1
|
Sun H, Liu X, Lu J, Fan H, Lu D, Sun H, Zhou Z, Li Y, Yin X, Song Y, Wang S, Xin T. A multi-omics target study for glioblastoma multiforme (GBM) based on Mendelian randomization analysis. IBRO Neurosci Rep 2025; 18:400-408. [PMID: 40124114 PMCID: PMC11928806 DOI: 10.1016/j.ibneur.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/23/2025] [Indexed: 03/25/2025] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most frequent type of primary malignant brain tumor. This study utilized Mendelian randomization (MR) analysis to explore the causal link between proteins in plasma and cerebrospinal fluid and GBM. Aims This study aimed to identify proteins in both plasma and cerebrospinal fluid (CSF) that could serve as potential therapeutic targets for GBM. Methods We employed previously published protein quantitative trait loci (pQTL) data from CSF and plasma as the exposure data, alongside aggregated Genome-Wide Association Study (GWAS) data on GBM for our MR analysis. Furthermore, we conducted Bayesian co-localization analysis and examined the protein-protein interaction (PPI) networks of CSF and plasma proteins related to GBM risk. Results MR identified three key proteins linked to GBM risk: ribophorin I (RPN1) in plasma, von Willebrand factor (vWF) and macrophage-stimulating protein (MSP). in CSF. Elevated RPN1 and MSP were associated with decreased GBM risk, while increased vWF was linked to higher risk. External validation confirmed that RPN1 served as a key protein in GBM development. Bayesian co-localization showed a 10.35 % probability of a shared causal variant between RPN1 and GBM. Protein-protein interaction analysis further highlighted related proteins for RPN1. Conclusions In summary, the plasma protein RPN1 and the CSF proteins vWF and MSP are causally associated with the risk of GBM. Further research is needed to clarify the roles of these candidate proteins in GBM. Notably, RPN1 may serve as a potential therapeutic target for GBM. Future clinical studies on GBM treatment could explore drugs targeting RPN1.
Collapse
Affiliation(s)
- Hao Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Xiangyin Liu
- Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250000, China
| | - Jiaze Lu
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Hao Fan
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Dongxiao Lu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Haohan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Zijian Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Yuming Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Xianyong Yin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Yuwen Song
- Department of Opthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Shan Wang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Tao Xin
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| |
Collapse
|
2
|
Shen C, Zuo Q, Shao Z, Lin Y, Chen S. Research progress in myocardial function and diseases related to muscarinic acetylcholine receptor (Review). Int J Mol Med 2025; 55:86. [PMID: 40183403 PMCID: PMC12005369 DOI: 10.3892/ijmm.2025.5527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Muscarinic acetylcholine (ACh) receptors (also known as M receptors) are widely distributed in all organs and tissues of the body, mainly playing a role in cholinergic nerve conduction. There are five known subtypes of muscarinic ACh receptors, but their pharmacological mechanisms of action on myocardial function have remained to be clearly defined. Functional myocardial diseases and myocardial injuries, such as arrhythmia, myocardial ischemia, myocarditis and myocardial fibrosis, may be affected by muscarinic ACh receptors. This article reviews the research progress of the regulation of myocardial function by muscarinic ACh receptors and related diseases, with the aim of developing better strategies and providing references for further revealing and clarifying the signal transduction and mechanisms of muscarinic ACh receptors in cardiomyocytes, and finding potential myocardial protective drugs that act on muscarinic ACh receptors.
Collapse
Affiliation(s)
- Chuqiao Shen
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Qiang Zuo
- Department of Cardiology, First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Zhengbin Shao
- Department of Cardiology, First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Yixuan Lin
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Shuo Chen
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230038, P.R. China
| |
Collapse
|
3
|
Li C, Zhang K, Zhao J. Genome-wide Mendelian randomization mapping the influence of plasma proteome on major depressive disorder. J Affect Disord 2025; 376:1-9. [PMID: 39892755 DOI: 10.1016/j.jad.2025.01.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Plasma proteins play critical roles in a series of biological processes and represent a major source of translational biomarkers and drug targets. In this study, we performed Mendelian randomization (MR) to explore potential causal associations of protein quantitative trait loci (pQTL, n = 54,219) with major depressive disorder (MDD) using summary statistics from the PGC (n = 143,265) and further replicated in FinnGen cohort (n = 406,986). Subsequently, gene expression quantitative trait loci (eQTL) of identified proteins were leveraged to validate the primary findings in both PGC and FinnGen cohorts. We implemented reverse causality detection using bidirectional MR analysis, Steiger test, Bayesian co-localization and phenotype scanning to further strengthen the MR findings. In primary analyses, MR analysis revealed 2 plasma protein significantly associated with MDD risk at Bonferroni correction (P < 3.720 × 10-5), including butyrophilin subfamily 2 member A1 (BTN2A1, OR = 0.860; 95 % CI, 0.825-0.895; P = 1.79 × 10-5) and butyrophilin subfamily 3 member A2 (BTN3A2, OR = 1.071; 95 % CI, 1.056-1.086; P = 3.89 × 10-6). Both the identified proteins had no reverse causality. Bayesian co-localization indicated that BTN2A1 (coloc.abf-PPH4 = 0.620) and BTN3A2 (coloc.abf-PPH4 = 0.872) exhibited a shared variant with MDD, a finding that was subsequently validated by HEIDI test. In the replication stage, BTN2A1 and BTN3A2 were successfully validated in the FinnGen cohort. This study genetically determined BTN2A1 and BTN3A2 were associated with MDD and these findings may have clinical implications for MDD prevention.
Collapse
Affiliation(s)
- Chong Li
- Department of Psychiatry, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue Zhong, Guangzhou, Guangdong 510220, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Dadao Road North, Guangzhou, Guangdong 510515, China
| | - Jiubo Zhao
- Department of Psychiatry, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue Zhong, Guangzhou, Guangdong 510220, China; Department of Psychology, School of Public Health, Southern Medical University, No. 1838 Guangzhou Dadao Road North, Guangzhou, Guangdong 510220, China.
| |
Collapse
|
4
|
Jayakody T, Budagoda DK, Mendis K, Dilshan WD, Bethmage D, Dissasekara R, Dawe GS. Biased agonism in peptide-GPCRs: A structural perspective. Pharmacol Ther 2025; 269:108806. [PMID: 39889970 DOI: 10.1016/j.pharmthera.2025.108806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/13/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
G protein-coupled receptors (GPCRs) are dynamic membrane receptors that transduce extracellular signals to the cell interior by forming a ligand-receptor-effector (ternary) complex that functions via allosterism. Peptides constitute an important class of ligands that interact with their cognate GPCRs (peptide-GPCRs) to form the ternary complex. "Biased agonism", a therapeutically relevant phenomenon exhibited by GPCRs owing to their allosteric nature, has also been observed in peptide-GPCRs, leading to the development of selective therapeutics with fewer side effects. In this review, we have focused on the structural basis of signalling bias at peptide-GPCRs of classes A and B, and reviewed the therapeutic relevance of bias at peptide-GPCRs, with the hope of contributing to the discovery of novel biased peptide drugs.
Collapse
Affiliation(s)
- Tharindunee Jayakody
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | | | - Krishan Mendis
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | | | - Duvindu Bethmage
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | - Rashmi Dissasekara
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka; The Graduate School, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
5
|
Yao P, Mazidi M, Pozarickij A, Iona A, Wright N, Lin K, Millwood I, Fry H, Kartsonaki C, Chen Y, Yang L, Du H, Avery D, Schmidt D, Sun D, Lv J, Yu C, Hill M, Bennett D, Walters R, Li L, Clarke R, Chen Z. Proteome-Wide Genetic Study in East Asians and Europeans Identified Multiple Therapeutic Targets for Ischemic Stroke. Stroke 2025. [PMID: 40304040 DOI: 10.1161/strokeaha.125.050982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/18/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Analyses of genomic and proteomics data in prospective biobank studies in diverse populations may discover novel or repurposing drug targets for stroke. METHODS We extracted individual cis-protein quantitative trait locus for 2923 proteins measured using Olink Explore panel from a genome-wide association study in prospective China Kadoorie Biobank and UK Biobank, both established ≈20 years ago. These cis-protein quantitative trait loci were used in ancestry-specific 2-sample Mendelian randomization analyses of ischemic stroke (IS) in East Asians (n=22 664 cases) and Europeans (n=62 100 cases). We further undertook colocalization analyses to examine the shared causal variants of cis-protein quantitative trait locus with stroke, along with various downstream analyses (eg, phenome-wide association study, drug development lookups) to clarify mechanisms of action and druggability. RESULTS In Mendelian randomization analyses, the genetically predicted plasma levels of 10 proteins were significantly associated with IS in East Asians (n=2) and Europeans (n=9), with 6 proteins (FGF5 [fibroblast growth factor 5], TMPRSS5 [transmembrane protease serine 5], FURIN, F11 [coagulation factor XI], ALDH2 [aldehyde dehydrogenase 2], and ABO) showing positive and 4 (GRK5 [G protein-coupled receptor kinase 5], KIAA0319 [dyslexia-associated protein KIAA0319], PROCR [endothelial protein C receptor], and MMP12 [macrophage metalloelastase 12]) showing inverse associations, all directionally consistent between East Asians and Europeans. Colocalization analyses provided strong evidence (posterior probabilities for the H4 hypothesis ≥0.7) of shared genetic variants with IS for 9 out of 10 proteins (except ABO). Moreover, 8 proteins were also causally associated, in the expected directions, with systolic blood pressure (positive/inverse: 4/2), low-density lipoprotein cholesterol (1 positive), body mass index (1 inverse), type 2 diabetes (2/1), or atrial fibrillation (3/1). Phenome-wide association study analyses and lookups in knock-out mouse models confirmed their importance for IS or stroke-related traits (eg, hematologic phenotypes). Of these 10 proteins, 1 was not druggable (ABO), 3 had known primary (F11) or potentially repurposed (ALDH2, MMP12) drug targets for stroke, and 6 (PROCR, GRK5, FGF5, FURIN, KIAA0319, and TMPRSS5) had no evidence of any drug targets. CONCLUSIONS Proteogenomic investigation in diverse ancestry populations identified the causal relevance of 10 proteins for IS, with several being potentially novel or repurposed targets that could be prioritized for further investigation.
Collapse
Affiliation(s)
- Pang Yao
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Mohsen Mazidi
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Alfred Pozarickij
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Andri Iona
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Neil Wright
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Kuang Lin
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Iona Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Hannah Fry
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Huaidong Du
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Daniel Avery
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Dan Schmidt
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Michael Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Derrick Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Robin Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Robert Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| |
Collapse
|
6
|
Yao T, Luo J, Han X, Yi H, Zhang H, Pan W, Xue Q, Liu X, Fu J, Zhang A. From Nuclear Receptors to GPCRs: a Deep Transfer Learning Approach for Enhanced Environmental Estrogen Recognition. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025. [PMID: 40306980 DOI: 10.1021/acs.est.5c02014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Environmental estrogens (EEs), as typical endocrine-disrupting chemicals (EDCs), can bind to classic estrogen receptors (ERs) to induce genomic effects, as well as to G protein-coupled estrogen receptor (GPER) located on the membrane, thereby inducing downstream nongenomic effects rapidly. However, due to the relatively scarce ligand data, receptor-based or ligand-based screening models for GPER are challenging. Inspired by functional similarity between GPER and ER, this study constructs a deep transfer learning model named GPNET to predict potential GPER-binding ligands by using three-dimensional (3D) molecular surface electrostatic potential point clouds (SepPC) as input. The model retains a part of molecular structural knowledge learned from the ER ligands and then trains the remaining parameters of the model using the GPER ligands, ultimately obtaining the GPNET model, which effectively predicts the binding activity of compounds with GPER. GPNET outperforms From Scratch (nontransfer) model on the small data set, achieving the area under the receiver operating characteristic (ROC) curve (AUCROC) of 0.898 on the validation set and 0.863 on the test set, respectively. Furthermore, by visualizing the critical points and extracting the features from activation points of active ligands, the study provides a more in-depth interpretation of the molecular mechanism of two bisphenol A (BPA) alternatives binding to GPER.
