Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Virol ; 95(13): e0004621, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33762416

ABSTRACT

Adenoviruses (AdVs) are etiological agents of gastrointestinal, heart, eye, and respiratory tract infections that can be lethal for immunosuppressed people. Many AdVs use the coxsackievirus and adenovirus receptor (CAR) as a primary receptor. The CAR isoform resulting from alternative splicing that includes the eighth exon, CAREx8, localizes to the apical surface of polarized epithelial cells and is responsible for the initiation of AdV infection. We have shown that the membrane level of CAREx8 is tightly regulated by two MAGI-1 PDZ domains, PDZ2 and PDZ4, resulting in increased or decreased AdV transduction, respectively. We hypothesized that targeting the interactions between the MAGI-1 PDZ2 domain and CAREx8 would decrease the apical CAREx8 expression level and prevent AdV infection. Decoy peptides that target MAGI-1 PDZ2 were synthesized (TAT-E6 and TAT-NET1). PDZ2 binding peptides decreased CAREx8 expression and reduced AdV transduction. CAREx8 degradation was triggered by the activation of the regulated intramembrane proteolysis (RIP) pathway through a disintegrin and metalloproteinase (ADAM17) and γ-secretase. Further analysis revealed that ADAM17 interacts directly with the MAGI-1 PDZ3 domain, and blocking the PDZ2 domain enhanced the accessibility of ADAM17 to the substrate (CAREx8). Finally, we validated the efficacy of TAT-PDZ2 peptides in protecting the epithelia from AdV transduction in vivo using a novel transgenic animal model. Our data suggest that TAT-PDZ2 binding peptides are novel anti-AdV molecules that act by enhanced RIP of CAREx8 and decreased AdV entry. This strategy has additional translational potential for targeting other viral receptors that have PDZ binding domains, such as the angiotensin-converting enzyme 2 receptor. IMPORTANCE Adenovirus is a common threat in immunosuppressed populations and military recruits. There are no currently approved treatments/prophylactic agents that protect from most AdV infections. Here, we developed peptide-based small molecules that can suppress AdV infection of polarized epithelia by targeting the AdV receptor, coxsackievirus and adenovirus receptor (CAREx8). The newly discovered peptides target a specific PDZ domain of the CAREx8-interacting protein MAGI-1 and decrease AdV transduction in multiple polarized epithelial models. Peptide-induced CAREx8 degradation is triggered by extracellular domain (ECD) shedding through ADAM17 followed by γ-secretase-mediated nuclear translocation of the C-terminal domain. The enhanced shedding of the CAREx8 ECD further protected the epithelium from AdV infection. Taken together, these novel molecules protect the epithelium from AdV infection. This approach may be applicable to the development of novel antiviral molecules against other viruses that use a receptor with a PDZ binding domain.


Subject(s)
ADAM17 Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adenoviridae Infections/prevention & control , Cell Adhesion Molecules/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/antagonists & inhibitors , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Guanylate Kinases/metabolism , 3T3 Cells , Adenoviridae/immunology , Amyloid Precursor Protein Secretases/metabolism , Animals , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Protein Domains
2.
Article in English | MEDLINE | ID: mdl-28989932

ABSTRACT

Liver X receptor alpha (LXRα) is crucial for the maintenance of lipid and cholesterol homeostasis. Ligand binding and dimerization with retinoid X receptor (RXR) or peroxisome proliferator-activated receptor (PPAR) is required for forming active DNA binding complexes leading to gene regulation. Structure based prediction and solvent accessibility of LXRα LBD shows that residues H383, E387, H390, L414, and R415 which are located in helices 9 and 10 may be critical for mediating protein-protein interactions. In this study, LXRα interface residues were individually mutated to determine their effects on ligand binding, protein-protein association, subcellular localization, and transactivation activity. LXRα L414R and R415A lacked binding to T-0901317, but retained binding to 25-Hydroxycholesterol. In vitro assay and a cell based assay demonstrated that LXRα L414R was specifically impaired for interactions with RXRα but not PPARα suggesting that charge reversal at the interface provides selectivity to LXRα dimerization. Furthermore, binding of LXRα L414R or R415A with PPARα exhibited minimal conformational changes in the dimer secondary structure. Interestingly, all LXRα mutants exhibited lower levels of ligand dependent luciferase activity driven by the SREBP-1c or ApoA1 promoter. Taken together, our data demonstrates that intact hydrophobic interactions and salt bridges at the interface mediate efficient ligand-dependent transactivation activities.

