Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Biochem Pharmacol ; 182: 114219, 2020 12.
Article in English | MEDLINE | ID: mdl-32931773

ABSTRACT

Isoprenylcysteine carboxylmethyltransferase (ICMT) has been reported to regulate the inflammatory response through the Ras/MAPK/AP-1 pathway. Nevertheless, the potential of ICMT inhibitors as therapeutic agents against inflammatory diseases has not been examined. Therefore, in this study, we investigated the anti-inflammatory properties of two ICMT inhibitors, cysmethynil (CyM) and 3-methoxy-N-[2-2,2,6,6-tetramethyl-4-phenyltetrahydropyran-4-yl)ethyl]aniline (MTPA), using in vitro analyses and in vivo analyses (lipopolysaccharide (LPS)/D-GalN-triggered hepatitis and DSS-induced colitis mouse models). CyM and MTPA inhibited the production of nitric oxide (NO) and prostaglandin E (PGE)2 and the expression of cyclooxygenase (COX)-2, tumor necrosis factor (TNF)-α and interleukin (IL)-1ß in LPS-induced RAW264.7 cells and peritoneal macrophages without cytotoxicity. CyM also reduced AP-1-mediated luciferase activity in LPS-stimulated RAW264.7 cells and MyD88- and TRIF-expressing HEK293 cells. In addition, CyM and MTPA suppressed the translocation of Ras to the cell membrane and ER as well as phosphorylation of Ras-dependent AP-1 signaling molecules including Raf, MEK1/2, ERK p38, and JNK. Consistent with these results, CyM diminished the expression of inflammatory genes (COX-2, TNF-α, IL-1ß, and IL-6), AP-1-Luc activity, and phosphorylation of Ras-mediated signaling enzymes in Ras-overexpressing HEK 293 cells. Moreover, CyM and MTPA ameliorated symptoms of hepatitis and colitis in mice and restrained the ICMT/Ras-dependent AP-1 pathway in inflammatory lesions of the mouse model systems. Taken together, our results indicate that CyM and MTPA alleviate the LPS-induced ICMT/Ras/AP-1 signaling pathway, thereby inhibiting the inflammatory response as promising anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Indoles/pharmacology , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Hepatitis/drug therapy , Hepatitis/metabolism , Humans , Indoles/chemistry , Indoles/therapeutic use , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , RAW 264.7 Cells
2.
J Med Chem ; 62(13): 6035-6046, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31181882

ABSTRACT

Blockade of Ras activity by inhibiting its post-translational methylation catalyzed by isoprenylcysteine carboxylmethyltransferase (ICMT) has been suggested as a promising antitumor strategy. However, the paucity of inhibitors has precluded the clinical validation of this approach. In this work we report a potent ICMT inhibitor, compound 3 [UCM-1336, IC50 = 2 µM], which is selective against the other enzymes involved in the post-translational modifications of Ras. Compound 3 significantly impairs the membrane association of the four Ras isoforms, leading to a decrease of Ras activity and to inhibition of Ras downstream signaling pathways. In addition, it induces cell death in a variety of Ras-mutated tumor cell lines and increases survival in an in vivo model of acute myeloid leukemia. Because ICMT inhibition impairs the activity of the four Ras isoforms regardless of its activating mutation, compound 3 surmounts many of the common limitations of available Ras inhibitors described so far. In addition, these results validate ICMT as a valuable target for the treatment of Ras-driven tumors.


Subject(s)
Alanine/therapeutic use , Amides/therapeutic use , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Protein Methyltransferases/antagonists & inhibitors , Alanine/analogs & derivatives , Alanine/chemical synthesis , Alanine/pharmacology , Amides/chemical synthesis , Amides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Mice , Xenograft Model Antitumor Assays
3.
Biochem Biophys Res Commun ; 516(3): 784-789, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31253403

ABSTRACT

Development of chemo-resistance in nasopharyngeal carcinoma (NPC) poses the therapeutic challenge and its mechanisms are still poorly understood. In this work, we demonstrate that targeting isoprenylcysteine carboxylmethyltransferase (Icmt) is a therapeutic strategy to overcome NPC chemo-resistance. We found that Icmt mRNA and protein levels were increased in NPC cells after prolonged exposure to chemotherapy. Using pharmacological inhibitor cysmethynil or genetic siRNA approaches, we showed that Icmt inhibition was more effective against chemoresistant compared to chemosensitive NPC cells, suggesting that chemoresistant NPC cells is more dependent on Icmt function. The combination of Icmt inhibition with 5-FU or cisplatin resulted in greater efficacy than single chemotherapeutic agent alone in NPC. Notably, we demonstrated that the in vitro observations were translatable to in vivo NPC cancer xenograft mouse model. Mechanism analysis indicated that Icmt inhibition decreased Ras and RhoA activities, leading to the suppression of Ras and RhoA-mediated downstream signaling in NPC cells. The reverse of the inhibitory effects of cysmethynil by constitutively active Ras suggests that Ras is the critical effector of Icmt in NPC cells. Our work is the first to show that Icmt plays an important role in the development of NPC chemoresistance. Our findings also suggest that targeting Icmt represents a promising strategy to inhibit Ras function.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/genetics , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Protein Methyltransferases/genetics , ras Proteins/genetics , Animals , Cell Line, Tumor , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fluorouracil/administration & dosage , Humans , Indoles/administration & dosage , Mice, Nude , Mice, SCID , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/metabolism , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/metabolism , RNA Interference , Xenograft Model Antitumor Assays/methods , ras Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
4.
Nat Commun ; 10(1): 19, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604761

ABSTRACT

Protein methyltransferases (PMTs) comprise a major class of epigenetic regulatory enzymes with therapeutic relevance. Here we present a collection of chemical probes and associated reagents and data to elucidate the function of human and murine PMTs in cellular studies. Our collection provides inhibitors and antagonists that together modulate most of the key regulatory methylation marks on histones H3 and H4, providing an important resource for modulating cellular epigenomes. We describe a comprehensive and comparative characterization of the probe collection with respect to their potency, selectivity, and mode of inhibition. We demonstrate the utility of this collection in CD4+ T cell differentiation assays revealing the potential of individual probes to alter multiple T cell subpopulations which may have implications for T cell-mediated processes such as inflammation and immuno-oncology. In particular, we demonstrate a role for DOT1L in limiting Th1 cell differentiation and maintaining lineage integrity. This chemical probe collection and associated data form a resource for the study of methylation-mediated signaling in epigenetics, inflammation and beyond.


Subject(s)
Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Histones/metabolism , Protein Methyltransferases/antagonists & inhibitors , Protein Processing, Post-Translational/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Enzyme Assays/methods , Epigenomics/methods , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Jurkat Cells , Methylation/drug effects , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Mice, Inbred C57BL , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational/genetics , Th1 Cells/drug effects , Th1 Cells/physiology
5.
Stem Cells ; 37(5): 640-651, 2019 05.
Article in English | MEDLINE | ID: mdl-30681750

ABSTRACT

Understanding the mechanisms that promote the specification of pancreas progenitors and regulate their self-renewal and differentiation will help to maintain and expand pancreas progenitor cells derived from human pluripotent stem (hPS) cells. This will improve the efficiency of current differentiation protocols of hPS cells into ß-cells and bring such cells closer to clinical applications for the therapy of diabetes. Aldehyde dehydrogenase 1b1 (Aldh1b1) is a mitochondrial enzyme expressed specifically in progenitor cells during mouse pancreas development, and we have shown that its functional inactivation leads to accelerated differentiation and deficient ß-cells. In this report, we aimed to identify small molecule inducers of Aldh1b1 expression taking advantage of a mouse embryonic stem (mES) cell Aldh1b1 lacZ reporter line and a pancreas differentiation protocol directing mES cells into pancreatic progenitors. We identified AMI-5, a protein methyltransferase inhibitor, as an Aldh1b1 inducer and showed that it can maintain Aldh1b1 expression in embryonic pancreas explants. This led to a selective reduction in endocrine specification. This effect was due to a downregulation of Ngn3, and it was mediated through Aldh1b1 since the effect was abolished in Aldh1b1 null pancreata. The findings implicated methyltransferase activity in the regulation of endocrine differentiation and showed that methyltransferases can act through specific regulators during pancreas differentiation. Stem Cells 2019;37:640-651.


Subject(s)
Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Cell Differentiation/genetics , Diabetes Mellitus/therapy , Pluripotent Stem Cells/transplantation , Protein Methyltransferases/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Benzoates/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Insulin-Secreting Cells/metabolism , Mice , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/enzymology , Nerve Tissue Proteins/genetics , Pancreas/drug effects , Pancreas/growth & development , Protein Methyltransferases/antagonists & inhibitors , Xanthenes/pharmacology
6.
Chembiochem ; 20(8): 976-984, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30479015

ABSTRACT

Protein α-N-terminal methylation is catalyzed by protein N-terminal methyltransferases. The prevalent occurrence of this methylation in ribosomes, myosin, and histones implies its function in protein-protein interactions. Although its full spectrum of function has not yet been outlined, recent discoveries have revealed the emerging roles of α-N-terminal methylation in protein-chromatin interactions, DNA damage repair, and chromosome segregation. Herein, an overview of the discovery of protein N-terminal methyltransferases and functions of α-N-terminal methylation is presented. In addition, substrate recognition, mechanisms, and inhibition of N-terminal methyltransferases are reviewed. Opportunities and gaps in protein α-N-terminal methylation are also discussed.


Subject(s)
Protein Methyltransferases/metabolism , Catalysis , DNA Damage , DNA Repair , Enzyme Inhibitors/pharmacology , Humans , Methylation , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/chemistry , Protein Processing, Post-Translational , Substrate Specificity
7.
Chem Rec ; 18(12): 1660-1671, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30324709

ABSTRACT

Physiological regulatory mechanisms of protein, RNA, and DNA functions include small chemical modifications, such as methylation, which are introduced or removed in a highly chemo-, regio-, and site-selective manner by methyltransferases and demethylases, respectively. However, mimicking or controlling these modifications by using labeling reagents and inhibitors remains challenging. In this Personal Account, we introduce our nascent interdisciplinary collaboration between chemists and biologists aimed at developing a basic strategy to analyse and control the methylation reactions regulated by protein methyltransferases (PMTs). We focus in particular on the structural development of chaetocin and S-adenosylmethionine to obtain PMT inhibitors and PMT substrate detectors.


Subject(s)
Protein Methyltransferases/metabolism , Proteins/metabolism , S-Adenosylmethionine/analogs & derivatives , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Methylation , Piperazines/chemistry , Piperazines/metabolism , Protein Methyltransferases/antagonists & inhibitors , Proteomics , S-Adenosylmethionine/metabolism
8.
Oral Oncol ; 81: 100-108, 2018 06.
Article in English | MEDLINE | ID: mdl-29884408

ABSTRACT

Squamous cell carcinoma of the head and neck is a lethal disease with suboptimal survival outcomes and standard therapies with significant comorbidities. Whole exome sequencing data recently revealed an abundance of genetic and expression alterations in a family of enzymes known as protein methyltransferases in a variety of cancer types, including squamous cell carcinoma of the head and neck. These enzymes are mostly known for their chromatin-modifying functions through methylation of various histone substrates, though evidence supports their function also through methylation of non-histone substrates. This review summarizes the current knowledge on the function of protein methyltransferases in squamous cell carcinoma of the head and neck and highlights their promising potential as the next generation of therapeutic targets in this disease.


Subject(s)
Head and Neck Neoplasms/drug therapy , Protein Methyltransferases/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/drug therapy , Clinical Trials as Topic , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic , Head and Neck Neoplasms/enzymology , Humans , Methylation , Mutation , Protein Methyltransferases/genetics , Squamous Cell Carcinoma of Head and Neck/enzymology
9.
Biochem Biophys Res Commun ; 501(2): 556-562, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29746868

ABSTRACT

Inhibition of isoprenylcysteine carboxylmethyltransferase (Icmt), which catalyzes the final step of oncoproteins' prenylation, targets growth and survival of various cancers. In this work, we systematically studied the expression, functions and molecular signaling of Icmt in ovarian cancer. We show that the upregulation of Icmt expression is a common feature in patients with epithelial ovarian cancer regardless of age and disease stage. In line with the observations in ovarian cancer patients, a panel of epithelial ovarian cancer cell lines also demonstrates the significant increase on Icmt transcript and protein levels than normal ovarian epithelial cells. In addition, ovarian cancer cell lines with higher Icmt levels are more resistant to chemotherapeutic agents. We further show that Icmt inhibition by siRNA or inhibitor cysmethynil suppresses growth and induces apoptosis in ovarian cancer cells. Importantly, Icmt inhibition significantly augments chemotherapeutic agent's efficacy in vitro and in vivo, demonstrating the translational potential of Icmt inhibition in ovarian cancer. Mechanistically, we show that Ras activation is a critical effector of Icmt in ovarian cancer cells. Using cell culturing system, mouse model and patient samples, our work is the first to demonstrate the essential roles of Icmt in ovarian cancer via Ras signaling, particularly on its response to chemotherapy. Our findings suggest that Icmt inhibition is a promising therapeutic strategy to overcome chemoresistance in ovarian cancer, in particular, those patients with high Icmt expression.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Protein Methyltransferases/metabolism , ras Proteins/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Humans , Indoles/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Protein Methyltransferases/analysis , Protein Methyltransferases/antagonists & inhibitors
10.
Article in English | MEDLINE | ID: mdl-29685962

ABSTRACT

The protein methyltransferases (PMTs) represent a large class of enzymes that catalyse the methylation of side chain nitrogen atoms of the amino acids lysine or arginine at specific locations along the primary sequence of target proteins. These enzymes play a key role in the spatio-temporal control of gene transcription by performing site-specific methylation of lysine or arginine residues within the histone proteins of chromatin, thus effecting chromatin conformational changes that activate or repress gene transcription. Over the past decade, it has become clear that the dysregulated activity of some PMTs plays an oncogenic role in a number of human cancers. Here we review research of the past decade that has identified specific PMTs as oncogenic drivers of cancers and progress toward the discovery and development of selective, small molecule inhibitors of these enzymes as precision cancer therapeutics.This article is part of a discussion meeting issue 'Frontiers in epigenetic chemical biology'.


Subject(s)
Enzyme Inhibitors/chemistry , Neoplasms , Protein Methyltransferases , Humans , Neoplasms/genetics , Neoplasms/prevention & control , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/genetics
11.
Cell Stem Cell ; 22(2): 177-190.e7, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395054

ABSTRACT

The development of cell therapy for repairing damaged or diseased skeletal muscle has been hindered by the inability to significantly expand immature, transplantable myogenic stem cells (MuSCs) in culture. To overcome this limitation, a deeper understanding of the mechanisms regulating the transition between activated, proliferating MuSCs and differentiation-primed, poorly engrafting progenitors is needed. Here, we show that methyltransferase Setd7 facilitates such transition by regulating the nuclear accumulation of ß-catenin in proliferating MuSCs. Genetic or pharmacological inhibition of Setd7 promotes in vitro expansion of MuSCs and increases the yield of primary myogenic cell cultures. Upon transplantation, both mouse and human MuSCs expanded with a Setd7 small-molecule inhibitor are better able to repopulate the satellite cell niche, and treated mouse MuSCs show enhanced therapeutic potential in preclinical models of muscular dystrophy. Thus, Setd7 inhibition may help bypass a key obstacle in the translation of cell therapy for muscle disease.


Subject(s)
Muscle Development , Protein Methyltransferases/antagonists & inhibitors , Stem Cell Transplantation , Stem Cells/cytology , Active Transport, Cell Nucleus/drug effects , Animals , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cells, Cultured , Gene Deletion , Histone-Lysine N-Methyltransferase , Mice , Muscle, Skeletal/physiology , MyoD Protein/metabolism , Protein Binding/drug effects , Protein Methyltransferases/metabolism , Pyrrolidines/pharmacology , Regeneration/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Sulfonamides/pharmacology , Tetrahydroisoquinolines/pharmacology , beta Catenin/metabolism
12.
Biomed Pharmacother ; 99: 169-175, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29331763

ABSTRACT

Isoprenylcysteine carboxylmethyltransferase (Icmt) catalyzes the last step of post-translational protein prenylation, which is essential for the stability and proper functions of many oncogenic proteins, such as Ras. Despite extensive studies on the roles of Icmt in tumor transformation and progression, little is known on the involvement ofIcmt in the development of tumor resistance to chemotherapy. Here we show the upregulation of Icmt as a persistent response to chemotherapy in cervical cancer cells. In-depth functional analysis demonstrated that Icmt inhibition significantly inhibited growth, induced apoptosis and augmented the inhibitory effects of chemotherapy drugs in cervical cancer in cell culture system and xenograft mouse model. Importantly, combination of Icmt specific inhibitor cysmethynil with doxorubicin or paclitaxel at sublethal concentration achieved almost full inhibition of tumor cell growth and survival. The remarkable synergy between chemotherapy drugs and Icmt inhibition in cervical cancer cells is likely due to the additional suppression of Ras and its downstream signaling pathways. We are the first to demonstrate the contribution of Icmt in tumor cells in response to chemotherapy. Our work also highlights Icmt inhibition as a sensitizing strategy for the treatment of cervical cancer or other Ras-driven tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Protein Methyltransferases/antagonists & inhibitors , Signal Transduction , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/enzymology , ras Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Female , Humans , Mice, SCID , Protein Methyltransferases/metabolism , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
Nature ; 553(7689): 526-529, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29342140

ABSTRACT

The maturation of RAS GTPases and approximately 200 other cellular CAAX proteins involves three enzymatic steps: addition of a farnesyl or geranylgeranyl prenyl lipid to the cysteine (C) in the C-terminal CAAX motif, proteolytic cleavage of the AAX residues and methylation of the exposed prenylcysteine residue at its terminal carboxylate. This final step is catalysed by isoprenylcysteine carboxyl methyltransferase (ICMT), a eukaryote-specific integral membrane enzyme that resides in the endoplasmic reticulum. ICMT is the only cellular enzyme that is known to methylate prenylcysteine substrates; methylation is important for the biological functions of these substrates, such as the membrane localization and subsequent activity of RAS, prelamin A and RAB. Inhibition of ICMT has potential for combating progeria and cancer. Here we present an X-ray structure of ICMT, in complex with its cofactor, an ordered lipid molecule and a monobody inhibitor, at 2.3 Å resolution. The active site spans cytosolic and membrane-exposed regions, indicating distinct entry routes for the cytosolic methyl donor, S-adenosyl-l-methionine, and for prenylcysteine substrates, which are associated with the endoplasmic reticulum membrane. The structure suggests how ICMT overcomes the topographical challenge and unfavourable energetics of bringing two reactants that have different cellular localizations together in a membrane environment-a relatively uncharacterized but defining feature of many integral membrane enzymes.


Subject(s)
Protein Methyltransferases/chemistry , Protein Methyltransferases/metabolism , Tribolium/enzymology , Animals , Catalytic Domain , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , Cysteine/analogs & derivatives , Cysteine/chemistry , Cysteine/metabolism , Drug Design , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Models, Molecular , Protein Methyltransferases/antagonists & inhibitors , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Substrate Specificity
14.
Chem Rev ; 118(3): 989-1068, 2018 02 14.
Article in English | MEDLINE | ID: mdl-28338320

ABSTRACT

Post-translational modifications of histones by protein methyltransferases (PMTs) and histone demethylases (KDMs) play an important role in the regulation of gene expression and transcription and are implicated in cancer and many other diseases. Many of these enzymes also target various nonhistone proteins impacting numerous crucial biological pathways. Given their key biological functions and implications in human diseases, there has been a growing interest in assessing these enzymes as potential therapeutic targets. Consequently, discovering and developing inhibitors of these enzymes has become a very active and fast-growing research area over the past decade. In this review, we cover the discovery, characterization, and biological application of inhibitors of PMTs and KDMs with emphasis on key advancements in the field. We also discuss challenges, opportunities, and future directions in this emerging, exciting research field.


Subject(s)
Enzyme Inhibitors/metabolism , Histone Demethylases/metabolism , Protein Methyltransferases/metabolism , Enzyme Inhibitors/chemistry , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/classification , Humans , Kinetics , Lysine/metabolism , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/classification , Protein Processing, Post-Translational , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
15.
Sci Adv ; 3(11): eaao5268, 2017 11.
Article in English | MEDLINE | ID: mdl-29109980

ABSTRACT

Metabolic reprogramming is a major factor in transformation, and particular metabolic phenotypes correlate with oncogenotype, tumor progression, and metastasis. By profiling metabolites in 17 patient-derived xenograft melanoma models, we identified durable metabolomic signatures that correlate with biological features of the tumors. BRAF mutant tumors had metabolomic and metabolic flux features of enhanced glycolysis compared to BRAF wild-type tumors. Tumors that metastasized efficiently from their primary sites had elevated levels of metabolites related to protein methylation, including trimethyllysine (TML). TML levels correlated with histone H3 trimethylation at Lys9 and Lys27, and methylation at these sites was also enhanced in efficiently metastasizing tumors. Erasing either of these marks by genetically or pharmacologically silencing the histone methyltransferase SETDB1 or EZH2 had no effect on primary tumor growth but reduced cellular invasiveness and metastatic spread. Thus, metabolite profiling can uncover targetable epigenetic requirements for the metastasis of human melanoma cells.


Subject(s)
Melanoma/pathology , Metabolome , Methylation , Animals , Cell Movement/drug effects , Down-Regulation/drug effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Indoles/pharmacology , Indoles/therapeutic use , Melanoma/drug therapy , Melanoma/metabolism , Metabolomics , Methylation/drug effects , Mice , Mice, Inbred NOD , Neoplasm Metastasis , Principal Component Analysis , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyridones/pharmacology , Pyridones/therapeutic use , RNA Interference , RNA, Small Interfering/metabolism , Transplantation, Heterologous
16.
AIDS Res Hum Retroviruses ; 33(S1): S8-S22, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29140109

ABSTRACT

A successful HIV cure strategy may require reversing HIV latency to purge hidden viral reservoirs or enhancing HIV latency to permanently silence HIV transcription. Epigenetic modifying agents show promise as antilatency therapeutics in vitro and ex vivo, but also affect other steps in the viral life cycle. In this review, we summarize what we know about cellular DNA and protein methyltransferases (PMTs) as well as demethylases involved in HIV infection. We describe the biology and function of DNA methyltransferases, and their controversial role in HIV infection. We further explain the biology of PMTs and their effects on lysine and arginine methylation of histone and nonhistone proteins. We end with a focus on protein demethylases, their unique modes of action and their emerging influence on HIV infection. An outlook on the use of methylation-modifying agents in investigational HIV cure strategies is provided.


Subject(s)
Anti-HIV Agents/pharmacology , DNA Modification Methylases/antagonists & inhibitors , HIV Infections/drug therapy , Histone Demethylases/antagonists & inhibitors , Protein Methyltransferases/antagonists & inhibitors , Virus Latency/drug effects , DNA/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Epigenesis, Genetic/drug effects , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Virus Activation/drug effects
17.
J Comput Aided Mol Des ; 31(10): 877-889, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28879500

ABSTRACT

ERG-associated protein with the SET domain (ESET/SET domain bifurcated 1/SETDB1/KMT1E) is a histone lysine methyltransferase (HKMT) and it preferentially tri-methylates lysine 9 of histone H3 (H3K9me3). SETDB1/ESET leads to heterochromatin condensation and epigenetic gene silencing. These functional changes are reported to correlate with Huntington's disease (HD) progression and mood-related disorders which make SETDB1/ESET a viable drug target. In this context, the present investigation was performed to identify novel peptide-competitive small molecule inhibitors of the SETDB1/ESET by a combined in silico-in vitro approach. A ligand-based pharmacophore model was built and employed for the virtual screening of ChemDiv and Asinex database. Also, a human SETDB1/ESET homology model was constructed to supplement the data further. Biological evaluation of the selected 21 candidates singled out 5 compounds exhibiting a notable reduction of the H3K9me3 level via inhibitory potential of SETDB1/ESET activity in SETDB1/ESET-inducible cell line and HD striatal cells. Later on, we identified two compounds as final hits that appear to have neuronal effects without cytotoxicity based on the result from MTT assay. These compounds hold the calibre to become the future lead compounds and can provide structural insights into more SETDB1/ESET-focused drug discovery research. Moreover, these SETDB1/ESET inhibitors may be applicable for the preclinical study to ameliorate neurodegenerative disorders via epigenetic regulation.


Subject(s)
Benzene Derivatives/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Protein Methyltransferases/antagonists & inhibitors , Benzene Derivatives/pharmacology , Binding Sites , Cell Line , Cell Survival , Computer Simulation , Databases, Pharmaceutical , Humans , Ligands , Methylation , Molecular Docking Simulation , Neurons/cytology , Neurons/drug effects , Protein Binding , Protein Methyltransferases/metabolism , Structure-Activity Relationship
18.
Amino Acids ; 49(9): 1469-1485, 2017 09.
Article in English | MEDLINE | ID: mdl-28631011

ABSTRACT

Among the enzymes involved in the post-translational modification of Ras, isoprenyl carboxyl methyltransferase (ICMT) has been explored by a number of researchers as a significant enzyme controlling the activation of Ras. Indeed, inhibition of ICMT exhibited promising anti-cancer activity against various cancer cell lines. This paper reviews patents and research articles published between 2009 and 2016 that reported inhibitors of ICMT as potential chemotherapeutic agents targeting Ras-induced growth factor signaling. Since ICMT inhibitors can modulate Ras signaling pathway, it might be possible to develop a new class of anti-cancer drugs targeting Ras-related cancers. Researchers have discovered indole-based small-molecular ICMT inhibitors through high-throughput screening. Researchers at Duke University identified a prototypical inhibitor, cysmethynil. At Singapore University, Ramanujulu and his colleagues patented more potent compounds by optimizing cysmethynil. In addition, Rodriguez and Stevenson at Universidad Complutense De Madrid and Cancer Therapeutics CRC PTY Ltd., respectively, have developed inhibitors based on formulas other than the indole base. However, further optimization of chemicals targeted to functional groups is needed to improve the characteristics of ICMT inhibitors related to their application as drugs, such as solubility, effectiveness, and safety, to facilitate clinical use.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Indoles/pharmacology , Neoplasms/drug therapy , Protein Methyltransferases/antagonists & inhibitors , Protein Processing, Post-Translational , Animals , Antineoplastic Agents/chemical synthesis , Drug Design , Enzyme Inhibitors/chemical synthesis , Humans , Indoles/chemical synthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Patents as Topic , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Signal Transduction , Structure-Activity Relationship , ras Proteins/antagonists & inhibitors , ras Proteins/genetics , ras Proteins/metabolism
19.
Curr Opin Chem Biol ; 39: 100-108, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28662389

ABSTRACT

Mounting evidence suggests that protein methyltransferases (PMTs), which catalyze methylation of histones as well as non-histone proteins, play a crucial role in diverse biological pathways and human diseases. In particular, PMTs have been recognized as major players in regulating gene expression and chromatin state. There has been an increasingly growing interest in these enzymes as potential therapeutic targets and over the past two years tremendous progress has been made in the discovery of selective, small molecule inhibitors of protein lysine and arginine methyltransferases. Inhibitors of PMTs have been used extensively in oncology studies as tool compounds, and inhibitors of EZH2, DOT1L and PRMT5 are currently in clinical trials.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Protein Methyltransferases/antagonists & inhibitors , Animals , Humans
20.
Mol Cancer Ther ; 16(5): 914-923, 2017 05.
Article in English | MEDLINE | ID: mdl-28167504

ABSTRACT

Pancreatic cancer remains one of the most difficult to treat human cancers despite recent advances in targeted therapy. Inhibition of isoprenylcysteine carboxylmethyltransferase (ICMT), an enzyme that posttranslationally modifies a group of proteins including several small GTPases, suppresses proliferation of some human cancer cells. However, the efficacy of ICMT inhibition on human pancreatic cancer has not been evaluated. In this study, we have evaluated a panel of human pancreatic cancer cell lines and identified those that are sensitive to ICMT inhibition. In these cells, ICMT suppression inhibited proliferation and induced apoptosis. This responsiveness to ICMT inhibition was confirmed in in vivo xenograft tumor mouse models using both a small-molecule inhibitor and shRNA-targeting ICMT. Mechanistically, we found that, in sensitive pancreatic cancer cells, ICMT inhibition induced mitochondrial respiratory deficiency and cellular energy depletion, leading to significant upregulation of p21. Furthermore, we characterized the role of p21 as a regulator and coordinator of cell signaling that responds to cell energy depletion. Apoptosis, but not autophagy, that is induced via p21-activated BNIP3 expression accounts for the efficacy of ICMT inhibition in sensitive pancreatic cancer cells in both in vitro and in vivo models. In contrast, cells resistant to ICMT inhibition demonstrated no mitochondria dysfunction or p21 signaling changes under ICMT suppression. These findings not only identify pancreatic cancers as potential therapeutic targets for ICMT suppression but also provide an avenue for identifying those subtypes that would be most responsive to agents targeting this critical enzyme. Mol Cancer Ther; 16(5); 914-23. ©2017 AACR.


Subject(s)
Membrane Proteins/genetics , Pancreatic Neoplasms/drug therapy , Protein Methyltransferases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , rho GTP-Binding Proteins/genetics , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Methyltransferases/genetics , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...