Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Int J Pharm ; 657: 124132, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38641019

ABSTRACT

Targeting enzymes involved in lipid metabolism is increasingly recognized as a promising anticancer strategy. Efficient inhibition of diacylglycerol O-transferase 1 (DGAT1) can block fatty acid (FA) storage. This, in turn, triggers an increase in free polyunsaturated FA concentration, leading to peroxidation and ferroptosis. In this study, we report the development of a pH-sensitive peptide (pHLIP)-drug conjugate designed to selectively deliver DGAT1 inhibitors to cancer cells nested within the acidic microenvironment of tumors. We utilized two previously established pHLIP sequences for coupling with drugs. The study of DGAT1 conjugates in large unilamellar vesicles (LUVs) of different compositions did not reveal enhanced pH-dependent insertion compared to POPC LUVs. However, using in vitro 3D tumor spheroids, significant antiproliferative effects were observed upon exposure to pHLIP-T863 (DGAT1 inhibitor) conjugates, surpassing the inhibitory activity of T863 alone. In conclusion, our study provides the first evidence that pHLIP-based conjugates with DGAT1 inhibitors have the potential to specifically target the acidic compartment of tumors. Moreover, it sheds light on the limitations of LUV models in capturing the pH-dependency of such conjugates.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Diacylglycerol O-Acyltransferase , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Humans , Hydrogen-Ion Concentration , Cell Proliferation/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Peptides/chemistry , Peptides/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Spheroids, Cellular/drug effects , Tumor Microenvironment/drug effects , Membrane Proteins
2.
J Biol Chem ; 300(4): 107168, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490434

ABSTRACT

Lipids have been previously implicated in the lifecycle of neuroinvasive viruses. However, the role of lipids in programmed cell death and the relationship between programmed cell death and lipid droplets (LDs) in neuroinvasive virus infection remains unclear. Here, we found that the infection of neuroinvasive virus, such as rabies virus and encephalomyocarditis virus could enhance the LD formation in N2a cells, and decreasing LDs production by targeting diacylglycerol acyltransferase could suppress viral replication. The lipidomics analysis revealed that arachidonic acid (AA) was significantly increased after reducing LD formation by restricting diacylglycerol acyltransferase, and AA was further demonstrated to induce ferroptosis to inhibit neuroinvasive virus replication. Moreover, lipid peroxidation and viral replication inhibition could be significantly alleviated by a ferroptosis inhibitor, ferrostatin-1, indicating that AA affected neuroinvasive virus replication mainly through inducing ferroptosis. Furthermore, AA was demonstrated to activate the acyl-CoA synthetase long-chain family member 4-lysophosphatidylcholine acyltransferase 3-cytochrome P450 oxidoreductase axis to induce ferroptosis. Our findings highlight novel cross-talks among viral infection, LDs, and ferroptosis for the first time, providing a potential target for antiviral drug development.


Subject(s)
Arachidonic Acid , Ferroptosis , Lipid Droplets , Virus Replication , Ferroptosis/drug effects , Lipid Droplets/metabolism , Lipid Droplets/drug effects , Animals , Virus Replication/drug effects , Mice , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Encephalomyocarditis virus/drug effects , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Lipid Peroxidation/drug effects , Coenzyme A Ligases/metabolism , Cell Line, Tumor , Humans
3.
Oncogene ; 43(2): 136-150, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37973951

ABSTRACT

Impaired macroautophagy/autophagy flux has been implicated in the treatment of prostate cancer (PCa). However, the mechanism underlying autophagy dysregulation in PCa remains unknown. In the current study, we investigated the role of diacylglycerol acyltransferases 1 (DGAT1) and its potential effects on cellular energy homeostasis and autophagy flux in PCa. The results of immunohistochemical staining suggested that DGAT1 expression was positively corrected with tumor stage and node metastasis, indicating DGAT1 is an important factor involved in the development and progression of PCa. Furthermore, targeting DGAT1 remarkably inhibited cell proliferation in vitro and suppressed PCa growth in xenograft models by triggering severe oxidative stress and subsequently autophagy flux blockage. Mechanically, DGAT1 promoted PCa progression by maintaining cellular energy homeostasis, preserving mitochondrial function, protecting against reactive oxygen species, and subsequently promoting autophagy flux via regulating lipid droplet formation. Moreover, we found that fenofibrate exhibits as an upstream regulator of DGAT1. Fenofibrate performed its anti-PCa effect involved the aforementioned mechanisms, and partially dependent on the regulation of DGAT1. Collectively. These findings indicate that DGAT1 regulates PCa lipid droplets formation and is essential for PCa progression. Targeting DGAT1 might be a promising method to control the development and progression of PCa. Schematic representation of DGAT1 affects autophagy flux by regulating lipid homeostasis and maintaining mitochondrial function in prostate cancer (PCa). PCa is characterized up-regulation of DGAT1, leading to the translocation of free fatty acids into lipid droplets, thereby preventing PCa cell from lipotoxicity. Inhibition of DGAT1 suppresses growth of PCa by inducing oxidative stress and subsequently autophagy flux blockage. Further, the current results revealed that fenofibrate exhibits as an upstream regulator of DGAT1, and fenofibrate plays an anti-PCa role partially dependent on the regulation of DGAT1, suggesting a potential therapeutic approach to ameliorate this refractory tumor.


