Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Antimicrob Agents Chemother ; 66(9): e0025322, 2022 09 20.
Article in English | MEDLINE | ID: mdl-35920662

ABSTRACT

Taniborbactam, an investigational ß-lactamase inhibitor that is active against both serine- and metallo-ß-lactamases, is being developed in combination with cefepime to treat serious infections caused by multidrug-resistant Gram-negative bacteria. Anticipating the use of cefepime-taniborbactam in patients with impaired renal function, an open-label, single-dose clinical study was performed to examine the pharmacokinetics of both drugs in subjects with various degrees of renal function. Hemodialysis-dependent subjects were also studied to examine the amounts of cefepime and taniborbactam dialyzed. Single intravenous infusions of 2 g cefepime and 0.5 g taniborbactam coadministered over 2 h were examined, with hemodialysis-dependent subjects receiving doses both on- and off-dialysis. No subjects experienced serious adverse events or discontinued treatment due to adverse events. The majority of adverse events observed were mild in severity, and there were no trends in the safety of cefepime-taniborbactam related to declining renal function or the timing of hemodialysis. Clinically significant and similar decreases in drug clearance with declining renal function were observed for both cefepime and taniborbactam. The respective decreases in geometric mean clearance for subjects with mild, moderate, and severe renal impairment compared to subjects with normal renal function were 18%, 63%, and 78% for cefepime and 15%, 63%, and 81% for taniborbactam, respectively. Decreases in clearance were similar for both drugs and were shown to be proportional to decreases in renal function. Both cefepime and taniborbactam were dialyzable, with similar amounts removed during 4 h of hemodialysis. This study is registered at ClinicalTrials.gov as NCT03690362.


Subject(s)
Renal Insufficiency , beta-Lactamase Inhibitors , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Borinic Acids , Carboxylic Acids , Cefepime/therapeutic use , Humans , Renal Insufficiency/drug therapy , Serine , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases
2.
Curr Protoc ; 2(8): e527, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36001747

ABSTRACT

Histone post-translational modifications (PTMs) play important roles in many biological processes, including gene regulation and chromatin dynamics, and are thus of high interest across many fields of biological research. Chromatin immunoprecipitation coupled with sequencing (ChIP-seq) is a powerful tool to profile histone PTMs in vivo. This method, however, is largely dependent on the specificity and availability of suitable commercial antibodies. While mass spectrometry (MS)-based proteomic approaches to quantitatively measure histone PTMs have been developed in mammals and several other model organisms, such methods are currently not readily available in plants. One major challenge for the implementation of such methods in plants has been the difficulty in isolating sufficient amounts of pure, high-quality histones, a step rendered difficult by the presence of the cell wall. Here, we developed a high-yielding histone extraction and purification method optimized for Arabidopsis thaliana that can be used to obtain high-quality histones for MS. In contrast to other methods used in plants, this approach is relatively simple, and does not require membranes or additional specialized steps, such as gel excision or chromatography, to extract highly purified histones. We also describe methods for producing MS-ready histone peptides through chemical labeling and digestion. Finally, we describe an optimized method to quantify and analyze the resulting histone PTM data using a modified version of EpiProfile 2.0 for Arabidopsis. In all, the workflow described here can be used to measure changes to histone PTMs resulting from various treatments, stresses, and time courses, as well as in different mutant lines. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Nuclear isolation and histone acid extraction Basic Protocol 2: Peptide labeling, digestion, and desalting Basic Protocol 3: Histone HPLC-MS/MS and data analysis.


Subject(s)
Arabidopsis , Histones , Animals , Arabidopsis/genetics , Histone Code , Histones/chemistry , Mammals/genetics , Peptides/genetics , Protein Processing, Post-Translational , Proteomics/methods , Tandem Mass Spectrometry/methods
3.
Antimicrob Agents Chemother ; 65(11): e0105321, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34370573

ABSTRACT

Taniborbactam (formerly VNRX-5133), an investigational ß-lactamase inhibitor active against both serine- and metallo-ß-lactamases, is being developed in combination with cefepime to treat serious infections caused by multidrug-resistant Gram-negative bacteria. This first-in-human study evaluated the safety and pharmacokinetics of single and multiple doses of taniborbactam in healthy adult subjects. Single doses of 62.5 to 1,500 mg taniborbactam and multiple doses of 250 to 750 mg taniborbactam every 8 h (q8h) for 10 days were examined; all taniborbactam doses were administered as a 2-h intravenous infusion. No subjects experienced serious adverse events or discontinued treatment due to adverse events. The most common adverse event in both placebo- and taniborbactam-treated subjects was headache. The pharmacokinetics of taniborbactam were similar to the pharmacokinetics reported for cefepime. Taniborbactam demonstrated dose-proportional pharmacokinetics with low intersubject variability. Following single doses and with extended sampling, the mean terminal elimination half-life ranged from 3.4 to 5.8 h; however, the majority of exposure was characterized by an earlier phase with a half-life of about 2 h. Following multiple dosing, there was minimal accumulation of taniborbactam in plasma. At steady-state, approximately 90% of the administered dose of taniborbactam was recovered in urine as intact drug. There was no appreciable metabolism observed in either plasma or urine samples. (This study is registered at clinicaltrials.gov under registration number NCT02955459.).


Subject(s)
Borinic Acids , beta-Lactamase Inhibitors , Adult , Borinic Acids/adverse effects , Carboxylic Acids , Double-Blind Method , Half-Life , Healthy Volunteers , Humans , beta-Lactamase Inhibitors/adverse effects
4.
Pharmacotherapy ; 41(5): 451-456, 2021 05.
Article in English | MEDLINE | ID: mdl-33797776

ABSTRACT

STUDY OBJECTIVE: Lefamulin is a novel IV and oral pleuromutilin recently approved for the treatment of community-acquired bacterial pneumonia (CABP). Given that renal comorbidities are common in patients admitted for CABP, understanding the pharmacokinetics of lefamulin in the face of severe renal impairment, including those requiring hemodialysis, is needed. DESIGN: Open-label, Phase-1 pharmacokinetic study. SETTING: Research Study Center. PATIENTS: Twenty-three matched subjects were included, seven with "Normal" renal function (creatinine clearance >90 ml/min), eight with "Severe" renal impairment (glomerular filtration rate <30 ml/min/1.73 m2 ), and eight subjects requiring hemodialysis. MEASUREMENTS AND MAIN RESULTS: Subjects were administered a single dose of lefamulin IV 150 mg as a 1-h infusion. Subjects in the hemodialysis group started hemodialysis within 1 h after lefamulin infusion (On dialysis), as well as, on a non-dialysis day (Off dialysis). Plasma, urine, and dialysate fluid were collected for 36 h and analyzed for lefamulin and its major metabolite, BC-8041. Lefamulin was primarily excreted non-renally across groups. Statistical analyses revealed lefamulin and BC-8041 pharmacokinetics were similar between Normal and Severe groups, except for renal clearance, which decreased in Severe subjects (mean 1.3 L/h Normal vs. 0.4 L/h Severe). Likewise, lefamulin pharmacokinetics during on and off dialysis were unchanged, with lefamulin not measurably filtered in dialysate fluid. Two, three, and three subjects reported drug-related treatment-emergent adverse events (TEAE) in Normal, Severe, and Hemodialysis groups, respectively. All TEAEs were mild, except one (infusion-site reaction) that was classified as moderate. CONCLUSION: No dosage adjustment is required for patients with renal impairment, and lefamulin can be administered without regard to hemodialysis timing.


Subject(s)
Diterpenes , Polycyclic Compounds , Renal Dialysis , Renal Insufficiency , Thioglycolates , Administration, Intravenous , Diterpenes/administration & dosage , Diterpenes/adverse effects , Diterpenes/pharmacokinetics , Humans , Polycyclic Compounds/administration & dosage , Polycyclic Compounds/adverse effects , Polycyclic Compounds/pharmacokinetics , Renal Insufficiency/drug therapy , Renal Insufficiency/therapy , Thioglycolates/administration & dosage , Thioglycolates/adverse effects , Thioglycolates/pharmacokinetics
5.
Pharmacotherapy ; 41(5): 457-462, 2021 05.
Article in English | MEDLINE | ID: mdl-33811377

ABSTRACT

STUDY OBJECTIVE: Lefamulin is a novel pleuromutilin recently approved by the FDA for the treatment of community-acquired bacterial pneumonia. Given that, lefamulin is primarily metabolized by CYP450 Phase-1 reactions, this study evaluated the pharmacokinetics of IV lefamulin in subjects with various degrees of hepatic impairment as compared with matched healthy subjects. DESIGN: Open-label, Phase-1 clinical pharmacokinetic study. SETTING: Research Study Center. PATIENTS: Twenty-seven subjects; comprised of 11 individuals with normal hepatic function and eight each with moderate or severe hepatic impairment were included, as classified by Child-Pugh scores. MEASUREMENTS AND MAIN RESULTS: Subjects were administered a single dose of IV lefamulin 150 mg over 1 h. Plasma was collected for 48 h and analyzed for lefamulin and its major metabolite, BC-8041, concentrations in addition to assessing lefamulin plasma protein binding. Pharmacokinetics were evaluated by noncompartmental analysis. Pharmacokinetic parameters were compared using least square geometric mean ratios. Lefamulin was well tolerated in all hepatic function groups. Statistical analyses revealed reductions in Cmax and increases in renal clearance for Moderate and Severe groups, as well as, the increased volume of distribution for the Severe group. Lefamulin plasma AUC mean (SD) was similar across groups at 7615 (1554), 8233 (2286), and 8938 (1640) h.ng/mL for Normal, Moderate, and Severe groups, respectively, despite decreased clearance observed primarily during terminal elimination phases. Decreased plasma-protein binding was seen in hepatically-impaired versus normal subjects. CONCLUSION: Lefamulin was generally well tolerated. Differences in lefamulin and BC-8041 pharmacokinetics were small, relative to the overall variability, and any changes appear to be compensated by increases in renal clearance and decreased protein binding.


Subject(s)
Diterpenes , Liver Diseases , Polycyclic Compounds , Thioglycolates , Administration, Intravenous , Diterpenes/administration & dosage , Diterpenes/adverse effects , Diterpenes/pharmacology , Humans , Liver Diseases/drug therapy , Polycyclic Compounds/administration & dosage , Polycyclic Compounds/adverse effects , Polycyclic Compounds/pharmacology , Thioglycolates/administration & dosage , Thioglycolates/adverse effects , Thioglycolates/pharmacology
6.
ACS Omega ; 6(4): 2494-2504, 2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33553868

ABSTRACT

Previous benchmarking studies have demonstrated the importance of instrument acquisition methodology and statistical analysis on quantitative performance in label-free proteomics. However, the effects of these parameters in combination with replicate number and false discovery rate (FDR) corrections are not known. Using a benchmarking standard, we systematically evaluated the combined impact of acquisition methodology, replicate number, statistical approach, and FDR corrections. These analyses reveal a complex interaction between these parameters that greatly impacts the quantitative fidelity of protein- and peptide-level quantification. At a high replicate number (n = 8), both data-dependent acquisition (DDA) and data-independent acquisition (DIA) methodologies yield accurate protein quantification across statistical approaches. However, at a low replicate number (n = 4), only DIA in combination with linear models for microarrays (LIMMA) and reproducibility-optimized test statistic (ROTS) produced a high level of quantitative fidelity. Quantitative accuracy at low replicates is also greatly impacted by FDR corrections, with Benjamini-Hochberg and Storey corrections yielding variable true positive rates for DDA workflows. For peptide quantification, replicate number and acquisition methodology are even more critical. A higher number of replicates in combination with DIA and LIMMA produce high quantitative fidelity, while DDA performs poorly regardless of replicate number or statistical approach. These results underscore the importance of pairing instrument acquisition methodology with the appropriate replicate number and statistical approach for optimal quantification performance.

7.
Methods Mol Biol ; 1983: 79-106, 2019.
Article in English | MEDLINE | ID: mdl-31087294

ABSTRACT

Posttranslational modifications of proteins control many complex biological processes, including genome expression, chromatin dynamics, metabolism, and cell division through a language of chemical modifications. Improvements in mass spectrometry-based proteomics have demonstrated protein acetylation is a widespread and dynamic modification in the cell; however, many questions remain on the regulation and downstream effects, and an assessment of the overall acetylation stoichiometry is needed. In this chapter, we describe the determination of acetylation stoichiometry using data-independent acquisition mass spectrometry to expand the number of acetylation sites quantified. However, the increased depth of data-independent acquisition is limited by the spectral library used to deconvolute fragmentation spectra. We describe a powerful approach of subcellular fractionation in conjunction with offline prefractionation to increase the depth of the spectral library. This deep interrogation of subcellular compartments provides essential insights into the compartment-specific regulation and downstream functions of protein acetylation.


Subject(s)
Lysine/metabolism , Proteins/metabolism , Acetylation , Cell Culture Techniques , Cell Fractionation , Chromatography, Liquid , Chromatography, Reverse-Phase , Data Interpretation, Statistical , Hydrogen-Ion Concentration , Lysine/chemistry , Mass Spectrometry/methods , Mitochondria/metabolism , Protein Processing, Post-Translational , Proteins/chemistry , Proteomics/methods , Tandem Mass Spectrometry
8.
Neurosci Lett ; 678: 8-15, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29715546

ABSTRACT

Transactive response DNA-binding protein of 43 kDa (TDP-43) functions as a heterogeneous nuclear ribonucleoprotein and is the major pathological protein in frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis/motor neuron disease (ALS/MND). TDP-43 pathology may also be present as a comorbidity in approximately 20-50% of sporadic Alzheimer's disease cases. In a mouse model of MND, full-length TDP-43 increases association with the mitochondria and blocking the TDP-43/mitochondria interaction ameliorates motor dysfunction. Utilizing a proteomics screen, several mitochondrial TDP-43-interacting partners were identified, including voltage-gated anion channel 1 (VDAC1) and prohibitin 2 (PHB2), a crucial mitophagy receptor. Overexpression of TDP-43 led to an increase in PHB2 whereas TDP-43 knockdown reduced PHB2 expression in cells treated with carbonyl cyanide m-chlorophenylhydrazone (CCCP), an inducer of mitophagy. These results suggest that TDP-43 expression contributes to metabolism and mitochondrial function however we show no change in bioenergetics when TDP-43 is overexpressed and knocked down in HEK293T cells. Furthermore, the fusion protein mitofusin 2 (MFN2) interacts in complex with TDP-43 and selective expression of human TDP-43 in the hippocampus and cortex induced an age-dependent change in Mfn2 expression. Mitochondria morphology is altered in 9-month-old mice selectively expressing TDP-43 in an APP/PS1 background compared with APP/PS1 littermates. We further confirmed TDP-43 localization to the mitochondria using immunogold labeled TDP-43 transmission electron microscopy (TEM) and mitochondrial isolation methods There was no increase in full-length TDP-43 localized to the mitochondria in APP/PS1 mice compared to wild-type (littermates); however, using C- and N-terminal-specific TDP-43 antibodies, the N-terminal (27 kDa, N27) and C-terminal (30 kDa, C30) fragments of TDP-43 are greatly enriched in mitochondrial fractions. In addition, when the mitochondrial peptidase (PMPCA) is overexpressed there is an increase in the N-terminal fragment (N27). These results suggest that TDP-43 processing may contribute to metabolism and mitochondrial function.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Mitophagy , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , GTP Phosphohydrolases/metabolism , Hippocampus/metabolism , Hippocampus/ultrastructure , Mice , Mitochondria/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Prohibitins , Repressor Proteins/metabolism
9.
Neurobiol Dis ; 103: 154-162, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28416393

ABSTRACT

Although the main focus in Alzheimer's disease (AD) has been an investigation of mechanisms causing Aß plaque deposition and tau tangle formation, recent studies have shown that phosphorylated TDP-43 pathology is present in up to 50% of sporadic cases. Furthermore, elevated phosphorylated TDP-43 has been associated with more severe AD pathology. Therefore, we hypothesized that TDP-43 may regulate amyloid-beta precursor protein (APP) trafficking and tau phosphorylation/aggregation. In order to examine the role of TDP-43 in AD, we developed a transgenic mouse that overexpresses hippocampal and cortical neuronal TDP-43 in a mouse expressing familial mutations (K595N and M596L) in APP and presenilin 1 (PSEN1ΔE9). In our model, increased TDP-43 was related to increased tau aggregation as evidenced by thioflavin S-positive phosphorylated tau, which may implicate TDP-43 expression in pre-tangle formation. In addition, there was increased endosomal/lysosomal localization of APP and reduced Aß plaque formation with increased TDP-43. Furthermore, there was decreased calcineurin with elevated TDP-43 expression. Since calcineurin is a phosphatase for TDP-43, the decreased calcineurin expression may be one mechanism leading to an increase in accumulation of diffuse phosphorylated TDP-43 in the hippocampus and cortex. We further show that when TDP-43 is knocked down there is an increase in calcineurin. In our model of selective TDP-43 overexpression in an APP/PSEN1 background, we show that TDP-43 decreases Aß plaque deposition while increasing abnormal tau aggregation. These observations indicate that TDP-43 may play a role in regulating APP trafficking and tau aggregation. Our data suggest that TDP-43 could be a putative target for therapeutic intervention in AD affecting both Aß plaque formation and tauopathy.


Subject(s)
DNA-Binding Proteins/biosynthesis , Plaque, Amyloid/metabolism , Protein Aggregation, Pathological/metabolism , tau Proteins/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Presenilin-1/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , tau Proteins/genetics
10.
Plant Physiol ; 173(4): 2243-2252, 2017 04.
Article in English | MEDLINE | ID: mdl-28202597

ABSTRACT

Epigenetic modifications play critical roles in diverse biological processes. Histone Lys-to-Met (K-to-M) mutations act as gain-of-function mutations to inhibit a wide range of histone methyltransferases and are thought to promote tumorigenesis. However, it is largely unknown whether K-to-M mutations impact organismal development. Using Arabidopsis (Arabidopsis thaliana) as a model system, we discovered that a transgene exogenously expressing histone 3 Lys-36 to Met mutation (K36M) acts in a dominant-negative manner to cause global reduction of H3K36 methylation. Remarkably, this dominant repressive activity is dosage-dependent and causes strong developmental perturbations including extreme branching and early flowering by affecting the expression of genes involved in developmental and metabolic processes. Besides the established pathological roles of K-to-M mutations in tumor cells, we demonstrate a physiological outcome for K-to-M induced H3K36 hypomethylation. This study provides evidence for a conserved dominant-negative inhibitory role of histone K-to-M mutation across the plant and animal kingdoms. We also highlight the unique ability of K36M mutations to alter plant developmental processes leading to severe pleiotropic phenotypes. Finally, our data suggests K-to-M mutations may provide a useful strategy for altering epigenetic landscapes in organisms where histone methyltransferases are uncharacterized.


Subject(s)
Arabidopsis Proteins/genetics , Histones/genetics , Lysine/genetics , Methionine/genetics , Mutation , Amino Acid Substitution , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Epigenesis, Genetic , Flowers/genetics , Flowers/growth & development , Flowers/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Expression Regulation, Plant , Gene Ontology , Genetic Pleiotropy , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Immunoblotting , Lysine/metabolism , Methionine/metabolism , Methylation , Plants, Genetically Modified , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
J Biol Chem ; 291(40): 20827-20837, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27497435

ABSTRACT

Mitochondria are essential organelles that adapt to stress and environmental changes. Among the nutrient signals that affect mitochondrial form and function is iron, whose depletion initiates a rapid and reversible decrease in mitochondrial biogenesis through unclear means. Here we demonstrate that, unlike the canonical iron-induced alterations to transcript stability, loss of iron dampens the transcription of genes encoding mitochondrial proteins with no change to transcript half-life. Using mass spectrometry, we demonstrate that these transcriptional changes are accompanied by dynamic alterations to histone acetylation and methylation levels that are largely reversible upon readministration of iron. Moreover, histone deacetylase inhibition abrogates the decreased histone acetylation observed upon iron deprivation and restores normal transcript levels at genes encoding mitochondrial proteins. Collectively, we demonstrate that deprivation of an essential nutrient induces transcriptional repression of organellar biogenesis involving epigenetic alterations.


Subject(s)
Epigenesis, Genetic , Iron Deficiencies , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/biosynthesis , Transcription, Genetic , Acetylation , Animals , Histones/metabolism , Mice
12.
J Exp Med ; 213(7): 1267-84, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27242167

ABSTRACT

The import of acetyl-CoA into the lumen of the endoplasmic reticulum (ER) by AT-1/SLC33A1 regulates Nε-lysine acetylation of ER-resident and -transiting proteins. Specifically, lysine acetylation within the ER appears to influence the efficiency of the secretory pathway by affecting ER-mediated quality control. Mutations or duplications in AT-1/SLC33A1 have been linked to diseases such as familial spastic paraplegia, developmental delay with premature death, and autism spectrum disorder with intellectual disability. In this study, we generated an AT-1 Tg mouse model that selectively overexpresses human AT-1 in neurons. These animals demonstrate cognitive deficits, autistic-like social behavior, aberrations in synaptic plasticity, an increased number of dendritic spines and branches, and widespread proteomic changes. We also found that AT-1 activity regulates acetyl-CoA flux, causing epigenetic modulation of the histone epitope H3K27 and mitochondrial adaptation. In conclusion, our results indicate that increased expression of AT-1 can cause an autistic-like phenotype by affecting key neuronal metabolic pathways.


Subject(s)
Autism Spectrum Disorder/metabolism , Dendritic Spines/metabolism , Epigenesis, Genetic , Membrane Transport Proteins/biosynthesis , Phenotype , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Dendritic Spines/genetics , Dendritic Spines/pathology , Histones/genetics , Histones/metabolism , Humans , Membrane Transport Proteins/genetics , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology
13.
J Proteome Res ; 14(8): 3252-62, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26120868

ABSTRACT

Histone post-translational modifications (PTMs) are important regulators of chromatin structure and gene expression. Quantitative analysis of histone PTMs by mass spectrometry remains extremely challenging due to the complex and combinatorial nature of histone PTMs. The most commonly used mass spectrometry-based method for high-throughput histone PTM analysis is data-dependent acquisition (DDA). However, stochastic precursor selection and dependence on MS1 ions for quantification impede comprehensive interrogation of histone PTM states using DDA methods. To overcome these limitations, we utilized a data-independent acquisition (DIA) workflow that provides superior run-to-run consistency and postacquisition flexibility in comparison to DDA methods. In addition, we developed a novel DIA-based methodology to quantify isobaric, co-eluting histone peptides that lack unique MS2 transitions. Our method enabled deconvolution and quantification of histone PTMs that are otherwise refractory to quantitation, including the heavily acetylated tail of histone H4. Using this workflow, we investigated the effects of the histone deacetylase inhibitor SAHA (suberoylanilide hydroxamic acid) on the global histone PTM state of human breast cancer MCF7 cells. A total of 62 unique histone PTMs were quantified, revealing novel SAHA-induced changes in acetylation and methylation of histones H3 and H4.


Subject(s)
Histone Code/drug effects , Histones/metabolism , Hydroxamic Acids/pharmacology , Mass Spectrometry/methods , Protein Processing, Post-Translational/drug effects , Acetylation/drug effects , Amino Acid Sequence , Blotting, Western , Chromatography, Liquid , Histone Deacetylase Inhibitors/pharmacology , Humans , MCF-7 Cells , Methylation/drug effects , Molecular Sequence Data , Peptides/metabolism , Proteomics/methods , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Vorinostat
14.
J Biol Chem ; 289(31): 21326-38, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24917678

ABSTRACT

Acetylation of lysine ϵ-amino groups influences many cellular processes and has been mapped to thousands of sites across many organisms. Stoichiometric information of acetylation is essential to accurately interpret biological significance. Here, we developed and employed a novel method for directly quantifying stoichiometry of site-specific acetylation in the entire proteome of Escherichia coli. By coupling isotopic labeling and a novel pairing algorithm, our approach performs an in silico enrichment of acetyl peptides, circumventing the need for immunoenrichment. We investigated the function of the sole NAD(+)-dependent protein deacetylase, CobB, on both site-specific and global acetylation. We quantified 2206 peptides from 899 proteins and observed a wide distribution of acetyl stoichiometry, ranging from less than 1% up to 98%. Bioinformatic analysis revealed that metabolic enzymes, which either utilize or generate acetyl-CoA, and proteins involved in transcriptional and translational processes displayed the highest degree of acetylation. Loss of CobB led to increased global acetylation at low stoichiometry sites and induced site-specific changes at high stoichiometry sites, and biochemical analysis revealed altered acetyl-CoA metabolism. Thus, this study demonstrates that sirtuin deacetylase deficiency leads to both site-specific and global changes in protein acetylation stoichiometry, affecting central metabolism.


Subject(s)
Bacterial Proteins/metabolism , Lysine/metabolism , Proteome , Acetylation , Amino Acid Sequence , Bacterial Proteins/chemistry , Chromatography, High Pressure Liquid , Computational Biology , Escherichia coli/metabolism , Molecular Sequence Data , Tandem Mass Spectrometry
15.
J Neurochem ; 127(6): 772-81, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24047560

ABSTRACT

Fragile X syndrome (FXS) is caused by the loss of functional fragile X mental retardation protein (FMRP). Loss of FMRP results in an elevated basal protein expression profile of FMRP targeted mRNAs, a loss of local metabotropic glutamate receptor (mGluR)-regulated protein synthesis, exaggerated long-term depression and corresponding learning and behavioral deficits. Evidence shows that blocking mGluR signaling in FXS models ameliorates these deficits. Therefore, understanding the signaling mechanisms downstream of mGluR stimulation may provide additional therapeutic targets for FXS. Kinase cascades are an integral mechanism regulating mGluR-dependent protein translation. The c-Jun N-terminal kinase (JNK) pathway has been shown to regulate mGluR-dependent nuclear transcription; however, the involvement of JNK in local, synaptic signaling has not been explored. Here, we show that JNK is both necessary and sufficient for mGluR-dependent expression of a subset of FMRP target proteins. In addition, JNK activity is basally elevated in fmr1 knockout mouse synapses, and blocking JNK activity reduces the over-expression of post-synaptic proteins in these mice. Together, these data suggest that JNK may be an important signaling mechanism downstream of mGluR stimulation, regulating FMRP-dependent protein synthesis. Furthermore, local, post-synaptic dysregulation of JNK activity may provide a viable target to ameliorate the deficits involved in FXS. Expression of many FMRP target proteins is enhanced in FXS. Here, we evaluated the role of JNKs in FXS. We found that JNK signaling is activated upon mGluR stimulation in wild-type neurons. Conversely, JNK activity is basally elevated in fmr1 knockout. Inhibiting JNK reduced the expression of FMRP target proteins and driving JNK activity increased the expression of these proteins.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Dendrites/metabolism , Enzyme Activation , Fragile X Mental Retardation Protein/genetics , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mice , Mice, Knockout , Signal Transduction
16.
PLoS One ; 8(7): e70163, 2013.
Article in English | MEDLINE | ID: mdl-23922950

ABSTRACT

The Nrf2 (NF-E2 related factor 2)-ARE (antioxidant response element) pathway controls a powerful array of endogenous cellular antioxidant systems and is an important pathway in the detoxification of reactive oxygen species (ROS) in the brain. Using a combination of quantitative proteomics and siRNA screening, we have identified novel protective mechanisms of the Nrf2-ARE pathway against oxidative stress in astrocytes. Studies from our lab and others have shown Nrf2 overexpression protects astrocytes from oxidative stress. However, the exact mechanisms by which Nrf2 elicits these effects are unknown. In this study, we show that induction of Nrf2 reduces levels of reactive oxygen species (ROS) produced by various oxidative stressors and results in robust cytoprotection. To identify the enzymes responsible for these effects, we used stable isotope labeling by amino acids in cell culture (SILAC) and quantitative shotgun proteomics to identify 72 Nrf2-regulated proteins in astrocytes. We hypothesized a subset of these proteins might play a critical role in Nrf2 protection. In order to identify these critical proteins, we used bioinformatics to narrow our target list of proteins and then systematically screened each candidate with siRNA to assess the role of each in Nrf2 protection. We screened each target against H2O2, tert-butyl hydroperoxide, and 4-hydroxynonenal and subsequently identified three enzymes-catalase, prostaglandin reductase-1, and peroxiredoxin-6-that are critical for Nrf2-mediated protection in astrocytes.


Subject(s)
Astrocytes/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Proteomics , RNA, Small Interfering/genetics , Alcohol Dehydrogenase/metabolism , Aldehydes/metabolism , Animals , Antioxidant Response Elements , Astrocytes/drug effects , Catalase/metabolism , Gene Expression Regulation , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Mice , Oxidative Stress/genetics , Peroxiredoxin VI/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction , tert-Butylhydroperoxide/metabolism
17.
Methods Mol Biol ; 615: 217-26, 2010.
Article in English | MEDLINE | ID: mdl-20013212

ABSTRACT

Due to the complexity of the mammalian central nervous system, neuropeptidomic studies in mammals often yield very complicated mass spectra that make data analysis difficult. Careful sample preparation and extraction protocols must be employed in order to minimize spectral complexity and enable extraction of useful information on neuropeptides from a given sample. Controlling post-mortem protease activity is essential to simplifying mass spectra and to identifying low-abundance neuropeptides in tissue samples. Post-mortem microwave-irradiation coupled with cryostat dissection has proven to be effective in arresting protease activity to allow detection of endogenous neuropeptides instead of protein degradation products.


Subject(s)
Brain Chemistry , Brain , Mass Spectrometry/methods , Neuropeptides/analysis , Protease Inhibitors/metabolism , Proteomics/methods , Animals , Brain/anatomy & histology , Brain/metabolism , Neuropeptides/genetics , Rats
18.
J Proteome Res ; 8(8): 4135-43, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19469553

ABSTRACT

Astrocytes are important regulators of normal brain function in mammals, including roles in synaptic signaling, synapse formation, and neuronal health and survival. Many of these functions are executed via secreted proteins. To analyze the astrocyte secretome, a combination of shotgun proteomics and bioinformatics was employed to analyze conditioned media from primary murine astrocyte cultures. Both two- and one-dimensional LC-MS/MS were used to analyze astrocyte secreted proteins, resulting in the identification of over 420 proteins. To refine our results, the intracellular protein contaminants were removed in silico using a cytoplasmic control. In additional rounds of refinement, putative secreted proteins were subjected to analysis by SignalP, SecretomeP, and gene ontology analysis, yielding a refined list of 187 secreted proteins. In conclusion, the use of shotgun proteomics combined with multiple rounds of data refinement produced a high quality catalog of astrocyte secreted proteins.


Subject(s)
Astrocytes/metabolism , Computational Biology/methods , Nerve Tissue Proteins/analysis , Peptide Mapping/methods , Proteomics/methods , Animals , Cells, Cultured , Chromatography, Liquid , Computer Simulation , Databases, Protein , Mice , Nerve Tissue Proteins/metabolism , Specimen Handling , Tandem Mass Spectrometry
19.
J Clin Pharmacol ; 49(4): 465-76, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19318696

ABSTRACT

Three open-label studies assessed the safety, tolerability, and pharmacokinetics of intravenous dalbavancin in patients with hepatic or renal impairment, including patients with end-stage renal disease (ESRD) receiving dialysis. In each study, 4 to 10 patients with mild, moderate, or severe impairment and age-, sex-, and weight-matched controls were administered either a single dose (500 or 1000 mg) or 2 doses (1000 mg followed by 500 mg 1 week apart) of dalbavancin. Dalbavancin exposures were not increased due to mild renal impairment. The mean area under the concentration-time curve from time 0 to infinity (AUC0-infinity) values were approximately 50% higher in patients with moderate renal impairment or ESRD and 100% higher in patients with severe renal impairment. Dose adjustment is not considered necessary in patients with mild or moderate renal impairment or for patients with ESRD receiving hemodialysis; however, a lower dose of dalbavancin (750 mg followed 1 week later by 375 mg) may be considered for patients with severe renal impairment (creatinine clearance<30 mL/min). AUC0-infinity values were similar in patients with mild hepatic impairment and were about 27% to 36% lower in patients with moderate to severe hepatic impairment compared with controls. No dosage adjustment is recommended in patients with any degree of hepatic impairment. Dalbavancin was well tolerated in all impairment groups.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Hepatic Insufficiency/metabolism , Kidney Failure, Chronic/metabolism , Renal Insufficiency/metabolism , Teicoplanin/analogs & derivatives , Adult , Aged , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Audiometry , Female , Hearing/drug effects , Humans , Infusions, Intravenous , Male , Middle Aged , Teicoplanin/administration & dosage , Teicoplanin/adverse effects , Teicoplanin/pharmacokinetics , Time Factors
20.
Methods Mol Biol ; 492: 381-93, 2009.
Article in English | MEDLINE | ID: mdl-19241046

ABSTRACT

Because of their great biological significance, neuropeptides are the subject of intensive research. Mass spectrometry (MS) is a highly informative and sensitive method used for detecting and characterizing these compounds. Successful MS analysis of neuropeptides is dependent on careful sample preparation. Herein, we present two common sample preparation strategies: direct tissue analysis and pooled tissue extraction coupled with fractionation.


Subject(s)
Mass Spectrometry/methods , Neuropeptides/analysis , Neuropeptides/chemistry , Acetic Acid/chemistry , Amino Acid Sequence , Animals , Chemical Fractionation , Chromatography, High Pressure Liquid , Methanol/chemistry , Neuropeptides/isolation & purification , Organ Specificity , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...