Network


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

Hotspot


Dive into the research topics where Anuradha Ramamoorthy is active.

Publication


Featured researches published by Anuradha Ramamoorthy.


Drug Metabolism and Disposition | 2012

In Silico and In Vitro Identification of MicroRNAs That Regulate Hepatic Nuclear Factor 4α Expression

Anuradha Ramamoorthy; Lang Li; Andrea Gaedigk; L. DiAnne Bradford; Eric A. Benson; David A. Flockhart; Todd C. Skaar

Hepatic nuclear factor 4α (HNF4A) is a nuclear transcription factor that regulates the expression of many genes involved in drug disposition. To identify additional molecular mechanisms that regulate HNF4A, we identified microRNAs (miRNAs) that target HNF4A expression. In silico analyses suggested that HNF4A is targeted by many miRNAs. We conducted in vitro studies to validate several of these predictions. With use of an HNF4A 3′-untranslated region (UTR) luciferase reporter assay, five of six miRNAs tested significantly down-regulated (∼20–40%) the luciferase activity. In HepG2 cells, miR-34a and miR-449a also down-regulated the expression of both the HNF4A protein and an HNF4A target gene, PXR (∼30–40%). This regulation appeared without reduction in HNF4A mRNA expression, suggesting that they must be blocking HNF4A translation. Using additional bioinformatic algorithms, we identified polymorphisms that are predicted to alter the miRNA targeting of HNF4A. Luciferase assays indicated that miR-34a and miR-449a were less effective in regulating a variant (rs11574744) than the wild-type HNF4A 3′-UTR. In vivo, subjects with the variant HNF4A had lower CYP2D6 enzyme activity, although this result was not statistically significant (p = 0.16). In conclusion, our findings demonstrate strong evidence for a role of miRNAs in the regulation of HNF4A.


Xenobiotica | 2012

Stereoselective and regiospecific hydroxylation of ketamine and norketamine

Zeruesenay Desta; Ruin Moaddel; Evan T. Ogburn; Cong Xu; Anuradha Ramamoorthy; Swarajya Lakshmi Vattem Venkata; Mitesh Sanghvi; Michael E. Goldberg; Marc C. Torjman; Irving W. Wainer

The objective was to determine the cytochrome P450s (CYPs) responsible for the stereoselective and regiospecific hydroxylation of ketamine [(R,S)-Ket] to diastereomeric hydroxyketamines, (2S,6S;2R,6R)-HK (5a) and (2S,6R;2R,6S)-HK (5b) and norketamine [(R,S)-norKet] to hydroxynorketamines, (2S,6S;2R,6R)-HNK (4a), (2S,6R;2R,6S)-HNK (4b), (2S,5S;2R,5R)-HNK (4c), (2S,4S;2R,4R)-HNK (4d), (2S,4R;2R,4S)-HNK (4e), (2S,5R;2R,5S)-HNK (4f). The enantiomers of Ket and norKet were incubated with characterized human liver microsomes (HLMs) and expressed CYPs. Metabolites were identified and quantified using LC/MS/MS and apparent kinetic constants estimated using single-site Michaelis–Menten, Hill or substrate inhibition equation.  5a was predominantly formed from (S)-Ket by CYP2A6 and N-demethylated to 4a by CYP2B6. 5b was formed from (R)- and (S)-Ket by CYP3A4/3A5 and N-demethylated to 4b by multiple enzymes. norKet incubation produced 4a, 4c and 4f and minor amounts of 4d and 4e. CYP2A6 and CYP2B6 were the major enzymes responsible for the formation of 4a, 4d and 4f, and CYP3A4/3A5 for the formation of 4e. The 4b metabolite was not detected in the norKet incubates.  5a and 4b were detected in plasma samples from patients receiving (R,S)-Ket, indicating that 5a and 5b are significant Ket metabolites. Large variations in HNK concentrations were observed suggesting that pharmacogenetics and/or metabolic drug interactions may play a role in therapeutic response.


