56 results on '"Kruger RG"'
Search Results
2. Phase I trials of the lysine-specific demethylase 1 inhibitor, GSK2879552, as mono- and combination-therapy in relapsed/refractory acute myeloid leukemia or high-risk myelodysplastic syndromes
- Author
-
Roboz GJ, Yee K, Verma A, Borthakur G, de la Fuente Burguera A, Sanz G, Mohammad HP, Kruger RG, Karpinich NO, Ferron-Brady G, Acusta A, Del Buono H, Collingwood T, Ballas M, Dhar A, and Wei AH
- Published
- 2022
3. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia
- Author
-
Bell, CC, Fenne, KA, Chan, Y-C, Rambow, F, Yeung, MM, Vassiliadis, D, Lara, L, Yeh, P, Martelotto, LG, Rogiers, A, Kremer, BE, Barbash, O, Mohammad, HP, Johanson, TM, Burr, ML, Dhar, A, Karpinich, N, Tian, L, Tyler, DS, MacPherson, L, Shi, J, Pinnawala, N, Fong, CY, Papenfuss, AT, Grimmond, SM, Dawson, S-J, Allan, RS, Kruger, RG, Vakoc, CR, Goode, DL, Naik, SH, Gilan, O, Lam, EYN, Marine, J-C, Prinjha, RK, Dawson, MA, Bell, CC, Fenne, KA, Chan, Y-C, Rambow, F, Yeung, MM, Vassiliadis, D, Lara, L, Yeh, P, Martelotto, LG, Rogiers, A, Kremer, BE, Barbash, O, Mohammad, HP, Johanson, TM, Burr, ML, Dhar, A, Karpinich, N, Tian, L, Tyler, DS, MacPherson, L, Shi, J, Pinnawala, N, Fong, CY, Papenfuss, AT, Grimmond, SM, Dawson, S-J, Allan, RS, Kruger, RG, Vakoc, CR, Goode, DL, Naik, SH, Gilan, O, Lam, EYN, Marine, J-C, Prinjha, RK, and Dawson, MA
- Abstract
Non-genetic drug resistance is increasingly recognised in various cancers. Molecular insights into this process are lacking and it is unknown whether stable non-genetic resistance can be overcome. Using single cell RNA-sequencing of paired drug naïve and resistant AML patient samples and cellular barcoding in a unique mouse model of non-genetic resistance, here we demonstrate that transcriptional plasticity drives stable epigenetic resistance. With a CRISPR-Cas9 screen we identify regulators of enhancer function as important modulators of the resistant cell state. We show that inhibition of Lsd1 (Kdm1a) is able to overcome stable epigenetic resistance by facilitating the binding of the pioneer factor, Pu.1 and cofactor, Irf8, to nucleate new enhancers that regulate the expression of key survival genes. This enhancer switching results in the re-distribution of transcriptional co-activators, including Brd4, and provides the opportunity to disable their activity and overcome epigenetic resistance. Together these findings highlight key principles to help counteract non-genetic drug resistance.
- Published
- 2019
4. Inhibiting Type I Arginine Methyltransferase Activity Promotes T Cell-Mediated Antitumor Immune Responses.
- Author
-
Fedoriw A, Shi L, O'Brien S, Smitheman KN, Wang Y, Hou J, Sherk C, Rajapurkar S, Laraio J, Williams LJ, Xu C, Han G, Feng Q, Bedford MT, Wang L, Barbash O, Kruger RG, Hwu P, Mohammad HP, and Peng W
- Subjects
- Animals, Arginine, Humans, Immunity, Mice, Intracellular Signaling Peptides and Proteins, Protein-Arginine N-Methyltransferases genetics, Protein-Arginine N-Methyltransferases metabolism
- Abstract
Protein arginine methyltransferases (PRMT) are a widely expressed class of enzymes responsible for catalyzing arginine methylation on numerous protein substrates. Among them, type I PRMTs are responsible for generating asymmetric dimethylarginine. By controlling multiple basic cellular processes, such as DNA damage responses, transcriptional regulation, and mRNA splicing, type I PRMTs contribute to cancer initiation and progression. A type I PRMT inhibitor, GSK3368715, has been developed and has entered clinical trials for solid and hematologic malignancies. Although type I PRMTs have been reported to play roles in modulating immune cell function, the immunologic role of tumor-intrinsic pathways controlled by type I PRMTs remains uncharacterized. Here, our The Cancer Genome Atlas dataset analysis revealed that expression of type I PRMTs associated with poor clinical response and decreased immune infiltration in patients with melanoma. In cancer cell lines, inhibition of type I PRMTs induced an IFN gene signature, amplified responses to IFN and innate immune signaling, and decreased expression of the immunosuppressive cytokine VEGF. In immunocompetent mouse tumor models, including a model of T-cell exclusion that represents a common mechanism of anti-programmed cell death protein 1 (PD-1) resistance in humans, type I PRMT inhibition increased T-cell infiltration, produced durable responses dependent on CD8+ T cells, and enhanced efficacy of anti-PD-1 therapy. These data indicate that type I PRMT inhibition exhibits immunomodulatory properties and synergizes with immune checkpoint blockade (ICB) to induce durable antitumor responses in a T cell-dependent manner, suggesting that type I PRMT inhibition can potentiate an antitumor immunity in refractory settings., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
5. Discovery of a first-in-class reversible DNMT1-selective inhibitor with improved tolerability and efficacy in acute myeloid leukemia.
- Author
-
Pappalardi MB, Keenan K, Cockerill M, Kellner WA, Stowell A, Sherk C, Wong K, Pathuri S, Briand J, Steidel M, Chapman P, Groy A, Wiseman AK, McHugh CF, Campobasso N, Graves AP, Fairweather E, Werner T, Raoof A, Butlin RJ, Rueda L, Horton JR, Fosbenner DT, Zhang C, Handler JL, Muliaditan M, Mebrahtu M, Jaworski JP, McNulty DE, Burt C, Eberl HC, Taylor AN, Ho T, Merrihew S, Foley SW, Rutkowska A, Li M, Romeril SP, Goldberg K, Zhang X, Kershaw CS, Bantscheff M, Jurewicz AJ, Minthorn E, Grandi P, Patel M, Benowitz AB, Mohammad HP, Gilmartin AG, Prinjha RK, Ogilvie D, Carpenter C, Heerding D, Baylin SB, Jones PA, Cheng X, King BW, Luengo JI, Jordan AM, Waddell I, Kruger RG, and McCabe MT
- Subjects
- Animals, DNA metabolism, DNA Methylation, DNA Modification Methylases genetics, Decitabine pharmacology, Mice, Azacitidine pharmacology, Leukemia, Myeloid, Acute drug therapy
- Abstract
DNA methylation, a key epigenetic driver of transcriptional silencing, is universally dysregulated in cancer. Reversal of DNA methylation by hypomethylating agents, such as the cytidine analogs decitabine or azacytidine, has demonstrated clinical benefit in hematologic malignancies. These nucleoside analogs are incorporated into replicating DNA where they inhibit DNA cytosine methyltransferases DNMT1, DNMT3A and DNMT3B through irreversible covalent interactions. These agents induce notable toxicity to normal blood cells thus limiting their clinical doses. Herein we report the discovery of GSK3685032, a potent first-in-class DNMT1-selective inhibitor that was shown via crystallographic studies to compete with the active-site loop of DNMT1 for penetration into hemi-methylated DNA between two CpG base pairs. GSK3685032 induces robust loss of DNA methylation, transcriptional activation and cancer cell growth inhibition in vitro. Due to improved in vivo tolerability compared with decitabine, GSK3685032 yields superior tumor regression and survival mouse models of acute myeloid leukemia., Competing Interests: Competing interests M.B.P., K.K., W.A.K., C.S., K.W., J.B., M.S., A.G., C.F.M., N.C., A.P.G., T.W., L.R., D.T.F., C.Z., J.L.H., M.Muliaditan, M.Mebrahtu, J.P.J., D.E.M., H.C.E., A.N.T., T.H., S.M., S.W.F., A. Rutkowska, M.L., S.P.R., M.B., A.J.J., E.M., P.G., M.P., A.B.B., H.P.M., A.G.G., R.K.P., C.C., D.H., B.W.K., J.I.L., R.G.K. and M.T.M. are/were employees and/or shareholders of GlaxoSmithKline (GSK). The remaining authors declare no competing interests.
- Published
- 2021
6. Fragment-based Scaffold Hopping: Identification of Potent, Selective, and Highly Soluble Bromo and Extra Terminal Domain (BET) Second Bromodomain (BD2) Inhibitors.
- Author
-
Seal JT, Atkinson SJ, Bamborough P, Bassil A, Chung CW, Foley J, Gordon L, Grandi P, Gray JRJ, Harrison LA, Kruger RG, Matteo JJ, McCabe MT, Messenger C, Mitchell D, Phillipou A, Preston A, Prinjha RK, Rianjongdee F, Rioja I, Taylor S, Wall ID, Watson RJ, Woolven JM, Wyce A, Zhang XP, and Demont EH
- Subjects
- Dose-Response Relationship, Drug, Furans chemistry, Humans, Molecular Structure, Proteins metabolism, Pyrazoles chemistry, Structure-Activity Relationship, Furans pharmacology, Proteins antagonists & inhibitors, Pyrazoles pharmacology
- Abstract
The profound efficacy of pan-BET inhibitors is well documented, but these epigenetic agents have shown pharmacology-driven toxicity in oncology clinical trials. The opportunity to identify inhibitors with an improved safety profile by selective targeting of a subset of the eight bromodomains of the BET family has triggered extensive medicinal chemistry efforts. In this article, we disclose the identification of potent and selective drug-like pan-BD2 inhibitors such as pyrazole 23 (GSK809) and furan 24 (GSK743) that were derived from the pyrrole fragment 6 . We transpose the key learnings from a previous pyridone series (GSK620 2 as a representative example) to this novel class of inhibitors, which are characterized by significantly improved solubility relative to our previous research.
- Published
- 2021
- Full Text
- View/download PDF
7. In vitro and in vivo induction of fetal hemoglobin with a reversible and selective DNMT1 inhibitor.
- Author
-
Gilmartin AG, Groy A, Gore ER, Atkins C, Long ER, Montoute MN, Wu Z, Halsey W, McNulty DE, Ennulat D, Rueda L, Pappalardi M, Kruger RG, McCabe MT, Raoof A, Butlin R, Stowell A, Cockerill M, Waddell I, Ogilvie D, Luengo J, Jordan A, and Benowitz AB
- Subjects
- Animals, Azacitidine pharmacology, DNA Methylation, Mice, gamma-Globins genetics, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell genetics, Fetal Hemoglobin genetics
- Abstract
Pharmacological induction of fetal hemoglobin (HbF) expression is an effective therapeutic strategy for the management of beta-hemoglobinopathies such as sickle cell disease. DNA methyltransferase (DNMT) inhibitors 5-azacytidine (5-aza) and 5-aza-2'-deoxycytidine (decitabine) have been shown to induce fetal hemoglobin expression in both preclinical models and clinical studies, but are not currently approved for the management of hemoglobinopathies. We report here the discovery of a novel class of orally bioavailable DNMT1-selective inhibitors as exemplified by GSK3482364. This molecule potently inhibits the methyltransferase activity of DNMT1, but not DNMT family members DNMT3A or DNMT3B. In contrast with cytidine analog DNMT inhibitors, the DNMT1 inhibitory mechanism of GSK3482364 does not require DNA incorporation and is reversible. In cultured human erythroid progenitor cells (EPCs), GSK3482364 decreased overall DNA methylation resulting in de-repression of the gamma globin genes HBG1 and HBG2 and increased HbF expression. In a transgenic mouse model of sickle cell disease, orally administered GSK3482364 caused significant increases in both HbF levels and in the percentage HbF-expressing erythrocytes, with good overall tolerability. We conclude that in these preclinical models, selective, reversible inhibition of DNMT1 is sufficient for the induction of HbF, and is well-tolerated. We anticipate that GSK3482364 will be a useful tool molecule for the further study of selective DNMT1 inhibition both in vitro and in vivo.
- Published
- 2021
- Full Text
- View/download PDF
8. Phase I, Open-Label, Dose-Escalation Study of the Safety, Pharmacokinetics, Pharmacodynamics, and Efficacy of GSK2879552 in Relapsed/Refractory SCLC.
- Author
-
Bauer TM, Besse B, Martinez-Marti A, Trigo JM, Moreno V, Garrido P, Ferron-Brady G, Wu Y, Park J, Collingwood T, Kruger RG, Mohammad HP, Ballas MS, Dhar A, and Govindan R
- Subjects
- Adolescent, Adult, Aged, Benzoates pharmacokinetics, Cyclopropanes pharmacokinetics, Dose-Response Relationship, Drug, Female, Follow-Up Studies, Humans, Lung Neoplasms pathology, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Recurrence, Local pathology, Prognosis, Small Cell Lung Carcinoma pathology, Survival Rate, Tissue Distribution, Young Adult, Benzoates therapeutic use, Cyclopropanes therapeutic use, Drug Resistance, Neoplasm drug effects, Lung Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Salvage Therapy, Small Cell Lung Carcinoma drug therapy
- Abstract
Introduction: This first-time-in-humans study assessed the safety, pharmacokinetics (PK), pharmacodynamics (PD), and clinical activity of GSK2879552 in patients with relapsed or refractory SCLC., Methods: This phase I, multicenter, open-label study (NCT02034123) enrolled patients (≥18 years old) with relapsed or refractory SCLC (after ≥1 platinum-containing chemotherapy or refusal of standard therapy). Part 1 was a dose-escalation study; Part 2 was a dose-expansion study. Dose escalations were based on safety, PK, and PD. The primary end point (Part 1) was to determine the safety, tolerability, and recommended dose and regimen of GSK2879552. Secondary end points were to characterize PK and PD parameters and measure disease control rate at week 16. Part 2 was not conducted., Results: Between February 4, 2014, and April 18, 2017, a total of 29 patients were allocated to one of nine dose cohorts (0.25 mg-3 mg once daily and 3-mg or 4-mg intermittent dosing). In all, 22 patients completed the study; 7 withdrew, primarily owing to adverse events (AEs). Most patients (24 of 29 [83%]) had at least one treatment-related AE, most commonly thrombocytopenia (12 of 29 [41%]). Twelve serious AEs (SAEs) were reported by nine patients; six were considered treatment related, the most common of which was encephalopathy (four SAEs). Three patients died; one death was related to SAEs. PK was characterized by rapid absorption, slow elimination, and a dose-proportional increase in exposure., Conclusions: GSK2879552 is a potent, selective inhibitor of lysine demethylase 1A and has demonstrated favorable PK properties but provided poor disease control and a high AE rate in patients with SCLC. The study was terminated, as the risk-benefit profile did not favor continuation., (Copyright © 2019 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
9. Anti-tumor Activity of the Type I PRMT Inhibitor, GSK3368715, Synergizes with PRMT5 Inhibition through MTAP Loss.
- Author
-
Fedoriw A, Rajapurkar SR, O'Brien S, Gerhart SV, Mitchell LH, Adams ND, Rioux N, Lingaraj T, Ribich SA, Pappalardi MB, Shah N, Laraio J, Liu Y, Butticello M, Carpenter CL, Creasy C, Korenchuk S, McCabe MT, McHugh CF, Nagarajan R, Wagner C, Zappacosta F, Annan R, Concha NO, Thomas RA, Hart TK, Smith JJ, Copeland RA, Moyer MP, Campbell J, Stickland K, Mills J, Jacques-O'Hagan S, Allain C, Johnston D, Raimondi A, Porter Scott M, Waters N, Swinger K, Boriack-Sjodin A, Riera T, Shapiro G, Chesworth R, Prinjha RK, Kruger RG, Barbash O, and Mohammad HP
- Subjects
- Alternative Splicing, Antineoplastic Agents chemistry, Biomarkers, Cell Line, Tumor, Drug Synergism, Enzyme Inhibitors chemistry, Humans, Methylation, Models, Molecular, Molecular Conformation, Molecular Structure, Protein Binding, Protein-Arginine N-Methyltransferases chemistry, Substrate Specificity, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Purine-Nucleoside Phosphorylase deficiency
- Abstract
Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of arginines on proteins. Type I PRMTs and their substrates have been implicated in human cancers, suggesting inhibition of type I PRMTs may offer a therapeutic approach for oncology. The current report describes GSK3368715 (EPZ019997), a potent, reversible type I PRMT inhibitor with anti-tumor effects in human cancer models. Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer cell growth inhibition when combined with GSK3368715. Interestingly, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and correlates with sensitivity to GSK3368715 in cell lines. These data provide rationale to explore MTAP status as a biomarker strategy for patient selection., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
10. Rational Targeting of Cooperating Layers of the Epigenome Yields Enhanced Therapeutic Efficacy against AML.
- Author
-
Duy C, Teater M, Garrett-Bakelman FE, Lee TC, Meydan C, Glass JL, Li M, Hellmuth JC, Mohammad HP, Smitheman KN, Shih AH, Abdel-Wahab O, Tallman MS, Guzman ML, Muench D, Grimes HL, Roboz GJ, Kruger RG, Creasy CL, Paietta EM, Levine RL, Carroll M, and Melnick AM
- Subjects
- Animals, Azacitidine pharmacology, DNA (Cytosine-5-)-Methyltransferase 1 antagonists & inhibitors, DNA Methylation drug effects, DNA-Binding Proteins genetics, Dioxygenases, Enhancer Elements, Genetic, Epigenome, GATA2 Transcription Factor genetics, GATA2 Transcription Factor metabolism, Genes, Tumor Suppressor, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Mice, Mice, Inbred NOD, Mice, SCID, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Promoter Regions, Genetic drug effects, Proto-Oncogene Proteins genetics, Random Allocation, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Histone Demethylases antagonists & inhibitors, Histone Demethylases genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Disruption of epigenetic regulation is a hallmark of acute myeloid leukemia (AML), but epigenetic therapy is complicated by the complexity of the epigenome. Herein, we developed a long-term primary AML ex vivo platform to determine whether targeting different epigenetic layers with 5-azacytidine and LSD1 inhibitors would yield improved efficacy. This combination was most effective in TET2
mut AML, where it extinguished leukemia stem cells and particularly induced genes with both LSD1-bound enhancers and cytosine-methylated promoters. Functional studies indicated that derepression of genes such as GATA2 contributes to drug efficacy. Mechanistically, combination therapy increased enhancer-promoter looping and chromatin-activating marks at the GATA2 locus. CRISPRi of the LSD1-bound enhancer in patient-derived TET2mut AML was associated with dampening of therapeutic GATA2 induction. TET2 knockdown in human hematopoietic stem/progenitor cells induced loss of enhancer 5-hydroxymethylation and facilitated LSD1-mediated enhancer inactivation. Our data provide a basis for rational targeting of cooperating aberrant promoter and enhancer epigenetic marks driven by mutant epigenetic modifiers. SIGNIFICANCE: Somatic mutations of genes encoding epigenetic modifiers are a hallmark of AML and potentially disrupt many components of the epigenome. Our study targets two different epigenetic layers at promoters and enhancers that cooperate to aberrant gene silencing, downstream of the actions of a mutant epigenetic regulator. This article is highlighted in the In This Issue feature, p. 813 ., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
11. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia.
- Author
-
Bell CC, Fennell KA, Chan YC, Rambow F, Yeung MM, Vassiliadis D, Lara L, Yeh P, Martelotto LG, Rogiers A, Kremer BE, Barbash O, Mohammad HP, Johanson TM, Burr ML, Dhar A, Karpinich N, Tian L, Tyler DS, MacPherson L, Shi J, Pinnawala N, Yew Fong C, Papenfuss AT, Grimmond SM, Dawson SJ, Allan RS, Kruger RG, Vakoc CR, Goode DL, Naik SH, Gilan O, Lam EYN, Marine JC, Prinjha RK, and Dawson MA
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Bone Marrow pathology, CRISPR-Cas Systems genetics, Cell Line, Tumor, Epigenesis, Genetic drug effects, Female, HEK293 Cells, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Mice, Mice, Inbred C57BL, Sequence Analysis, RNA, Single-Cell Analysis, Trans-Activators genetics, Trans-Activators metabolism, Transcription, Genetic drug effects, Treatment Outcome, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Gene Expression Regulation, Leukemic drug effects, Leukemia, Myeloid, Acute drug therapy, Trans-Activators antagonists & inhibitors
- Abstract
Non-genetic drug resistance is increasingly recognised in various cancers. Molecular insights into this process are lacking and it is unknown whether stable non-genetic resistance can be overcome. Using single cell RNA-sequencing of paired drug naïve and resistant AML patient samples and cellular barcoding in a unique mouse model of non-genetic resistance, here we demonstrate that transcriptional plasticity drives stable epigenetic resistance. With a CRISPR-Cas9 screen we identify regulators of enhancer function as important modulators of the resistant cell state. We show that inhibition of Lsd1 (Kdm1a) is able to overcome stable epigenetic resistance by facilitating the binding of the pioneer factor, Pu.1 and cofactor, Irf8, to nucleate new enhancers that regulate the expression of key survival genes. This enhancer switching results in the re-distribution of transcriptional co-activators, including Brd4, and provides the opportunity to disable their activity and overcome epigenetic resistance. Together these findings highlight key principles to help counteract non-genetic drug resistance.
- Published
- 2019
- Full Text
- View/download PDF
12. Lysine specific demethylase 1 inactivation enhances differentiation and promotes cytotoxic response when combined with all- trans retinoic acid in acute myeloid leukemia across subtypes.
- Author
-
Smitheman KN, Severson TM, Rajapurkar SR, McCabe MT, Karpinich N, Foley J, Pappalardi MB, Hughes A, Halsey W, Thomas E, Traini C, Federowicz KE, Laraio J, Mobegi F, Ferron-Brady G, Prinjha RK, Carpenter CL, Kruger RG, Wessels L, and Mohammad HP
- Subjects
- Antineoplastic Agents administration & dosage, Apoptosis drug effects, Benzoates pharmacology, Caspases metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cyclopropanes pharmacology, Dose-Response Relationship, Drug, Drug Synergism, Gene Expression Profiling, Gene Expression Regulation, Neoplastic genetics, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Treatment Outcome, Tretinoin administration & dosage, Antineoplastic Agents pharmacology, Cell Differentiation drug effects, Histone Demethylases antagonists & inhibitors, Leukemia, Myeloid, Acute metabolism, Tretinoin pharmacology
- Abstract
Lysine specific demethylase 1 (LSD1) is a histone modifying enzyme that suppresses gene expression through demethylation of lysine 4 on histone H3. The anti-tumor activity of GSK2879552 and GSK-LSD1, potent, selective irreversible inactivators of LSD1, has previously been described. Inhibition of LSD1 results in a cytostatic growth inhibitory effect in a range of acute myeloid leukemia cell lines. To enhance the therapeutic potential of LSD1 inhibition in this disease setting, a combination of LSD1 inhibition and all- trans retinoic acid was explored. All- trans retinoic acid is currently approved for use in acute promyelocytic leukemia in which it promotes differentiation of abnormal blast cells into normal white blood cells. Combined treatment with all- trans retinoic acid and GSK2879552 results in synergistic effects on cell proliferation, markers of differentiation, and, most importantly, cytotoxicity. Ultimately the combination potential for LSD1 inhibition and ATRA will require validation in acute myeloid leukemia patients, and clinical studies to assess this are currently underway., (Copyright© 2019 Ferrata Storti Foundation.)
- Published
- 2019
- Full Text
- View/download PDF
13. CARM1 Is Essential for Myeloid Leukemogenesis but Dispensable for Normal Hematopoiesis.
- Author
-
Greenblatt SM, Man N, Hamard PJ, Asai T, Karl D, Martinez C, Bilbao D, Stathias V, Jermakowicz AM, Duffort S, Tadi M, Blumenthal E, Newman S, Vu L, Xu Y, Liu F, Schurer SC, McCabe MT, Kruger RG, Xu M, Yang FC, Tenen DG, Watts J, Vega F, and Nimer SD
- Published
- 2019
- Full Text
- View/download PDF
14. Histone demethylase LSD1 is required for germinal center formation and BCL6-driven lymphomagenesis.
- Author
-
Hatzi K, Geng H, Doane AS, Meydan C, LaRiviere R, Cardenas M, Duy C, Shen H, Vidal MNC, Baslan T, Mohammad HP, Kruger RG, Shaknovich R, Haberman AM, Inghirami G, Lowe SW, and Melnick AM
- Subjects
- Animals, CRISPR-Cas Systems, Carcinogenesis, DNA, Intergenic genetics, Germinal Center immunology, Histone Demethylases genetics, Hyperplasia, Immunological Synapses genetics, Introns genetics, Lymphoma genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Proto-Oncogene Proteins c-bcl-6 genetics, B-Lymphocytes physiology, Germinal Center pathology, Histone Demethylases metabolism, Lymphoma metabolism, Proto-Oncogene Proteins c-bcl-6 metabolism
- Abstract
Germinal center (GC) B cells feature repression of many gene enhancers to establish their characteristic transcriptome. Here we show that conditional deletion of Lsd1 in GCs significantly impaired GC formation, associated with failure to repress immune synapse genes linked to GC exit, which are also direct targets of the transcriptional repressor BCL6. We found that BCL6 directly binds LSD1 and recruits it primarily to intergenic and intronic enhancers. Conditional deletion of Lsd1 suppressed GC hyperplasia caused by constitutive expression of BCL6 and significantly delayed BCL6-driven lymphomagenesis. Administration of catalytic inhibitors of LSD1 had little effect on GC formation or GC-derived lymphoma cells. Using a CRISPR-Cas9 domain screen, we found instead that the LSD1 Tower domain was critical for dependence on LSD1 in GC-derived B cells. These results indicate an essential role for LSD1 in the humoral immune response, where it modulates enhancer function by forming repression complexes with BCL6.
- Published
- 2019
- Full Text
- View/download PDF
15. CARM1 Is Essential for Myeloid Leukemogenesis but Dispensable for Normal Hematopoiesis.
- Author
-
Greenblatt SM, Man N, Hamard PJ, Asai T, Karl D, Martinez C, Bilbao D, Stathias V, McGrew-Jermacowicz A, Duffort S, Tadi M, Blumenthal E, Newman S, Vu L, Xu Y, Liu F, Schurer SC, McCabe MT, Kruger RG, Xu M, Yang FC, Tenen D, Watts J, Vega F, and Nimer SD
- Published
- 2018
- Full Text
- View/download PDF
16. Activation of the p53-MDM4 regulatory axis defines the anti-tumour response to PRMT5 inhibition through its role in regulating cellular splicing.
- Author
-
Gerhart SV, Kellner WA, Thompson C, Pappalardi MB, Zhang XP, Montes de Oca R, Penebre E, Duncan K, Boriack-Sjodin A, Le B, Majer C, McCabe MT, Carpenter C, Johnson N, Kruger RG, and Barbash O
- Subjects
- Alternative Splicing genetics, Antineoplastic Agents, Arginine analogs & derivatives, Arginine metabolism, Cell Cycle drug effects, Cell Cycle genetics, Cell Cycle Proteins, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Enzyme Inhibitors pharmacology, Humans, Nuclear Proteins genetics, Protein Isoforms genetics, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Proto-Oncogene Proteins genetics, Tumor Suppressor Protein p53 genetics, snRNP Core Proteins metabolism, Nuclear Proteins metabolism, Protein-Arginine N-Methyltransferases metabolism, Proto-Oncogene Proteins metabolism, RNA Splicing genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Evasion of the potent tumour suppressor activity of p53 is one of the hurdles that must be overcome for cancer cells to escape normal regulation of cellular proliferation and survival. In addition to frequent loss of function mutations, p53 wild-type activity can also be suppressed post-translationally through several mechanisms, including the activity of PRMT5. Here we describe broad anti-proliferative activity of potent, selective, reversible inhibitors of protein arginine methyltransferase 5 (PRMT5) including GSK3326595 in human cancer cell lines representing both hematologic and solid malignancies. Interestingly, PRMT5 inhibition activates the p53 pathway via the induction of alternative splicing of MDM4. The MDM4 isoform switch and subsequent p53 activation are critical determinants of the response to PRMT5 inhibition suggesting that the integrity of the p53-MDM4 regulatory axis defines a subset of patients that could benefit from treatment with GSK3326595.
- Published
- 2018
- Full Text
- View/download PDF
17. MEK inhibitors overcome resistance to BET inhibition across a number of solid and hematologic cancers.
- Author
-
Wyce A, Matteo JJ, Foley SW, Felitsky DJ, Rajapurkar SR, Zhang XP, Musso MC, Korenchuk S, Karpinich NO, Keenan KM, Stern M, Mathew LK, McHugh CF, McCabe MT, Tummino PJ, Kruger RG, Carpenter C, and Barbash O
- Abstract
BET inhibitors exhibit broad activity in cancer models, making predictive biomarkers challenging to define. Here we investigate the biomarkers of activity of the clinical BET inhibitor GSK525762 (I-BET; I-BET762) across cancer cell lines and demonstrate that KRAS mutations are novel resistance biomarkers. This finding led us to combine BET with RAS pathway inhibition using MEK inhibitors to overcome resistance, which resulted in synergistic effects on growth and survival in RAS pathway mutant models as well as a subset of cell lines lacking RAS pathway mutations. GSK525762 treatment up-regulated p-ERK1/2 levels in both RAS pathway wild-type and mutant cell lines, suggesting that MEK/ERK pathway activation may also be a mechanism of adaptive BET inhibitor resistance. Importantly, gene expression studies demonstrated that the BET/MEK combination uniquely sustains down-regulation of genes associated with mitosis, leading to prolonged growth arrest that is not observed with either single agent therapy. These studies highlight a potential to enhance the clinical benefit of BET and MEK inhibitors and provide a strong rationale for clinical evaluation of BET/MEK combination therapies in cancer.
- Published
- 2018
- Full Text
- View/download PDF
18. LSD1 inhibition exerts its antileukemic effect by recommissioning PU.1- and C/EBPα-dependent enhancers in AML.
- Author
-
Cusan M, Cai SF, Mohammad HP, Krivtsov A, Chramiec A, Loizou E, Witkin MD, Smitheman KN, Tenen DG, Ye M, Will B, Steidl U, Kruger RG, Levine RL, Rienhoff HY Jr, Koche RP, and Armstrong SA
- Subjects
- Animals, CCAAT-Enhancer-Binding Proteins genetics, Histone Demethylases genetics, Histone Demethylases metabolism, Leukemia, Biphenotypic, Acute genetics, Leukemia, Biphenotypic, Acute metabolism, Leukemia, Biphenotypic, Acute pathology, Mice, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Proto-Oncogene Proteins genetics, Response Elements, Trans-Activators genetics, CCAAT-Enhancer-Binding Proteins metabolism, Enzyme Inhibitors pharmacology, Histone Demethylases antagonists & inhibitors, Leukemia, Biphenotypic, Acute drug therapy, Neoplasm Proteins antagonists & inhibitors, Neoplasms, Experimental drug therapy, Proto-Oncogene Proteins metabolism, Trans-Activators metabolism
- Abstract
Epigenetic regulators are recurrently mutated and aberrantly expressed in acute myeloid leukemia (AML). Targeted therapies designed to inhibit these chromatin-modifying enzymes, such as the histone demethylase lysine-specific demethylase 1 (LSD1) and the histone methyltransferase DOT1L, have been developed as novel treatment modalities for these often refractory diseases. A common feature of many of these targeted agents is their ability to induce myeloid differentiation, suggesting that multiple paths toward a myeloid gene expression program can be engaged to relieve the differentiation blockade that is uniformly seen in AML. We performed a comparative assessment of chromatin dynamics during the treatment of mixed lineage leukemia (MLL)-AF9-driven murine leukemias and MLL-rearranged patient-derived xenografts using 2 distinct but effective differentiation-inducing targeted epigenetic therapies, the LSD1 inhibitor GSK-LSD1 and the DOT1L inhibitor EPZ4777. Intriguingly, GSK-LSD1 treatment caused global gains in chromatin accessibility, whereas treatment with EPZ4777 caused global losses in accessibility. We captured PU.1 and C/EBPα motif signatures at LSD1 inhibitor-induced dynamic sites and chromatin immunoprecipitation coupled with high-throughput sequencing revealed co-occupancy of these myeloid transcription factors at these sites. Functionally, we confirmed that diminished expression of PU.1 or genetic deletion of C/EBPα in MLL-AF9 cells generates resistance of these leukemias to LSD1 inhibition. These findings reveal that pharmacologic inhibition of LSD1 represents a unique path to overcome the differentiation block in AML for therapeutic benefit., (© 2018 by The American Society of Hematology.)
- Published
- 2018
- Full Text
- View/download PDF
19. Identification of a CARM1 Inhibitor with Potent In Vitro and In Vivo Activity in Preclinical Models of Multiple Myeloma.
- Author
-
Drew AE, Moradei O, Jacques SL, Rioux N, Boriack-Sjodin AP, Allain C, Scott MP, Jin L, Raimondi A, Handler JL, Ott HM, Kruger RG, McCabe MT, Sneeringer C, Riera T, Shapiro G, Waters NJ, Mitchell LH, Duncan KW, Moyer MP, Copeland RA, Smith J, Chesworth R, and Ribich SA
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Cell Line, Tumor, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacokinetics, Humans, In Vitro Techniques, Isoxazoles pharmacokinetics, Male, Mice, Neoplasm Transplantation, Pyrimidines pharmacokinetics, Rats, Sprague-Dawley, Spiro Compounds pharmacokinetics, Antineoplastic Agents therapeutic use, CARD Signaling Adaptor Proteins antagonists & inhibitors, Enzyme Inhibitors therapeutic use, Guanylate Cyclase antagonists & inhibitors, Isoxazoles therapeutic use, Multiple Myeloma drug therapy, Pyrimidines therapeutic use, Spiro Compounds therapeutic use
- Abstract
CARM1 is an arginine methyltransferase with diverse histone and non-histone substrates implicated in the regulation of cellular processes including transcriptional co-activation and RNA processing. CARM1 overexpression has been reported in multiple cancer types and has been shown to modulate oncogenic pathways in in vitro studies. Detailed understanding of the mechanism of action of CARM1 in oncogenesis has been limited by a lack of selective tool compounds, particularly for in vivo studies. We describe the identification and characterization of, to our knowledge, the first potent and selective inhibitor of CARM1 that exhibits anti-proliferative effects both in vitro and in vivo and, to our knowledge, the first demonstration of a role for CARM1 in multiple myeloma (MM). EZM2302 (GSK3359088) is an inhibitor of CARM1 enzymatic activity in biochemical assays (IC
50 = 6 nM) with broad selectivity against other histone methyltransferases. Treatment of MM cell lines with EZM2302 leads to inhibition of PABP1 and SMB methylation and cell stasis with IC50 values in the nanomolar range. Oral dosing of EZM2302 demonstrates dose-dependent in vivo CARM1 inhibition and anti-tumor activity in an MM xenograft model. EZM2302 is a validated chemical probe suitable for further understanding the biological role CARM1 plays in cancer and other diseases.- Published
- 2017
- Full Text
- View/download PDF
20. Targeting Histone Methylation in Cancer.
- Author
-
McCabe MT, Mohammad HP, Barbash O, and Kruger RG
- Subjects
- Antimetabolites, Antineoplastic pharmacology, Antimetabolites, Antineoplastic therapeutic use, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Histone Demethylases genetics, Histone Demethylases metabolism, Histone-Lysine N-Methyltransferase genetics, Histones metabolism, Humans, Methylation drug effects, Neoplasms genetics, Protein Processing, Post-Translational drug effects, Epigenesis, Genetic, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histones genetics, Molecular Targeted Therapy methods, Neoplasms drug therapy
- Abstract
Most, if not all, human cancers exhibit altered epigenetic signatures that promote aberrant gene expression that contributes to cellular transformation. Historically, attempts to pharmacologically intervene in this process have focused on DNA methylation and histone acetylation. More recently, genome-wide studies have identified histone and chromatin regulators as one of the most frequently dysregulated functional classes in a wide range of cancer types. These findings have provided numerous potential therapeutic targets including many that affect histone methylation. These include histone lysine methyltransferases such as enhancer of zeste homolog 2 and DOT1L, protein arginine methyltransferases such as protein arginine methyltransferase 5, and histone lysine demethylases such as lysine-specific demethylase 1. This review presents the rationale for targeting histone methylation in oncology and provides an update on a few key targets that are being investigated in the clinic.
- Published
- 2017
- Full Text
- View/download PDF
21. Structure-Based Design of a Novel SMYD3 Inhibitor that Bridges the SAM-and MEKK2-Binding Pockets.
- Author
-
Van Aller GS, Graves AP, Elkins PA, Bonnette WG, McDevitt PJ, Zappacosta F, Annan RS, Dean TW, Su DS, Carpenter CL, Mohammad HP, and Kruger RG
- Subjects
- Binding Sites, Enzyme Inhibitors chemistry, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Humans, MAP Kinase Kinase Kinase 2 metabolism, Mutation, Protein Binding, S-Adenosylmethionine pharmacology, Enzyme Inhibitors pharmacology, Histone-Lysine N-Methyltransferase chemistry, Molecular Docking Simulation
- Abstract
SMYD3 is a lysine methyltransferase overexpressed in colorectal, breast, prostate, and hepatocellular tumors, and has been implicated as an oncogene in human malignancies. Methylation of MEKK2 by SMYD3 is important for regulation of the MEK/ERK pathway, suggesting the possibility of selectively targeting SMYD3 in RAS-driven cancers. Structural and kinetic characterization of SMYD3 was undertaken leading to a co-crystal structure of SMYD3 with a MEKK2-peptide substrate bound, and the observation that SMYD3 follows a partially processive mechanism. These insights allowed for the design of GSK2807, a potent and selective, SAM-competitive inhibitor of SMYD3 (Ki = 14 nM). A high-resolution crystal structure reveals that GSK2807 bridges the gap between the SAM-binding pocket and the substrate lysine tunnel of SMYD3. Taken together, our data demonstrate that small-molecule inhibitors of SMYD3 can be designed to prevent methylation of MEKK2 and these could have potential use as anticancer therapeutics., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
22. Antitumor activity of LSD1 inhibitors in lung cancer.
- Author
-
Mohammad HP and Kruger RG
- Abstract
Epigenetic machinery have become a major focus for new targeted cancer therapies. Our previous report described the discovery and biological activity of a potent, selective, orally bioavailable, irreversible inhibitor of Lysine Demethylase 1 (LSD1), GSK2879552. A proliferation screen of cell lines representing a number of tumor types indicated that small cell lung carcinoma (SCLC) was sensitive to LSD1 inhibition. The SCLC lines that undergo growth inhibition in response to GSK2879552 exhibit DNA hypomethylation of a signature set of probes suggesting this may be used as a predictive biomarker of activity. This targeted mechanism coupled with a novel predictive biomarker make LSD1 inhibition an exciting potential therapy for SCLC.
- Published
- 2016
- Full Text
- View/download PDF
23. Corrigendum: The promise and peril of chemical probes.
- Author
-
Arrowsmith CH, Audia JE, Austin C, Baell J, Bennett J, Blagg J, Bountra C, Brennan PE, Brown PJ, Bunnage ME, Buser-Doepner C, Campbell RM, Carter AJ, Cohen P, Copeland RA, Cravatt B, Dahlin JL, Dhanak D, Edwards AM, Frederiksen M, Frye SV, Gray N, Grimshaw CE, Hepworth D, Howe T, Huber KV, Jin J, Knapp S, Kotz JD, Kruger RG, Lowe D, Mader MM, Marsden B, Mueller-Fahrnow A, Müller S, O'Hagan RC, Overington JP, Owen DR, Rosenberg SH, Ross R, Roth B, Schapira M, Schreiber SL, Shoichet B, Sundström M, Superti-Furga G, Taunton J, Toledo-Sherman L, Walpole C, Walters MA, Willson TM, Workman P, Young RN, and Zuercher WJ
- Published
- 2015
- Full Text
- View/download PDF
24. The promise and peril of chemical probes.
- Author
-
Arrowsmith CH, Audia JE, Austin C, Baell J, Bennett J, Blagg J, Bountra C, Brennan PE, Brown PJ, Bunnage ME, Buser-Doepner C, Campbell RM, Carter AJ, Cohen P, Copeland RA, Cravatt B, Dahlin JL, Dhanak D, Edwards AM, Frederiksen M, Frye SV, Gray N, Grimshaw CE, Hepworth D, Howe T, Huber KV, Jin J, Knapp S, Kotz JD, Kruger RG, Lowe D, Mader MM, Marsden B, Mueller-Fahrnow A, Müller S, O'Hagan RC, Overington JP, Owen DR, Rosenberg SH, Roth B, Ross R, Schapira M, Schreiber SL, Shoichet B, Sundström M, Superti-Furga G, Taunton J, Toledo-Sherman L, Walpole C, Walters MA, Willson TM, Workman P, Young RN, and Zuercher WJ
- Subjects
- Biomedical Research instrumentation, Humans, Intellectual Property, Internet, Molecular Probes pharmacology, Molecular Weight, Sensitivity and Specificity, Small Molecule Libraries pharmacology, Biomedical Research methods, Information Dissemination ethics, Molecular Probes chemistry, Small Molecule Libraries chemistry
- Published
- 2015
- Full Text
- View/download PDF
25. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC.
- Author
-
Mohammad HP, Smitheman KN, Kamat CD, Soong D, Federowicz KE, Van Aller GS, Schneck JL, Carson JD, Liu Y, Butticello M, Bonnette WG, Gorman SA, Degenhardt Y, Bai Y, McCabe MT, Pappalardi MB, Kasparec J, Tian X, McNulty KC, Rouse M, McDevitt P, Ho T, Crouthamel M, Hart TK, Concha NO, McHugh CF, Miller WH, Dhanak D, Tummino PJ, Carpenter CL, Johnson NW, Hann CL, and Kruger RG
- Subjects
- Administration, Oral, Animals, Antineoplastic Agents pharmacology, Benzoates pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Cyclopropanes pharmacology, Enzyme Inhibitors pharmacology, Epigenesis, Genetic drug effects, Gene Expression Regulation, Neoplastic drug effects, Histone Demethylases genetics, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Mice, Molecular Sequence Data, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology, Xenograft Model Antitumor Assays, Antineoplastic Agents administration & dosage, Benzoates administration & dosage, Cyclopropanes administration & dosage, DNA Methylation drug effects, Enzyme Inhibitors administration & dosage, Histone Demethylases antagonists & inhibitors, Lung Neoplasms drug therapy, Small Cell Lung Carcinoma drug therapy
- Abstract
Epigenetic dysregulation has emerged as an important mechanism in cancer. Alterations in epigenetic machinery have become a major focus for targeted therapies. The current report describes the discovery and biological activity of a cyclopropylamine containing inhibitor of Lysine Demethylase 1 (LSD1), GSK2879552. This small molecule is a potent, selective, orally bioavailable, mechanism-based irreversible inactivator of LSD1. A proliferation screen of cell lines representing a number of tumor types indicated that small cell lung carcinoma (SCLC) is sensitive to LSD1 inhibition. The subset of SCLC lines and primary samples that undergo growth inhibition in response to GSK2879552 exhibit DNA hypomethylation of a signature set of probes, suggesting this may be used as a predictive biomarker of activity., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
26. A687V EZH2 is a driver of histone H3 lysine 27 (H3K27) hypertrimethylation.
- Author
-
Ott HM, Graves AP, Pappalardi MB, Huddleston M, Halsey WS, Hughes AM, Groy A, Dul E, Jiang Y, Bai Y, Annan R, Verma SK, Knight SD, Kruger RG, Dhanak D, Schwartz B, Tummino PJ, Creasy CL, and McCabe MT
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Binding Sites, Cell Line, Tumor, Cluster Analysis, Enhancer of Zeste Homolog 2 Protein, Gene Expression, Gene Expression Profiling, Gene Silencing, Heterozygote, Humans, Lysine metabolism, Methylation, Models, Molecular, Molecular Sequence Data, Polycomb Repressive Complex 2 chemistry, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Protein Conformation, Sequence Alignment, Substrate Specificity, Transcriptional Activation, Histones metabolism, Mutation, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism
- Abstract
The EZH2 methyltransferase silences gene expression through methylation of histone H3 on lysine 27 (H3K27). Recently, EZH2 mutations have been reported at Y641, A677, and A687 in non-Hodgkin lymphoma. Although the Y641F/N/S/H/C and A677G mutations exhibit clearly increased activity with substrates dimethylated at lysine 27 (H3K27me2), the A687V mutant has been shown to prefer a monomethylated lysine 27 (H3K27me1) with little gain of activity toward H3K27me2. Herein, we demonstrate that despite this unique substrate preference, A687V EZH2 still drives increased H3K27me3 when transiently expressed in cells. However, unlike the previously described mutants that dramatically deplete global H3K27me2 levels, A687V EZH2 retains normal levels of H3K27me2. Sequencing of B-cell-derived cancer cell lines identified an acute lymphoblastic leukemia cell line harboring this mutation. Similar to exogenous expression of A687V EZH2, this cell line exhibited elevated H3K27me3 while possessing H3K27me2 levels higher than Y641- or A677-mutant lines. Treatment of A687V EZH2-mutant cells with GSK126, a selective EZH2 inhibitor, was associated with a global decrease in H3K27me3, robust gene activation, caspase activation, and decreased proliferation. Structural modeling of the A687V EZH2 active site suggests that the increased catalytic activity with H3K27me1 may be due to a weakened interaction with an active site water molecule that must be displaced for dimethylation to occur. These findings suggest that A687V EZH2 likely increases global H3K27me3 indirectly through increased catalytic activity with H3K27me1 and cells harboring this mutation are highly dependent on EZH2 activity for their survival., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
27. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer.
- Author
-
Mazur PK, Reynoird N, Khatri P, Jansen PW, Wilkinson AW, Liu S, Barbash O, Van Aller GS, Huddleston M, Dhanak D, Tummino PJ, Kruger RG, Garcia BA, Butte AJ, Vermeulen M, Sage J, and Gozani O
- Subjects
- Adenocarcinoma enzymology, Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma pathology, Adenocarcinoma of Lung, Animals, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Disease Models, Animal, Humans, Lung Neoplasms enzymology, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, MAP Kinase Kinase Kinase 2 chemistry, MAP Kinase Kinase Kinases chemistry, Methylation, Mice, Mitogen-Activated Protein Kinases metabolism, Oncogene Protein p21(ras) genetics, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Protein Phosphatase 2 antagonists & inhibitors, Protein Phosphatase 2 metabolism, Proto-Oncogene Proteins A-raf metabolism, Signal Transduction, Cell Transformation, Neoplastic metabolism, Histone-Lysine N-Methyltransferase metabolism, Lysine metabolism, MAP Kinase Kinase Kinase 2 metabolism, MAP Kinase Kinase Kinases metabolism, Oncogene Protein p21(ras) metabolism
- Abstract
Deregulation of lysine methylation signalling has emerged as a common aetiological factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain containing protein 3) is overexpressed in numerous human tumours. However, the molecular mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signalling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumour development in response to oncogenic Ras. We used protein array technology to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signalling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key negative regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signalling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signalling.
- Published
- 2014
- Full Text
- View/download PDF
28. Long residence time inhibition of EZH2 in activated polycomb repressive complex 2.
- Author
-
Van Aller GS, Pappalardi MB, Ott HM, Diaz E, Brandt M, Schwartz BJ, Miller WH, Dhanak D, McCabe MT, Verma SK, Creasy CL, Tummino PJ, and Kruger RG
- Subjects
- Allosteric Regulation, Allosteric Site, Binding, Competitive, Dose-Response Relationship, Drug, Enhancer of Zeste Homolog 2 Protein, Enzyme Inhibitors chemistry, HeLa Cells, Humans, Indoles chemistry, Methylation, Nucleosomes drug effects, Nucleosomes enzymology, Point Mutation, Polycomb Repressive Complex 2 genetics, Protein Binding, Pyridones chemistry, Structure-Activity Relationship, Substrate Specificity, Time Factors, Enzyme Inhibitors pharmacology, Indoles pharmacology, Polycomb Repressive Complex 2 antagonists & inhibitors, Pyridones pharmacology
- Abstract
EZH2/PRC2 catalyzes transcriptionally repressive methylation at lysine 27 of histone H3 and has been associated with numerous cancer types. Point mutations in EZH2 at Tyr641 and Ala677 identified in non-Hodgkin lymphomas alter substrate specificity and result in increased trimethylation at histone H3K27. Interestingly, EZH2/PRC2 is activated by binding H3K27me3 marks on histones, and this activation is proposed as a mechanism for self-propagation of gene silencing. Recent work has identified GSK126 as a potent, selective, SAM-competitive inhibitor of EZH2 capable of globally decreasing H3K27 trimethylation in cells. Here we show that activation of PRC2 by an H3 peptide trimethylated at K27 is primarily an effect on the rate-limiting step (kcat) with no effect on substrate binding (Km). Additionally, GSK126 is shown to have a significantly longer residence time of inhibition on the activated form of EZH2/PRC2 as compared to unactivated EZH2/PRC2. Overall inhibition constant (Ki*) values for GSK126 were determined to be as low as 93 pM and appear to be driven by slow dissociation of inhibitor from the activated enzyme. The data suggest that activation of EZH2 allows the enzyme to adopt a conformation that possesses greater affinity for GSK126. The long residence time of GSK126 may be beneficial in vivo and may result in durable target inhibition after drug systemic clearance.
- Published
- 2014
- Full Text
- View/download PDF
29. EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation.
- Author
-
Béguelin W, Popovic R, Teater M, Jiang Y, Bunting KL, Rosen M, Shen H, Yang SN, Wang L, Ezponda T, Martinez-Garcia E, Zhang H, Zheng Y, Verma SK, McCabe MT, Ott HM, Van Aller GS, Kruger RG, Liu Y, McHugh CF, Scott DW, Chung YR, Kelleher N, Shaknovich R, Creasy CL, Gascoyne RD, Wong KK, Cerchietti L, Levine RL, Abdel-Wahab O, Licht JD, Elemento O, and Melnick AM
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Enhancer of Zeste Homolog 2 Protein, Gene Deletion, Gene Expression Regulation, Neoplastic, Germinal Center drug effects, Histones metabolism, Methylation, Mice, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Promoter Regions, Genetic, Proto-Oncogene Proteins c-bcl-2 metabolism, Proto-Oncogene Proteins c-bcl-2 physiology, B-Lymphocytes metabolism, Cell Transformation, Neoplastic genetics, Germinal Center metabolism, Mutation, Polycomb Repressive Complex 2 physiology
- Abstract
The EZH2 histone methyltransferase is highly expressed in germinal center (GC) B cells and targeted by somatic mutations in B cell lymphomas. Here, we find that EZH2 deletion or pharmacologic inhibition suppresses GC formation and functions. EZH2 represses proliferation checkpoint genes and helps establish bivalent chromatin domains at key regulatory loci to transiently suppress GC B cell differentiation. Somatic mutations reinforce these physiological effects through enhanced silencing of EZH2 targets. Conditional expression of mutant EZH2 in mice induces GC hyperplasia and accelerated lymphomagenesis in cooperation with BCL2. GC B cell (GCB)-type diffuse large B cell lymphomas (DLBCLs) are mostly addicted to EZH2 but not the more differentiated activated B cell (ABC)-type DLBCLs, thus clarifying the therapeutic scope of EZH2 targeting., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
30. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations.
- Author
-
McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, Liu Y, Graves AP, Della Pietra A 3rd, Diaz E, LaFrance LV, Mellinger M, Duquenne C, Tian X, Kruger RG, McHugh CF, Brandt M, Miller WH, Dhanak D, Verma SK, Tummino PJ, and Creasy CL
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Enhancer of Zeste Homolog 2 Protein, Gene Expression Regulation, Neoplastic drug effects, Gene Silencing drug effects, Histone Methyltransferases, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Histones chemistry, Histones metabolism, Humans, Lymphoma, Follicular enzymology, Lymphoma, Follicular genetics, Lymphoma, Follicular pathology, Lymphoma, Large B-Cell, Diffuse enzymology, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Methylation drug effects, Mice, Neoplasm Transplantation, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Repressor Proteins chemistry, Repressor Proteins metabolism, Transcriptional Activation drug effects, Transplantation, Heterologous, Indoles pharmacology, Indoles therapeutic use, Lymphoma, Follicular drug therapy, Lymphoma, Large B-Cell, Diffuse drug therapy, Mutation genetics, Polycomb Repressive Complex 2 antagonists & inhibitors, Pyridones pharmacology, Pyridones therapeutic use
- Abstract
In eukaryotes, post-translational modification of histones is critical for regulation of chromatin structure and gene expression. EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2) and is involved in repressing gene expression through methylation of histone H3 on lysine 27 (H3K27). EZH2 overexpression is implicated in tumorigenesis and correlates with poor prognosis in several tumour types. Additionally, somatic heterozygous mutations of Y641 and A677 residues within the catalytic SET domain of EZH2 occur in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma. The Y641 residue is the most frequently mutated residue, with up to 22% of germinal centre B-cell DLBCL and follicular lymphoma harbouring mutations at this site. These lymphomas have increased H3K27 tri-methylation (H3K27me3) owing to altered substrate preferences of the mutant enzymes. However, it is unknown whether specific, direct inhibition of EZH2 methyltransferase activity will be effective in treating EZH2 mutant lymphomas. Here we demonstrate that GSK126, a potent, highly selective, S-adenosyl-methionine-competitive, small-molecule inhibitor of EZH2 methyltransferase activity, decreases global H3K27me3 levels and reactivates silenced PRC2 target genes. GSK126 effectively inhibits the proliferation of EZH2 mutant DLBCL cell lines and markedly inhibits the growth of EZH2 mutant DLBCL xenografts in mice. Together, these data demonstrate that pharmacological inhibition of EZH2 activity may provide a promising treatment for EZH2 mutant lymphoma.
- Published
- 2012
- Full Text
- View/download PDF
31. Development and validation of reagents and assays for EZH2 peptide and nucleosome high-throughput screens.
- Author
-
Diaz E, Machutta CA, Chen S, Jiang Y, Nixon C, Hofmann G, Key D, Sweitzer S, Patel M, Wu Z, Creasy CL, Kruger RG, LaFrance L, Verma SK, Pappalardi MB, Le B, Van Aller GS, McCabe MT, Tummino PJ, Pope AJ, Thrall SH, Schwartz B, and Brandt M
- Subjects
- Drug Screening Assays, Antitumor methods, Enhancer of Zeste Homolog 2 Protein, Humans, Indicators and Reagents, Kinetics, Peptides antagonists & inhibitors, Polycomb Repressive Complex 2 antagonists & inhibitors, Polycomb Repressive Complex 2 chemistry, Reproducibility of Results, High-Throughput Screening Assays methods, Nucleosomes metabolism, Peptides metabolism, Polycomb Repressive Complex 2 metabolism
- Abstract
Histone methyltransferases (HMT) catalyze the methylation of histone tail lysines, resulting in changes in gene transcription. Misregulation of these enzymes has been associated with various forms of cancer, making this target class a potential new area for the development of novel chemotherapeutics. EZH2 is the catalytic component of the polycomb group repressive complex (PRC2), which selectively methylates histone H3 lysine 27 (H3K27). EZH2 is overexpressed in prostate, breast, bladder, brain, and other tumor types and is recognized as a molecular marker for cancer progression and aggressiveness. Several new reagents and assays were developed to aid in the identification of EZH2 inhibitors, and these were used to execute two high-throughput screening campaigns. Activity assays using either an H3K27 peptide or nucleosomes as substrates for methylation are described. The strategy to screen EZH2 with either a surrogate peptide or a natural substrate led to the identification of the same tractable series. Compounds from this series are reversible, are [(3)H]-S-adenosyl-L-methionine competitive, and display biochemical inhibition of H3K27 methylation.
- Published
- 2012
- Full Text
- View/download PDF
32. Identification of Potent, Selective, Cell-Active Inhibitors of the Histone Lysine Methyltransferase EZH2.
- Author
-
Verma SK, Tian X, LaFrance LV, Duquenne C, Suarez DP, Newlander KA, Romeril SP, Burgess JL, Grant SW, Brackley JA, Graves AP, Scherzer DA, Shu A, Thompson C, Ott HM, Aller GS, Machutta CA, Diaz E, Jiang Y, Johnson NW, Knight SD, Kruger RG, McCabe MT, Dhanak D, Tummino PJ, Creasy CL, and Miller WH
- Abstract
The histone H3-lysine 27 (H3K27) methyltransferase EZH2 plays a critical role in regulating gene expression, and its aberrant activity is linked to the onset and progression of cancer. As part of a drug discovery program targeting EZH2, we have identified highly potent, selective, SAM-competitive, and cell-active EZH2 inhibitors, including GSK926 (3) and GSK343 (6). These compounds are small molecule chemical tools that would be useful to further explore the biology of EZH2.
- Published
- 2012
- Full Text
- View/download PDF
33. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation.
- Author
-
Van Aller GS, Reynoird N, Barbash O, Huddleston M, Liu S, Zmoos AF, McDevitt P, Sinnamon R, Le B, Mas G, Annan R, Sage J, Garcia BA, Tummino PJ, Gozani O, and Kruger RG
- Subjects
- Animals, Blotting, Western, Chromatin genetics, Chromatin metabolism, Enzyme Activation, Fibroblasts metabolism, Fibroblasts pathology, Genetic Complementation Test, HeLa Cells, Histone-Lysine N-Methyltransferase genetics, Histones genetics, Humans, Methylation, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutagenesis, Site-Directed, Peptide Library, Phenotype, Plasmids genetics, Plasmids metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Recombinant Proteins genetics, Recombinant Proteins metabolism, Substrate Specificity, Gene Expression Regulation, Neoplastic, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Lysine metabolism
- Abstract
Smyd3 is a lysine methyltransferase implicated in chromatin and cancer regulation. Here we show that Smyd3 catalyzes histone H4 methylation at lysine 5 (H4K5me). This novel histone methylation mark is detected in diverse cell types and its formation is attenuated by depletion of Smyd3 protein. Further, Smyd3-driven cancer cell phenotypes require its enzymatic activity. Thus, Smyd3, via H4K5 methylation, provides a potential new link between chromatin dynamics and neoplastic disease.
- Published
- 2012
- Full Text
- View/download PDF
34. Mutation of A677 in histone methyltransferase EZH2 in human B-cell lymphoma promotes hypertrimethylation of histone H3 on lysine 27 (H3K27).
- Author
-
McCabe MT, Graves AP, Ganji G, Diaz E, Halsey WS, Jiang Y, Smitheman KN, Ott HM, Pappalardi MB, Allen KE, Chen SB, Della Pietra A 3rd, Dul E, Hughes AM, Gilbert SA, Thrall SH, Tummino PJ, Kruger RG, Brandt M, Schwartz B, and Creasy CL
- Subjects
- Amino Acid Sequence, Base Sequence, Binding Sites, Cell Line, Tumor, DNA Mutational Analysis, DNA-Binding Proteins chemistry, DNA-Binding Proteins metabolism, Enhancer of Zeste Homolog 2 Protein, Gene Expression Regulation, Neoplastic, Glycine genetics, Heterozygote, Histone Methyltransferases, Histone-Lysine N-Methyltransferase chemistry, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Humans, Methylation, Molecular Sequence Data, Mutant Proteins chemistry, Mutant Proteins metabolism, Polycomb Repressive Complex 2, Substrate Specificity, Transcription Factors chemistry, Transcription Factors metabolism, Alanine genetics, DNA-Binding Proteins genetics, Histones metabolism, Lymphoma, B-Cell enzymology, Lymphoma, B-Cell genetics, Lysine metabolism, Mutation genetics, Transcription Factors genetics
- Abstract
Trimethylation of histone H3 on lysine 27 (H3K27me3) is a repressive posttranslational modification mediated by the histone methyltransferase EZH2. EZH2 is a component of the polycomb repressive complex 2 and is overexpressed in many cancers. In B-cell lymphomas, its substrate preference is frequently altered through somatic mutation of the EZH2 Y641 residue. Herein, we identify mutation of EZH2 A677 to a glycine (A677G) among lymphoma cell lines and primary tumor specimens. Similar to Y641 mutant cell lines, an A677G mutant cell line revealed aberrantly elevated H3K27me3 and decreased monomethylated H3K27 (H3K27me1) and dimethylated H3K27 (H3K27me2). A677G EZH2 possessed catalytic activity with a substrate specificity that was distinct from those of both WT EZH2 and Y641 mutants. Whereas WT EZH2 displayed a preference for substrates with less methylation [unmethylated H3K27 (H3K27me0):me1:me2 k(cat)/K(m) ratio = 9:6:1] and Y641 mutants preferred substrates with greater methylation (H3K27me0:me1:me2 k(cat)/K(m) ratio = 1:2:13), the A677G EZH2 demonstrated nearly equal efficiency for all three substrates (H3K27me0:me1:me2 k(cat)/K(m) ratio = 1.1:0.6:1). When transiently expressed in cells, A677G EZH2, but not WT EZH2, increased global H3K27me3 and decreased H3K27me2. Structural modeling of WT and mutant EZH2 suggested that the A677G mutation acquires the ability to methylate H3K27me2 through enlargement of the lysine tunnel while preserving activity with H3K27me0/me1 substrates through retention of the Y641 residue that is crucial for orientation of these smaller substrates. This mutation highlights the interplay between Y641 and A677 residues in the substrate specificity of EZH2 and identifies another lymphoma patient population that harbors an activating mutation of EZH2.
- Published
- 2012
- Full Text
- View/download PDF
35. GSK1838705A inhibits the insulin-like growth factor-1 receptor and anaplastic lymphoma kinase and shows antitumor activity in experimental models of human cancers.
- Author
-
Sabbatini P, Korenchuk S, Rowand JL, Groy A, Liu Q, Leperi D, Atkins C, Dumble M, Yang J, Anderson K, Kruger RG, Gontarek RR, Maksimchuk KR, Suravajjala S, Lapierre RR, Shotwell JB, Wilson JW, Chamberlain SD, Rabindran SK, and Kumar R
- Subjects
- Anaplastic Lymphoma Kinase, Animals, Blood Glucose metabolism, Cell Proliferation drug effects, Enzyme Activation drug effects, Humans, Mice, Phosphorylation drug effects, Protein-Tyrosine Kinases metabolism, Receptor Protein-Tyrosine Kinases, Receptor, IGF Type 1 metabolism, Receptor, Insulin metabolism, Signal Transduction drug effects, Antineoplastic Agents pharmacology, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Pyrimidines pharmacology, Pyrroles pharmacology, Receptor, IGF Type 1 antagonists & inhibitors, Xenograft Model Antitumor Assays
- Abstract
The insulin-like growth factor-I receptor (IGF-IR) signaling pathway is activated in various tumors, and inhibition of IGF-IR kinase provides a therapeutic opportunity in these patients. GSK1838705A is a small-molecule kinase inhibitor that inhibits IGF-IR and the insulin receptor with IC(50)s of 2.0 and 1.6 nmol/L, respectively. GSK1838705A blocks the in vitro proliferation of cell lines derived from solid and hematologic malignancies, including multiple myeloma and Ewing's sarcoma, and retards the growth of human tumor xenografts in vivo. Despite the inhibitory effect of GSK1838705A on insulin receptor, minimal effects on glucose homeostasis were observed at efficacious doses. GSK1838705A also inhibits the anaplastic lymphoma kinase (ALK), which drives the aberrant growth of anaplastic large-cell lymphomas, some neuroblastomas, and a subset of non-small cell lung cancers. GSK1838705A inhibits ALK, with an IC(50) of 0.5 nmol/L, and causes complete regression of ALK-dependent tumors in vivo at well-tolerated doses. GSK1838705A is therefore a promising antitumor agent for therapeutic use in human cancers.
- Published
- 2009
- Full Text
- View/download PDF
36. Distinct concentration-dependent effects of the polo-like kinase 1-specific inhibitor GSK461364A, including differential effect on apoptosis.
- Author
-
Gilmartin AG, Bleam MR, Richter MC, Erskine SG, Kruger RG, Madden L, Hassler DF, Smith GK, Gontarek RR, Courtney MP, Sutton D, Diamond MA, Jackson JR, and Laquerre SG
- Subjects
- Adenosine Triphosphate metabolism, Animals, Biomarkers, Tumor, Cell Cycle Proteins metabolism, Cell Line, Tumor, Clinical Trials as Topic, Dose-Response Relationship, Drug, G2 Phase drug effects, Humans, Mice, Mice, Nude, Mitosis drug effects, Neoplasms drug therapy, Neoplasms pathology, Neoplasms, Experimental drug therapy, Neoplasms, Experimental enzymology, Neoplasms, Experimental pathology, Protein Kinase Inhibitors therapeutic use, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Thiophenes therapeutic use, Polo-Like Kinase 1, Apoptosis drug effects, Cell Cycle Proteins antagonists & inhibitors, Neoplasms enzymology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors, Thiophenes pharmacology
- Abstract
Polo-like kinase 1 (Plk1) is a conserved serine/threonine kinase that plays an essential role in regulating the many processes involved in mitotic entry and progression. In humans, Plk1 is expressed primarily during late G(2) and M phases and, in conjunction with Cdk1/cyclin B1, acts as master regulatory kinases for the myriad protein substrates involved in mitosis. Plk1 overexpression is strongly associated with cancer and has been correlated with poor prognosis in a broad range of human tumor types. We have identified a potent, selective, reversible, ATP-competitive inhibitor of Plk1, GSK461364A, capable of inhibiting cell growth of most proliferating cancer cell lines tested. We observe distinct cell cycle effects of GSK461364A depending on the dose used. The predominant phenotype for cells treated with GSK461364A is prometaphase arrest with characteristic collapsed polar polo spindle. At high concentrations, GSK461364A delays mitotic entry in G(2) followed by gradual progression into terminal mitosis; in some cell lines, this correlates with decreased apoptosis. Cell culture growth inhibition by GSK461364A can be cytostatic or cytotoxic but leads to tumor regression in xenograft tumor models under proper dose scheduling. Finally, we describe pharmacodynamic biomarkers of GSK461364A activity (pHH3 and Plk1) that are currently being evaluated in human cancer clinical trials.
- Published
- 2009
- Full Text
- View/download PDF
37. Kinetic analysis of teicoplanin glycosyltransferases and acyltransferase reveal ordered tailoring of aglycone scaffold to reconstitute mature teicoplanin.
- Author
-
Howard-Jones AR, Kruger RG, Lu W, Tao J, Leimkuhler C, Kahne D, and Walsh CT
- Subjects
- Acylation, Acyltransferases metabolism, Catalysis, Glycosylation, Glycosyltransferases metabolism, Kinetics, Teicoplanin metabolism, Acyltransferases chemistry, Glycosyltransferases chemistry, Teicoplanin chemistry
- Published
- 2007
- Full Text
- View/download PDF
38. Staphylococcus aureus sortase transpeptidase SrtA: insight into the kinetic mechanism and evidence for a reverse protonation catalytic mechanism.
- Author
-
Frankel BA, Kruger RG, Robinson DE, Kelleher NL, and McCafferty DG
- Subjects
- Aminoacyltransferases metabolism, Bacillus anthracis enzymology, Bacillus anthracis pathogenicity, Bacterial Proteins metabolism, Catalysis, Cell Wall enzymology, Cysteine chemistry, Cysteine Endopeptidases, Enzyme Activation, Hydrogen-Ion Concentration, Kinetics, Spectrometry, Mass, Electrospray Ionization methods, Staphylococcus aureus pathogenicity, Aminoacyltransferases chemistry, Bacterial Proteins chemistry, Staphylococcus aureus enzymology
- Abstract
The Staphylococcus aureus transpeptidase SrtA catalyzes the covalent attachment of LPXTG-containing virulence and colonization-associated proteins to cell-wall peptidoglycan in Gram-positive bacteria. Recent structural characterizations of staphylococcal SrtA, and related transpeptidases SrtB from S. aureus and Bacillus anthracis, provide many details regarding the active site environment, yet raise questions with regard to the nature of catalysis and active site cysteine thiol activation. Here we re-evaluate the kinetic mechanism of SrtA and shed light on aspects of its catalytic mechanism. Using steady-state, pre-steady-state, bisubstrate kinetic studies, and high-resolution electrospray mass spectrometry, revised steady-state kinetic parameters and a ping-pong hydrolytic shunt kinetic mechanism were determined for recombinant SrtA. The pH dependencies of kinetic parameters k(cat)/K(m) and k(cat) for the substrate Abz-LPETG-Dap(Dnp)-NH(2) were bell-shaped with pK(a) values of 6.3 +/- 0.2 and 9.4 +/- 0.2 for k(cat) and 6.2 +/- 0.2 and 9.4 +/- 0.2 for k(cat)/K(m). Solvent isotope effect (SIE) measurements revealed inverse behavior, with a (D)2(O)k(cat) of 0.89 +/- 0.01 and a (D)2(O)(k(cat)/K(m)) of 0.57 +/- 0.03 reflecting an equilibrium SIE. In addition, SIE measurements strongly implicated Cys184 participation in the isotope-sensitive rate-determining chemical step when considered in conjunction with an inverse linear proton inventory for k(cat). Last, the pH dependence of SrtA inactivation by iodoacetamide revealed a single ionization for inactivation. These studies collectively provide compelling evidence for a reverse protonation mechanism where a small fraction (ca. 0.06%) of SrtA is competent for catalysis at physiological pH, yet is highly active with an estimated k(cat)/K(m) of >10(5) M(-)(1) s(-)(1).
- Published
- 2005
- Full Text
- View/download PDF
39. A systematic investigation of the synthetic utility of glycopeptide glycosyltransferases.
- Author
-
Oberthür M, Leimkuhler C, Kruger RG, Lu W, Walsh CT, and Kahne D
- Subjects
- Anti-Bacterial Agents biosynthesis, Glycosylation, Glycosyltransferases isolation & purification, Glycosyltransferases metabolism, Hexosamines chemistry, Hexosamines metabolism, Isoenzymes, Kinetics, Substrate Specificity, Vancomycin chemical synthesis, Vancomycin chemistry, Vancomycin metabolism, Anti-Bacterial Agents chemical synthesis, Glycosyltransferases chemistry, Vancomycin analogs & derivatives
- Abstract
Glycosyltransferases involved in the biosynthesis of bacterial secondary metabolites may be useful for the generation of sugar-modified analogues of bioactive natural products. Some glycosyltransferases have relaxed substrate specificity, and it has been assumed that promiscuity is a feature of the class. As part of a program to explore the synthetic utility of these enzymes, we have analyzed the substrate selectivity of glycosyltransferases that attach similar 2-deoxy-L-sugars to glycopeptide aglycons of the vancomycin-type, using purified enzymes and chemically synthesized TDP beta-2-deoxy-L-sugar analogues. We show that while some of these glycopeptide glycosyltransferases are promiscuous, others tolerate only minor modifications in the substrates they will handle. For example, the glycosyltransferases GtfC and GtfD, which transfer 4-epi-L-vancosamine and L-vancosamine to C-2 of the glucose unit of vancomycin pseudoaglycon and chloroorienticin B, respectively, show moderately relaxed donor substrate specificities for the glycosylation of their natural aglycons. In contrast, GtfA, a transferase attaching 4-epi-L-vancosamine to a benzylic position, only utilizes donors that are closely related to its natural TDP sugar substrate. Our data also show that the spectrum of donors utilized by a given enzyme can depend on whether the natural acceptor or an analogue is used, and that GtfD is the most versatile enzyme for the synthesis of vancomycin analogues.
- Published
- 2005
- Full Text
- View/download PDF
40. Assembly of the SIR complex and its regulation by O-acetyl-ADP-ribose, a product of NAD-dependent histone deacetylation.
- Author
-
Liou GG, Tanny JC, Kruger RG, Walz T, and Moazed D
- Subjects
- Binding Sites physiology, Histone Deacetylases genetics, Lysine metabolism, Macromolecular Substances metabolism, O-Acetyl-ADP-Ribose biosynthesis, Phosphoglycerate Dehydrogenase, Protein Binding physiology, Protein Structure, Tertiary physiology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Silent Information Regulator Proteins, Saccharomyces cerevisiae genetics, Sirtuin 2, Sirtuins genetics, Sirtuins metabolism, Histone Deacetylases metabolism, Histones metabolism, NAD metabolism, O-Acetyl-ADP-Ribose metabolism, Silent Information Regulator Proteins, Saccharomyces cerevisiae metabolism
- Abstract
Assembly of silent chromatin domains in budding yeast involves the deacetylation of histone tails by Sir2 and the association of the Sir3 and Sir4 proteins with hypoacetylated histone tails. Sir2 couples deacetylation to NAD hydrolysis and the synthesis of a metabolite, O-acetyl-ADP-ribose (AAR), but the functional significance of NAD hydrolysis or AAR, if any, is unknown. Here we examine the association of the Sir2, Sir3, and Sir4 proteins with each other and histone tails. Our analysis reveals that deacetylation of histone H4-lysine 16 (K16), which is critical for silencing in vivo, is also critical for the binding of Sir3 and Sir4 to histone H4 peptides in vitro. Moreover, AAR itself promotes the association of multiple copies of Sir3 with Sir2/Sir4 and induces a dramatic structural rearrangement in the SIR complex. These results suggest that Sir2 activity modulates the assembly of the SIR complex through both histone deacetylation and AAR synthesis.
- Published
- 2005
- Full Text
- View/download PDF
41. AknT is an activating protein for the glycosyltransferase AknS in L-aminodeoxysugar transfer to the aglycone of aclacinomycin A.
- Author
-
Lu W, Leimkuhler C, Gatto GJ Jr, Kruger RG, Oberthür M, Kahne D, and Walsh CT
- Subjects
- Aclarubicin chemistry, Aclarubicin metabolism, Anthracyclines chemistry, Bacterial Proteins genetics, Bacterial Proteins isolation & purification, Disaccharides metabolism, Escherichia coli genetics, Escherichia coli metabolism, Genetic Vectors, Glycosides chemistry, Glycosylation, Glycosyltransferases genetics, Glycosyltransferases isolation & purification, Naphthacenes chemistry, Naphthacenes metabolism, Nucleoside Diphosphate Sugars metabolism, Protein Binding, Streptomyces genetics, Streptomyces metabolism, Anthracyclines metabolism, Bacterial Proteins metabolism, Glycosides metabolism, Glycosyltransferases metabolism
- Abstract
During biosynthesis of the anthracycline antitumor agents daunomycin, adriamycin, and aclacinomycin, the polyketide-derived tetracyclic aglycone is enzymatically glycosylated at the C7-OH by dedicated glycosyltransferases (Gtfs) that transfer L-2,3,6-trideoxy-3-aminohexoses. In aclacinomycins, the first deoxyhexose is predicted to be transferred via AknS action, then subjected to further elongation to a trisaccharide by the subsequent Gtf, AknK. We report here that purified AknS has very low activity in the absence of the adjacently encoded AknT; however, at a 3:1 ratio, AknT stimulates AknS k(cat) by 40-fold up to 0.22 min(-1) for transfer of L-2-deoxyfucose (2-dF) to the aglycone aklavinone. It is likely that several other Gtfs that glycosylate polyketide aglycones also act as two-component catalytic systems. Incubations of purified AknS/AknT/AknK with two aglycones and two dTDP-2-deoxyhexoses produced previously uncharacterized anthracycline disaccharides.
- Published
- 2005
- Full Text
- View/download PDF
42. Tailoring of glycopeptide scaffolds by the acyltransferases from the teicoplanin and A-40,926 biosynthetic operons.
- Author
-
Kruger RG, Lu W, Oberthür M, Tao J, Kahne D, and Walsh CT
- Subjects
- Acyl Coenzyme A chemistry, Acyltransferases genetics, Acyltransferases isolation & purification, Cloning, Molecular, Glycopeptides chemistry, Hydrogen-Ion Concentration, Kinetics, Molecular Conformation, Sensitivity and Specificity, Stereoisomerism, Teicoplanin chemical synthesis, Time Factors, Acyltransferases chemistry, Anti-Bacterial Agents chemistry, Glycopeptides chemical synthesis, Operon, Teicoplanin chemistry
- Abstract
The teicoplanin acyltransferase (Atf) responsible for N-acylation of the glucosamine moiety to create the teicoplanin lipoglycopeptide scaffold has recently been identified. Here we use that enzyme (tAtf) and the cognate acyltransferase from the related A-40,926 biosynthetic cluster (aAtf) to evaluate specificity for glycopeptide scaffolds and for the acyl-CoA donor. In addition to acylation of 2-aminoglucosyl glycopeptide scaffolds with k(cat) values of 400-2000 min(-1), both Atfs transfer acyl groups to regioisomeric 6-aminoglucosyl scaffolds and to glucosyl scaffolds at rates of 0.2-0.5 min(-1) to create variant lipoglycopeptides. Using the teicoplanin glycosyltransferase tGtfA, tAtf, and GtfD, a glycosyltransferase from the vancomycin producer, it is possible to assemble a novel lipoglycopeptide with GlcNAc at beta-OH-Tyr(6) and an N(6)-acyl-glucosaminyl-vancosamine at Phegly(4). This study illustrates the utility of chemo- and regioselective acyltransferases and glycosyltransferases to create novel lipoglycopeptides.
- Published
- 2005
- Full Text
- View/download PDF
43. Inhibition of the Staphylococcus aureus sortase transpeptidase SrtA by phosphinic peptidomimetics.
- Author
-
Kruger RG, Barkallah S, Frankel BA, and McCafferty DG
- Subjects
- Amino Acid Sequence, Aminoacyltransferases metabolism, Bacterial Proteins, Biomimetic Materials chemistry, Biomimetic Materials pharmacology, Catalysis, Cysteine Endopeptidases, Enzyme Inhibitors chemistry, Kinetics, Molecular Structure, Phosphopeptides chemical synthesis, Staphylococcus aureus chemistry, Aminoacyltransferases antagonists & inhibitors, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors pharmacology, Phosphopeptides chemistry, Phosphopeptides pharmacology, Staphylococcus aureus drug effects, Staphylococcus aureus enzymology
- Abstract
During pathogenesis, Gram-positive bacteria utilize surface protein virulence factors such as the MSCRAMMs (microbial surface components recognizing adhesive matrix molecules) to aid the initiation and propagation of infection through adherence to host endothelial tissue and immune system evasion. These virulence-associated proteins generally contain a C-terminal LPXTG motif that becomes covalently anchored to the peptidoglycan biosynthesis intermediate lipid II. In Staphylococcus aureus, deletion of the sortase isoform SrtA results in marked reduction in virulence and infection potential, making it an important antivirulence target. Here we describe the chemical synthesis and kinetic characterization of a nonhydrolyzable phosphinic peptidomimetic inhibitor of SrtA derived from the LPXTG substrate sequence.
- Published
- 2004
- Full Text
- View/download PDF
44. Alanine scanning mutagenesis of the testosterone binding site of rat 3 alpha-hydroxysteroid dehydrogenase demonstrates contact residues influence the rate-determining step.
- Author
-
Heredia VV, Cooper WC, Kruger RG, Jin Y, and Penning TM
- Subjects
- 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific) isolation & purification, Amino Acid Substitution genetics, Animals, Binding Sites genetics, Catalysis, Kinetics, Models, Chemical, NADP metabolism, Oxidation-Reduction, Progesterone metabolism, Protein Binding genetics, Rats, Recombinant Proteins biosynthesis, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Spectrometry, Fluorescence, Spectrophotometry, Ultraviolet, Substrate Specificity, 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific) genetics, 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific) metabolism, Alanine genetics, Mutagenesis, Site-Directed, Testosterone metabolism
- Abstract
Aldo-keto reductase (AKR1C) isoforms can regulate ligand access to nuclear receptors by acting as hydroxysteroid dehydrogenases. The principles that govern steroid hormone binding and steroid turnover by these enzymes were analyzed using rat 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD, AKR1C9) as the protein model. Systematic alanine scanning mutagenesis was performed on the substrate-binding pocket as defined by the crystal structure of the 3alpha-HSD.NADP(+).testosterone ternary complex. T24, L54, F118, F129, T226, W227, N306, and Y310 were individually mutated to alanine, while catalytic residues Y55 and H117 were unaltered. The effects of these mutations on the ordered bi-bi mechanism were examined. No mutations changed the affinity for NADPH by more than 2-3-fold. Fluorescence titrations of the energy transfer band of the E.NADPH complex with competitive inhibitors testosterone and progesterone showed that the largest effect was a 23-fold decrease in the affinity for progesterone in the W227A mutant. By contrast, changes in the K(d) for testosterone were negligible. Examination of the k(cat)/K(m) data for these mutants indicated that, irrespective of steroid substrate, the bimolecular rate constant was more adversely affected when alanine replaced an aromatic hydrophobic residue. By far, the greatest effects were on k(cat) (decreases of more than 2 log units), suggesting that the rate-determining step was either altered or slowed significantly. Single- and multiple-turnover experiments for androsterone oxidation showed that while the wild-type enzyme demonstrated a k(lim) and burst kinetics consistent with slow product release, the W227A and F118A mutants eliminated this kinetic profile. Instead, single- and multiple-turnover experiments gave k(lim) and k(max) values identical with k(cat) values, respectively, indicating that chemistry was now rate-limiting overall. Thus, conserved residues within the steroid-binding pocket affect k(cat) more than K(d) by influencing the rate-determining step of steroid oxidation. These findings support the concept of enzyme catalysis in which the correct positioning of reactants is essential; otherwise, k(cat) will be limited by the chemical event.
- Published
- 2004
- Full Text
- View/download PDF
45. Vinyl sulfones: inhibitors of SrtA, a transpeptidase required for cell wall protein anchoring and virulence in Staphylococcus aureus.
- Author
-
Frankel BA, Bentley M, Kruger RG, and McCafferty DG
- Subjects
- Aminoacyltransferases metabolism, Bacterial Proteins, Cell Wall enzymology, Cell Wall physiology, Cysteine Endopeptidases, Staphylococcus aureus drug effects, Staphylococcus aureus pathogenicity, Virulence, Aminoacyltransferases antagonists & inhibitors, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Staphylococcus aureus enzymology, Sulfones chemistry, Sulfones pharmacology
- Abstract
Several small molecule vinyl sulfones were found to exhibit irreversible time-dependent inhibition of the Staphylococcus aureus sortase SrtA in vitro. A representative of these compounds was shown to impair the ability of S. aureus bacteria to bind fibronectin-coated surfaces through in vivo inhibition of SrtA-mediated linkage of fibronectin to the cell surface. These data highlight the potential use of small molecule vinyl sulfones as chemotherapeutics to prevent adhesion to and colonization of host tissues during S. aureus infection.
- Published
- 2004
- Full Text
- View/download PDF
46. Development of a high-performance liquid chromatography assay and revision of kinetic parameters for the Staphylococcus aureus sortase transpeptidase SrtA.
- Author
-
Kruger RG, Dostal P, and McCafferty DG
- Subjects
- Bacterial Proteins, Cell Membrane metabolism, Cysteine Endopeptidases, Kinetics, Molecular Structure, Peptides chemical synthesis, Peptides chemistry, Peptides metabolism, Spectrometry, Fluorescence, Aminoacyltransferases analysis, Aminoacyltransferases metabolism, Chromatography, High Pressure Liquid methods, Staphylococcus aureus enzymology
- Abstract
The SrtA isoform of the Staphylococcus aureus sortase transpeptidase is responsible for the covalent attachment of virulence- and colonization-associated proteins to the bacterial peptidoglycan. Sortase utilizes two substrates, undecaprenol-pyrophosphoryl-MurNAc(GlcNAc)-Ala-d-isoGlu-Lys(-Gly5)-d-Ala-d-Ala (branched Lipid II) and secreted proteins containing a highly conserved LPXTG sequence near their C termini. SrtA simultaneously cleaves the Thr-Gly bond of the LPXTG-containing protein and forms a new amide bond with the nucleophilic amino group of the Gly5 portion of branched Lipid II, anchoring the protein to this key intermediate that is subsequently polymerized into peptidoglycan. Here we show that reported fluorescence quenching activity assays for SrtA are subject to marked fluorescence inner filter effect quenching, resulting in prematurely hyperbolic velocity versus substrate profiles and underestimates of the true kinetic parameters kcat and Km. We therefore devised a discontinuous high-performance liquid chromatography (HPLC)-based assay to monitor the SrtA reaction employing the same substrates used in the fluorescence quenching assay: Gly5 and Abz-LPETG-Dap(Dnp)-NH2. Fluorescence or UV detection using these substrates facilitates separate analysis of both the acylation and the transpeptidation steps of the reaction. Because HPLC was performed using fast-flow analytical columns (<8min/run), high-throughput applications of this assay for analysis of SrtA substrate specificity, kinetic mechanism, and inhibition are now feasible. Kinetic analysis using the HPLC assay revealed that the kinetic parameters for SrtA with Abz-LPETG-Dap(Dnp)-NH2 are 5.5mM for Km and 0.27s-1 for kcat. The Km for Gly5 was determined to be 140microM. These values represent a 300-fold increase in Km for the LPXTG substrate and a 12,000-fold increase in kcat over literature-reported values, suggesting that SrtA is more a robust enzyme than previous analyses indicated.
- Published
- 2004
- Full Text
- View/download PDF
47. Analysis of the substrate specificity of the Staphylococcus aureus sortase transpeptidase SrtA.
- Author
-
Kruger RG, Otvos B, Frankel BA, Bentley M, Dostal P, and McCafferty DG
- Subjects
- Amino Acid Motifs, Aminoacyltransferases deficiency, Aminoacyltransferases genetics, Aminoacyltransferases ultrastructure, Bacterial Adhesion, Bacterial Proteins, Cell Wall chemistry, Cell Wall genetics, Cell Wall ultrastructure, Cloning, Molecular, Conserved Sequence, Cysteine Endopeptidases, Enzyme Activation genetics, Histidine chemistry, Isoenzymes chemistry, Isoenzymes deficiency, Isoenzymes genetics, Isoenzymes ultrastructure, Mutation, Peptide Library, Staphylococcus aureus genetics, Staphylococcus aureus pathogenicity, Staphylococcus aureus ultrastructure, Substrate Specificity, Virulence, Aminoacyltransferases chemistry, Staphylococcus aureus enzymology
- Abstract
The Staphylococcus aureus sortase transpeptidase SrtA isoform is responsible for the covalent attachment of virulence and colonization-associated proteins to the bacterial peptidoglycan. SrtA utilizes two substrates, undecaprenol-pyrophosphoryl-MurNAc(GlcNAc)-Ala-D-isoGlu-Lys(epsilon-Gly(5))-D-Ala-D-Ala (branched Lipid II) and secreted proteins containing a highly conserved C-terminal LPXTG sequence. SrtA simultaneously cleaves the Thr-Gly bond of the LPXTG-containing protein and forms a new amide bond with the nucleophilic amino group of the Gly(5) portion of branched Lipid II, anchoring the protein to this key intermediate that is subsequently polymerized into peptidoglycan. Here we describe the development of a general in vitro method for elucidating the substrate specificity of sortase enzymes. In addition, using immunofluorescence, cell adhesion assays, and transmission electron microscopy, we establish links between in vitro substrate specificity and in vivo function of the S. aureus sortase isoforms. Results from these studies provide strong supporting evidence of a primary role of the SrtA isoform in S. aureus adhesion and host colonization, illustrate a lack of specificity cross talk between SrtA and SrtB isoforms, and highlight the potential of SrtA as a target for the development of antivirulence chemotherapeutics against Gram-positive bacterial pathogens.
- Published
- 2004
- Full Text
- View/download PDF
48. Steroid-binding site residues dictate optimal substrate positioning in rat 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD or AKR1C9).
- Author
-
Heredia VV, Kruger RG, and Penning TM
- Subjects
- Alanine genetics, Animals, Binding Sites, Hydroxysteroid Dehydrogenases chemistry, Hydroxysteroid Dehydrogenases genetics, Kinetics, Rats, Substrate Specificity, Hydroxysteroid Dehydrogenases metabolism, Progesterone metabolism, Testosterone metabolism
- Abstract
Rat liver 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD or AKR1C9), a member of the aldo-keto reductase (AKR) superfamily, plays a pivotal role in the inactivation of circulating steroid hormones. It is the most thoroughly characterized HSD of the AKR superfamily and can be used as a template for structure-function studies in other AKR members such as rodent and human 3alpha-, 17beta- and 20alpha-HSDs. Based on the crystal structure of the E.NADP(+) testosterone ternary complex, there are ten residues that line the testosterone binding cavity: T24, L54, Y55, H117, F118, F129, T226, W227, N306 and Y310. Each residue in the cavity, except for the catalytic residues Y55 and H117, was systematically mutated to alanine to determine the role of the individual residues in steroid recognition. Binding data and kinetic parameters (K(d), k(cat), K(m) and k(cat)/K(m)) of the homogeneous mutants were compared with that of the wild type (WT) enzyme. Titration of the intrinsic tryptophan fluorescence with NADPH demonstrated that cofactor binding was unaltered. However, binding of the steroid hormones testosterone and progesterone to the E.NADPH binary complex was affected to varying degrees. The largest effects on K(d) were an 8-fold decrease in affinity for testosterone and a 50-fold decrease in affinity for progesterone. The mutants bound both hormones in the same rank-order except for W227A, where the binding of progesterone was more adversely affected. A series of 3alpha-hydroxysteroid substrates (A/B trans- and cis-ring fused C(19) and C(21) steroids) were used to determine the ability of each mutant to catalyze steroid turnover. The alanine mutants that retained k(cat)/K(m) values similar to WT were those in which alanine substituted short polar residues such as T24A and T226A. The mutants with the lowest catalytic efficiencies were those in which alanine substituted aromatic residues such as W227A and F129A. The loss in catalytic efficiency was due to large changes in k(cat) (up to 1000-fold), but not K(m). Molecular modeling of the alanine mutants showed that changes in the reaction trajectory defined by the angles and distances by groups that participate in catalysis correlate with changes in k(cat). These results highlight the importance of steroid binding site residues in dictating the proper orientation of substrates to achieve high catalytic turnover while having minimal effects on hormone affinity.
- Published
- 2003
- Full Text
- View/download PDF
49. An economical and preparative orthogonal solid phase synthesis of fluorescein and rhodamine derivatized peptides: FRET substrates for the Staphylococcus aureus sortase SrtA transpeptidase reaction.
- Author
-
Kruger RG, Dostal P, and McCafferty DG
- Subjects
- Aminoacyltransferases chemistry, Bacterial Proteins, Cysteine Endopeptidases, Energy Transfer, Fluorescence Polarization, Fluorescent Dyes chemistry, Isoenzymes chemistry, Isoenzymes metabolism, Molecular Probe Techniques, Peptidyl Transferases chemistry, Staphylococcus aureus enzymology, Substrate Specificity, Aminoacyltransferases metabolism, Fluorescein chemistry, Fluorescent Dyes chemical synthesis, Peptides chemical synthesis, Peptidyl Transferases metabolism, Rhodamines chemistry
- Abstract
An economical and preparative-scale orthogonal solid-phase method of incorporating carboxyrhodamine and carboxyfluorescein fluorescence resonance energy transfer (FRET) probes site-specifically into synthetic peptide substrates for the S. aureus Sortase transpeptidase SrtA has been developed.
- Published
- 2002
- Full Text
- View/download PDF
50. Functional analysis of the lipoglycodepsipeptide antibiotic ramoplanin.
- Author
-
Cudic P, Behenna DC, Kranz JK, Kruger RG, Wand AJ, Veklich YI, Weisel JW, and McCafferty DG
- Subjects
- Anti-Bacterial Agents pharmacology, Dimerization, Drug Design, Drug Stability, Glycosylation, Molecular Conformation, Ornithine, Peptides, Cyclic pharmacology, Peptidoglycan biosynthesis, Peptidoglycan drug effects, Peptidoglycan metabolism, Structure-Activity Relationship, Anti-Bacterial Agents chemistry, Depsipeptides, Peptides, Cyclic chemistry
- Abstract
The peptide antibiotic ramoplanin is highly effective against several drug-resistant gram-positive bacteria, including vancomycin-resistant Enterococcus faecium (VRE) and methicillin-resistant Staphylococcus aureus (MRSA), two important opportunistic human pathogens. Ramoplanin inhibits bacterial peptidoglycan (PG) biosynthesis by binding to Lipid intermediates I and II at a location different than the N-acyl-D-Ala-D-Ala dipeptide site targeted by vancomycin. Lipid I/II capture physically occludes these substrates from proper utilization by the late-stage PG biosynthesis enzymes MurG and the transglycosylases. Key structural features of ramoplanin responsible for antibiotic activity and PG molecular recognition have been discovered by antibiotic semisynthetic modification in conjunction with NMR analyses. These results help define a minimalist ramoplanin pharmacophore and introduce the possibility of generating ramoplanin-derived peptide or peptidomimetic antibiotics for use against VRE, MRSA, and related pathogens.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.