3.
J Lipid Res ; 58(2): 393-402, 2017 02.
Article in English | MEDLINE | ID: mdl-28011707

ABSTRACT

Liver X receptor (LXR)α is a nuclear receptor that responds to oxysterols and cholesterol overload by stimulating cholesterol efflux, transport, conversion to bile acids, and excretion. LXRα binds to and is regulated by synthetic (T-0901317, GW3695) and endogenous (oxysterols) ligands. LXRα activity is also modulated by FAs, but the ligand binding specificity of FA and acyl-CoA derivatives for LXRα remains unknown. We investigated whether LXRα binds FA or FA acyl-CoA with affinities that mimic in vivo concentrations, examined the effect of FA chain length and the degree of unsaturation on binding, and investigated whether FAs regulate LXRα activation. Saturated medium-chain FA (MCFA) displayed binding affinities in the low nanomolar concentration range, while long-chain fatty acyl-CoA did not bind or bound weakly to LXRα. Circular dichroic spectra and computational docking experiments confirmed that MCFA bound to the LXRα ligand binding pocket similar to the known synthetic agonist of LXRα (T0901317), but with limited change to the conformation of the receptor. Transactivation assays showed that MCFA activated LXRα, whereas long-chain FA caused no effect. Our results suggest that LXRα functions as a receptor for saturated FA or acyl-CoA of C10 and C12 in length.


Subject(s)
Acyl Coenzyme A/metabolism , Cholesterol/metabolism , Fatty Acids/metabolism , Liver X Receptors/metabolism , Acyl Coenzyme A/chemistry , Animals , COS Cells , Chlorocebus aethiops , Cholesterol/chemistry , Fatty Acids/chemistry , Humans , Hydrocarbons, Fluorinated/chemistry , Hydrocarbons, Fluorinated/metabolism , Ligands , Oxysterols/chemistry , Oxysterols/metabolism , Protein Binding , Sulfonamides/chemistry , Sulfonamides/metabolism
4.
Nanotechnology ; 27(14): 145603, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26916727

ABSTRACT

The effectiveness of nanoparticle-based functional devices depends strongly on the surface morphology and area of the support. An emerging powerful approach of increasing the available surface area without decreasing strength or increasing bulk is to attach arrays of suitable nanotubes on the surface, and to attach the necessary nanoparticles to them. Earlier publications by this team have shown that carpet-like arrays of carbon nanotubes (CNTs) can be successfully grown on a variety of larger carbon substrates such as graphite, foams and fabric, which offer hierarchical multiscale supporting architecture suitable for the attachment of silver nanoparticles (AgNPs). A limiting factor of pure CNT arrays in fluid-based applications is their hydrophobicity, which can reduce the percolation of an aqueous medium through individual nanotubes. Previous studies have demonstrated that the treatment of CNT carpets with dry (oxygen) plasma can induce reversible wettability, and treatment with wet (sol-gel) coating can impart permanent wettability. In this paper, we report the influence of such treatments on the attachment of AgNPs, and their effectiveness in water disinfection treatments. Both types of hydrophilic surface treatment show an increase in silver loading on the CNT carpets. Oxygen-plasma treated surfaces (O-CNT) show fine and densely packed AgNPs, whereas silica-coated nanotubes (silica-CNT) show uneven clusters of AgNPs. However, O-CNT surfaces lose their hydrophilicity during AgNP deposition, whereas silica-CNT surfaces remain hydrophilic. This difference significantly impacts the antibacterial effectiveness of these materials, as tested in simulated water containing Gram negative Escherichia coli (E. coli, JM109). AgNPs on silica-coated CNT substrates showed significantly higher reduction rates of E. coli compared to AgNPs on plasma-treated CNT substrates, despite the finer and better dispersed AgNP distribution in the latter. These results provide important insights into different aspects of surface modification approaches that can control the wettability of CNT carpets, and their applicability in water treatment applications.


Subject(s)
Anti-Bacterial Agents/chemistry , Escherichia coli/isolation & purification , Metal Nanoparticles/chemistry , Nanotubes, Carbon/chemistry , Silver/chemistry , Water Microbiology , Water Purification/methods , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/growth & development , Hydrophobic and Hydrophilic Interactions , Metal Nanoparticles/microbiology , Metal Nanoparticles/ultrastructure , Nanotechnology/methods , Nanotubes, Carbon/microbiology , Nanotubes, Carbon/ultrastructure , Oxygen/chemistry , Silicon Dioxide/chemistry , Silver/pharmacology , Surface Properties
5.
Sci Rep ; 5: 15379, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26493292

