26 results on '"Ribich S"'
Search Results
2. Characterization of IS1676 from Rhodococcus erythropolis SQ1
- Author
-
Lessard, P. A., O'Brien, X. M., Ahlgren, N. A., Ribich, S. A., and Sinskey, A. J.
- Published
- 1999
- Full Text
- View/download PDF
3. INTERIM REPORT FROM A PHASE 2 MULTICENTER STUDY OF TAZEMETOSTAT, AN EZH2 INHIBITOR, IN PATIENTS WITH RELAPSED OR REFRACTORY B-CELL NON-HODGKIN LYMPHOMAS
- Author
-
Morschhauser, F., primary, Salles, G., additional, McKay, P., additional, Tilly, H., additional, Schmitt, A., additional, Gerecitano, J., additional, Johnson, P., additional, Le Gouill, S., additional, Dickinson, M.J., additional, Fruchart, C., additional, Lamy, T., additional, Chaidos, A., additional, Jurczak, W., additional, Opat, S., additional, Radford, J., additional, Zinzani, P.L., additional, Assouline, S., additional, Cartron, G., additional, Clawson, A., additional, Picazio, N., additional, Ribich, S., additional, Blakemore, S.J., additional, Larus, J., additional, Miao, H., additional, Ho, P.T., additional, and Ribrag, V., additional
- Published
- 2017
- Full Text
- View/download PDF
4. A Medium-Throughput Single Cell CRISPR-Cas9 Assay to Assess Gene Essentiality
- Author
-
Grassian, A. R., primary, Scales, T. M. E., additional, Knutson, S. K., additional, Kuntz, K. W., additional, McCarthy, N. J., additional, Lowe, C. E., additional, Moore, J. D., additional, Copeland, R. A., additional, Keilhack, H., additional, Smith, J. J., additional, Wickenden, J. A., additional, and Ribich, S., additional
- Published
- 2015
- Full Text
- View/download PDF
5. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling
- Author
-
Gereben, B., Zavacki, A.M., Ribich, S., Kim, B.W., Huang, S.A., Simonides, W.S., Zeold, A., Bianco, A.C., Physiology, and ICaR - Heartfailure and pulmonary arterial hypertension
- Published
- 2008
6. Characterization of IS 1676 from Rhodococcus erythropolis SQ1
- Author
-
Lessard, P. A., primary, O'Brien, X. M., additional, Ahlgren, N. A., additional, Ribich, S. A., additional, and Sinskey, A. J., additional
- Published
- 1999
- Full Text
- View/download PDF
7. Membrane association and protein conformation of alpha-synuclein in intact neurons. Effect of Parkinson's disease-linked mutations.
- Author
-
McLean, P J, Kawamata, H, Ribich, S, and Hyman, B T
- Abstract
Two missense mutations (Ala-30 --> Pro and Ala-53 --> Thr) in the gene encoding alpha-synuclein are associated with rare autosomal dominant forms of familial Parkinson's disease. In addition, alpha-synuclein is an abundant component of Lewy bodies in sporadic Parkinson's disease and diffuse Lewy body disease. However, the normal conformation of alpha-synuclein, its cellular localization in neurons, and the effects of the mutations remain to be determined. In the present study, we examine these questions using sensitive fluorescence resonance energy transfer techniques. Transient transfection of alpha-synuclein expression constructs into primary cortical neurons and counterstaining with the lipophilic fluorescent marker, DiI, demonstrates a close association between alpha-synuclein and cellular membranes. Both the N- and C-terminal regions of alpha-synuclein are tightly associated with membranes. A weak interaction also occurs between the N and C termini themselves. The Parkinson's disease-associated mutations have no effect on membrane interaction; however, the Ala-30 --> Pro mutation alters the three-dimensional conformation of alpha-synuclein, as measured by significantly increased fluorescence resonance energy transfer between the N and C termini.
- Published
- 2000
8. CDK2 regulates collapsed replication fork repair in CCNE1-amplified ovarian cancer cells via homologous recombination.
- Author
-
Brown VE, Moore SL, Chen M, House N, Ramsden P, Wu HJ, Ribich S, Grassian AR, and Choi YJ
- Abstract
CCNE1 amplification is a common alteration in high-grade serous ovarian cancer and occurs in 15-20% of these tumors. These amplifications are mutually exclusive with homologous recombination deficiency, and, as they have intact homologous recombination, are intrinsically resistant to poly (ADP-ribose) polymerase inhibitors or chemotherapy agents. Understanding the molecular mechanisms that lead to this mutual exclusivity may reveal therapeutic vulnerabilities that could be leveraged in the clinic in this still underserved patient population. Here, we demonstrate that CCNE1 -amplified high-grade serous ovarian cancer cells rely on homologous recombination to repair collapsed replication forks. Cyclin-dependent kinase 2, the canonical partner of cyclin E1, uniquely regulates homologous recombination in this genetic context, and as such cyclin-dependent kinase 2 inhibition synergizes with DNA damaging agents in vitro and in vivo . We demonstrate that combining a selective cyclin-dependent kinase 2 inhibitor with a DNA damaging agent could be a powerful tool in the clinic for high-grade serous ovarian cancer., (© The Author(s) 2023. Published by Oxford University Press on behalf of NAR Cancer.)
- Published
- 2023
- Full Text
- View/download PDF
9. A Mass Spectrometric Assay of METTL3/METTL14 Methyltransferase Activity.
- Author
-
Buker SM, Gurard-Levin ZA, Wheeler BD, Scholle MD, Case AW, Hirsch JL, Ribich S, Copeland RA, and Boriack-Sjodin PA
- Subjects
- Adenosine analogs & derivatives, Adenosine genetics, Adenosine pharmacology, Drug Discovery trends, Gene Expression Regulation, Developmental drug effects, Humans, Methylation drug effects, Multiprotein Complexes antagonists & inhibitors, Multiprotein Complexes genetics, RNA Processing, Post-Transcriptional genetics, RNA Stability drug effects, RNA Stability genetics, RNA, Messenger drug effects, RNA, Messenger genetics, S-Adenosylhomocysteine pharmacology, Mass Spectrometry methods, Methyltransferases genetics, RNA Processing, Post-Transcriptional drug effects
- Abstract
A variety of covalent modifications of RNA have been identified and demonstrated to affect RNA processing, stability, and translation. Methylation of adenosine at the N6 position (m
6 A) in messenger RNA (mRNA) is currently the most well-studied RNA modification and is catalyzed by the RNA methyltransferase complex METTL3/METTL14. Once generated, m6 A can modulate mRNA splicing, export, localization, degradation, and translation. Although potent and selective inhibitors exist for several members of the Type I S -adenosylmethionine (SAM)-dependent methyltransferase family, no inhibitors have been reported for METTL3/METTL14 to date. To facilitate drug discovery efforts, a sensitive and robust mass spectrometry-based assay for METTL3/METTL14 using self-assembled monolayer desorption/ionization (SAMDI) technology has been developed. The assay uses an 11-nucleotide single-stranded RNA compared to a previously reported 27-nucleotide substrate. IC50 values of mechanism-based inhibitors S -adenosylhomocysteine (SAH) and sinefungin (SFG) are comparable between the SAMDI and radiometric assays that use the same substrate. This work demonstrates that SAMDI technology is amenable to RNA substrates and can be used for high-throughput screening and compound characterization for RNA-modifying enzymes.- Published
- 2020
- Full Text
- View/download PDF
10. Small molecule inhibitors and CRISPR/Cas9 mutagenesis demonstrate that SMYD2 and SMYD3 activity are dispensable for autonomous cancer cell proliferation.
- Author
-
Thomenius MJ, Totman J, Harvey D, Mitchell LH, Riera TV, Cosmopoulos K, Grassian AR, Klaus C, Foley M, Admirand EA, Jahic H, Majer C, Wigle T, Jacques SL, Gureasko J, Brach D, Lingaraj T, West K, Smith S, Rioux N, Waters NJ, Tang C, Raimondi A, Munchhof M, Mills JE, Ribich S, Porter Scott M, Kuntz KW, Janzen WP, Moyer M, Smith JJ, Chesworth R, Copeland RA, and Boriack-Sjodin PA
- Subjects
- A549 Cells, Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung pathology, CRISPR-Cas Systems, Carcinogenesis drug effects, Cell Proliferation drug effects, Enzyme Inhibitors pharmacology, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histone-Lysine N-Methyltransferase chemistry, Humans, Methylation drug effects, Methyltransferases antagonists & inhibitors, RNA Interference, Small Molecule Libraries pharmacology, Adenocarcinoma of Lung drug therapy, Carcinogenesis genetics, Histone-Lysine N-Methyltransferase genetics
- Abstract
A key challenge in the development of precision medicine is defining the phenotypic consequences of pharmacological modulation of specific target macromolecules. To address this issue, a variety of genetic, molecular and chemical tools can be used. All of these approaches can produce misleading results if the specificity of the tools is not well understood and the proper controls are not performed. In this paper we illustrate these general themes by providing detailed studies of small molecule inhibitors of the enzymatic activity of two members of the SMYD branch of the protein lysine methyltransferases, SMYD2 and SMYD3. We show that tool compounds as well as CRISPR/Cas9 fail to reproduce many of the cell proliferation findings associated with SMYD2 and SMYD3 inhibition previously obtained with RNAi based approaches and with early stage chemical probes., Competing Interests: This study was funded by Epizyme Inc. All authors are employees of, and/or hold equity in Epizyme, Inc. EPZ028862 and EPZ033294 are products in development and the compounds as well as their use are subject to several U.S. and international patents and patent applications, including but not limited to “Substituted Piperidine Compounds” (PCT/US2015/049235) and “SMYD Inhibitors” (PCT/US2015/049221), both filed on September 9th, 2015. This does not alter the authors’ adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2018
- Full Text
- View/download PDF
11. RNA-modifying proteins as anticancer drug targets.
- Author
-
Boriack-Sjodin PA, Ribich S, and Copeland RA
- Subjects
- Animals, Humans, Methylation drug effects, RNA, Messenger genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Neoplasms genetics, RNA genetics
- Abstract
All major biological macromolecules (DNA, RNA, proteins and lipids) undergo enzyme-catalysed covalent modifications that impact their structure, function and stability. A variety of covalent modifications of RNA have been identified and demonstrated to affect RNA stability and translation to proteins; these mechanisms of translational control have been termed epitranscriptomics. Emerging data suggest that some epitranscriptomic mechanisms are altered in human cancers as well as other human diseases. In this Review, we examine the current understanding of RNA modifications with a focus on mRNA methylation, highlight their possible roles in specific cancer indications and discuss the emerging potential of RNA-modifying proteins as therapeutic targets.
- Published
- 2018
- Full Text
- View/download PDF
12. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study.
- Author
-
Italiano A, Soria JC, Toulmonde M, Michot JM, Lucchesi C, Varga A, Coindre JM, Blakemore SJ, Clawson A, Suttle B, McDonald AA, Woodruff M, Ribich S, Hedrick E, Keilhack H, Thomson B, Owa T, Copeland RA, Ho PTC, and Ribrag V
- Subjects
- Aged, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Benzamides adverse effects, Benzamides pharmacokinetics, Biphenyl Compounds, Dose-Response Relationship, Drug, Enhancer of Zeste Homolog 2 Protein metabolism, Enzyme Inhibitors adverse effects, Enzyme Inhibitors pharmacokinetics, Female, France, Humans, Lymphoma, B-Cell enzymology, Lymphoma, B-Cell pathology, Male, Maximum Tolerated Dose, Middle Aged, Morpholines, Pyridones adverse effects, Pyridones pharmacokinetics, Time Factors, Treatment Outcome, Antineoplastic Agents administration & dosage, Benzamides administration & dosage, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Enzyme Inhibitors administration & dosage, Lymphoma, B-Cell drug therapy, Pyridones administration & dosage
- Abstract
Background: Activating enhancer of zeste homolog 2 (EZH2) mutations or aberrations of the switch/sucrose non-fermentable (SWI/SNF) complex (eg, mutations or deletions of the subunits INI1 or SMARCA4) can lead to aberrant histone methylation, oncogenic transformation, and a proliferative dependency on EZH2 activity. In this first-in-human study, we aimed to investigate the safety, clinical activity, pharmacokinetics, and pharmacodynamics of tazemetostat, a first-in-class selective inhibitor of EZH2., Methods: We did an open-label, multicentre, dose-escalation, phase 1 study using a 3 + 3 design with planned cohort expansion at the two highest doses below the maximally tolerated dose. The study was done at two centres in France: Institut Gustave Roussy (Villejuif, Val de Marne) and Institut Bergonié (Bordeaux, Gironde). Eligible patients had relapsed or refractory B-cell non-Hodgkin lymphoma or an advanced solid tumour and were older than 18 years, with Eastern Cooperative Oncology Group performance status of 0 or 1, and adequate end-organ function. Tazemetostat was administered orally from 100 mg twice daily to 1600 mg twice daily in 28-day cycles. The primary endpoint was to establish the maximum tolerated dose or recommended phase 2 dose of tazemetostat, as determined by dose-limiting toxicities, laboratory values, and other safety or pharmacokinetic measures in cycle one according to local investigator assessment. Safety was assessed in patients who received at least one dose of tazemetostat; antitumour activity was assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT01897571. The phase 1 part of the study is complete, and phase 2 is ongoing., Findings: Between June 13, 2013, and Sept 21, 2016, 64 patients (21 with B-cell non-Hodgkin lymphoma, and 43 with advanced solid tumours) received doses of tazemetostat. The most common treatment-related adverse events, regardless of attribution, were asthenia (21 [33%] of 64 treatment-related events), anaemia (nine [14%]), anorexia (four [6%]), muscle spasms (nine [14%]), nausea (13 [20%]), and vomiting (six [9%]), usually grade 1 or 2 in severity. A single dose-limiting toxicity of grade 4 thrombocytopenia was identified at the highest dose of 1600 mg twice daily. No treatment-related deaths occurred; seven (11%) patients had non-treatment-related deaths (one at 200 mg twice daily, four at 400 mg twice daily, and two at 1600 mg twice daily). The recommended phase 2 dose was determined to be 800 mg twice daily. Durable objective responses, including complete responses, were observed in eight (38%) of 21 patients with B-cell non-Hodgkin lymphoma and two (5%) of 43 patients with solid tumours., Interpretation: Tazemetostat showed a favourable safety profile and antitumour activity in patients with refractory B-cell non-Hodgkin lymphoma and advanced solid tumours, including epithelioid sarcoma. Further clinical investigation of tazemetostat monotherapy is ongoing in phase 2 studies in adults and a phase 1 study for children, which are currently enrolling patients who have B-cell non-Hodgkin lymphoma and INI1-negative or SMARCA4-negative tumours., Funding: Epizyme and Eisai., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
13. Drug Discovery and Chemical Biology of Cancer Epigenetics.
- Author
-
Ribich S, Harvey D, and Copeland RA
- Subjects
- Acetylation drug effects, DNA (Cytosine-5-)-Methyltransferases antagonists & inhibitors, DNA (Cytosine-5-)-Methyltransferases metabolism, Endosomal Sorting Complexes Required for Transport antagonists & inhibitors, Endosomal Sorting Complexes Required for Transport genetics, Endosomal Sorting Complexes Required for Transport metabolism, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Histones metabolism, Humans, Methylation drug effects, Neoplasms drug therapy, Neoplasms genetics, Drug Discovery, Epigenesis, Genetic, Neoplasms pathology
- Abstract
Comprehensive whole-exome sequencing, DNA copy-number determination, and transcriptomic analyses of diverse cancers have greatly expanded our understanding of the biology of many tumor types. In addition to mutations in the common cell-of-origin specific driver mutations, these studies have also revealed a large number of loss-of-function and gain-of-function mutations in chromatin-modifying proteins (CMPs). This has revealed that epigenetic dysregulation is a common feature of most pediatric and adult cancers. Many specific and potent inhibitors have been developed for multiple CMP classes, which have assisted in elucidating the role of epigenetics as well as epigenetic vulnerabilities in these cancer types. Clinical trials with numerous CMP inhibitors are also currently in progress to evaluate the therapeutic potential of epigenetic inhibitors. In this review, we aim to provide a summary of genetic mutations in epigenetic genes and a review of CMP inhibitors suitable for preclinical studies or currently in clinical trials. Additionally, we highlight the CMPs for which potent inhibitors have not been developed and additional research focus should be dedicated., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
14. Chemosensitive Relapse in Small Cell Lung Cancer Proceeds through an EZH2-SLFN11 Axis.
- Author
-
Gardner EE, Lok BH, Schneeberger VE, Desmeules P, Miles LA, Arnold PK, Ni A, Khodos I, de Stanchina E, Nguyen T, Sage J, Campbell JE, Ribich S, Rekhtman N, Dowlati A, Massion PP, Rudin CM, and Poirier JT
- Subjects
- Animals, Drug Resistance, Neoplasm, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Humans, Mice, Nuclear Proteins analysis, Nuclear Proteins genetics, Twist-Related Protein 1 physiology, Enhancer of Zeste Homolog 2 Protein physiology, Lung Neoplasms drug therapy, Nuclear Proteins physiology, Small Cell Lung Carcinoma drug therapy
- Abstract
Small cell lung cancer is initially highly responsive to cisplatin and etoposide but in almost every case becomes rapidly chemoresistant, leading to death within 1 year. We modeled acquired chemoresistance in vivo using a series of patient-derived xenografts to generate paired chemosensitive and chemoresistant cancers. Multiple chemoresistant models demonstrated suppression of SLFN11, a factor implicated in DNA-damage repair deficiency. In vivo silencing of SLFN11 was associated with marked deposition of H3K27me3, a histone modification placed by EZH2, within the gene body of SLFN11, inducing local chromatin condensation and gene silencing. Inclusion of an EZH2 inhibitor with standard cytotoxic therapies prevented emergence of acquired resistance and augmented chemotherapeutic efficacy in both chemosensitive and chemoresistant models of small cell lung cancer., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
15. Correction: Preclinical Evidence of Anti-Tumor Activity Induced by EZH2 Inhibition in Human Models of Synovial Sarcoma.
- Author
-
Kawano S, Grassian AR, Tsuda M, Knutson SK, Warholic NM, Kuznetsov G, Xu S, Xiao Y, Pollock RM, Smith JJ, Kuntz KW, Ribich S, Minoshima Y, Matsui J, Copeland RA, Tanaka S, and Keilhack H
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0158888.].
- Published
- 2017
- Full Text
- View/download PDF
16. Preclinical Evidence of Anti-Tumor Activity Induced by EZH2 Inhibition in Human Models of Synovial Sarcoma.
- Author
-
Kawano S, Grassian AR, Tsuda M, Knutson SK, Warholic NM, Kuznetsov G, Xu S, Xiao Y, Pollock RM, Smith JS, Kuntz KK, Ribich S, Minoshima Y, Matsui J, Copeland RA, Tanaka S, and Keilhack H
- Subjects
- Animals, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, SMARCB1 Protein genetics, SMARCB1 Protein metabolism, Sarcoma, Synovial genetics, Sarcoma, Synovial metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Sarcoma, Synovial drug therapy
- Abstract
The catalytic activities of covalent and ATP-dependent chromatin remodeling are central to regulating the conformational state of chromatin and the resultant transcriptional output. The enzymes that catalyze these activities are often contained within multiprotein complexes in nature. Two such multiprotein complexes, the polycomb repressive complex 2 (PRC2) methyltransferase and the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeler have been reported to act in opposition to each other during development and homeostasis. An imbalance in their activities induced by mutations/deletions in complex members (e.g. SMARCB1) has been suggested to be a pathogenic mechanism in certain human cancers. Here we show that preclinical models of synovial sarcoma-a cancer characterized by functional SMARCB1 loss via its displacement from the SWI/SNF complex through the pathognomonic SS18-SSX fusion protein-display sensitivity to pharmacologic inhibition of EZH2, the catalytic subunit of PRC2. Treatment with tazemetostat, a clinical-stage, selective and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity reverses a subset of synovial sarcoma gene expression and results in concentration-dependent cell growth inhibition and cell death specifically in SS18-SSX fusion-positive cells in vitro. Treatment of mice bearing either a cell line or two patient-derived xenograft models of synovial sarcoma leads to dose-dependent tumor growth inhibition with correlative inhibition of trimethylation levels of the EZH2-specific substrate, lysine 27 on histone H3. These data demonstrate a dependency of SS18-SSX-positive, SMARCB1-deficient synovial sarcomas on EZH2 enzymatic activity and suggests the potential utility of EZH2-targeted drugs in these genetically defined cancers.
- Published
- 2016
- Full Text
- View/download PDF
17. Structural Insights into Ternary Complex Formation of Human CARM1 with Various Substrates.
- Author
-
Boriack-Sjodin PA, Jin L, Jacques SL, Drew A, Sneeringer C, Scott MP, Moyer MP, Ribich S, Moradei O, and Copeland RA
- Subjects
- Binding Sites, Cell Line, Crystallography, X-Ray, Gene Expression Regulation, Enzymologic, Humans, Models, Molecular, Protein Conformation, Protein-Arginine N-Methyltransferases chemistry, Protein-Arginine N-Methyltransferases genetics, Substrate Specificity, Protein-Arginine N-Methyltransferases metabolism
- Abstract
Coactivator-associated arginine methyltransferase 1 (CARM1) is a protein arginine N-methyltransferase (PRMT) enzyme that has been implicated in a variety of cancers. CARM1 is known to methylate histone H3 and nonhistone substrates. To date, several crystal structures of CARM1 have been solved, including structures with small molecule inhibitors, but no ternary structures with nucleoside and peptide substrates have been reported. Here, the crystal structures of human CARM1 with the S-adenosylmethione (SAM) mimic sinefungin and three different peptide sequences from histone H3 and PABP1 are presented, with both nonmethylated and singly methylated arginine residues exemplified. This is the first example of multiple substrate sequences solved in a single PRMT enzyme and demonstrates how the CARM1 binding site is capable of accommodating a variety of peptide sequences while maintaining a core binding mode for the unmethylated and monomethylated substrates. Comparison of these with other PRMT enzyme-peptide structures shows hydrogen bonding patterns that may be thematic of these binding sites.
- Published
- 2016
- Full Text
- View/download PDF
18. Structural and Kinetic Characterization of a Novel N-acetylated Aliphatic Amine Metabolite of the PRMT Inhibitor, EPZ011652.
- Author
-
Rioux N, Mitchell LH, Tiller P, Plant K, Shaw J, Frost K, Ribich S, Moyer MP, Copeland RA, Chesworth R, and Waters NJ
- Subjects
- Animals, Arylamine N-Acetyltransferase metabolism, Bile metabolism, Biotransformation, Dogs, Gas Chromatography-Mass Spectrometry, Hepatocytes metabolism, Humans, Isoenzymes metabolism, Kinetics, Liver metabolism, Male, Metabolic Networks and Pathways, Rats, Rats, Sprague-Dawley, Amines metabolism, Enzyme Inhibitors metabolism, Ethylenediamines metabolism, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Pyrazoles metabolism
- Abstract
Pharmacokinetic and metabolite identification studies were conducted to understand the clearance pathways of EPZ011652 [(2-aminoethyl)(methyl)({3-[4-(propan-2-yloxy)phenyl]-1H-pyrazol-4-yl}methyl)amine], a potent protein arginine N-methyltransferase inhibitor. Metabolic clearance was the major pathway of EPZ011652 elimination in rats with structural elucidation of metabolites via liquid chromatography - mass spectrometry (LC-MS(n)) accurate mass measurement revealing the formation of a novel aliphatic N-acetylated metabolite (M1) located on the terminal nitrogen of the ethylene-diamine side chain. EPZ015564, a synthetic standard of the N-acetyl product, was prepared and was also generated by human and rat, but not dog hepatocytes. In rat hepatocytes, on incubation with EPZ011652, the concentration of EPZ015564 initially increased before decreasing with incubation time, suggesting that the metabolite is itself a substrate for other metabolizing enzymes, in agreement with the identification of metabolites M2, M3, and M4 in rat bile, all N-acetylated metabolites, undergoing sequential phase I (demethylation, oxidation) or phase II (sulfation) reactions. Reaction phenotyping with recombinant human N-acetyltransferase (NAT) isoforms revealed that both NAT1 and NAT2 are capable of acetylating EPZ011652, although with different catalytic efficiencies. Kinetic profiles of EPZ015564 formation followed classic Michaelis-Menten behavior with apparent Km values of >1000 μM for NAT1 and 165 ± 14.1 µM for NAT2. The in vitro intrinsic clearance for EPZ011652 by NAT2 (110 μL/min/mg) was 500-fold greater than by NAT1. In summary, we report the unusual N-acetylation of an aliphatic amine and discuss the implications for drug discovery and clinical development., (Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
19. The chemical chaperones tauroursodeoxycholic and 4-phenylbutyric acid accelerate thyroid hormone activation and energy expenditure.
- Author
-
da-Silva WS, Ribich S, Arrojo e Drigo R, Castillo M, Patti ME, and Bianco AC
- Subjects
- Adipocytes, Brown drug effects, Adipocytes, Brown metabolism, Animals, Cell Line, Cells, Cultured, Dietary Fats adverse effects, Gene Expression Regulation drug effects, Gene Knockout Techniques, Glucose Intolerance prevention & control, Humans, Iodide Peroxidase genetics, Lipid Metabolism drug effects, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Oxygen Consumption drug effects, RNA, Messenger metabolism, Iodothyronine Deiodinase Type II, Energy Metabolism drug effects, Iodide Peroxidase metabolism, Phenylbutyrates pharmacology, Taurochenodeoxycholic Acid pharmacology, Triiodothyronine metabolism
- Abstract
Exposure of cell lines endogenously expressing the thyroid hormone activating enzyme type 2 deiodinase (D2) to the chemical chaperones tauroursodeoxycholic acid (TUDCA) or 4-phenylbutiric acid (4-PBA) increases D2 expression, activity and T3 production. In brown adipocytes, TUDCA or 4-PBA induced T3-dependent genes and oxygen consumption (∼2-fold), an effect partially lost in D2 knockout cells. In wild type, but not in D2 knockout mice, administration of TUDCA lowered the respiratory quotient, doubled brown adipose tissue D2 activity and normalized the glucose intolerance associated with high fat feeding. Thus, D2 plays a critical role in the metabolic effects of chemical chaperones., (Copyright © 2011 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
20. Absence of thyroid hormone activation during development underlies a permanent defect in adaptive thermogenesis.
- Author
-
Hall JA, Ribich S, Christoffolete MA, Simovic G, Correa-Medina M, Patti ME, and Bianco AC
- Subjects
- Acclimatization genetics, Acclimatization physiology, Adipocytes cytology, Adipocytes metabolism, Adipogenesis genetics, Adipogenesis physiology, Adipose Tissue, Brown embryology, Adipose Tissue, Brown growth & development, Animals, Blotting, Western, Cell Differentiation genetics, Cell Differentiation physiology, Cells, Cultured, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Embryo, Mammalian physiology, Female, Gene Expression Regulation, Developmental, Iodide Peroxidase genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Oxygen Consumption genetics, Oxygen Consumption physiology, Reverse Transcriptase Polymerase Chain Reaction, Temperature, Thermogenesis genetics, Thyroid Hormones blood, Time Factors, Iodothyronine Deiodinase Type II, Adipose Tissue, Brown metabolism, Iodide Peroxidase metabolism, Thermogenesis physiology, Thyroid Hormones metabolism
- Abstract
Type 2 deiodinase (D2), which is highly expressed in brown adipose tissue (BAT), is an enzyme that amplifies thyroid hormone signaling in individual cells. Mice with inactivation of the D2 pathway (D2KO) exhibit dramatically impaired thermogenesis in BAT, leading to hypothermia during cold exposure and a greater susceptibility to diet-induced obesity. This was interpreted as a result of defective acute activation of BAT D2. Here we report that the adult D2KO BAT has a permanent thermogenic defect that stems from impaired embryonic BAT development. D2KO embryos have normal serum T3 but due to lack of D2-generated T3 in BAT, this tissue exhibits decreased expression of genes defining BAT identity [i.e. UCP1, PGC-1alpha and Dio2 (nonfunctional)], which results in impaired differentiation and oxidative capacity. Coinciding with a reduction of these T3-responsive genes, there is oxidative stress that in a cell model of brown adipogenesis can be linked to decreased insulin signaling and decreased adipogenesis. This discovery highlights the importance of deiodinase-controlled thyroid hormone signaling in BAT development, where it has important metabolic repercussions for energy homeostasis in adulthood.
- Published
- 2010
- Full Text
- View/download PDF
21. Conserved role of SIRT1 orthologs in fasting-dependent inhibition of the lipid/cholesterol regulator SREBP.
- Author
-
Walker AK, Yang F, Jiang K, Ji JY, Watts JL, Purushotham A, Boss O, Hirsch ML, Ribich S, Smith JJ, Israelian K, Westphal CH, Rodgers JT, Shioda T, Elson SL, Mulligan P, Najafi-Shoushtari H, Black JC, Thakur JK, Kadyk LC, Whetstine JR, Mostoslavsky R, Puigserver P, Li X, Dyson NJ, Hart AC, and Näär AM
- Subjects
- Acetylation, Animals, Benzamides pharmacology, Caenorhabditis elegans, Cell Line, Cholesterol biosynthesis, HeLa Cells, Heterocyclic Compounds, 4 or More Rings pharmacology, Humans, Lipids biosynthesis, Mice, Naphthols pharmacology, Niacinamide pharmacology, Protein Stability drug effects, Sirtuins antagonists & inhibitors, Down-Regulation drug effects, Fasting physiology, Sirtuin 1 metabolism, Sterol Regulatory Element Binding Protein 1 metabolism, Sterol Regulatory Element Binding Protein 2 metabolism
- Abstract
The sterol regulatory element-binding protein (SREBP) transcription factor family is a critical regulator of lipid and sterol homeostasis in eukaryotes. In mammals, SREBPs are highly active in the fed state to promote the expression of lipogenic and cholesterogenic genes and facilitate fat storage. During fasting, SREBP-dependent lipid/cholesterol synthesis is rapidly diminished in the mouse liver; however, the mechanism has remained incompletely understood. Moreover, the evolutionary conservation of fasting regulation of SREBP-dependent programs of gene expression and control of lipid homeostasis has been unclear. We demonstrate here a conserved role for orthologs of the NAD(+)-dependent deacetylase SIRT1 in metazoans in down-regulation of SREBP orthologs during fasting, resulting in inhibition of lipid synthesis and fat storage. Our data reveal that SIRT1 can directly deacetylate SREBP, and modulation of SIRT1 activity results in changes in SREBP ubiquitination, protein stability, and target gene expression. In addition, chemical activators of SIRT1 inhibit SREBP target gene expression in vitro and in vivo, correlating with decreased hepatic lipid and cholesterol levels and attenuated liver steatosis in diet-induced and genetically obese mice. We conclude that SIRT1 orthologs play a critical role in controlling SREBP-dependent gene regulation governing lipid/cholesterol homeostasis in metazoans in response to fasting cues. These findings may have important biomedical implications for the treatment of metabolic disorders associated with aberrant lipid/cholesterol homeostasis, including metabolic syndrome and atherosclerosis.
- Published
- 2010
- Full Text
- View/download PDF
22. Mice lacking Pctp /StarD2 exhibit increased adaptive thermogenesis and enlarged mitochondria in brown adipose tissue.
- Author
-
Kang HW, Ribich S, Kim BW, Hagen SJ, Bianco AC, and Cohen DE
- Subjects
- Adaptation, Physiological, Adipocytes cytology, Adipocytes metabolism, Adipose Tissue, Brown cytology, Adipose Tissue, Brown ultrastructure, Animals, Blotting, Western, Body Temperature, Cells, Cultured, Gene Expression Profiling, Lipid Metabolism drug effects, Mice, Mice, Knockout, Microscopy, Electron, Microscopy, Fluorescence, Mitochondria ultrastructure, Norepinephrine pharmacology, Oxygen Consumption, Phospholipid Transfer Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction, Thiolester Hydrolases genetics, Thiolester Hydrolases metabolism, Adipose Tissue, Brown metabolism, Mitochondria metabolism, Phospholipid Transfer Proteins metabolism, Thermogenesis
- Abstract
Pctp(-/-) mice that lack phosphatidylcholine transfer protein (Pctp) exhibit a marked shift toward utilization of fatty acids for oxidative phosphorylation, suggesting that Pctp may regulate the entry of fatty acyl-CoAs into mitochondria. Here, we examined the influence of Pctp expression on the function and structure of brown adipose tissue (BAT), a mitochondrial-rich, oxidative tissue that mediates nonshivering thermogenesis. Consistent with increased thermogenesis, Pctp(-/-) mice exhibited higher core body temperatures than wild-type controls at room temperature. During a 24 h cold challenge, Pctp(-/-) mice defended core body temperature efficiently enough that acute, full activation of BAT thermogenic genes did not occur. Brown adipocytes lacking Pctp harbored enlarged and elongated mitochondria. Consistent with increased fatty acid utilization, brown adipocytes cultured from Pctp(-/-) mice exhibited higher oxygen consumption rates in response to norepinephrine. The absence of Pctp expression during brown adipogenesis in vitro altered the expression of key transcription factors, which could be corrected by adenovirus-mediated overexpression of Pctp early but not late during the differentiation. Collectively, these findings support a key role for Pctp in limiting mitochondrial oxidation of fatty acids and thus regulating adaptive thermogenesis in BAT.
- Published
- 2009
- Full Text
- View/download PDF
23. Type 2 deiodinase expression is induced by peroxisomal proliferator-activated receptor-gamma agonists in skeletal myocytes.
- Author
-
Grozovsky R, Ribich S, Rosene ML, Mulcahey MA, Huang SA, Patti ME, Bianco AC, and Kim BW
- Subjects
- Animals, Cells, Cultured, Chromans pharmacology, Enzyme Activation drug effects, Immunohistochemistry, Insulin pharmacology, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle Fibers, Skeletal metabolism, Myoblasts drug effects, Myoblasts metabolism, Pioglitazone, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Thiazolidinediones pharmacology, Troglitazone, Iodide Peroxidase metabolism, Muscle Fibers, Skeletal drug effects, Muscle Fibers, Skeletal enzymology, PPAR gamma agonists
- Abstract
The thyroid hormone activating type 2 deiodinase (D2) is known to play a role in brown adipose tissue-mediated adaptive thermogenesis in rodents, but the finding of D2 in skeletal muscle raises the possibility of a broader metabolic role. In the current study, we examined the regulation of the D2 pathway in primary skeletal muscle myoblasts taken from both humans and mice. We found that pioglitazone treatment led to a 1.6- to 1.9-fold increase in primary human skeletal myocyte D2 activity; this effect was seen with other peroxisomal proliferator-activated receptor-gamma agonists. D2 activity in primary murine skeletal myotubes increased 2.8-fold in response to 5 microM pioglitazone and 1.6-fold in response to 5 nM insulin and increased in a dose-dependent manner in response to lithocholic acid (maximum response at 25 microM was approximately 3.8-fold). We compared Akt phosphorylation in primary myotubes derived from wild-type and D2 knockout (D2KO) mice: phospho-Akt was reduced by 50% in the D2KO muscle after 1 nM insulin exposure. Expression of T(3)-responsive muscle genes via quantitative RT-PCR suggests that D2KO cells have decreased thyroid hormone signaling, which could contribute to the abnormalities in insulin signaling. D2 activity in skeletal muscle fragments from both murine and human sources was low, on the order of about 0.01 fmol/min . mg of muscle protein. The phenotypic changes seen with D2KO cells support a metabolic role for D2 in muscle, hinting at a D2-mediated linkage between thyroid hormone and insulin signaling, but the low activity calls into question whether skeletal muscle D2 is a major source of plasma T(3).
- Published
- 2009
- Full Text
- View/download PDF
24. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling.
- Author
-
Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, Simonides WS, Zeöld A, and Bianco AC
- Subjects
- Animals, Humans, Signal Transduction, Thyroid Diseases metabolism, Thyroid Gland enzymology, Iodide Peroxidase metabolism, Thyroid Gland metabolism, Thyroxine metabolism, Triiodothyronine metabolism
- Abstract
The iodothyronine deiodinases initiate or terminate thyroid hormone action and therefore are critical for the biological effects mediated by thyroid hormone. Over the years, research has focused on their role in preserving serum levels of the biologically active molecule T(3) during iodine deficiency. More recently, a fascinating new role of these enzymes has been unveiled. The activating deiodinase (D2) and the inactivating deiodinase (D3) can locally increase or decrease thyroid hormone signaling in a tissue- and temporal-specific fashion, independent of changes in thyroid hormone serum concentrations. This mechanism is particularly relevant because deiodinase expression can be modulated by a wide variety of endogenous signaling molecules such as sonic hedgehog, nuclear factor-kappaB, growth factors, bile acids, hypoxia-inducible factor-1alpha, as well as a growing number of xenobiotic substances. In light of these findings, it seems clear that deiodinases play a much broader role than once thought, with great ramifications for the control of thyroid hormone signaling during vertebrate development and metamorphosis, as well as injury response, tissue repair, hypothalamic function, and energy homeostasis in adults.
- Published
- 2008
- Full Text
- View/download PDF
25. An inside job.
- Author
-
Bianco AC, Ribich S, and Kim BW
- Subjects
- Animals, Brain metabolism, Iodide Peroxidase genetics, Mice, Mice, Knockout, Models, Biological, Neuropsychological Tests, Pituitary Gland metabolism, Thyroid Hormones blood, Thyroid Hormones metabolism, Thyroxine blood, Thyroxine metabolism, Thyroxine physiology, Triiodothyronine blood, Triiodothyronine metabolism, Triiodothyronine physiology, Iodothyronine Deiodinase Type II, Iodide Peroxidase metabolism, Thyroid Hormones physiology
- Published
- 2007
- Full Text
- View/download PDF
26. Identification of long-range regulatory elements in the protocadherin-alpha gene cluster.
- Author
-
Ribich S, Tasic B, and Maniatis T
- Subjects
- Animals, Deoxyribonuclease I metabolism, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Gene Expression Regulation, Humans, Mice, RNA, Messenger genetics, Regulatory Sequences, Nucleic Acid, Cadherins genetics, Cadherins metabolism, Multigene Family genetics
- Abstract
The clustered protocadherins (Pcdh) are encoded by three closely linked gene clusters (Pcdh-alpha, -beta, and -gamma) that span nearly 1 million base pairs of DNA. The Pcdh-alpha gene cluster encodes a family of 14 distinct cadherin-like cell surface proteins that are expressed in neurons and are present at synaptic junctions. Individual Pcdh-alpha mRNAs are assembled from one of 14 "variable" (V) exons and three "constant" exons in a process that involves both differential promoter activation and alternative pre-mRNA splicing. In individual neurons, only one (and rarely two) of the Pcdh alpha1-12 promoters is independently and randomly activated on each chromosome. Thus, in most cells, this unusual form of monoallelic expression leads to the expression of two different Pcdh-alpha 1-12 V exons, one from each chromosome. The two remaining V exons in the cluster (Pcdh-alphaC1 and alphaC2) are expressed biallelically in every neuron. The mechanisms that underlie promoter choice and monoallelic expression in the Pcdh-alpha gene cluster are not understood. Here we report the identification of two long-range cis-regulatory elements in the Pcdh-alpha gene cluster, HS5-1 and HS7. We show that HS5-1 is required for maximal levels of expression from the Pcdh alpha1-12 and alphaC1 promoters, but not the Pcdh-alphaC2 promoter. The nearly cluster-wide requirement of the HS5-1 element is consistent with the possibility that the monoallelic expression of Pcdh-alpha V exons is a consequence of competition between individual V exon promoters for the two regulatory elements.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.