Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Methods Mol Biol ; 2648: 187-206, 2023.
Article in English | MEDLINE | ID: mdl-37039992

ABSTRACT

Molecular oxygen is essential for all multicellular life forms. In humans, the hypoxia-inducible factor (HIF) prolyl hydroxylase domain-containing enzymes (PHDs) serve as important oxygen sensors by regulating the activity of HIF, the master regulator that mediates cellular oxygen homeostasis, in an oxygen-dependent manner. In normoxia, PHDs catalyze the prolyl hydroxylation of HIF, which leads to its degradation and prevents cellular hypoxic response to be triggered. PHDs are current inhibition targets for the potential treatments of a number of diseases. In this chapter, we discuss in vitro and cell-based methods to study the modulation of PHD2, the most important human PHD isoform in normoxia and mild hypoxia. These include the production and purification of recombinant PHD2, the use of mass spectrometry to follow PHD2-catalyzed reactions and the studies of HIF stabilization in cells by immunoblotting.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Oxygen , Humans , Oxygen/metabolism , Hypoxia/metabolism , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/metabolism , Protein Isoforms
2.
Biochem Pharmacol ; 205: 115262, 2022 11.
Article in English | MEDLINE | ID: mdl-36191627

ABSTRACT

The role of epidermal growth factor receptor (EGFR) in non-small cell lung cancer (NSCLC) has been vastly studied over the last decade. This has led to the rapid development of many generations of EGFR tyrosine kinase inhibitors (EGFR-TKIs). However, patients treated with third-generation TKIs (osimertinib, avitinib and rociletinib) targeting the EGFR T790M mutation have shown emerging resistances and relapses. Therefore, further molecular understanding of NSCLC mutations, bypass signalling, tumour microenvironment and the existence of cancer stem cells to overcome such resistances is warranted. This will pave the way for designing novel and effective chemotherapies to improve patients' overall survival. In this review, we provide an overview of the multifaceted mechanisms of resistance towards EGFR-TKIs, as well as the challenges and perspectives that should be addressed in strategising chemotherapeutic treatments to overcome the ever-evolving and adaptive nature of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Protein Kinase Inhibitors/adverse effects , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/chemically induced , Neoplasm Recurrence, Local/drug therapy , Tumor Microenvironment
3.
Cells ; 10(9)2021 08 24.
Article in English | MEDLINE | ID: mdl-34571823

ABSTRACT

Cancer increases the global disease burden substantially, but it remains a challenge to manage it. The search for novel biomarkers is essential for risk assessment, diagnosis, prognosis, prediction of treatment response, and cancer monitoring. This paper examined NEDD8 ultimate buster-1 (NUB1) and F-adjacent transcript 10 (FAT10) proteins as novel biomarkers in cancer. This literature review is based on the search of the electronic database, PubMed. NUB1 is an interferon-inducible protein that mediates apoptotic and anti-proliferative actions in cancer, while FAT10 is a ubiquitin-like modifier that promotes cancer. The upregulated expression of both NUB1 and FAT10 has been observed in various cancers. NUB1 protein binds to FAT10 non-covalently to promote FAT10 degradation. An overexpressed FAT10 stimulates nuclear factor-kappa ß, activates the inflammatory pathways, and induces the proliferation of cancer. The FAT10 protein interacts with the mitotic arrest deficient 2 protein, causing chromosomal instability and breast tumourigenesis. FAT10 binds to the proliferating cell nuclear antigen protein and inhibits the DNA damage repair response. In addition, FAT10 involves epithelial-mesenchymal transition, invasion, apoptosis, and multiplication in hepatocellular carcinoma. Our knowledge about them is still limited. There is a need to further develop NUB1 and FAT10 as novel biomarkers.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Ubiquitins/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation/physiology , Humans , Neoplasms/pathology
4.
Chembiochem ; 21(11): 1647-1655, 2020 06 02.
Article in English | MEDLINE | ID: mdl-31919953

ABSTRACT

The hypoxia-inducible factors (HIFs) are key transcription factors in determining cellular responses involving alterations in protein levels in response to limited oxygen availability in animal cells. 2-Oxoglutarate-dependent oxygenases play key roles in regulating levels of HIF and its transcriptional activity. We describe MS-based proteomics studies in which we compared the results of subjecting human breast cancer MCF-7 cells to hypoxia or treating them with a cell-penetrating derivative (dimethyl N-oxalylglycine; DMOG) of the stable 2OG analogue N-oxalylglycine. The proteomic results are consistent with reported transcriptomic analyses and support the proposed key roles of 2OG-dependent HIF prolyl- and asparaginyl-hydroxylases in the hypoxic response. Differences between the data sets for hypoxia and DMOG might reflect context-dependent effects or HIF-independent effects of DMOG.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Cell Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Oxygen/pharmacology , Proteome/genetics , Transcriptome , Atlases as Topic , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MCF-7 Cells , Metabolic Networks and Pathways/genetics , Molecular Sequence Annotation , Proteome/classification , Proteome/metabolism , Proteomics/methods
5.
J Biol Chem ; 291(39): 20661-73, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27502280

ABSTRACT

The hypoxia-inducible factor (HIF) system orchestrates cellular responses to hypoxia in animals. HIF is an α/ß-heterodimeric transcription factor that regulates the expression of hundreds of genes in a tissue context-dependent manner. The major hypoxia-sensing component of the HIF system involves oxygen-dependent catalysis by the HIF hydroxylases; in humans there are three HIF prolyl hydroxylases (PHD1-3) and an asparaginyl hydroxylase (factor-inhibiting HIF (FIH)). PHD catalysis regulates HIFα levels, and FIH catalysis regulates HIF activity. How differences in HIFα hydroxylation status relate to variations in the induction of specific HIF target gene transcription is unknown. We report studies using small molecule HIF hydroxylase inhibitors that investigate the extent to which HIF target gene expression is induced by PHD or FIH inhibition. The results reveal substantial differences in the role of prolyl and asparaginyl hydroxylation in regulating hypoxia-responsive genes in cells. PHD inhibitors with different structural scaffolds behave similarly. Under the tested conditions, a broad-spectrum 2-oxoglutarate dioxygenase inhibitor is a better mimic of the overall transcriptional response to hypoxia than the selective PHD inhibitors, consistent with an important role for FIH in the hypoxic transcriptional response. Indeed, combined application of selective PHD and FIH inhibitors resulted in the transcriptional induction of a subset of genes not fully responsive to PHD inhibition alone. Thus, for the therapeutic regulation of HIF target genes, it is important to consider both PHD and FIH activity, and in the case of some sets of target genes, simultaneous inhibition of the PHDs and FIH catalysis may be preferable.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/biosynthesis , Transcription, Genetic/physiology , Cell Hypoxia/physiology , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , MCF-7 Cells
6.
Elife ; 5: e11469, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26901438

ABSTRACT

Transcription factor (TF) networks determine cell-type identity by establishing and maintaining lineage-specific expression profiles, yet reconstruction of mammalian regulatory network models has been hampered by a lack of comprehensive functional validation of regulatory interactions. Here, we report comprehensive ChIP-Seq, transgenic and reporter gene experimental data that have allowed us to construct an experimentally validated regulatory network model for haematopoietic stem/progenitor cells (HSPCs). Model simulation coupled with subsequent experimental validation using single cell expression profiling revealed potential mechanisms for cell state stabilisation, and also how a leukaemogenic TF fusion protein perturbs key HSPC regulators. The approach presented here should help to improve our understanding of both normal physiological and disease processes.


Subject(s)
Gene Regulatory Networks , Hematopoiesis , Hematopoietic Stem Cells/physiology , Transcription Factors/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , Computer Simulation , Gene Expression Profiling , Mice , Models, Theoretical , Sequence Analysis, DNA
7.
Mol Aspects Med ; 47-48: 54-75, 2016.
Article in English | MEDLINE | ID: mdl-26791432

ABSTRACT

In human cells oxygen levels are 'sensed' by a set of ferrous iron and 2-oxoglutarate dependent dioxygenases. These enzymes regulate a broad range of cellular and systemic responses to hypoxia by catalysing the post-translational hydroxylation of specific residues in the alpha subunits of hypoxia inducible factor (HIF) transcriptional complexes. The HIF hydroxylases are now the subject of pharmaceutical targeting by small molecule inhibitors that aim to activate or augment the endogenous HIF transcriptional response for the treatment of anaemia and other hypoxic human diseases. Here we consider the rationale for this therapeutic strategy from the biochemical, biological and medical perspectives. We outline structural and mechanistic considerations that are relevant to the design of HIF hydroxylase inhibitors, including likely determinants of specificity, and review published reports on their activity in pre-clinical models and clinical trials.


Subject(s)
Drug Delivery Systems , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , Anemia/drug therapy , Animals , Disease Models, Animal , Erythropoietin/deficiency , Erythropoietin/metabolism , Humans , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Inflammation/drug therapy , Ischemia/drug therapy , Mixed Function Oxygenases/metabolism , Oxygen/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Protein Conformation , Randomized Controlled Trials as Topic
8.
PLoS One ; 10(7): e0132004, 2015.
Article in English | MEDLINE | ID: mdl-26147748

ABSTRACT

As part of the cellular adaptation to limiting oxygen availability in animals, the expression of a large set of genes is activated by the upregulation of the hypoxia-inducible transcription factors (HIFs). Therapeutic activation of the natural human hypoxic response can be achieved by the inhibition of the hypoxia sensors for the HIF system, i.e. the HIF prolyl-hydroxylases (PHDs). Here, we report studies on tricyclic triazole-containing compounds as potent and selective PHD inhibitors which compete with the 2-oxoglutarate co-substrate. One compound (IOX4) induces HIFα in cells and in wildtype mice with marked induction in the brain tissue, revealing that it is useful for studies aimed at validating the upregulation of HIF for treatment of cerebral diseases including stroke.


Subject(s)
Brain/enzymology , Enzyme Inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Stroke/drug therapy , Triazoles , Animals , Brain/pathology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , MCF-7 Cells , Mice , Stroke/enzymology , Stroke/pathology , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology
9.
Eur J Med Chem ; 94: 509-16, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25023609

ABSTRACT

Protein-protein interactions between the hypoxia inducible factor (HIF) and the transcriptional coactivators p300/CBP are potential cancer targets due to their role in the hypoxic response. A natural product based screen led to the identification of indandione and benzoquinone derivatives that reduce the tight interaction between a HIF-1α fragment and the CH1 domain of p300. The indandione derivatives were shown to fragment to give ninhydrin, which was identified as the active species. Both the naphthoquinones and ninhydrin were observed to induce Zn(II) ejection from p300 and the catalytic domain of the histone demethylase KDM4A. Together with previous reports on the effects of related compounds on HIF-1α and other systems, the results suggest that care should be taken in interpreting biological results obtained with highly electrophilic/thiol modifying compounds.


Subject(s)
E1A-Associated p300 Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indans/pharmacology , Organometallic Compounds/pharmacology , Quinones/pharmacology , Zinc/pharmacology , Dose-Response Relationship, Drug , Humans , Indans/chemistry , Models, Molecular , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Protein Binding/drug effects , Quinones/chemistry , Structure-Activity Relationship , Zinc/chemistry
10.
Nat Commun ; 5: 3423, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24594748

ABSTRACT

Accumulation of (R)-2-hydroxyglutarate in cells results from mutations to isocitrate dehydrogenase that correlate with cancer. A recent study reports that (R)-, but not (S)-2-hydroxyglutarate, acts as a co-substrate for the hypoxia-inducible factor prolyl hydroxylases via enzyme-catalysed oxidation to 2-oxoglutarate. Here we investigate the mechanism of 2-hydroxyglutarate-enabled activation of 2-oxoglutarate oxygenases, including prolyl hydroxylase domain 2, the most important human prolyl hydroxylase isoform. We observe that 2-hydroxyglutarate-enabled catalysis by prolyl hydroxylase domain 2 is not enantiomer-specific and is stimulated by ferrous/ferric ion and reducing agents including L-ascorbate. The results reveal that 2-hydroxyglutarate is oxidized to 2-oxoglutarate non-enzymatically, likely via iron-mediated Fenton-chemistry, at levels supporting in vitro catalysis by 2-oxoglutarate oxygenases. Succinic semialdehyde and succinate are also identified as products of 2-hydroxyglutarate oxidation. Overall, the results rationalize the reported effects of 2-hydroxyglutarate on catalysis by prolyl hydroxylases in vitro and suggest that non-enzymatic 2-hydroxyglutarate oxidation may be of biological interest.


Subject(s)
Alcohol Oxidoreductases/metabolism , Glutarates/chemistry , Glutarates/metabolism , Oxygenases/metabolism , Ascorbic Acid/metabolism , Succinic Acid/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/metabolism
11.
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
12.
ACS Chem Biol ; 8(7): 1488-96, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23683440

ABSTRACT

The hypoxia inducible factor (HIF) system is central to the signaling of low oxygen (hypoxia) in animals. The levels of HIF-α isoforms are regulated in an oxygen-dependent manner by the activity of the HIF prolyl-hydroxylases (PHD or EGLN enzymes), which are Fe(II) and 2-oxoglutarate (2OG) dependent oxygenases. Here, we describe biochemical, crystallographic, cellular profiling, and animal studies on PHD inhibitors including selectivity studies using a representative set of human 2OG oxygenases. We identify suitable probe compounds for use in studies on the functional effects of PHD inhibition in cells and in animals.


Subject(s)
Heterocyclic Compounds/chemical synthesis , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Small Molecule Libraries/chemistry , Animals , Animals, Genetically Modified , Biological Assay , Cell Line , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Signal Transduction , Small Molecule Libraries/pharmacology , Zebrafish/embryology , Zebrafish/genetics
13.
Org Biomol Chem ; 11(5): 732-745, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23151668

ABSTRACT

Inhibition of the hypoxia-inducible factor (HIF) prolyl hydroxylases (PHD or EGLN enzymes) is of interest for the treatment of anemia and ischemia-related diseases. Most PHD inhibitors work by binding to the single ferrous ion and competing with 2-oxoglutarate (2OG) co-substrate for binding at the PHD active site. Non-specific iron chelators also inhibit the PHDs, both in vitro and in cells. We report the identification of dual action PHD inhibitors, which bind to the active site iron and also induce the binding of a second iron ion at the active site. Following analysis of small-molecule iron complexes and application of non-denaturing protein mass spectrometry to assess PHD2·iron·inhibitor stoichiometry, selected diacylhydrazines were identified as PHD2 inhibitors that induce the binding of a second iron ion. Some compounds were shown to inhibit the HIF hydroxylases in human hepatoma and renal carcinoma cell lines.


Subject(s)
Hydrazines/chemistry , Hydrazines/pharmacology , Iron/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/metabolism , Catalytic Domain , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Molecular Docking Simulation , Procollagen-Proline Dioxygenase/chemistry , Protein Binding/drug effects , Spectrometry, Mass, Electrospray Ionization
14.
Chem Sci ; 4(8): 3110-3117, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-26682036

ABSTRACT

2-Oxoglutarate and iron dependent oxygenases are therapeutic targets for human diseases. Using a representative 2OG oxygenase panel, we compare the inhibitory activities of 5-carboxy-8-hydroxyquinoline (IOX1) and 4-carboxy-8-hydroxyquinoline (4C8HQ) with that of two other commonly used 2OG oxygenase inhibitors, N-oxalylglycine (NOG) and 2,4-pyridinedicarboxylic acid (2,4-PDCA). The results reveal that IOX1 has a broad spectrum of activity, as demonstrated by the inhibition of transcription factor hydroxylases, representatives of all 2OG dependent histone demethylase subfamilies, nucleic acid demethylases and γ-butyrobetaine hydroxylase. Cellular assays show that, unlike NOG and 2,4-PDCA, IOX1 is active against both cytosolic and nuclear 2OG oxygenases without ester derivatisation. Unexpectedly, crystallographic studies on these oxygenases demonstrate that IOX1, but not 4C8HQ, can cause translocation of the active site metal, revealing a rare example of protein ligand-induced metal movement.

15.
J Med Chem ; 55(14): 6639-43, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22724510

ABSTRACT

The JmjC oxygenases catalyze the N-demethylation of N(ε)-methyl lysine residues in histones and are current therapeutic targets. A set of human 2-oxoglutarate analogues were screened using a unified assay platform for JmjC demethylases and related oxygenases. Results led to the finding that daminozide (N-(dimethylamino)succinamic acid, 160 Da), a plant growth regulator, selectively inhibits the KDM2/7 JmjC subfamily. Kinetic and crystallographic studies reveal that daminozide chelates the active site metal via its hydrazide carbonyl and dimethylamino groups.


Subject(s)
Enzyme Inhibitors/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Plant Growth Regulators/pharmacology , Succinates/pharmacology , Humans , Inhibitory Concentration 50 , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Molecular , Protein Conformation , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...