Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David B. Lloyd is active.

Publication


Featured researches published by David B. Lloyd.


Gene | 1991

Modulation of firefly luciferase stability and impact on studies of gene regulation

John F. Thompson; Lisa S. Hayes; David B. Lloyd

Two of the reporter enzymes most commonly used in studies of eukaryotic gene expression are chloramphenicol acetyl-transferase (CAT) and firefly luciferase (Luc). CAT has a half-life of about 50 h in mammalian cells, making it useful for transient transfection assays but less suitable for assays with stable cell lines. Luc has a half-life of only 3 h in mammalian cells, making it much more responsive in stable cell lines. Luc instability arises from its sensitivity to proteolysis both in vivo and in vitro. Compounds that resemble its natural substrate, luciferin, act as effective competitive inhibitors in vitro. When these compounds (e.g., phenylbenzothiazole) are added to either prokaryotic or eukaryotic cells, more than tenfold increases in Luc activity can be observed. This increased activity results from a lower rate of degradation of the enzyme in vivo and can be mimicked in vitro as phenylbenzothiazole protects Luc from trypsin digestion while it has no effect on the rate of digestion of alkaline phosphatase.


Nature Structural & Molecular Biology | 2007

Crystal structure of cholesteryl ester transfer protein reveals a long tunnel and four bound lipid molecules

Xiayang Qiu; Anil Mistry; Mark Ammirati; Boris A. Chrunyk; Ronald W. Clark; Yang Cong; Jeffrey S. Culp; Dennis E. Danley; Thomas B. Freeman; Kieran F. Geoghegan; Matthew C. Griffor; Steven J. Hawrylik; Cheryl Myers Hayward; Preston Hensley; Lise R. Hoth; George A. Karam; Maruja E. Lira; David B. Lloyd; Katherine M McGrath; Kim Jonelle Stutzman-Engwall; Ann Subashi; Timothy A. Subashi; John F. Thompson; Ing-Kae Wang; Honglei Zhao; Andrew P. Seddon

Cholesteryl ester transfer protein (CETP) shuttles various lipids between lipoproteins, resulting in the net transfer of cholesteryl esters from atheroprotective, high-density lipoproteins (HDL) to atherogenic, lower-density species. Inhibition of CETP raises HDL cholesterol and may potentially be used to treat cardiovascular disease. Here we describe the structure of CETP at 2.2-Å resolution, revealing a 60-Å-long tunnel filled with two hydrophobic cholesteryl esters and plugged by an amphiphilic phosphatidylcholine at each end. The two tunnel openings are large enough to allow lipid access, which is aided by a flexible helix and possibly also by a mobile flap. The curvature of the concave surface of CETP matches the radius of curvature of HDL particles, and potential conformational changes may occur to accommodate larger lipoprotein particles. Point mutations blocking the middle of the tunnel abolish lipid-transfer activities, suggesting that neutral lipids pass through this continuous tunnel.


Pharmacogenomics Journal | 2005

An association study of 43 SNPs in 16 candidate genes with atorvastatin response

John F. Thompson; M. Man; Keith Johnson; Linda S. Wood; Maruja E. Lira; David B. Lloyd; Poulabi Banerjee; Patrice M. Milos; S. P. Myrand; J. Paulauskis; M. A. Milad; W. J. Sasiela

Variation in individual response to statin therapy has been widely studied for a potential genetic component. Multiple genes have been identified as potential modulators of statin response, but few study findings have replicated. To further examine these associations, 2735 individuals on statin therapy, half on atorvastatin and the other half divided among fluvastatin, lovastatin, pravastatin and simvastatin were genotyped for 43 SNPs in 16 genes that have been implicated in statin response. Associations with low-density lipoprotein cholesterol (LDL-C) lowering, total cholesterol lowering, HDL-C elevation and triglyceride lowering were examined. The only significant associations with LDL-C lowering were found with apoE2 in which carriers of the rare allele who took atorvastatin lowered their LDL-C by 3.5% more than those homozygous for the common allele and with rs2032582 (S893A in ABCB1) in which the two groups of homozygotes differed by 3% in LDL-C lowering. These genetic effects were smaller than those observed with the demographic variables of age and gender. The magnitude of all the differences found is sufficiently small that genetic data from these genes should not influence clinical decisions on statin administration.