Subject(s)
Fenofibrate , Prostatic Neoplasms , Humans , Male , Autophagy , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Fenofibrate/metabolism , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Oxidative Stress , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
4.
Microbiologyopen ; 11(6): e1334, 2022 12.
Article in English | MEDLINE | ID: mdl-36479627

ABSTRACT

Dysregulation of lipid metabolism is associated with obesity and metabolic diseases but there is also increasing evidence of a relationship between lipid body excess and cancer. Lipid body synthesis requires diacylglycerol acyltransferases (DGATs) which catalyze the last step of triacylglycerol synthesis from diacylglycerol and acyl-coenzyme A. The DGATs and in particular DGAT2, are therefore considered potential therapeutic targets for the control of these pathologies. Here, the murine and the human DGAT2 were overexpressed in the oleaginous yeast Yarrowia lipolytica deleted for all DGAT activities, to evaluate the functionality of the enzymes in this heterologous host and DGAT activity inhibitors. This work provides evidence that mammalian DGATs expressed in Y. lipolytica are a useful tool for screening chemical libraries to identify potential inhibitors or activators of these enzymes of therapeutic interest.


Subject(s)
Diacylglycerol O-Acyltransferase , Enzyme Inhibitors , Animals , Humans , Mice , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Yarrowia , Enzyme Inhibitors/pharmacology
5.
Bioorg Med Chem Lett ; 62: 128632, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35189320

ABSTRACT

A series of novel spirocyclic DGAT1 inhibitors containing the oxadiazole motif were designed and synthesized for biological evaluation. Several compounds exhibited potent diacylglycerol acyltransferase 1 (DGAT1) inhibitory activity. Optimization of the series led to the identification of five lead compounds 8, 9, 10, 11 and 12 that showed excellent in-vitro activity with IC50 values ranging from 7 to 20 nM against human DGAT1. All compounds demonstrated good druggability as well as microsomal stability and safety profiles such as hERG and CYP. Compound 12 significantly reduced plasma triglyceride levels in-vivo in the mouse model of acute lipid challenge. Significant reduction in plasma TG excursion was observed, thus indicating DGAT1 inhibition in-vivo.


Subject(s)
Carboxylic Acids , Diacylglycerol O-Acyltransferase , Enzyme Inhibitors , Animals , Carboxylic Acids/pharmacology , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Disease Models, Animal , Drug Design , Enzyme Inhibitors/pharmacology , Mice , Oxadiazoles/pharmacology , Triglycerides
6.
Front Immunol ; 12: 722735, 2021.
Article in English | MEDLINE | ID: mdl-34603294

ABSTRACT

Lipid metabolism plays a complex and dynamic role in host-pathogen interaction during Mycobacterium tuberculosis infection. While bacterial lipid metabolism is key to the success of the pathogen, the host also offers a lipid rich environment in the form of necrotic caseous granulomas, making this association beneficial for the pathogen. Accumulation of the neutral lipid triglyceride, as lipid droplets within the cellular cuff of necrotic granulomas, is a peculiar feature of pulmonary tuberculosis. The role of triglyceride synthesis in the TB granuloma and its impact on the disease outcome has not been studied in detail. Here, we identified diacylglycerol O-acyltransferase 1 (DGAT1) to be essential for accumulation of triglyceride in necrotic TB granulomas using the C3HeB/FeJ murine model of infection. Treatment of infected mice with a pharmacological inhibitor of DGAT1 (T863) led to reduction in granuloma triglyceride levels and bacterial burden. A decrease in bacterial burden was associated with reduced neutrophil infiltration and degranulation, and a reduction in several pro-inflammatory cytokines including IL1ß, TNFα, IL6, and IFNß. Triglyceride lowering impacted eicosanoid production through both metabolic re-routing and via transcriptional control. Our data suggests that manipulation of lipid droplet homeostasis may offer a means for host directed therapy in Tuberculosis.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Granuloma/drug therapy , Mycobacterium tuberculosis/immunology , Triglycerides/biosynthesis , Tuberculosis/drug therapy , Animals , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Granuloma/microbiology , Granuloma/pathology , Host-Pathogen Interactions , Male , Mice , Mice, Inbred C3H , Mycobacterium tuberculosis/drug effects , Neutrophil Infiltration , Tuberculosis/complications , Tuberculosis/immunology , Tuberculosis/microbiology
7.
Nat Med ; 27(10): 1836-1848, 2021 10.
Article in English | MEDLINE | ID: mdl-34635855

