Background and purpose: The intestine plays a key role in the initiation of sepsis. The gut barrier impedes the translocation of commensal bacteria to the liver in sepsis. Previous studies have reported that angiotensin-(1-7) [Ang-(1-7)] attenuated sepsis-induced organ injury and mortality. However, its role in sepsis-induced intestinal barrier dysfunction remains unclear. Here we have investigated therapeutic effects of Ang-(1-7) on the intestinal barrier dysfunction and dysbiosis in a murine model of sepsis.
Experimental approach: We used a model of sepsis in C57BL/6 mice with caecal ligation and puncture (CLP), to assess mortality and histological and biochemical changes in the gut and liver tissues. Faecal microbiota transplantation (FMT) was used to assess the role of the gut microbiome. 16-s rDNA and metabolomics analyses were performed to characterize differences in the gut microbiome signatures and metabolic profiles.
Key results: Plasma Ang-(1-7) was decreased in patients with sepsis. In CLP mice, exogenous Ang-(1-7) attenuated intestinal barrier dysfunction and liver damage. FMT experiments showed that the protective effects of Ang-(1-7) on the gut depended on the gut microbiota. Furthermore, 16-s ribosomal DNA analysis revealed that Ang-(1-7) treatment increased the abundance of Lactobacillus gasseri (L. gasseri) among commensal bacteria. Mechanistically, L. gasseri regulated the production of antimicrobial peptides in intestinal epithelia by activating NLRP6 inflammation.
Conclusion and implications: Ang-(1-7) protected against sepsis-induced intestine barrier dysfunction and liver injury in mice by modulating gut homeostasis and NLRP6 inflammasome. Ang-(1-7) is a promising candidate drug for protecting intestinal homeostasis in sepsis, offering new insights for clinical treatment.
{"title":"Angiotensin-(1-7) alleviates intestinal barrier dysfunction and dysbiosis in mice with polymicrobial sepsis.","authors":"Jun Wang, Jierui Li, Yuhan Li, Weichang Huang, Chongyang Huang, Qihan Xu, Jing Sun, Jiacheng Gong, Xiaoxin Ma, Guozhen Wang, Ying Meng, Xu Li","doi":"10.1111/bph.70248","DOIUrl":"https://doi.org/10.1111/bph.70248","url":null,"abstract":"<p><strong>Background and purpose: </strong>The intestine plays a key role in the initiation of sepsis. The gut barrier impedes the translocation of commensal bacteria to the liver in sepsis. Previous studies have reported that angiotensin-(1-7) [Ang-(1-7)] attenuated sepsis-induced organ injury and mortality. However, its role in sepsis-induced intestinal barrier dysfunction remains unclear. Here we have investigated therapeutic effects of Ang-(1-7) on the intestinal barrier dysfunction and dysbiosis in a murine model of sepsis.</p><p><strong>Experimental approach: </strong>We used a model of sepsis in C57BL/6 mice with caecal ligation and puncture (CLP), to assess mortality and histological and biochemical changes in the gut and liver tissues. Faecal microbiota transplantation (FMT) was used to assess the role of the gut microbiome. 16-s rDNA and metabolomics analyses were performed to characterize differences in the gut microbiome signatures and metabolic profiles.</p><p><strong>Key results: </strong>Plasma Ang-(1-7) was decreased in patients with sepsis. In CLP mice, exogenous Ang-(1-7) attenuated intestinal barrier dysfunction and liver damage. FMT experiments showed that the protective effects of Ang-(1-7) on the gut depended on the gut microbiota. Furthermore, 16-s ribosomal DNA analysis revealed that Ang-(1-7) treatment increased the abundance of Lactobacillus gasseri (L. gasseri) among commensal bacteria. Mechanistically, L. gasseri regulated the production of antimicrobial peptides in intestinal epithelia by activating NLRP6 inflammation.</p><p><strong>Conclusion and implications: </strong>Ang-(1-7) protected against sepsis-induced intestine barrier dysfunction and liver injury in mice by modulating gut homeostasis and NLRP6 inflammasome. Ang-(1-7) is a promising candidate drug for protecting intestinal homeostasis in sepsis, offering new insights for clinical treatment.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145767250","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Willem Abma, Sven-Erik Dahlén, Craig E Wheelock, Mikael Adner, Mamdoh Al-Amerie, Erik Sachs, Kasra Vali Jalali, Leonardo De Maria, Henric Olsson, Jesper Säfholm
Background and purpose: Interleukin (IL)-13 is implicated in airway hyperreactivity (AHR), a key feature of asthma. We explored the potential anti-AHR activity of selected specialised pro-resolving mediators (SPMs) in IL-13-induced AHR models, using human bronchial smooth muscle cells (BSMCs) and human isolated bronchi.
Experimental approach: Calcium flux responses induced by histamine or LTD4 were assessed in BSMCs preconditioned with IL-13 and SPMs for 24 h. Human bronchi were isolated from lung tissue and preconditioned for 48 h in the presence or absence of IL-13 and SPMs. Concentration-response relationships for histamine and LTD4 were established using myography to determine efficacy (Emax) and potency (pEC50) following interventions.
Key results: In BSMCs, exposure to IL-13 increased calcium flux (Emax) triggered by histamine and LTD4. Protectin D1 (PD1) and maresin 1 (MaR1) reversed this effect, but not lipoxin A4, resolvin D2, and maresin-conjugate in tissue repair 3 (MCTR3). In bronchi, IL-13 exposure amplified contractions to histamine and LTD4, and this enhancement was reversed by PD1 and MaR1. In contrast, PD1 and MaR1 added acutely during myography had no effect on agonist-induced contractility. PD1 attenuated IL-13-induced enhancement of airway contractions triggered by mast cell activation. CysLT1 antagonism did not influence the anti-hyperreactive effect of SPMs. Chemo-informatics revealed structural similarities between PD1 and MaR1 that may explain the anti-hyperreactive action of these two SPMs.
Conclusion and implications: This new anti-hyperreactive action of PD1 and MaR1 encourages further research into their potential as therapies for the treatment of airway hyperreactivity.
{"title":"Protectin D1 and maresin 1 attenuate airway hyperreactivity induced by IL-13 in human isolated small bronchi.","authors":"Willem Abma, Sven-Erik Dahlén, Craig E Wheelock, Mikael Adner, Mamdoh Al-Amerie, Erik Sachs, Kasra Vali Jalali, Leonardo De Maria, Henric Olsson, Jesper Säfholm","doi":"10.1111/bph.70298","DOIUrl":"https://doi.org/10.1111/bph.70298","url":null,"abstract":"<p><strong>Background and purpose: </strong>Interleukin (IL)-13 is implicated in airway hyperreactivity (AHR), a key feature of asthma. We explored the potential anti-AHR activity of selected specialised pro-resolving mediators (SPMs) in IL-13-induced AHR models, using human bronchial smooth muscle cells (BSMCs) and human isolated bronchi.</p><p><strong>Experimental approach: </strong>Calcium flux responses induced by histamine or LTD<sub>4</sub> were assessed in BSMCs preconditioned with IL-13 and SPMs for 24 h. Human bronchi were isolated from lung tissue and preconditioned for 48 h in the presence or absence of IL-13 and SPMs. Concentration-response relationships for histamine and LTD<sub>4</sub> were established using myography to determine efficacy (E<sub>max</sub>) and potency (pEC<sub>50</sub>) following interventions.</p><p><strong>Key results: </strong>In BSMCs, exposure to IL-13 increased calcium flux (E<sub>max</sub>) triggered by histamine and LTD<sub>4</sub>. Protectin D1 (PD1) and maresin 1 (MaR1) reversed this effect, but not lipoxin A<sub>4</sub>, resolvin D2, and maresin-conjugate in tissue repair 3 (MCTR3). In bronchi, IL-13 exposure amplified contractions to histamine and LTD<sub>4</sub>, and this enhancement was reversed by PD1 and MaR1. In contrast, PD1 and MaR1 added acutely during myography had no effect on agonist-induced contractility. PD1 attenuated IL-13-induced enhancement of airway contractions triggered by mast cell activation. CysLT<sub>1</sub> antagonism did not influence the anti-hyperreactive effect of SPMs. Chemo-informatics revealed structural similarities between PD1 and MaR1 that may explain the anti-hyperreactive action of these two SPMs.</p><p><strong>Conclusion and implications: </strong>This new anti-hyperreactive action of PD1 and MaR1 encourages further research into their potential as therapies for the treatment of airway hyperreactivity.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145767326","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Maria Fernanda Pessano Fialho, Raquel Tonello, Evelyne Silva Brum, Gabriela Becker, Nigel W Bunnett, Sara Marchesan Oliveira
Background and purpose: Anastrozole, an aromatase inhibitor, is used to treat postmenopausal women with hormone receptor-positive breast cancer, but also induces musculoskeletal pain and can lead to therapeutic regimen suspension. Aromatase inhibitors promote the release of pro-inflammatory substances from sensitised nerve fibres, which might lead to peripheral mast cell activation.