Collapse
Affiliation(s)
- Tingji Yao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
| | - Jiaqi Luo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Xiaoxiao Han
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Hang Yi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Huazhou Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Wenxiao Pan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Qiao Xue
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Xian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Jianjie Fu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, P.R. China
| | - Aiqian Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P. R. China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310012, P. R. China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, P.R. China
| |
Collapse
|
7
|
DePasquale O, O'Brien C, Gordon B, Barker DJ. The Orphan Receptor GPR151: Discovery, Expression, and Emerging Biological Significance. ACS Chem Neurosci 2025. [PMID: 40295925 DOI: 10.1021/acschemneuro.4c00780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are among the most prominent druggable targets in the human genome, accounting for approximately 40% of marketed drugs. Despite this, current GPCR-targeted therapies address only about 10% of the GPCRs encoded in the genome. Expanding our knowledge of the remaining "orphan" GPCRs represents a critical frontier in drug discovery. GPR151 emerges as a compelling target due to its distinct expression in the habenula complex, spinal cord neurons, and dorsal root ganglia. This receptor is highly conserved across mammals and possesses orthologs in species such as zebrafish and chickens, underscoring its evolutionarily conserved role in fundamental mammalian processes. Although the precise function of GPR151 remains unknown, it has been strongly implicated in pain modulation and reward-seeking behavior. These attributes position GPR151 as a promising candidate for the development of targeted and specialized pharmacological therapies. This review summarizes the current literature on GPR151, including its discovery, structure, mechanisms, anatomical distribution, and functional roles, while also exploring potential directions for future research.
Collapse
Affiliation(s)
- Olivia DePasquale
- Department of Psychology, Rutgers, The State University of New Jersey, 152 Frelinghuysen Road, Piscataway, New Jersey 08854, United States
| | - Chris O'Brien
- Department of Psychology, Rutgers, The State University of New Jersey, 152 Frelinghuysen Road, Piscataway, New Jersey 08854, United States
| | - Baila Gordon
- Department of Psychology, Rutgers, The State University of New Jersey, 152 Frelinghuysen Road, Piscataway, New Jersey 08854, United States
| | - David J Barker
- Department of Psychology, Rutgers, The State University of New Jersey, 152 Frelinghuysen Road, Piscataway, New Jersey 08854, United States
- Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854, United States
- Rutgers Addiction Research Center, Piscataway, New Jersey 08854, United States
| |
Collapse
|
8
|
Abu-Helo A, Daubeuf F, Tranchant T, Lehalle C, Elhabazi K, Zeder-Lutz G, Kugler V, Lugnier C, Frossard N, Lecat S, Simonin F. GPRASP1 deletion in mice abrogates adverse side effects associated with chronic stimulation of Beta2-adrenoceptor. Biomed Pharmacother 2025; 187:118073. [PMID: 40286598 DOI: 10.1016/j.biopha.2025.118073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
GPCR associated sorting protein 1 (GPRASP1) interacts with numerous GPCRs including the Beta2-adrenoceptor (B2AR) and has been proposed to be involved in adaptations associated with chronic stimulation of those receptors. In clinic, long acting B2AR agonists (LABAs) such as formoterol are used in the treatment of asthma as potent bronchodilators but with adverse side effects including the development of tolerance and airway hyperresponsiveness upon chronic administration. Here, we investigated the role of GPRASP1 on B2AR activity and on B2AR agonists-associated side effects in vitro and in vivo. To this purpose, we set-up a model of chronic formoterol administration in mouse leading to B2AR down-regulation as well as to the development of airway hyperreactivity and bronchodilator tolerance and studied the phenotype of GPRASP1 knockout animals. We show in cells that GPRASP1 expression has no impact on agonist-induced B2AR down-regulation but strongly modulate B2AR-associated signalling. Moreover, wild-type mice chronically treated with formoterol developed airway hyperresponsiveness to methacholine and bronchodilator tolerance to formoterol that were absent in GPRASP1 KO mice while B2AR down-regulation was similar in both genotypes. These adverse side effects were correlated with an increase in the number of cells and in collagen levels in the lungs of wild-type but not of GPRASP1 KO mice. Collectively, our data show that GPRASP1 is critically involved in adaptations to chronic activation of B2AR that leads to lung tissue remodelling, development of bronchial hyperresponsiveness and bronchodilator tolerance to B2AR agonist formoterol and could therefore represent an interesting target to limit side effects associated with LABAs.
Collapse
Affiliation(s)
- Alaa Abu-Helo
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - François Daubeuf
- Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Thibaud Tranchant
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Christine Lehalle
- Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Khadija Elhabazi
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France
| | - Gabrielle Zeder-Lutz
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Valérie Kugler
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Claire Lugnier
- Section de Structures Biologiques, Pharmacologie et Enzymologie, CNRS/Unistra, CRBS, UR, Strasbourg 3072, France.
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Sandra Lecat
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
9
|
Fu R, Ling D, Zhang Q, Jiang A, Pang H. Harnessing Nur77's mitochondrial apoptotic pathway: A promising therapeutic strategy for targeted disease intervention. Biomed Pharmacother 2025; 187:118091. [PMID: 40286599 DOI: 10.1016/j.biopha.2025.118091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025] Open
Abstract
The role of mitochondria in disease development cannot be overlooked, and the targeting of mitochondria for the treatment of disease has emerged as a significant area of research in recent years. Mitochondria are the control center of the intrinsic apoptotic pathway, and their normal functions are finely regulated by a series of complex mechanisms. The nuclear receptor Nur77 is closely related to the functions of the mitochondria and is an active pro-apoptotic member of the nuclear receptor superfamily. The translocation of Nur77 to the mitochondria can promote the conversion of the anti-apoptotic protein Bcl-2 to a pro-apoptotic state, disrupt the balance between mitochondrial fission and fusion, and inhibit mitophagy. These effects lead to irreversible damage to mitochondria and apoptosis, ultimately accelerating the progression of the disease. Here, we review the mechanism and targeted drug development of the mitochondrial apoptosis pathway activated by Nur77 in human diseases, helping to understand the new advances in disease treatment.
Collapse
Affiliation(s)
- Ruihai Fu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, PR China
| | - Dandan Ling
- Clinical Research Center for Placental Medicine In Hunan Province, Changsha City, Hunan Province, PR China; Department of Obstetrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha City, Hunan Province, PR China
| | - Qiqi Zhang
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, PR China
| | - Aifang Jiang
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, PR China; School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, PR China
| | - Haiyan Pang
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, PR China; School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, PR China.
| |
Collapse
|
10
|
Leung K, Schaefer K, Lin Z, Yao Z, Wells JA. Engineered Proteins and Chemical Tools to Probe the Cell Surface Proteome. Chem Rev 2025; 125:4069-4110. [PMID: 40178992 PMCID: PMC12022999 DOI: 10.1021/acs.chemrev.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 02/05/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
The cell surface proteome, or surfaceome, is the hub for cells to interact and communicate with the outside world. Many disease-associated changes are hard-wired within the surfaceome, yet approved drugs target less than 50 cell surface proteins. In the past decade, the proteomics community has made significant strides in developing new technologies tailored for studying the surfaceome in all its complexity. In this review, we first dive into the unique characteristics and functions of the surfaceome, emphasizing the necessity for specialized labeling, enrichment, and proteomic approaches. An overview of surfaceomics methods is provided, detailing techniques to measure changes in protein expression and how this leads to novel target discovery. Next, we highlight advances in proximity labeling proteomics (PLP), showcasing how various enzymatic and photoaffinity proximity labeling techniques can map protein-protein interactions and membrane protein complexes on the cell surface. We then review the role of extracellular post-translational modifications, focusing on cell surface glycosylation, proteolytic remodeling, and the secretome. Finally, we discuss methods for identifying tumor-specific peptide MHC complexes and how they have shaped therapeutic development. This emerging field of neo-protein epitopes is constantly evolving, where targets are identified at the proteome level and encompass defined disease-associated PTMs, complexes, and dysregulated cellular and tissue locations. Given the functional importance of the surfaceome for biology and therapy, we view surfaceomics as a critical piece of this quest for neo-epitope target discovery.
Collapse
Affiliation(s)
- Kevin
K. Leung
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
| | - Kaitlin Schaefer
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
| | - Zhi Lin
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
| | - Zi Yao
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
| | - James A. Wells
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
- Department
of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
11
|
Catacuzzeno L, Michelucci A, Franciolini F. The crucial decade that ion channels were proven to exist : The vision of Bertil Hille and Clay Armstrong and how it came through. Pflugers Arch 2025:10.1007/s00424-025-03085-5. [PMID: 40261416 DOI: 10.1007/s00424-025-03085-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025]
Abstract
This retrospective begins with the first recording of the Na+ and K+ currents underlying the action potential in the giant squid axon reported by Hodgkin and Huxley in 1952, which made the question of where ions pass through the membrane more compelling. The notion of channels in the membrane had been around for quite some time but was so vague and contested that even the recording of Na+ and K+ currents through the membrane was not considered sufficient proof of their existence. In fact, Hodgkin and Huxley never referred to ion channels in their papers, only currents and conductances. The word "channel" remained somewhat left out from the scientific debate for almost another two decades, even though its idea was slowly making its way into the minds of discerning scientists. It is precisely this period that the present retrospective focuses on to understand the evolution of the ion channel concept from a speculative functional entity to a physical transmembrane object that serves the efficient and selective passage of ions. In this regard, the fundamental contribution of Bertil Hille and Clay Armstrong in promoting this idea, in the cold attitude, when not open aversion, of much of the scientific community, is fully acknowledged. Mention should also be made of Erwin Neher and Bert Sakmann's patch-clamp technique, which made it possible to directly measure ion currents through individual channels, thus conclusively demonstrating their presence in cell membranes. The retrospective goes on to briefly show how the cloning of ion channels in the 1980s and the first X-ray crystallographic structures at the turn of the century fully confirmed the initial suggestions, and closes by illustrating the relevance of ion channels in biology and medicine.
Collapse
Affiliation(s)
- Luigi Catacuzzeno
- Dipartimento di Chimica Biologia e Biotecnologie, Universita' di Perugia, Perugia, Italy.
| | - Antonio Michelucci
- Dipartimento di Chimica Biologia e Biotecnologie, Universita' di Perugia, Perugia, Italy
| | - Fabio Franciolini
- Dipartimento di Chimica Biologia e Biotecnologie, Universita' di Perugia, Perugia, Italy.
| |
Collapse
|
12
|
Namba S, Iwata M, Nureki SI, Yuyama Otani N, Yamanishi Y. Therapeutic target prediction for orphan diseases integrating genome-wide and transcriptome-wide association studies. Nat Commun 2025; 16:3355. [PMID: 40251160 PMCID: PMC12008218 DOI: 10.1038/s41467-025-58464-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/19/2025] [Indexed: 04/20/2025] Open
Abstract
Therapeutic target identification is challenging in drug discovery, particularly for rare and orphan diseases. Here, we propose a disease signature, TRESOR, which characterizes the functional mechanisms of each disease through genome-wide association study (GWAS) and transcriptome-wide association study (TWAS) data, and develop machine learning methods for predicting inhibitory and activatory therapeutic targets for various diseases from target perturbation signatures (i.e., gene knockdown and overexpression). TRESOR enables highly accurate identification of target candidate proteins that counteract disease-specific transcriptome patterns, and the Bayesian optimization with omics-based disease similarities achieves the performance enhancement for diseases with few or no known targets. We make comprehensive predictions for 284 diseases with 4345 inhibitory target candidates and 151 diseases with 4040 activatory target candidates, and elaborate the promising targets using several independent cohorts. The methods are expected to be useful for understanding disease-disease relationships and identifying therapeutic targets for rare and orphan diseases.
Collapse
Affiliation(s)
- Satoko Namba
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, 820-8502, Japan
- Department of Complex Systems Science, Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Aichi, 464-8601, Japan
| | - Michio Iwata
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, 820-8502, Japan
| | - Shin-Ichi Nureki
- Department of Respiratory Medicine and Infectious Diseases, Oita University Faculty of Medicine, Yufu, Oita, 879-5593, Japan
| | - Noriko Yuyama Otani
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, 820-8502, Japan
- Department of Complex Systems Science, Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Aichi, 464-8601, Japan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, 820-8502, Japan.
- Department of Complex Systems Science, Graduate School of Informatics, Nagoya University, Chikusa, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
13
|
Beaudoin CA, Norget S, Omran Z, Hala S, Daqeeq AH, Burnet PWJ, Blundell TL, van Tonder AJ. Similarity of drug targets to human microbiome metaproteome promotes pharmacological promiscuity. THE PHARMACOGENOMICS JOURNAL 2025; 25:9. [PMID: 40246834 PMCID: PMC12006021 DOI: 10.1038/s41397-025-00367-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025]
Abstract
Similarity between candidate drug targets and human proteins is commonly assessed to minimize the occurrence of side effects. Although numerous drugs have been found to disrupt the health of the human microbiome, no comprehensive comparison between established drug targets and the human microbiome metaproteome has yet been conducted. Therefore, herein, sequence and structure alignments between human and pathogen drug targets and representative human gut, oral, and vaginal microbiome metaproteomes were performed. Both human and pathogen drug targets were found to be similar in sequence, function, structure, and drug binding capacity to proteins in diverse pathogenic and non-pathogenic bacteria from all three microbiomes. The gut metaproteome was identified as particularly susceptible overall to off-target effects. Certain symptoms, such as infections and immune disorders, may be more common among drugs that non-selectively target host microbiota. These findings suggest that similarities between human microbiome metaproteomes and drug target candidates should be routinely checked.
Collapse
Affiliation(s)
| | - Shannon Norget
- Department of Psychology, Health & Technology, University of Twente, Enschede, the Netherlands
| | - Ziad Omran
- King Abdullah International Medical Research Center, King Saud Bin Abdelaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Sharif Hala
- Biothreat Department, Public Health Laboratory, Public Health Authority, Riyadh, Saudi Arabia
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Abdullah H Daqeeq
- Department of Anesthesia, International Medical Center, Jeddah, Kingdom of Saudi Arabia
| | | | - Tom L Blundell
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Biomedical Campus, Trumpington, Cambridge, UK
| | | |
Collapse
|
14
|
Poudel P, Miteva MA, Alexov E. Strategies for in Silico Drug Discovery to Modulate Macromolecular Interactions Altered by Mutations. FRONT BIOSCI-LANDMRK 2025; 30:26339. [PMID: 40302318 DOI: 10.31083/fbl26339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/22/2024] [Accepted: 10/09/2024] [Indexed: 05/02/2025]
Abstract
Most human diseases have genetic components, frequently single nucleotide variants (SNVs), which alter the wild type characteristics of macromolecules and their interactions. A straightforward approach for correcting such SNVs-related alterations is to seek small molecules, potential drugs, that can eliminate disease-causing effects. Certain disorders are caused by altered protein-protein interactions, for example, Snyder-Robinson syndrome, the therapy for which focuses on the development of small molecules that restore the wild type homodimerization of spermine synthase. Other disorders originate from altered protein-nucleic acid interactions, as in the case of cancer; in these cases, the elimination of disease-causing effects requires small molecules that eliminate the effect of mutation and restore wild type p53-DNA affinity. Overall, especially for complex diseases, pathogenic mutations frequently alter macromolecular interactions. This effect can be direct, i.e., the alteration of wild type affinity and specificity, or indirect via alterations in the concentration of the binding partners. Here, we outline progress made in methods and strategies to computationally identify small molecules capable of altering macromolecular interactions in a desired manner, reducing or increasing the binding affinity, and eliminating the disease-causing effect. When applicable, we provide examples of the outlined general strategy. Successful cases are presented at the end of the work.
Collapse
Affiliation(s)
- Pitambar Poudel
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Maria A Miteva
- Université Paris Cité, CNRS UMR 8038 CiTCoM, Inserm, U1268 MCTR Paris, France
| | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
15
|
Raavi, Koehler AN, Vegas AJ. At The Interface: Small-Molecule Inhibitors of Soluble Cytokines. Chem Rev 2025. [PMID: 40233276 DOI: 10.1021/acs.chemrev.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytokines are crucial regulators of the immune system that orchestrate interactions between cells and, when dysregulated, contribute to the progression of chronic inflammation, cancer, and autoimmunity. Numerous biologic-based clinical agents, mostly monoclonal antibodies, have validated cytokines as important clinical targets and are now part of the standard of care for a number of diseases. These agents, while impactful, still suffer from limitations including a lack of oral bioavailability, high cost of production, and immunogenicity. Small-molecule cytokine inhibitors are attractive alternatives that can address these limitations. Although targeting cytokine-cytokine receptor complexes with small molecules has been a challenging research endeavor, multiple small-molecule inhibitors have now been identified, with a number of them undergoing clinical evaluation. In this review, we highlight the recent advancements in the discovery and development of small-molecule inhibitors targeting soluble cytokines. The strategies for identifying these novel ligands as well as the structural and mechanistic insights into their activity represent important milestones in tackling these challenging and clinically important protein-protein interactions.
Collapse
Affiliation(s)
- Raavi
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Arturo J Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02115, United States
| |
Collapse
|
16
|
Deng C, Xie C, Li Z, Mei J, Wang K. Multi-omics analysis identifies diagnostic circulating biomarkers and potential therapeutic targets, revealing IQGAP1 as an oncogene in gastric cancer. NPJ Precis Oncol 2025; 9:105. [PMID: 40229327 PMCID: PMC11997149 DOI: 10.1038/s41698-025-00895-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
This study employed a multi-omics integration approach to identify circulating biomarkers for gastric cancer (GC). We analyzed plasma and tumor tissue single-cell RNA sequencing data, along with gene and protein quantitative trait loci analyses. Leveraging data from UK Biobank and FinnGen, we investigated genetic associations with GC. Through colocalization, Mendelian Randomization, and various filtering analyses, we identified four genes (IQGAP1, KRTCAP2, PARP1, MLF2) and four proteins (EGFL9 [DLK2], ECM1, PDIA5, TIMP4) as potential GC biomarkers. These were selected based on significant genetic colocation probabilities and significant associations with GC. Seven of these biomarkers demonstrated predictive capability for GC occurrence, with AUC ranging from 0.61 to 0.99. Drug prediction analysis identified seven protein biomarkers as potential targets for immunotherapy, targeted therapies, and tumor chemotherapy. Further scRNA-seq analysis revealed significant expression differences between gastric tumor and normal tissues, particularly the upregulation of IQGAP1, which highlights its role in tumor growth.
Collapse
Affiliation(s)
- Chao Deng
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chenjun Xie
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China
| | - Zixi Li
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China
| | - Jie Mei
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China.
| | - Kewei Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
17
|
Liu W, Li G, Shi J, Gao Y, Fang P, Zhao Y, Zhong F, Guo X, Lyu Y, Da X, Li Z, Fa J, Hu L, Yuan A, Chen L, Liu J, Chen AF, Sheng B, Ji Y, Lu X, Pu J. NR4A1 Acts as a Novel Regulator of Platelet Activation and Thrombus Formation. Circ Res 2025; 136:809-826. [PMID: 40035146 PMCID: PMC11984555 DOI: 10.1161/circresaha.124.325645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Mounting evidence indicates that nuclear receptors play a critical regulatory role in platelet pathophysiology and thrombotic disorders. Although NR4A (the nuclear receptor subfamily 4 group A) plays an important role in cardiovascular pathophysiology, the expression profile and biological function of NR4A member 1 (NR4A1) in platelets have never been reported. METHODS We evaluated the functions and the underlying mechanisms of NR4A1 in platelet activation and thrombus formation using platelet-specific NR4A1-deficient mice and NR4A1-specific agonists. Using a hyperlipidemic mouse model and platelets from patients with hypercholesterolemia, we explored the influence of hypercholesterolemia on NR4A1 expression and the effects of NR4A1-specific agonists on platelet hyperreactivity induced by hypercholesterolemia. RESULTS NR4A1 was expressed in both human and mouse platelets. Platelet-specific NR4A1 deletion accelerated FeCl3-induced carotid arterial occlusive thrombus formation, enhanced collagen/epinephrine-induced pulmonary thromboembolism, and exacerbated microvascular microthrombi obstruction and infarct expansion in an acute myocardial infarction model. NR4A1-deficient platelets exhibited enhanced agonist-induced aggregation responses, integrin αIIbβ3 activation, dense granule release, α-granule release, platelet spreading, and clot retraction. Consistently, pharmacological activation of NR4A1 by specific agonists decreased platelet activation in both mouse and human platelets. Mechanistically, CAP1 (adenylyl cyclase-associated protein 1) was identified as the direct downstream interacting protein of NR4A1. NR4A1 deletion decreased cAMP levels and phosphorylation of VASP (vasodilator-stimulated phosphoprotein), while NR4A1-specific agonists increased cAMP levels and phosphorylation of VASP in platelets. Importantly, NR4A1 expression in platelets was upregulated in the setting of hypercholesterolemia, which was derived from its upregulation in megakaryocytes in a reactive oxygen species-dependent manner. Platelets from hypercholesterolemic patients and mice exhibited hyperreactivity. However, NR4A1-specific agonists significantly inhibited the activation of hypercholesterolemic platelets to the levels of healthy control platelets. CONCLUSIONS We provide the first evidence that nuclear receptor NR4A1 negatively regulates platelet activation and thrombus formation. NR4A1 may serve as a novel therapeutic target for managing thrombosis-based cardiovascular diseases, especially with hypercholesterolemia.
Collapse
MESH Headings
- Animals
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/blood
- Platelet Activation/physiology
- Humans
- Thrombosis/metabolism
- Thrombosis/blood
- Thrombosis/genetics
- Blood Platelets/metabolism
- Mice
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Hypercholesterolemia/blood
- Hypercholesterolemia/genetics
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
- Wenhua Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Gaoxiang Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jianfeng Shi
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yu Gao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Peiliang Fang
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yichao Zhao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Fangyuan Zhong
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xiao Guo
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yuyan Lyu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xingwen Da
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Zhaoyan Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jingjing Fa
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Baoshan Branch (J.F.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Liuhua Hu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Ancai Yuan
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Lei Chen
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Junling Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education (J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Alex F. Chen
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, China (B.S.)
| | - Yong Ji
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China(Y.J.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Heilongjiang, China (Y.J.)
| | - Xiyuan Lu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jun Pu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|
18
|
Zhou Y, Ding Y, Xu B, Fei H, Wang Z. Genetically druggable targets for MAPK-activated colorectal cancer by a two-sample mendelian randomization analysis. Sci Rep 2025; 15:12239. [PMID: 40210889 PMCID: PMC11986099 DOI: 10.1038/s41598-024-82567-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 12/06/2024] [Indexed: 04/12/2025] Open
Abstract
Colorectal cancer (CRC) is a significant worldwide health issue, ranking second in women and third in men. Predictions estimate a rise to 2.5 million cases by 2035, with CRC being the fourth deadliest cancer due to delayed diagnosis and the scarcity of effective treatment options. Over 60% of CRC cases involve MAPK-activated signal pathways, particularly driven by RAS oncogene mutations, which hinder treatment responses, making them 'undruggable.' This study conducts a two-sample Mendelian randomization protein quantitative trait loci (pQTL) analysis to investigate the causal association between plasma proteins and MAPK-activated CRCs. The study indicates that four plasma proteins-MHC class I polypeptide-related sequence B (MICB), complement C4A, C4B, and interleukin-21 (IL-21) are associated with an increased risk of MAPK-activated CRCs. These findings highlight the possibility of utilizing plasma proteins as therapeutic targets and diagnostic markers to advance the fight against CRCs, indicating promising results for more effective interventions. To ascertain and expand upon these discoveries, further research is imperative to fully harness the potential of these discoveries.
Collapse
Affiliation(s)
- Yuxuan Zhou
- Department of Gastrointestinal Surgery/Hernia Surgery, Jilin Province People's Hospital, No. 1183 Gongnong Road, Changchun, Jilin, China
| | - Yunlong Ding
- Department of Emergency General Surgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Bangyue Xu
- Jilin Central General Hospital, Changchun, Jilin, China
| | - Hongyang Fei
- Department of Hepatobiliary and Pancreatic Surgery, Jilin Province People's Hospital, No. 1183 Gongnong Road, Changchun, Jilin, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery/Hernia Surgery, Jilin Province People's Hospital, No. 1183 Gongnong Road, Changchun, Jilin, China.
| |
Collapse
|
19
|
Bucknam A, Nicholson JM, Radomska HS, Young GM, Coss CC, Micalizio GC. An Estrogen Receptor β Agonist with AR Antagonist Activity from a Modern Asymmetric De Novo Steroid Synthesis. ACS Med Chem Lett 2025; 16:631-637. [PMID: 40236556 PMCID: PMC11995240 DOI: 10.1021/acsmedchemlett.5c00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025] Open
Abstract
While the androgen receptor (AR) has long been a molecular target of prostate cancer therapeutics, the estrogen receptor β (ERβ) has more recently become of interest in this area of chemotherapeutic space due to its demonstrated role as a tumor suppressor. Here, we report studies that build on our earlier discovery of an exquisitely potent and selective steroidal agonist of ERβ that have resulted in the identification of a synthetic substituted estrane that, in reporter assays, is a potent and selective agonist of ERβ over ERα while also possessing activity as an antagonist of the AR that has potency within 7-fold of enzalutamide. Notably, this discovery was made possible by leveraging the power of our modern asymmetric de novo chemical synthesis of estranes that obviates the typical dependence on semisynthesis (natural product derivatization) for steroid-based medicinal chemistry.
Collapse
Affiliation(s)
- Andrea
R. Bucknam
- Department
of Chemistry, Dartmouth College, Burke Laboratory, Hanover, New Hampshire 03755, United States
| | - Joshua M. Nicholson
- Department
of Chemistry, Dartmouth College, Burke Laboratory, Hanover, New Hampshire 03755, United States
| | - Hanna S. Radomska
- Division
of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Greg M. Young
- Division
of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Christopher C. Coss
- Division
of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Glenn C. Micalizio
- Department
of Chemistry, Dartmouth College, Burke Laboratory, Hanover, New Hampshire 03755, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, California 92697, United States
| |
Collapse
|
20
|
Syahputra EW, Lee H, Cho H, Park HJ, Park KS, Hwang D. PROTAC Delivery Strategies for Overcoming Physicochemical Properties and Physiological Barriers in Targeted Protein Degradation. Pharmaceutics 2025; 17:501. [PMID: 40284496 PMCID: PMC12030311 DOI: 10.3390/pharmaceutics17040501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Proteolysis targeting chimeras (PROTACs), heterobifunctional molecules that hijack the ubiquitin-proteasome system (UPS) to degrade specific proteins, hold great promise in treating diseases driven by traditionally "undruggable" targets. However, their large molecular weight, high hydrophobicity, and other physicochemical hurdles contribute to their limited bioavailability, suboptimal pharmacokinetics, and attenuated therapeutic efficacy. Consequently, diverse formulation innovations have been investigated to optimize PROTAC delivery. This review examines current challenges and advances in specialized drug delivery approaches designed to bolster PROTAC pharmacological performance. We first outline the fundamental limitations of PROTACs-their low aqueous solubility, poor cell permeability, rapid clearance, and concentration-dependent "hook effect". We then discuss how various enabling formulations address these issues, including polymeric micelles, emulsions, amorphous solid dispersions, lipid-based nanoparticles, liposomes, and exosomes. Collectively, these delivery technologies substantially improve the therapeutic outcomes of PROTACs in preclinical cancer models. Future applications may extend beyond oncology to address other complex diseases using newly emerging heterobifunctional molecules. By integrating advanced formulation science with innovative degrader design, the field stands poised to unlock the clinical potential of PROTACs for protein degradation therapies.
Collapse
Affiliation(s)
- Endry Wahyu Syahputra
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; (E.W.S.); (H.C.); (H.J.P.)
| | - Hyunji Lee
- College of Pharmacy, Kyungsung University, Busan 48434, Republic of Korea;
| | - Hyukjun Cho
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; (E.W.S.); (H.C.); (H.J.P.)
| | - Hyun Jin Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; (E.W.S.); (H.C.); (H.J.P.)
| | - Kwang-Su Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; (E.W.S.); (H.C.); (H.J.P.)
| | - Duhyeong Hwang
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; (E.W.S.); (H.C.); (H.J.P.)
| |
Collapse
|
21
|
Dai Y, Yang M, Luo W, Qiu Y, Zhou F, Zheng X, Zhao F, Yao X, Zhao S, Tao H. "Living Detergents": an in Situ Detergent Tailoring Strategy for Efficient Membrane Protein Stabilization and Analysis. Chemistry 2025:e202501128. [PMID: 40192258 DOI: 10.1002/chem.202501128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
Detergents are essential molecular tools for membrane protein (MP) research, yet traditional detergents with static properties often fail to address the diverse and evolving needs of MP studies. To this end, this study introduces "living detergents", an innovative class of detergents equipped with functional tags that enable bioorthogonal modifications with externally introduced structural elements. This approach allows for not only the parallel generation of new detergents, but also in situ tuning of MP samples within freshly formed detergents. The efficacy of this strategy was demonstrated through the rapid identification of optimal detergents for high-quality electron microscopy studies of A2AAR. Overall, this flexible and robust platform enables efficient tailoring of detergents, advancing the exploration of detergent structure-function relationships in MP research and opening pathways for more specialized solutions for diverse experimental demands.
Collapse
Affiliation(s)
- Yili Dai
- Institute of Pharmacy and Pharmacology, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Meifang Yang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weiling Luo
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Yanli Qiu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Fang Zhou
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Xing Zheng
- Institute of Pharmacy and Pharmacology, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
- Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha, Hunan, 410004, China
| | - Fei Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Xu Yao
- Institute of Pharmacy and Pharmacology, Hengyang Medicinal School, University of South China, Hengyang, Hunan, 421001, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, P. R. China
| | - Houchao Tao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
22
|
Zhou Z, Chen Z, Li Y, Mao X, Chen J, Zhou X, Zhang B. Advances in solubilization and stabilization techniques for structural and functional studies of membrane proteins. PeerJ 2025; 13:e19211. [PMID: 40196297 PMCID: PMC11974516 DOI: 10.7717/peerj.19211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Membrane proteins (MPs) are indispensable in various biological processes, including material transport, signal transduction, immune response, and cell recognition. Unraveling the intricate interplay between MP structure and function is pivotal for advancing fundamental biology and pharmaceutical research. However, the inherent hydrophobicity and complex lipid interactions of MPs pose significant challenges in determining their three-dimensional configurations. In recent years, cryo-electron microscopy (cryo-EM) has emerged as a powerful alternative for structural elucidation, overcoming the challenges faced by traditional techniques such as X-ray crystallography and nuclear magnetic resonance (NMR). This review centers on advanced solubilization and stabilization techniques for MPs, as well as MP functions and expression systems, highlighting the strengths and limitations of conventional detergents, liposomes, bicelles, and nanodiscs, alongside emerging alternatives like styrene-maleic acid (SMA) and diisobutylene-maleic acid (DIBMA). Notably, SMA and its derivatives provide promising detergent-free alternatives that preserve protein stability and native conformation, which is particularly valuable for accurate cryo-EM characterization of complex MPs. This work is designed to serve as both an updated resource for researchers already immersed in the field and an accessible entry point for those new to MP research. By consolidating recent advancements and highlighting critical gaps, this review aims to inspire future investigations that push the boundaries of MP structural and functional studies, ultimately driving innovations in drug discovery and therapeutic development.
Collapse
Affiliation(s)
- Zhuanghan Zhou
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Zheng Chen
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Yiran Li
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Xingyue Mao
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Junjie Chen
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Xuan Zhou
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Bo Zhang
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
- Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University of New Jersey, Union, NJ, United States of America
| |
Collapse
|
23
|
Yang Z, Zhou SH, Zhang QY, Song ZC, Liu WW, Sun Y, Wang MW, Fu XL, Zhu KK, Guan Y, Qi JY, Wang XH, Sun YN, Lu Y, Ping YQ, Xi YT, Teng ZX, Xu L, Xiao P, Xu ZG, Xiong W, Qin W, Yang W, Yi F, Chai RJ, Yu X, Sun JP. A force-sensitive adhesion GPCR is required for equilibrioception. Cell Res 2025; 35:243-264. [PMID: 39966628 PMCID: PMC11958651 DOI: 10.1038/s41422-025-01075-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Equilibrioception (sensing of balance) is essential for mammals to perceive and navigate the three-dimensional world. A rapid mechanoelectrical transduction (MET) response in vestibular hair cells is crucial for detecting position and motion. Here, we identify the G protein-coupled receptor (GPCR) LPHN2/ADGRL2, expressed on the apical membrane of utricular hair cells, as essential for maintaining normal balance. Loss of LPHN2 specifically in hair cells impaired both balance behavior and the MET response in mice. Functional analyses using hair-cell-specific Lphn2-knockout mice and an LPHN2-specific inhibitor suggest that LPHN2 regulates tip-link-independent MET currents at the apical surface of utricular hair cells. Mechanistic studies in a heterologous system show that LPHN2 converts force stimuli into increased open probability of transmembrane channel-like protein 1 (TMC1). LPHN2-mediated force sensation triggers glutamate release and calcium signaling in utricular hair cells. Importantly, reintroducing LPHN2 into the hair cells of Lphn2-deficient mice restores vestibular function and MET response. Our data reveal that a mechanosensitive GPCR is required for equilibrioception.
Collapse
Affiliation(s)
- Zhao Yang
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shu-Hua Zhou
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu, China
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qi-Yue Zhang
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhi-Chen Song
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wen-Wen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ming-Wei Wang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiao-Long Fu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kong-Kai Zhu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ying Guan
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jie-Yu Qi
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu, China
| | - Xiao-Hui Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu-Nan Sun
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yan Lu
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yu-Qi Ping
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yue-Tong Xi
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhen-Xiao Teng
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Peng Xiao
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhi-Gang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Shandong University School of Life Sciences, Qingdao, Shandong, China
| | - Wei Xiong
- Chinese Institute for Brain Research, Beijing, China
| | - Wei Qin
- School of Physics, State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, China
| | - Wei Yang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China.
| | - Ren-Jie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu, China.
| | - Xiao Yu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Jin-Peng Sun
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, and New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
24
|
Fournier L, Guarnera E, Kolmar H, Becker S. Allosteric antibodies: a novel paradigm in drug discovery. Trends Pharmacol Sci 2025; 46:311-323. [PMID: 39562213 DOI: 10.1016/j.tips.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/30/2024] [Accepted: 10/18/2024] [Indexed: 11/21/2024]
Abstract
Allostery represents a fundamental mechanism in protein regulation, enabling modulation of protein function from sites distal to the active site. While traditionally explored in the context of small molecules, allosteric modulation is gaining traction as a main mode of action in the realm of antibodies, which offer enhanced specificity and reduced toxicity. This review delves into the rapidly growing field of allosteric antibodies, highlighting recent therapeutic advancements and novel druggability avenues. We also explore the potential of these antibodies as innovative tools in drug discovery and discuss contemporary strategies for designing novel allosteric antibodies, leveraging state-of-the-art computational approaches.
Collapse
Affiliation(s)
- Léxane Fournier
- Early Protein Supply and Characterization, Merck Healthcare KGaA, Darmstadt, Germany; Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Enrico Guarnera
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany.
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Stefan Becker
- Early Protein Supply and Characterization, Merck Healthcare KGaA, Darmstadt, Germany.
| |
Collapse
|
25
|
Conflitti P, Lyman E, Sansom MSP, Hildebrand PW, Gutiérrez-de-Terán H, Carloni P, Ansell TB, Yuan S, Barth P, Robinson AS, Tate CG, Gloriam D, Grzesiek S, Eddy MT, Prosser S, Limongelli V. Functional dynamics of G protein-coupled receptors reveal new routes for drug discovery. Nat Rev Drug Discov 2025; 24:251-275. [PMID: 39747671 PMCID: PMC11968245 DOI: 10.1038/s41573-024-01083-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 01/04/2025]
Abstract
G protein-coupled receptors (GPCRs) are the largest human membrane protein family that transduce extracellular signals into cellular responses. They are major pharmacological targets, with approximately 26% of marketed drugs targeting GPCRs, primarily at their orthosteric binding site. Despite their prominence, predicting the pharmacological effects of novel GPCR-targeting drugs remains challenging due to the complex functional dynamics of these receptors. Recent advances in X-ray crystallography, cryo-electron microscopy, spectroscopic techniques and molecular simulations have enhanced our understanding of receptor conformational dynamics and ligand interactions with GPCRs. These developments have revealed novel ligand-binding modes, mechanisms of action and druggable pockets. In this Review, we highlight such aspects for recently discovered small-molecule drugs and drug candidates targeting GPCRs, focusing on three categories: allosteric modulators, biased ligands, and bivalent and bitopic compounds. Although studies so far have largely been retrospective, integrating structural data on ligand-induced receptor functional dynamics into the drug discovery pipeline has the potential to guide the identification of drug candidates with specific abilities to modulate GPCR interactions with intracellular effector proteins such as G proteins and β-arrestins, enabling more tailored selectivity and efficacy profiles.
Collapse
Affiliation(s)
- Paolo Conflitti
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Edward Lyman
- Department of Physics and Astronomy, University of Delaware, Newark, DE, USA
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Peter W Hildebrand
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Hugo Gutiérrez-de-Terán
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Paolo Carloni
- INM-9/IAS-5 Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - T Bertie Ansell
- Department of Biochemistry, University of Oxford, Oxford, UK
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Shuguang Yuan
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - David Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark
| | - Stephan Grzesiek
- Focal Area Structural Biology and Biophysics, Biozentrum, University of Basel, Basel, Switzerland
| | - Matthew T Eddy
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Scott Prosser
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - Vittorio Limongelli
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland.
| |
Collapse
|
26
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
27
|
Orzeł U, Barreto CAV, Filipek S, Moreira IS. GPCR oligomerization across classes: A2AR-mediated regulation of mGlu5R activation. Int J Biol Macromol 2025; 299:139880. [PMID: 39842585 DOI: 10.1016/j.ijbiomac.2025.139880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
The adenosine A2A receptor (A2AR), a class A GPCR, is a known player in neurological diseases, including Parkinson's disease and Alzheimer's disease, and is also implicated in SARS-CoV-2 infection. Recent studies have revealed its oligomerization with metabotropic glutamate receptor type 5 (mGlu5R), a class C G protein coupled receptor (GPCR) that exists in the homodimeric form. Simultaneous activation of both receptors synergistically enhances mGlu5R-mediated effects in the hippocampus. Despite their importance, the molecular mechanisms governing these interactions remain unclear. In this study, we used molecular modelling techniques, including molecular docking, extensive molecular dynamics (MD) simulations, and detailed analysis, to elucidate the interactions between mGlu5R and A2AR in the inactive and active states. Our findings provide molecular-level insights into the permissive role of A2AR in mGlu5R activation, demonstrating that the inactive A2AR interface within the oligomer blocks the mGlu5R transmembrane helix 6 (TM6), which is crucial for activation. Upon A2AR activation, the oligomer interface undergoes conformational rearrangement, exposing mGlu5R-TM6 and allowing for mGlu5R activation. Furthermore, we identified a pivotal role of the mGlu5R-TM4:A2AR-TM4 interface in facilitating mGlu5R activation. These results highlight the intricate architecture of the mGlu5R:A2AR oligomer, advancing our understanding of GPCR oligomerization and its regulatory mechanisms on receptor activity.
Collapse
Affiliation(s)
- Urszula Orzeł
- PhD Programme in Biosciences, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland; Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | - Carlos A V Barreto
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Sławomir Filipek
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland; Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | - Irina S Moreira
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| |
Collapse
|
28
|
Yan Y, Zhang Y, Liu J, Chen B, Wang Y. Emerging magic bullet: subcellular organelle-targeted cancer therapy. MEDICAL REVIEW (2021) 2025; 5:117-138. [PMID: 40224364 PMCID: PMC11987508 DOI: 10.1515/mr-2024-0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/27/2024] [Indexed: 04/15/2025]
Abstract
The therapeutic efficacy of anticancer drugs heavily relies on their concentration and retention at the corresponding target site. Hence, merely increasing the cellular concentration of drugs is insufficient to achieve satisfactory therapeutic outcomes, especially for the drugs that target specific intracellular sites. This necessitates the implementation of more precise targeting strategies to overcome the limitations posed by diffusion distribution and nonspecific interactions within cells. Consequently, subcellular organelle-targeted cancer therapy, characterized by its exceptional precision, have emerged as a promising approach to eradicate cancer cells through the specific disruption of subcellular organelles. Owing to several advantages including minimized dosage and side effect, optimized efficacy, and reversal of multidrug resistance, subcellular organelle-targeted therapies have garnered significant research interest in recent years. In this review, we comprehensively summarize the distribution of drug targets, targeted delivery strategies at various levels, and sophisticated strategies for targeting specific subcellular organelles. Additionally, we highlight the significance of subcellular targeting in cancer therapy and present essential considerations for its clinical translation.
Collapse
Affiliation(s)
- Yue Yan
- Department of Central Laboratory, Peking University First Hospital, Beijing, China
| | - Yimeng Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianxiong Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Department of Central Laboratory, Peking University First Hospital, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Chemical Biology Center, Peking University, Beijing, China
| |
Collapse
|
29
|
Caña-Bozada VH, Dawoud AAZ, Ramos-de la Cruz I, Flores-Méndez LC, Barrera-Redondo J, Briones-Mendoza J, Yañez-Guerra LA. Global analysis of ligand-gated ion channel conservation across Platyhelminthes. Gen Comp Endocrinol 2025; 366:114718. [PMID: 40157577 DOI: 10.1016/j.ygcen.2025.114718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Ligand-gated ion channels (LGICs) are critical for neurotransmission, mediating responses to neurotransmitters and hormones, and influencing diverse physiological processes. This study identifies and classifies LGICs across Platyhelminthes, with a particular focus on parasitic neodermatans, which impact human and animal health. Using bioinformatics tools, we analyzed LGICs from 41 neodermatan species and expanded our investigation to encompass vertebrates, other invertebrates, and non-bilaterians to trace LGIC evolutionary pathways across Metazoa. We identified 2,269 putative LGICs within neodermatan species, which we classified into the cys-loop, ASIC/Deg/ENaC, iGluR, and P2X families. Our phylogenetic and clustering analyses reveal lineage-specific patterns with distinct evolutionary trajectories for each LGIC family in neodermatans compared to free-living platyhelminths and other taxa. Notably, the ASIC/Deg/ENaC family displayed the greatest degree of neodermatan-specific divergence, while cys-loop and P2X families were more conserved across taxa. To provide insight into their potential physiological roles, we analyzed LGIC expression patterns in Schistosoma mansoni, revealing widespread expression across neuronal and muscle cell types. The distribution of acid-sensing ion channels (ASICs) in both neurons and muscles suggests a role in neuromuscular signalling, while the P2X receptor (Smp_333600) exhibited sex-specific expression, potentially indicating distinct functional roles in males and females. Additionally, several cys-loop acetylcholine and GABA receptors showed differential neuronal and muscle expression, highlighting their likely contributions to cholinergic and inhibitory neurotransmission. These findings underscore the relevance of LGICs in parasite physiology, particularly in neuromuscular and sensory processes, and suggest potential targets for antiparasitic interventions.
Collapse
Affiliation(s)
- Víctor Hugo Caña-Bozada
- Centro de Investigación en Alimentación y Desarrollo, A.C. Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán 82112 Sinaloa, Mexico; Centro de Investigación para la Salud en América Latina (CISeAL), Pontificia Universidad Católica del Ecuador (PUCE), Quito, Ecuador.
| | - Ahmed A Z Dawoud
- School of biology. University of Southampton, University Road, SO17 1BJ Southampton, UK
| | - Ivana Ramos-de la Cruz
- Centro de Investigación en Alimentación y Desarrollo, A.C. Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán 82112 Sinaloa, Mexico
| | - Lizeth C Flores-Méndez
- Universidad Autónoma de Occidente, Unidad Regional Mazatlán. Av. del Mar, Tellería, Mazatlán 82100 Sinaloa, Mexico
| | - Josué Barrera-Redondo
- Department of Algal Development and Evolution, Max Planck Institute for Biology Tübingen 72076 Tübingen, Germany
| | - Jesús Briones-Mendoza
- Carrera de Biología, Facultad de Ciencias de la Vida y Tecnologías, Universidad Laica "Eloy Alfaro" de Manabí, Ciudadela Universitaria vía San Mateo, Manta, Ecuador
| | - Luis A Yañez-Guerra
- School of biology. University of Southampton, University Road, SO17 1BJ Southampton, UK; Institute for Life Sciences. University of Southampton, University Road SO17 1BJ Southampton, UK.
| |
Collapse
|
30
|
Qin ZX, Zuo L, Zeng Z, Ma R, Xie W, Zhu X, Zhou X. GalNac-siRNA conjugate delivery technology promotes the treatment of typical chronic liver diseases. Expert Opin Drug Deliv 2025; 22:455-469. [PMID: 39939158 DOI: 10.1080/17425247.2025.2466767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
INTRODUCTION Nucleic acid-based therapeutics have become a key pillar of the 'third wave' of modern medicine, following the eras of small molecule inhibitors and antibody drugs. Their rapid progress is heavily dependent on delivery technologies, with the development of N-acetylgalactosamine (GalNAc) conjugates marking a breakthrough in targeting liver diseases. This technology has gained significant attention for its role in addressing chronic conditions like chronic hepatitis B (CHB) and nonalcoholic steatohepatitis (NASH), which are challenging to treat with conventional methods. AREAS COVERED This review explores the origins, mechanisms, and advantages of GalNAc-siRNA delivery systems, highlighting their ability to target hepatocytes via the asialoglycoprotein receptor (ASGPR). The literature reviewed covers preclinical and clinical advancements, particularly in CHB and NASH. Key developments in stabilization chemistry and conjugation technologies are examined, emphasizing their impact on enhancing therapeutic efficacy and patient compliance. EXPERT OPINION GalNAc-siRNA technology represents a transformative advancement in RNA interference (RNAi) therapies, addressing unmet needs in liver-targeted diseases. While significant progress has been made, challenges remain, including restricted targeting scope and scalability concerns. Continued innovation is expected to expand applications, improve delivery efficiency, and overcome limitations, establishing GalNAc-siRNA as a cornerstone for future nucleic acid-based treatments.
Collapse
Affiliation(s)
- Zhen-Xin Qin
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Ling Zuo
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Ziran Zeng
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Rongguan Ma
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Wenyan Xie
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
31
|
Sun D, Li X, Yuan Q, Wang Y, Shi P, Zhang H, Wang T, Sun W, Ling S, Liu Y, Lai J, Xie W, Yin W, Liu L, Xu HE, Tian C. Molecular mechanism of the arrestin-biased agonism of neurotensin receptor 1 by an intracellular allosteric modulator. Cell Res 2025; 35:284-295. [PMID: 40118988 PMCID: PMC11958688 DOI: 10.1038/s41422-025-01095-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/02/2025] [Indexed: 03/24/2025] Open
Abstract
Biased allosteric modulators (BAMs) of G protein-coupled receptors (GPCRs) have been at the forefront of drug discovery owing to their potential to selectively stimulate therapeutically relevant signaling and avoid on-target side effects. Although structures of GPCRs in complex with G protein or GRK in a BAM-bound state have recently been resolved, revealing that BAM can induce biased signaling by directly modulating interactions between GPCRs and these two transducers, no BAM-bound GPCR-arrestin complex structure has yet been determined, limiting our understanding of the full pharmacological profile of BAMs. Herein, we developed a chemical protein synthesis strategy to generate neurotensin receptor 1 (NTSR1) with defined hexa-phosphorylation at its C-terminus and resolved high-resolution cryo-EM structures (2.65-2.88 Å) of NTSR1 in complex with both β-arrestin1 and the BAM SBI-553. These structures revealed a unique "loop engagement" configuration of β-arrestin1 coupling to NTSR1 in the presence of SBI-553, markedly different from the typical "core engagement" configuration observed in the absence of BAMs. This configuration is characterized by the engagement of the intracellular loop 3 of NTSR1 with a cavity in the central crest of β-arrestin1, representing a previously unobserved, arrestin-selective conformation of GPCR. Our findings fill the critical knowledge gap regarding the regulation of GPCR-arrestin interactions and biased signaling by BAMs, which would advance the development of safer and more efficacious GPCR-targeted therapeutics.
Collapse
Affiliation(s)
- Demeng Sun
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Xiang Li
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Qingning Yuan
- State Key Laboratory of Drug Research, Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yuanxia Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Pan Shi
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Huanhuan Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Tao Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wenjing Sun
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shenglong Ling
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- School of Biomedical Engineering and Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China
| | - Yuanchun Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China
| | - Jinglin Lai
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenqin Xie
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanchao Yin
- State Key Laboratory of Drug Research, Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Lei Liu
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, China.
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Changlin Tian
- Hefei National Laboratory for Physical Sciences at the Microscale, Joint Center for Biological Analytical Chemistry, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- School of Biomedical Engineering and Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu, China.
- School of Chemistry and Chemical Engineering & Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
32
|
Liu X, Deng C, Deng Y, Luo X, Zhang W. Molecule-rich solutions for achieving novel non-opioid analgesics. Drug Discov Today 2025; 30:104329. [PMID: 40081520 DOI: 10.1016/j.drudis.2025.104329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Despite their efficacy, opioids have long been associated with risks of addiction, tolerance, and dependence, leaving an unmet clinical need for pain treatment. Efforts have been devoted to developing novel classes of pain-relieving medication that outperform current options in terms of pain relief, side-effect profiles, and potential for abuse, but with limited success. Recent advances in the neurobiology of pain have shed light on the potential of targeting non-opioid receptors involved in pain processing. In this review, we identify avenues, ranging from molecular-based approaches to molecule-rich solutions, for effectively identifying non-opioid analgesics free from the side effects associated with opioids.
Collapse
Affiliation(s)
- Xingxing Liu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Chaoyi Deng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research, Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Deng
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research, Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xudong Luo
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Pharmacy, West China Tianfu Hospital, Sichuan University, Chengdu 610213, China
| | - Wensheng Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research, Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
33
|
Jackson CC, Liu J(J, Liu HY, Williams SG, Anees A, Noor Z, Lucas N, Xavier D, Hains PG, Bucio-Noble D, Aref AT, Porceddu SV, Ladwa R, Whitfield J, Reddel RR, Zhong Q, Panizza BJ, Robinson PJ. A Proteomic Signature for Human Papillomavirus-Associated Oropharyngeal Squamous Cell Carcinoma Predicts Patients at High Risk of Recurrence. CANCER RESEARCH COMMUNICATIONS 2025; 5:580-593. [PMID: 40014866 PMCID: PMC11979894 DOI: 10.1158/2767-9764.crc-23-0460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/21/2023] [Accepted: 02/25/2025] [Indexed: 03/01/2025]
Abstract
SIGNIFICANCE HPV+OPSCC incidence is increasing, with heterogeneous treatment outcomes despite favorable prognosis. Current de-escalation strategies show inferior results, highlighting the need for precise risk stratification. Using data-independent acquisition mass spectrometry proteomics, we identified a 26-peptide signature that stratifies patients into risk categories, potentially enabling personalized treatment decisions and optimal patient selection for de-escalation trials.
Collapse
Affiliation(s)
- Christopher C. Jackson
- Department of Otolaryngology, Head and Neck Surgery, Princess Alexandra Hospital, Brisbane, Australia
- Queensland Head and Neck Cancer Centre, Princess Alexandra Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Jia (Jenny) Liu
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
- The Kinghorn Cancer Centre, St. Vincent’s Hospital, Darlinghurst, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Sydney, Australia
| | - Howard Y. Liu
- Faculty of Medicine, University of Queensland, Brisbane, Australia
- Department of Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - Steven G. Williams
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Asim Anees
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Zainab Noor
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Natasha Lucas
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Dylan Xavier
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Peter G. Hains
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Daniel Bucio-Noble
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Adel T. Aref
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Sandro V. Porceddu
- Department of Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - Rahul Ladwa
- Department of Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - Joseph Whitfield
- Pathology Queensland, Princess Alexandra Hospital, Brisbane, Australia
| | - Roger R. Reddel
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Qing Zhong
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Benedict J. Panizza
- Department of Otolaryngology, Head and Neck Surgery, Princess Alexandra Hospital, Brisbane, Australia
- Queensland Head and Neck Cancer Centre, Princess Alexandra Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Phillip J. Robinson
- ProCan, Children’s Medical Research Institute, The University of Sydney, Sydney, Australia
| |
Collapse
|
34
|
Li J, Bao J, Liu Y, Chen M, Chen Y, Tuolihong L, Jiang F, Xie S, Lyu F, Sun Y, Cao Y, Chen H, Chen Z, Zeng Z. Lentinan enhances microbiota-derived isoursodeoxycholic acid levels to alleviate hepatic ischemia-reperfusion injury in mice. Int J Biol Macromol 2025; 304:140717. [PMID: 39920949 DOI: 10.1016/j.ijbiomac.2025.140717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is an essential clinical concern caused by liver transplantation, resection, trauma, and shock that must be addressed immediately. Although the mechanisms underlying HIRI are well-documented, effective prevention and treatment strategies are still lacking. Inflammation is a central mechanism of HIRI, with macrophages playing a crucial role in initiating and amplifying the inflammatory response. Numerous plant polysaccharides exhibit substantial anti-inflammatory and hepatoprotective properties. However, the function of Lentinan (LNT) in HIRI has not been fully explored. Thus, this study aims to investigate the preventive potential of LNT in HIRI. Here, we reveal that oral administration of LNT considerably reduces hepatic inflammation and improves liver pathology in mice with HIRI by modulating gut microbiota. Specifically, LNT considerably increased microbiota-derived isoursodeoxycholic acid (IsoUDCA). Further experiments showed that IsoUDCA alleviates hepatic injury by suppressing macrophage inflammation. Mechanistically, IsoUDCA directly binds to and activates the neuron-derived clone 77 (Nur77) transcription factor, inhibiting the NF-κB signaling pathway in macrophages. Our findings shed light on the significant role of the LNT-microbiota-IsoUDCA-Nur77 axis in attenuating macrophage inflammation during HIRI, offering novel insights into potential therapeutic targets and avenues for preventing HIRI.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jingna Bao
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yihong Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Meiling Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuqi Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lina Tuolihong
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fuhui Jiang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shihao Xie
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fengyuan Lyu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ye Sun
- Department of Critical Care Medicine, Yuebei People's Hospital, Shaoguan 512000, Guangdong, China
| | - Yan Cao
- Department of Critical Care Medicine, Yuebei People's Hospital, Shaoguan 512000, Guangdong, China
| | - Huarong Chen
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong.
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
35
|
Matthews I, Mehra P, Suárez-Calvet X, Piñol-Jurado P, Cox D, Justian V, Carrasco-Rozas A, Laidler Z, Bowey A, Rushton P, López-Fernández S, Díaz-Manera J, Fernández-Simón E. Strategy for drug repurposing in fibroadipogenic replacement during muscle wasting: application to duchenne muscular dystrophy. Front Cell Dev Biol 2025; 13:1505697. [PMID: 40206397 PMCID: PMC11979640 DOI: 10.3389/fcell.2025.1505697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Background Understanding the cell functionality during disease progression or drugs' mechanism are major challenges for precision medicine. Predictive models describing biological phenotypes can be challenging to obtain, particularly in scenarios where sample availability is limited, such as in the case of rare diseases. Here we propose a new method that reproduces the fibroadipogenic expansion that occurs in muscle wasting. Methods We used immortalized fibroadipogenic progenitor cells (FAPs) and differentiated them into fibroblasts or adipocytes. The method successfully identified FAPs cell differentiation fate using accurate measurements of changes in specific proteins, which ultimately constitute a valid cellular in vitro platform for drug screening. Results were confirmed using primary FAPs differentiation as well as comparison with omics data from proteomics and genomic studies. Results Our method allowed us to screen 508 different drugs from 2 compounds libraries. Out of these 508, we identified 4 compounds that reduced fibrogenesis and adipogenesis of ≥30% of fibrogenesis and adipogenesis using immortalized cells. After selecting the optimal dose of each compound, the inhibitory effect on FAP differentiation was confirmed by using primary FAPs from healthy subjects (n = 3) and DMD patients (n = 3). The final 4 selected hits reduced fibrogenic differentiation in healthy and DMD samples. The inhibition of adipogenesis was more evident in DMD samples than healthy samples. After creating an inhibitory map of the tested drugs, we validated the signalling pathways more involved in FAPs differentiation analysing data from proteomic and genomic studies. Conclusion We present a map of molecular targets of approved drugs that helps in predicting which therapeutic option may affect FAP differentiation. This method allows to study the potential effect of signalling circuits on FAP differentiation after drug treatment providing insights into molecular mechanism of action of muscle degeneration. The accuracy of the method is demonstrated by comparing the signal pathway activity obtained after drug treatment with proteomic and genomic data from patient-derived cells.
Collapse
Affiliation(s)
- Izzy Matthews
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
| | - Priyanka Mehra
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Xavier Suárez-Calvet
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Patricia Piñol-Jurado
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dan Cox
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Vellia Justian
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ana Carrasco-Rozas
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Zoe Laidler
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew Bowey
- Great North Children’s Hospital, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Paul Rushton
- Great North Children’s Hospital, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Susana López-Fernández
- Plastic Surgery Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Díaz-Manera
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Esther Fernández-Simón
- John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
36
|
Yuan Y, Yu L, Bi C, Huang L, Su B, Nie J, Dou Z, Yang S, Li Y. A new paradigm for drug discovery in the treatment of complex diseases: drug discovery and optimization. Chin Med 2025; 20:40. [PMID: 40122800 PMCID: PMC11931805 DOI: 10.1186/s13020-025-01075-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
In the past, the drug research and development has predominantly followed a "single target, single disease" model. However, clinical data show that single-target drugs are difficult to interfere with the complete disease network, are prone to develop drug resistance and low safety in clinical use. The proposal of multi-target drug therapy (also known as "cocktail therapy") provides a new approach for drug discovery, which can affect the disease and reduce adverse reactions by regulating multiple targets. Natural products are an important source for multi-target innovative drug development, and more than half of approved small molecule drugs are related to natural products. However, there are many challenges in the development process of natural products, such as active drug screening, target identification and preclinical dosage optimization. Therefore, how to develop multi-target drugs with good drug resistance from natural products has always been a challenge. This article summarizes the applications and shortcomings of related technologies such as natural product bioactivity screening, clarify the mode of action of the drug (direct/indirect target), and preclinical dose optimization. Moreover, in response to the challenges faced by natural products in the development process and the trend of interdisciplinary and multi-technology integration, and a multi-target drug development strategy of "active substances - drug action mode - drug optimization" is proposed to solve the key challenges in the development of natural products from multiple dimensions and levels.
Collapse
Affiliation(s)
- Yu Yuan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lulu Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Chenghao Bi
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Liping Huang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Buda Su
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Collaborative Innovation Center of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, 010110, China
| | - Jiaxuan Nie
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhiying Dou
- School of Traditional Chinese Medicine, Tianjin University of Chinese Medicine, Tianjin, 301617, China.
| | - Shenshen Yang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yubo Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
37
|
Barkdull AP, Holcomb M, Forli S. A quantitative analysis of ligand binding at the protein-lipid bilayer interface. Commun Chem 2025; 8:89. [PMID: 40121339 PMCID: PMC11929912 DOI: 10.1038/s42004-025-01472-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
The majority of drugs target membrane proteins, and many of these proteins contain ligand binding sites embedded within the lipid bilayer. However, targeting these therapeutically relevant sites is hindered by limited characterization of both the sites and the molecules that bind to them. Here, we introduce the Lipid-Interacting LigAnd Complexes Database (LILAC-DB), a curated dataset of 413 structures of ligands bound at the protein-bilayer interface. Analysis of these structures reveals that ligands binding to lipid-exposed sites exhibit distinct chemical properties, such as higher calculated partition coefficient (clogP), molecular weight, and a greater number of halogen atoms, compared to ligands that bind to soluble proteins. Additionally, we demonstrate that the atomic properties of these ligands vary significantly depending on their depth within and exposure to the lipid bilayer. We also find that ligand binding sites exposed to the bilayer have distinct amino acid compositions compared to other protein regions, which may aid in the identification of lipid-exposed binding sites. This analysis provides valuable guidelines for researchers pursuing structure-based drug discovery targeting underexploited ligand binding sites at the protein-lipid bilayer interface.
Collapse
Affiliation(s)
- Allison Pearl Barkdull
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Matthew Holcomb
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
38
|
Yu X, He Y, Kamenecka TM, Kojetin DJ. Towards a unified molecular mechanism for liganddependent activation of NR4A-RXR heterodimers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.642122. [PMID: 40166180 PMCID: PMC11956975 DOI: 10.1101/2025.03.19.642122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
A subset of nuclear receptors (NRs) function as permissive heterodimers with retinoid X receptor (RXR), defined by transcriptional activation in response to binding RXR agonist ligands. Permissive NR-RXR activation operates via a classical pharmacological mechanism, where binding of an RXR agonist increases coactivator recruitment to the heterodimer. However, we previously demonstrated that transcriptional activation of permissive Nurr1-RXRα (NR4A2-NR2B1) heterodimers by an RXR ligand set, which included pharmacological RXR agonists and selective Nurr1-RXRα agonists that function as antagonists of RXRα homodimers, occurs via a non-classical mechanism: ligand-binding domain (LBD) heterodimer dissociation (Yu et al., 2023). Here, we extend mechanistic ligand profiling of the same RXR ligand set to Nur77-RXRγ (NR4A1-NR2B3), which is evolutionarily related to Nurr1-RXRα. Biochemical and NMR protein-protein interaction profiling along with cellular transcription studies indicate that the RXR ligand set, which lacks selective Nur77-RXRγ agonists, may influence Nur77-RXRγ transcriptional activation through both classical pharmacological activation and LBD heterodimer dissociation. However, upon reanalyzing our previously published data for Nurr1-RXRα, we found that the inclusion of selective Nurr1-RXRα agonists was essential for elucidating the LBD heterodimer dissociation mechanism. Our findings underscore the need for a more functionally diverse RXR ligand set to explore Nur77-RXRγ activation and unify LBD heterodimer dissociation as a potential targeting mechanism for NR4A-RXR heterodimers in neurodegenerative and inflammatory diseases.
Collapse
Affiliation(s)
- Xiaoyu Yu
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
- Department of Integrative Structural and Computational Biology, Scripps Research and The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida, United States
| | - Yuanjun He
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Theodore M. Kamenecka
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Douglas J. Kojetin
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
- Department of Integrative Structural and Computational Biology, Scripps Research and The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida, United States
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, United States
- Center for Applied AI in Protein Dynamics, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
39
|
Kopac T. Leveraging Artificial Intelligence and Machine Learning for Characterizing Protein Corona, Nanobiological Interactions, and Advancing Drug Discovery. Bioengineering (Basel) 2025; 12:312. [PMID: 40150776 PMCID: PMC11939375 DOI: 10.3390/bioengineering12030312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Proteins are essential for all living organisms, playing key roles in biochemical reactions, structural support, signal transduction, and gene regulation. Their importance in biomedical research is highlighted by their role as drug targets in various diseases. The interactions between proteins and nanoparticles (NPs), including the protein corona's formation, significantly affect NP behavior, biodistribution, cellular uptake, and toxicity. Comprehending these interactions is pivotal for advancing the design of NPs to augment their efficacy and safety in biomedical applications. While traditional nanomedicine design relies heavily on experimental work, the use of data science and machine learning (ML) is on the rise to predict the synthesis and behavior of nanomaterials (NMs). Nanoinformatics combines computational simulations with laboratory studies, assessing risks and revealing complex nanobio interactions. Recent advancements in artificial intelligence (AI) and ML are enhancing the characterization of the protein corona and improving drug discovery. This review discusses the advantages and limitations of these approaches and stresses the importance of comprehensive datasets for better model accuracy. Future developments may include advanced deep-learning models and multimodal data integration to enhance protein function prediction. Overall, systematic research and advanced computational tools are vital for improving therapeutic outcomes and ensuring the safe use of NMs in medicine.
Collapse
Affiliation(s)
- Turkan Kopac
- Department of Chemistry, Zonguldak Bülent Ecevit University, 67100 Zonguldak, Türkiye
| |
Collapse
|
40
|
Skiba MA, Canavan C, Nemeth GR, Liu J, Kanso A, Kruse AC. Epitope-directed selection of GPCR nanobody ligands with evolvable function. Proc Natl Acad Sci U S A 2025; 122:e2423931122. [PMID: 40067891 PMCID: PMC11929449 DOI: 10.1073/pnas.2423931122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Antibodies have the potential to target G protein-coupled receptors (GPCRs) with high receptor, cellular, and tissue selectivity; however, few antibody ligands for GPCRs exist. Here, we describe a generalizable selection method to enrich for GPCR ligands from a synthetic camelid antibody fragment (nanobody) library. Our strategy yielded multiple nanobody ligands for the angiotensin II type I receptor (AT1R), a prototypical GPCR and important drug target. We found that nanobodies readily act as allosteric modulators, encoding selectivity for both the receptor and chemical features of GPCR ligands. We then used structure-guided design to convert two nanobodies from allosteric ligands to competitive AT1R inhibitors through simple mutations. This work demonstrates that nanobodies can encode multiple pharmacological behaviors and have great potential as evolvable scaffolds for the development of next-generation GPCR therapeutics.
Collapse
Affiliation(s)
- Meredith A. Skiba
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Clare Canavan
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Genevieve R. Nemeth
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Jinghan Liu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Ali Kanso
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Andrew C. Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| |
Collapse
|
41
|
Ro SY, Jao C, Oh A, Kschonsak M, Li T, Austin D, Greiner DMZ, Zhou L, Zhang Y, Chen J, Quinn JG. Fab-Induced Stabilization of an Ion Channel Receptor Enables Mechanistic Characterization of Small-Molecule Therapeutics. Anal Chem 2025; 97:5102-5108. [PMID: 39993138 PMCID: PMC11913017 DOI: 10.1021/acs.analchem.4c06339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025]
Abstract
Developing small-molecule (SM) therapeutics that target membrane proteins (MPs) is often challenging, because few biophysical methods can handle the detergents required to maintain target stability. Here, we report a surface plasmon resonance (SPR)-based methodology that enables the characterization of interactions between SMs and an ion channel receptor (MP1) in complex with a stabilizing antibody fragment (Fab) and surfactant. Briefly, a stable MP1-Fab complex was formed by coimmobilizing MP1 with an anti-MP1-Fab within the hydrogel film to study the interactions of MP1 with SMs. The critical micelle concentration (CMC) is the concentration at which 50% of the surfactant monomers are assembled into micelles. Micelles readily absorb compounds resulting in compound-loaded micelles that generate high nonspecific binding and hinder resolution of SM binding responses. This micelle-induced interference was avoided by utilizing a weak detergent at a concentration below its CMC, allowing for the resolution of compound binding to a solvent-exposed pocket. Additional Fab stabilization was required to rescue binding at a second pocket buried within the transmembrane region of MP1. The resulting SPR-based assay proved invaluable during hit-to-lead optimization by progressing structure-activity relationship (SAR) studies and resolving the mechanism of action (MoA).
Collapse
Affiliation(s)
- Soo Y Ro
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Christine Jao
- Department of Protein Sciences, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Angela Oh
- Department of Protein Sciences, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Marc Kschonsak
- Department of Protein Sciences, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Tianbo Li
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel Austin
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Demi Maria Z Greiner
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Lijuan Zhou
- Department of Early Discovery Biochemistry, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Yingnan Zhang
- Department of Early Discovery Biochemistry, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - Jun Chen
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| | - John G Quinn
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc. All located at 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
42
|
Kyro GW, Smaldone AM, Shee Y, Xu C, Batista VS. T-ALPHA: A Hierarchical Transformer-Based Deep Neural Network for Protein-Ligand Binding Affinity Prediction with Uncertainty-Aware Self-Learning for Protein-Specific Alignment. J Chem Inf Model 2025; 65:2395-2415. [PMID: 39965912 DOI: 10.1021/acs.jcim.4c02332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
There is significant interest in targeting disease-causing proteins with small molecule inhibitors to restore healthy cellular states. The ability to accurately predict the binding affinity of small molecules to a protein target in silico enables the rapid identification of candidate inhibitors and facilitates the optimization of on-target potency. In this work, we present T-ALPHA, a novel deep learning model that enhances protein-ligand binding affinity prediction by integrating multimodal feature representations within a hierarchical transformer framework to capture information critical to accurately predicting binding affinity. T-ALPHA outperforms all existing models reported in the literature on multiple benchmarks designed to evaluate protein-ligand binding affinity scoring functions. Remarkably, T-ALPHA maintains state-of-the-art performance when utilizing predicted structures rather than crystal structures, a powerful capability in real-world drug discovery applications where experimentally determined structures are often unavailable or incomplete. Additionally, we present an uncertainty-aware self-learning method for protein-specific alignment that does not require additional experimental data and demonstrate that it improves T-ALPHA's ability to rank compounds by binding affinity to biologically significant targets such as the SARS-CoV-2 main protease and the epidermal growth factor receptor. To facilitate implementation of T-ALPHA and reproducibility of all results presented in this paper, we made all of our software available at https://github.com/gregory-kyro/T-ALPHA.
Collapse
Affiliation(s)
- Gregory W Kyro
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, Unites States
| | - Anthony M Smaldone
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, Unites States
| | - Yu Shee
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, Unites States
| | - Chuzhi Xu
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, Unites States
| | - Victor S Batista
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, Unites States
| |
Collapse
|
43
|
Kelly-Laubscher R, Koenig J, Cunningham M, Aljofan M, Babey AM, Hawes M, Hinton T, Karpa K, Karunaratne N, Nicolazzo J, Liang W, Mraiche F, Restini C, Santiago M, Volbrecht K, Guilding C, White PJ. Evaluating student understanding of pharmacodynamics core concepts. Eur J Pharmacol 2025; 990:177257. [PMID: 39788406 DOI: 10.1016/j.ejphar.2025.177257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
Pharmacodynamics is an essential subdiscipline of pharmacology that underpins safe and effective prescribing and therapeutic decision-making, as well as drug discovery and development. The exponential increase in the number of therapeutic drugs has prompted members of the pharmacology educator community to question existing pharmacology curricula focused on individual drugs and move toward a curriculum focused on conceptual understanding. A first step towards conceptual understanding is to establish what students currently know about pharmacodynamic core concepts. A total of 218 students from 10 universities were invited to complete a questionnaire that assessed their understanding of drug efficacy, drug-target interaction, drug tolerance, and structure-activity relationship. Pairs of pharmacology experts independently assessed each student's response and flagged any misconceptions that arose. The experts then compared their evaluations, achieved a consensus decision, and grouped the misconceptions into themes. Less than 25% of students provided core concept meanings that fully aligned with those of the expert group. By contrast, more than 75% of students could apply the core concept to a novel scenario at least in part. Overall, 480 misconceptions were identified and grouped into 55 misconception themes. The concept of drug efficacy was the core concept with which students struggled most. It is unclear why students were better able to apply their knowledge than to define the core concepts, although this might reflect a focus on active learning in pharmacology courses globally. The deficits in defining and understanding pharmacodynamic core concepts, and the misconceptions revealed in student responses, can be used by educators to guide their efforts.
Collapse
Affiliation(s)
- Roisin Kelly-Laubscher
- Dept. Pharmacology & Therapeutics, School of Medicine, College of Medicine and Health, University College Cork, Ireland
| | - Jennifer Koenig
- Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| | - Margaret Cunningham
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Mohamad Aljofan
- Department of Biomedical Science, School of Medicine Nazarbayev University, Astana, Kazakhstan
| | - Anna-Marie Babey
- School of Science & Technology, University of New England, Australia
| | - Martin Hawes
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, University of Surrey, UK
| | - Tina Hinton
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kelly Karpa
- East Tennessee State University, Quillen College of Medicine, USA
| | - Nilushi Karunaratne
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Australia
| | - Joseph Nicolazzo
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Australia
| | - Willmann Liang
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Fatima Mraiche
- Department of Pharmacology, Faculty of Medicine & Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Carolina Restini
- Department of Pharmacology and Toxicology, Michigan State University, College of Osteopathic Medicine, MI, USA
| | - Marina Santiago
- Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Kieran Volbrecht
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Australia
| | - Clare Guilding
- School of Medicine, Faculty of Medical Science, Newcastle University, UK
| | - Paul J White
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Australia.
| |
Collapse
|
44
|
Qi Y, Jiang H, Lun Y, Gang Q, Shen S, Zhang H, Liu M, Wang Y, Zhang J. Protein Drug Targets for Abdominal Aortic Aneurysm and Proteomic Associations Between Modifiable Risk Factors and Abdominal Aortic Aneurysm. J Am Heart Assoc 2025; 14:e037802. [PMID: 40008516 DOI: 10.1161/jaha.124.037802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a severe aortic disease for which no pharmacological interventions have yet been developed. This investigation focused on identifying protein-based therapeutic targets and assessing how proteins mediate the interplay between modifiable risk factors and AAA development. METHODS Causal inferences between plasma proteins and AAA were drawn using 2-sample Mendelian randomization, followed by comprehensive sensitivity testing, colocalization, and replication efforts. Further analyses included database interrogation, single-cell RNA data analysis, enrichment analysis, protein-protein interaction networks, and immunohistochemistry to map the tissue-specific expression of these proteins, their expression within AAA tissues, and their biological roles. Mediation Mendelian randomization was employed to evaluate the mediating effects of AAA-related proteins on the associations between AAA and 3 risk factors: hypertension, smoking, and obesity. RESULTS A total of 43 proteins were identified as having causal links to AAA. Colocalization analysis pinpointed 13 proteins with strong evidence of colocalization with AAA. Of these, the causal involvement of 10 proteins was substantiated by external validation data. Consistent evidence for PCSK9 (proprotein convertase subtilisin/kexin type 9), IL6R (interleukin-6R), ECM1 (extracellular matrix protein 1), and ANGPTL4 (angiopoietin-related protein 4) was further validated through tissue immunohistochemistry and blood data. Moreover, Mendelian randomization analysis identified 10 proteins as mediators of the influence of hypertension, smoking, and obesity on AAA development. CONCLUSIONS This analysis identifies 4 proteins (PCSK9, IL6R, ECM1, and ANGPTL4) as high-priority therapeutic targets for AAA and emphasizes the intermediary role of plasma proteins in linking hypertension, smoking, obesity, and AAA. Further investigations are needed to clarify the specific roles of these proteins in AAA pathology.
Collapse
Affiliation(s)
- Yao Qi
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Jiang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yu Lun
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Qingwei Gang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Shikai Shen
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Mingyu Liu
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yixian Wang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| |
Collapse
|
45
|
Lorente JS, Sokolov AV, Ferguson G, Schiöth HB, Hauser AS, Gloriam DE. GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov 2025:10.1038/s41573-025-01139-y. [PMID: 40033110 DOI: 10.1038/s41573-025-01139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 03/05/2025]
Abstract
G protein-coupled receptors (GPCRs) form one of the largest drug target families, reflecting their involvement in numerous pathophysiological processes. In this Review, we analyse drug discovery trends for the GPCR superfamily, covering compounds, targets and indications that have reached regulatory approval or that are being investigated in clinical trials. We find that there are 516 approved drugs targeting GPCRs, making up 36% of all approved drugs. These drugs act on 121 GPCR targets, one-third of all non-sensory GPCRs. Furthermore, 337 agents targeting 133 GPCRs, including 30 novel targets, are being investigated in clinical trials. Notably, 165 of these agents are approved drugs being tested for additional indications and novel agents are increasingly allosteric modulators and biologics. Remarkably, diabetes and obesity drugs targeting GPCRs had sales of nearly US $30 billion in 2023 and the numbers of clinical trials for GPCR modulators in the metabolic diseases, oncology and immunology areas are increasing strongly. Finally, we highlight the potential of untapped target-disease associations and pathway-biased signalling. Overall, this Review provides an up-to-date reference for the drugged and potentially druggable GPCRome to inform future GPCR drug discovery and development.
Collapse
Affiliation(s)
- Javier Sánchez Lorente
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aleksandr V Sokolov
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Gavin Ferguson
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- ALPX S.A.S., Grenoble, France
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Alexander S Hauser
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David E Gloriam
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
46
|
Gong J, Williams DM, Scholes S, Assaad S, Bu F, Hayat S, Zaninotto P, Steptoe A. Unraveling the role of proteins in dementia: insights from two UK cohorts with causal evidence. Brain Commun 2025; 7:fcaf097. [PMID: 40092369 PMCID: PMC11906402 DOI: 10.1093/braincomms/fcaf097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/16/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025] Open
Abstract
Population-based proteomics offers a groundbreaking avenue to predict future disease risks, enhance our understanding of disease mechanisms, and discover novel therapeutic targets and biomarkers. The role of plasma proteins in dementia, however, requires further exploration. This study investigated 276 protein-dementia associations in 229 incident all-cause dementia, 89 Alzheimer's disease, and 41 vascular dementia among 3249 participants (55% women, 97.2% white ethnicity) from the English Longitudinal Study of Ageing (ELSA) over a median 9.8-year follow-up. We used Cox proportional hazard regression for the analysis. Receiver operating characteristic analyses were conducted to assess the precision of the identified proteins from the fully adjusted Cox regression models in predicting incident all-cause dementia, both individually and in combination with demographic predictors, APOE genotype, and memory score, to estimate the area under the curve. Additionally, the eXtreme Gradient Boosting machine learning algorithm was used to identify the most important features predictive of future all-cause dementia onset. These associations were then validated in 1506 incident all-cause dementia, 732 Alzheimer's disease, 281 vascular dementia, and 111 frontotemporal dementia cases among 52 745 individuals (53.9% women, 93.3% White ethnicity) from the UK Biobank over a median 13.7-year follow-up. Two-sample bi-directional Mendelian randomization and drug target Mendelian randomization were further employed to determine the causal direction between protein concentration and dementia. NEFL (hazard ratio [HR] [95% confidence intervals (CIs)]: 1.54 [1.29, 1.84]) and RPS6KB1 (HR [95% CI]: 1.33 [1.16, 1.52]) were robustly associated with incident all-cause dementia; MMP12 (HR [95% CI]: 2.06 [1.41, 2.99]) was associated with vascular dementia in ELSA, after correcting for multiple testing. Additional markers EDA2R and KIM1 were identified from subgroup and sensitivity analyses. Combining NEFL and RPS6KB1 with other predictors yielded high predictive accuracy (area under the curve = 0.871) for incident all-cause dementia. The eXtreme Gradient Boosting machine learning algorithm also identified RPS6KB1, NEFL, and KIM1 as the most important protein features for predicting future all-cause dementia. Sex difference was evident for the association between RPS6KB1 and all-cause dementia, with stronger association in men (P for interaction = 0.037). Replication in the UK Biobank confirmed the associations between the identified proteins and various dementia subtypes. The results from Mendelian randomization in the reverse direction indicated that several proteins serve as early markers for dementia, rather than being direct causes of the disease. These findings provide insights into putative mechanisms for dementia. Future studies are needed to validate the findings on RPS6KB1 in relation to dementia risk.
Collapse
Affiliation(s)
- Jessica Gong
- Department of Epidemiology and Public Health, University College London, London WC1E 7HB, UK
- George Institute for Global Health, Imperial College London, London W12 7RZ, UK
| | - Dylan M Williams
- MRC Unit for Lifelong Health & Ageing, University College London, London WC1E 7HB, UK
| | - Shaun Scholes
- Department of Epidemiology and Public Health, University College London, London WC1E 7HB, UK
| | - Sarah Assaad
- Department of Epidemiology and Public Health, University College London, London WC1E 7HB, UK
| | - Feifei Bu
- Department of Behavioural Science and Health, University College London, London WC1E 7HB, UK
| | - Shabina Hayat
- Department of Epidemiology and Public Health, University College London, London WC1E 7HB, UK
| | - Paola Zaninotto
- Department of Epidemiology and Public Health, University College London, London WC1E 7HB, UK
| | - Andrew Steptoe
- Department of Behavioural Science and Health, University College London, London WC1E 7HB, UK
| |
Collapse
|
47
|
Belbasis L, Morris S, van Duijn C, Bennett D, Walters R. Mendelian randomization identifies proteins involved in neurodegenerative diseases. Brain 2025:awaf018. [PMID: 40037332 DOI: 10.1093/brain/awaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 10/26/2024] [Accepted: 12/20/2024] [Indexed: 03/06/2025] Open
Abstract
Proteins are involved in multiple biological functions. High-throughput technologies have allowed the measurement of thousands of proteins in population biobanks. In this study, we aimed to identify proteins related to Alzheimer's disease, Parkinson's disease, multiple sclerosis and amyotrophic lateral sclerosis by leveraging large-scale genetic and proteomic data. We performed a two-sample cis Mendelian randomization study by selecting instrumental variables for the abundance of >2700 proteins measured by either Olink or SomaScan platforms in plasma from the UK Biobank and the deCODE Health Study. We also used the latest publicly available genome-wide association studies for the neurodegenerative diseases of interest. The potentially causal effect of proteins on neurodegenerative diseases was estimated based on the Wald ratio. We tested 13 377 protein-disease associations, identifying 169 associations that were statistically significant (5% false discovery rate). Evidence of co-localization between plasma protein abundance and disease risk (posterior probability > 0.80) was identified for 61 protein-disease pairs, leading to 50 unique protein-disease associations. Notably, 23 of 50 protein-disease associations corresponded to genetic loci not previously reported by genome-wide association studies. The two-sample Mendelian randomization and co-localization analysis also showed that APOE abundance in plasma was associated with three subcortical volumes (hippocampus, amygdala and nucleus accumbens) and white matter hyper-intensities, whereas PILRA and PILRB abundance in plasma was associated with caudate nucleus volume. Our study provided a comprehensive assessment of the effect of the human proteome that is currently measurable through two different platforms on neurodegenerative diseases. The newly associated proteins indicated the involvement of complement (C1S and C1R), microglia (SIRPA, SIGLEC9 and PRSS8) and lysosomes (CLN5) in Alzheimer's disease; the interleukin-6 pathway (CTF1) in Parkinson's disease; lysosomes (TPP1), blood-brain barrier integrity (MFAP2) and astrocytes (TNFSF13) in amyotrophic lateral sclerosis; and blood-brain barrier integrity (VEGFB), oligodendrocytes (PARP1), node of Ranvier and dorsal root ganglion (NCS1, FLRT3 and CDH15) and the innate immune system (CR1, AHSG and WARS) in multiple sclerosis. Our study demonstrates how harnessing large-scale genomic and proteomic data can yield new insights into the role of the plasma proteome in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lazaros Belbasis
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Sam Morris
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Derrick Bennett
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Robin Walters
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| |
Collapse
|
48
|
Xia B, Shaheen N, Chen H, Zhao J, Guo P, Zhao Y. RNA aptamer-mediated RNA nanotechnology for potential treatment of cardiopulmonary diseases. Pharmacol Res 2025; 213:107659. [PMID: 39978660 DOI: 10.1016/j.phrs.2025.107659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/14/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Ribonucleic acid (RNA) aptamers are single-stranded RNAs that bind to target proteins or other molecules with high specificity and affinity, modulating biological functions through distinct mechanisms. These aptamers can act n as antagonists to block pathological interactions, agonists to activate signaling pathways, or delivery vehicles for therapeutic cargos such as siRNAs and miRNAs. The advances in RNA nanotechnology further enhances the versatility of RNA aptamers, offering scalable platforms for engineering. In this review, we have summarized recent developments in RNA aptamer-mediated RNA nanotechnology and provide an overview of its potential in treating cardiovascular and respiratory disorders, including atherosclerosis, acute coronary syndromes, heart failure, lung cancer, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), acute lung injury, viral respiratory infections, and pulmonary fibrosis. By integrating aptamer technologies with innovative delivery systems, RNA aptamers hold the potential to revolutionize the treatment landscape for cardiopulmonary diseases.
Collapse
Affiliation(s)
- Boyu Xia
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nargis Shaheen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Huilong Chen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
49
|
Shen C, Zhang R, Yu J, Sahakian BJ, Cheng W, Feng J. Plasma proteomic signatures of social isolation and loneliness associated with morbidity and mortality. Nat Hum Behav 2025; 9:569-583. [PMID: 39753750 PMCID: PMC11936835 DOI: 10.1038/s41562-024-02078-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 10/31/2024] [Indexed: 03/27/2025]
Abstract
The biology underlying the connection between social relationships and health is largely unknown. Here, leveraging data from 42,062 participants across 2,920 plasma proteins in the UK Biobank, we characterized the proteomic signatures of social isolation and loneliness through proteome-wide association study and protein co-expression network analysis. Proteins linked to these constructs were implicated in inflammation, antiviral responses and complement systems. More than half of these proteins were prospectively linked to cardiovascular disease, type 2 diabetes, stroke and mortality during a 14 year follow-up. Moreover, Mendelian randomization (MR) analysis suggested causal relationships from loneliness to five proteins, with two proteins (ADM and ASGR1) further supported by colocalization. These MR-identified proteins (GFRA1, ADM, FABP4, TNFRSF10A and ASGR1) exhibited broad associations with other blood biomarkers, as well as volumes in brain regions involved in interoception and emotional and social processes. Finally, the MR-identified proteins partly mediated the relationship between loneliness and cardiovascular diseases, stroke and mortality. The exploration of the peripheral physiology through which social relationships influence morbidity and mortality is timely and has potential implications for public health.
Collapse
Affiliation(s)
- Chun Shen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ruohan Zhang
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Jintai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Barbara J Sahakian
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Department of Computer Science, University of Warwick, Coventry, UK.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- School of Data Science, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
Lundquist KP, Romeo I, Puglielli RB, Pestalozzi M, Gram ML, Hudson ES, Levi O, Arava YS, Gotfredsen CH, Clausen MH. Design, synthesis, and screening of an RNA optimized fluorinated fragment library. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100215. [PMID: 39828142 DOI: 10.1016/j.slasd.2025.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Fragment-based screening is an efficient method for early-stage drug discovery. In this study, we aimed to create a fragment library optimized for producing high hit rates against RNA targets. RNA has historically been an underexplored target, but recent research suggests potential for optimizing small molecule libraries for RNA binding. We extended this concept to fragment libraries to produce an RNA optimized fluorinated fragment library. We then screened this library, alongside two non-RNA optimized fragment libraries, against three RNA targets: the human cytoplasmic A-site and the S. cerevisiae tRNAAsp anticodon stem loop with and without nucleobase modifications. The screens yielded 24, 31, and 20 hits against the respective targets. Importantly, statistical analysis confirmed a significant overrepresentation of hits in our RNA optimized library. Based on these findings, we propose guidelines for developing RNA optimized fragment libraries. We hope the guidelines will help expediting fragment-based ligand discovery for RNA targets and contribute to presenting RNA as a promising target in drug discovery.
Collapse
Affiliation(s)
- Kasper P Lundquist
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Isabella Romeo
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | | | - Maëlle Pestalozzi
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Marie L Gram
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Emily S Hudson
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Ofri Levi
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa 3200001, Israel
| | - Yoav S Arava
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa 3200001, Israel
| | - Charlotte H Gotfredsen
- NMR Center • DTU, DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark.
| | - Mads H Clausen
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark.
| |
Collapse
|