ABSTRACT

Alterations in lipid metabolism might contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). However, no pharmacological agents are currently approved in the United States or the European Union for the treatment of NAFLD. Two parallel phase 2a studies investigated the effects of liver-directed ACC1/2 inhibition in adults with NAFLD. The first study ( NCT03248882 ) examined the effects of monotherapy with a novel ACC1/2 inhibitor, PF-05221304 (2, 10, 25 and 50 mg once daily (QD)), versus placebo at 16 weeks of treatment; the second study ( NCT03776175 ) investigated the effects of PF-05221304 (15 mg twice daily (BID)) co-administered with a DGAT2 inhibitor, PF-06865571 (300 mg BID), versus placebo after 6 weeks of treatment. The primary endpoint in both studies was percent change from baseline in liver fat assessed by magnetic resonance imaging-proton density fat fraction. Dose-dependent reductions in liver fat reached 50-65% with PF-05221304 monotherapy doses ≥10 mg QD; least squares mean (LSM) 80% confidence interval (CI) was -7.2 (-13.9, 0.0), -17.1 (-22.7, -11.1), -49.9 (-53.3, -46.2), -55.9 (-59.0, -52.4) and -64.8 (-67.5, -62.0) with 16 weeks placebo and PF-05221304 2, 10, 25 and 50 mg QD, respectively. The overall incidence of adverse events (AEs) did not increase with increasing PF-05221304 dose, except for a dose-dependent elevation in serum triglycerides (a known consequence of hepatic acetyl-coenzyme A carboxylase (ACC) inhibition) in 23/305 (8%) patients, leading to withdrawal in 13/305 (4%), and a dose-dependent elevation in other serum lipids. Co-administration of PF-05221304 and PF-06865571 lowered liver fat compared to placebo (placebo-adjusted LSM (90% CI) -44.6% (-54.8, -32.2)). Placebo-adjusted LSM (90% CI) reduction in liver fat was -44.5% (-55.0, -31.7) and -35.4% (-47.4, -20.7) after 6 weeks with PF-05221304 or PF-06865571 alone. AEs were reported for 10/28 (36%) patients after co-administered PF-05221304 and PF-06865571, with no discontinuations due to AEs, and the ACC inhibitor-mediated effect on serum triglycerides was mitigated, suggesting that PF-05221304 and PF-06865571 co-administration has the potential to address some of the limitations of ACC inhibition alone.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Liver/enzymology , Non-alcoholic Fatty Liver Disease/drug therapy , Acetyl-CoA Carboxylase/genetics , Diacylglycerol O-Acyltransferase/genetics , Double-Blind Method , Drug Synergism , Enzyme Inhibitors/adverse effects , Female , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/ultrastructure , Magnetic Resonance Imaging , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Placebos
8.
Dev Comp Immunol ; 124: 104197, 2021 11.
Article in English | MEDLINE | ID: mdl-34228994

ABSTRACT

DGAT1 and DGAT2 are two acyl-CoA:diacylglycerol O-acyltransferase (DGAT) enzymes that catalyze the final step in triglyceride (TG) synthesis. TGs are the primary constituents of lipid droplets (LDs). Although it has been demonstrated that LDs modulate immune and inflammatory responses in CIK cells, little is known about whether DGAT1 and DGAT2 involve in this process. Firstly, grass carp DGAT2 was isolated and characterized, encoding 361 amino acids, and all DGAT2 proteins in genomic structures are conserved in vertebrates. Then, using TLR7 agonist, we induced LDs accumulation in CIK cells. Only DGAT1b and DGAT2 were upregulated in forming TLR7 agonist induced-LDs. Next, we utilized small-molecule inhibitors of DGAT1 and DGAT2. The results indicated that DGAT1 inactivation attenuated TG content and the relative expressions of IFNα3, NF-κB, IL-1ß, and TNFα genes, whereas DGAT2 inhibition decreased TG content and the relative expressions of MyD88, IRF7, IFNα3, NF-κB, IL-1ß, and TNFα genes, implying that DGAT1-generated LDs and DGAT2-generated LDs contribute to TLR7-induced immune response via different signaling pathways. Finally, inhibiting ATF6 effectively decreased DGAT-generated LDs accumulation and the expression of TLR7 signaling-related genes induced by TLR7 agonist, implying that ATF6 UPR pathway may mediate the role of DGAT-generated LDs in TLR7 signaling. Overall, we demonstrate that DGAT1 and DGAT2-catalyzed TAG synthesis may generate different LDs to provide distinct signaling platforms for innate TLR7 signaling.


