Network


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

Hotspot


Dive into the research topics where Claire M. Brett is active.

Publication


Featured researches published by Claire M. Brett.


Journal of Clinical Investigation | 2007

Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action

Yan Shu; Steven A. Sheardown; Chaline Brown; Ryan P. Owen; Shuzhong Zhang; Richard A. Castro; Alexandra G. Ianculescu; Lin Yue; Joan C. Lo; Esteban G. Burchard; Claire M. Brett; Kathleen M. Giacomini

Metformin is among the most widely prescribed drugs for the treatment of type 2 diabetes. Organic cation transporter 1 (OCT1) plays a role in the hepatic uptake of metformin, but its role in the therapeutic effects of the drug, which involve activation of AMP-activated protein kinase (AMPK), is unknown. Recent studies have shown that human OCT1 is highly polymorphic. We investigated whether OCT1 plays a role in the action of metformin and whether individuals with OCT1 polymorphisms have reduced response to the drug. In mouse hepatocytes, deletion of Oct1 resulted in a reduction in the effects of metformin on AMPK phosphorylation and gluconeogenesis. In Oct1-deficient mice the glucose-lowering effects of metformin were completely abolished. Seven nonsynonymous polymorphisms of OCT1 that exhibited reduced uptake of metformin were identified. Notably, OCT1-420del (allele frequency of about 20% in white Americans), previously shown to have normal activity for model substrates, had reduced activity for metformin. In clinical studies, the effects of metformin in glucose tolerance tests were significantly lower in individuals carrying reduced function polymorphisms of OCT1. Collectively, the data indicate that OCT1 is important for metformin therapeutic action and that genetic variation in OCT1 may contribute to variation in response to the drug.


Clinical Pharmacology & Therapeutics | 2008

Effect of Genetic Variation in the Organic Cation Transporter 1, OCT1, on Metformin Pharmacokinetics

Y Shu; Chaline Brown; Richard A. Castro; Rj Shi; Emil T. Lin; Ryan P. Owen; Sa Sheardown; L Yue; Esteban G. Burchard; Claire M. Brett; Kathleen M. Giacomini

The goal of this study was to determine the effects of genetic variation in the organic cation transporter 1, OCT1, on the pharmacokinetics of the antidiabetic drug, metformin. Twenty healthy volunteers with known OCT1 genotype agreed to participate in the study. Each subject received two oral doses of metformin followed by collection of blood and urine samples. OCT1 genotypes had a significant (P<0.05) effect on metformin pharmacokinetics, with a higher area under the plasma concentration–time curve (AUC), higher maximal plasma concentration (Cmax), and lower oral volume of distribution (V/F) in the individuals carrying a reduced function OCT1 allele (R61C, G401S, 420del, or G465R). The effect of OCT1 on metformin pharmacokinetics in mice was less than in humans possibly reflecting species differences in hepatic expression level of the transporter. Our studies suggest that OCT1 genotype is a determinant of metformin pharmacokinetics.


Clinical Pharmacology & Therapeutics | 2007

The Pharmacogenetics Research Network: From SNP Discovery to Clinical Drug Response

Kathleen M. Giacomini; Claire M. Brett; Russ B. Altman; Neal L. Benowitz; M E Dolan; David A. Flockhart; Julie A. Johnson; Daniel F. Hayes; Teri E. Klein; Ronald M. Krauss; Deanna L. Kroetz; Howard L. McLeod; Anne Nguyen; Mark J. Ratain; Mary V. Relling; Victor I. Reus; Dan M. Roden; C A Schaefer; Alan R. Shuldiner; Todd C. Skaar; Kelan G. Tantisira; Rachel F. Tyndale; L. Wang; Richard M. Weinshilboum; Scott T. Weiss; Issam Zineh

The NIH Pharmacogenetics Research Network (PGRN) is a collaborative group of investigators with a wide range of research interests, but all attempting to correlate drug response with genetic variation. Several research groups concentrate on drugs used to treat specific medical disorders (asthma, depression, cardiovascular disease, addiction of nicotine, and cancer), whereas others are focused on specific groups of proteins that interact with drugs (membrane transporters and phase II drug‐metabolizing enzymes). The diverse scientific information is stored and annotated in a publicly accessible knowledge base, the Pharmacogenetics and Pharmacogenomics Knowledge base (PharmGKB). This report highlights selected achievements and scientific approaches as well as hypotheses about future directions of each of the groups within the PGRN. Seven major topics are included: informatics (PharmGKB), cardiovascular, pulmonary, addiction, cancer, transport, and metabolism.


Pharmacogenetics and Genomics | 2009

Effect of genetic variation in the organic cation transporter 2 on the renal elimination of metformin

Ying Chen; Shuanglian Li; Chaline Brown; Stephen Cheatham; Richard A. Castro; Maya K. Leabman; Thomas J. Urban; Ligong Chen; Sook Wah Yee; Ji Ha Choi; Yong Huang; Claire M. Brett; Esteban G. Burchard; Kathleen M. Giacomini

Objective The goal of this study was to determine the effect of a genetic variant in the organic cation transporter 2 (OCT2), OCT2-808G/T, which results in an amino acid change, A270S, on the pharmacokinetics of the antidiabetic drug, metformin. Methods The uptake of metformin was performed in stably transfected HEK-293 cells expressing the empty vector (MOCK), the reference OCT2-808G, and the variant OCT2-808T. Healthy individuals with known OCT2 genotypes [14 homozygous for the OCT2 reference allele (808G/G) and nine heterozygous for the variant allele (808G/T, *3D)] were recruited to this study. Metformin concentrations in plasma and urine were measured by liquid chromatography–tandem mass spectrometry method. Creatinine levels were also measured in plasma and urine. Pharmacokinetic parameters were evaluated for both the groups. Results We observed that in HEK-293 stably transfected cells, OCT2-808T had a greater capacity to transport metformin than did the reference OCT2. Metformin pharmacokinetics was characterized in 23 healthy volunteers of Caucasian and African-American ancestries. We observed that the renal clearance (CLR) and the net secretion (SrCLR) of metformin were significantly different between the volunteers heterozygous for the variant allele (808G/T), and the volunteers homozygous for the reference allele (808G/G) (P<0.005). Multivariate analysis revealed that OCT2 genotype was a significant predictor of CLR and SrCLR of metformin (P<0.01). Conclusion We conclude that genetic variation in OCT2 plays an important role in the CLR and SrCLR of metformin in healthy volunteers.


Journal of Pharmacology and Experimental Therapeutics | 2006

Interaction of Methotrexate with Organic-Anion Transporting Polypeptide 1A2 and Its Genetic Variants

Ilaria Badagnani; Richard A. Castro; Travis R. Taylor; Claire M. Brett; Conrad C. Huang; Douglas Stryke; Michiko Kawamoto; Susan J. Johns; Thomas E. Ferrin; Elaine J. Carlson; Esteban G. Burchard; Kathleen M. Giacomini

Methotrexate (MTX) is used in patients with malignant and autoimmune diseases. This drug is primarily excreted unchanged in the urine, and its net excretion occurs via active secretory and reabsorptive processes. We characterized the interaction of MTX with human organic-anion transporting polypeptide transporter (OATP) 1A2, which is expressed in tissues important for MTX disposition and toxicity, such as the intestine, kidney, liver, and endothelial cells of the blood-brain barrier. In Xenopus laevis oocytes expressing OATP1A2, the uptake of the model substrate, estrone-3-sulfate (ES), was enhanced 30-fold compared with uninjected oocytes. MTX uptake in oocytes expressing OATP1A2 was saturable (Km = 457 ± 118 μM; Vmax = 17.5 ± 4.9 pmol/oocyte/60 min) and sensitive to extracellular pH. That is, acidic pHs stimulated MTX uptake by as much as 7-fold. Seven novel protein-altering variants were identified in 270 ethnically diverse DNA samples. Four protein-altering variants in OATP1A2 exhibited altered transport of ES and/or MTX. The common variant, protein reference sequence (p.) Ile13Thr, was hyperfunctional for ES and MTX and showed a 2-fold increase in the Vmax for ES. The common variant, p. Glu172Asp, exhibited reduced maximal transport capacity for ES and MTX. p. Arg168Cys was hypofunctional, and p. Asn277DEL was nonfunctional. Because of its expression on the apical membrane of the distal tubule and in tissues relevant to MTX disposition and toxicity, these findings suggest that OATP1A2 may play a role in active tubular reabsorption of MTX and in MTX-induced toxicities. Furthermore, genetic variation in OATP1A2 may contribute to variation in MTX disposition and response.