ABSTRACT

The expression of the epidermal growth factor receptor (EGFR) is highly regulated in normal cells, whereas some cancer cells have high constitutive levels. Understanding naturally-occurring ways of downregulating EGFR in cancer cells was investigated. Phosphatidic acid (PA) or Nuclear Receptors (NR) PPARα/RXRα/LXRα, enhance EGFR expression, mediated by the promoter region -856(A) to -226(T). Unexpectedly, the combination of NRs and PA caused repression. PA induces a conformational change in the nuclear receptor PPARα (increase of alpha-helices at the expense of decreasing beta-sheets), as evidenced by circular dichroism. This represses the naturally-enhancing capability of PPARα on EGFR transcription. PPARα-overexpressing cells in the presence of PA > 300 nM or the enzyme that produces it, phospholipase D (PLD), downregulate EGFR expression. The reasons are two-fold. First, PA displaces PPARα binding to the EGFR promoter at those concentrations. Second, NR heterodimer-dependent promoter activity is weakened in the presence of PA in vivo. Since other genes considered (ß-catenin, cyclin D3, PLD2 and ACOX-1) are also downregulated with a PA + PPARα combination, the transrepression appears to be a global phenomenon. Lastly, the reported effect is greater in MCF-7 than in MDA-MB-231 breast cancer cells, which could provide a novel basis for regulating excessive expression of EGFR in luminal cancer cells.


Subject(s)
ErbB Receptors/genetics , Orphan Nuclear Receptors/metabolism , PPAR alpha/metabolism , Phosphatidic Acids/pharmacology , Promoter Regions, Genetic , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Circular Dichroism , DNA , Female , Humans , Liver X Receptors , Molecular Sequence Data
6.
Int J Biochem Cell Biol ; 61: 29-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25622559

ABSTRACT

The Coxsackievirus and adenovirus receptor (CAR) is an essential cellular protein that is involved in cell-cell adhesion, protein trafficking, and viral infection. The major isoform of CAR is selectively sorted to the basolateral membrane of polarized epithelial cells where it co-localizes with the cellular scaffolding protein membrane-associated guanylate kinase with inverted domain structure-1 (MAGI-1). Previously, we demonstrated CAR interacts with MAGI-1 through a PDZ-domain dependent interaction. Here, we show that the PDZ3 domain of MAGI-1 is exclusively responsible for the high affinity interaction between the seven exon isoform of CAR and MAGI-1 using yeast-two-hybrid analysis and confirming this interaction biochemically and in cellular lysates by in vitro pull down assay and co-immunoprecipitation. The high affinity interaction between the PDZ3 domain and CAR C-terminus was measured by fluorescence resonance energy transfer. Further, we investigated the biological relevance of this high affinity interaction between CAR and the PDZ3 domain of MAGI-1 and found that it does not alter CAR-mediated adenovirus infection. By contrast, interruption of this high affinity interaction altered the localization of MAGI-1 indicating that CAR is able to traffic MAGI-1 to cell junctions. These data deepen the molecular understanding of the interaction between CAR and MAGI-1 and indicate that although CAR plays a role in trafficking PDZ-based scaffolding proteins to cellular junctions, association with a high affinity intracellular binding partner does not significantly alter adenovirus binding and entry via CAR.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Animals , CHO Cells , COS Cells , Cell Adhesion Molecules, Neuronal/genetics , Cell Membrane/metabolism , Chlorocebus aethiops , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Cricetulus , Models, Molecular , PDZ Domains , Transfection
7.
J Mol Graph Model ; 51: 27-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24858253

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) is an important regulator of hepatic lipid metabolism which functions through ligand binding. Despite high amino acid sequence identity (>90%), marked differences in PPARα ligand binding, activation and gene regulation have been noted across species. Similar to previous observations with synthetic agonists, we have recently reported differences in ligand affinities and extent of activation between human PPARα (hPPARα) and mouse PPARα (mPPARα) in response to long chain fatty acids (LCFA). The present study was aimed to determine if structural alterations could account for these differences. The binding of PPARα to LCFA was examined through in silico molecular modeling and docking simulations. Modeling suggested that variances at amino acid position 272 are likely to be responsible for differences in saturated LCFA binding to hPPARα and mPPARα. To confirm these results experimentally, LCFA binding, circular dichroism, and transactivation studies were performed using a F272I mutant form of mPPARα. Experimental data correlated with in silico docking simulations, further confirming the importance of amino acid 272 in LCFA binding. Although the driving force for evolution of species differences at this position are yet unidentified, this study enhances our understanding of ligand-induced regulation by PPARα and demonstrates the efficacy of molecular modeling and docking simulations.


Subject(s)
Fatty Acids/chemistry , PPAR alpha/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Fatty Acids/physiology , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Molecular Docking Simulation , Molecular Sequence Data , PPAR alpha/physiology , Phenylalanine/chemistry , Phenylalanine/genetics , Protein Binding , Protein Structure, Secondary , Retinoid X Receptor alpha/physiology , Sequence Homology, Amino Acid , Thermodynamics , Transcriptional Activation
8.
Biochemistry ; 53(16): 2632-43, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24713062

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) and liver X receptor α (LXRα) are members of the nuclear receptor superfamily that function to regulate lipid metabolism. Complex interactions between the LXRα and PPARα pathways exist, including competition for the same heterodimeric partner, retinoid X receptor α (RXRα). Although data have suggested that PPARα and LXRα may interact directly, the role of endogenous ligands in such interactions has not been investigated. Using in vitro protein-protein binding assays, circular dichroism, and co-immunoprecipitation of endogenous proteins, we established that full-length human PPARα and LXRα interact with high affinity, resulting in altered protein conformations. We demonstrated for the first time that the affinity of this interaction and the resulting conformational changes could be altered by endogenous PPARα ligands, namely long chain fatty acids (LCFA) or their coenzyme A thioesters. This heterodimer pair was capable of binding to PPARα and LXRα response elements (PPRE and LXRE, respectively), albeit with an affinity lower than that of the respective heterodimers formed with RXRα. LCFA had little effect on binding to the PPRE but suppressed binding to the LXRE. Ectopic expression of PPARα and LXRα in mammalian cells yielded an increased level of PPRE transactivation compared to overexpression of PPARα alone and was largely unaffected by LCFA. Overexpression of both receptors also resulted in transactivation from an LXRE, with decreased levels compared to that of LXRα overexpression alone, and LCFA suppressed transactivation from the LXRE. These data are consistent with the hypothesis that ligand binding regulates heterodimer choice and downstream gene regulation by these nuclear receptors.


Subject(s)
Orphan Nuclear Receptors/chemistry , Orphan Nuclear Receptors/metabolism , PPAR alpha/chemistry , PPAR alpha/metabolism , Circular Dichroism , Coenzyme A/chemistry , Coenzyme A/metabolism , Fatty Acids/metabolism , Hep G2 Cells , Humans , Immunoprecipitation , Ligands , Liver X Receptors , Orphan Nuclear Receptors/genetics , PPAR alpha/genetics , Palmitoyl Coenzyme A/chemistry , Palmitoyl Coenzyme A/metabolism , Protein Conformation , Protein Multimerization , Response Elements
9.
FEBS Lett ; 587(23): 3787-91, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24140341

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) regulates liver type fatty acid binding protein (L-FABP) gene expression. Conversely as shown herein, L-FABP structurally and functionally also interacts with HNF4α. Fluorescence resonance energy transfer (FRET) between Cy3-HNF4α (donor) and Cy5-L-FABP (acceptor) as well as FRET microscopy detected L-FABP in close proximity (~80 Å) to HNF4α, binding with high affinity Kd ~250-300 nM. Circular dichroism (CD) determined that the HNF4α/L-FABP interaction altered protein secondary structure. Finally, L-FABP potentiated transactivation of HNF4α in COS7 cells. Taken together, these data suggest that L-FABP provides a signaling path to HNF4α activation in the nucleus.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Fatty Acid-Binding Proteins/chemistry , Gene Expression Regulation , Hepatocyte Nuclear Factor 4/chemistry , Hepatocyte Nuclear Factor 4/genetics , Protein Binding , Protein Structure, Secondary , Rats
10.
J Lipid Res ; 54(9): 2354-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23797899

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) belongs to the family of ligand-dependent nuclear transcription factors that regulate energy metabolism. Although there exists remarkable overlap in the activities of PPARα across species, studies utilizing exogenous PPARα ligands suggest species differences in binding, activation, and physiological effects. While unsaturated long-chain fatty acids (LCFA) and their thioesters (long-chain fatty acyl-CoA; LCFA-CoA) function as ligands for recombinant mouse PPARα (mPPARα), no such studies have been conducted with full-length human PPARα (hPPARα). The objective of the current study was to determine whether LCFA and LCFA-CoA constitute high-affinity endogenous ligands for hPPARα or whether there exist species differences for ligand specificity and affinity. Both hPPARα and mPPARα bound with high affinity to LCFA-CoA; however, differences were noted in LCFA affinities. A fluorescent LCFA analog was bound strongly only by mPPARα, and naturally occurring saturated LCFA was bound more strongly by hPPARα than mPPARα. Similarly, unsaturated LCFA induced transactivation of both hPPARα and mPPARα, whereas saturated LCFA induced transactivation only in hPPARα-expressing cells. These data identified LCFA and LCFA-CoA as endogenous ligands of hPPARα, demonstrated species differences in binding specificity and activity, and may help delineate the role of PPARα as a nutrient sensor in metabolic regulation.


Subject(s)
PPAR alpha/metabolism , Amino Acids, Aromatic/chemistry , Animals , Boron Compounds/metabolism , COS Cells , Chlorocebus aethiops , Esters , Fatty Acids/chemistry , Fatty Acids/metabolism , Humans , Ligands , Mice , PPAR alpha/chemistry , PPAR alpha/genetics , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Retinoid X Receptor alpha/chemistry , Species Specificity , Substrate Specificity , Transcriptional Activation
11.
Chem Biol ; 19(7): 806-18, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22840769

ABSTRACT

Hepatocyte nuclear factor (HNF)4α is a central regulator of gene expression in cell types that play a critical role in metabolic homeostasis, including hepatocytes, enterocytes, and pancreatic ß cells. Although fatty acids were found to occupy the HNF4α ligand-binding pocket and were proposed to act as ligands, there is controversy about both the nature of HNF4α ligands as well as the physiological role of the binding. Here, we report the discovery of potent synthetic HNF4α antagonists through a high-throughput screen for effectors of the human insulin promoter. These molecules bound to HNF4α with high affinity and modulated the expression of known HNF4α target genes. Notably, they were found to be selectively cytotoxic to cancer cell lines in vitro and in vivo, although in vivo potency was limited by suboptimal pharmacokinetic properties. The discovery of bioactive modulators for HNF4α raises the possibility that diseases involving HNF4α, such as diabetes and cancer, might be amenable to pharmacologic intervention by modulation of HNF4α activity.


Subject(s)
Benzimidazoles/pharmacology , Drug Discovery , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , High-Throughput Screening Assays , Insulin/genetics , Promoter Regions, Genetic/genetics , Sulfonamides/pharmacology , Benzimidazoles/chemistry , Dose-Response Relationship, Drug , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Models, Molecular , Molecular Structure , PPAR gamma/agonists , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Structure-Activity Relationship , Sulfonamides/chemistry
12.
J Amino Acids ; 2012: 575180, 2012.
Article in English | MEDLINE | ID: mdl-22500212

ABSTRACT

Rotavirus (RV) NSP4, the first described viral enterotoxin, is a multifunctional glycoprotein that contributes to viral pathogenesis, morphogenesis, and replication. NSP4 binds both termini of caveolin-1 and is isolated from caveolae fractions that are rich in anionic phospholipids and cholesterol. These interactions indicate that cholesterol/caveolin-1 plays a role in NSP4 transport to the cell surface, which is essential to its enterotoxic activity. Synthetic peptides were utilized to identify target(s) of intervention by exploring the NSP4-caveolin-1 and -cholesterol interactions. NSP4(112-140) that overlaps the caveolin-1 binding domain and a cholesterol recognition amino acid consensus (CRAC) motif and both termini of caveolin-1 (N-caveolin-1(2-20), (19-40) and C-caveolin-1(161-180)) were synthesized. Direct fluorescence-binding assays were employed to determine binding affinities of the NSP4-caveolin-1 peptides and cholesterol. Intracellular cholesterol alteration revealed a redistribution of NSP4 and disintegration of viroplasms. These data further imply interruption of NSP4(112-140)-N-caveolin-1(19-40) and cholesterol interactions may block NSP4 intracellular transport, hence enterotoxicity.

13.
Mol Cell Biochem ; 355(1-2): 135-48, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21541677

ABSTRACT

Although the rate limiting step in mitochondrial fatty acid oxidation, catalyzed by carnitine palmitoyl transferase I (CPTI), utilizes long-chain fatty acyl-CoAs (LCFA-CoA) as a substrate, how LCFA-CoA is transferred to CPTI remains elusive. Based on secondary structural predictions and conserved tryptophan residues, the cytoplasmic C-terminal domain was hypothesized to be the LCFA-CoA binding site and important for interaction with cytoplasmic LCFA-CoA binding/transport proteins to provide a potential route for LCFA-CoA transfer. To begin to address this question, the cytoplasmic C-terminal region of liver CPTI (L-CPTI) was recombinantly expressed and purified. Data herein showed for the first time that the L-CPTI C-terminal 89 residues were sufficient for high affinity binding of LCFA-CoA (K (d) = 2-10 nM) and direct interaction with several cytoplasmic LCFA-CoA binding proteins (K (d) < 10 nM), leading to enhanced CPTI activity. Furthermore, alanine substitutions for tryptophan in L-CPTI (W391A and W452A) altered secondary structure, decreased binding affinity for LCFA-CoA, and almost completely abolished L-CPTI activity, suggesting that these amino acids may be important for ligand stabilization necessary for L-CPTI activity. Moreover, while decreased activity of the W452A mutant could be explained by decreased binding of lipid binding proteins, W391 itself seems to be important for activity. These data suggest that both interactions with lipid binding proteins and the peptide itself are important for optimal enzyme activity.


Subject(s)
Acyl Coenzyme A/chemistry , Carnitine O-Palmitoyltransferase/chemistry , Mitochondria/enzymology , Peptide Fragments/chemistry , Recombinant Proteins/chemistry , Animals , Binding Sites , Binding, Competitive , Circular Dichroism , Enzyme Assays , Fluorescence Resonance Energy Transfer , Humans , Mice , Protein Binding , Protein Structure, Secondary , Rats , Spectrometry, Fluorescence
14.
J Lipid Res ; 51(11): 3103-16, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20628144

ABSTRACT

Although the pathophysiology of diabetes is characterized by elevated levels of glucose and long-chain fatty acids (LCFA), nuclear mechanisms linking glucose and LCFA metabolism are poorly understood. As the liver fatty acid binding protein (L-FABP) shuttles LCFA to the nucleus, where L-FABP directly interacts with peroxisome proliferator-activated receptor-α (PPARα), the effect of glucose on these processes was examined. In vitro studies showed that L-FABP strongly bound glucose and glucose-1-phosphate (K(d) = 103 ± 19 nM and K(d) = 20 ± 3 nM, respectively), resulting in altered L-FABP conformation, increased affinity for lipid ligands, and enhanced interaction with PPARα. In living cells, glucose stimulated cellular uptake and nuclear localization of a nonmetabolizable fluorescent fatty acid analog (BODIPY C-16), particularly in the presence of L-FABP. These data suggest for the first time a direct role of glucose in facilitating L-FABP-mediated uptake and distribution of lipidic ligands to the nucleus for regulation of PPARα transcriptional activity.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Glucose/pharmacology , Lipid Metabolism/drug effects , PPAR alpha/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Fatty Acid-Binding Proteins/chemistry , Fatty Acids/chemistry , Fatty Acids/metabolism , Glucose/metabolism , Mice , PPAR alpha/chemistry , Protein Binding/drug effects , Protein Structure, Secondary/drug effects , Rats
15.
J Nutr Biochem ; 21(11): 1015-32, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20537520

ABSTRACT

While low levels of unesterified long chain fatty acids (LCFAs) are normal metabolic intermediates of dietary and endogenous fat, LCFAs are also potent regulators of key receptors/enzymes and at high levels become toxic detergents within the cell. Elevated levels of LCFAs are associated with diabetes, obesity and metabolic syndrome. Consequently, mammals evolved fatty acid-binding proteins (FABPs) that bind/sequester these potentially toxic free fatty acids in the cytosol and present them for rapid removal in oxidative (mitochondria, peroxisomes) or storage (endoplasmic reticulum, lipid droplets) organelles. Mammals have a large (15-member) family of FABPs with multiple members occurring within a single cell type. The first described FABP, liver-FABP (L-FABP or FABP1), is expressed in very high levels (2-5% of cytosolic protein) in liver as well as in intestine and kidney. Since L-FABP facilitates uptake and metabolism of LCFAs in vitro and in cultured cells, it was expected that abnormal function or loss of L-FABP would reduce hepatic LCFA uptake/oxidation and thereby increase LCFAs available for oxidation in muscle and/or storage in adipose. This prediction was confirmed in vitro with isolated liver slices and cultured primary hepatocytes from L-FABP gene-ablated mice. Despite unaltered food consumption when fed a control diet ad libitum, the L-FABP null mice exhibited age- and sex-dependent weight gain and increased fat tissue mass. The obese phenotype was exacerbated in L-FABP null mice pair fed a high-fat diet. Taken together with other findings, these data suggest that L-FABP could have an important role in preventing age- or diet-induced obesity.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Lipid Metabolism/genetics , Obesity/metabolism , Animals , Cells, Cultured , Diet , Fatty Acid-Binding Proteins/genetics , Female , Hepatocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Models, Biological , Obesity/genetics , Peroxisomes/metabolism , Rats , Weight Gain
16.
J Lipid Res ; 50(8): 1663-75, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19289416