Subject(s)
Activating Transcription Factor 6/metabolism , Carps/immunology , Diacylglycerol O-Acyltransferase/metabolism , Immunity, Innate , Toll-Like Receptor 7/immunology , Activating Transcription Factor 6/antagonists & inhibitors , Animals , Carps/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Humans , Kidney , Lipid Droplets/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Tissue Distribution , Toll-Like Receptor 7/agonists
9.
Mol Divers ; 25(3): 1481-1495, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34160713

ABSTRACT

DGAT1 plays a crucial controlling role in triglyceride biosynthetic pathways, which makes it an attractive therapeutic target for obesity. Thus, development of DGAT1 inhibitors with novel chemical scaffolds is desired and important in the drug discovery. In this investigation, the multistep virtual screening methods, including machine learning methods and common feature pharmacophore model, were developed and used to identify novel DGAT1 inhibitors from BioDiversity database with 30,000 compounds. 531 compounds were predicted as DGAT1 inhibitors by combination of machine learning methods comprising of SVM, NB and RP models. Then, 12 agents were filtered from 531 compounds by using the common feature pharmacophore model. The 3D chemical structures of the 12 hits coordinated with surface charges and isosurface have been carefully analyzed by the established 3D-QSAR model. Finally, 8 compounds with desired properties were retained from the final hits and have been assigned to another research group to complete the follow-up compound synthesis and biologic evaluation.


Subject(s)
Diacylglycerol O-Acyltransferase/chemistry , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Machine Learning , Models, Molecular , Quantitative Structure-Activity Relationship , Algorithms , Cheminformatics/methods , Databases, Chemical , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , ROC Curve , Reproducibility of Results
10.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070938

ABSTRACT

The excessive accumulation of lipids in hepatocytes induces a type of cytotoxicity called hepatic lipotoxicity, which is a fundamental contributor to liver metabolic diseases (such as NAFLD). Magnesium isoglycyrrhizinate (MGIG), a magnesium salt of the stereoisomer of natural glycyrrhizic acid, is widely used as a safe and effective liver protectant. However, the mechanism by which MGIG protects against NAFLD remains unknown. Based on the significant correlation between NAFLD and the reprogramming of liver metabolism, we aimed to explore the beneficial effects of MGIG from a metabolic viewpoint in this paper. We treated HepaRG cells with palmitic acid (PA, a saturated fatty acid of C16:0) to induce lipotoxicity and then evaluated the antagonistic effect of MGIG on lipotoxicity by investigating the cell survival rate, DNA proliferation rate, organelle damage, and endoplasmic reticulum stress (ERS). Metabolomics, lipidomics, and isotope tracing were used to investigate changes in the metabolite profile, lipid profile, and lipid flux in HepaRG cells under different intervention conditions. The results showed that MGIG can indeed protect hepatocytes against PA-induced cytotoxicity and ERS. In response to the metabolic abnormality of lipotoxicity, MGIG curtailed the metabolic activation of lipids induced by PA. The content of total lipids and saturated lipids containing C16:0 chains increased significantly after PA stimulation and then decreased significantly or even returned to normal levels after MGIG intervention. Lipidomic data show that glycerides and glycerophospholipids were the two most affected lipids. For excessive lipid accumulation in hepatocytes, MGIG can downregulate the expression of the metabolic enzymes (GPATs and DAGTs) involved in triglyceride biosynthesis. In conclusion, MGIG has a positive regulatory effect on the metabolic disorders that occur in hepatocytes under lipotoxicity, and the main mechanisms of this effect are in lipid metabolism, including reducing the total lipid content, reducing lipid saturation, inhibiting glyceride and glycerophospholipid metabolism, and downregulating the expression of metabolic enzymes in lipid synthesis.


Subject(s)
Hepatocytes/drug effects , Lipid Metabolism/drug effects , Metabolome/drug effects , Palmitic Acid/antagonists & inhibitors , Protective Agents/pharmacology , Saponins/pharmacology , Triterpenes/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation , Glycerides/classification , Glycerides/metabolism , Glycerol-3-Phosphate O-Acyltransferase/antagonists & inhibitors , Glycerol-3-Phosphate O-Acyltransferase/genetics , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Glycerophospholipids/classification , Glycerophospholipids/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lipid Metabolism/genetics , Lipidomics , Palmitic Acid/toxicity
11.
Anal Chem ; 93(15): 6071-6079, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33819010

ABSTRACT

The primary goal of high-throughput screening (HTS) is to rapidly survey a broad collection of compounds, numbering from tens of thousands to millions of members, and identify those that modulate the activity of a therapeutic target of interest. For nearly two decades, mass spectrometry has been used as a label-free, direct-detection method for HTS and is widely acknowledged as being less susceptible to interferences than traditional optical techniques. Despite these advantages, the throughput of conventional MS-based platforms like RapidFire or parallel LC-MS, which typically acquire data at speeds of 6-30 s/sample, can still be limiting for large HTS campaigns. To overcome this bottleneck, the field has recently turned to chromatography-free approaches including MALDI-TOF-MS and acoustic droplet ejection-MS, both of which are capable of throughputs of 1 sample/second or faster. In keeping with these advances, we report here on our own characterization of an acoustic droplet ejection, open port interface (ADE-OPI)-MS system as a platform for HTS using the membrane-associated, lipid metabolizing enzyme diacylglycerol acyltransferase 2 (DGAT2) as a model system. We demonstrate for the first time that the platform is capable of ejecting droplets from phase-separated samples, allowing direct coupling of liquid-liquid extraction with OPI-MS analysis. By applying the platform to screen a 6400-member library, we further demonstrate that the ADE-OPI-MS assay is suitable for HTS and also performs comparably to LC-MS, but with an efficiency gain of >20-fold.


Subject(s)
Diacylglycerol O-Acyltransferase , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Acoustics , Chromatography, Liquid , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
BMC Cancer ; 21(1): 252, 2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33750350

ABSTRACT

BACKGROUND: Diacylglycerol-acyltransferase 1 (DGAT1) plays an important role in the energy storage and is involved in cancer progression. A growing number of evidences showed that elevated expression of DGAT1 in cancer tissue indicated a poor outcome in cancer patients. However, the relationship between DGAT1 and gastric cancer is still unclear. Thus, Transcriptomic analysis and in vitro experiments were performed to investigate the role of DGAT1 in gastric cancer, as well as the potential therapy target in gastric cancer treatment. METHODS: We screened the public cancer datasets to identify the expression and function of DGAT1 in gastric cancer and tumor infiltrating lymphocytes. Then we testified the DGAT1 expression and function after sodium oleate treatment in AGS and MKN45 cell line. Finally, we analyzed ration of apoptosis, necrosis in gastric cancer cells by using flow cytometry after administration of DGAT1 inhibitor. RESULTS: Our results showed a highly expression of DGAT1 in gastric cancer tissues (n = 5, p = 0.0004), and tumor-infiltrating macrophages with elevated DGAT1 expression is associated with poor overall survival in gastric cancer patients. In addition, gastric cell lines AGS (n = 3, p < 0.05) and MKN45 (n = 3, p < 0.01) expressed higher level of DGAT1 than human gastric mucosal epithelial cell line GES-1. Administration of DGAT1 inhibitor effectively suppressed functional factors expression and induced cell death in MKN45. CONCLUSION: The findings of this research provide an in-depth insight into the potential role and influences involved in DGAT1 in the gastric cancer patients. And higher expression of DGAT1 leads to lower overall survival (OS) rate in patients with poorly differentiated gastric cancer. Our findings suggest a potential role for DGAT1 in the gastric cancer progression and inhibiting DGAT1 might be a promising strategy in gastric cancer treatment.


Subject(s)
Diacylglycerol O-Acyltransferase/physiology , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/mortality , Tumor-Associated Macrophages/physiology , Apoptosis , Cell Line, Tumor , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , NADPH Oxidase 2/physiology , Prognosis , Stomach Neoplasms/pathology
13.
Fitoterapia ; 151: 104881, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33713740

ABSTRACT

Five new meroterpenes, 12α-Psoracorylifol F (1), 7ß,8α-hydroxy-12ß-Psoracorylifol F (2), 8-ketone-Cyclobakuchiol C (3), 7α,8ß-hydroxy-12ß-Cyclobakuchiol C (4) and 8α-hydroxy-Cyclobakuchiol C (5) together with six known compounds (6-11) were isolated from seeds of Psoralea corylifolia, and their structures were elucidated on the basis of spectroscopic and physicochemical analyses. All the isolates were evaluated for in vitro inhibitory activity against DGAT1/2. Among them, compounds 1-6 were found to exhibit selective inhibitory activity on DGAT1 with IC50 values ranging from 61.5 ± 1.1 to 89.1 ± 1.2 µM.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Psoralea/chemistry , Terpenes/pharmacology , Animals , China , HEK293 Cells , Humans , Molecular Structure , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Seeds/chemistry , Sf9 Cells , Structure-Activity Relationship , Terpenes/isolation & purification
14.
Acta Pharmacol Sin ; 42(2): 264-271, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32737468

ABSTRACT

Previous studies demonstrated that prolonged exposure to elevated levels of free fatty acids (FFA), especially saturated fatty acids, could lead to pancreatic ß-cell apoptosis, which plays an important role in the progression of type 2 diabetes (T2D). Diacylglycerol acyltransferase 1 (DGAT1), an enzyme that catalyzes the final step of triglyceride (TG) synthesis, has been reported as a novel target for the treatment of multiple metabolic diseases. In this study we evaluated the potential beneficial effects of DGAT1 inhibitors on pancreatic ß-cells, and further verified their antidiabetic effects in db/db mice. We showed that DGAT1 inhibitors (4a and LCQ908) at the concentration of 1 µM significantly ameliorated palmitic acid (PA)-induced apoptosis in MIN6 pancreatic ß-cells and primary cultured mouse islets; oral administration of a DGAT1 inhibitor (4a) (100 mg/kg) for 4 weeks significantly reduced the apoptosis of pancreatic islets in db/db mice. Meanwhile, 4a administration significantly decreased fasting blood glucose and TG levels, and improved glucose tolerance and insulin tolerance in db/db mice. Furthermore, we revealed that pretreatment with 4a (1 µM) significantly alleviated PA-induced intracellular lipid accumulation, endoplasmic reticulum (ER) stress, and proinflammatory responses in MIN6 cells, which might contribute to the protective effects of DGAT1 inhibitors on pancreatic ß-cells. These findings provided a better understanding of the antidiabetic effects of DGAT1 inhibitors.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Animals , Apoptosis/drug effects , Blood Glucose/drug effects , Cell Line , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Endoplasmic Reticulum Stress/drug effects , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Palmitic Acid/toxicity
15.
Mol Metab ; 45: 101150, 2021 03.
Article in English | MEDLINE | ID: mdl-33359403