The New England Journal of Medicine | 1982

Prophylactic Indomethacin Therapy for Patent Ductus Arteriosus in Very-Low-Birth-Weight Infants

Lynn Mahony; Violetta Carnero; Claire M. Brett; Michael A. Heymann; Ronald I. Clyman

We performed a double-blind, controlled study of prophylactic indomethacin therapy in 47 premature infants (less than 1700 g) who had subclinical patent ductus arteriosus. They received either indomethacin or placebo at a mean age of 2.9 days. Among the 25 infants weighing more than 1000 g, a hemodynamically important ductus shunt developed in only four of the 14 given placebo. The incidence of important shunts, the number of surgical ligations, and the duration of oxygen therapy were not appreciably different between the study groups. In contrast, among the 22 infants who weighed 1000 g or less, a major ductus shunt developed in 10 of the 12 given placebo. In the smaller infants indomethacin therapy was associated with a significantly lower incidence of major shunts, fewer surgical ligations, a decreased duration of oxygen therapy, and fewer days necessary to regain birth weight. We conclude that prophylactic indomethacin therapy in infants weighing under 1000 g prevents the later development of large ductus shunts and decreases morbidity.


Pediatric Research | 1985

Developmental Changes in Myocardial Contractile Reserve in the Lamb

David F. Teitel; Daniel Sidi; Tom D. Y. Chin; Claire M. Brett; Michael A. Heymann; Abraham M. Rudolph

ABSTRACT: We have assessed serial changes in myocardial contractility and reserve in the normal lamb over the first month of life using an in vivo adaptation of the endsystolic pressure-volume relationship. Via a left thoracotomy, we insert a catheter tip pressure transducer into the left ventricle, affix an echo transducer onto the left ventricular epicardium, place an electromagnetic flow transducer around the pulmonary artery, and insert catheters for monitoring and infusions. We measure contractility by generating left ventricular wall stress-volume index (the cube of dimension) curves, at the same time increasing afterload by infusing phenylephrine. The slope of the endsystolic wall stress-volume index relationship is our index of contractility. Weekly studies were performed at rest and during isoproterenol infusion in 12 animals, and after propranolol administration in four. The data showed a progressive decrease in resting contractility but no change in maximal contractility during isoproterenol infusion over the 4 wk. Taking each week separately, the average increase in contractility during isoproterenol infusion was small at 1 wk (13%), moderate at 2 and 3 wk (24 and 26%, respectively), and large at 4 wk (79%). β-Adrenergic blockade with propranolol caused a significant decrease in contractility in three of four animals studied at 1 wk, in only one of four animals at 2 wk, and in none of four animals at 3 or 4 wk. Thus, the newborn lamb shows a limited reserve in contractility that increases progressively with age; the limited reserve appears secondary to a high resting β-adrenergic state.


Clinical Pharmacology & Therapeutics | 2008

Effects of Genetic Variation in the Novel Organic Cation Transporter, OCTN1, on the Renal Clearance of Gabapentin

Thomas J. Urban; Chaline Brown; Richard A. Castro; N Shah; R Mercer; Yong Huang; Claire M. Brett; Esteban G. Burchard; Kathleen M. Giacomini

Gabapentin is an anticonvulsant that is widely prescribed for epilepsy and other neuropathic disorders. The pharmacokinetics, particularly the absorption and renal elimination, of gabapentin appear to involve membrane transporters. In this study, we tested the hypothesis that organic cation transporter 1 (OCTN1), a multispecific transporter expressed at the apical membrane in intestine and kidney, plays a role in gabapentin pharmacokinetics and that the common variant of OCTN1, OCTN1‐L503F, contributes to variation in the pharmacokinetics of the drug. We observed that OCTN1 facilitates the Na+‐independent transport of gabapentin, and that the OCTN1‐L503F variant is deficient in gabapentin transport activity in stably transfected HEK‐293 cells (fourfold enhanced uptake of gabapentin by OCTN1‐503L vs twofold enhanced uptake by OCTN1‐L503F, compared to mock‐transfected cells). In clinical studies, we found that in subjects homozygous for the L503F variant, gabapentin renal clearance (CLR) approximates the glomerular filtration rate (mean±SE: 110±12 ml/min, n=9), whereas in subjects homozygous for the reference allele, gabapentin undergoes net secretion in the kidney (141±7.8 ml/min, n=11, P<0.05). Creatinine clearance and OCTN1 genotype accounted for 56% of the variation in CLR and were the only significant predictors of CLR (P<0.05). Importantly, OCTN1 genotype was the only significant predictor of net secretion of gabapentin (P<0.008). Oral bioavailability of gabapentin was not affected by OCTN1 genotype. We conclude that OCTN1 contributes to active tubular secretion of gabapentin, and that this effect may be diminished or absent in individuals carrying the OCTN1‐L503F polymorphism. These results provide clinical evidence of the role of genetic variation in renal drug transporters in active drug secretion in vivo.


