Network


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

Hotspot


Dive into the research topics where Chaline Brown is active.

Publication


Featured researches published by Chaline Brown.


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.


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.


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.


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.


Molecular Pharmacology | 2006

Functional Genetic Diversity in the High-Affinity Carnitine Transporter OCTN2 (SLC22A5)

Thomas J. Urban; Renata C. Gallagher; Chaline Brown; Richard A. Castro; Leah L. Lagpacan; Claire M. Brett; Travis R. Taylor; Elaine J. Carlson; Thomas E. Ferrin; Esteban G. Burchard; Seymour Packman; Kathleen M. Giacomini

Systemic carnitine deficiency (SCD) is a rare autosomal recessive disease resulting from defects in the OCTN2 (SLC22A5) gene, which encodes the high-affinity plasma membrane carnitine transporter. Although OCTN2 is fairly well studied in its relationship with SCD, little is known about the carrier frequency of disease-causing alleles of OCTN2, or of more common functional polymorphisms in this gene. To address these issues, we screened for genetic variants in the OCTN2 coding region by direct sequencing of the exons and flanking intronic region of OCTN2 in a large sample (n = 276) of ethnically diverse subjects. In addition, we established lymphoblastoid cell lines from subjects homozygous for either allele of the previously identified promoter region variant, -207G>C. We found eight amino acid sequence variants of OCTN2, of which three (Phe17Leu, Leu144Phe, and Pro549Ser) were polymorphic in at least one ethnic group. When assayed for functional activity by expression in human embryonic kidney 293 cells, using as probes both the endogenous substrate (l-carnitine) and the organic cation tetraethylammonium, three variants showed functional differences from the reference OCTN2 (Phe17Leu, Tyr449Asp, Val481Phe; p < 0.05). Further studies of the Phe17Leu polymorphism showed a reduced Vmax for l-carnitine transport to approximately 50% of the reference OCTN2. Confocal microscopy studies using an OCTN2-GFP fusion protein showed that Phe17Leu had distinct subcellular localization from the reference OCTN2, with diffuse cytoplasmic retention of Phe17Leu, in contrast to reference OCTN2, which localized specifically to the plasma membrane. Lymphoblasts from subjects homozygous for the -207G allele showed increased l-carnitine transport compared with the -207C/C homozygotes (p < 0.05). This study suggests that although loss-of-function mutations in OCTN2 are likely to be rare, common variants of OCTN2 found in healthy populations may contribute to variation in the disposition of carnitine and some clinically used drugs.


Journal of Pharmaceutical Sciences | 2013

Reduced Renal Clearance of Cefotaxime in Asians with a Low-Frequency Polymorphism of OAT3 (SLC22A8)

Sook Wah Yee; Anh Nguyet Nguyen; Chaline Brown; Radojka M. Savic; Youcai Zhang; Richard A. Castro; Cheryl D. Cropp; Ji Ha Choi; Diment Singh; Harunobu Tahara; Sophie L. Stocker; Yong Huang; Claire M. Brett; Kathleen M. Giacomini

Organic anion transporter 3 (OAT3, SLC22A8), a transporter expressed on the basolateral membrane of the proximal tubule, plays a critical role in the renal excretion of organic anions including many therapeutic drugs. The goal of this study was to evaluate the in vivo effects of the OAT3-Ile305Phe variant (rs11568482), present at 3.5% allele frequency in Asians, on drug disposition with a focus on cefotaxime, a cephalosporin antibiotic. In HEK293-Flp-In cells, the OAT3-Ile305Phe variant had a lower maximum cefotaxime transport activity, Vmax , [159 ± 3 nmol*(mg protein)(-1) /min (mean ± SD)] compared with the reference OAT3 [305 ± 28 nmol*(mg protein)(-1) /min, (mean ± SD), p < 0.01], whereas the Michaelis-Menten constant values (Km ) did not differ. In healthy volunteers, we found volunteers that were heterozygous for the Ile305Phe variant and had a significantly lower cefotaxime renal clearance (CLR ; mean ± SD: 84.8 ± 32.1 mL/min, n = 5) compared with volunteers that were homozygous for the reference allele (158 ± 44.1 mL/min, n = 10; p = 0.006). Furthermore, the net secretory component of cefotaxime renal clearance (CLsec ) was reduced in volunteers heterozygous for the variant allele [33.3 ± 31.8 mL/min (mean ± SD)] compared with volunteers homozygous for the OAT3 reference allele [97.0 ± 42.2 mL/min (mean ± SD), p = 0.01]. In summary, our study suggests that a low-frequency reduced-function polymorphism of OAT3 associates with reduced cefotaxime CLR and CL(sec) .


Clinical Pharmacology & Therapeutics | 2005

Heritability of metformin renal clearance

Maya K. Leabman; Chaline Brown; J. Chung; Richard A. Castro; Gary E. Swan; Emil T. Lin; Kathleen M. Giacomini

Twin studies have been used to determine the genetic contribution to variation in hepatic drug metabolism. In contrast, little is known about the contribution of genetic factors to variation in renal drug elimination. In this study, we used monozygotic twin pairs to assess the heritability of the clearance of the anti‐diabetic agent, metformin, a drug that is eliminated exclusively in the kidneys.


Clinical Pharmacology & Therapeutics | 2005

Functional heterogeneity of polymorphisms in the human organic anion transporter, OAT3 (SLC22A8)

Andrew R. Erdman; Chaline Brown; Richard A. Castro; Travis R. Taylor; Elaine J. Carlson; Kathleen M. Giacomini

The human organic anion transporter, OAT3 (SLC22A8), mediates renal excretion of various drugs including cephalosporins. To test whether genetic variants of OAT3 exhibit altered function in vitro and in vivo, we conducted a series of studies.


Pharmacogenetics and Genomics | 2007

Functional effects of protein sequence polymorphisms in the organic cation/ergothioneine transporter OCTN1 (SLC22A4)

Thomas J. Urban; Chen Yang; Leah L. Lagpacan; Chaline Brown; Richard A. Castro; Travis R. Taylor; Conrad C. Huang; Douglas Stryke; Susan J. Johns; Michiko Kawamoto; Elaine J. Carlson; Thomas E. Ferrin; Esteban G. Burchard; Kathleen M. Giacomini

Collaboration


Dive into the Chaline Brown's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge