Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ChemMedChem ; 15(3): 270-273, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31751494

ABSTRACT

The 2-oxoglutarate-dependent hypoxia inducible factor prolyl hydroxylases (PHDs) are targets for treatment of a variety of diseases including anaemia. One PHD inhibitor is approved for use for the treatment of renal anaemia and others are in late stage clinical trials. The number of reported templates for PHD inhibition is limited. We report structure-activity relationship and crystallographic studies on a promising class of 4-hydroxypyrimidine-containing PHD inhibitors.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Prolyl-Hydroxylase Inhibitors/pharmacology , Pyrimidinones/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Models, Molecular , Molecular Structure , Prolyl-Hydroxylase Inhibitors/chemistry , Pyrimidinones/chemistry , Structure-Activity Relationship
2.
ACS Chem Biol ; 14(8): 1737-1750, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31287655

ABSTRACT

Fe(II)- and 2-oxoglutarate (2OG)-dependent JumonjiC domain-containing histone demethylases (JmjC KDMs) are "epigenetic eraser" enzymes involved in the regulation of gene expression and are emerging drug targets in oncology. We screened a set of clinically used iron chelators and report that they potently inhibit JMJD2A (KDM4A) in vitro. Mode of action investigations revealed that one compound, deferasirox, is a bona fide active site-binding inhibitor as shown by kinetic and spectroscopic studies. Synthesis of derivatives with improved cell permeability resulted in significant upregulation of histone trimethylation and potent cancer cell growth inhibition. Deferasirox was also found to inhibit human 2OG-dependent hypoxia inducible factor prolyl hydroxylase activity. Therapeutic effects of clinically used deferasirox may thus involve transcriptional regulation through 2OG oxygenase inhibition. Deferasirox might provide a useful starting point for the development of novel anticancer drugs targeting 2OG oxygenases and a valuable tool compound for investigations of KDM function.


Subject(s)
Deferasirox/pharmacology , Enzyme Inhibitors/pharmacology , Iron Chelating Agents/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Catalytic Domain/drug effects , Cell Line, Tumor , Demethylation/drug effects , Epigenesis, Genetic/drug effects , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/chemistry
3.
ChemMedChem ; 14(1): 94-99, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30380199

ABSTRACT

Prolyl hydroxylation domain (PHD) enzymes catalyze the hydroxylation of the transcription factor hypoxia-inducible factor (HIF) and serve as cellular oxygen sensors. HIF and the PHD enzymes regulate numerous potentially tissue-protective target genes which can adapt cells to metabolic and ischemic stress. We describe a fluorescent PHD inhibitor (1-chloro-4-hydroxybenzo[g]isoquinoline-3-carbonyl)glycine which is suited to fluorescence-based detection assays and for monitoring PHD inhibitors in biological systems. In cell-based assays, application of the fluorescent PHD inhibitor allowed co-localization with a cellular PHD enzyme and led to live cell imaging of processes involved in cellular oxygen sensing.


Subject(s)
Benzylisoquinolines/pharmacology , Fluorescent Dyes/pharmacology , Molecular Imaging/methods , Optical Imaging/methods , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Benzylisoquinolines/chemical synthesis , Benzylisoquinolines/chemistry , Biocatalysis/drug effects , Dose-Response Relationship, Drug , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Molecular Structure , Prolyl-Hydroxylase Inhibitors/chemical synthesis , Prolyl-Hydroxylase Inhibitors/chemistry , Structure-Activity Relationship
4.
Chem Commun (Camb) ; 55(8): 1020-1023, 2019 Jan 22.
Article in English | MEDLINE | ID: mdl-30452037

ABSTRACT

We describe covalently binding modulators of the activity of human prolyl hydroxylase domain 2 (PHD2) and studies towards a strategy for photocapture of PHD2 substrates. Reversible active site binding of electrophile bearing compounds enables susbsequent covalent reaction with a lysine residue (K408) in the flexible C-terminal region of PHD2 to give a modified protein that retains catalytic activity.


Subject(s)
Enzyme Inhibitors/metabolism , Hippurates/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Azides/chemistry , Azides/radiation effects , Catalysis , Catalytic Domain , Enzyme Inhibitors/chemistry , HeLa Cells , Hippurates/chemistry , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/chemistry , Ligands , Lysine/chemistry , Protein Binding , Ultraviolet Rays
5.
Chemistry ; 25(8): 2019-2024, 2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30427558

ABSTRACT

Human prolyl hydroxylases are involved in the modification of transcription factors, procollagen, and ribosomal proteins, and are current medicinal chemistry targets. To date, there are few reports on inhibitors selective for the different types of prolyl hydroxylases. We report a structurally informed template-based strategy for the development of inhibitors selective for the human ribosomal prolyl hydroxylase OGFOD1. These inhibitors did not target the other human oxygenases tested, including the structurally similar hypoxia-inducible transcription factor prolyl hydroxylase, PHD2.


Subject(s)
Prolyl Hydroxylases , Prolyl-Hydroxylase Inhibitors , Ribosomes/drug effects , Carrier Proteins/antagonists & inhibitors , Drug Design , Humans , Nuclear Proteins/antagonists & inhibitors , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/chemistry , Prolyl-Hydroxylase Inhibitors/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Ribosomes/metabolism , Structure-Activity Relationship , Substrate Specificity
6.
Chem Sci ; 8(11): 7651-7668, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29435217

ABSTRACT

Inhibition of the human 2-oxoglutarate (2OG) dependent hypoxia inducible factor (HIF) prolyl hydroxylases (human PHD1-3) causes upregulation of HIF, thus promoting erythropoiesis and is therefore of therapeutic interest. We describe cellular, biophysical, and biochemical studies comparing four PHD inhibitors currently in clinical trials for anaemia treatment, that describe their mechanisms of action, potency against isolated enzymes and in cells, and selectivities versus representatives of other human 2OG oxygenase subfamilies. The 'clinical' PHD inhibitors are potent inhibitors of PHD catalyzed hydroxylation of the HIF-α oxygen dependent degradation domains (ODDs), and selective against most, but not all, representatives of other human 2OG dependent dioxygenase subfamilies. Crystallographic and NMR studies provide insights into the different active site binding modes of the inhibitors. Cell-based results reveal the inhibitors have similar effects on the upregulation of HIF target genes, but differ in the kinetics of their effects and in extent of inhibition of hydroxylation of the N- and C-terminal ODDs; the latter differences correlate with the biophysical observations.

7.
Sci Rep ; 6: 30986, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27507219

ABSTRACT

The mechanism of Amifostine (WR-2721) mediated radioprotection is poorly understood. The effects of amifostine on human basal metabolism, mouse liver metabolism and on normal and tumor hepatic cells were studied. Indirect calorimetric canopy tests showed significant reductions in oxygen consumption and of carbon dioxide emission in cancer patients receiving amifostine. Glucose levels significantly decreased and lactate levels increased in patient venous blood. Although amifostine in vitro did not inhibit the activity of the prolyl-hydroxylase PHD2, experiments with mouse liver showed that on a short timescale WR-1065 induced expression of the Hypoxia Inducible Factor HIF1α, lactate dehydrogenase LDH5, glucose transporter GLUT2, phosphorylated pyruvate dehydrogenase pPDH and PDH-kinase. This effect was confirmed on normal mouse NCTC hepatocytes, but not on hepatoma cells. A sharp reduction of acetyl-CoA and ATP levels in NCTC cells indicated reduced mitochondrial usage of pyruvate. Transient changes of mitochondrial membrane potential and reactive oxygen species ROS production were evident. Amifostine selectively protects NCTC cells against radiation, whilst HepG2 neoplastic cells are sensitized. The radiation protection was correlates with HIF levels. These findings shed new light on the mechanism of amifostine cytoprotection and encourage clinical research with this agent for the treatment of primary and metastatic liver cancer.


Subject(s)
Amifostine/pharmacology , Breast Neoplasms/radiotherapy , Radiation-Protective Agents/pharmacology , Adenosine Triphosphate/metabolism , Animals , Basal Metabolism/drug effects , Blood Glucose/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Glucose Transporter Type 2/metabolism , Glycolysis/drug effects , Glycolysis/radiation effects , Hepatocytes/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/drug effects , Liver/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mice, Inbred BALB C , Oxygen/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
8.
Sci Transl Med ; 8(328): 328ra29, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26936506

ABSTRACT

Disability or death due to intracerebral hemorrhage (ICH) is attributed to blood lysis, liberation of iron, and consequent oxidative stress. Iron chelators bind to free iron and prevent neuronal death induced by oxidative stress and disability due to ICH, but the mechanisms for this effect remain unclear. We show that the hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) family of iron-dependent, oxygen-sensing enzymes are effectors of iron chelation. Molecular reduction of the three HIF-PHD enzyme isoforms in the mouse striatum improved functional recovery after ICH. A low-molecular-weight hydroxyquinoline inhibitor of the HIF-PHD enzymes, adaptaquin, reduced neuronal death and behavioral deficits after ICH in several rodent models without affecting total iron or zinc distribution in the brain. Unexpectedly, protection from oxidative death in vitro or from ICH in vivo by adaptaquin was associated with suppression of activity of the prodeath factor ATF4 rather than activation of an HIF-dependent prosurvival pathway. Together, these findings demonstrate that brain-specific inactivation of the HIF-PHD metalloenzymes with the blood-brain barrier-permeable inhibitor adaptaquin can improve functional outcomes after ICH in several rodent models.


Subject(s)
Activating Transcription Factor 4/metabolism , Brain/pathology , Intracranial Hemorrhages/pathology , Molecular Targeted Therapy , Neurons/pathology , Oxygen/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Animals , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Genes, Reporter , Hemin/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracranial Hemorrhages/physiopathology , Iron/pharmacology , Iron Chelating Agents/pharmacology , Mice , Neurons/drug effects , Neuroprotective Agents/pharmacology , Procollagen-Proline Dioxygenase/metabolism , Protein Domains , Protein Isoforms/metabolism , Rats , Recovery of Function/drug effects
9.
ChemMedChem ; 9(3): 566-71, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24504543

ABSTRACT

The 2-oxoglutarate (2OG)-dependent Jumonji C domain (JmjC) family is the largest family of histone lysine demethylases. There is interest in developing small-molecule probes that modulate JmjC activity to investigate their biological roles. 5-Carboxy-8-hydroxyquinoline (IOX1) is the most potent broad-spectrum inhibitor of 2OG oxygenases, including the JmjC demethylases, reported to date; however, it suffers from low cell permeability. Here, we describe structure-activity relationship studies leading to the discovery of an n-octyl ester form of IOX1 with improved cellular potency (EC50 value of 100 to 4 µM). These findings are supported by in vitro inhibition and selectivity studies, docking studies, activity versus toxicity analysis in cell cultures, and intracellular uptake measurements. The n-octyl ester was found to have improved cell permeability; it was found to inhibit some JmjC demethylases in its intact ester form and to be more selective than IOX1. The n-octyl ester of IOX1 should find utility as a starting point for the development of JmjC inhibitors and as a use as a cell-permeable tool compound for studies investigating the roles of 2OG oxygenases in epigenetic regulation.


Subject(s)
Cell Membrane Permeability/drug effects , Enzyme Inhibitors/pharmacology , Esters/pharmacology , Hydroxyquinolines/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Esters/chemistry , HeLa Cells , Humans , Hydroxyquinolines/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship
10.
Mol Cell ; 44(2): 225-34, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-22017871

ABSTRACT

Regulation of gene expression plays an integral role in adaptation of cells to hypoxic stress. In mammals, prolyl hydroxylases control levels of the central transcription factor hypoxia inducible factor (HIF) through regulation of HIFα subunit stability. Here, we report that the hydroxylase Ofd1 regulates the Sre1 hypoxic transcription factor in fission yeast by controlling DNA binding. Prolyl hydroxylases require oxygen as a substrate, and the activity of Ofd1 regulates Sre1-dependent transcription. In the presence of oxygen, Ofd1 binds the Sre1 N-terminal transcription factor domain (Sre1N) and inhibits Sre1-dependent transcription by blocking DNA binding. In the absence of oxygen, the inhibitor Nro1 binds Ofd1, thereby releasing Sre1N and leading to activation of genes required for hypoxic growth. In contrast to the HIF system, where proline hydroxylation is essential for regulation, Ofd1 inhibition of Sre1N does not require hydroxylation and, thus, defines a new mechanism for hypoxic gene regulation.


Subject(s)
Oxygen/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/metabolism , Binding Sites , Cell Hypoxia/physiology , DNA, Fungal/metabolism , Hydroxylation , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces pombe Proteins/metabolism
11.
Structure ; 19(4): 503-14, 2011 Apr 13.
Article in English | MEDLINE | ID: mdl-21481773

ABSTRACT

Fission yeast protein Sre1, the homolog of the mammalian sterol regulatory element-binding protein (SREBP), is a hypoxic transcription factor required for sterol homeostasis and low-oxygen growth. Nro1 regulates the stability of the N-terminal transcription factor domain of Sre1 (Sre1N) by inhibiting the action of the prolyl 4-hydroxylase-like Ofd1 in an oxygen-dependent manner. The crystal structure of Nro1 determined at 2.2 Å resolution shows an all-α-helical fold that can be divided into two domains: a small N-terminal domain, and a larger C-terminal HEAT-repeat domain. Follow-up studies showed that Nro1 defines a new class of nuclear import adaptor that functions both in Ofd1 nuclear localization and in the oxygen-dependent inhibition of Ofd1 to control the hypoxic response.


Subject(s)
Cell Nucleus/metabolism , Nuclear Proteins/chemistry , Protein Structure, Secondary , Schizosaccharomyces pombe Proteins/chemistry , Sterols/biosynthesis , Active Transport, Cell Nucleus , Amino Acid Sequence , Anaerobiosis , Crystallography, X-Ray , Immunoprecipitation , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxygen/metabolism , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Protein Binding , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Sequence Homology, Amino Acid , Two-Hybrid System Techniques
12.
J Biol Chem ; 286(10): 8188-8196, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21209075

ABSTRACT

Huntington disease results from an expanded polyglutamine region in the N terminus of the huntingtin protein. HD pathology is characterized by neuronal degeneration and protein inclusions containing N-terminal fragments of mutant huntingtin. Structural information is minimal, though it is believed that mutant huntingtin polyglutamine adopts ß structure upon conversion to a toxic form. To this end, we designed mammalian cell expression constructs encoding compact ß variants of Htt exon 1 N-terminal fragment and tested their ability to aggregate and induce toxicity in cultured neuronal cells. In parallel, we performed molecular dynamics simulations, which indicate that constructs with expanded polyglutamine ß-strands are stabilized by main-chain hydrogen bonding. Finally, we found a correlation between the reactivity to 3B5H10, an expanded polyglutamine antibody that recognizes a compact ß rich hairpin structure, and the ability to induce cell toxicity. These data are consistent with an important role for a compact ß structure in mutant huntingtin-induced cell toxicity.


Subject(s)
Models, Biological , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Cell Line , Humans , Huntingtin Protein , Hydrogen Bonding , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...