Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Metabolites ; 12(8)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36005620

ABSTRACT

Worldwide, obesity rates have doubled since the 1980s and in the USA alone, almost 40% of adults are obese, which is closely associated with a myriad of metabolic diseases such as type 2 diabetes and arteriosclerosis. Obesity is derived from an imbalance between energy intake and consumption, therefore balancing energy homeostasis is an attractive target for metabolic diseases. One therapeutic approach consists of increasing the number of brown-like adipocytes in the white adipose tissue (WAT). Whereas WAT stores excess energy, brown adipose tissue (BAT) can dissipate this energy overload in the form of heat, increasing energy expenditure and thus inhibiting metabolic diseases. To facilitate BAT production a high-throughput screening approach was developed on previously known drugs using human Simpson-Golabi-Behmel Syndrome (SGBS) preadipocytes. The screening allowed us to discover that zafirlukast, an FDA-approved small molecule drug commonly used to treat asthma, was able to differentiate adipocyte precursors and white-biased adipocytes into functional brown adipocytes. However, zafirlukast is toxic to human cells at higher dosages. Drug-Initiated Activity Metabolomics (DIAM) was used to investigate zafirlukast as a BAT inducer, and the endogenous metabolite myristoylglycine was then discovered to mimic the browning properties of zafirlukast without impacting cell viability. Myristoylglycine was found to be bio-synthesized upon zafirlukast treatment and was unique in inducing brown adipocyte differentiation, raising the possibility of using endogenous metabolites and bypassing the exogenous drugs to potentially alleviate disease, in this case, obesity and other related metabolic diseases.

2.
Nat Commun ; 13(1): 4099, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835746

ABSTRACT

Hypertension and kidney disease have been repeatedly associated with genomic variants and alterations of lysine metabolism. Here, we combined stable isotope labeling with untargeted metabolomics to investigate lysine's metabolic fate in vivo. Dietary 13C6 labeled lysine was tracked to lysine metabolites across various organs. Globally, lysine reacts rapidly with molecules of the central carbon metabolism, but incorporates slowly into proteins and acylcarnitines. Lysine metabolism is accelerated in a rat model of hypertension and kidney damage, chiefly through N-alpha-mediated degradation. Lysine administration diminished development of hypertension and kidney injury. Protective mechanisms include diuresis, further acceleration of lysine conjugate formation, and inhibition of tubular albumin uptake. Lysine also conjugates with malonyl-CoA to form a novel metabolite Nε-malonyl-lysine to deplete malonyl-CoA from fatty acid synthesis. Through conjugate formation and excretion as fructoselysine, saccharopine, and Nε-acetyllysine, lysine lead to depletion of central carbon metabolites from the organism and kidney. Consistently, lysine administration to patients at risk for hypertension and kidney disease inhibited tubular albumin uptake, increased lysine conjugate formation, and reduced tricarboxylic acid (TCA) cycle metabolites, compared to kidney-healthy volunteers. In conclusion, lysine isotope tracing mapped an accelerated metabolism in hypertension, and lysine administration could protect kidneys in hypertensive kidney disease.


Subject(s)
Hypertension , Kidney , Lysine , Albumins/metabolism , Animals , Carbon/metabolism , Disease Models, Animal , Hypertension/metabolism , Kidney/metabolism , Lysine/metabolism , Malonyl Coenzyme A/metabolism , Rats
3.
Nat Commun ; 13(1): 608, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35105890

ABSTRACT

In obesity, signaling through the IRE1 arm of the unfolded protein response exerts both protective and harmful effects. Overexpression of the IRE1-regulated transcription factor XBP1s in liver or fat protects against obesity-linked metabolic deterioration. However, hyperactivation of IRE1 engages regulated IRE1-dependent decay (RIDD) and TRAF2/JNK pro-inflammatory signaling, which accelerate metabolic dysfunction. These pathologic IRE1-regulated processes have hindered efforts to pharmacologically harness the protective benefits of IRE1/XBP1s signaling in obesity-linked conditions. Here, we report the effects of a XBP1s-selective pharmacological IRE1 activator, IXA4, in diet-induced obese (DIO) mice. IXA4 transiently activates protective IRE1/XBP1s signaling in liver without inducing RIDD or TRAF2/JNK signaling. IXA4 treatment improves systemic glucose metabolism and liver insulin action through IRE1-dependent remodeling of the hepatic transcriptome that reduces glucose production and steatosis. IXA4-stimulated IRE1 activation also enhances pancreatic function. Our findings indicate that systemic, transient activation of IRE1/XBP1s signaling engenders multi-tissue benefits that integrate to mitigate obesity-driven metabolic dysfunction.


Subject(s)
Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/pharmacology , X-Box Binding Protein 1/metabolism , Animals , Fatty Liver/metabolism , Gene Expression Regulation , Glucose/metabolism , Homeostasis , Liver/metabolism , Membrane Proteins/genetics , Mice , Mice, Obese , Molecular Medicine , Obesity/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Transcription Factors/metabolism , Unfolded Protein Response , X-Box Binding Protein 1/genetics
4.
Sci Signal ; 14(702): eabf6584, 2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34582249

ABSTRACT

Untargeted metabolomics of disease-associated intestinal microbiota can detect quantitative changes in metabolite profiles and complement other methodologies to reveal the full effect of intestinal dysbiosis. Here, we used the T cell transfer mouse model of colitis to identify small-molecule metabolites with altered abundance due to intestinal inflammation. We applied untargeted metabolomics to detect metabolite signatures in cecal, colonic, and fecal samples from healthy and colitic mice and to uncover differences that would aid in the identification of colitis-associated metabolic processes. We provided an unbiased spatial survey of the GI tract for small molecules, and we identified the likely source of metabolites and biotransformations. Several prioritized metabolites that we detected as being altered in colitis were evaluated for their ability to induce inflammatory signaling in cultured macrophages, such as NF-κB signaling and the expression of cytokines and chemokines upon LPS stimulation. Multiple previously uncharacterized anti-inflammatory and inflammation-augmenting metabolites were thus identified, with phytosphingosine showing the most effective anti-inflammatory activity in vitro. We further demonstrated that oral administration of phytosphingosine decreased inflammation in a mouse model of colitis induced by the compound TNBS. The collection of distinct metabolites we identified and characterized, many of which have not been previously associated with colitis, may offer new biological insight into IBD-associated inflammation and disease pathogenesis.


Subject(s)
Colitis , T-Lymphocytes , Anti-Inflammatory Agents , Humans , Metabolomics
5.
J Vis Exp ; (167)2021 01 25.
Article in English | MEDLINE | ID: mdl-33554974

ABSTRACT

The understanding of the mechanisms underlying adipocyte differentiation and function has greatly benefited from the use of immortalized white preadipocyte cell lines. These cultured cell lines, however, have limitations. They do not fully capture the diverse functional spectrum of the heterogenous adipocyte populations that are now known to exist within white adipose depots. To provide a more physiologically relevant model to study the complexity of white adipose tissue, a protocol has been developed and optimized to enable simultaneous isolation of primary white and brown adipocyte progenitors from newborn mice, their rapid expansion in culture, and their differentiation in vitro into mature, fully functional adipocytes. The primary advantage of isolating primary cells from newborn, rather than adult mice, is that the adipose depots are actively developing and are, therefore, a rich source of proliferating preadipocytes. Primary preadipocytes isolated using this protocol differentiate rapidly upon reaching confluence and become fully mature in 4-5 days, a temporal window that accurately reflects the appearance of developed fat pads in newborn mice. Primary cultures prepared using this strategy can be expanded and studied with high reproducibility, making them suitable for genetic and phenotypic screens and enabling the study of the cell-autonomous adipocyte phenotypes of genetic mouse models. This protocol offers a simple, rapid, and inexpensive approach to study the complexity of adipose tissue in vitro.


Subject(s)
Adipocytes, Brown/cytology , Adipocytes, White/cytology , Cell Differentiation , Cell Separation/methods , Animals , Animals, Newborn , Cells, Cultured , Energy Metabolism , Mice , Reproducibility of Results
6.
Nat Chem Biol ; 16(9): 997-1005, 2020 09.
Article in English | MEDLINE | ID: mdl-32514184

ABSTRACT

Activity-based protein profiling (ABPP) has been used extensively to discover and optimize selective inhibitors of enzymes. Here, we show that ABPP can also be implemented to identify the converse-small-molecule enzyme activators. Using a kinetically controlled, fluorescence polarization-ABPP assay, we identify compounds that stimulate the activity of LYPLAL1-a poorly characterized serine hydrolase with complex genetic links to human metabolic traits. We apply ABPP-guided medicinal chemistry to advance a lead into a selective LYPLAL1 activator suitable for use in vivo. Structural simulations coupled to mutational, biochemical and biophysical analyses indicate that this compound increases LYPLAL1's catalytic activity likely by enhancing the efficiency of the catalytic triad charge-relay system. Treatment with this LYPLAL1 activator confers beneficial effects in a mouse model of diet-induced obesity. These findings reveal a new mode of pharmacological regulation for this large enzyme family and suggest that ABPP may aid discovery of activators for additional enzyme classes.


Subject(s)
Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Lysophospholipase/metabolism , Small Molecule Libraries/pharmacology , Animals , Drug Discovery , Enzyme Activators/pharmacokinetics , Fluorescence Polarization , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Insulin Resistance , Lysophospholipase/chemistry , Lysophospholipase/genetics , Male , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Mice, Inbred C57BL , Mice, Obese , Molecular Dynamics Simulation , Molecular Structure , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics , Structure-Activity Relationship
7.
J Biol Chem ; 295(18): 5891-5905, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32152231

ABSTRACT

Fatty acid esters of hydroxy fatty acids (FAHFAs) are a newly discovered class of signaling lipids with anti-inflammatory and anti-diabetic properties. However, the endogenous regulation of FAHFAs remains a pressing but unanswered question. Here, using MS-based FAHFA hydrolysis assays, LC-MS-based lipidomics analyses, and activity-based protein profiling, we found that androgen-induced gene 1 (AIG1) and androgen-dependent TFPI-regulating protein (ADTRP), two threonine hydrolases, control FAHFA levels in vivo in both genetic and pharmacologic mouse models. Tissues from mice lacking ADTRP (Adtrp-KO), or both AIG1 and ADTRP (DKO) had higher concentrations of FAHFAs particularly isomers with the ester bond at the 9th carbon due to decreased FAHFA hydrolysis activity. The levels of other lipid classes were unaltered indicating that AIG1 and ADTRP specifically hydrolyze FAHFAs. Complementing these genetic studies, we also identified a dual AIG1/ADTRP inhibitor, ABD-110207, which is active in vivo Acute treatment of WT mice with ABD-110207 resulted in elevated FAHFA levels, further supporting the notion that AIG1 and ADTRP activity control endogenous FAHFA levels. However, loss of AIG1/ADTRP did not mimic the changes associated with pharmacologically administered FAHFAs on extent of upregulation of FAHFA levels, glucose tolerance, or insulin sensitivity in mice, indicating that therapeutic strategies should weigh more on FAHFA administration. Together, these findings identify AIG1 and ADTRP as the first endogenous FAHFA hydrolases identified and provide critical genetic and chemical tools for further characterization of these enzymes and endogenous FAHFAs to unravel their physiological functions and roles in health and disease.


Subject(s)
Esterases/metabolism , Esters/chemistry , Fatty Acids/chemistry , Fatty Acids/metabolism , Membrane Proteins/metabolism , Animals , Esterases/deficiency , Esterases/genetics , Gene Knockout Techniques , Hydrolysis , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice
8.
Nature ; 576(7785): 138-142, 2019 12.
Article in English | MEDLINE | ID: mdl-31748741

ABSTRACT

Haem is an essential prosthetic group of numerous proteins and a central signalling molecule in many physiologic processes1,2. The chemical reactivity of haem means that a network of intracellular chaperone proteins is required to avert the cytotoxic effects of free haem, but the constituents of such trafficking pathways are unknown3,4. Haem synthesis is completed in mitochondria, with ferrochelatase adding iron to protoporphyrin IX. How this vital but highly reactive metabolite is delivered from mitochondria to haemoproteins throughout the cell remains poorly defined3,4. Here we show that progesterone receptor membrane component 2 (PGRMC2) is required for delivery of labile, or signalling haem, to the nucleus. Deletion of PGMRC2 in brown fat, which has a high demand for haem, reduced labile haem in the nucleus and increased stability of the haem-responsive transcriptional repressors Rev-Erbα and BACH1. Ensuing alterations in gene expression caused severe mitochondrial defects that rendered adipose-specific PGRMC2-null mice unable to activate adaptive thermogenesis and prone to greater metabolic deterioration when fed a high-fat diet. By contrast, obese-diabetic mice treated with a small-molecule PGRMC2 activator showed substantial improvement of diabetic features. These studies uncover a role for PGRMC2 in intracellular haem transport, reveal the influence of adipose tissue haem dynamics on physiology and suggest that modulation of PGRMC2 may revert obesity-linked defects in adipocytes.


Subject(s)
Adipocytes/metabolism , Heme/metabolism , Membrane Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Homeostasis , Humans , Intracellular Space/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Molecular Chaperones/metabolism , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Transcription, Genetic
9.
Mol Metab ; 16: 76-87, 2018 10.
Article in English | MEDLINE | ID: mdl-30120064

ABSTRACT

OBJECTIVES: Extracts of the hops plant have been shown to reduce weight and insulin resistance in rodents and humans, but elucidation of the mechanisms responsible for these benefits has been hindered by the use of heterogeneous hops-derived mixtures. Because hop extracts are used as flavoring agents for their bitter properties, we hypothesized that bitter taste receptors (Tas2rs) could be mediating their beneficial effects in metabolic disease. Studies have shown that exposure of cultured enteroendocrine cells to bitter tastants can stimulate release of hormones, including glucagon-like peptide 1 (GLP-1). These findings have led to the suggestion that activation of Tas2rs may be of benefit in diabetes, but this tenet has not been tested. Here, we have assessed the ability of a pure derivative of a hops isohumulone with anti-diabetic properties, KDT501, to signal through Tas2rs. We have further used this compound as a tool to systematically assess the impact of bitter taste receptor activation in obesity-diabetes. METHODS: KDT501 was tested in a panel of bitter taste receptor signaling assays. Diet-induced obese mice (DIO) were dosed orally with KDT501 and acute effects on glucose homeostasis determined. A wide range of metabolic parameters were evaluated in DIO mice chronically treated with KDT501 to establish the full impact of activating gut bitter taste signaling. RESULTS: We show that KDT501 signals through Tas2r108, one of 35 mouse Tas2rs. In DIO mice, acute treatment stimulated GLP-1 secretion and enhanced glucose tolerance. Chronic treatment caused weight and fat mass loss, increased energy expenditure, enhanced glucose tolerance and insulin sensitivity, normalized plasma lipids, and induced broad suppression of inflammatory markers. Chronic KDT501 treatment altered enteroendocrine hormone levels and bile acid homeostasis and stimulated sustained GLP-1 release. Combined treatment with a dipeptidyl peptidase IV inhibitor amplified the incretin-based benefits of this pure isohumulone. CONCLUSIONS: Activation of Tas2r108 in the gut results in a remodeling of enteroendocrine hormone release and bile acid metabolism that ameliorates multiple features of metabolic syndrome. Targeting extraoral bitter taste receptors may be useful in metabolic disease.


Subject(s)
Cyclopentanes/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Body Weight/drug effects , Cyclopentanes/pharmacology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/drug effects , Glucagon-Like Peptide 1/metabolism , Humulus/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Intestinal Mucosa/metabolism , Intestines/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Receptors, G-Protein-Coupled/drug effects , Signal Transduction/drug effects
10.
Methods Mol Biol ; 1787: 115-127, 2018.
Article in English | MEDLINE | ID: mdl-29736714

ABSTRACT

Defects in adipocyte function associated with obesity drive the development of systemic insulin resistance and type 2 diabetes. Agents that correct obesity-linked adipocyte dysfunction serve as useful insulin sensitizers in humans, as is exemplified by the thiazolidinediones (TZDs). We have developed a new platform that integrates advanced chemoproteomics with phenotypic screening to identify small molecules that promote differentiation and lipid storage in adipocytes, and, in tandem, their molecular target(s). These molecules mimic the activity of TZDs in culture and thus may also serve as insulin sensitizers in vivo. Central to this platform is the use of fully functionalized fragment (FFF) probes that consist of a variable, fragment-like recognition element linked to an alkyne-diazirine group that enables the photoactivated capture of probe-bound proteins directly in living cells and subsequent copper-catalyzed azide-alkyne cycloaddition to reporter tags for enrichment and identification of these probe-bound proteins by mass spectrometry. This platform, which can be adapted to diverse screens and cell types beyond adipocytes, has the potential to uncover new biological pathways amenable to pharmacological modulation that may impact human disease.


Subject(s)
Adipocytes/drug effects , Adipocytes/physiology , Drug Discovery , 3T3-L1 Cells , Adipogenesis/drug effects , Animals , Drug Discovery/methods , Drug Evaluation, Preclinical , Genetic Variation , Humans , Mass Spectrometry , Mice , Phenotype , Proteome , Proteomics/methods , Small Molecule Libraries , Thiazolidinediones/pharmacology
11.
Nat Chem Biol ; 14(1): 22-28, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29131145

ABSTRACT

Endogenous metabolites play essential roles in the regulation of cellular identity and activity. Here we have investigated the process of oligodendrocyte precursor cell (OPC) differentiation, a process that becomes limiting during progressive stages of demyelinating diseases, including multiple sclerosis, using mass-spectrometry-based metabolomics. Levels of taurine, an aminosulfonic acid possessing pleotropic biological activities and broad tissue distribution properties, were found to be significantly elevated (∼20-fold) during the course of oligodendrocyte differentiation and maturation. When added exogenously at physiologically relevant concentrations, taurine was found to dramatically enhance the processes of drug-induced in vitro OPC differentiation and maturation. Mechanism of action studies suggest that the oligodendrocyte-differentiation-enhancing activities of taurine are driven primarily by its ability to directly increase available serine pools, which serve as the initial building block required for the synthesis of the glycosphingolipid components of myelin that define the functional oligodendrocyte cell state.


Subject(s)
Cell Differentiation/physiology , Metabolomics/methods , Oligodendrocyte Precursor Cells , Taurine/metabolism , Cell Differentiation/drug effects , Glycosphingolipids/biosynthesis , Metabolic Networks and Pathways , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/physiology , Serine/metabolism , Taurine/pharmacology
12.
Cell ; 171(3): 696-709.e23, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28965760

ABSTRACT

The transcription factor NRF2 is a master regulator of the cellular antioxidant response, and it is often genetically activated in non-small-cell lung cancers (NSCLCs) by, for instance, mutations in the negative regulator KEAP1. While direct pharmacological inhibition of NRF2 has proven challenging, its aberrant activation rewires biochemical networks in cancer cells that may create special vulnerabilities. Here, we use chemical proteomics to map druggable proteins that are selectively expressed in KEAP1-mutant NSCLC cells. Principal among these is NR0B1, an atypical orphan nuclear receptor that we show engages in a multimeric protein complex to regulate the transcriptional output of KEAP1-mutant NSCLC cells. We further identify small molecules that covalently target a conserved cysteine within the NR0B1 protein interaction domain, and we demonstrate that these compounds disrupt NR0B1 complexes and impair the anchorage-independent growth of KEAP1-mutant cancer cells. Our findings designate NR0B1 as a druggable transcriptional regulator that supports NRF2-dependent lung cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Proteome/analysis , Transcriptome , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cysteine/metabolism , DAX-1 Orphan Nuclear Receptor/metabolism , Gene Regulatory Networks , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Ligands , Lung Neoplasms/metabolism
13.
Article in English | MEDLINE | ID: mdl-29033896

ABSTRACT

OBJECTIVE: In a phase II clinical trial in nine obese, insulin-resistant humans, we observed that treatment with KDT501, a novel isohumulone drug, increased total and high-molecular weight (HMW) adiponectin in plasma. The objective was to determine whether KDT501 increased adiponectin secretion from subcutaneous white adipose tissue (SC WAT) and the underlying mechanism(s). METHODS: Nine obese participants with either prediabetes or with normal glucose tolerance plus three features of metabolic syndrome were part of the study. SC WAT biopsies were performed before and after 28 days of KDT501 treatment in a clinical research setting. In addition, a cold stimulus was used to induce thermogenic gene expression. Adiponectin secretion was measured, and gene expression of 130 genes involved in adipose tissue function was determined. The effect of KDT501 on adipocyte mitochondrial function was analyzed in vitro. RESULTS: SC WAT explants secreted more total and HMW adiponectin after KDT501 treatment (P < 0.05). After KDT501 treatment, a number of genes involved in thermogenesis and lipolysis were induced by cold (P < 0.05). KDT501 also potentiated ß-adrenergic signaling (P < 0.001) and enhanced mitochondrial function in adipocytes (P < 0.001). CONCLUSION: KDT501 induced adiponectin secretion posttranscriptionally and increased gene expression of thermogenic and lipolytic genes in response to cold stimulation. These beneficial effects on SC WAT may be explained by the ability of KDT501 to potentiate ß-adrenergic signaling and enhance mitochondrial function in adipocytes. CLINICAL TRIAL REGISTRATION: https://www.ClinicalTrials.gov, ID number: NCT02444910.

14.
Chem Biol ; 21(8): 917-8, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126986

ABSTRACT

Diterpene derivatives of the natural product acanthoic acid have potent anti-inflammatory effects in vivo. In this issue of Chemistry & Biolgy, Través and colleagues report that the primary molecular mechanism of action of diterpenes structurally related to acanthoic acid is the direct activation of PI3-kinase signaling in macrophages, which in turn inhibits NF-κB activation and suppresses proinflammatory gene expression.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Diterpenes/pharmacology , NF-kappa B/antagonists & inhibitors , Protein Subunits/metabolism , Animals
15.
FASEB J ; 28(6): 2655-66, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24599971

ABSTRACT

Autotaxin is a secreted enzyme that produces most extracellular lysophosphatidate, which stimulates 6 G-protein-coupled receptors. Lysophosphatidate promotes cancer cell survival, growth, migration, invasion, metastasis, and resistance to chemotherapy and radiotherapy. The present work investigated whether inhibiting autotaxin could decrease breast tumor growth and metastasis. We used a new autotaxin inhibitor (ONO-8430506; IC90=100 nM), which decreased plasma autotaxin activity by >60% and concentrations of unsaturated lysophosphatidates by >75% for 24 h compared with vehicle-treated mice. The effects of ONO-8430506 on tumor growth were determined in a syngeneic orthotopic mouse model of breast cancer following injection of 20,000 BALB/c mouse 4T1 or 4T1-12B cancer cells. We show for the first time that inhibiting autotaxin decreases initial tumor growth and subsequent lung metastatic nodules both by 60% compared with vehicle-treated mice. Significantly, 4T1 cells express negligible autotaxin compared with the mammary fat pad. Autotaxin activity in the fat pad of nontreated mice was increased 2-fold by tumor growth. Our results emphasize the importance of tumor interaction with its environment and the role of autotaxin in promoting breast cancer growth and metastasis. We also established that autotaxin inhibition could provide a novel therapeutic approach to blocking the adverse effects of lysophosphatidate in cancer.


Subject(s)
Lung Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/drug effects , Animals , Carbolines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Lung Neoplasms/secondary , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/pharmacology , Mammary Neoplasms, Experimental/pathology , Mice
16.
J Biol Chem ; 289(15): 10876-10886, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24558042

ABSTRACT

Lipin-1 is a phosphatidate phosphatase in glycerolipid biosynthesis and signal transduction. It also serves as a transcriptional co-regulator to control lipid metabolism and adipogenesis. These functions are controlled partly by its subcellular distribution. Hyperphosphorylated lipin-1 remains sequestered in the cytosol, whereas hypophosphorylated lipin-1 translocates to the endoplasmic reticulum and nucleus. The serine/threonine protein phosphatase-1 catalytic subunit (PP-1c) is a major protein dephosphorylation enzyme. Its activity is controlled by interactions with different regulatory proteins, many of which contain conserved RVXF binding motifs. We found that lipin-1 binds to PP-1cγ through a similar HVRF binding motif. This interaction depends on Mg(2+) or Mn(2+) and is competitively inhibited by (R/H)VXF-containing peptides. Mutating the HVRF motif in the highly conserved N terminus of lipin-1 greatly decreases PP-1cγ interaction. Moreover, mutations of other residues in the N terminus of lipin-1 also modulate PP-1cγ binding. PP-1cγ binds poorly to a phosphomimetic mutant of lipin-1 and binds well to the non-phosphorylatable lipin-1 mutant. This indicates that lipin-1 is dephosphorylated before PP-1cγ binds to its HVRF motif. Importantly, mutating the HVRF motif also abrogates the nuclear translocation and phosphatidate phosphatase activity of lipin-1. In conclusion, we provide novel evidence of the importance of the lipin-1 N-terminal domain for its catalytic activity, nuclear localization, and binding to PP-1cγ.


Subject(s)
Active Transport, Cell Nucleus , Lipid Metabolism , Phosphatidate Phosphatase/metabolism , Protein Phosphatase 1/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Conserved Sequence , Gene Expression Regulation , Genetic Vectors , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid
17.
J Lipid Res ; 54(6): 1662-1677, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23505321

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) and PPARγ coactivator 1α (PGC-1α) are crucial transcriptional regulators for genes involved in FA oxidation. Lipin-1 is essential for this increased capacity for ß-oxidation in fasted livers, and it is also a phosphatidate phosphatase involved in triacylglycerol and phospholipid synthesis. Little is known about the regulation of these proteins in the heart during fasting, where there is increased FA esterification and oxidation. Lipin-1, lipin-2, lipin-3, carnitine palmitoyltransferase-1b (Cpt1b), and PGC-1α-b mRNA were increased by glucocorticoids and cAMP in neonatal rat cardiomyocytes. However, Cpt1b upregulation was caused by increased PPARα activation rather than expression. By contrast, the effects of PPARα in fasted livers are mediated through increased expression. During fasting, the expressions of PGC-1α-b and PGC-1α-c are increased in mouse hearts, and this is explained by increased cAMP-dependent signaling. By contrast, PGC-1α-a expression is increased in liver. Contrary to our expectations, lipin-1 expression was decreased and lipin-2 remained unchanged in hearts compared with increases in fasted livers. Our results identify novel differences in the regulation of lipins, PPARα, and PGC-1α splice variants during fasting in heart versus liver, even though the ultimate outcome in both tissues is to increase FA turnover and oxidation.


Subject(s)
Alternative Splicing/physiology , Gene Expression Regulation/physiology , Liver/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , PPAR alpha/biosynthesis , Transcription Factors/biosynthesis , Animals , Fasting/metabolism , Fatty Acids/metabolism , Organ Specificity/physiology , Organic Chemicals/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Sprague-Dawley
18.
Heart Fail Clin ; 8(4): 643-61, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22999246

ABSTRACT

Management of diabetes and insulin resistance in the setting of cardiovascular disease has become an important issue in an increasingly obese society. Besides the development of hypertension and buildup of atherosclerotic plaques, the derangement of fatty acid and lipid metabolism in the heart plays an important role in promoting cardiac dysfunction and oxidative stress. This review discusses the mechanisms by which metabolic inflexibility in the use of fatty acids as the preferred cardiac substrate in diabetes produces detrimental effects on mechanical efficiency, mitochondrial function, and recovery from ischemia. Lipid accumulation and the consequences of toxic lipid metabolites are also discussed.


Subject(s)
Diabetic Cardiomyopathies/etiology , Fatty Acids/metabolism , Hyperglycemia/complications , Hyperlipidemias/complications , Insulin Resistance , Myocardium/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Endoplasmic Reticulum , Humans , Hyperglycemia/pathology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Mitochondria , Myocardium/pathology , Myocytes, Cardiac , Oxidative Stress , Risk Factors
20.
J Lipid Res ; 53(1): 105-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22058427

ABSTRACT

Lipin-1 is the major phosphatidate phosphatase (PAP) in the heart and a transcriptional coactivator that regulates fatty acid (FA) oxidation in the liver. As the control of FA metabolism is essential for maintaining cardiac function, we investigated whether lipin-1 deficiency affects cardiac metabolism and performance. Cardiac PAP activity in lipin-1 deficient [fatty liver dystrophy (fld)] mice was decreased by >80% compared with controls. Surprisingly, oleate oxidation and incorporation in triacylglycerol (TG), as well as glucose oxidation, were not significantly different in perfused working fld hearts. Despite this, [³H]oleate accumulation in phosphatidate and phosphatidylinositol was increased in fld hearts, reflecting the decreased PAP activity. Phosphatidate accumulation was linked to increased cardiac mammalian target of rapamycin complex 1 (mTORC1) signaling and endoplasmic reticulum (ER) stress. Transthoracic echocardiography showed decreased cardiac function in fld mice; however, cardiac dysfunction was not observed in ex vivo perfused working fld hearts. This showed that changes in systemic factors due to the global absence of lipin-1 could contribute to the decreased cardiac function in vivo. Collectively, this study shows that fld hearts exhibit unchanged oleate esterification, as well as oleate and glucose oxidation, despite the absence of lipin-1. However, lipin-1 deficiency increases the accumulation of newly synthesized phosphatidate and induces aberrant cell signaling.


Subject(s)
Glucose/metabolism , Heart/physiology , Nuclear Proteins/deficiency , Oleic Acid/metabolism , Phosphatidate Phosphatase/deficiency , Animals , Fatty Liver/physiopathology , In Vitro Techniques , Male , Mice , Myocardium/metabolism , Triglycerides/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...