Network


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

Hotspot


Dive into the research topics where Elizabeth M. Laurenzana is active.

Publication


Featured researches published by Elizabeth M. Laurenzana.


Toxicological Sciences | 2010

Kynurenic Acid Is a Potent Endogenous Aryl Hydrocarbon Receptor Ligand that Synergistically Induces Interleukin-6 in the Presence of Inflammatory Signaling

Brett C. DiNatale; Iain A. Murray; Jennifer C. Schroeder; Colin A. Flaveny; Tejas S. Lahoti; Elizabeth M. Laurenzana; Curtis J. Omiecinski; Gary H. Perdew

Inflammatory signaling plays a key role in tumor progression, and the pleiotropic cytokine interleukin-6 (IL-6) is an important mediator of protumorigenic properties. Activation of the aryl hydrocarbon receptor (AHR) with exogenous ligands coupled with inflammatory signals can lead to synergistic induction of IL6 expression in tumor cells. Whether there are endogenous AHR ligands that can mediate IL6 production remains to be established. The indoleamine-2,3-dioxygenase pathway is a tryptophan oxidation pathway that is involved in controlling immune tolerance, which also aids in tumor escape. We screened the metabolites of this pathway for their ability to activate the AHR; results revealed that kynurenic acid (KA) is an efficient agonist for the human AHR. Structure-activity studies further indicate that the carboxylic acid group is required for significant agonist activity. KA is capable of inducing CYP1A1 messenger RNA levels in HepG2 cells and inducing CYP1A-mediated metabolism in primary human hepatocytes. In a human dioxin response element-driven stable reporter cell line, the EC(25) was observed to be 104nM, while in a mouse stable reporter cell line, the EC(25) was 10muM. AHR ligand competition binding assays revealed that KA is a ligand for the AHR. Treatment of MCF-7 cells with interleukin-1beta and a physiologically relevant concentration of KA (e.g., 100nM) leads to induction of IL6 expression that is largely dependent on AHR expression. Our findings have established that KA is a potent AHR endogenous ligand that can induce IL6 production and xenobiotic metabolism in cells at physiologically relevant concentrations.


Biochemistry | 2010

The uremic toxin 3-indoxyl sulfate is a potent endogenous agonist for the human aryl hydrocarbon receptor.

Jennifer C. Schroeder; Brett C. DiNatale; Iain A. Murray; Colin A. Flaveny; Qiang Liu; Elizabeth M. Laurenzana; Jyh-Ming Lin; Stephen C. Strom; Curtis J. Omiecinski; Shantu Amin; Gary H. Perdew

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor involved in the regulation of multiple cellular pathways, such as xenobiotic metabolism and Th17 cell differentiation. Identification of key physiologically relevant ligands that regulate AHR function remains to be accomplished. Screening of indole metabolites has identified indoxyl 3-sulfate (I3S) as a potent endogenous ligand that selectively activates the human AHR at nanomolar concentrations in primary human hepatocytes, regulating transcription of multiple genes, including CYP1A1, CYP1A2, CYP1B1, UGT1A1, UGT1A6, IL6, and SAA1. Furthermore, I3S exhibits an approximately 500-fold greater potency in terms of transcriptional activation of the human AHR relative to the mouse AHR in cell lines. Structure-function studies reveal that the sulfate group is an important determinant for efficient AHR activation. This is the first phase II enzymatic product identified that can significantly activate the AHR, and ligand competition binding assays indicate that I3S is a direct AHR ligand. I3S failed to activate either CAR or PXR. The physiological importance of I3S lies in the fact that it is a key uremic toxin that accumulates to high micromolar concentrations in kidney dialysis patients, but its mechanism of action is unknown. I3S represents the first identified relatively high potency endogenous AHR ligand that plays a key role in human disease progression. These studies provide evidence that the production of I3S can lead to AHR activation and altered drug metabolism. Our results also suggest that prolonged activation of the AHR by I3S may contribute to toxicity observed in kidney dialysis patients and thus represent a possible therapeutic target.


Pharmacology, Biochemistry and Behavior | 2007

Sex differences in (+)-amphetamine- and (+)-methamphetamine-induced behavioral response in male and female Sprague-Dawley rats.

Alessandra Milesi-Hallé; Donald E. McMillan; Elizabeth M. Laurenzana; Kelly A. Byrnes-Blake; S. Michael Owens

(+)-Methamphetamine (METH) and (+)-amphetamine (AMP) are structurally similar drugs that are reported to induce similar pharmacological effects in rats of the same sex. Because pharmacokinetic data suggest female rats should be more affected than males, the current studies sought to test the hypothesis that the behavioral and temporal actions of METH and AMP should be greater in female Sprague-Dawley rats than in males. Using a dosing regimen designed to reduce the possibility of tolerance and sensitization, rats were administered 1.0 and 3.0 mg/kg intravenous drug doses. Distance traveled, rearing events and focal stereotypies (e.g., head weaving, sniffing) were measured. Female rats traveled significantly greater distances and displayed a greater number of rearing events than males after both doses. Analysis of stereotypy ratings after 3.0 mg/kg revealed that focal stereotypies were more pronounced and lasted longer in females. The second study compared the potencies of METH and AMP in inducing locomotor activity and focal stereotypies in each sex. No differences in potency were found when METH and AMP effects were compared within males or females. In summary, these studies showed female rats displayed greater and longer-lasting locomotor activity and more stereotypic behaviors, supporting earlier evidence of significant sexual dimorphism in pharmacokinetics.


European Journal of Pharmacology | 2003

Pharmacodynamic mechanisms of monoclonal antibody-based antagonism of (+)-methamphetamine in rats

Kelly A. Byrnes-Blake; Elizabeth M. Laurenzana; F. Ivy Carroll; Philip Abraham; W. Brooks Gentry; Reid D. Landes; S. Michael Owens

Our studies examined pharmacokinetic mechanisms involved in high-affinity (K(d) approximately 11 nM) monoclonal antibody-based antagonism of (+)-methamphetamine-induced locomotor effects. Male rats received (+)-methamphetamine (0.3, 1, or 3 mg/kg i.v.) followed 30 min later by saline or anti-(+)-methamphetamine monoclonal antibody. All groups received a constant dose of monoclonal antibody that was equimolar in binding sites to the body burden of a 1 mg/kg i.v. (+)-methamphetamine dose 30 min after administration. The monoclonal antibody antagonized locomotor effects due to 0.3 and 1 mg/kg (+)-methamphetamine. In contrast, monoclonal antibody treatment increased locomotor activity due to 3 mg/kg (+)-methamphetamine. We also investigated the serum and brain pharmacokinetics of (+)-methamphetamine without and with the monoclonal antibody. Rats received (+)-methamphetamine (1 mg/kg i.v.) followed by saline or monoclonal antibody treatment at 30 min. The monoclonal antibody significantly increased serum methamphetamine concentrations and significantly decreased brain methamphetamine concentrations. These data indicate that anti-(+)-methamphetamine monoclonal antibody-induced pharmacodynamics are complex, but are related to time-dependent changes in (+)-methamphetamine brain distribution.


Pharmacology, Biochemistry and Behavior | 2004

(+)-Methamphetamine-induced spontaneous behavior in rats depends on route of (+)METH administration

W. Brooks Gentry; Abid U. Ghafoor; William D. Wessinger; Elizabeth M. Laurenzana; Howard P. Hendrickson; S. Michael Owens

These studies examined the role of (+)-methamphetamine ((+)METH) administration route on spontaneous behavioral activity vs. time relationships, and pharmacokinetic mechanisms for differences in effects. Male Sprague-Dawley rats (n=6 per administration route) received saline and three doses (0.3, 1.0 and 3.0 mg/kg) of (+)METH in a mixed-sequence design by intravenous (iv), subcutaneous (sc) or intraperitoneal (ip) administration. Locomotion and stereotypy were quantified by video-tracking analysis. The effects of (+)METH on spontaneous behavior were dose- and route-dependent. In particular, total locomotor activity was greatest following 3.0 mg/kg intraperitoneally (P<0.05) and stereotypy ratings were greatest following 3.0 mg/kg subcutaneously (P<0.05). In addition, the duration of locomotor effects was greatest after 3.0 mg/kg subcutaneously (P<0.05). Serum pharmacokinetic parameters were determined in separate rats given 3.0 mg/kg by subcutaneous and intraperitoneal administration (n=4 per administration route). The (+)METH elimination half-life was not different between the routes, but the (+)METH AUC was greater (P<0.05), and the (+)METH and (+)-amphetamine (AMP) maximum concentrations occurred later following subcutaneous than after intraperitoneal dosing (P<0.05), increasing and prolonging drug exposure. In conclusion, the overall pattern of (+)METH effects on locomotor activity depend on dose and the route of administration, which affects serum concentration and the time course of behavioral effects.


Pharmacogenetics | 1998

Post-transcriptional regulation of human microsomal epoxide hydrolase

Elizabeth M. Laurenzana; Christopher Hassett; Curtis J. Omiecinski

Microsomal epoxide hydrolase (mEH) is a key biotransformation enzyme that is variably expressed in humans. Genetic polymorphisms in the mEH gene have been identified that result in amino acid substitutions in the corresponding enzyme. Results of expression analyses of the mEH allelic variants in vitro suggest that the mutations do not affect the specific activity of the mEH enzyme, but may alter post-transcriptional regulation of mEH. To identify potential post-transcriptional mechanisms that influence mEH expression, the translational efficiency, mRNA half-life, and protein half-life of mEH allelic variants were determined. Constructs encoding each of the four mEH alleles were transcribed in vitro and translated. No differences were detected in the rate of protein synthesis among the variant transcripts, indicating that the previously characterized coding region polymorphisms do not appear to affect translational efficiency. mEH variant RNA half-lives were determined in transfected COS-1 cells, but no differences in decay rates were apparent among the polymorphic constructs. Half-lives of the polymorphic mEH proteins were determined in transiently transfected COS-1 cells treated with the protein synthesis inhibitor cycloheximide. Calculated protein half-lives were: Y113/H139, 15.2 h; H113/H139, 10.7 h, Y113/H139, 16.9 h and H113/R139, 16.0 h. The protein half-lives calculated for the polymorphic variants exhibited the same rank order as mEH protein and activity levels determined previously from expression experiments in vitro and therefore suggest that polymorphic amino acid substitution may result in altered protein stability. However, the differences noted were not statistically significant at the P < 0.05 level, and therefore additional study is required to firmly establish causative relationships.


Journal of Pharmacology and Experimental Therapeutics | 2008

Development and Preclinical Testing of a High-Affinity Single-Chain Antibody against (+)-Methamphetamine

Eric C. Peterson; Elizabeth M. Laurenzana; William T. Atchley; Howard P. Hendrickson; S M Owens

Chronic or excessive (+)-methamphetamine (METH) use often leads to addiction and toxicity to critical organs like the brain. With medical treatment as a goal, a novel single-chain variable fragment (scFv) against METH was engineered from anti-METH monoclonal antibody mAb6H4 (IgG, κ light chain, Kd = 11 nM) and found to have similar ligand affinity (Kd = 10 nM) and specificity as mAb6H4. The anti-METH scFv (scFv6H4) was cloned, expressed in yeast, purified, and formulated as a naturally occurring mixture of monomer (∼75%) and dimer (∼25%). To test the in vivo efficacy of the scFv6H4, male Sprague-Dawley rats (n = 5) were implanted with 3-day s.c. osmotic pumps delivering 3.2 mg/kg/day METH. After reaching steady-state METH concentrations, an i.v. dose of scFv6H4 (36.5 mg/kg, equimolar to the METH body burden) was administered along with a [3H]scFv6H4 tracer. Serum pharmacokinetic analysis of METH and [3H]scFv6H4 showed that the scFv6H4 caused an immediate 65-fold increase in the METH concentrations and a 12-fold increase in the serum METH area under the concentration-time curve from 0 to 480 min after scFv6H4 administration. The scFv6H4 monomer was quickly cleared or converted to multivalent forms with an apparent t1/2λz of 5.8 min. In contrast, the larger scFv6H4 multivalent forms (dimers, trimers, etc.) showed a much longer t1/2λz (228 min), and the significantly increased METH serum molar concentrations correlated directly with scFv6H4 serum molar concentrations. Considered together, these data suggested that the scFv6H4 multimers (and not the monomer) were responsible for the prolonged redistribution of METH into the serum.


Vaccine | 2009

Functional and biological determinants affecting the duration of action and efficacy of anti-(+)-methamphetamine monoclonal antibodies in rats

Elizabeth M. Laurenzana; Howard P. Hendrickson; Dylan Carpenter; Eric C. Peterson; W. Brooks Gentry; Michael West; Yingni Che; F. Ivy Carroll; S. Michael Owens

These studies examined the in vivo pharmacokinetics and efficacy of five anti-methamphetamine monoclonal antibodies (mAbs, K(D) values from 11 to 250 nM) in rats. While no substantive differences in mAb systemic clearance (t(1/2)=6.1-6.9 days) were found, in vivo function was significantly reduced within 1-3 days for four of the five mAbs. Only mAb4G9 was capable of prolonged efficacy, as judged by prolonged high methamphetamine serum concentrations. MAb4G9 also maintained high amphetamine serum concentrations, along with reductions in methamphetamine and amphetamine brain concentrations, indicating neuroprotection. The combination of broad specificity for methamphetamine-like drugs, high affinity, and prolonged action in vivo suggests mAb4G9 is a potentially efficacious medication for treating human methamphetamine-related medical diseases.