Electrophoresis | 2013

Optimization and validation of a capillary electrophoresis laser-induced fluorescence method for amino acids determination in human plasma: application to bipolar disorder study.

Mª Paz Lorenzo; Alma Villaseñor; Anuradha Ramamoorthy; Antonia García

Quantitative and qualitative analysis of amino acids in biofluids offers relevant information in diagnosis of diseases, evaluation of nutritional state, and in elucidating metabolic influences on physiology. A simple, rapid, and robust procedure in terms of sample treatment, separation, and quantitation based on CE‐LIF has been optimized for use in human plasma samples. Time required for derivatization was 15 min and analysis time was 35 min. 4‐Fluoro‐7‐nitro‐2,1,3‐benzoxadiazole (NBD‐F) was the labeling agent used for obtaining fluorescent derivatives. Electrophoretic conditions were: 175 mM borate buffer at pH 10.25 prepared with 12.5 mM β‐cyclodextrin. The voltage applied was +21 kV. Fourteen amino acids could be quantified: l‐proline, l‐phenylalanine, l‐leucine, l‐isoleucine, l‐ornithine, d‐ornithine, l‐glutamine, l‐alanine, l‐threonine, glycine, l‐serine, d‐serine, taurine and l‐glutamate. With this chiral CE‐LIF method, l‐ and d‐amino acids are adequately separated. The method was validated for a representative group of amino acids in human plasma: l‐proline, l‐isoleucine, l‐ornithine, l‐glutamine, l‐alanine l‐threonine, glycine, l‐serine, d‐serine, and glutamate. The method has been successfully applied to human plasma from patients with bipolar disorder, all of whom were taking lithium as a mood stabilizer. Eleven amino acids were quantified in plasma from nine patients, aged 24–55 years. The results were in accordance to published values for the bipolar patients. The method is useful particularly in studies where plasma amino acid levels can be used as biomarkers for diagnosis of diseases, evaluating the disease progression, and monitoring response to drug therapy.


Xenobiotica | 2012

Breast cancer resistance protein (BCRP/ABCG2) localises to the nucleus in glioblastoma multiforme cells.

Prateek Bhatia; Michel Bernier; Mitesh Sanghvi; Ruin Moaddel; Roland Schwarting; Anuradha Ramamoorthy; Irving W. Wainer

The breast cancer resistance protein (BCRP), an ATP binding cassette (ABC) efflux transporter, plays a role in multiple drug resistance (MDR). Previous studies of the subcellular location of the ABC transporter P-glycoprotein indicated that this protein is expressed in nuclear membranes. This study examines the nuclear distribution of BCRP in seven human-derived glioblastoma (GBM) and astrocytoma cell lines. BCRP expression was observed in the nuclear extracts of 6/7 cell lines. Using the GBM LN229 cell line as a model, nuclear BCRP protein was detected by immunoblotting and confocal laser microscopy. Importantly, nuclear BCRP staining was found in a subpopulation of tumour cells in a human brain GBM biopsy. Mitoxantrone cytotoxicity in the LN229 cell line was determined with and without the BCRP inhibitor fumitremorgin C (FTC) and after downregulation of BCRP with small interfering RNA (siRNA). FTC inhibition of BCRP increased mitoxantrone cytotoxicity with a ~7-fold reduction in the IC50 and this effect was further potentiated in the siRNA -treated cells. In conclusion, BCRP is expressed in the nuclear extracts of select GBM and astrocytoma cell lines and in a human GBM tumour biopsy. Its presence in the nucleus of cancer cells suggests new role for BCRP in MDR.


Pharmacogenetics and Genomics | 2010

Differential quantification of CYP2D6 gene copy number by four different quantitative real-time PCR assays

Anuradha Ramamoorthy; David A. Flockhart; Naoya Hosono; Michiaki Kubo; Yusuke Nakamura; Todd C. Skaar

Copy number variations (CNVs) in the CYP2D6 gene contribute to interindividual variation in drug metabolism. As the most common duplicated allele in Asian populations is the nonfunctional CYP2D6*36 allele, the goal of this study was to identify CNV assays that can differentiate between multiple copies of the CYP2D6*36 allele and multiple copies of other CYP2D6 alleles. We determined CYP2D6 gene copy numbers in 32 individuals with known CYP2D6 CNVs from the Coriell Japanese–Chinese panel using four quantitative real-time PCR assays. These assays target different regions of the CYP2D6 gene: 5′-flanking region, intron 2, intron 6, and exon 9 (Ex9). The specific target site of the Ex9 assay was verified by sequencing the PCR amplicon. Three of the CYP2D6 CNV assays (5′-flanking region, intron 2, and intron 6) estimated CYP2D6 copy numbers that were concordant for all 32 individuals. However, the Ex9 assay was concordant in only 10 of 32 samples. The 10 concordant samples did not contain any CYP2D6*36 alleles and the 22 discordant samples contained at least one CYP2D6*36 allele. In addition, the Ex9 assay accurately quantified all of the non-CYP2D6*36 alleles in all samples. Ex9 amplicon sequencing indicated that it targets a region of CYP2D6 exon 9 that undergoes partial gene-conversion in the CYP2D6*36 allele. In conclusion, CYP2D6 Ex9 CNV assay can be used to determine the copy number of non-CYP2D6*36 alleles. Selective amplification of non-CYP2D6*36 sequence by the Ex9 assay should be useful in determining the number of functional copies of CYP2D6 in Asian populations.


Cellular Signalling | 2013

Nicotinic acetylcholine receptor antagonists alter the function and expression of serine racemase in PC-12 and 1321N1 cells.

Nagendra S. Singh; Rajib K. Paul; Anuradha Ramamoorthy; Marc C. Torjman; Ruin Moaddel; Michel Bernier; Irving W. Wainer

Western blot analysis demonstrated that PC-12 cells express monomeric and dimeric forms of serine racemase (m-SR, d-SR) and that 1321N1 cells express m-SR. Quantitative RT-PCR and functional studies demonstrated that PC-12 cells express homomeric and heteromeric forms of nicotinic acetylcholine receptors (nAChR) while 1321N1 cells primarily express the α7-nAChR subtype. The effect of nAChR agonists and antagonists on SR activity and expression was examined by following concentration-dependent changes in intracellular d-Ser levels and SR protein expression. Incubation with (S)-nicotine increased d-Ser levels, which were attenuated by the α7-nAChR antagonist methyllycaconitine (MLA). Treatment of PC-12 cells with mecamylamine (MEC) produced a bimodal reduction of d-Ser reflecting MEC inhibition of homomeric and heteromeric nAChRs, while a unimodal curve was observed with 1321N1 cells, reflecting predominant expression of α7-nAChR. The nAChR subtype selectivity was probed using α7-nAChR selective inhibitors MLA and (R,S)-dehydronorketamine and α3β4-nAChR specific inhibitor AT-1001. The compounds reduced d-Ser in PC-12 cells, but only MLA and (R,S)-dehydronorketamine were effective in 1321N1 cells. Incubation of PC-12 and 1321N1 cells with (S)-nicotine, MEC and AT-1001 did not affect m-SR or d-SR expression, while MLA and (R,S)-dehydronorketamine increased m-SR expression but not SR mRNA levels. Treatment with cycloheximide indicated that increased m-SR was due to de novo protein synthesis associated with phospho-active forms of ERK1/2, MARCKS, Akt and rapamycin-sensitive mTOR. This effect was attenuated by treatment with the pharmacological inhibitors U0126, LY294002 and rapamycin, which selectively block the activation of ERK1/2, Akt and mTOR, respectively, and siRNAs directed against ERK1/2, Akt and mTOR. We propose that nAChR-associated changes in Ca(2+) flux affect SR activity, but not expression, and that MLA and (R,S)-dehydronorketamine bind to allosteric sites on the α7-nAChR and promote multiple signaling cascades that converge at mTOR to increase m-SR levels.


Drug Metabolism and Disposition | 2013

Regulation of microRNA expression by rifampin in human hepatocytes.

Anuradha Ramamoorthy; Yunlong Liu; Santosh Philips; Zeruesenay Desta; Hai Lin; Chirayu Goswami; Andrea Gaedigk; Lang Li; David A. Flockhart; Todd C. Skaar

Rifampin causes drug interactions by altering hepatic drug metabolism. Because microRNAs (miRNAs) have been shown to regulate genes involved in drug metabolism, we determined the effect of rifampin on the expression of hepatic miRNAs. Primary human hepatocytes from seven subjects were treated with rifampin, and the expression of miRNA and cytochrome P450 (P450) mRNAs was measured by TaqMan assays and RNA-seq, respectively. Rifampin induced the expression of 10 clinically important and 13 additional P450 genes and repressed the expression of 9 other P450 genes (P < 0.05). Rifampin induced the expression of 33 miRNAs and repressed the expression of 35 miRNAs (P < 0.05). Several of these changes were highly negatively correlated with the rifampin-induced changes in the expression of their predicted target P450 mRNAs, supporting the possibility of miRNA-induced regulation of P450 mRNA expression. In addition, several other miRNA changes were positively correlated with the changes in P450 mRNA expression, suggesting similar regulatory mechanisms. Despite the interindividual variability in the rifampin effects on miRNA expression, principal components analysis clearly separated the rifampin-treated samples from the controls. In conclusion, rifampin treatment alters miRNA expression patterns in human hepatocytes, and some of the changes were correlated with the rifampin-induced changes in expression of the P450 mRNAs they are predicted to target.


Pharmacogenomics | 2011

Gene copy number variations: it is important to determine which allele is affected

Anuradha Ramamoorthy; Todd C. Skaar

Copy number variations (CNVs) are one of the primary sources of variation found in the human genome. They involve both the deletion and multi plication of DNA segments [1]. In the human genome, there are thousands of copy number variable regions that range from 100 bp to several Mb pairs [2]. The human genome project revealed that many regions of the genome have CNVs and that some are more common than others [1,3]. According to the Database of Genomic Variants [3], there are 66,741 catalogued CNVs from a total of 15,963 CNV loci (accessed in December, 2010). CNVs have been increasingly implicated in a number of diseases including cancer, developmental diseases, mental illness, autoimmune diseases and infectious diseases [4,5]. The importance of CNVs in genes controlling drug metabolism has been appreciated for many years. A partial list of those affected include UDP-glucuronosyltransferase 2B17 and 2B28 genes (UGT2B17 and UGT2B28) [6], sulfotransferase 1A1 (SULTA1) [7], glutathione S-transferase M1 and T1 genes (GSTT1 and GSTM1) [8], CYP2A6 [9] and CYP2D6 [10]. In fact, UGT2B28, UGT2B17, CYP2A6, GSTM1 and GSTT1 are among the most commonly deleted genes in the human genome [6].


PLOS Genetics | 2014

Genome-Wide Discovery of Drug-Dependent Human Liver Regulatory Elements

Robin P. Smith; Walter L. Eckalbar; Kari M. Morrissey; Marcelo R. Luizon; Thomas J. Hoffmann; Xuefeng Sun; Stacy L. Jones; Shelley Force Aldred; Anuradha Ramamoorthy; Zeruesenay Desta; Yunlong Liu; Todd C. Skaar; Nathan D. Trinklein; Kathleen M. Giacomini; Nadav Ahituv

Inter-individual variation in gene regulatory elements is hypothesized to play a causative role in adverse drug reactions and reduced drug activity. However, relatively little is known about the location and function of drug-dependent elements. To uncover drug-associated elements in a genome-wide manner, we performed RNA-seq and ChIP-seq using antibodies against the pregnane X receptor (PXR) and three active regulatory marks (p300, H3K4me1, H3K27ac) on primary human hepatocytes treated with rifampin or vehicle control. Rifampin and PXR were chosen since they are part of the CYP3A4 pathway, which is known to account for the metabolism of more than 50% of all prescribed drugs. We selected 227 proximal promoters for genes with rifampin-dependent expression or nearby PXR/p300 occupancy sites and assayed their ability to induce luciferase in rifampin-treated HepG2 cells, finding only 10 (4.4%) that exhibited drug-dependent activity. As this result suggested a role for distal enhancer modules, we searched more broadly to identify 1,297 genomic regions bearing a conditional PXR occupancy as well as all three active regulatory marks. These regions are enriched near genes that function in the metabolism of xenobiotics, specifically members of the cytochrome P450 family. We performed enhancer assays in rifampin-treated HepG2 cells for 42 of these sequences as well as 7 sequences that overlap linkage-disequilibrium blocks defined by lead SNPs from pharmacogenomic GWAS studies, revealing 15/42 and 4/7 to be functional enhancers, respectively. A common African haplotype in one of these enhancers in the GSTA locus was found to exhibit potential rifampin hypersensitivity. Combined, our results further suggest that enhancers are the predominant targets of rifampin-induced PXR activation, provide a genome-wide catalog of PXR targets and serve as a model for the identification of drug-responsive regulatory elements.


Pharmacology Research & Perspectives | 2015

The distribution and clearance of (2S,6S)-hydroxynorketamine, an active ketamine metabolite, in Wistar rats

Ruin Moaddel; Mitesh Sanghvi; Katina Sourou Sylvestre Dossou; Anuradha Ramamoorthy; Carol E. Green; James E. Bupp; Robert Swezey; Kathleen O'Loughlin; Irving W. Wainer

The distribution, clearance, and bioavailability of (2S,6S)‐hydroxynorketamine has been studied in the Wistar rat. The plasma and brain tissue concentrations over time of (2S,6S)‐hydroxynorketamine were determined after intravenous (20 mg/kg) and oral (20 mg/kg) administration of (2S,6S)‐hydroxynorketamine (n = 3). After intravenous administration, the pharmacokinetic parameters were estimated using noncompartmental analysis and the half‐life of drug elimination during the terminal phase (t1/2) was 8.0 ± 4.0 h and the apparent volume of distribution (Vd) was 7352 ± 736 mL/kg, clearance (Cl) was 704 ± 139 mL/h per kg, and the bioavailability was 46.3%. Significant concentrations of (2S,6S)‐hydroxynorketamine were measured in brain tissues at 10 min after intravenous administration, ~30 μg/mL per g tissue which decreased to 6 μg/mL per g tissue at 60 min. The plasma and brain concentrations of (2S,6S)‐hydroxynorketamine were also determined after the intravenous administration of (S)‐ketamine, where significant plasma and brain tissue concentrations of (2S,6S)‐hydroxynorketamine were observed 10 min after administration. The (S)‐ketamine metabolites (S)‐norketamine, (S)‐dehydronorketamine, (2S,6R)‐hydroxynorketamine, (2S,5S)‐hydroxynorketamine and (2S,4S)‐hydroxynorketamine were also detected in both plasma and brain tissue. The enantioselectivity of the conversion of (S)‐ketamine and (R)‐ketamine to the respective (2,6)‐hydroxynorketamine metabolites was also investigated over the first 60 min after intravenous administration. (S)‐Ketamine produced significantly greater plasma and brain tissue concentrations of (2S,6S)‐hydroxynorketamine relative to the (2R,6R)‐hydroxynorketamine observed after the administration of (R)‐ketamine. However, the relative brain tissue: plasma concentrations of the enantiomeric (2,6)‐hydroxynorketamine metabolites were not significantly different indicating that the penetration of the metabolite is not enantioselective.

Collaboration


Dive into the Anuradha Ramamoorthy's collaboration.

Top Co-Authors

Avatar

Irving W. Wainer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ruin Moaddel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michel Bernier

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mitesh Sanghvi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marc C. Torjman

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Rajib K. Paul

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Luckenbaugh

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge