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










Publication year range
1.
Chembiochem ; 24(5): e202200455, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36538283

ABSTRACT

The blue biliprotein phycocyanin, produced by photo-autotrophic cyanobacteria including spirulina (Arthrospira) and marketed as a natural food supplement or "nutraceutical," is reported to have anti-inflammatory, antioxidant, immunomodulatory, and anticancer activity. These diverse biological activities have been specifically attributed to the phycocyanin chromophore, phycocyanobilin (PCB). However, the mechanism of action of PCB and the molecular targets responsible for the beneficial properties of PCB are not well understood. We have developed a procedure to rapidly cleave the PCB pigment from phycocyanin by ethanolysis and then characterized it as an electrophilic natural product that interacts covalently with thiol nucleophiles but lacks any appreciable cytotoxicity or antibacterial activity against common pathogens and gut microbes. We then designed alkyne-bearing PCB probes for use in chemical proteomics target deconvolution studies. Target identification and validation revealed the cysteine protease legumain (also known as asparaginyl endopeptidase, AEP) to be a target of PCB. Inhibition of this target may account for PCB's diverse reported biological activities.


Subject(s)
Cysteine Proteases , Spirulina , Phycocyanin/pharmacology , Phycocyanin/chemistry , Phycobilins/pharmacology , Phycobilins/chemistry , Spirulina/chemistry , Dietary Supplements
2.
Angew Chem Int Ed Engl ; 62(9): e202212111, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36495310

ABSTRACT

Heme is a cofactor with myriad roles and essential to almost all living organisms. Beyond classical gas transport and catalytic functions, heme is increasingly appreciated as a tightly controlled signalling molecule regulating protein expression. However, heme acquisition, biosynthesis and regulation is poorly understood beyond a few model organisms, and the heme-binding proteome has not been fully characterised in bacteria. Yet as heme homeostasis is critical for bacterial survival, heme-binding proteins are promising drug targets. Herein we report a chemical proteomics method for global profiling of heme-binding proteins in live cells for the first time. Employing a panel of heme-based clickable and photoaffinity probes enabled the profiling of 32-54 % of the known heme-binding proteomes in Gram-positive and Gram-negative bacteria. This simple-to-implement profiling strategy could be interchangeably applied to different cell types and systems and fuel future research into heme biology.


Subject(s)
Proteome , Proteomics , Proteome/metabolism , Bacterial Proteins/metabolism , Heme-Binding Proteins/metabolism , Gram-Negative Bacteria/metabolism , Anti-Bacterial Agents/metabolism , Gram-Positive Bacteria , Bacteria/metabolism , Heme/chemistry
3.
iScience ; 25(8): 104787, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35992086

ABSTRACT

Despite much progress in developing better drugs, many patients with acute myeloid leukemia (AML) still die within a year of diagnosis. This is partly because it is difficult to identify therapeutic targets that are effective across multiple AML subtypes. One common factor across AML subtypes is the presence of a block in differentiation. Overcoming this block should allow for the identification of therapies that are not dependent on a specific mutation for their efficacy. Here, we used a phenotypic screen to identify compounds that stimulate differentiation in genetically diverse AML cell lines. Lead compounds were shown to decrease tumor burden and to increase survival in vivo. Using multiple complementary target deconvolution approaches, these compounds were revealed to be anti-mitotic tubulin disruptors that cause differentiation by inducing a G2-M mitotic arrest. Together, these results reveal a function for tubulin disruptors in causing differentiation of AML cells.

4.
Angew Chem Int Ed Engl ; 61(29): e202201136, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35286003

ABSTRACT

Cofactors are required for almost half of all enzyme reactions, but their functions and binding partners are not fully understood even after decades of research. Functionalised cofactor mimics that bind in place of the unmodified cofactor can provide answers, as well as expand the scope of cofactor activity. Through chemical proteomics approaches such as activity-based protein profiling, the interactome and localisation of the native cofactor in its physiological environment can be deciphered and previously uncharacterised proteins annotated. Furthermore, cofactors that supply functional groups to substrate biomolecules can be hijacked by mimics to site-specifically label targets and unravel the complex biology of post-translational protein modification. The diverse activity of cofactors has inspired the design of mimics for use as inhibitors, antibiotic therapeutics, and chemo- and biosensors, and cofactor conjugates have enabled the generation of novel enzymes and artificial DNAzymes.


Subject(s)
DNA, Catalytic , Proteomics
5.
Eur J Med Chem ; 220: 113431, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33915371

ABSTRACT

Duchenne muscular dystrophy is a fatal disease with no cure, caused by lack of the cytoskeletal protein dystrophin. Upregulation of utrophin, a dystrophin paralogue, offers a potential therapy independent of mutation type. The failure of first-in-class utrophin modulator ezutromid/SMT C1100 in Phase II clinical trials necessitates development of compounds with better efficacy, physicochemical and ADME properties and/or complementary mechanisms. We have discovered and performed a preliminary optimisation of a novel class of utrophin modulators using an improved phenotypic screen, where reporter expression is derived from the full genomic context of the utrophin promoter. We further demonstrate through target deconvolution studies, including expression analysis and chemical proteomics, that this compound series operates via a novel mechanism of action, distinct from that of ezutromid.


Subject(s)
Drug Discovery , Hydrazines/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Pyrimidines/pharmacology , Utrophin/metabolism , Dose-Response Relationship, Drug , Humans , Hydrazines/chemical synthesis , Hydrazines/chemistry , Molecular Structure , Muscular Dystrophy, Duchenne/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , RNA, Messenger/metabolism , Structure-Activity Relationship
6.
Bioorg Med Chem ; 28(22): 115724, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33128909

ABSTRACT

We have previously reported the discovery of a series of rhodanine-based inhibitors of the PIM family of serine/threonine kinases. Here we described the optimisation of those compounds to improve their physicochemical and ADME properties as well as reducing their off-targets activities against other kinases. Through molecular modeling and systematic structure activity relationship (SAR) studies, advanced molecules with high inhibitory potency, reduced off-target activity and minimal efflux were identified as new pan-PIM inhibitors. One example of an early lead, OX01401, was found to inhibit PIMs with nanomolar potency (15 nM for PIM1), inhibit proliferation of two PIM-expressing leukaemic cancer cell lines, MV4-11 and K562, and to reduce intracellular phosphorylation of a PIM substrate in a concentration dependent manner.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Thiazoles/pharmacology , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-pim-1/metabolism , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
7.
Drug Discov Today ; 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32971235

ABSTRACT

Investment in phenotypic drug discovery has led to increased demand for rapid and robust target deconvolution to aid successful drug development. Although methods for target identification and mechanism of action (MoA) discovery are flourishing, they typically lead to lists of putative targets. Validating which target(s) are involved in the therapeutic mechanism of a compound poses a significant challenge, requiring direct binding, target engagement, and functional studies in relevant physiological contexts. A combination of orthogonal approaches can allow target identification beyond the proteome as well as aid prioritisation for resource-intensive target validation studies.

8.
Biochem Soc Trans ; 48(3): 813-821, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32597486

ABSTRACT

Genetic approaches for the diagnosis and treatment of inherited muscle diseases have advanced rapidly in recent years. Many of the advances have occurred in the treatment of Duchenne muscular dystrophy (DMD), a muscle wasting disease where affected boys are typically wheelchair bound by age 12 years and generally die in their twenties from respiratory failure or cardiomyopathy. Dystrophin is a 421 kD protein which links F-actin to the extracellular matrix via the dystrophin-associated protein complex (DAPC) at the muscle membrane. In the absence of dystrophin, the DAPC is lost, making the muscle membrane more susceptible to contraction-induced injury. The identification of the gene causing DMD in 1986 resulted in improved diagnosis of the disease and the identification of hotspots for mutation. There is currently no effective treatment. However, there are several promising genetic therapeutic approaches at the preclinical stage or in clinical trials including read-through of stop codons, exon skipping, delivery of dystrophin minigenes and the modulation of expression of the dystrophin related protein, utrophin. In spite of significant progress, the problem of targeting all muscles, including diaphragm and heart at sufficiently high levels, remains a challenge. Any therapy also needs to consider the immune response and some treatments are mutation specific and therefore limited to a subgroup of patients. This short review provides a summary of the current status of DMD therapy with a particular focus on those genetic strategies that have been taken to the clinic.


Subject(s)
Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/therapy , CRISPR-Cas Systems , Codon, Terminator , Dystrophin/genetics , Exons , Genetic Therapy/methods , Humans , Male , Muscular Dystrophy, Duchenne/genetics
9.
Angew Chem Int Ed Engl ; 59(6): 2420-2428, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31755636

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease arising from mutations in the dystrophin gene. Upregulation of utrophin to compensate for the missing dystrophin offers a potential therapy independent of patient genotype. The first-in-class utrophin modulator ezutromid/SMT C1100 was developed from a phenotypic screen through to a Phase 2 clinical trial. Promising efficacy and evidence of target engagement was observed in DMD patients after 24 weeks of treatment, however trial endpoints were not met after 48 weeks. The objective of this study was to understand the mechanism of action of ezutromid which could explain the lack of sustained efficacy and help development of new generations of utrophin modulators. Using chemical proteomics and phenotypic profiling we show that the aryl hydrocarbon receptor (AhR) is a target of ezutromid. Several lines of evidence demonstrate that ezutromid binds AhR with an apparent KD of 50 nm and behaves as an AhR antagonist. Furthermore, other reported AhR antagonists also upregulate utrophin, showing that this pathway, which is currently being explored in other clinical applications including oncology and rheumatoid arthritis, could also be exploited in future DMD therapies.


Subject(s)
Benzoxazoles/chemistry , Naphthalenes/chemistry , Proteomics/methods , Receptors, Aryl Hydrocarbon/metabolism , Utrophin/metabolism , Animals , Benzoxazoles/metabolism , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Cycloaddition Reaction , Drug Design , Humans , Kinetics , Mice , Molecular Probes/chemistry , Muscular Dystrophy, Duchenne/drug therapy , Myoblasts/cytology , Myoblasts/metabolism , Naphthalenes/metabolism , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Protein Binding , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Up-Regulation/drug effects , Utrophin/agonists , Utrophin/genetics
10.
Bioorg Chem ; 94: 103395, 2020 01.
Article in English | MEDLINE | ID: mdl-31733898

ABSTRACT

Firefly luciferase (FLuc) is a powerful tool for molecular and cellular biology, and popular in high-throughput screening and drug discovery. However, FLuc assays have been plagued with positive and negative artefacts due to stabilisation and inhibition by small molecules from a range of chemical classes. Here we disclose Phase II clinical compound SMT C1100 for the treatment of Duchenne muscular dystrophy as an FLuc inhibitor (KD of 0.40 ±â€¯0.15 µM). Enzyme kinetic studies using SMT C1100 and other non-competitive inhibitors including resveratrol and NFκBAI4 identified previously undescribed modes of inhibition with respect to FLuc's luciferyl adenylate intermediate. Employing a photoaffinity strategy to identify SMT C1100's binding site, a photolabelled SMT C1100 probe instead underwent FLuc-dependent photooxidation. Our findings support novel binding sites on FLuc for non-competitive inhibitors.


Subject(s)
Benzoxazoles/pharmacology , Enzyme Inhibitors/pharmacology , Fireflies/enzymology , Luciferases, Firefly/antagonists & inhibitors , Animals , Benzoxazoles/chemical synthesis , Benzoxazoles/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Kinetics , Luciferases, Firefly/metabolism , Molecular Structure , Structure-Activity Relationship
11.
J Med Chem ; 63(5): 2547-2556, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31599580

ABSTRACT

5-(Ethylsulfonyl)-2-(naphthalen-2-yl)benzo[d]oxazole (ezutromid, 1) is a first-in-class utrophin modulator that has been evaluated in a phase 2 clinical study for the treatment of Duchenne muscular dystrophy (DMD). Ezutromid was found to undergo hepatic oxidation of its 2-naphthyl substituent to produce two regioisomeric 1,2-dihydronaphthalene-1,2-diols, DHD1 and DHD3, as the major metabolites after oral administration in humans and rodents. In many patients, plasma levels of the DHD metabolites were found to exceed those of ezutromid. Herein, we describe the structural elucidation of the main metabolites of ezutromid, the regio- and relative stereochemical assignments of DHD1 and DHD3, their de novo chemical synthesis, and their production in systems in vitro. We further elucidate the likely metabolic pathway and CYP isoforms responsible for DHD1 and DHD3 production and characterize their physicochemical, ADME, and pharmacological properties and their preliminary toxicological profiles.


Subject(s)
Benzoxazoles/metabolism , Muscular Dystrophy, Duchenne/drug therapy , Naphthalenes/metabolism , Naphthols/metabolism , Utrophin/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Benzoxazoles/adverse effects , Humans , Liver/drug effects , Liver/metabolism , Metabolic Networks and Pathways , Metabolome , Mice , Muscular Dystrophy, Duchenne/metabolism , Naphthalenes/adverse effects , Naphthols/adverse effects , Naphthols/analysis , Naphthols/chemical synthesis , Rats , Stereoisomerism
12.
J Tissue Viability ; 14(4): 124, 126, 128, passim, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15516100

ABSTRACT

Clinical assessment and management for anyone who has diabetes may be influenced by the development of the National Service Framework (NSF) for Diabetes. Through a case study, this article explains how the NSF for Diabetes and other recent NHS documentation has influenced our approach to managing a type 2 diabetic patient whose feet are categorised as 'high risk'. Some of the potential shortfalls of the NSF for Diabetes are also discussed in this context.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/diagnosis , Diabetic Neuropathies/therapy , Podiatry/methods , Aged , Humans , Male , Podiatry/standards , Practice Guidelines as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...