ABSTRACT

Although studies with liver type fatty acid binding protein (L-FABP) gene ablated mice demonstrate a physiological role for L-FABP in hepatic fatty acid metabolism, little is known about the mechanisms whereby L-FABP elicits these effects. Studies indicate that L-FABP may function to shuttle lipids to the nucleus, thereby increasing the availability of ligands of nuclear receptors, such as peroxisome proliferator-activated receptor-alpha (PPARalpha). The data herein suggest that such mechanisms involve direct interaction of L-FABP with PPARalpha. L-FABP was shown to directly interact with PPARalpha in vitro through co-immunoprecipitation (co-IP) of pure proteins, altered circular dichroic (CD) spectra, and altered fluorescence spectra. In vitro fluorescence resonance energy transfer (FRET) between Cy3-labeled PPARalpha and Cy5-labeled L-FABP proteins showed that these proteins bound with high affinity (Kd approximately 156 nM) and in close proximity (intermolecular distance of 52A). This interaction was further substantiated by co-IP of both proteins from liver homogenates of wild-type mice. Moreover, double immunogold electron microscopy and FRET confocal microscopy of cultured primary hepatocytes showed that L-FABP was in close proximity to PPARalpha (intermolecular distance 40-49A) in vivo. Taken together, these studies were consistent with L-FABP regulating PPARalpha transcriptional activity in hepatocytes through direct interaction with PPARalpha. Our in vitro and imaging experiments demonstrate high affinity, structural molecular interaction of L-FABP with PPARalpha and suggest a functional role for L-FABP interaction with PPARalpha in long chain fatty acid (LCFA) metabolism.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Hepatocytes/metabolism , PPAR alpha/metabolism , Animals , Cell Compartmentation , Cell Nucleus/metabolism , Cells, Cultured , Fatty Acid-Binding Proteins/chemistry , Fatty Acids/metabolism , Hepatocytes/ultrastructure , Ligands , Male , Mice , Mice, Knockout , PPAR alpha/chemistry , Palmitic Acid/metabolism , Protein Binding , Protein Conformation , Protein Structure, Quaternary , Recombinant Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
17.
Arch Biochem Biophys ; 485(2): 160-73, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19285478

ABSTRACT

The effect of liver type fatty acid binding protein (L-FABP) gene ablation on the uptake and distribution of long chain fatty acids (LCFA) to the nucleus by real-time laser scanning confocal imaging and peroxisome proliferator-activated receptor-alpha (PPARalpha) activity was examined in cultured primary hepatocytes from livers wild-type L-FABP+/+ and gene ablated L-FABP-/- mice. Cultured primary hepatocytes from livers of L-FABP-/- mice exhibited: (i) reduced oxidation of palmitic acid, a common dietary long chain fatty acid (LCFA); (ii) reduced expression of fatty acid oxidative enzymes-proteins transcriptionally regulated by PPARalpha; (iii) reduced palmitic acid-induced PPARalpha co-immunoprecipitation with coactivator SRC-1 concomitant with increased PPARalpha co-immunoprecipitation with coinhibitor N-CoR; (iv) reduced palmitic acid-induced PPARalpha. Diminished PPARalpha activation in L-FABP null hepatocytes was associated with lower uptake of common dietary LCFA (palmitic acid as well as its fluorescent derivative BODIPY FL C(16)), reduced level of total unesterified LCFA, and real-time redistribution of BODIPY FL C(16) from the central nucleoplasm to the nuclear envelope. Taken together, these studies support the hypothesis that L-FABP may facilitate ligand (LCFA)-activated PPARalpha transcriptional activity at least in part by increasing total LCFA ligand available to PPARalpha for inducing PPARalpha-mediated transcription of proteins involved in LCFA metabolism.


Subject(s)
Fatty Acid-Binding Proteins/physiology , Hepatocytes/metabolism , PPAR alpha/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Fatty Acid-Binding Proteins/genetics , Immunoprecipitation , Ligands , Male , Mice , Mice, Knockout , Microscopy, Confocal
18.
J Biomol Screen ; 13(9): 855-61, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18812576

ABSTRACT

As natural peroxisome proliferator-activated receptor-alpha (PPARalpha) ligands, high levels of fatty acids and glucose could lead to hyperactivation of PPARalpha, like that seen in diabetes. Important diabetes research goals are to uncover new metabolic or signaling pathways involved in hyperglycemic cellular injury and to develop therapeutics for preventing or reversing this injury. Consequently, 1040 putative antidiabetic agents were screened for their ability to 1) affect PPARalpha lipid binding, 2) directly bind PPARalpha, and 3) alter PPARalpha transactivation in the presence of high glucose. A high-throughput fluorescent binding assay was developed to examine each compound's ability to restore fatty acyl-CoA binding to PPARalpha in the presence of high glucose concentrations. Approximately 1% of the compounds restored acyl-CoA binding by 60% or more. These compounds directly interacted with PPARalpha with high affinity (nM K(d)s), validating the primary screen. Furthermore, these compounds altered PPARalpha transactivation, and 1 strongly reversed the hyperactivation of PPARalpha found in the presence of clofibrate and high glucose levels.