British Journal of Pharmacology | 2005

Pentamidine reduces hERG expression to prolong the QT interval

Jason S. Cordes; Zhuoqian Sun; David B. Lloyd; Jenifer Bradley; Alan Opsahl; Mark W. Tengowski; Xian Chen; Jun Zhou

1 Pentamidine, an antiprotozoal agent, has been traditionally known to cause QT prolongation and arrhythmias; however, its ionic mechanism has not been illustrated. 2 In a stable HEK‐293 cell line, we observed a concentration‐dependent inhibition of the hERG current with an IC50 of 252 μM. 3 In freshly isolated guinea‐pig ventricular myocytes, pentamidine showed no effect on the L‐type calcium current at concentrations up to 300 μM, with a slight prolongation of the action potential duration at this concentration. 4 Since the effective concentrations of pentamidine on the hERG channel and APD were much higher than clinically relevant exposures (∼1 μM free or lower), we speculated that this drug might not prolong the QT interval through direct inhibition of IKr channel. We therefore incubated hERG‐HEK cells in 1 and 10 μM pentamidine‐containing media (supplemented with 10% serum) for 48 h, and examined the hERG current densities in the vehicle control and pentamidine‐treated cells. 5 In all, 36 and 85% reductions of the current densities were caused by 1‐ and 10‐μM pentamidine treatment (P<0.001 vs control), respectively. A similar level of reduction of the hERG polypeptides and a reduced intensity of the hERG protein on the surface membrane in treated cells were observed by Western blot analysis and laser‐scanning confocal microscopy, respectively. 6 Taken together, our data imply that chronic administration of pentamidine at clinically relevant exposure reduces the membrane expression of the hERG channel, which may most likely be the major mechanism of QT prolongation and torsade de pointes reported in man.


Journal of Biological Chemistry | 1997

Mutation of a Protease-sensitive Region in Firefly Luciferase Alters Light Emission Properties

John F. Thompson; Kieran F. Geoghegan; David B. Lloyd; Anthony J. Lanzetti; Rachelle A. Magyar; Shannon M. Anderson; Bruce R. Branchini

Luciferase (EC 1.13.12.7) from the North American firefly, Photinus pyralis, is widely used as a reporter enzyme in cell biology. One of its distinctive properties is a pronounced susceptibility to proteolytic degradation that causes luciferase to have a very short intracellular half-life. To define the structural basis for this behavior and possibly facilitate the design of more stable forms of luciferase, limited proteolysis studies were undertaken using trypsin and chymotrypsin to identify regions of the protein whose accessible and flexible character rendered them especially sensitive to cleavage. Regions of amino acid sequence 206–220 and 329–341 were found to be sensitive, and because the region around 206–220 had high homology with other luciferases, CoA ligases, and peptidyl synthetases, this region was selected for mutagenesis experiments intended to determine which of its amino acids were essential for activity. Surprisingly, many highly conserved residues including Ser198, Ser201, Thr202, and Gly203 could be mutated with little effect on the luminescent activity of P. pyralisluciferase. One mutation, however, S198T, caused several alterations in enzymatic properties including shifting the pH optimum from 8.1 to 8.7, lowering the K m for Mg-ATP by a factor of 4 and increasing the half-time for light emission decay by a factor of up to 150. While the S198T luciferase was less active than wild type, activity could be restored by the introduction of the additional L194F and N197Y mutations. In addition to indicating the involvement of this region in ATP binding, these results provide a new form of the enzyme that affords a more versatile reporter system.


Journal of Biological Chemistry | 2012

Crystal Structures of Cholesteryl Ester Transfer Protein in Complex with Inhibitors

Shenping Liu; Anil Mistry; Jennifer M. Reynolds; David B. Lloyd; Matthew C. Griffor; David Austen Perry; Roger Benjamin Ruggeri; Ronald W. Clark; Xiayang Qiu

Background: Human cholesteryl ester transfer protein (CETP) transfers cholesteryl esters from high-density to low-density lipoprotein particles. Results: Crystallographic, mutagenesis, and biochemical studies illuminated inhibition mechanisms of CETP by torcetrapib and a structurally distinct compound, ((2R)-3-{[4-(4-chloro-3-ethylphenoxy)pyrimidin-2-yl][3-(1,1,2,2-tetrafluoroethoxy)benzyl]amino}-1,1,1-trifluoropropan-2-ol. Conclusion: These small molecules inhibit CETP through blocking its lipid tunnel. Significance: Potential polar interactions at compound binding site may be utilized in design of inhibitors with improved physical properties. Human plasma cholesteryl ester transfer protein (CETP) transports cholesteryl ester from the antiatherogenic high-density lipoproteins (HDL) to the proatherogenic low-density and very low-density lipoproteins (LDL and VLDL). Inhibition of CETP has been shown to raise human plasma HDL cholesterol (HDL-C) levels and is potentially a novel approach for the prevention of cardiovascular diseases. Here, we report the crystal structures of CETP in complex with torcetrapib, a CETP inhibitor that has been tested in phase 3 clinical trials, and compound 2, an analog from a structurally distinct inhibitor series. In both crystal structures, the inhibitors are buried deeply within the protein, shifting the bound cholesteryl ester in the N-terminal pocket of the long hydrophobic tunnel and displacing the phospholipid from that pocket. The lipids in the C-terminal pocket of the hydrophobic tunnel remain unchanged. The inhibitors are positioned near the narrowing neck of the hydrophobic tunnel of CETP and thus block the connection between the N- and C-terminal pockets. These structures illuminate the unusual inhibition mechanism of these compounds and support the tunnel mechanism for neutral lipid transfer by CETP. These highly lipophilic inhibitors bind mainly through extensive hydrophobic interactions with the protein and the shifted cholesteryl ester molecule. However, polar residues, such as Ser-230 and His-232, are also found in the inhibitor binding site. An enhanced understanding of the inhibitor binding site may provide opportunities to design novel CETP inhibitors possessing more drug-like physical properties, distinct modes of action, or alternative pharmacological profiles.


Journal of Biological Chemistry | 2005

Cholesteryl ester transfer protein variants have differential stability but uniform inhibition by torcetrapib

David B. Lloyd; Maruja E. Lira; Linda S. Wood; L. Kathryn Durham; Thomas B. Freeman; Gregory M. Preston; Xiayang Qiu; Eliot Sugarman; Peter C. Bonnette; Anthony J. Lanzetti; Patrice M. Milos; John F. Thompson

Cholesteryl ester transfer protein (CETP) is an important modulator of high density lipoprotein cholesterol in humans and thus considered to be a therapeutic target for preventing cardiovascular disease. The gene encoding CETP has been shown to be highly variable, with multiple single nucleotide polymorphisms responsible for altering both its transcription and sequence. Examining nine missense variants of CETP, we found some had significant associations with CETP mass and high density lipoprotein cholesterol levels. Two variants, Pro-373 and Gln-451, appear to be more stable in vivo, an observation mirrored by partial proteolysis studies performed in vitro. Because these naturally occurring variant proteins are potentially present in clinical populations that will be treated with CETP inhibitors, all commonly occurring haplotypes were tested to determine whether the proteins they encode could be inhibited by torcetrapib, a compound currently in clinical trials in combination with atorvastatin. Torcetrapib behaved similarly with all variants, with no significant differences in inhibition.


Clinical Genetics | 2004

Cholesteryl ester transfer protein promoter single-nucleotide polymorphisms in Sp1-binding sites affect transcription and are associated with high-density lipoprotein cholesterol

John F. Thompson; David B. Lloyd; Maruja E. Lira; Patrice M. Milos

Genetic variation in the human cholesteryl ester transfer protein (CETP) promoter has been shown to be associated with high‐density lipoprotein cholesterol (HDL‐C) levels and cardiovascular disease. Some of this variation occurs in Sp1/Sp3 binding sites in the proximal promoter. We find that both the known promoter polymorphism at −629 and the previously uncharacterized polymorphism at −38 are associated with HDL‐C levels in vivo and affect transcription in vitro. While the −629 polymorphism is common in all ethnic groups, the −38 polymorphism is found at significant levels (6.4%) only among African Americans. Those homozygous for the less common −38A allele have higher HDL‐C levels than those with the more frequent −38G allele. This association was found in a population of African Americans at risk of cardiovascular disease and then replicated in a different population chosen from among patients with extremes of HDL‐C. When studied in vitro, the most transcriptionally active allele (−629C/−38G) yields 51% more reporter protein than the least active allele (−629A/−38A) in HepG2 cells. These transcriptional effects reflect the projected impact of increased CETP expression on HDL‐C phenotypes seen in vivo.


Journal of Biological Chemistry | 1995

Transcriptional modulators affect in vivo protein binding to the low density lipoprotein receptor and 3-hydroxy-3-methylglutaryl coenzyme A reductase promoters.

David B. Lloyd; John F. Thompson

Treatment of HepG2 cells with known effectors of low density lipoprotein receptor (LDLR) gene expression altered the in vivo pattern of protein-DNA interactions in the promoter. The observed changes are consistent with proteins binding in vivo to the sterol regulatory element (SRE), to Sp1-like sites, as well as to other regions. Protein bound to the SRE in all conditions, but the nature of the dimethyl sulfate reactivity changed depending on the physiological state of the cell. Hypermethylation within the SRE of the low density lipoprotein receptor promoter was observed when cells were treated with cholesterol synthesis inhibitors, insulin, or phorbol 12-myristate 13-acetate, suggesting that the SRE regulates this promoter through sterol-independent as well as sterol-dependent mechanisms. No significant changes were observed in binding to the Sp1-like sites, suggesting that differential binding to these sites does not play a role in altered transcription levels. Analysis of the 3-hydroxy-3-methylglutaryl coenzyme A reductase promoter also revealed protections that varied in a cell type-specific manner. Binding to the 3-hydroxy-3-methylglutaryl coenzyme A reductase SRE and putative nuclear factor 1 sites could be observed but varied little in different physiological conditions.


Journal of Lipid Research | 2010

Assessment of cholesteryl ester transfer protein inhibitors for interaction with proteins involved in the immune response to infection

Ronald W. Clark; David Cunningham; Yang Cong; Timothy A. Subashi; George T. Tkalcevic; David B. Lloyd; James G. Boyd; Boris A. Chrunyk; George A. Karam; Xiayang Qiu; Ing-Kae Wang; Omar L. Francone

The CETP inhibitor, torcetrapib, was prematurely terminated from phase 3 clinical trials due to an increase in cardiovascular and noncardiovascular mortality. Because nearly half of the latter deaths involved patients with infection, we have tested torcetrapib and other CETPIs to see if they interfere with lipopolysaccharide binding protein (LBP) or bactericidal/permeability increasing protein (BPI). No effect of these potent CETPIs on LPS binding to either protein was detected. Purified CETP itself bound weakly to LPS with a Kd ≥ 25 uM compared with 0.8 and 0.5 nM for LBP and BPI, respectively, and this binding was not blocked by torcetrapib. In whole blood, LPS induced tumor necrosis factor-α normally in the presence of torcetrapib. Furthermore, LPS had no effect on CETP activity. We conclude that the sepsis-related mortality of the ILLUMINATE trial was unlikely due to a direct effect of torcetrapib on LBP or BPI function, nor to inhibition of an interaction of CETP with LPS. Instead, we speculate that the negative outcome seen for patients with infections might be related to the changes in plasma lipoprotein composition and metabolism, or alternatively to the known off-target effects of torcetrapib, such as aldosterone elevation, which may have aggravated the effects of sepsis.

Collaboration


Dive into the David B. Lloyd's collaboration.

Researchain Logo
Decentralizing Knowledge