Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
bioRxiv ; 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37873072

ABSTRACT

Computational modelling of microbiome metabolism has proved instrumental to catalyse our understanding of diet-host-microbiome-disease interactions through the interrogation of mechanistic, strain- and molecule-resolved metabolic models. We present APOLLO, a resource of 247,092 human microbial genome-scale metabolic reconstructions spanning 19 phyla and accounting for microbial genomes from 34 countries, all age groups, and five body sites. We explored the metabolic potential of the reconstructed strains and developed a machine learning classifier able to predict with high accuracy the taxonomic strain assignments. We also built 14,451 sample-specific microbial community models, which could be stratified by body site, age, and disease states. Finally, we predicted faecal metabolites enriched or depleted in gut microbiomes of people with Crohn's disease, Parkinson disease, and undernourished children. APOLLO is compatible with the human whole-body models, and thus, provide unprecedented opportunities for systems-level modelling of personalised host-microbiome co-metabolism. APOLLO will be freely available under https://www.vmh.life/.

2.
ChemMedChem ; 17(1): e202100398, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34581506

ABSTRACT

Studies on the inhibition of the human 2-oxoglutarate dependent oxygenase JMJD6, which is a cancer target, by 2-oxoglutarate mimics / competitors, including human drugs, drug candidates, and metabolites relevant to cancer are described. JMJD6 assays employed NMR to monitor inhibitor binding and use of mass spectrometry to monitor JMJD6-catalysed lysine hydroxylation. Notably, some clinically applied prolyl hydroxylase inhibitors also inhibit JMJD6. The results will help enable the development of inhibitors selective for human oxygenases, including JMJD6.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Ketoglutaric Acids/pharmacology , Prolyl-Hydroxylase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Ketoglutaric Acids/chemistry , Molecular Structure , Prolyl-Hydroxylase Inhibitors/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship
3.
Annu Rev Microbiol ; 75: 199-222, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34314593

ABSTRACT

The human microbiome plays an important role in human health and disease. Meta-omics analyses provide indispensable data for linking changes in microbiome composition and function to disease etiology. Yet, the lack of a mechanistic understanding of, e.g., microbiome-metabolome links hampers the translation of these findings into effective, novel therapeutics. Here, we propose metabolic modeling of microbial communities through constraint-based reconstruction and analysis (COBRA) as a complementary approach to meta-omics analyses. First, we highlight the importance of microbial metabolism in cardiometabolic diseases, inflammatory bowel disease, colorectal cancer, Alzheimer disease, and Parkinson disease. Next, we demonstrate that microbial community modeling can stratify patients and controls, mechanistically link microbes with fecal metabolites altered in disease, and identify host pathways affected by the microbiome. Finally, we outline our vision for COBRA modeling combined with meta-omics analyses and multivariate statistical analyses to inform and guide clinical trials, yield testable hypotheses, and ultimately propose novel dietary and therapeutic interventions.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Precision Medicine
4.
BMC Biol ; 18(1): 62, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32517799

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a systemic disease clinically defined by the degeneration of dopaminergic neurons in the brain. While alterations in the gut microbiome composition have been reported in PD, their functional consequences remain unclear. Herein, we addressed this question by an analysis of stool samples from the Luxembourg Parkinson's Study (n = 147 typical PD cases, n = 162 controls). RESULTS: All individuals underwent detailed clinical assessment, including neurological examinations and neuropsychological tests followed by self-reporting questionnaires. Stool samples from these individuals were first analysed by 16S rRNA gene sequencing. Second, we predicted the potential secretion for 129 microbial metabolites through personalised metabolic modelling using the microbiome data and genome-scale metabolic reconstructions of human gut microbes. Our key results include the following. Eight genera and seven species changed significantly in their relative abundances between PD patients and healthy controls. PD-associated microbial patterns statistically depended on sex, age, BMI, and constipation. Particularly, the relative abundances of Bilophila and Paraprevotella were significantly associated with the Hoehn and Yahr staging after controlling for the disease duration. Furthermore, personalised metabolic modelling of the gut microbiomes revealed PD-associated metabolic patterns in the predicted secretion potential of nine microbial metabolites in PD, including increased methionine and cysteinylglycine. The predicted microbial pantothenic acid production potential was linked to the presence of specific non-motor symptoms. CONCLUSION: Our results suggest that PD-associated alterations of the gut microbiome can translate into substantial functional differences affecting host metabolism and disease phenotype.


Subject(s)
Gastrointestinal Microbiome/physiology , Parkinson Disease/metabolism , Aged , Case-Control Studies , Female , Humans , Luxembourg , Male , Middle Aged , Parkinson Disease/microbiology , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/analysis
5.
Cell Rep ; 29(7): 1767-1777.e8, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31722195

ABSTRACT

Parkinson's disease (PD) exhibits systemic effects on the human metabolism, with emerging roles for the gut microbiome. Here, we integrate longitudinal metabolome data from 30 drug-naive, de novo PD patients and 30 matched controls with constraint-based modeling of gut microbial communities derived from an independent, drug-naive PD cohort, and prospective data from the general population. Our key results are (1) longitudinal trajectory of metabolites associated with the interconversion of methionine and cysteine via cystathionine differed between PD patients and controls; (2) dopaminergic medication showed strong lipidomic signatures; (3) taurine-conjugated bile acids correlated with the severity of motor symptoms, while low levels of sulfated taurolithocholate were associated with PD incidence in the general population; and (4) computational modeling predicted changes in sulfur metabolism, driven by A. muciniphila and B. wadsworthia, which is consistent with the changed metabolome. The multi-omics integration reveals PD-specific patterns in microbial-host sulfur co-metabolism that may contribute to PD severity.


Subject(s)
Gastrointestinal Microbiome , Parkinson Disease/microbiology , Sulfur/metabolism , Aged , Female , Humans , Longitudinal Studies , Male , Middle Aged
6.
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
7.
Cell Chem Biol ; 24(11): 1377-1387.e3, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28965728

ABSTRACT

The human retrovirus HTLV-1 causes a hematological malignancy or neuroinflammatory disease in ∼10% of infected individuals. HTLV-1 primarily infects CD4+ T lymphocytes and persists as a provirus integrated in their genome. HTLV-1 appears transcriptionally latent in freshly isolated cells; however, the chronically active anti-HTLV-1 cytotoxic T cell response observed in infected individuals indicates frequent proviral expression in vivo. The kinetics and regulation of HTLV-1 proviral expression in vivo are poorly understood. By using hypoxia, small-molecule hypoxia mimics, and inhibitors of specific metabolic pathways, we show that physiologically relevant levels of hypoxia, as routinely encountered by circulating T cells in the lymphoid organs and bone marrow, significantly enhance HTLV-1 reactivation from latency. Furthermore, culturing naturally infected CD4+ T cells in glucose-free medium or chemical inhibition of glycolysis or the mitochondrial electron transport chain strongly suppresses HTLV-1 plus-strand transcription. We conclude that glucose metabolism and oxygen tension regulate HTLV-1 proviral latency and reactivation in vivo.


Subject(s)
Glucose/metabolism , Human T-lymphotropic virus 1/physiology , Oxygen/metabolism , Amino Acids, Dicarboxylic/pharmacology , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Hypoxia , Cells, Cultured , Citric Acid Cycle/drug effects , Electron Transport Chain Complex Proteins/metabolism , Epigenesis, Genetic , Glycolysis/drug effects , Histones/genetics , Histones/metabolism , Human T-lymphotropic virus 1/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ketoglutaric Acids/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Mitochondria/drug effects , Mitochondria/metabolism , Virus Latency , Virus Replication/drug effects
8.
Kidney Int ; 92(4): 900-908, 2017 10.
Article in English | MEDLINE | ID: mdl-28506759

ABSTRACT

Complement C1q is part of the C1 macromolecular complex that mediates the classical complement activation pathway: a major arm of innate immune defense. C1q is composed of A, B, and C chains that require post-translational prolyl 4-hydroxylation of their N-terminal collagen-like domain to enable the formation of the functional triple helical multimers. The prolyl 4-hydroxylase(s) that hydroxylate C1q have not previously been identified. Recognized prolyl 4-hydroxylases include collagen prolyl-4-hydroxylases (CP4H) and the more recently described prolyl hydroxylase domain (PHD) enzymes that act as oxygen sensors regulating hypoxia-inducible factor (HIF). We show that several small-molecule prolyl hydroxylase inhibitors that activate HIF also potently suppress C1q secretion by human macrophages. However, reducing oxygenation to a level that activates HIF does not compromise C1q hydroxylation. In vitro studies showed that a C1q A chain peptide is not a substrate for PHD2 but is a substrate for CP4H1. Circulating levels of C1q did not differ between wild-type mice or mice with genetic deficits in PHD enzymes, but were reduced by prolyl hydroxylase inhibitors. Thus, C1q is hydroxylated by CP4H, but not the structurally related PHD hydroxylases. Hence, reduction of C1q levels may be an important off-target side effect of small molecule PHD inhibitors developed as treatments for renal anemia.


Subject(s)
Complement C1q/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia/metabolism , Macrophages/metabolism , Procollagen-Proline Dioxygenase/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Anemia/drug therapy , Anemia/etiology , Animals , Cell Line , Complement C1q/analysis , Complement Pathway, Classical , Female , Humans , Hydroxylation , Kidney Diseases/blood , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Prolyl-Hydroxylase Inhibitors/therapeutic use , Protein Processing, Post-Translational
9.
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.

10.
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
11.
Structure ; 23(4): 639-52, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25728928

ABSTRACT

Post-translational ribosomal protein hydroxylation is catalyzed by 2-oxoglutarate (2OG) and ferrous iron dependent oxygenases, and occurs in prokaryotes and eukaryotes. OGFOD1 catalyzes trans-3 prolyl hydroxylation at Pro62 of the small ribosomal subunit protein uS12 (RPS23) and is conserved from yeasts to humans. We describe crystal structures of the human uS12 prolyl 3-hydroxylase (OGFOD1) and its homolog from Saccharomyces cerevisiae (Tpa1p): OGFOD1 in complex with the broad-spectrum 2OG oxygenase inhibitors; N-oxalylglycine (NOG) and pyridine-2,4-dicarboxylate (2,4-PDCA) to 2.1 and 2.6 Å resolution, respectively; and Tpa1p in complex with NOG, 2,4-PDCA, and 1-chloro-4-hydroxyisoquinoline-3-carbonylglycine (a more selective prolyl hydroxylase inhibitor) to 2.8, 1.9, and 1.9 Å resolution, respectively. Comparison of uS12 hydroxylase structures with those of other prolyl hydroxylases, including the human hypoxia-inducible factor (HIF) prolyl hydroxylases (PHDs), reveals differences between the prolyl 3- and prolyl 4-hydroxylase active sites, which can be exploited for developing selective inhibitors of the different subfamilies.


Subject(s)
Carrier Proteins/chemistry , Nuclear Proteins/chemistry , Prolyl-Hydroxylase Inhibitors/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Amino Acid Sequence , Binding Sites , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Humans , Molecular Docking Simulation , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Binding , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
12.
Biochim Biophys Acta ; 1839(12): 1416-32, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24859458

ABSTRACT

N-Methylation of lysine and arginine residues has emerged as a major mechanism of transcriptional regulation in eukaryotes. In humans, N(ε)-methyllysine residue demethylation is catalysed by two distinct subfamilies of demethylases (KDMs), the flavin-dependent KDM1 subfamily and the 2-oxoglutarate- (2OG) dependent JmjC subfamily, which both employ oxidative mechanisms. Modulation of histone methylation status is proposed to be important in epigenetic regulation and has substantial medicinal potential for the treatment of diseases including cancer and genetic disorders. This article provides an introduction to the enzymology of the KDMs and the therapeutic possibilities and challenges associated with targeting them, followed by a review of reported KDM inhibitors and their mechanisms of action from kinetic and structural perspectives.


Subject(s)
Histone Demethylases/metabolism , Molecular Targeted Therapy/methods , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/chemistry , Humans , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Binding
13.
Bioorg Med Chem ; 22(11): 3030-54, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24758871

ABSTRACT

A naphthoquinone inhibitor of human arylamine N-acetyltransferase 1 (hNAT1), a potential cancer biomarker and therapeutic target, has been reported which undergoes a distinctive concomitant color change from red to blue upon binding to the enzyme. Here we describe the use of in silico modeling alongside structure-activity relationship studies to advance the hit compound towards a potential probe to quantify hNAT1 levels in tissues. Derivatives with both a fifty-fold higher potency against hNAT1 and a two-fold greater absorption coefficient compared to the initial hit have been synthesized; these compounds retain specificity for hNAT1 and its murine homologue mNat2 over the isoenzyme hNAT2. A relationship between pKa, inhibitor potency and colorimetric properties has also been uncovered. The high potency of representative examples against hNAT1 in ZR-75-1 cell extracts also paves the way for the development of inhibitors with improved intrinsic sensitivity which could enable detection of hNAT1 in tissue samples and potentially act as tools for elucidating the unknown role hNAT1 plays in ER+ breast cancer; this could in turn lead to a therapeutic use for such inhibitors.


Subject(s)
Arylamine N-Acetyltransferase/antagonists & inhibitors , Biomarkers, Tumor/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Naphthoquinones/pharmacology , Arylamine N-Acetyltransferase/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colorimetry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Isoenzymes/metabolism , Models, Molecular , Molecular Structure , Naphthoquinones/chemical synthesis , Naphthoquinones/chemistry , Structure-Activity Relationship
14.
PLoS One ; 8(8): e70600, 2013.
Article in English | MEDLINE | ID: mdl-23940600

ABSTRACT

Human arylamine N-acetyltransferase 1 (hNAT1) has become an attractive potential biomarker for estrogen-receptor-positive breast cancers. We describe here the mechanism of action of a selective non-covalent colorimetric biosensor for the recognition of hNAT1 and its murine homologue, mNat2, over their respective isoenzymes, leading to new opportunities in diagnosis. On interaction with the enzyme, the naphthoquinone probe undergoes an instantaneous and striking visible color change from red to blue. Spectroscopic, chemical, molecular modelling and biochemical studies reported here show that the color change is mediated by selective recognition between the conjugate base of the sulfonamide group within the probe and the conjugate acid of the arginine residue within the active site of both hNAT1 and mNat2. This represents a new mechanism for selective biomarker sensing and may be exploited as a general approach to the specific detection of biomarkers in disease.


Subject(s)
Arylamine N-Acetyltransferase/chemistry , Arylamine N-Acetyltransferase/metabolism , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Color , Isoenzymes/chemistry , Isoenzymes/metabolism , Naphthoquinones/chemistry , Naphthoquinones/metabolism , Animals , Catalytic Domain , Female , Humans , Mice , Protein Binding
15.
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.

SELECTION OF CITATIONS
SEARCH DETAIL
...