Subject(s)
Drug Evaluation, Preclinical/instrumentation , Hypoglycemic Agents/pharmacology , PPAR alpha/metabolism , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Circular Dichroism , Drug Evaluation, Preclinical/methods , Fluorescent Dyes/pharmacology , Glucose/chemistry , Insulin Resistance , Kinetics , Ligands , Protein Binding , Transcriptional Activation
19.
Biochemistry ; 47(22): 5915-34, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18465878

ABSTRACT

Although sterol carrier protein-2 (SCP-2) is encoded as a precursor protein (proSCP-2), little is known regarding the structure and function of the 20-amino acid N-terminal presequence. As shown herein, the presequence contains significant secondary structure and alters SCP-2: (i) secondary structure (CD), (ii) tertiary structure (aqueous exposure of Trp shown by UV absorbance, fluorescence, and fluorescence quenching), (iii) ligand binding site [Trp response to ligands, peptide cross-linked by photoactivatable free cholesterol (FCBP)], (iv) selectivity for interaction with anionic phospholipid-rich membranes, (v) interaction with a peroxisomal import protein [FRET studies of Pex5p(C) binding], the N-terminal presequence increased SCP-2's affinity for Pex5p(C) by 10-fold, and (vi) intracellular targeting in living and fixed cells (confocal microscopy). Nearly 5-fold more SCP-2 than proSCP-2 colocalized with plasma membrane lipid rafts and caveolae (AF488-CTB); 2.8-fold more SCP-2 than proSCP-2 colocalized with a mitochondrial marker (Mitotracker), but nearly 2-fold less SCP-2 than proSCP-2 colocalized with peroxisomes (AF488 antibody to PMP70). These data indicate the importance of the N-terminal presequence in regulating SCP-2 structure, cholesterol localization within the ligand binding site, membrane association, and, potentially, intracellular targeting.


Subject(s)
Carrier Proteins/chemistry , Protein Precursors/chemistry , Binding Sites , Carrier Proteins/metabolism , Fluorescence Resonance Energy Transfer , Humans , Ligands , Protein Precursors/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Structure-Activity Relationship , Tryptophan/chemistry , Tryptophan/metabolism
20.
Protein Expr Purif ; 58(2): 184-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18178100

ABSTRACT

Acyl coenzyme A binding protein (ACBP) has been proposed to transport fatty acyl CoAs intracellularly, facilitating their metabolism. In this study, a new mouse recombinant ACBP was produced by insertion of a histidine (his) tag at the C-terminus to allow efficient purification by Ni-affinity chromatography. The his-tag was inserted at the C-terminus since ACBP is a small molecular size (10 kDa) protein whose structure and activity are sensitive to amino acid substitutions in the N-terminus. The his-tag had no or little effect on ACBP structure or ligand binding affinity and specificity. His-ACBP bound the naturally occurring fluorescent cis-parinaroyl-CoA with very high affinity (K(d)=2.15 nM), but exhibited no affinity for non-esterified cis-parinaric acid. To determine if the presence of the C-terminal his-tag altered ACBP interactions with other proteins, direct binding to hepatocyte nuclear factor-4alpha (HNF-4alpha), a nuclear receptor regulating transcription of genes involved in lipid metabolism, was examined. His-ACBP and HNF-4alpha were labeled with Cy5 and Cy3, respectively, and direct interaction was determined by a novel fluorescence resonance energy transfer (FRET) binding assay. FRET analysis showed that his-ACBP directly interacted with HNF-4alpha (intermolecular distance of 73 A) at high affinity (K(d)=64-111 nM) similar to native ACBP. The his-tag also had no effect on ACBPs ability to interact with and stimulate microsomal enzymes utilizing or forming fatty acyl CoA. Thus, C-terminal his-tagged-ACBP maintained very similar structural and functional features of the untagged native protein and can be used in further in vitro experiments that require pure recombinant ACBP.


Subject(s)
Diazepam Binding Inhibitor/chemistry , Diazepam Binding Inhibitor/metabolism , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Histidine/chemistry , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Carbocyanines/chemistry , Circular Dichroism , Cloning, Molecular , Coenzyme A Ligases/metabolism , Diazepam Binding Inhibitor/isolation & purification , Escherichia coli/metabolism , Fatty Acids, Unsaturated/metabolism , Fluorescence Resonance Energy Transfer , Hepatocyte Nuclear Factor 4/metabolism , Mice , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL