Immunity, Cellular/drug effects , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Macrophage Activation/drug effects , Macrophage Activation/physiology , Macrophages/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Transferases (Other Substituted Phosphate Groups)/genetics , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology
4.
Mar Drugs ; 18(7)2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32629787

ABSTRACT

Given our previous finding that fluorination at the C18 position of largazole showed reasonably good tolerance towards inhibitory activity and selectivity of histone deacetylases (HDACs), further modification on the valine residue in the fluoro-largazole's macrocyclic moiety with S-Me l-Cysteine or Glycine residue was performed. While the Glycine-modified fluoro analog showed poor activity, the S-Me l-Cysteine-modified analog emerged to be a very potent HDAC inhibitor. Unlike all previously reported C2-modified compounds in the largazole family (including our recent fluoro-largazole analogs) where replacement of the Val residue has failed to provide any potency improvement, the S-Me l-Cysteine-modified analog displayed significantly enhanced (five-nine-fold) inhibition of all the tested HDACs while maintaining the selectivity of HDAC1 over HDAC6, as compared to largazole thiol. A molecular modeling study provided rational explanation and structural evidence for the enhanced inhibitory activity. This new finding will aid the design of novel potent HDAC inhibitors.


Subject(s)
Depsipeptides/chemistry , Depsipeptides/pharmacology , Histone Deacetylases/metabolism , Thiazoles/chemistry , Thiazoles/pharmacology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Molecular Structure , Structure-Activity Relationship
5.
FEBS Open Bio ; 9(10): 1734-1743, 2019 10.
Article in English | MEDLINE | ID: mdl-31376210

ABSTRACT

Lysophosphatidylcholine acyltransferase 3 (LPCAT3) is an important enzyme in phospholipid remodeling, a process that influences the biophysical properties of cell membranes and thus cell function. Multiple lines of evidence suggest that LPCAT3 is involved in several diseases, including atherosclerosis, non-alcoholic steatohepatitis, and carcinoma. Thus, LPCAT3 may have potential as a therapeutic target for these diseases. In the present study, we devised an assay based on reversed-phase HPLC to measure LPCAT3 activity, which may facilitate the identification of LPCAT3 inhibitors and activators. We found that optimal pH and temperature of recombinant human LPCAT3 are 6.0 and 30 °C, respectively. The enzyme Km values for substrates NBD-labelled lysophosphatidylcholine and arachidonoyl CoA were 266.84 ± 3.65 and 11.03 ± 0.51 µmol·L-1 , respectively, and the Vmax was 39.76 ± 1.86 pmol·min-1 ·U-1 . Moreover, we used our new method to determine the IC50 of a known LPCAT inhibitor, TSI-10. In conclusion, this novel assay can be used to measure the effects of compounds on LPCAT3 activity.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/analysis , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Enzyme Assays/methods , 1-Acylglycerophosphocholine O-Acyltransferase/antagonists & inhibitors , Animals , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Mice , Mice, Inbred C57BL , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Temperature
7.
Eur J Med Chem ; 163: 864-882, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30580239

ABSTRACT

The sphingomyelin synthase 2 (SMS2) is a potential target for pharmacological intervention in atherosclerosis. However, so far, few selective SMS2 inhibitors and their pharmacological activities were reported. In this study, a class of 2-benzyloxybenzamides were discovered as novel SMS2 inhibitors through scaffold hopping and structural optimization. Among them, Ly93 as one of the most potent inhibitors exhibited IC50 values of 91 nM and 133.9 µM against purified SMS2 and SMS1 respectively. The selectivity ratio of Ly93 was more than 1400-fold for purified SMS2 over SMS1. The in vitro studies indicated that Ly93 not only dose-dependently diminished apoB secretion from Huh7 cells, but also significantly reduced the SMS activity and increased cholesterol efflux from macrophages. Meanwhile, Ly93 inhibited the secretion of LPS-mediated pro-inflammatory cytokine and chemokine in macrophages. The pharmacokinetic profiles of Ly93 performed on C57BL/6J mice demonstrated that Ly93 was orally efficacious. As a potent selective SMS2 inhibitor, Ly93 significantly decreased the plasma SM levels of C57BL/6J mice. Furthermore, Ly93 was capable of dose-dependently attenuating the atherosclerotic lesions in the root and the entire aorta as well as macrophage content in lesions, in apolipoprotein E gene knockout mice treated with Ly93. In conclusion, we discovered a novel selective SMS2 inhibitor Ly93 and demonstrated its anti-atherosclerotic activities in vivo. The preliminary molecular mechanism-of-action studies revealed its function in lipid homeostasis and inflammation process, which indicated that the selective inhibition of SMS2 would be a promising treatment for atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Benzamides/therapeutic use , Drug Discovery , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Animals , Apolipoproteins E/genetics , Benzamides/chemistry , Benzamides/pharmacology , Cell Line , Homeostasis/drug effects , Humans , Inflammation/drug therapy , Lipid Metabolism , Macrophages/metabolism , Mice , Mice, Knockout
8.
J Biol Chem ; 293(47): 18328-18336, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30305392

ABSTRACT

2-Hydroxy-oleic acid (2OHOA) is a potent anticancer drug that induces cancer cell cycle arrest and apoptosis. Previous studies have suggested that 2OHOA's anticancer effect is mediated by SMS activation in cancer cells, including A549 and U118 cells. To confirm this phenomenon, in this study, we treated both A549 and U118 cells with 2OHOA and measured SMS activity. To our surprise, we found neither 2OHOA-mediated SMS activation nor sphingomyelin accumulation in the cells. However, we noted that 2OHOA significantly reduces phosphatidylcholine in these cells. We also did not observe 2OHOA-mediated SMS activation in mouse tissue homogenates. Importantly, 2OHOA inhibited rather than activated recombinant SMS1 (rSMS1) and rSMS2 in a dose-dependent fashion. Intra-gastric treatment of C57BL/6J mice with 2OHOA for 10 days had no effects on liver and small intestine SMS activities and plasma sphingomyelin levels. The treatment inhibited lysophosphatidylcholine acyltransferase (LPCAT) activity, consistent with the aforementioned reduction in plasma phosphatidylcholine. Because total cellular phosphatidylcholine is used as a predictive biomarker for monitoring tumor responses, the previously reported 2OHOA-mediated cancer suppression could be related to this phosphatidylcholine reduction, which may influence cell membrane structure and properties. We conclude that 2OHOA is not a SMS activator and that its anticancer property may be related to an effect on phosphatidylcholine metabolism.


Subject(s)
Antineoplastic Agents/metabolism , Neoplasms/enzymology , Oleic Acids/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Enzyme Activation , Enzyme Activators/administration & dosage , Enzyme Activators/chemistry , Enzyme Activators/metabolism , Humans , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Oleic Acids/administration & dosage , Oleic Acids/chemistry , Phosphatidylcholines/metabolism , Sphingomyelins/metabolism , Transferases (Other Substituted Phosphate Groups)/chemistry , Transferases (Other Substituted Phosphate Groups)/genetics
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(8): 834-843, 2018 08.
Article in English | MEDLINE | ID: mdl-29673706

ABSTRACT

Levels of polyunsaturated phosphatidylcholine (PC) influence plasma membrane structure and function. Phosphatidylcholine (PC) is synthesized de novo in the Kennedy pathway and then undergoes extensive deacylation/reacylation remodeling via Lands' cycle (non-Kennedy pathway). The reacylation is catalyzed by lysophosphatidylcholine acyltransferase (LPCAT), which adds a polyunsaturated fatty acid at the sn-2 position. Four LPCAT isoforms have been described to date, among which we found LPCAT3 to be the major isoform in adipose tissue, but its exact role in adipogenesis is unclear. In this study, we aimed to investigate whether LPCAT3 activity affects 3T3L1 cell adipogenic differentiation potential and its underline mechanism. Lentivirus-mediated LPCAT3 shRNA expression stably knocked down LPCAT3 in 3T3L1 preadipocytes and LPCAT3 deficiency dramatically reduced the levels of cellular polyunsaturated PCs. Importantly, we found that this deficiency activated the ß-catenin dependent Wnt signaling pathway, which suppressed the expression of adipogenesis-related genes, thereby inhibiting 3T3L1 preadipocyte differentiation and lipid accumulation. Moreover, three different Wnt/ß-catenin pathway inhibitors reversed the effect of LPCAP3 deficiency, suggesting that Wnt/ß-catenin pathway activation is one of the causes for the observed phenotypes. To the best of our knowledge, we show here for the first time that PC remodeling is an important regulator of adipocyte differentiation.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/deficiency , Adipocytes/physiology , Adipogenesis/physiology , Wnt Signaling Pathway/physiology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 3T3-L1 Cells , Acylation/physiology , Animals , Cell Membrane/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Knockdown Techniques , Lipogenesis/physiology , Mice , Phosphatidylcholines/metabolism , RNA, Small Interfering/metabolism
10.
Acta Pharmacol Sin ; 38(10): 1381-1393, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28504248

ABSTRACT

The flavonoid quercetin exhibits significant anticancer activities with few side effects. In the current study, we characterized TL-2-8, a quercetin derivative, as a novel anticancer agent in vitro and in vivo. Cell proliferation and viability were assessed using Cell Counting Kit-8 and CellTiter-Blue assay, respectively. Cell death was examined using PI staining or a TUNEL assay. Mitophagy was determined by measuring autophagic flux and by confocal imaging. Protein expression was examined by Western blotting. We found that TL-2-8 selectively inhibited the proliferation and decreased the viability of various cancer cells (the anti-proliferation IC50 values in MDA-MB-231, MDA-MB-468 and MCF-7 breast cancer cells at 72 h were 8.28, 8.56, and 9.58 µmol/L, respectively), and it displayed only slight cytotoxicity against normal MCF-10A and HEK-293 cells. In MDA-MB-231 and MDA-MB-468 breast cancer cells, TL-2-8 treatment induced the degradation of multiple Hsp90 client proteins without inducing Hsp70. TL-2-8 (3, 6, 12 µmol/L) dose-dependently inhibited the expression of AHA1, an activator of Hsp90 ATPase, and decreased Hsp90-AHA1 complex formation, leading to decreased Hsp90 chaperone function and reduced polo-like kinase 1 (PLK1) signaling. Consequently, impaired mitophagy was induced via the downregulation of lysosomal-associated membrane protein 2 (LAMP2). The in vivo anticancer effects of TL-2-8 were evaluated in an MDA-MB-231 breast cancer xenograft model, which was treated with TL-2-8 (25, 50, 100 mg·kg-1·d-1, po). Administration of TL-2-8 resulted in tumor growth inhibition rates of 37.9%, 58.9% and 70.9%, respectively, whereas quercetin treatment (100 mg·kg-1·d-1, po) produced only a lower tumor growth inhibition rate (49.5%). Furthermore, TL-2-8 treatment significantly extended the lifespan of mice bearing MDA-MB-231 breast cancer cell xenografts. Our results demonstrate that TL-2-8 induces significant cell death and immature mitophagy in breast cancer cells in vitro and in vivo via AHA1 abrogation.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Flavonoids/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Female , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , Humans , In Situ Nick-End Labeling , Inhibitory Concentration 50 , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Mitophagy/drug effects , Molecular Chaperones/metabolism , Xenograft Model Antitumor Assays
11.
Bioorg Med Chem ; 23(18): 6173-84, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26314925

ABSTRACT

Sphingomyelin synthase (SMS) has been proved to be a potential drug target for the treatment of atherosclerosis. However, few SMS inhibitors have been reported. In this paper, structure-based virtual screening was performed on hSMS1. SAPA 1a was discovered as a novel SMS1 inhibitor with an IC50 value of 5.2 µM in enzymatic assay. A series of 2-(4-(N-phenethylsulfamoyl)phenoxy)acetamides (SAPAs) were synthesized and their biological activities toward SMS1 were evaluated. Among them, SAPA 1j was found to be the most potent SMS1 inhibitor with an IC50 value of 2.1 µM in in vitro assay. The molecular docking studies suggested the interaction modes of SMS1 inhibitors and PC with the active site of SMS1. Site-directed mutagenesis validated the involvement of residues Arg342 and Tyr338 in enzymatic sphingomyelin production. The discovery of SAPA derivatives as a novel class of SMS1 inhibitors would advance the development of more effective SMS1 inhibitors.


Subject(s)
Acetamides/chemistry , Enzyme Inhibitors/chemical synthesis , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Acetamides/chemical synthesis , Acetamides/metabolism , Binding Sites , Catalytic Domain , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Docking Simulation , Mutagenesis, Site-Directed , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Structure-Activity Relationship , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism
12.
Gastroenterology ; 149(6): 1519-29, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26226572

ABSTRACT

BACKGROUND & AIMS: Phosphatidylcholines (PCs) are structural and functional constituents of cell membranes. The activity of acyltransferase (lysophosphatidylcholine acyltransferase [LPCAT]) is required for addition of polyunsaturated fatty acids to the sn-2 position of PCs and is therefore required to maintain cell membrane structure and function. LPCAT3 is the most abundant isoform of LPCAT in the small intestine and liver, which are important sites of plasma lipoprotein metabolism. We investigated the effects of Lpcat3 disruption on lipid metabolism in mice. METHODS: We disrupted the gene Lpcat3 in C57BL/6J mice to create LPCAT3 knockout (KO) mice. Livers and small intestinal tissues were collected from LPCAT3 KO and C57BL/6J parental strain (controls), and levels of LPCAT messenger RNAs and protein were measured. Levels of lipids and lipoproteins were measured in plasma samples. We isolated enterocytes from mice and measured levels of RNAs and proteins involved in lipid uptake by real-time polymerase chain reaction and immunoblot assays, respectively. We assessed lipid absorption and PC subspecies in the enterocyte plasma membrane using liquid chromatography with tandem mass spectometry. RESULTS: LPCAT3 KO mice survived only 3 weeks after birth. Oil Red O staining showed that the control but not LPCAT3 KO mice accumulated lipids in the small intestine; levels of Niemann-Pick C1-like 1 (NPC1L1) and fatty acid transporter protein 4 (FATP4), which regulate lipid uptake, were greatly reduced in the small intestines of LPCAT3 KO mice. Oral administration of PC and olive oil allowed the LPCAT3 KO mice to survive with the same body weights as controls, but the KO mice had shorter and wider small-intestinal villi and longer and bigger small intestines. Plasma membranes of enterocytes from LPCAT3 KO mice also had significant reductions in the composition of polyunsaturated PCs and reduced levels of NPC1L1, CD36, and FATP4 proteins. These reductions were associated with reduced intestinal uptake of lipid by the small intestine and reduced plasma levels of cholesterol, phospholipid, and triglyceride. CONCLUSIONS: LPCAT3 KO mice have longer and larger small intestines than control mice, with shorter wide villi, reduced lipid absorption, and lower levels NPC1L1, CD36, and FATP4 proteins. Inhibition of LPCAT3 in the small intestine could be developed as an approach to treat hyperlipidemia.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Enterocytes/metabolism , Intestinal Absorption/physiology , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Lipid Metabolism/physiology , 1-Acylglycerophosphocholine O-Acyltransferase/deficiency , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Body Weight/physiology , CD36 Antigens/metabolism , Cholesterol/blood , Chromatography, Liquid , Fatty Acid Transport Proteins/metabolism , Immunoblotting , Intestinal Absorption/genetics , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Lipid Metabolism/genetics , Liver/metabolism , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Olive Oil/administration & dosage , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/metabolism , Phospholipids/metabolism , RNA, Messenger , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry , Triglycerides/blood
13.
J Lipid Res ; 56(3): 537-545, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25605874

ABSTRACT

Sphingomyelin synthase-related protein (SMSr) synthesizes the sphingomyelin analog ceramide phosphoethanolamine (CPE) in cells. Previous cell studies indicated that SMSr is involved in ceramide homeostasis and is crucial for cell function. To further examine SMSr function in vivo, we generated Smsr KO mice that were fertile and had no obvious phenotypic alterations. Quantitative MS analyses of plasma, liver, and macrophages from the KO mice revealed only marginal changes in CPE and ceramide as well as other sphingolipid levels. Because SMS2 also has CPE synthase activity, we prepared Smsr/Sms2 double KO mice. We found that CPE levels were not significantly changed in macrophages, suggesting that CPE levels are not exclusively dependent on SMSr and SMS2 activities. We then measured CPE levels in Sms1 KO mice and found that Sms1 deficiency also reduced plasma CPE levels. Importantly, we found that expression of Sms1 or Sms2 in SF9 insect cells significantly increased not only SM but also CPE formation, indicating that SMS1 also has CPE synthase activity. Moreover, we measured CPE synthase Km and Vmax for SMS1, SMS2, and SMSr using different NBD ceramides. Our study reveals that all mouse SMS family members (SMSr, SMS1, and SMS2) have CPE synthase activity. However, neither CPE nor SMSr appears to be a critical regulator of ceramide levels in vivo.


Subject(s)
Sphingomyelins/biosynthesis , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , Mice , Mice, Knockout , Sphingomyelins/genetics , Transferases (Other Substituted Phosphate Groups)/genetics
14.
PLoS One ; 9(7): e102641, 2014.
Article in English | MEDLINE | ID: mdl-25032960

ABSTRACT

Sphingomyelin synthase (SMS) plays an important role in plasma atherogenic lipoprotein metabolism, inflammation, and the development of atherosclerosis. To understand whether the impaired apoB secretion and inflammation response is a direct result from lack of SMS activity, in this study, we prepared a series of compounds that inhibit SMS activity. Further, we characterized Dy105, the most potent inhibitor. We found that Dy105 treatment significantly reduces SM levels in SM-rich microdomain on cell membranes. Moreover, we found that SMS inhibition reduces apoB secretion in a human hepatoma cell line and reduces the activation of NFκB and p38, a MAP kinase, in bone marrow derived macrophages. These studies provided further evidence that SMS activity regulates atherogenic lipoprotein metabolism and inflammatory responses. Pharmacologic inhibition of SMS may be a new therapy for atherosclerosis by reducing apoB secretion, and reducing inflammation.


Subject(s)
Apolipoproteins B/metabolism , Hepatocytes/metabolism , Macrophages/immunology , Sphingomyelins/metabolism , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Acetonitriles/pharmacology , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Endotoxins , Enzyme Activation , Humans , Inflammation/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Eur J Med Chem ; 73: 1-7, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24374347

ABSTRACT

Sphingomyelin synthase (SMS), which catalyzes ceramide as one of the substrates to produce sphingomyelin, is a critical factor in the sphingolipid biosynthesis pathway. Recent studies indicated that SMS could serve as a novel potential drug target for the treatment of various metabolic diseases such as insulin resistance and atherosclerosis. However, very few small-molecule inhibitors of SMS are known. In this study, we performed structure-based virtual screening in combination with chemical synthesis and bioassay and discovered a class of small-molecule SMS inhibitors. The most potent compound exhibited an IC50 value lower than 20 µM in an in vitro enzymatic assay. To the best of our knowledge, this is the first time that small-molecule SMS inhibitors with potency close to the micromolar range are publicly revealed. The structure-activity relationship demonstrated by this class of compounds provides insights into the structural features that are essential for effective SMS inhibition.


Subject(s)
Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Animals , Binding Sites , Cell Survival/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Female , Humans , Mice , Mice, Inbred ICR , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry , Small Molecule Libraries/toxicity , Structure-Activity Relationship
16.
Arterioscler Thromb Vasc Biol ; 33(7): 1513-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23640498

ABSTRACT

OBJECTIVE: Sphingolipid de novo biosynthesis is related to nonalcoholic fatty liver disease or hepatic steatosis. However, the mechanism is still unclear. Sphingomyelin synthase (SMS), using ceramide as one of the substrates to produce sphingomyelin, sits at the crossroads of sphingolipid biosynthesis. SMS has 2 isoforms: SMS1 and SMS2. SMS2 is the major isoform in liver. APPROACH AND RESULTS: To investigate the relationship between liver SMS2 activity-mediated sphingolipid changes and hepatic steatosis, we used 2 mouse models: Sms2 liver-specific transgenic and Sms2 knockout mice. We found that Sms2 liver-specific transgenic livers have lower ceramide and higher sphingomyelin, whereas Sms2 knockout livers have higher ceramide and lower sphingomyelin. We also found that liver Sms2 overexpression promoted fatty acid uptake and liver steatosis, whereas Sms2 deficiency had an opposite effect in comparison with their respective controls. Importantly, the exogenous ceramide supplementation to Huh7 cells, a human hepatoma cell line, reduced the expression of peroxisome proliferator-activated receptor γ2 and its target genes, Cd36 and Fsp27. Peroxisome proliferator-activated receptor γ reporter analysis confirmed this phenomenon. Furthermore, peroxisome proliferator-activated receptor γ antagonist treatment significantly decreased triglyceride accumulation in Sms2 liver-specific transgenic liver. CONCLUSIONS: We attributed these effects to ceramide that can suppress peroxisome proliferator-activated receptor γ2, thus reducing the expression of Cd36 and Fsp27 and reducing liver steatosis. After all, SMS2 inhibition in the liver could diminish liver steatosis.


Subject(s)
Ceramides/metabolism , Fatty Liver/enzymology , Liver/enzymology , PPAR gamma/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , CD36 Antigens/metabolism , Cell Line, Tumor , Cholesterol, Dietary , Disease Models, Animal , Down-Regulation , Fatty Liver/genetics , Fatty Liver/pathology , Fatty Liver/prevention & control , Female , Genes, Reporter , Humans , Liver/drug effects , Liver/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Phosphatidylcholines/metabolism , Sphingomyelins/metabolism , Time Factors , Transfection , Transferases (Other Substituted Phosphate Groups)/deficiency , Transferases (Other Substituted Phosphate Groups)/genetics
17.
J Biol Chem ; 287(24): 20122-31, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22511767

ABSTRACT

After de novo biosynthesis phospholipids undergo extensive remodeling by the Lands' cycle. Enzymes involved in phospholipid biosynthesis have been studied extensively but not those involved in reacylation of lysophosphopholipids. One key enzyme in the Lands' cycle is fatty acyl-CoA:lysophosphatidylcholine acyltransferase (LPCAT), which utilizes lysophosphatidylcholine (LysoPC) and fatty acyl-CoA to produce various phosphatidylcholine (PC) species. Four isoforms of LPCAT have been identified. In this study we found that LPCAT3 is the major hepatic isoform, and its knockdown significantly reduces hepatic LPCAT activity. Moreover, we report that hepatic LPCAT3 knockdown increases certain species of LysoPCs and decreases certain species of PC. A surprising observation was that LPCAT3 knockdown significantly reduces hepatic triglycerides. Despite this, these mice had higher plasma triglyceride and apoB levels. Lipoprotein production studies indicated that reductions in LPCAT3 enhanced assembly and secretion of triglyceride-rich apoB-containing lipoproteins. Furthermore, these mice had higher microsomal triglyceride transfer protein (MTP) mRNA and protein levels. Mechanistic studies in hepatoma cells revealed that LysoPC enhances secretion of apoB but not apoA-I in a concentration-dependent manner. Moreover, LysoPC increased MTP mRNA, protein, and activity. In short, these results indicate that hepatic LPCAT3 modulates VLDL production by regulating LysoPC levels and MTP expression.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Carrier Proteins/biosynthesis , Gene Expression Regulation/physiology , Lipoproteins, VLDL/biosynthesis , Liver/metabolism , Lysophosphatidylcholines/metabolism , Triglycerides/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Apolipoprotein B-100 , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Carrier Proteins/genetics , Gene Knockdown Techniques , Lipoproteins, VLDL/genetics , Male , Mice , Protein Biosynthesis/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
18.
Nutr Metab (Lond) ; 8: 89, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22168400

ABSTRACT

Sphingomyelin synthase (SMS) sits at the crossroads of sphingomyelin (SM), ceramide, diacylglycerol (DAG) metabolism. It utilizes ceramide and phosphatidylcholine as substrates to produce SM and DAG, thereby regulating lipid messengers which play a role in cell survival and apoptosis. Furthermore, its product SM has been implicated in atherogenic processes such as retention of lipoproteins in the blood vessel intima. There are two mammalian sphingomyelin synthases: SMS1 and SMS2. SMS1 is found exclusively in the Golgi at steady state, whereas SMS2 exists in the Golgi and plasma membrane. Conventional motifs responsible for protein targeting to the plasma membrane or Golgi are either not present in, or unique to, SMS1 and SMS2. In this study, we examined how SMS1 and SMS2 achieve their respective subcellular localization patterns. Brefeldin A treatment prevented SMS1 and SMS2 from exiting the ER, demonstrating that they transit through the classical secretory pathway. We created truncations and chimeras of SMS1 and SMS2 to define their targeting signals. We found that SMS1 contains a C-terminal Golgi targeting signal and that SMS2 contains a C-terminal plasma membrane targeting signal.

19.
Lipids Health Dis ; 10: 46, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21418611

ABSTRACT

BACKGROUND: Studies have shown that plasma high density lipoprotein cholesterol levels are negatively correlated with the development of atherosclerosis, whereas epidemiological studies have also shown that plasma sphingomyelin level is an independent risk factor for atherosclerosis. METHODS: To evaluate the relationship between cellular sphingomyelin level and cholesterol metabolism, we created two cell lines that overexpressed sphingomyelin synthase 1 or 2 (SMS1 or SMS2), using the Tet-off expression system. RESULTS: We found that SMS1 or SMS2 overexpression in Huh7 cells, a human hepatoma cell line, significantly increased the levels of intracellular sphingomyelin, cholesterol, and apolipoprotein A-I and decreased levels of apolipoprotein A-I and cholesterol in the cell culture medium, implying a defect in both processes. CONCLUSIONS: Our findings indicate that the manipulation of sphingomyelin synthase activity could influence the metabolism of sphingomyelin, cholesterol and apolipoprotein A-I.


Subject(s)
Cholesterol/metabolism , Liver/metabolism , Transferases (Other Substituted Phosphate Groups)/biosynthesis , Apolipoprotein A-I/metabolism , Cell Line, Tumor , Humans , Membrane Microdomains/metabolism , Sphingomyelins , Transferases (Other Substituted Phosphate Groups)/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...