Experimental approach: We explore if tryptase released by peripheral mast cells could activate the protease-activated receptor 2 (PAR2) to sustain anastrozole-induced painful symptoms.
Key results: Anastrozole caused mechanical allodynia, muscle strength loss, and increased mast cell number and tryptase levels in the plantar tissue of male C57BL/6 mice. Depletion (using compound 48/80) or stabilisation (using ketotifen fumarate) of mast cells prevented anastrozole-induced mechanical allodynia and muscle strength loss. Compound 48/80 also prevented the increase in the number of mast cells in the plantar tissue. Tryptase inhibitors nafamostat and gabexate, or PAR2 inhibitors ENMD-1068 and AZ3451, reduced the anastrozole-induced mechanical allodynia and muscle strength loss. Furthermore, anastrozole did not cause mechanical allodynia and muscle strength loss in global (PAR2-/-) or sensory neuron-specific (PAR2 Nav 1.8-/-) PAR2 knockout mice. Local sub-nociceptive doses of the PAR2 agonists (tryptase, trypsin or 2F) enhanced the anastrozole-induced mechanical sensitivity in wild-type mice, which was reduced by pre-treatment with AZ3451. PAR2-/- or PAR2 Nav 1.8-/- mice treated with anastrozole did not respond to local sub-nociceptive doses of PAR2 agonists.
Conclusions and implications: Our results provide a new mechanism underlying anastrozole-induced pain, highlighting the mast cell/tryptase/PAR2 axis as a therapeutic target to manage painful symptoms.
背景和目的:阿那曲唑是一种芳香酶抑制剂,用于治疗绝经后激素受体阳性乳腺癌妇女,但也会引起肌肉骨骼疼痛,并可能导致治疗方案暂停。芳香酶抑制剂促进促炎物质从敏感的神经纤维释放,这可能导致外周肥大细胞活化。实验方法:我们探索外周肥大细胞释放的胰蛋白酶是否可以激活蛋白酶激活受体2 (PAR2)以维持阿那曲唑诱导的疼痛症状。关键结果:阿那曲唑引起雄性C57BL/6小鼠的机械异常性疼痛,肌肉力量下降,足底组织肥大细胞数量和胰蛋白酶水平升高。肥大细胞耗竭(使用化合物48/80)或稳定(使用富马酸酮替芬)可防止阿那曲唑引起的机械异常性疼痛和肌肉力量丧失。化合物48/80也阻止了足底组织中肥大细胞数量的增加。胰蛋白酶抑制剂纳莫司他和加贝酸酯,或PAR2抑制剂ENMD-1068和AZ3451,减少了阿那曲唑引起的机械异常性疼痛和肌肉力量损失。此外,阿那曲唑不会引起整体(PAR2-/-)或感觉神经元特异性(PAR2 Nav 1.8-/-) PAR2敲除小鼠的机械异常性疼痛和肌肉力量丧失。局部亚伤害性剂量的PAR2激动剂(胰蛋白酶、胰蛋白酶或2F)增强了野生型小鼠阿那曲唑诱导的机械敏感性,而AZ3451预处理降低了这种敏感性。用阿那曲唑治疗的PAR2-/-或PAR2 Nav 1.8-/-小鼠对局部亚伤害性剂量的PAR2激动剂没有反应。结论和意义:我们的研究结果提供了阿那曲唑诱导疼痛的新机制,突出了肥大细胞/胰蛋白酶/PAR2轴作为治疗疼痛症状的治疗靶点。
{"title":"Critical role of the mast cell/tryptase/PAR2 axis in anastrozole-induced pain.","authors":"Maria Fernanda Pessano Fialho, Raquel Tonello, Evelyne Silva Brum, Gabriela Becker, Nigel W Bunnett, Sara Marchesan Oliveira","doi":"10.1111/bph.70280","DOIUrl":"https://doi.org/10.1111/bph.70280","url":null,"abstract":"<p><strong>Background and purpose: </strong>Anastrozole, an aromatase inhibitor, is used to treat postmenopausal women with hormone receptor-positive breast cancer, but also induces musculoskeletal pain and can lead to therapeutic regimen suspension. Aromatase inhibitors promote the release of pro-inflammatory substances from sensitised nerve fibres, which might lead to peripheral mast cell activation.</p><p><strong>Experimental approach: </strong>We explore if tryptase released by peripheral mast cells could activate the protease-activated receptor 2 (PAR2) to sustain anastrozole-induced painful symptoms.</p><p><strong>Key results: </strong>Anastrozole caused mechanical allodynia, muscle strength loss, and increased mast cell number and tryptase levels in the plantar tissue of male C57BL/6 mice. Depletion (using compound 48/80) or stabilisation (using ketotifen fumarate) of mast cells prevented anastrozole-induced mechanical allodynia and muscle strength loss. Compound 48/80 also prevented the increase in the number of mast cells in the plantar tissue. Tryptase inhibitors nafamostat and gabexate, or PAR2 inhibitors ENMD-1068 and AZ3451, reduced the anastrozole-induced mechanical allodynia and muscle strength loss. Furthermore, anastrozole did not cause mechanical allodynia and muscle strength loss in global (PAR2<sup>-/-</sup>) or sensory neuron-specific (PAR2 Na<sub>v</sub> 1.8<sup>-/-</sup>) PAR2 knockout mice. Local sub-nociceptive doses of the PAR2 agonists (tryptase, trypsin or 2F) enhanced the anastrozole-induced mechanical sensitivity in wild-type mice, which was reduced by pre-treatment with AZ3451. PAR2<sup>-/-</sup> or PAR2 Na<sub>v</sub> 1.8<sup>-/-</sup> mice treated with anastrozole did not respond to local sub-nociceptive doses of PAR2 agonists.</p><p><strong>Conclusions and implications: </strong>Our results provide a new mechanism underlying anastrozole-induced pain, highlighting the mast cell/tryptase/PAR2 axis as a therapeutic target to manage painful symptoms.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145755013","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
María Elena Angarita-Planchez, Camila Leiva-Castro, Ana M Múnera-Rodríguez, Macarena Martínez-Bailén, Ana T Carmona, Soledad López-Enríquez, Francisca Palomares
Background and purpose: Immune tolerance prevents inflammation and autoimmunity, with dendritic cells (DCs) playing a key role. Reprogramming DCs towards a tolerogenic state represents a promising therapeutic strategy. Sulforaphane (SFN) has known immunomodulatory effects, but its clinical application is limited by poor stability and bioavailability. To enhance its therapeutic potential, SFN was conjugated with mannose (SFNMan) or fucose (SFNFuc), aiming to induce a tolerogenic phenotype in human monocyte-derived DCs (moDCs) under inflammation and to explore NFATc1's involvement.
Experimental approach: moDCs were exposed to inflammatory conditions and treated with SFN, SFNMan or SFNFuc. Their phenotype, cytokine profile, T cell-modulating capacity and NFATc1 signalling were evaluated.
Key results: SFNMan selectively induced a tolerogenic phenotype, characterised by an increased PD-L1/CD86 ratio and IL-10 production; up-regulation of SOCS1 and IDO transcripts; and Treg expansion and reduced proliferation of cytotoxic T cell proliferation. Functional assays and confocal microscopy revealed that SFNMan, but not SFNFuc, promoted NFATc1 nuclear translocation. Pharmacological inhibition of NFATc1 with cyclosporin A (CsA) abolished these effects, confirming NFATc1 as a central mediator of SFNMan-induced immune tolerance.
Conclusions and implications: Our findings identify NFATc1 as a key transcriptional switch in moDCs tolerogenic programming and highlight the carbohydrate-dependent specificity of SFN conjugates. SFNMan represents a novel carbohydrate-engineered immunomodulator capable of driving immune tolerance through NFATc1 activation. These results provide a mechanistic framework for the development of precision therapies targeting inflammatory and autoimmune diseases.
{"title":"Monovalent mannose-glycoconjugates of sulforaphane reprogram human dendritic cells via NFATc1 to induce immune tolerance under inflammatory conditions.","authors":"María Elena Angarita-Planchez, Camila Leiva-Castro, Ana M Múnera-Rodríguez, Macarena Martínez-Bailén, Ana T Carmona, Soledad López-Enríquez, Francisca Palomares","doi":"10.1111/bph.70291","DOIUrl":"https://doi.org/10.1111/bph.70291","url":null,"abstract":"<p><strong>Background and purpose: </strong>Immune tolerance prevents inflammation and autoimmunity, with dendritic cells (DCs) playing a key role. Reprogramming DCs towards a tolerogenic state represents a promising therapeutic strategy. Sulforaphane (SFN) has known immunomodulatory effects, but its clinical application is limited by poor stability and bioavailability. To enhance its therapeutic potential, SFN was conjugated with mannose (SFNMan) or fucose (SFNFuc), aiming to induce a tolerogenic phenotype in human monocyte-derived DCs (moDCs) under inflammation and to explore NFATc1's involvement.</p><p><strong>Experimental approach: </strong>moDCs were exposed to inflammatory conditions and treated with SFN, SFNMan or SFNFuc. Their phenotype, cytokine profile, T cell-modulating capacity and NFATc1 signalling were evaluated.</p><p><strong>Key results: </strong>SFNMan selectively induced a tolerogenic phenotype, characterised by an increased PD-L1/CD86 ratio and IL-10 production; up-regulation of SOCS1 and IDO transcripts; and Treg expansion and reduced proliferation of cytotoxic T cell proliferation. Functional assays and confocal microscopy revealed that SFNMan, but not SFNFuc, promoted NFATc1 nuclear translocation. Pharmacological inhibition of NFATc1 with cyclosporin A (CsA) abolished these effects, confirming NFATc1 as a central mediator of SFNMan-induced immune tolerance.</p><p><strong>Conclusions and implications: </strong>Our findings identify NFATc1 as a key transcriptional switch in moDCs tolerogenic programming and highlight the carbohydrate-dependent specificity of SFN conjugates. SFNMan represents a novel carbohydrate-engineered immunomodulator capable of driving immune tolerance through NFATc1 activation. These results provide a mechanistic framework for the development of precision therapies targeting inflammatory and autoimmune diseases.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741218","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background and purpose: Pulmonary vascular remodelling is the key pathological feature of pulmonary arterial hypertension (PAH), but treatments targeting this process are lacking. Recent studies suggest that sodium-glucose cotransporter 2 (SGLT2) inhibitors, particularly empagliflozin, may improve PAH outcomes, although the underlying mechanisms remain largely unexplored.
Experimental approach: PAH models were induced in Sprague-Dawley rats with monocrotaline or SU5416-hypoxia (SU-Hx), and empagliflozin (10 mg kg-1 day-1) or saline was administered orally. At the end point, haemodynamic, electrocardiographic parameters and pulmonary vascular remodelling were evaluated to investigate effects of empagliflozin in vivo. Effects of empagliflozin in vitro, were assessed using PDGF-BB-/hypoxia-induced proliferation and migration assays on human pulmonary arterial smooth muscle cells (PASMCs). Network pharmacology, molecular docking and surface plasmon resonance (SPR) were performed to explore potential mechanism(s) of empagliflozin treatment.
Key results: Empagliflozin improved haemodynamic, electrocardiographic parameters and pulmonary vascular remodelling in monocrotaline-/SU-Hx-induced PAH models. Empagliflozin inhibited PDGF-BB/hypoxia-stimulated proliferation and migration of human PASMCs and arrested cells in the G0/G1 phase in a concentration-dependent manner. Network pharmacology, biological and SPR results suggested that empagliflozin ameliorated PAH by suppressing excessive proliferation and migration of PASMCs, partly through direct binding to TYR-740, GLY-738 and ASP-737 in the tyrosine kinase effector domain of PDGFRβ, inhibiting PDGFRβ phosphorylation and downstream signalling.
Conclusions and implications: The results highlight a novel mechanism underlying the beneficial effects of empagliflozin in PAH, through direct binding to the tyrosine kinase effector domain of PDGFRβ. This interaction inhibits PDGFRβ phosphorylation, offering new insights into therapeutic strategies for PAH.
{"title":"The sodium-glucose co-transporter 2 inhibitor, empagliflozin, attenuates pulmonary vascular remodelling by inhibiting the phosphorylation of PDGF receptor-β.","authors":"Ting-Ting Lyu, Jing-Yang Wang, Jiang-Shan Tan, Tian-Qi Li, Yu-Yuan Shu, Yanmin Yang","doi":"10.1111/bph.70222","DOIUrl":"https://doi.org/10.1111/bph.70222","url":null,"abstract":"<p><strong>Background and purpose: </strong>Pulmonary vascular remodelling is the key pathological feature of pulmonary arterial hypertension (PAH), but treatments targeting this process are lacking. Recent studies suggest that sodium-glucose cotransporter 2 (SGLT2) inhibitors, particularly empagliflozin, may improve PAH outcomes, although the underlying mechanisms remain largely unexplored.</p><p><strong>Experimental approach: </strong>PAH models were induced in Sprague-Dawley rats with monocrotaline or SU5416-hypoxia (SU-Hx), and empagliflozin (10 mg kg<sup>-1</sup> day<sup>-1</sup>) or saline was administered orally. At the end point, haemodynamic, electrocardiographic parameters and pulmonary vascular remodelling were evaluated to investigate effects of empagliflozin in vivo. Effects of empagliflozin in vitro, were assessed using PDGF-BB-/hypoxia-induced proliferation and migration assays on human pulmonary arterial smooth muscle cells (PASMCs). Network pharmacology, molecular docking and surface plasmon resonance (SPR) were performed to explore potential mechanism(s) of empagliflozin treatment.</p><p><strong>Key results: </strong>Empagliflozin improved haemodynamic, electrocardiographic parameters and pulmonary vascular remodelling in monocrotaline-/SU-Hx-induced PAH models. Empagliflozin inhibited PDGF-BB/hypoxia-stimulated proliferation and migration of human PASMCs and arrested cells in the G0/G1 phase in a concentration-dependent manner. Network pharmacology, biological and SPR results suggested that empagliflozin ameliorated PAH by suppressing excessive proliferation and migration of PASMCs, partly through direct binding to TYR-740, GLY-738 and ASP-737 in the tyrosine kinase effector domain of PDGFRβ, inhibiting PDGFRβ phosphorylation and downstream signalling.</p><p><strong>Conclusions and implications: </strong>The results highlight a novel mechanism underlying the beneficial effects of empagliflozin in PAH, through direct binding to the tyrosine kinase effector domain of PDGFRβ. This interaction inhibits PDGFRβ phosphorylation, offering new insights into therapeutic strategies for PAH.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145713372","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p><b>50</b></p><p><b>Leveraging artificial intelligence for drug repurposing: predictive modelling to identify novel anti-inflammatory and neuroprotective candidates for central nervous system disorders</b></p><p>Sheetal Thapliyal</p><p><i>Department of Pharmacology, Sardar Bhagwan Singh University</i></p><p><b>Introduction</b></p><p>Neuroinflammation plays a central role in the pathogenesis of numerous central nervous system (CNS) disorders, including Alzheimer's disease, multiple sclerosis, and Parkinson's disease. Current pharmacological strategies often fail to address the multifactorial interplay between inflammatory and neurodegenerative mechanisms, leaving a substantial unmet therapeutic need. Drug repurposing offers a rapid, cost-effective approach to identify novel therapeutic applications for existing agents. The present study aimed to apply an artificial intelligence (AI)–driven predictive modelling pipeline to systematically screen FDA-approved and investigational drugs for dual anti-inflammatory and neuroprotective potential, with a focus on blood–brain barrier (BBB) penetrance and multi-target activity. We hypothesized that a graph neural network (GNN)–based approach integrated with molecular docking and pathway enrichment analysis could reveal high-value candidates that modulate both inflammatory cascades and neurotrophic pathways.</p><p><b>Methods</b></p><p>A curated dataset of 2485 FDA-approved drugs and 1120 investigational compounds was assembled from DrugBank and ChEMBL. Known neuroprotective and anti-inflammatory drugs (n = 327) were used to train a GNN classifier using molecular graph embeddings (radius = 3, hidden layers = 256, dropout = 0.2). The model was optimized using Adam optimizer (learning rate = 1e−4) and evaluated by 5-fold cross-validation. Compounds were ranked by probability scores and subjected to BBB permeability prediction using an ensemble of support vector machine (SVM) and deep-learning models. Molecular docking was performed against TNF-α, NF-κB p65, Nrf2-Keap1, and TrkB (BDNF receptor) using AutoDock Vina, with binding energy thresholds set at ≤–9.0 kcal/mol. Pathway enrichment analysis of top candidates was conducted using Reactome. Statistical analyses included AUROC, F1-score, and bootstrapped confidence intervals.</p><p><b>Results</b></p><p>The GNN achieved an AUROC of 0.91 (95% CI, 0.89–0.93) and an F1-score of 0.87, significantly outperforming baseline random forest models (P < 0.001). Of the top 15 ranked candidates, nilvadipine (antihypertensive), pioglitazone (antidiabetic), and clemastine fumarate (antihistamine) emerged as unexpected high-probability hits. Nilvadipine was predicted to inhibit microglial activation via TNF-α suppression; pioglitazone stabilized Nrf2–Keap1 interaction, enhancing antioxidant defences; clemastine fumarate promoted oligodendrocyte differentiation and remyelination.</p><p>Pathway analysis revealed that 9 of the 15 top candidates co-modulated NF-κB-driven inflam
{"title":"Selected Abstracts from Pharmacology 2025","authors":"","doi":"10.1111/bph.70263","DOIUrl":"10.1111/bph.70263","url":null,"abstract":"<p><b>50</b></p><p><b>Leveraging artificial intelligence for drug repurposing: predictive modelling to identify novel anti-inflammatory and neuroprotective candidates for central nervous system disorders</b></p><p>Sheetal Thapliyal</p><p><i>Department of Pharmacology, Sardar Bhagwan Singh University</i></p><p><b>Introduction</b></p><p>Neuroinflammation plays a central role in the pathogenesis of numerous central nervous system (CNS) disorders, including Alzheimer's disease, multiple sclerosis, and Parkinson's disease. Current pharmacological strategies often fail to address the multifactorial interplay between inflammatory and neurodegenerative mechanisms, leaving a substantial unmet therapeutic need. Drug repurposing offers a rapid, cost-effective approach to identify novel therapeutic applications for existing agents. The present study aimed to apply an artificial intelligence (AI)–driven predictive modelling pipeline to systematically screen FDA-approved and investigational drugs for dual anti-inflammatory and neuroprotective potential, with a focus on blood–brain barrier (BBB) penetrance and multi-target activity. We hypothesized that a graph neural network (GNN)–based approach integrated with molecular docking and pathway enrichment analysis could reveal high-value candidates that modulate both inflammatory cascades and neurotrophic pathways.</p><p><b>Methods</b></p><p>A curated dataset of 2485 FDA-approved drugs and 1120 investigational compounds was assembled from DrugBank and ChEMBL. Known neuroprotective and anti-inflammatory drugs (n = 327) were used to train a GNN classifier using molecular graph embeddings (radius = 3, hidden layers = 256, dropout = 0.2). The model was optimized using Adam optimizer (learning rate = 1e−4) and evaluated by 5-fold cross-validation. Compounds were ranked by probability scores and subjected to BBB permeability prediction using an ensemble of support vector machine (SVM) and deep-learning models. Molecular docking was performed against TNF-α, NF-κB p65, Nrf2-Keap1, and TrkB (BDNF receptor) using AutoDock Vina, with binding energy thresholds set at ≤–9.0 kcal/mol. Pathway enrichment analysis of top candidates was conducted using Reactome. Statistical analyses included AUROC, F1-score, and bootstrapped confidence intervals.</p><p><b>Results</b></p><p>The GNN achieved an AUROC of 0.91 (95% CI, 0.89–0.93) and an F1-score of 0.87, significantly outperforming baseline random forest models (P < 0.001). Of the top 15 ranked candidates, nilvadipine (antihypertensive), pioglitazone (antidiabetic), and clemastine fumarate (antihistamine) emerged as unexpected high-probability hits. Nilvadipine was predicted to inhibit microglial activation via TNF-α suppression; pioglitazone stabilized Nrf2–Keap1 interaction, enhancing antioxidant defences; clemastine fumarate promoted oligodendrocyte differentiation and remyelination.</p><p>Pathway analysis revealed that 9 of the 15 top candidates co-modulated NF-κB-driven inflam","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":"183 3","pages":"644-907"},"PeriodicalIF":7.7,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://bpspubs.onlinelibrary.wiley.com/doi/epdf/10.1111/bph.70263","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145713294","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Agnieszka Michalak, Łukasz Kurach, Artur Wnorowski, Anita Kowal, Dominika Przygodzka, Vardan T Karamyan
Background and purpose: Recognition and procedural memory are modulated by the noradrenergic system, whose terminals release noradrenaline along with neuropeptide Y, acting via Y1 and Y2 receptors. This study aims to investigate the role of Y1 and Y2 receptor antagonists (BMS-193885 and SF-11) on recognition and procedural memory, including their co-administration with clenbuterol, a β2-adrenoceptor agonist, with additional focus on the molecular mechanisms underlying memory-related behavioural changes.
Experimental approach: Recognition memory was assessed in mice using the novel object recognition task. Motor learning and procedural memory were evaluated in the accelerating rotarod test. Memory-related behavioural outcomes were complemented by analyses of NPY levels, the phosphorylation status of CaMKII and ERK1/2, along with the concentrations of 5-HT, dopamine, noradrenaline and its metabolite in selected brain regions, including the prefrontal cortex, hippocampus and cerebellum.
Key results: SF-11 at 5 mg·kg-1 impaired recognition memory, prolonged procedural memory retention and inhibited CaMKII, but at 20 mg·kg-1, SF-11 disrupted both recognition and procedural memory increasing NPY. BMS-193885, at 5 mg·kg-1, prolonged procedural memory retention, but at 20 mg·kg-1, it impaired long-term recognition memory, prolonged procedural memory retention, increased NPY and decreased phospho-CaMKII. Clenbuterol enhanced procedural memory retention, but this pro-cognitive effect was diminished by co-administration with either Y receptor antagonist, which correlated with decreased noradrenaline levels.
Conclusions and implications: NPY receptor antagonists modulate memory processes in a dose- and memory-type-dependent manner, and these effects appear to be linked to hippocampal function. NPY receptors may serve as therapeutic targets to treat cognitive impairments.
{"title":"NPY Y<sub>1</sub> and Y<sub>2</sub> receptors in the regulation of mouse recognition and procedural memory: Interplay with cellular signalling, noradrenergic transmission and monoamine concentrations.","authors":"Agnieszka Michalak, Łukasz Kurach, Artur Wnorowski, Anita Kowal, Dominika Przygodzka, Vardan T Karamyan","doi":"10.1111/bph.70287","DOIUrl":"https://doi.org/10.1111/bph.70287","url":null,"abstract":"<p><strong>Background and purpose: </strong>Recognition and procedural memory are modulated by the noradrenergic system, whose terminals release noradrenaline along with neuropeptide Y, acting via Y1 and Y2 receptors. This study aims to investigate the role of Y1 and Y2 receptor antagonists (BMS-193885 and SF-11) on recognition and procedural memory, including their co-administration with clenbuterol, a β<sub>2</sub>-adrenoceptor agonist, with additional focus on the molecular mechanisms underlying memory-related behavioural changes.</p><p><strong>Experimental approach: </strong>Recognition memory was assessed in mice using the novel object recognition task. Motor learning and procedural memory were evaluated in the accelerating rotarod test. Memory-related behavioural outcomes were complemented by analyses of NPY levels, the phosphorylation status of CaMKII and ERK1/2, along with the concentrations of 5-HT, dopamine, noradrenaline and its metabolite in selected brain regions, including the prefrontal cortex, hippocampus and cerebellum.</p><p><strong>Key results: </strong>SF-11 at 5 mg·kg<sup>-1</sup> impaired recognition memory, prolonged procedural memory retention and inhibited CaMKII, but at 20 mg·kg<sup>-1</sup>, SF-11 disrupted both recognition and procedural memory increasing NPY. BMS-193885, at 5 mg·kg<sup>-1</sup>, prolonged procedural memory retention, but at 20 mg·kg<sup>-1</sup>, it impaired long-term recognition memory, prolonged procedural memory retention, increased NPY and decreased phospho-CaMKII. Clenbuterol enhanced procedural memory retention, but this pro-cognitive effect was diminished by co-administration with either Y receptor antagonist, which correlated with decreased noradrenaline levels.</p><p><strong>Conclusions and implications: </strong>NPY receptor antagonists modulate memory processes in a dose- and memory-type-dependent manner, and these effects appear to be linked to hippocampal function. NPY receptors may serve as therapeutic targets to treat cognitive impairments.</p>","PeriodicalId":9262,"journal":{"name":"British Journal of Pharmacology","volume":" ","pages":""},"PeriodicalIF":7.7,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145707381","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}