ABSTRACT

OBJECTIVE: Medium-chain fatty acids (MCFAs) play an increasing role in human nutrition. In the liver, one fraction is used for synthesis of MCFA-containing triacylglycerol (MCFA-TG), and the rest is used for oxidative energy production or ketogenesis. We investigated which enzymes catalyse the synthesis of MCFA-TG and how inhibition of MCFA-TG synthesis or fatty acid (FA) oxidation influences the metabolic fate of the MCFAs. METHODS: FA metabolism was followed by time-resolved tracing of alkyne-labelled FAs in freshly isolated mouse hepatocytes. Quantitative data were obtained by mass spectrometry of several hundred labelled lipid species. Wild-type hepatocytes and cells from diacylglycerol acyltransferase (DGAT)1-/- mice were treated with inhibitors against DGAT1, DGAT2, or FA ß-oxidation. RESULTS: Inhibition or deletion of DGAT1 resulted in a reduction of MCFA-TG synthesis by 70%, while long-chain (LC)FA-TG synthesis was reduced by 20%. In contrast, DGAT2 inhibition increased MCFA-TG formation by 50%, while LCFA-TG synthesis was reduced by 5-25%. Inhibition of ß-oxidation by the specific inhibitor teglicar strongly increased MCFA-TG synthesis. In contrast, the widely used ß-oxidation inhibitor etomoxir blocked MCFA-TG synthesis, phenocopying DGAT1 inhibition. CONCLUSIONS: DGAT1 is the major enzyme for hepatic MCFA-TG synthesis. Its loss can only partially be compensated by DGAT2. Specific inhibition of ß-oxidation leads to a compensatory increase in MCFA-TG synthesis, whereas etomoxir blocks both ß-oxidation and MCFA-TG synthesis, indicating a strong off-target effect on DGAT1.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/metabolism , Epoxy Compounds/pharmacology , Fatty Acids/metabolism , Liver/metabolism , Triglycerides/metabolism , Animals , Diacylglycerol O-Acyltransferase/genetics , Hepatocytes/metabolism , Lipid Metabolism , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction
16.
Eur J Pharm Sci ; 158: 105683, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33347980

ABSTRACT

Diacylglycerol acyltransferase 1 (DGAT1) plays a pivotal role in lipid metabolism by catalyzing the committed step in triglyceride (TG) synthesis and has been considered as a potential therapeutic target of multiple metabolic diseases, including dyslipidemia, obesity and type 2 diabetes. Here we report a novel DGAT1 inhibitor, Yhhu2407, which showed a stronger DGAT1 inhibitory activity (IC50 = 18.24 ± 4.72 nM) than LCQ908 (IC50 = 78.24 ± 8.16 nM) in an enzymatic assay and led to a significant reduction in plasma TG after an acute lipid challenge in mice. Pharmacokinetic studies illustrated that Yhhu2407 displayed a low systemic, liver- and intestine-targeted distribution pattern, which is consistent with the preferential tissue expression pattern of DGAT1 and therefore might help to maximize the beneficial pharmacological effects and prevent the occurrence of side effects. Cell-based investigations demonstrated that Yhhu2407 inhibited free fatty acid (FFA)-induced TG accumulation and apolipoprotein B (ApoB)-100 secretion in HepG2 cells. In vivo study also disclosed that Yhhu2407 exerted a beneficial effect on regulating plasma TG and lipoprotein levels in rats, and effectively ameliorated high-fat diet (HFD)-induced dyslipidemia in hamsters. In conclusion, we identified Yhhu2407 as a novel DGAT1 inhibitor with potent efficacy on improving lipid metabolism in rats and HFD-fed hamsters without causing obvious adverse effects.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Lipid Metabolism/drug effects , Animals , Diacylglycerol O-Acyltransferase/metabolism , Mice , Obesity , Rats , Triglycerides
17.
Expert Opin Ther Pat ; 30(11): 873-896, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32878484

ABSTRACT

INTRODUCTION: DGAT and MGAT enzymes play an important role in triacylglycerol (TGA) biosynthesis. Overexpression of these enzymes may lead to accumulation of TGA in adipose tissues causing development of diseases such as obesity and diabetes. High triglyceride levels increase risk factors for atherosclerosis, and increase the risk of heart attack, stroke and other heart diseases. DGAT and MGAT inhibitors are used for the treatment of such metabolic diseases. A number of DGAT and MGAT inhibitors entered into clinical and preclinical stages. However, some adverse effects are associated with them. Thus there is need to develop new, potent and safe DGAT and MGAT inhibitors. AREA COVERED: In this review, the authors carefully searched patent literature and reviewed recent advances since the year 2014. Diverse chemical classes reported in the patents belonging to the category DGAT and MGAT inhibitors have been highlighted. EXPERT OPINION: DGAT and MGAT inhibitors are now gaining significant importance in the treatment of metabolic diseases. Fused heterocycles with a combination of aromatic and aliphatic hydrophobic substituents could offer more potent DGAT and MGAT inhibitors. Previously reported chemical scaffolds and their DGAT and MGAT inhibitory activity could be employed as an input for some in silico studies to discover novel, potent and safe DGAT and MGAT inhibitors.


Subject(s)
Acyltransferases/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Acyltransferases/metabolism , Adipose Tissue/metabolism , Animals , Diacylglycerol O-Acyltransferase/metabolism , Drug Development , Drug Discovery , Humans , Metabolic Diseases/drug therapy , Metabolic Diseases/enzymology , Metabolic Diseases/physiopathology , Patents as Topic , Triglycerides/biosynthesis
18.
Physiol Rep ; 8(15): e14542, 2020 08.
Article in English | MEDLINE | ID: mdl-32786057

ABSTRACT

The intestinal metabolism and transport of triacylglycerol (TAG) play a critical role in dietary TAG absorption, and defects in the process are associated with congenital diarrhea. The final reaction in TAG synthesis is catalyzed by diacylglycerol acyltransferase (DGAT1 and DGAT2), which uses activated fatty acids (FA) as substrates. Loss-of-function mutations in DGAT1 cause watery diarrhea in humans, but mechanisms underlying the relationship between altered DGAT activity and diarrhea remain largely unclear. Here, the effects of DGAT1 and DGAT2 inhibition, alone or in combination, on dietary TAG absorption and diarrhea in mice were investigated by using a selective DGAT1 inhibitor (PF-04620110) and DGAT2 inhibitor (PF-06424439). Simultaneous administration of a single dosing of these inhibitors drastically decreased intestinal TAG secretion into the blood circulatory system and TAG accumulation in the duodenum at 60 min after lipid gavage. Under 60% high-fat diet (HFD) feeding, their repeated simultaneous administration for 2 days induced severe watery diarrhea and occasionally led to death. The diarrhea was accompanied by enhanced fecal FA excretion, intestinal injury and barrier failure. DGAT1 or DGAT2 inhibition alone did not induce the phenotypic changes observed in DGAT1/2 inhibitor-treated mice. The results demonstrate that DGAT1/2 inhibition alters TAG absorption and results in watery diarrhea in mice. DGAT1/2 inhibition-induced diarrhea may be caused by intestinal barrier dysfunction due to dysregulation of the cytotoxic FA metabolism. These findings suggest that DGAT-mediated intestinal TAG synthesis is a vital step for maintaining intestinal barrier integrity under HFD feeding.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diarrhea/metabolism , Fatty Acids/metabolism , Intestinal Mucosa/drug effects , Animals , Diacylglycerol O-Acyltransferase/metabolism , Diarrhea/etiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Imidazoles/pharmacology , Imidazoles/toxicity , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Pyridines/pharmacology , Pyridines/toxicity
19.
Lancet Gastroenterol Hepatol ; 5(9): 829-838, 2020 09.
Article in English | MEDLINE | ID: mdl-32553151

ABSTRACT

BACKGROUND: Diacylglycerol-O-acyltransferase 2 (DGAT2) is one of two enzyme isoforms that catalyse the final step in the synthesis of triglycerides. IONIS-DGAT2Rx is an antisense oligonucleotide inhibitor of DGAT2 that is under clinical investigation for the treatment of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). The aim of this trial was to examine the safety, tolerability, and efficacy of IONIS-DGAT2Rx versus placebo in reducing liver fat in patients with type 2 diabetes and NAFLD. METHODS: This double-blind, randomised, placebo-controlled, phase 2 study consisted of a 2-week screening period, a run-in period of up to 4 weeks, a 13-week treatment period of once-weekly dosing, and a 13-week post-treatment follow-up period. The study was done at 16 clinical research sites in Canada, Poland, and Hungary. Eligible participants were aged 18-75 years, had a body-mass index at screening between 27 kg/m2 and 39 kg/m2, haemoglobin A1c (HbA1c) levels from 7·3% to 9·5%, and liver fat content 10% or greater before randomisation, and agreed to maintain a stable diet and exercise routine throughout the study. Enrolled participants were stratified on the basis of liver fat content during the run-in period (<20% or ≥20%) and then centrally randomised (2:1) to receive once weekly subcutaneous injection of 250 mg IONIS-DGAT2Rx or placebo for 13 weeks. Participants, investigators, funder personnel, and the clinical research organisation staff, including central readers of MRI scans, were all masked to treatment identity. The primary endpoints were the safety, tolerability, and pharmacodynamic effect of IONIS-DGAT2Rx on hepatic steatosis, according to absolute reduction from baseline in liver fat percentage as quantified by MRI-estimated proton density fat fraction and assessed in the per-protocol population. Pharmacodynamic performance was determined in the per-protocol population by the change in liver fat content from baseline to 2 weeks after the last dose. The per-protocol population included all randomised participants who received at least ten doses of study drug, with the first four doses administered in the first 5 weeks, did not miss more than three consecutive weekly doses, and who had no protocol deviations that might affect efficacy. All randomised participants who received at least one dose of study drug were included in the safety analysis. This study is registered with ClinicalTrials.gov, NCT03334214. FINDINGS: Between Nov 3, 2017, and Nov 28, 2018, we screened 173 people for eligibility. 44 were enrolled and randomly assigned to receive either IONIS-DGAT2Rx (29 participants) or placebo (15 participants). After 13 weeks of treatment, the mean absolute reduction from baseline was -5·2% (SD 5·4) in the IONIS-DGAT2Rx group compared with -0·6% (6·1) in the placebo group (treatment difference -4·2%, 95% CI -7·8 to -0·5, p=0·026). Reductions in liver fat were not accompanied by hyperlipidaemia, elevations in serum aminotransferases or plasma glucose, changes in bodyweight, or gastrointestinal side-effects compared with placebo. Six serious adverse events occurred in four patients treated with IONIS-DGAT2Rx. No serious adverse events were reported in the placebo group. One of four patients reported three serious adverse events: acute exacerbation of chronic obstructive pulmonary disease, cardiac arrest, and ischaemic cerebral infarction, each considered severe and not related to study drug. Three of four patients reported one serious adverse event of increased blood triglycerides (severe, unrelated to study drug), deep-vein thrombosis (severe, unlikely to be related to study drug), and acute pancreatitis (mild, unrelated to study drug). INTERPRETATION: Our results suggest that DGAT2 antisense inhibition could be a safe and efficacious strategy for treatment of NAFLD and support further investigation in patients with biopsy-proven NASH. Based on the pharmacological target, the response to treatment observed in this study population could extend to the broader population of patients with NAFLD. FUNDING: Ionis Pharmaceuticals.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/drug therapy , Oligonucleotides, Antisense/antagonists & inhibitors , Aged , Body Mass Index , Canada/epidemiology , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diacylglycerol O-Acyltransferase/administration & dosage , Diacylglycerol O-Acyltransferase/adverse effects , Diacylglycerol O-Acyltransferase/pharmacology , Double-Blind Method , Drug Tolerance , Female , Humans , Hungary/epidemiology , Injections, Subcutaneous , Intra-Abdominal Fat/diagnostic imaging , Intra-Abdominal Fat/drug effects , Magnetic Resonance Imaging/methods , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Non-alcoholic Fatty Liver Disease/pathology , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/pharmacology , Placebos/administration & dosage , Poland/epidemiology , Safety , Treatment Outcome
20.
Article in English | MEDLINE | ID: mdl-32126480

ABSTRACT

Polyunsaturated fatty acids (PUFA) are important components of cellular membranes, serving both structural and signaling functions. Investigation of the functional responses of cells to various PUFA often involves cell culture experiments, which can then inform or guide subsequent in vivo and clinical investigations. In this study, human carcinoma and leukemia cell lines (MCF-7, HepG2, THP-1, Jurkat) were incubated for 3 days in the presence of up to 150 µM of exogenous arachidonic or eicosapentaenoic acids. At concentrations up to 20 µM these PUFA were enriched in cellular phospholipids, but at concentrations of 20 µM or higher cells accumulated large quantities of these PUFA and their elongation products into triglycerides. This coincided with decreased cell proliferation and enhanced apoptosis. Inhibition of DGAT1 but not DGAT2 enhanced the cytotoxic effect of exogenous PUFA suggesting a protective role of PUFA sequestration into TGs. Lower (10 µM) and higher (50 µM) exogenous PUFA concentrations also had different impacts on the expression of PUFA metabolizing enzymes. Overall, these results indicate that caution must be exercised when planning in vitro experiments since elevated concentrations of PUFA can lead to dysfunctional cellular responses that are not predictive of in vivo responses to dietary PUFA.


Subject(s)
Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Cell Culture Techniques/methods , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Protective Agents/metabolism , Protective Agents/pharmacology , Apoptosis/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Hep G2 Cells , Humans , Imidazoles/pharmacology , Jurkat Cells , MCF-7 Cells , Phospholipids/metabolism , Pyridines/pharmacology , THP-1 Cells , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...