Clinical Pharmacology & Therapeutics | 2013

The effect of novel promoter variants in MATE1 and MATE2 on the pharmacokinetics and pharmacodynamics of metformin.

Sophie L. Stocker; Kari M. Morrissey; Sook Wah Yee; Richard A. Castro; Lu Xu; Amber Dahlin; Andrea H. Ramirez; Dan M. Roden; Russ A. Wilke; Catherine A. McCarty; Robert L. Davis; Claire M. Brett; Kathleen M. Giacomini

Interindividual variation in response to metformin, first‐line therapy for type 2 diabetes, is substantial. Given that transporters are determinants of metformin pharmacokinetics, we examined the effects of promoter variants in both multidrug and toxin extrusion protein 1 (MATE1) (g.–66T→C, rs2252281) and MATE2 (g.–130G→A, rs12943590) on variation in metformin disposition and response. The pharmacokinetics and glucose‐lowering effects of metformin were assessed in healthy volunteers (n = 57) receiving metformin. The renal and secretory clearances of metformin were higher (22% and 26%, respectively) in carriers of variant MATE2 who were also MATE1 reference (P < 0.05). Both MATE genotypes were associated with altered post‐metformin glucose tolerance, with variant carriers of MATE1 and MATE2 having an enhanced (P < 0.01) and reduced (P < 0.05) response, respectively. Consistent with these results, patients with diabetes (n = 145) carrying the MATE1 variant showed enhanced metformin response. These findings suggest that promoter variants of MATE1 and MATE2 are important determinants of metformin disposition and response in healthy volunteers and diabetic patients.


Pharmacogenetics and Genomics | 2005

Functional analysis of polymorphisms in the organic anion transporter, SLC22A6 (OAT1)

Tomoe Fujita; Chaline Brown; Elaine J. Carlson; Travis R. Taylor; Melanie De La Cruz; Susan J. Johns; Doug Stryke; Michiko Kawamoto; Kazumi Fujita; Richard A. Castro; Chung-Wen Chen; Emil T. Lin; Claire M. Brett; Esteban G. Burchard; Thomas E. Ferrin; Conrad C. Huang; Maya K. Leabman; Kathleen M. Giacomini

Objectives The organic anion transporter, OAT1 (SLC22A6), plays a role in the renal elimination of many drugs and environmental toxins. The goal of this study was to identify and functionally characterize OAT1 variants as a first step towards understanding whether genetic variation in OAT1 may contribute to interindividual differences in renal elimination of xenobiotics. Methods As part of a larger study, 276 DNA samples from an ethnically diverse population were screened and 12 coding region variants of OAT1 were identified. The non-synonymous variants were then constructed and characterized in Xenopus laevis oocytes. A small family-based clinical study was conducted to determine the renal elimination of a model OAT1 substrate, adefovir (an antiviral agent) in human subjects who possessed a non-functional variant, OAT1-R454Q. Results Six non-synonymous variants were identified; two (OAT1-R50 H and OAT1-R293W) were present at ≥1% in at least one ethnic population. These two variants exhibited normal uptake of p-aminohippurate, ochratoxin A and methotrexate assayed in X. laevis oocytes. One variant, OAT1-R454Q, was non-functional with respect to the above substrates. In the clinical study, there was no significant decrease in the renal secretory clearance of adefovir in family members heterozygous for OAT1-454Q in comparison to those with the reference transporter, OAT1-454R. Conclusions These data indicate that the coding region of OAT1 has low genetic and functional diversity and suggest that coding region variants of OAT1 may not contribute substantially to interindividual differences in renal elimination of xenobiotics.

Collaboration


Dive into the Claire M. Brett's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chaline Brown

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sook Wah Yee

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge