skip to content

Department of Pharmacology

 

Reversing the triad of anaesthesia in a cannot intubate, cannot oxygenate emergency. Comment on Br J Anaesth 2024; 133: 190-2

Fri, 06/06/2025 - 11:00

Br J Anaesth. 2025 Jun 5:S0007-0912(25)00294-6. doi: 10.1016/j.bja.2025.04.044. Online ahead of print.

NO ABSTRACT

PMID:40480913 | DOI:10.1016/j.bja.2025.04.044

Computationally designed haemagglutinin with nanocage plug-and-display elicits pan-H5 influenza vaccine responses

Fri, 06/06/2025 - 11:00

Emerg Microbes Infect. 2025 Jun 6:2511132. doi: 10.1080/22221751.2025.2511132. Online ahead of print.

ABSTRACT

The increasing spread of highly pathogenic avian influenza (HPAI) A/H5 viruses poses a pandemic threat. Circulating clade 2.3.4.4b viruses have demonstrated rapid transcontinental dissemination, extensive reassortment, epizootic spread and potential sustained mammal-to-mammal transmission, signifying a heightened risk of becoming a human pathogen of high consequence. A broadly protective, future-proof vaccine against multiple clades of H5 influenza is urgently needed for pandemic preparedness. Here, we combine two novel vaccine technologies to generate a Digitally Immune Optimised and Selected H5 antigen (DIOSvax-H5inter) displayed multivalently on the mi3 nanocage using the SpyTag003/SpyCatcher003 conjugation system. Mice immunised with DIOSvax-H5inter Homotypic Nanocages at low doses demonstrate potent, cross-clade neutralising antibody and T cell responses against diverse H5 strains. DIOSvax-H5inter Homotypic Nanocages provide a scalable vaccine candidate with the potential for pan-H5 protection against drifted or newly emergent H5 strains. This World Health Organization preferred characteristic is essential for prospective strategic stockpiling in the pre-pandemic phase.Trial registration: ClinicalTrials.gov identifier: NCT06145178..

PMID:40476519 | DOI:10.1080/22221751.2025.2511132

Functional characterization of the store-operated calcium entry pathway in naked mole-rat cells

Tue, 03/06/2025 - 11:00

Open Biol. 2025 Jun;15(6):250052. doi: 10.1098/rsob.250052. Epub 2025 Jun 4.

ABSTRACT

Naked mole-rats (NMRs, Heterocephalus glaber) are highly unusual rodents exhibiting remarkable adaptations to their subterranean habitat and resistance to developing various age-related diseases such as those related to abnormal cell proliferation or cancer, neurodegeneration and inflammation. In other rodents, as well as humans, a ubiquitous Ca2+ influx pathway, namely the store-operated Ca2+ entry (SOCE), has been implicated in all these diseases. SOCE is triggered by intracellular Ca2+ store depletion resulting in interaction of Stim proteins with Orai proteins, the putative homologues of which appear to be present in the NMR genome, but no functional characterization of SOCE in NMRs has yet been conducted. In this study, we provide the first functional and pharmacological characterization of SOCE in NMR using both excitable and non-excitable cells.

PMID:40460874 | DOI:10.1098/rsob.250052

Mpox multiprotein virus-like nanoparticle vaccine induces neutralizing and protective antibodies in mice and non-human primates

Wed, 21/05/2025 - 11:00

Nat Commun. 2025 May 21;16(1):4726. doi: 10.1038/s41467-025-59826-8.

ABSTRACT

The upsurge of mpox in Africa and the recent global outbreak have stimulated the development of new vaccines and therapeutics. We describe the construction of virus-like particle (VLP) vaccines in which modified M1, A35 and B6 proteins from monkeypox virus (MPXV) clade Ia are conjugated individually or together to a scaffold that accommodates up to 60 ligands using the SpyTag/SpyCatcher nanoparticle system. Immunisation of female mice with VLPs induces higher anti-MPXV and anti-vaccinia virus (VACV) neutralizing antibodies than their soluble protein (SP) counterparts or modified VACV Ankara (MVA). Vaccination with individual single protein VLPs provides partial protection against lethal respiratory infections with VACV or MPXV clade IIa, whereas combinations or a chimeric VLP with all three antigens provide complete protection that is superior to SPs. Additionally, the VLP vaccine reduces the replication and spread of the virus at intranasal and intrarectal sites of inoculation. VLPs induce higher neutralizing activity than the Jynneos vaccine in rhesus macaques, and the VLP-induced antiserum provides better protection against MPXV and VACV than the Jynneos-induced antiserum when passively transferred to female mice. These data demonstrate that an mpox VLP vaccine derived from three MPXV clade Ia proteins protects against clade IIa MPXV and VACV, indicating cross-reactivity for orthopoxviruses.

PMID:40399314 | DOI:10.1038/s41467-025-59826-8

Loss-of-function Gα<sub>s</sub> rare disease variants exert mutation-specific effects on GPCR signaling

Tue, 20/05/2025 - 11:00

Sci Signal. 2025 May 20;18(887):eado7543. doi: 10.1126/scisignal.ado7543. Epub 2025 May 20.

ABSTRACT

G protein-coupled receptors (GPCRs) are transmembrane detectors of extracellular signals that activate heterotrimeric G proteins to regulate intracellular responses. Because there are only 16 Gα proteins that can couple to GPCRs, variation in a single Gα can affect the function of numerous receptors. Here, we investigated two mutant forms of Gαs (L388R and E392K) that are associated with pseudohypoparathyroidism type Ic (PHPIc), a maternally inherited rare disease. Gαs is encoded by an imprinted gene, resulting in the mutant form of Gαs being the only version of the protein present in certain tissues, which leads to tissue-specific disease manifestations. By integrating data from three-dimensional structures, GPCR-G protein coupling specificity, transcriptomics, biophysics, and molecular dynamics with systems pharmacology modeling, we identified GPCRs whose signaling could be altered by Gαs mutations in the kidney, a tissue involved in the pathophysiology of PHPIc. Analysis of G protein activation by the parathyroid hormone receptor 1 (PTH1R) revealed that L388R impaired Gαs interaction with the receptor, whereas E392K reduced the receptor-induced activation of heterotrimeric Gs. This indicates that different signal transduction steps can be altered by specific Gαs mutants associated with the same disease. These findings highlight the importance of investigating mutation-specific perturbations in GPCR signaling to suggest patient-specific treatment strategies. Furthermore, our methods provide a blueprint for interrogating GPCR signaling diversity in different physiological and pathophysiological contexts.

PMID:40392940 | DOI:10.1126/scisignal.ado7543

A platform for SpyCatcher conjugation to native antibodies

Mon, 19/05/2025 - 11:00

Chem Sci. 2025 May 6. doi: 10.1039/d5sc02286j. Online ahead of print.

ABSTRACT

Protein-antibody conjugates represent major advancements in targeted therapeutics. However, platforms enabling 'off-the-shelf' antibody conjugation are seldom reported. The SpyTag/SpyCatcher system, known for its stable isopeptide bond formation, is widely used to engineer protein architectures and study protein folding. This work introduces the fusion of SpyCatcher with native antibodies using cysteine-reactive tetra-divinylpyrimidine (TetraDVP)-SpyTag linkers. This platform allows for the rapid and stable conjugation of a native antibody with SpyCatcher proteins. As a proof of concept, the HER2-targeting antibody trastuzumab was conjugated to different SpyCatcher proteins using a TetraDVP-SpyTag linker, producing robust conjugates that retained specific binding to HER2-positive cells with excellent conversion rates. To demonstrate the platform's broader applicability, the TetraDVP-SpyTag linker was successfully conjugated to additional native IgG1 and IgG4 antibodies (durvalumab, brentuximab, cetuximab, and gemtuzumab) with similarly high efficiency as trastuzumab. Moreover, a scalable solid-phase synthesis of TetraDVP linkers has been developed, achieving high yields and purity. This innovative platform enables precise, single-step antibody bioconjugation, offering strong potential for protein-antibody conjugate synthesis. With applications across therapeutics and diagnostics, this method advances antibody-based drug development.

PMID:40386161 | PMC:PMC12080404 | DOI:10.1039/d5sc02286j

Tandem-repeat proteins introduce tuneable properties to engineered biomolecular condensates

Fri, 16/05/2025 - 11:00

Chem Sci. 2025 May 5. doi: 10.1039/d5sc00903k. Online ahead of print.

ABSTRACT

The cell's ability to rapidly partition biomolecules into biomolecular condensates is linked to a diverse range of cellular functions. Understanding how the structural attributes of biomolecular condensates are linked with their biological roles can be facilitated by the development of synthetic condensate systems that can be manipulated in a controllable and predictable way. Here, we design and characterise a tuneable synthetic biomolecular condensate platform fusing modular consensus-designed tetratricopeptide repeat (CTPR) proteins to intrinsically-disordered domains. Trends between the CTPR structural attributes and condensate propensity were recapitulated across different experimental conditions and by in silico modelling, demonstrating that the CTPR domain can systematically affect the condensates in a predictable manner. Moreover, we show that incorporating short binding motifs into the CTPR domain results in specific target-protein recruitment into the condensates. Our model system can be rationally designed in a versatile manner to both tune condensate propensity and endow the condensates with new functions.

PMID:40375868 | PMC:PMC12076082 | DOI:10.1039/d5sc00903k

Using a stable protein scaffold to display peptides that bind to alpha-synuclein fibrils

Thu, 15/05/2025 - 11:00

Protein Sci. 2025 Jun;34(6):e70150. doi: 10.1002/pro.70150.

ABSTRACT

Amyloid fibrils are ordered aggregates that are a pathological hallmark of many neurodegenerative disorders including Alzheimer's disease and Parkinson's disease. The process of amyloid formation involves a complex cascade by which soluble monomeric protein converts to insoluble, ordered aggregates (amyloid fibrils). Although inhibiting the aggregation pathway is a key target for therapeutic development, the heterogeneous collection of aggregation-prone species formed in this process, including oligomers, protofibrils, and fibrils, represents other targets for modifying disease pathology. Developing molecules that can bind to amyloid fibrils and potentially disrupt the harmful interactions between the fibrils and the cellular components would be advantageous. Designing peptide modulators for α-synuclein aggregation is of great interest; however, effective inhibitory peptides are often hydrophobic and hence difficult to handle. Therefore, developing strategies to display these peptides in a soluble scaffold would be very beneficial. Here we demonstrate that the ultra-stable consensus-designed tetratricopeptide repeat (CTPR) protein scaffold can be grafted with "KLVFF" derived peptides previously identified to inhibit protein aggregation and interact with amyloid fibrils to produce proteins that bind along the surface of α-synuclein fibrils with micromolar affinity. Given the ability to insert hydrophobic peptides to produce soluble, CTPR-based binders, this method may prove beneficial in screening for peptide modulators of protein aggregation.

PMID:40371781 | DOI:10.1002/pro.70150

CHD8 interacts with BCL11A to induce oncogenic transcription in triple negative breast cancer

Tue, 06/05/2025 - 11:00

EMBO J. 2025 May 6. doi: 10.1038/s44318-025-00447-8. Online ahead of print.

ABSTRACT

The identification of tumour-specific protein-protein interactions remains a challenge for the development of targeted cancer therapies. In this study we describe our approach for the identification of triple negative breast cancer (TNBC)-specific protein-protein interactions focusing on the oncogene BCL11A. We used a proteomic approach to identify the BCL11A protein networks in TNBC and compared it to its network in B-cells, a cell type in which BCL11A plays crucial roles. This approach identified the chromatin remodeller CHD8 as a TNBC-specific interaction partner of BCL11A. We show that CHD8 also plays a key role in TNBC pathogenesis, with detailed multi-omics analysis revealing that BCL11A and CHD8 co-regulate several targets and synergise to drive tumour development and progression. Using a battery of biophysical assays, we confirm that the BCL11A-CHD8 interaction is direct and identify chemical fragments that disrupt this interaction and affect downstream targets, decreasing proliferation in 3D colony assays. Our study provides a proof-of-principle approach for investigating tumour-specific protein-protein interactions and identifies lead chemical compounds that could be developed into novel therapeutics for TNBC.

PMID:40328966 | DOI:10.1038/s44318-025-00447-8

Nanoparticle-Mediated Targeted Protein Degradation: An Emerging Therapeutics Technology

Mon, 05/05/2025 - 11:00

Angew Chem Int Ed Engl. 2025 May 5:e202503958. doi: 10.1002/anie.202503958. Online ahead of print.

ABSTRACT

Targeted protein degradation (TPD) has emerged as a transformative therapeutic strategy for eliminating disease-associated proteins, with relevance across disorders ranging from cancer to neurodegeneration. Since its inception nearly two decades ago, TPD has attracted strong academic and commercial interest, with multiple candidates advancing into clinical trials. Despite this progress, the field faces persistent challenges, including limited solubility, poor cellular uptake, and unpredictable structure-activity relationship of small-molecule degraders, which complicate rational design. To address these limitations, alternative platforms such as nanoparticle-mediated protein degraders (NanoPDs) have gained attention. First reported 17 years ago, NanoPDs harness a diverse array of materials, degradation mechanisms, and linker chemistries to achieve protein clearance through novel pathways. While promising, their clinical translation remains constrained by barriers such as lysosomal entrapment, protein corona formation, and biocompatibility concerns. In this review, we present a comprehensive overview of the current landscape of nanoparticle-mediated TPD. We emphasize the design principles underlying nano-bio interfaces and explore the role of proximity-induced biology as a mechanism for orchestrating protein interactions. Finally, we highlight critical challenges and key questions that must be addressed to fully realise the therapeutic potential of NanoPDs.

PMID:40324952 | DOI:10.1002/anie.202503958

Nanomolar therapeutic concentrations of statins rapidly induce cerebral artery vasoconstriction by stimulating L-type calcium channels

Sun, 04/05/2025 - 11:00

Biochem Pharmacol. 2025 May 2:116970. doi: 10.1016/j.bcp.2025.116970. Online ahead of print.

ABSTRACT

All commonly prescribed statins have been reported to cause reversible memory loss within weeks of therapy, though the exact molecular mechanism remains unknown. However, whether therapeutic concentrations of statins can directly regulate the contractility of resistance cerebral arteries that control cerebrovascular perfusion remains unexplored. Here, we examined the acute vascular effects of statins on rat cerebral arteries and the underlying molecular mechanisms. Our pressure myography data demonstrate that, at therapeutically-relevant nanomolar concentrations, statins produced a robust and rapid vasoconstriction, appearing within 2-3 min of drug application. Interestingly, such vasoconstriction was largely absent in female rat cerebral arteries. Endothelial denudation or mevalonate supplementation did not alter statin-induced vasoconstriction, suggesting an endothelium- and cholesterol-independent mechanism. In contrast, such vasoconstriction was abolished upon removal of extracellular Ca2+, pharmacological blockade of the smooth muscle cell voltage-gated Ca2+ channel, CaV1.2, or siRNA knockdown of CaV1.2 - all of which reduced [Ca2+]i, indicating that Ca2+ entry through CaV1.2 plays a critical role in cerebral artery vasoconstriction. Arterial biotinylation revealed that acute statin exposure did not alter the surface expression, distribution, or function of CaV1.2 channels. Altogether, our data unveil an unexpected role of statins in rapidly inducing constriction of resistance cerebral arteries by directly stimulating CaV1.2 in smooth muscle cells. These findings offer a plausible explanation for statin-associated reversible memory impairment, its mitigation by calcium channel blockers, and why such effects may not be observed in all subjects, particularly those concurrently taking antihypertensive agents.

PMID:40320051 | DOI:10.1016/j.bcp.2025.116970

Recent developments in probing the levels and flux of selected organellar cations as well as organellar mechanosensitivity

Sun, 04/05/2025 - 11:00

Curr Opin Chem Biol. 2025 May 3;87:102600. doi: 10.1016/j.cbpa.2025.102600. Online ahead of print.

ABSTRACT

Electrochemical gradients exist not only across the plasma membrane (PM) but also across membranes of organelles. Various endomembrane-localised ion channels and transporters have been identified, the activity of which is critical for organellar (and also cellular) ionic homeostasis that underpins diverse cellular processes. Aberrant organellar ion flux underlies several diseases, identifying organellar channels and transporters as potential drug targets. Therefore, the need for probing the functions of these proteins in situ cannot be overemphasised. The acidic interior of a few organelles as well as the dynamic nature of most organelles historically presented challenges for reliable estimation of luminal ionic concentrations. But there have been significant methodological and technical advancements by now, allowing measurement of levels of specific ions within these organelles as well as their flux across endomembranes with increasing precision. Evidence also continues to amass reporting mechanosensitivity of the endomembranes and its physiological significance. Here we highlight some recent developments in tools and techniques for measuring the levels and movement of some selected organellar cations as well as organellar mechanosensitivity.

PMID:40319567 | DOI:10.1016/j.cbpa.2025.102600

Cellular parameters shaping pathways of targeted protein degradation

Fri, 02/05/2025 - 11:00

Commun Biol. 2025 May 2;8(1):691. doi: 10.1038/s42003-025-08104-w.

ABSTRACT

In recent years the development of proteolysis-targeting chimeras (PROTACs) has enhanced the field of ubiquitin signalling through advancing therapeutic targeted protein degradation (TPD) strategies and generating tools to explore the ubiquitin landscape. However, the interplay between PROTACs and their substrates, and other components of the ubiquitin proteasome system (UPS), raises fundamental questions about cellular parameters that might influence the action of PROTACs and the amenability of a given target to PROTAC-mediated degradation. In this perspective we discuss examples of cellular parameters that have been shown to influence PROTAC sensitivity and consider others likely to be important for PROTAC-mediated target degradation but not yet routinely considered in design of novel TPD strategies: Target localisation and accessibility on the one hand, and expression patterns, localisation and activity of E3 ligases, deubiquitinases (DUBs) and wider ubiquitin machinery on the other, are critical parameters in the exploitation of PROTACs, and establishing a better understanding of these parameters will facilitate the rational design of PROTACs.

PMID:40316744 | DOI:10.1038/s42003-025-08104-w

Agonist efficacy at the β<sub>2</sub>AR is driven by the faster association rate of the G<sub>s</sub> protein

Fri, 11/04/2025 - 11:00

Front Pharmacol. 2025 Mar 27;16:1367991. doi: 10.3389/fphar.2025.1367991. eCollection 2025.

ABSTRACT

INTRODUCTION: The β2-adrenoceptor (β2AR) is a class A G protein-coupled receptor (GPCR). It is therapeutically relevant in asthma and chronic obstructive pulmonary disease (COPD), where β2AR agonists relieve bronchoconstriction. The β2AR is a prototypical GPCR for structural and biophysical studies. However, the molecular basis of agonist efficacy at the β2AR is not understood. We hypothesised that the kinetics of GPCR-G protein interactions could play a role in determining ligand efficacy. By studying a range of agonists with varying efficacy, we examined the relationship between ligand-induced mini-Gs binding to the β2AR and ligand efficacy, along with the ability of individual ligands to activate the G protein in cells.

METHODS: We used NanoBRET technology to measure ligand-induced binding of purified Venus-mini-Gs to β2AR-nLuc in membrane preparations under both equilibrium and kinetic conditions. In addition, we examined the ability of these β2AR agonists to activate the heterotrimeric Gs protein, measured using the Gs-CASE protein biosensor in living cells. This assay detects a reduction in NanoBRET between the nano-luciferase (nLuc) donor on the Gα subunit and Venus acceptor on the Gγ upon Gs protein activation.

RESULTS: The 12 β2AR agonists under study revealed a broad range of ligand potency and efficacy values in the cellular Gs-CASE assays. Kinetic characterisation of mini-Gs binding to the agonist β2AR complex revealed a strong correlation between ligand efficacy values (Emax) and mini-Gs affinity (K d) and its association rate (k on). In contrast, there was no correlation between ligand efficacy and reported ligand dissociation rates (or residence times).

CONCLUSION: The association rate (k on) of the G protein to the agonist β2AR complex is directly correlated with ligand efficacy. These data support a model in which higher-efficacy agonists induce the β2AR to adopt a conformation that is more likely to recruit G protein. Conversely, these data did not support the role of agonist binding kinetics in determining the molecular basis of efficacy.

PMID:40213684 | PMC:PMC11983327 | DOI:10.3389/fphar.2025.1367991

Targeting Bacterial Cells with DNA Nanostructures

Wed, 02/04/2025 - 11:00

Methods Mol Biol. 2025;2901:103-115. doi: 10.1007/978-1-0716-4394-5_8.

ABSTRACT

DNA origami nanostructures offer wide potential for controlled functionalization with molecules of interest, such as peptides, enzymes, small-molecule drugs, and fluorophores. Here, we describe a protocol for the synthesis and characterization of aptamer-modified DNA origami nanostructures that can act as a vehicle for delivering antimicrobials to Gram-positive and Gram-negative bacterial targets in a specific and efficient manner.

PMID:40175870 | DOI:10.1007/978-1-0716-4394-5_8

Differential neuropilin isoform expressions highlight plasticity in macrophages in the heterogenous TME through <em>in-silico</em> profiling

Wed, 26/03/2025 - 10:00

Front Immunol. 2025 Mar 11;16:1547330. doi: 10.3389/fimmu.2025.1547330. eCollection 2025.

ABSTRACT

INTRODUCTION: The nuanced roles of neuropilin (NRP) isoforms, NRP1 and NRP2, have attracted considerable scientific interest regarding cancer progression. Their differential expressions across various cancer types are specific to NRP isoforms which are shown in a cancer type-dependent manner. It accounts for the different mechanisms involved, driven by a co-expression of gene-sets associated with overexpressed NRP1 or NRP2. Their different expressions on tumour-associated macrophages (TAMs) with disparate markers are associated with the heterogenous tumour microenvironment (TME) through their plasticity and pro-tumorigenic activities.

METHODS: Single-cell RNA sequencing (scRNA-seq) analyses were performed on tumours from clear cell Renal Cell Carcinoma (ccRCC) and skin cutaneous melanoma (SKCM) which exhibit the highest expressions of NRP1 and NRP2, respectively. Datasets were processed using established bioinformatics pipelines, including clustering algorithms, to determine cellular heterogeneity and quantify NRP isoform expression within distinct macrophage populations. Using differential gene expression analysis (DEGA) alongside co-enrichment studies, we explored gene-sets associated with NRP1 or NRP2 overexpression in TAMs.

RESULTS: Our analysis revealed a marked upregulation of NRP1 in TAMs isolated from ccRCC and elevated NRP2 expression in SKCM-derived TAMs. Both NRP1+ and NRP2+ macrophages showed an M2-like polarisation characterised by immune suppression and extracellular matrix degradation. Coupled with the previously uncharacterised NRP isoform specific- subpopulations within these cancers identified by DEGA, co-enrichment analyses demonstrated that the upregulation of gene-sets associated with NRP1 is associated with angiogenesis and tumour progression through VEGF signalling, while gene-sets with NRP2 showed dual functionality in the TME-dependent manner. Their distinct roles in regulating macrophage plasticity, tumour invasion, and metastasis were highlighted.

DISCUSSION: These findings underscore distinct isoform-specific mechanisms by which NRP1 and NRP2 contribute to TAM-mediated cancer progression. This study aims to establish a foundation for future research, leading to biological experiments with focused gene-sets derived from our findings. This approach can contribute to the development of immunomodulatory strategies targeting specific NRP isoforms in macrophages, tailored to individual cancer types and abnormal expressions of those gene markers, potentially offering a more effective therapeutic approach compared to broad-spectrum NRP inhibition strategies.

PMID:40134439 | PMC:PMC11933088 | DOI:10.3389/fimmu.2025.1547330

Pro-inflammatory mediators sensitise Transient Receptor Potential Melastatin 3 cation channel (TRPM3) function in mouse sensory neurons

Sun, 02/03/2025 - 11:00

Neuropharmacology. 2025 Feb 28:110391. doi: 10.1016/j.neuropharm.2025.110391. Online ahead of print.

ABSTRACT

Pro-inflammatory mediators can directly activate pain-sensing neurons, known as nociceptors. Additionally, these mediators can sensitise ion channels and receptors expressed by these cells through transcriptional and post-translational modulation, leading to nociceptor hypersensitivity. A well-characterised group of ion channels that subserve nociceptor sensitisation is the transient receptor potential (TRP) superfamily of cation channels. For example, the roles of TRP channels vanilloid 1 (TRPV1) and ankyrin 1 (TRPA1) in nociceptor sensitisation and inflammatory pain have been extensively documented. In the case of TRP melastatin 3 (TRPM3), however, despite the increasing recognition of this channel's role in inflammatory pain, the mediators driving its sensitisation during inflammation remain poorly characterised. Here, using Ca2+ imaging, we found that an inflammatory soup of bradykinin, interleukin 1β (IL-1β) and tumour necrosis factor α (TNFα) sensitised TRPM3 function in isolated mouse sensory neurons; IL-1β and TNFα, but not bradykinin, independently potentiated TRPM3 function. TRPM3 expression and translocation to the membrane remained unchanged upon individual or combined exposure to these inflammatory mediators, which suggests that post-translational modification might occur. Finally, using the complete Freund's adjuvant-induced model of knee inflammation, we found that systemic pharmacological blockade of TRPM3 does not alleviate inflammatory pain (as assessed through evaluation of digging behaviour and dynamic weight bearing), which contrasts with previous reports using different pain models. We propose that the nuances of the immune response may determine the relative contribution of TRPM3 to nociceptive signalling in different neuro-immune contexts. Collectively, our findings improve insight into the role of TRPM3 sensitisation in inflammatory pain.

PMID:40024472 | DOI:10.1016/j.neuropharm.2025.110391

Large scale investigation of GPCR molecular dynamics data uncovers allosteric sites and lateral gateways

Thu, 27/02/2025 - 11:00

Nat Commun. 2025 Feb 27;16(1):2020. doi: 10.1038/s41467-025-57034-y.

ABSTRACT

G protein-coupled receptors (GPCRs) constitute a functionally diverse protein family and are targets for a broad spectrum of pharmaceuticals. Technological progress in X-ray crystallography and cryogenic electron microscopy has enabled extensive, high-resolution structural characterisation of GPCRs in different conformational states. However, as highly dynamic events underlie GPCR signalling, a complete understanding of GPCR functionality requires insights into their conformational dynamics. Here, we present a large dataset of molecular dynamics simulations covering 60% of currently available GPCR structures. Our analysis reveals extensive local "breathing" motions of the receptor on a nano- to microsecond timescale and provides access to numerous previously unexplored receptor conformational states. Furthermore, we reveal that receptor flexibility impacts the shape of allosteric drug binding sites, which frequently adopt partially or completely closed states in the absence of a molecular modulator. We demonstrate that exploring membrane lipid dynamics and their interaction with GPCRs is an efficient approach to expose such hidden allosteric sites and even lateral ligand entrance gateways. The obtained insights and generated dataset on conformations, allosteric sites and lateral entrance gates in GPCRs allows us to better understand the functionality of these receptors and opens new therapeutic avenues for drug-targeting strategies.

PMID:40016203 | DOI:10.1038/s41467-025-57034-y