Network


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

Hotspot


Dive into the research topics where Bonnie S. Coats is active.

Publication


Featured researches published by Bonnie S. Coats.


Molecular Genetics and Metabolism | 2013

Long-term safety of dichloroacetate in congenital lactic acidosis

Monica Abdelmalak; Alicia Lew; Ryan Ramezani; Albert L. Shroads; Bonnie S. Coats; Taimour Y. Langaee; Meena N. Shankar; Richard E. Neiberger; Sankarasubramon H. Subramony; Peter W. Stacpoole

We followed 8 patients (4 males) with biochemically and/or molecular genetically proven deficiencies of the E1α subunit of the pyruvate dehydrogenase complex (PDC; 3 patients) or respiratory chain complexes I (1 patient), IV (3 patients) or I+IV (1 patient) who received oral dichloroacetate (DCA; 12.5 mg/kg/12 h) for 9.7 to 16.5 years. All subjects originally participated in randomized controlled trials of DCA and were continued on an open-label chronic safety study. Patients (1 adult) ranged in age from 3.5 to 40.2 years at the start of DCA administration and are currently aged 16.9 to 49.9 years (mean ± SD: 23.5 ± 10.9 years). Subjects were either normal or below normal body weight for age and gender. The 3 PDC deficient patients did not consume high fat (ketogenic) diets. DCA maintained normal blood lactate concentrations, even in PDC deficient children on essentially unrestricted diets. Hematological, electrolyte, renal and hepatic status remained stable. Nerve conduction either did not change or decreased modestly and led to reduction or temporary discontinuation of DCA in 3 patients, although symptomatic worsening of peripheral neuropathy did not occur. We conclude that chronic DCA administration is generally well-tolerated in patients with congenital causes of lactic acidosis and is effective in maintaining normal blood lactate levels, even in PDC-deficient children not consuming strict ketogenic diets.


The Journal of Clinical Pharmacology | 2012

Human Polymorphisms in the Glutathione Transferase Zeta 1/Maleylacetoacetate Isomerase Gene Influence the Toxicokinetics of Dichloroacetate

Albert L. Shroads; Taimour Y. Langaee; Bonnie S. Coats; Tracie L. Kurtz; John R. Bullock; David Weithorn; Yan Gong; David Wagner; David A. Ostrov; Julie A. Johnson; Peter W. Stacpoole

Dichloroacetate (DCA), a chemical relevant to environmental science and allopathic medicine, is dehalogenated by the bifunctional enzyme glutathione transferase zeta (GSTz1)/maleylacetoacetate isomerase (MAAI), the penultimate enzyme in the phenylalanine/tyrosine catabolic pathway. The authors postulated that polymorphisms in GSTz1/MAAI modify the toxicokinetics of DCA. GSTz1/MAAI haplotype significantly affected the kinetics and biotransformation of 1,2‐13C‐DCA when it was administered at either environmentally (μg/kg/d) or clinically (mg/kg/d) relevant doses. GSTz1/MAAI haplotype also influenced the urinary accumulation of potentially toxic tyrosine metabolites. Atomic modeling revealed that GSTz1/MAAI variants associated with the slowest rates of DCA metabolism induced structural changes in the enzyme homodimer, predicting protein instability or abnormal protein‐protein interactions. Knowledge of the GSTz1/MAAI haplotype can be used prospectively to identify individuals at potential risk of DCAs adverse side effects from environmental or clinical exposure or who may exhibit aberrant amino acid metabolism in response to dietary protein.


The Journal of Clinical Pharmacology | 2015

Haplotype Variations in Glutathione Transferase Zeta 1 Influence the Kinetics and Dynamics of Chronic Dichloroacetate in Children

Albert L. Shroads; Bonnie S. Coats; Caitrin W. McDonough; Taimour Y. Langaee; Peter W. Stacpoole

Dichloroacetate (DCA) is biotransformed by glutathione transferase zeta 1 (GSTZ1), a bifunctional enzyme that, as maleylacetoacetate isomerase (MAAI), catalyzes the penultimate step in tyrosine catabolism. DCA inhibits GSTZ1/MAAI, leading to delayed plasma drug clearance and to accumulation of potentially toxic tyrosine intermediates. Haplotype variability in GSTZ1 influences short‐term DCA kinetics in healthy adults, but the impact of genotype in children treated chronically with DCA is unknown. Drug kinetics was studied in 17 children and adolescents with congenital mitochondrial diseases administered 1,2‐13C‐DCA. Plasma drug half‐life and trough levels varied 3–6‐fold, depending on GSTZ1/MAAI haplotype and correlated directly with urinary maleylacetone, a substrate for MAAI. However, chronic DCA exposure did not lead to progressive accumulation of plasma drug concentration; instead, kinetics parameters plateaued, consistent with the hypothesis that equipoise is established between the inhibitory effect of DCA on GSTZ1/MAAI and new enzyme synthesis. GSTZ1/MAAI haplotype variability affects DCA kinetics and biotransformation. However, these differences appear to be stable in most individuals and are not associated with DCA plasma accumulation or drug‐associated toxicity in young children.


The Journal of Clinical Pharmacology | 2006

Human Kinetics of Orally and Intravenously Administered Low-Dose 1,2-13C-Dichloroacetate

Minghong Jia; Bonnie S. Coats; Monisha Chadha; Barbara Frentzen; Javier Perez‐Rodriguez; Paul A. Chadik; Richard A. Yost; George N. Henderson; Peter W. Stacpoole

Dichloroacetate (DCA) is a putative environmental hazard, owing to its ubiquitous presence in the biosphere and its association with animal and human toxicity. We sought to determine the kinetics of environmentally relevant concentrations of 1,2‐13C‐DCA administered to healthy adults. Subjects received an oral or intravenous dose of 2.5 μg/kg of 1,2‐13C‐DCA. Plasma and urine concentrations of 1,2‐13C‐DCA were measured by a modified gas chromatography‐tandem mass spectrometry method. 1,2‐13C‐DCA kinetics was determined by modeling using WinNonlin 4.1 software. Plasma concentrations of 1,2‐13C‐DCA peaked 10 minutes and 30 minutes after intravenous or oral administration, respectively. Plasma kinetic parameters varied as a function of dose and duration. Very little unchanged 1,2‐13C‐DCA was excreted in urine. Trace amounts of DCA alter its own kinetics after short‐term exposure. These findings have important implications for interpreting the impact of this xenobiotic on human health.


Drug metabolism and personalized therapy | 2015

Chloral hydrate, through biotransformation to dichloroacetate, inhibits maleylacetoacetate isomerase and tyrosine catabolism in humans

Albert L. Shroads; Bonnie S. Coats; Taimour Y. Langaee; Jonathan J. Shuster; Peter W. Stacpoole

Abstract Background: Chloral hydrate (CH), a sedative and metabolite of the environmental contaminant trichloroethylene, is metabolized to trichloroacetic acid, trichloroethanol, and possibly dichloroacetate (DCA). DCA is further metabolized by glutathione transferase zeta 1 (GSTZ1), which is identical to maleylacetoacetate isomerase (MAAI), the penultimate enzyme in tyrosine catabolism. DCA inhibits its own metabolism through depletion/inactivation of GSTZ1/MAAI with repeated exposure, resulting in lower plasma clearance of the drug and the accumulation of the urinary biomarker maleylacetone (MA), a metabolite of tyrosine. It is unknown if GSTZ1/MAAI may participate in the metabolism of CH or any of its metabolites and, therefore, affect tyrosine catabolism. Stable isotopes were utilized to determine the biotransformation of CH, the kinetics of its major metabolites, and the influence, if any, of GSTZ1/MAAI. Methods: Eight healthy volunteers (ages 21–40 years) received a dose of 1 g of CH (clinical dose) or 1.5 μg/kg (environmental) for five consecutive days. Plasma and urinary samples were analyzed by gas chromatography-mass spectrometry. Results: Plasma DCA (1.2–2.4 μg/mL), metabolized from CH, was measured on the fifth day of the 1 g/day CH dosage but was undetectable in plasma at environmentally relevant doses. Pharmacokinetic measurements from CH metabolites did not differ between slow and fast GSTZ1 haplotypes. Urinary MA levels increased from undetectable to 0.2–0.7 μg/g creatinine with repeated CH clinical dose exposure. Kinetic modeling of a clinical dose of 25 mg/kg DCA administered after 5 days of 1 g/day CH closely resembled DCA kinetics obtained in previously naïve individuals. Conclusions: These data indicate that the amount of DCA produced from clinically relevant doses of CH, although insufficient to alter DCA kinetics, is sufficient to inhibit MAAI and tyrosine catabolism, as evidenced by the accumulation of urinary MA.


American Journal of Physiology-endocrinology and Metabolism | 2004

Tracer-derived total and folate-dependent homocysteine remethylation and synthesis rates in humans indicate that serine is the main one-carbon donor

Steven R. Davis; Peter W. Stacpoole; Jerry Williamson; Lilia S. Kick; Eoin P. Quinlivan; Bonnie S. Coats; Barry Shane; Lynn B. Bailey; Jesse F. Gregory


Investigational New Drugs | 2014

Phase 1 trial of dichloroacetate (DCA) in adults with recurrent malignant brain tumors

E. M. Dunbar; Bonnie S. Coats; Albert L. Shroads; Taimour Y. Langaee; Alicia Lew; J. R. Forder; Jonathan J. Shuster; D. A. Wagner; Peter W. Stacpoole


Journal of Nutrition | 2005

The Methylenetetrahydrofolate Reductase 677C→T Polymorphism and Dietary Folate Restriction Affect Plasma One-Carbon Metabolites and Red Blood Cell Folate Concentrations and Distribution in Women

Steven R. Davis; Eoin P. Quinlivan; Karla Shelnutt; David R. Maneval; Haifa Ghandour; Antonieta Capdevila; Bonnie S. Coats; Conrad Wagner; Jacob Selhub; Lynn B. Bailey; Jonathan J. Shuster; Peter W. Stacpoole; Jesse F. Gregory


The American Journal of Clinical Nutrition | 2005

Dietary vitamin B-6 restriction does not alter rates of homocysteine remethylation or synthesis in healthy young women and men

Steven R. Davis; Jennifer B. Scheer; Eoin P. Quinlivan; Bonnie S. Coats; Peter W. Stacpoole; Jesse F. Gregory


Journal of Nutrition | 2013

Metabolite Profile Analysis Reveals Functional Effects of 28-Day Vitamin B-6 Restriction on One-Carbon Metabolism and Tryptophan Catabolic Pathways in Healthy Men and Women

Vanessa da Silva; Luisa Rios-Avila; Yvonne Lamers; Maria Ralat; Øivind Midttun; Eoin P. Quinlivan; Timothy J. Garrett; Bonnie S. Coats; Meena N. Shankar; Susan S. Percival; Yueh-Yun Chi; Keith E. Muller; Per Magne Ueland; Peter W. Stacpoole; Jesse F. Gregory

Collaboration


Dive into the Bonnie S. Coats'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