Drug Metabolism and Disposition | 2006

ANTI-PHENCYCLIDINE MONOCLONAL ANTIBODY BINDING CAPACITY IS NOT THE ONLY DETERMINANT OF EFFECTIVENESS, DISPROVING THE CONCEPT THAT ANTIBODY CAPACITY IS EASILY SURMOUNTED

Grzegorz Pitas; Elizabeth M. Laurenzana; D. Keith Williams; S. Michael Owens; W. Brooks Gentry

The effectiveness of a high-affinity monoclonal antibody (mAb) antagonist against chronic phencyclidine (PCP) use has been demonstrated in rats. In this study, we tested the hypothesis that intravenous doses of PCP in excess of the binding capacity of an anti-PCP mAb cannot easily surmount the beneficial effects of the mAb, even in the presence of a high body burden of the drug. One day after steady-state PCP concentrations were achieved in male rats by continuous s.c. infusion (18 mg/kg/day), a single i.v. dose of saline or the anti-PCP mAb (KD = 1.3 nM; at one-third the molar dose of the PCP body burden), treatment was administered. In an attempt to further surmount the effects of the mAb, rats were challenged with a single 1.0 mg/kg i.v. bolus PCP dose (along with a [3H]PCP tracer) 3 days after the mAb or saline treatment. Total (i.v. bolus + s.c. infusion) PCP concentrations were measured in serum, brain, and testis by radioimmunoassay before and after the challenge, and [3H]PCP concentrations were measured by liquid scintillation spectrometry. The anti-PCP mAb protected against adverse health effects, significantly increased the serum total and bolus PCP concentrations (p < 0.05), and significantly decreased brain total and bolus PCP concentrations (p < 0.05) after the i.v. challenge. These results showed the antibody can counteract extreme and potentially fatal PCP challenges and disproved the hypothesis that attempts to surmount the effects of the antibody with extremely high PCP doses would have immediate adverse health effects.


Aaps Journal | 2006

Quantitative determination of total methamphetamine and active metabolites in rat tissue by liquid chromatography with tandem mass spectrometric detection

Howard P. Hendrickson; Elizabeth M. Laurenzana; S. Michael Owens

High-throughput liquid chromatography with tandem mass spectrometric detection (LC-MS/MS) methodology for the determination of methamphetamine (METH), amphetamine (AMP), 4-hydroxymethamphetamine (4-OH-METH), and 4-hydroxyamphetamine (4-OH-AMP) was developed and validated using simple trichloroacetic acid sample treatment. The method was validated in rat serum, brain, and testis. Lower limits-of-quantitation (LOQ) for METH and AMP were 1 ng·mL−1 using positive ion electrospray tandem mass spectrometry (MS/MS). The accuracy of the method was within 25% of the actual values over a wide range of analyte concentrations. The within-assay precision was better than 12% (coefficient of variation). The method was linear over a wide dynamic range (0.3–1000 ng·mL−1). Quantitation was possible in all 3 matrices using only serum standards because of minimal matrix-associated ion effects or the use of an internal standard. Finally, the LC-MS/MS method was used to determine serum, brain, and testis METH and AMP concentrations during a subcutaneous infusion (5.6 mg kg−1 day−1) of METH in rats. Concentrations of 4-OH-AMP and 4-OH-METH were below the LOQ in experimental samples. The bias introduced by using serum calibrators for the determination of METH and AMP concentrations in testis and brain was less than 8% and insignificant relative to the interanimal variability.

Collaboration


Dive into the Elizabeth M. Laurenzana's collaboration.

Top Co-Authors

Avatar

S. Michael Owens

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Howard P. Hendrickson

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

W. Brooks Gentry

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Curtis J. Omiecinski

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

S M Owens

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

William Brooks Gentry

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Alessandra Milesi-Hallé

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

D. Keith Williams

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

David K. Williams

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Kelly A. Byrnes-Blake

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge