51
|
Chen L, Bai D, Du J, Zhao J, Zhou C, Gu C, Wang Y, Zhang L, Lu N, Zhao Y. GL-V9 inhibits Caspase-11 activation-induced pyroptosis by suppressing ALOX12-mediated lipid peroxidation to alleviate sepsis. Br J Pharmacol 2025. [PMID: 40233936 DOI: 10.1111/bph.70053] [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: 08/31/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND AND PURPOSE Sepsis, caused by pathogen infection, poses a serious threat to human life. While the link between sepsis and pyroptosis via Caspase-11 non-canonical inflammasome activation is known, effective treatments remain lacking. Previous studies have confirmed that GL-V9 has antifibrotic and antitumor activities, but whether it has a therapeutic effect on sepsis is unclear. The aim of this study was to investigate the anti-inflammatory activity of GL-V9 and its possible mechanism. EXPERIMENTAL APPROACH The caecal ligation and puncture (CLP) model was used to assess the antiseptic effects of GL-V9 in vivo. Mouse bone marrow derived macrophages (BMDMs) and murine macrophages line J774A.1 also served as an in vitro Caspase-11 activation induced pyroptosis model. Cellular functions and molecular mechanism were analysed using cell viability assay, PI uptake assay, western blotting, immunofluorescence and co-immunoprecipitation. KEY RESULTS GL-V9 reduced tissue damage and mortality in mice with sepsis, and decreased the secretion of inflammatory factors in vivo. In vitro, GL-V9 suppressed Caspase-11-induced pyroptosis and prevented the release of LPS from early endosomes. Mechanistic studies revealed that GL-V9 limits Caspase-11 activation by inhibiting ALOX12-mediated lipid peroxidation. Further studies confirmed that GL-V9 did not further alleviate the symptoms and inflammatory response of septic mice in Alox12 deficient mice. CONCLUSION AND IMPLICATIONS GL-V9 exerts a powerful anti-sepsis effect in vivo, which is associated with the inhibition of Caspase-11 activation. Mechanistically, GL-V9 may block LPS release from early endosomes by inhibiting ALOX12-mediated lipid peroxidation. This suggests that GL-V9 is a potential candidate for the treatment of sepsis.
Collapse
Affiliation(s)
- Li Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Dongsheng Bai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jiaying Du
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jiawei Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chen Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chunyang Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yuxiang Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Lulu Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
52
|
Martens N, Zhan N, Yam SC, Palumbo M, Pontini L, Leijten FPJ, van Vark-van der Zee L, Voortman G, Friedrichs S, Gerding A, Marinozzi M, Jonker JW, Kuipers F, Lütjohann D, Vanmierlo T, Mulder MT. Role for the liver X receptor agonist 22-ketositosterol in preventing disease progression in an Alzheimer's disease mouse model. Br J Pharmacol 2025. [PMID: 40233928 DOI: 10.1111/bph.70031] [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: 05/29/2024] [Revised: 01/29/2025] [Accepted: 02/09/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND AND PURPOSE Liver X receptors (LXRs) are promising therapeutic targets for alleviating Alzheimer's disease (AD) symptoms. We assessed the impact of the semi-synthetic LXR agonist 22-ketositosterol on disease progression in an AD mouse model. EXPERIMENTAL APPROACH From 5.5 months of age, APPswePS1ΔE9 (AD) mice and wild-type (WT) littermates received a regular or 22-ketositosterol-supplemented diet (0.017% w/w). Cognition was assessed with object location and recognition tasks and a spontaneous alternation Y-maze test. Amyloid β was quantified using immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA), microglia (Iba1, CD68) and astrocyte (GFAP) markers using IHC. Sterols were determined in food, serum, liver and cerebellum. KEY RESULTS 22-Ketositosterol activated both liver X receptors-α and -β and promoted cholesterol efflux in cell cultures. Diet supplementation with 22-ketositosterol prevented a decline in the performance of APPswePS1ΔE9 mice in the object location task but not in the other two tasks. Without affecting amyloid β deposition, 22-ketositosterol decreased microglia (Iba1, CD68) and astrocyte (GFAP) markers in the cortex and hippocampus of APPswePS1ΔE9, suggesting potential anti-inflammatory effects. No lipid accumulation was detected in the liver or serum upon 22-ketositosterol supplementation. CONCLUSIONS AND IMPLICATIONS Diet supplementation with 22-ketositosterol prevented the decline in spatial memory of APPswePS1ΔE9 mice. Our data suggest therapeutic benefits of 22-ketositosterol possibly by enhancing cholesterol efflux and mitigating inflammatory responses, without inducing hepatosteatosis or hypertriglyceridemia.
Collapse
Affiliation(s)
- Nikita Martens
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Na Zhan
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Sammie C Yam
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Lorenzo Pontini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Frank P J Leijten
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Leonie van Vark-van der Zee
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gardi Voortman
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Silvia Friedrichs
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Albert Gerding
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maura Marinozzi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Johan W Jonker
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Tim Vanmierlo
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute, Maastricht University, Maastricht, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
53
|
Xuan Y, Gao X, Wang J, Li H, Zhou Z, Liao M, Wen Z, Wang DW. Hydroxychloroquine cures autoimmune myocarditis by inhibiting the innate immune system via the C-X-C motif chemokine ligand 16 and C-X-C motif receptor 6 axis between macrophages and T cells. Br J Pharmacol 2025. [PMID: 40222951 DOI: 10.1111/bph.70028] [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: 01/07/2024] [Revised: 12/31/2024] [Accepted: 03/02/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Myocarditis is a life-threatening inflammatory disease, but lacks effective treatment options. Hydroxychloroquine (HCQ), an established antimalarial agent, is used widely to manage rheumatic disorders. This research aimed to evaluate the efficacy of HCQ in treating myocarditis. EXPERIMENTAL APPROACH A mouse model of experimental autoimmune myocarditis (EAM) was used to evaluate the therapeutic effects of HCQ on cardiac function, inflammation and fibrosis. Echocardiography, histology and cytokine assays were performed to assess cardiac function and inflammatory responses. Single-cell RNA sequencing was employed to analyse immune cell populations and chemotactic activity. C-X-C motif chemokine ligand 16 (CXCL16) levels were measured in cardiac tissue and serum, while YY1 expression was measured by western blotting in macrophages and cardiac tissue. Flow cytometry was used to evaluate immune cell infiltration and migration. KEY RESULTS HCQ improved cardiac function in acute and chronic myocarditis. HCQ treatment reduced inflammation, fibrosis and immune cell infiltration in myocarditis models. Single-cell RNA sequencing revealed that HCQ lowered inflammatory cell proportions and suppressed macrophage chemotaxis. HCQ reduced YY1 levels, leading to the down-regulation of CXCL16 expression in macrophages and inhibition of CXCL16-mediated chemotaxis to Th17 and natural killer T (NKT) cells. CXCL16 neutralizing antibodies improved cardiac function and reduced inflammation in myocarditis. CONCLUSION AND IMPLICATIONS HCQ improves cardiac function and reduces inflammation in myocarditis by inhibiting CXCL16 expression in macrophages, by suppressing its transcription factor YY1, which in turn reduced the chemotaxis of Th17 and NKT cells. HCQ is a promising therapeutic agent for myocarditis.
Collapse
Affiliation(s)
- Yunling Xuan
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Gao
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Wang
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huihui Li
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Zhou
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minyu Liao
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Wen
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
54
|
Mu SY, Xu R, Wu XF, Cheng YY, Sun ZM, Liu HT, Shao HB, Zhang XN, Zhang XN, Yang M, Tan MY, Liang WS, Wan SB, Cui SX, Qu XJ. Inhibition of sphingosine-1-phosphate receptor-2 attenuates idiopathic pulmonary fibrosis by preventing its binding to dapper1 in bronchial epithelial cells. Br J Pharmacol 2025. [PMID: 40222913 DOI: 10.1111/bph.70043] [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: 10/17/2024] [Revised: 01/20/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Activation of the sphingosine-1-phosphate receptor-2 (S1P2 receptor) promotes idiopathic pulmonary fibrosis (IPF). However, the mechanisms associated with IPF development via S1P2 receptor signalling are poorly understood and no S1P2 receptor antagonists have been approved for clinical use. EXPERIMENTAL APPROACH Western blotting and immunohistochemical assays analysed inflammatory factors and epithelial-mesenchymal transition (EMT) markers. Co-immunoprecipitation and immunofluorescence analysed the binding of S1P2 receptor to dapper1 (Dpr1) and cyclic AMP response-binding protein 1 (CREB1). X-ray-based computed tomography diagnosed IPF in bleomycin (BLM)-treated mice. Barometric whole-body plethysmography tested pulmonary function of mice. Masson's trichrome and Sirius red staining analysed extracellular matrix deposition. Enzyme-linked immunosorbent assays analysed inflammatory factors and hydroxyproline. KEY RESULTS Activation of S1P2 receptors promoted IPF through the binding of S1P2 receptor to Dpr1, decreasing dishevelled (Dvl) degradation to accumulate β-catenin. The β-catenin accumulated in the nucleus, upregulating its target genes by binding to T-cell factor/lymphoid enhancer factor. The binding of S1P2 receptor to Dpr1 also led to S1P2 receptor translocation to the nucleus, where it promoted EMT by activating CREB1. BLM-induced IPF in mice was characterised by activated-S1P2 receptor signalling. Inhibition of S1P2 receptor prevented the binding of S1P2 receptor to Dpr1, resulting in decreased β-catenin accumulation and blocking nuclear translocation of S1P2 receptor. The S1P2 receptor antagonist S118 was more effective than pirfenidone in attenuating IPF through anti-inflammatory, anti-fibrosis, and anti-EMT effects. CONCLUSIONS AND IMPLICATIONS Activation of S1P2 receptors promotes IPF through the binding of S1P2 receptor to Dpr1 and the nuclear translocation of S1P2 receptor to activate CREB1. Thus, the S1P2 receptor antagonist S118 has potential clinical application in attenuating IPF.
Collapse
Affiliation(s)
- Si-Yuan Mu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Rui Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xin-Feng Wu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yu-Yao Cheng
- Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Zhi-Meng Sun
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han-Tao Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han-Bing Shao
- Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Xiao-Nan Zhang
- Department of Pharmacology, Marine Biomedical Research Institute of Qingdao, Qingdao, China
| | - Xi-Nan Zhang
- Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Ming Yang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ming-Yong Tan
- Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Wei-Shi Liang
- Joint Laboratory for Research and Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Sheng-Biao Wan
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shu-Xiang Cui
- Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Xian-Jun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
55
|
Olivencia MA, Climent B, Barreira B, Morales-Cano D, Sánchez A, Fernández A, García-Gómez B, Romero-Otero J, Rodríguez C, Moreno L, Prieto D, Larriba MJ, Cogolludo A, Angulo J, Perez-Vizcaino F. Vitamin D deficiency induces erectile dysfunction: Role of superoxide and Slpi. Br J Pharmacol 2025. [PMID: 40222751 DOI: 10.1111/bph.70034] [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: 09/11/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Epidemiological studies suggest a relationship between vitamin D deficiency and erectile dysfunction (ED). We hypothesized that vitamin D deficiency or vitamin D receptor (VDR) knockout causes ED and analysed the underlying molecular mechanisms. EXPERIMENTAL APPROACH Erectile function was assessed in vivo in anaesthetized male mice or rats by evaluating intracavernosal pressure (ICP) and in vitro in male Vdr-/- mice, and rat or human isolated corpora cavernosa (CCs) mounted in a myograph. Bulk RNA-sequencing (RNA-seq) transcriptomic analysis was performed in rat CCs. Vitamin D deficiency was induced in rats fed a vitamin D-free diet for 5 months. KEY RESULTS CCs from human donors with low plasma vitamin D exhibited reduced nitric oxide (NO)-dependent erectile function. This ED was also reproduced in vitamin D-deficient rats and VDR knockout mice, in vivo and ex vivo, and is associated with penile fibrosis and reduced response to the phosphodiesterase 5 inhibitor (PDE5i) sildenafil. CCs from deficient rats show increased superoxide levels, and their impaired erectile function was restored by superoxide scavengers. Transcriptomic analysis, real-time polymerase chain reaction (RT-PCR) and Western blot showed down-regulated secretory leukocyte protease inhibitor (Slpi). Moreover, recombinant SLPI prevented superoxide-induced ED, while Slpi gene silencing led to reduced erectile function in a superoxide-dependent manner. CONCLUSION AND IMPLICATIONS Vitamin D deficiency or VDR knockout reduces erectile function. We suggest that this effect is mediated by increased superoxide levels and down-regulation of SLPI. Vitamin D deficiency might be an aetiological factor for vascular ED and for the therapeutic failure of PDE5i.
Collapse
Affiliation(s)
- Miguel A Olivencia
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Belén Climent
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Department of Physiology, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Daniel Morales-Cano
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Ana Sánchez
- Department of Physiology, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Argentina Fernández
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Borja García-Gómez
- Servicio de Urología, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Javier Romero-Otero
- Servicio de Urología, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Claudia Rodríguez
- Department of Physiology, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Dolores Prieto
- Department of Physiology, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - María Jesús Larriba
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Javier Angulo
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
56
|
Chen W, Jin T, Xie Y, Zhong C, Gao H, Zhang L, Ju J, Cheng T, Li M, Wang H, Yang Z, Deng Q, Du Z, Liang H. Berberine partially ameliorates cardiolipotoxicity in diabetic cardiomyopathy by modulating SIRT3-mediated lipophagy to remodel lipid droplets homeostasis. Br J Pharmacol 2025. [PMID: 40222752 DOI: 10.1111/bph.70035] [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: 12/11/2023] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Emerging evidence indicated that the excessive lipid droplets (LDs) accumulation and lipotoxicity play a significant role in the development of diabetic cardiomyopathy (DCM), yet the regulatory mechanisms governing the function of cardiac LDs are still unknown. Lipophagy has been shown to be involved in the maintenance of LDs homeostasis. The objective of this study was to explore the mechanism of lipophagy in cardiomyocytes and investigate whether berberine could mitigate DCM by modulating this pathway. EXPERIMENTAL APPROACH Bioinformatics analysis identified disorders of lipid metabolism and autophagy in DCM. To carry out further research, db/db mice were utilized. Furthermore, H9C2 cells treated with palmitic acid were employed as a model to explore the molecular mechanisms involved in myocardial lipotoxicity. KEY RESULTS The results showed that lipophagy was impaired in DCM. Mechanistically, sirtuin 3 (SIRT3) was demonstrated to regulate lipophagy in cardiomyocytes. SIRT3 was down-regulated in DCM. Conversely, activation of SIRT3 by the activator nicotinamide riboside (NR) could promote lipophagy to alleviate PA-induced lipotoxicity in H9C2 cells. Moreover, berberine administration markedly mitigated diabetes-induced cardiac dysfunction and hypertrophy in db/db mice, which dependent on SIRT3-mediated lipophagy. CONCLUSION AND IMPLICATIONS Collectively, SIRT3 could moderate cardiac lipotoxicity in DCM by promoting lipophagy, suggesting that the regulation of SIRT3-mediated lipophagy may be a promising strategy for treating DCM. The findings indicate that the therapeutic potential of berberine for DCM is associated with lipophagy.
Collapse
Affiliation(s)
- Wenxian Chen
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University, Zhuhai, China
- School of Pharmacy, Health Science Center, Shenzhen University, Shen Zhen, China
| | - Tongzhu Jin
- Department of Pharmacy at the Second Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yilin Xie
- School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Changsheng Zhong
- School of Pharmacy, Health Science Center, Shenzhen University, Shen Zhen, China
| | - Huiying Gao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lei Zhang
- School of Pharmacy, Health Science Center, Shenzhen University, Shen Zhen, China
| | - Jin Ju
- School of Pharmacy, Health Science Center, Shenzhen University, Shen Zhen, China
| | - Ting Cheng
- School of Pharmacy, Health Science Center, Shenzhen University, Shen Zhen, China
| | - Mengyang Li
- School of Pharmacy, Health Science Center, Shenzhen University, Shen Zhen, China
| | - Huifang Wang
- School of Pharmacy, Health Science Center, Shenzhen University, Shen Zhen, China
| | - Zhenbo Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Qin Deng
- School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhimin Du
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University, Zhuhai, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Haihai Liang
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Clinical Medical College of Jinan University, Zhuhai, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| |
Collapse
|
57
|
Pantke S, Steinberg JH, Weber LKH, Fricke TC, Carvalheira Arnaut Pombeiro Stein I, Oprita G, Herzog C, Leffler A. High Concentrations of the Antidepressant Amitriptyline Activate and Desensitize the Capsaicin Receptor TRPV1. Pharmaceuticals (Basel) 2025; 18:560. [PMID: 40283995 PMCID: PMC12030456 DOI: 10.3390/ph18040560] [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: 03/12/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background: A large number of patients suffer from neuropathic pain, and systemic therapy often remains ineffective while inducing severe side effects. Topical therapy with the TRPV1-agonist capsaicin is an established alternative, and the identification of co-therapeutics that modulate TRPV1 may be a promising approach to reduce the dose of capsaicin while maintaining efficacy. Here, we aimed to determine if the antidepressant amitriptyline displays properties rendering it a potential co-therapeutic agent. Methods: We performed patch clamp and calcium imaging experiments on HEK293T cells expressing human (h) TRPV1 as well as on dorsal root ganglion (DRG) neurons from adult mice. Results: Amitriptyline induced an increase in intracellular calcium in both HEK293T and mouse DRG neurons expressing TRPV1. Patch clamp experiments revealed a concentration-dependent activation of hTRPV1 by amitriptyline that was also evident in cell-free inside-out patches. When hTRPV1 was fully activated by capsaicin, amitriptyline induced concentration-dependent and partly reversible current inhibition. In contrast, amitriptyline potentiated small responses to capsaicin, heat and protons. We also found that amitriptyline desensitized hTRPV1 to capsaicin. This effect was reduced by the intracellular application of the strong calcium chelator BAPTA. Furthermore, the non-desensitizing mutant hTRPV1-Y672K displayed a reduced amitriptyline-induced desensitization. Conclusions: Our data showed that amitriptyline can activate, sensitize, desensitize and even inhibit TRPV1. Together with its property as a strong local anesthetic, our data suggest that amitriptyline may be a promising adjunct to topical capsaicin.
Collapse
Affiliation(s)
- Sebastian Pantke
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
- PRACTIS Clinician Scientist Program, Dean’s Office for Academic Career Development, Hannover Medical School, 30625 Hannover, Germany
| | - Johanna H. Steinberg
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
| | - Lucas K. H. Weber
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
| | - Tabea C. Fricke
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
- PRACTIS Clinician Scientist Program, Dean’s Office for Academic Career Development, Hannover Medical School, 30625 Hannover, Germany
| | - Inês Carvalheira Arnaut Pombeiro Stein
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
- PRACTIS Clinician Scientist Program, Dean’s Office for Academic Career Development, Hannover Medical School, 30625 Hannover, Germany
| | - George Oprita
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
| | - Christine Herzog
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.P.); (J.H.S.); (L.K.H.W.); (I.C.A.P.S.); (G.O.); (C.H.)
| |
Collapse
|
58
|
Fan BQ, Xia JM, Yi XW, Yuan TJ, Zhou Y, Xu R, Wang L, Wang D, Xia Y, Yi WJ, Ding JH, Qu WM, Li WX, Huang ZL, Han Y. Ventral pallidum GABAergic and glutamatergic neurons modulate arousal during sevoflurane general anaesthesia in male mice. Br J Pharmacol 2025. [PMID: 40205920 DOI: 10.1111/bph.70019] [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: 09/15/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The induction and emergence of general anaesthesia involve an altered process of states of consciousness, yet the central nervous system mechanisms remain inadequately understood. The ventral pallidum (VP) within the basal ganglia is crucial in sleep-wake modulation. However, its involvement in general anaesthesia and the underlying neuronal mechanisms are not well elucidated. EXPERIMENTAL APPROACH In vivo electrophysiological recordings were conducted to examine changes in the activity of different types of VP neurons before and after sevoflurane exposure. Fibre photometry, combined with electroencephalogram and electromyography recordings, was employed to analyse neuronal activity during both the induction and recovery phases of sevoflurane anaesthesia. Chemogenetics was implemented to investigate the impact of modulated neuronal activity on anaesthesia induction and emergence, whereas optogenetics was used for real time activation of neurons at different depths of anaesthesia. KEY RESULTS Sevoflurane exposure reduced the firing activity of both VP GABAergic (VPGABA) and VP glutamatergic (VPglu) neurons, without affecting cholinergic neurons. VPGABA and VPglu neuronal activity decreased during sevoflurane anaesthesia induction and increased during emergence. Manipulation of VPGABA neurons bidirectionally influenced the duration of induction and emergence. Inhibiting VPglu neurons accelerated induction. Real time activation of VPGABA neurons triggered cortical activation and behavioural emergence during steady-state sevoflurane anaesthesia and reduced the burst suppression ratio during deep anaesthesia. CONCLUSION AND IMPLICATIONS These findings highlight the role of VPGABA and VPglu neurons in modulating transitions between anaesthesia stages, providing valuable insights into the neuronal mechanisms underlying sevoflurane-induced anaesthesia.
Collapse
Affiliation(s)
- Bing-Qian Fan
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology and Perioperative Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun-Ming Xia
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Xiu-Wen Yi
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Tian-Jie Yuan
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Yu Zhou
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Rui Xu
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Lu Wang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
| | - Di Wang
- Department of Anaesthesiology and Perioperative Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Xia
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Wen-Jing Yi
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Jia-Hui Ding
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Xian Li
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yuan Han
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| |
Collapse
|
59
|
Asano S, Ozasa K, Uehara T, Yokoyama R, Nakazawa T, Yanamoto S, Ago Y. Dimerisation of the VIP receptor VIPR2 is essential to its binding VIP and Gα i proteins, and to its functions in breast cancer cells. Br J Pharmacol 2025. [PMID: 40203889 DOI: 10.1111/bph.70039] [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: 09/24/2024] [Revised: 02/02/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Several G protein-coupled receptors (GPCRs) are known to homodimerise. Dimeric GPCRs may have different properties from their monomers, but the molecular basis and functional significance of GPCR dimerisation remain largely unknown. We recently found that signalling by the vasoactive intestinal peptide receptor, VIPR2, regulates breast cancer cell migration and proliferation. However, it is unclear whether VIPR2 monomers directly interact with each other and what function the dimeric receptor has. Here, we showed that VIPR2 dimerises and investigated their role in breast cancer progression. EXPERIMENTAL APPROACH Dimerisation of VIPR2 was assessed by fluorescence resonance energy transfer (FRET) analysis and a pull-down assay. Breast cancer progression was analysed by orthotopic growth and metastasis of human breast cancers into proper axillary and subiliac lymph-nodes in mice. KEY RESULTS VIPR2 monomers directly interacted with each other through transmembrane domains (TM)3-4. FRET analysis revealed that VIPR2 moved further apart in cells expressing TM3-4-peptides, suggesting that TM3-4 prevents VIPR2 dimerisation. Breast cancer cells stably expressing TM3-4 region exhibited suppressed tumour growth and lymph-node metastasis. Furthermore, ligand-receptor binding assays revealed that VIP-FITC bound to cells dose-dependently, and VIPR2 de-dimerisation by TM3-4 expression decreased VIP's affinity to cells. Additionally, TM3-4 expression decreased Gαi-VIPR2 interactions. CONCLUSION AND IMPLICATIONS Dimeric VIPR2 forms the minimal functional unit that effectively promotes growth and metastasis of breast cancer. Therefore, dimeric VIPR2 is a potential therapeutic target for breast cancer, and TM3-4-peptides are potential anti-cancer drug candidates to suppress cancer progression.
Collapse
Affiliation(s)
- Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| | - Kairi Ozasa
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| | - Teru Uehara
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rei Yokoyama
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| | - Takanobu Nakazawa
- Department of Bioscience, Graduate School of Life Sciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Souichi Yanamoto
- School of Dentistry, Hiroshima University, Hiroshima, Japan
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
60
|
Bose S, Saha P, Alam MT, Chatterjee B, Sarkar M, Dixit AK, Kumar D, Pathak RK, Tripathi PP, Srivastava AK. Inhibition of DNA polymerase eta-mediated translesion DNA synthesis with small molecule sensitises ovarian cancer stem-like cells to chemotherapy. Br J Pharmacol 2025. [PMID: 40194519 DOI: 10.1111/bph.70037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/07/2025] [Accepted: 02/26/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND AND PURPOSE Chemoresistance and tumour relapse pose significant challenges in achieving successful chemotherapy outcomes. Targeting DNA polymerase eta (Pol ƞ)-mediated mutagenic translesion DNA synthesis (TLS) has emerged as a promising strategy for improving chemotherapy. However, the identification of small molecule inhibitors targeting Pol ƞ -mediated TLS with high in vivo efficacy remains a challenge. EXPERIMENTAL APPROACH The small molecule was identified through in silico screening. Pol η inhibitory potential of the identified small molecule was validated by a fluorescent-based reporter strand displacement assay. Flow cytometry was conducted to analyse the CD44 + CD117 + cancer stem-like cell (CSC) population and live-dead cell population. Xenograft mouse models were used to test the CSC sensitising potential. KEY RESULTS We screened and identified chrysin as a small-molecule inhibitor that sensitises ovarian cancer stem-like cells to cisplatin treatment by inhibiting Pol ƞ -mediated TLS. Chrysin effectively inhibits Pol ƞ expression, mitigates cancer stem-like cell enrichment and enhances cisplatin-induced cell death both in vitro and in vivo. Furthermore, chrysin treatment reduces spontaneous and cisplatin-induced mutagenesis. Pre-treatment with chrysin attenuates cisplatin-induced haematological toxicity and suppresses tumour growth in human ovarian cancer xenografts. CONCLUSIONS AND IMPLICATIONS These results establish chrysin as a novel class of TLS inhibitors and highlight its potential as a chemotherapy adjuvant for overcoming chemoresistance and improving treatment outcomes in ovarian cancer.
Collapse
Affiliation(s)
- Subhankar Bose
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priyanka Saha
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Md Tanjim Alam
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Bilash Chatterjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Mrinmoy Sarkar
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Amit Kumar Dixit
- Department of Biochemistry, Central Ayurveda Research Institute, Kolkata, India
| | - Deepak Kumar
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Rakesh Kumar Pathak
- Department of Chemical Science, Indian Institute of Science Education and Research Berhampur, Odisha, India
| | - Prem Prakash Tripathi
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Amit Kumar Srivastava
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
61
|
Li J, Zhang J, Zou L, Cao M, Zhu Z, Yu L, Zhou M, Fu W, Dong Z, Gao H, Jiang B. Fibrin induces infiltration of macrophages and neutrophils via integrin αMβ2 and triggers aortic dissection. Br J Pharmacol 2025. [PMID: 40194543 DOI: 10.1111/bph.70033] [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: 09/04/2024] [Revised: 02/18/2025] [Accepted: 03/02/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND AND PURPOSE Infiltration of macrophages and neutrophils plays a crucial role in the occurrence of aortic dissection (AD), while the mechanism elucidating their infiltration remains unknown. The present study aimed to delineate the underlying mechanism and provide a potential therapeutic strategy to attenuate AD progression. EXPERIMENTAL APPROACH A model of AD was established in male mice using β-aminopropionitrile and angiotensin II. Proteomic analysis, histological evaluation, flow cytometry, western blot, multiple fluorescence staining and adhesion assays were used to evaluate fibrin and inflammatory cells during AD progression. Fibrinogen-lowering drugs and fibrinogen γ-chain knockout (Fgg+/-) mice were also used to evaluate the fibrin-integrin αMβ2 interaction. KEY RESULTS Fibrin deposition was confirmed by proteomic analysis and histological staining, accompanied by infiltration of macrophages and neutrophils detected by flow cytometry during the progression of AD. After confirming that macrophages and neutrophils infiltrated at the sites where fibrin was deposited by immunofluorescence, an association between fibrin and the integrin αMβ2 was disclosed using protein-protein interaction analysis and immunofluorescence. The pivotal role of interactions between fibrin and integrin αMβ2 in AD progression was confirmed by cell adhesion in vitro, down-regulation of fibrin using batroxobin and Fgg+/- mice in vivo. The relevance of fibrin and integrin αMβ2 was also found in patients with AD. CONCLUSION AND IMPLICATIONS Fibrin plays a crucial role in triggering AD through recruiting macrophages and neutrophils via integrin αMβ2. Regulation of fibrin deposition or inhibition of the interaction between fibrin and integrin αMβ2 provide a potential therapy against AD.
Collapse
Affiliation(s)
- Jie Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Jixiu Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shenyang Pharmaceutical University, Shenyang, China
| | - Lingwei Zou
- Departments of Vascular Surgery of Zhongshan Hospital & Institute of Vascular Surgery, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Meifang Cao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhonghui Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Li Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Min Zhou
- Departments of Vascular Surgery of Zhongshan Hospital & Institute of Vascular Surgery, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Weiguo Fu
- Departments of Vascular Surgery of Zhongshan Hospital & Institute of Vascular Surgery, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zhihui Dong
- Departments of Vascular Surgery of Zhongshan Hospital & Institute of Vascular Surgery, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Huiyuan Gao
- Shenyang Pharmaceutical University, Shenyang, China
| | - Baohong Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
62
|
Clunas H, Walpole S, Babic I, Nair M, May N, Huang XF, Solowij N, Newell KA, Weston-Green K. Improved recognition memory and reduced inflammation following β-caryophyllene treatment in the Wistar-Kyoto rodent model of treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111312. [PMID: 40049345 DOI: 10.1016/j.pnpbp.2025.111312] [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: 10/04/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/12/2025]
Abstract
Persistent low mood, anxiety and cognitive deficits are common symptoms of depression and highly efficacious treatments that address symptoms including cognitive dysfunction are still required. β-caryophyllene (BCP) is a terpene with anti-inflammatory and pro-cognitive properties; however, its efficacy on cognition in depression remains unclear. This study aimed to investigate acute and chronic BCP treatment effects on cognitive, depressive- and anxiety-like behaviours, and inflammation in male and female Wistar-Kyoto (WKY) rats, a rodent model of treatment-resistant depression. Rats were administered either BCP (50 mg/kg) or vehicle (control). Open field (OFT), social interaction, sucrose preference, novel object recognition (NOR) and elevated plus maze (EPM) tests were conducted after acute (1 h) and chronic (2 weeks) treatment. Peripheral plasma inflammatory cytokine levels were examined. BCP acutely increased locomotor activity in the OFT but did not improve social interaction, whereas chronic BCP prevented increased latency to first interaction in females (not males). BCP did not improve sucrose preference or prevent anxiety-like behaviours in the EPM. BCP significantly increased novel object discrimination in the NOR test in male and female WKY rats and reduced cytokine levels after chronic treatment. This study shows for the first time that chronic BCP treatment improved recognition memory and exerted anti-inflammatory properties in a rodent model of depressive-like behaviours. BCP did not significantly improve anxiety-like behaviours, social interaction or anhedonia in WKY rats of either sex. These findings demonstrate the pro-cognitive effects of BCP in a rodent model of treatment-resistant depression worthy of further investigation.
Collapse
Affiliation(s)
- Helen Clunas
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia
| | - Samara Walpole
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Ilijana Babic
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Mayank Nair
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Naomi May
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Xu-Feng Huang
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia
| | - Nadia Solowij
- Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia; School of Psychology, Faculty of the Arts, Social Sciences and Humanities, University of Wollongong, NSW 2522, Australia
| | - Kelly A Newell
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Katrina Weston-Green
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
63
|
Haridevamuthu B, Bharti AK, Nayak SPRR, Narayanan D, Loganathan Sumathi D, Chagaleti BK, Saravanan V, Rajagopal R, Alfarhan A, Muthu Kumaradoss K, Arockiaraj J. Hydroxyl chalcone derivative DK02 as a multi-target-directed ligand for Alzheimer's disease: A preclinical study in zebrafish. Br J Pharmacol 2025; 182:1582-1598. [PMID: 39710579 DOI: 10.1111/bph.17426] [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: 08/24/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is a widespread neurodegenerative condition characterized by amyloid-beta (Aβ) plaques and tau protein aggregates, leading to significant cognitive decline. Existing treatments primarily offer symptomatic relief, underscoring the urgent need for novel therapies that address multiple AD pathways. This study evaluates the efficacy of DK02, a hydroxyl chalcone derivative, in a scopolamine-induced dementia model in zebrafish, hypothesizing that it targets several neurodegenerative mechanisms simultaneously. EXPERIMENTAL APPROACH We employed a blend of experiments, including in silico docking, in vitro enzyme inhibition assays and in vivo zebrafish models, to assess therapeutic effects of DK02. Methods included molecular docking to forecast interaction sites, acetylcholinesterase (AChE) inhibition testing, and various behavioural and histopathological analyses to gauge DK02's cognitive and neuroprotective impacts. KEY RESULTS DK02 emerged as a potent AChE inhibitor via virtual screening, and significantly enhanced cognitive functions in zebrafish, by improving memory retention and reducing anxiety-like behaviours. DK02 also displayed strong antioxidant properties, reducing oxidative stress-induced neuronal damage. Histopathological analysis confirmed its neuroprotective effects by showing decreased amyloid plaque burden and mitigated structural brain damage. CONCLUSION AND IMPLICATIONS DK02 shows promise as a multi-target-directed ligand for AD, offering a new therapeutic path by simultaneously addressing cholinergic, oxidative and amyloid pathways. Its potential to enhance cognitive functions and curtail neurodegeneration suggests advantages over current symptomatic treatments. Further research into DK02 mechanisms and long-term impacts is essential for its development in AD therapy.
Collapse
Affiliation(s)
- Balasubramanian Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ankit Kumar Bharti
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Santosh Pushpa Ramya Ranjan Nayak
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Dhaareeshwar Narayanan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Dhivya Loganathan Sumathi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Dr. M.G.R. Educational and Research Institute, Chennai, Tamil Nadu, India
| | - Bharath Kumar Chagaleti
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Venkatesan Saravanan
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Alfarhan
- Department of Botany and Microbiology, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Kathiravan Muthu Kumaradoss
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
64
|
Jiao B, An C, Du H, Tran M, Yang D, Zhao Y, Wang P, Hu Z, Zhou D, Wang Y. Genetic deficiency or pharmacological inhibition of cGAS-STING signalling suppresses kidney inflammation and fibrosis. Br J Pharmacol 2025; 182:1741-1762. [PMID: 39833988 DOI: 10.1111/bph.17412] [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: 10/24/2023] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Chronic kidney disease (CKD) is characterised by inflammation, which can lead to tubular atrophy and fibrosis. The molecular mechanisms are not well understood. In this study, we investigated the functional role of the cyclic GMP-AMP synthase (cGAS)- stimulator of interferon genes (STING) signalling in renal inflammation and fibrosis. EXPERIMENTAL APPROACH Mice with global cGAS deficiency or global or myeloid cell-specific STING deficiency or wild-type mice treated with RU.521, a selective cGAS inhibitor, were used to examine the role of cGAS-STING signalling in renal inflammation and fibrosis in a preclinical model of obstructive nephropathy in vivo. Bone marrow-derived macrophages were used to determine whether tubular epithelial cell-derived DNA can activate cGAS-STING signalling in vitro. KEY RESULTS Following obstructive injury, cGAS-STING signalling was activated in the kidneys during the development of renal fibrosis. Mice with deficiency of cGAS or STING exhibited significantly less macrophage proinflammatory activation, myofibroblast formation, total collagen deposition, and extracellular matrix (ECM) protein production in the kidneys following obstructive injury. Pharmacological inhibition of cGAS with RU.521 reduced macrophage proinflammatory activation, suppressed myofibroblast formation, and attenuated kidney fibrosis following obstructive injury. Mechanistically, cGAS-STING signalling in macrophages is activated by double-stranded DNA released from damaged tubular epithelial cells, which induces inflammatory responses. CONCLUSIONS AND IMPLICATIONS Our study identifies the cGAS-STING signalling pathway as a critical regulator of macrophage proinflammatory activation during the development of renal fibrosis. Therefore, inhibition of cGAS-STING signalling may represent a novel therapeutic strategy for CKD.
Collapse
Affiliation(s)
- Baihai Jiao
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Changlong An
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Hao Du
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Melanie Tran
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Duomeng Yang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Yuqi Zhao
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Monrovia, California, USA
| | - Penghua Wang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Zhaoyong Hu
- Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut, USA
- Renal Section, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
65
|
Yang D, Peng N, Zhang H, Qiu Z, Xu L, Pan M. Cordycepin ameliorates autoimmunity by promoting STING degradation via autophagy pathway. Br J Pharmacol 2025; 182:1546-1560. [PMID: 39675775 DOI: 10.1111/bph.17425] [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: 07/15/2024] [Revised: 10/23/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Stimulator of interferon response cGAMP interactor 1 (STING), a central hub protein of cyclic GMP-AMP synthase (cGAS)-STING signalling pathway, has a crucial role in regulating type I interferons (IFNs) production and response. Recent studies indicate that excessive activation of STING is strongly associated with autoimmune diseases, including systemic lupus erythematosus (SLE). Searching immunomodulators that negatively regulate STING might greatly contribute to the suppression of autoimmunity. EXPERIMENTAL APPROACH The peripheral blood mononuclear cells (PBMCs) of SLE patients, Hela cells, L929 cells and bone marrow-derived macrophages (BMDMs) from mice were used as in vitro models. While, Trex1 KO mouse autoimmune disease model was used as in vivo model. After treatment with cordycepin, a nucleoside from Cordyceps mushrooms, type I IFNs production and response were determined by western blotting, real-time polymerase chain reaction (PCR), dual-luciferase assay, enzyme-linked immunosorbent assay (ELISA), haematoxylin-eosin staining and RNA-seq. KEY RESULTS Cordycepin inhibited type I IFNs production and response in human and murine systems following cGAS-STING signalling activation. Importantly, cordycepin markedly attenuates the autoinflammatory and autoimmune responses in Trex1 KO BMDMs and Trex1 KO mice. Furthermore, cordycepin effectively suppressed the production of type I IFNs and interferon-stimulated genes (ISGs) in the PBMCs of SLE patients. Mechanistically, cordycepin promoted STING degradation via autophagy pathway upon DNA stimulation. CONCLUSION AND IMPLICATIONS This study shows that cordycepin promotes STING autophagic degradation to alleviate autoimmunity upon DNA stimulation. Cordycepin might be a potential therapeutic candidate for alleviating aberrant type I IFNs in autoimmune and autoinflammatory diseases.
Collapse
Affiliation(s)
- Daidi Yang
- Department of Ophthalmology, The First People's Hospital of Wuhu, Wuhu, China
| | - Niannian Peng
- School of Pharmacy, Jiangsu Food and Pharmaceutical Science College, Huaian, China
| | - Hongqian Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zuocheng Qiu
- Guangdong Provincial Key Laboratory of Speed Capability Research, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lingxiao Xu
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingyu Pan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Department of Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
66
|
Yuan L, Cai Y, Wang G, Liu X, Chen B, Zhou D, Wu Y, Qu N, Li X, Zhou W. SGK3 promotes estrogen receptor-positive breast cancer proliferation by activating STAT3/ZMIZ2 pathway to stabilise β-catenin. Br J Pharmacol 2025; 182:1856-1875. [PMID: 39876548 DOI: 10.1111/bph.17453] [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: 05/17/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND PURPOSE Breast cancer is a leading threat to women's health, with approximately 70% of cases being estrogen receptor-positive. SGK3 is regulated by estrogen and is positively associated with estrogen receptor expression, although its molecular role remains unclear. EXPERIMENTAL APPROACH Proteomics was used to identify SGK3's downstream targets. Tissue microarray immunofluorescence evaluated SGK3 and ZMIZ2 expression in ER+ breast cancer. Lentiviral-mediated knockdown and overexpression of SGK3 and/or ZMIZ2 assessed their effects on cell proliferation in vitro and in vivo. Chromatin immunoprecipitation (ChIP) analyzed p-STAT3 binding to the ZMIZ2 promoter, and Co-immunoprecipitation (Co-IP) examined ZMIZ2-β-catenin interaction. KEY RESULTS SGK3 expression was elevated in breast tumour tissues correlating with reduced patient survival. Proteomic analysis identified ZMIZ2 as a downstream target of SGK3. Overexpression of SGK3 promoted the proliferation of estrogen receptor-positive breast cancer in MCF-7 and T47D cells. Inhibition had the opposite effects. ZMIZ2 overexpression rescued the proliferation deficit in SGK3 knockdown cells. ZMIZ2 was found to bind and stabilises β-catenin. Knockdown of SGK3 led to β-catenin degradation via polyubiquitination, a process reversed by ZMIZ2 overexpression. STAT3 was identified as a downstream effector of SGK3 and its knockdown reduced cytoplasmic and nuclear p-STAT3 and STAT3, and inhibited ZMIZ2 and β-catenin expression. Celastrol suppressed estrogen receptor-positive breast cancer cell proliferation by inhibiting the SGK3/STAT3/ZMIZ2/β-catenin pathway. CONCLUSIONS AND IMPLICATIONS SGK3 expression is associated with poorer survival rates, thus SGK3 is a potential therapeutic target. As celastrol can inhibit SGK3 expression it could be an effective therapeutic agent.
Collapse
Affiliation(s)
- Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Gang Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xu Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Department of Pharmacy, The Third People's Hospital of Chengdu, Sichuan, China
| | - Bo Chen
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Duanfang Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yuanli Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Na Qu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Weiying Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
67
|
Mangutov E, Dripps I, Siegersma K, Zhang Y, Bocian R, Asif S, Halbesma T, Witkowski W, Pradhan AA. Activation of δ-opioid receptors blocks allodynia in a model of headache induced by PACAP. Br J Pharmacol 2025; 182:1630-1643. [PMID: 39797405 DOI: 10.1111/bph.17424] [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: 05/03/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND AND PURPOSE Pituitary adenylate cyclase activating polypeptide (PACAP) is a human migraine trigger that is being targeted for migraine. The δ-opioid receptor (δ-receptor) is a novel target for the treatment of migraine, but its mechanism remains unclear. The goals of this study were to develop a mouse PACAP-headache model using clinically significant doses of PACAP; determine the effects of δ-receptor activation in this model; and investigate the co-expression of δ-receptors, PACAP and PACAP-PAC1 receptor. EXPERIMENTAL APPROACH Cephalic allodynia to low doses of acute and chronic PACAP were tested. A triptan (sumatriptan) and a CGRP receptor antagonist (olcegepant) were tested in this model. The δ-receptor agonist SNC80 was tested in PACAP and CGRP-induced headache models. Expression of PACAP, PAC1, CRLR and δ-receptors was determined using in situ hybridisation. KEY RESULTS Low doses of PACAP produced dose-dependent acute and chronic cephalic allodynia, blocked by sumatriptan but not by olcegepant. The PAC1 antagonist (M65) did not inhibit CGRP-induced allodynia. There was moderate co-expression of PAC1 and CRLR transcripts in migraine-related regions. SNC80 blocked PACAP- and CGRP-induced allodynia. There was low co-expression of PACAP and δ-receptors in brain regions measured. However, there was high co-expression of PAC1 and δ-receptors in somatosensory cortex, hippocampus and trigeminal nucleus caudalis. CONCLUSION AND IMPLICATIONS We developed a translationally significant model of PACAP-induced headache, which was mechanistically distinct from CGRP. Activation of δ-receptors blocked PACAP- and CGRP-induced allodynia, and δ-receptors were highly co-expressed with the PACAP-ergic system. Future studies will examine the functional relationship between δ-receptors and PAC1.
Collapse
MESH Headings
- Animals
- Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, delta/genetics
- Hyperalgesia/chemically induced
- Hyperalgesia/metabolism
- Hyperalgesia/drug therapy
- Male
- Disease Models, Animal
- Mice
- Mice, Inbred C57BL
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/genetics
- Azepines/pharmacology
- Sumatriptan/pharmacology
- Migraine Disorders/drug therapy
- Migraine Disorders/chemically induced
- Migraine Disorders/metabolism
- Quinazolines/pharmacology
- Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology
- Benzamides/pharmacology
Collapse
Affiliation(s)
- Elizaveta Mangutov
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kendra Siegersma
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yanping Zhang
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rebecca Bocian
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Sarah Asif
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Timothy Halbesma
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Wiktor Witkowski
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Amynah A Pradhan
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
68
|
de Sousa N, Correia-Silva A, Pinho AG, Vidinha-Mira A, Cainé L, Lima MF, Santos DJ, Cibrão JR, Campos J, Cavaleiro H, Pinho TS, Afonso JL, Sampaio-Marques B, Monteiro S, Silva NA, Barreiro-Iglesias A, Salgado AJ. Baclofen modulates the immune response after spinal cord injury with locomotor benefits. Br J Pharmacol 2025; 182:1783-1802. [PMID: 39842440 DOI: 10.1111/bph.17438] [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: 11/20/2023] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND AND PURPOSE Spinal cord injury (SCI) is a neurological condition that affects motor and sensory functions below the injury site. The consequences of SCI are devastating for the patients, and although significant efforts have been done in the last years, there is no effective therapy. Baclofen has emerged in the last few years as an interesting drug in the SCI field. Already used in the SCI clinical setting to control spasticity, baclofen has shown important impact on SCI recovery in animal models, such as lampreys and mice. EXPERIMENTAL APPROACH AND KEY RESULTS Herein, we proposed to go deeper into baclofen's mechanism of action and to study its role on the modulation of the immune response after SCI, a major process associated with the severeness of the lesion. Using a SCI compression mice model, we confirmed that baclofen leads to higher locomotor performance, but only at 1 mg·kg-1 and not in higher concentrations, as 5 mg·kg-1. Moreover, we found that baclofen at 1 mg·kg-1 can strongly modulate the immune response after SCI at local, systemic and peripheric levels. This is interesting and intriguingly at the same time, since now, additional studies should be performed to understand if the modulation of the immune response is the responsible for the locomotor outcomes observed on Baclofen treated animals. CONCLUSION AND IMPLICATIONS Our findings showed, for the first time, that baclofen can modulate the immune response after SCI, becoming a relevant drug in the field of the immunomodulators.
Collapse
Affiliation(s)
- Nídia de Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Ariana Correia-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Andreia G Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - André Vidinha-Mira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Laura Cainé
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Marta F Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Diogo J Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Jorge R Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Helena Cavaleiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - João L Afonso
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Antón Barreiro-Iglesias
- Department of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| |
Collapse
|
69
|
Shalomov B, Friesacher T, Yakubovich D, Combista JC, Reddy HP, Dabbah S, Bernsteiner H, Zangerl-Plessl EM, Stary-Weinzinger A, Dascal N. Ethosuximide: Subunit- and Gβγ-dependent blocker and reporter of allosteric changes in GIRK channels. Br J Pharmacol 2025; 182:1704-1718. [PMID: 39814556 DOI: 10.1111/bph.17446] [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: 08/28/2024] [Revised: 11/07/2024] [Accepted: 11/30/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND AND PURPOSE The antiepileptic drug ethosuximide (ETX) suppresses epileptiform activity in a mouse model of GNB1 syndrome, caused by mutations in Gβ1 protein, likely through the inhibition of G-protein gated K+ (GIRK) channels. Here, we investigated the mechanism of ETX inhibition (block) of different GIRKs. EXPERIMENTAL APPROACH We studied ETX inhibition of GIRK channels expressed in Xenopus oocytes with or without their physiological activator, the G protein subunit dimer Gβγ. ETX binding site and mode of action were analysed using molecular dynamic (MD) simulations and kinetic modelling, and the predictions were tested by mutagenesis and functional testing. KEY RESULTS We show that ETX is a subunit-selective, allosteric blocker of GIRKs. The potency of ETX block is increased by Gβγ, in parallel with channel activation. MD simulations and mutagenesis locate the ETX binding site in GIRK2 to a region associated with phosphatidylinositol-4,5-bisphosphate (PIP2) regulation, and suggest that ETX acts by closing the helix bundle crossing (HBC) gate and altering channel's interaction with PIP2. The apparent affinity of ETX block is highly sensitive to changes in channel gating caused by mutations in Gβ1 or GIRK subunits. CONCLUSION AND IMPLICATIONS ETX block of GIRKs is allosteric, subunit-specific, and enhanced by Gβγ through an intricate network of allosteric interactions within the channel molecule. Our findings pose GIRK as a potential therapeutic target for ETX and ETX as a potent allosteric GIRK blocker and a tool for probing gating-related conformational changes in GIRK.
Collapse
Affiliation(s)
- Boris Shalomov
- Department of Physiology and Pharmacology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Theres Friesacher
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | | | - J Carlo Combista
- Department of Physiology and Pharmacology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Haritha P Reddy
- Department of Physiology and Pharmacology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shoham Dabbah
- Department of Physiology and Pharmacology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Harald Bernsteiner
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Eva-Maria Zangerl-Plessl
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Anna Stary-Weinzinger
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Nathan Dascal
- Department of Physiology and Pharmacology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
70
|
Al-Hosni R, Agostinelli E, Ilkan Z, Scofano L, Kaye R, Dinsdale RL, Acheson K, MacDonald A, Rivers D, Biosa A, Gunthorpe MJ, Platt F, Tammaro P. Pharmacological profiling of small molecule modulators of the TMEM16A channel and their implications for the control of artery and capillary function. Br J Pharmacol 2025; 182:1719-1740. [PMID: 39829151 DOI: 10.1111/bph.17383] [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: 03/19/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND PURPOSE TMEM16A chloride channels constitute a depolarising mechanism in arterial smooth muscle cells (SMCs) and contractile cerebral pericytes. TMEM16A pharmacology is incompletely defined. We elucidated the mode of action and selectivity of a recently identified positive allosteric modulator of TMEM16A (PAM_16A) and of a range of TMEM16A inhibitors. We also explore the consequences of selective modulation of TMEM16A activity on arterial and capillary function. EXPERIMENTAL APPROACH Patch-clamp electrophysiology, isometric tension recordings, live imaging of cerebral cortical capillaries and assessment of cell death were employed to explore the effect of selective pharmacological control of TMEM16A on vascular function. KEY RESULTS In low intracellular free Ca2+ concentrations ([Ca2+]i), nanomolar concentrations of PAM_16A activated heterologous TMEM16A channels, while being almost ineffective on the closely related TMEM16B channel. In either the absence of Ca2+ or in saturating [Ca2+]i, PAM_16A had no effect on TMEM16A currents at physiological potentials. PAM_16A selectively activated TMEM16A currents in SMCs and enhanced aortic contraction caused by phenylephrine or angiotensin-II and capillary (pericyte) constriction evoked by endothelin-1 or oxygen-glucose deprivation (OGD) to simulate cerebral ischaemia. Conversely, selective TMEM16A inhibition with Ani9 facilitated aortic, mesenteric and pericyte relaxation, and protected against OGD-mediated pericyte cell death. Unlike PAM_16A and Ani9, a range of other available modulators were found to interfere with endogenous cationic currents in SMCs. CONCLUSIONS AND IMPLICATIONS Arterial tone and capillary diameter can be controlled with TMEM16A modulators, highlighting TMEM16A as a target for disorders with a vascular component, including hypertension, stroke, Alzheimer's disease and vascular dementia.
Collapse
Affiliation(s)
| | | | - Zeki Ilkan
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lara Scofano
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Rachel Kaye
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Ria L Dinsdale
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Kathryn Acheson
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Andrew MacDonald
- Autifony Therapeutics Ltd, Stevenage Bioscience Catalyst, Stevenage, UK
| | - Dean Rivers
- Autifony Therapeutics Ltd, Stevenage Bioscience Catalyst, Stevenage, UK
| | - Alice Biosa
- Autifony Srl, Istituto di Ricerca Pediatrica Citta' della Speranza, Padua, Italy
| | | | - Frances Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Paolo Tammaro
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
71
|
Hasegawa M, Kunisawa K, Wulaer B, Kubota H, Kurahashi H, Sakata T, Ando H, Fujigaki S, Fujigaki H, Yamamoto Y, Nagai T, Saito K, Nabeshima T, Mouri A. Chronic stress induces behavioural changes through increased kynurenic acid by downregulation of kynurenine-3-monooxygenase with microglial decline. Br J Pharmacol 2025; 182:1466-1486. [PMID: 39658392 DOI: 10.1111/bph.17407] [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: 08/14/2024] [Revised: 10/06/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Alterations in tryptophan-kynurenine (TRP-KYN) pathway are implicated in major depressive disorder (MDD). α7 nicotinic acetylcholine (α7nACh) receptor regulates the hypothalamic-pituitary-adrenal (HPA) axis. We have shown that deficiency of kynurenine 3-monooxygenase (KMO) induces depression-like behaviour via kynurenic acid (KYNA; α7nACh antagonist). In this study, we investigated the involvement of the TRP-KYN pathway in stress-induced behavioural changes and the regulation of the HPA axis. EXPERIMENTAL APPROACH Mice were exposed to chronic unpredictable mild stress (CUMS) and subjected to behavioural tests. We measured TRP-KYN metabolites and the expression of their enzymes in the hippocampus. KMO heterozygous mice were used to investigate stress vulnerability. We also evaluated the effect of nicotine (s.c.) on CUMS-induced behavioural changes and an increase in serum corticosterone (CORT) concentration. KEY RESULTS CUMS decreased social interaction time but increased immobility time under tail suspension associated with increased serum corticosterone concentration. CUMS increased KYNA levels via KMO suppression with microglial decline in the hippocampus. Kmo+/- mice were vulnerable to stress: they exhibited social impairment and increased serum corticosterone concentration even after short-term CUMS. Nicotine attenuated CUMS-induced behavioural changes and increased serum corticosterone concentration by inhibiting the increase in corticotropin-releasing hormone. Methyllycaconitine (α7nACh antagonist) inhibited the attenuating effect of nicotine. CONCLUSIONS AND IMPLICATIONS CUMS-induced behavioural changes and the HPA axis dysregulation could be induced by the increased levels of KYNA via KMO suppression. KYNA plays an important role in the pathophysiology of MDD as an α7nACh antagonist. Therefore, α7nACh receptor is an attractive therapeutic target for MDD.
Collapse
Affiliation(s)
- Masaya Hasegawa
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Bolati Wulaer
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Hitomi Kurahashi
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Takatoshi Sakata
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Honomi Ando
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Suwako Fujigaki
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Hidetsugu Fujigaki
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Yasuko Yamamoto
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Taku Nagai
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Kuniaki Saito
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Toshitaka Nabeshima
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| |
Collapse
|
72
|
Bate ST, Stanford SC, Page L. A three-stage strategy for conducting an experimental investigation: A recommendation to improve the reproducibility of reported conclusions. J Psychopharmacol 2025; 39:301-312. [PMID: 40071732 DOI: 10.1177/02698811251319453] [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] [Indexed: 04/03/2025]
Abstract
The reproducibility of the results from preclinical research rests on many factors, including the selection of appropriate experimental designs for the individual experiments that constitute the investigation. The design of each of these experiments depends on their purpose within the entire investigation and the information to be gained from conducting them. Here, we explain and justify a three-stage strategy comprising a series of different types of experiment, each with a different purpose and design: a pilot study, a hypothesis-generating experiment and a final hypothesis-confirming experiment. Compliance with this three-stage strategy, over the course of an entire investigation, will not only strengthen its reproducibility but, importantly, can save time and other resources, including the total number of animals used.
Collapse
Affiliation(s)
| | - S Clare Stanford
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Lee Page
- British Pharmacology Society, The Schild Plot, London, UK
| |
Collapse
|
73
|
Li HF, Lin H, Liu HT, Lin TJ, Tseng TL. Activating transcription factor-3 orchestrates the modulation of vascular anti-contractile activity and relaxation by governing the secretion of HDL-bound sphingosine-1-phosphate in perivascular adipose tissue. Br J Pharmacol 2025; 182:1763-1782. [PMID: 39843165 DOI: 10.1111/bph.17433] [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: 05/10/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND AND PURPOSE Perivascular adipose tissues (PVATs) play a critical role in modulating vascular homeostasis and protecting against cardiovascular dysfunction-mediated blood pressure dysregulation. We demonstrated that the activating transcription factor-3 (Atf3) gene in the PVAT is crucial for improving vascular wall tension abnormalities; however, its protective mechanism remains unclear. Herein, we aim to determine whether ATF3 regulates PVAT-derived relaxing factor (PVDRF) biosynthesis and if its secretion contributes to vasorelaxation. EXPERIMENTAL APPROACH This study employed an in vivo animal model using global Atf3-deficient mice, in vitro blood vessel myography, and biochemical analyses to evaluate ATF3-mediated PVDRF release and reactivity in the vasculature. KEY RESULTS Wild-type (WT) mouse thoracic aortic PVAT extracts significantly induced resting tone dilation and attenuated vasoconstrictor-induced contractile responses compared to Atf3-/- mice. Heat-stable PVAT extracts from WT mice caused sustained and reproducible vasodilation without tachyphylaxis in control aortic rings. Biochemical evaluation of PVDRF release revealed that Atf3-/- mice had lower sphingosine-1-phosphate (S1P) and HDL cholesterol (HDL-C) levels than WT mice. Furthermore, PVAT extracts from WT mice induced long-lasting vasorelaxation, which was significantly inhibited by the S1P3 receptor antagonist TY52156 and scavenger receptor class B type 1 receptor antagonist glyburide. CONCLUSION AND IMPLICATIONS ATF3 within the PVAT can modulate vascular function by strengthening sphingosine kinase 1 (sphk1)-S1P-S1P3 receptor lipid signalling and stimulating S1P binding to HDL to form the vasodilator HDL-S1P. ATF3 is an essential modulator for maintaining the physiological function of PVAT, providing a novel target for treatment of obesity-related cardiovascular diseases.
Collapse
Affiliation(s)
- Hsiao-Fen Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Heng Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Jen Lin
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- CardioVascular Research Center, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Tzu-Ling Tseng
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- CardioVascular Research Center, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
- Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
74
|
Zhu Z, Guan Q, Xu B, Bahriz S, Shen A, West TM, Zhang Y, Deng B, Wei W, Han Y, Wang Q, Xiang YK. Inhibition of the upregulated phosphodiesterase 4D isoforms improves SERCA2a function in diabetic cardiomyopathy. Br J Pharmacol 2025; 182:1487-1507. [PMID: 39662482 DOI: 10.1111/bph.17411] [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: 12/27/2023] [Revised: 09/04/2024] [Accepted: 10/15/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND AND PURPOSE Sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) is impaired in heart failure. Phosphodiesterases (PDEs) are implicated in the modulation of local cAMP signals and protein kinase A (PKA) activity essential for cardiac function. We characterise PDE isoforms that underlie decreased activities of SERCA2a and reduced cardiac contractile function in diabetic cardiomyopathy. EXPERIMENTAL APPROACH Wild type mice were fed with either normal chow or a high-fat diet (HFD). Cardiomyocytes were isolated for excitation-contraction coupling (ECC), fluorescence resonant energy transfer PKA biosensor and proximity ligation assays. KEY RESULTS The upregulated PDE4D3 and PDE4D9 isoforms in HFD cardiomyocytes specifically bound to SERCA2a but not ryanodine receptor 2 (RyR2) on the sarcoplasmic reticulum (SR). The increased association of PDE4D isoforms with SERCA2a in HFD cardiomyocytes led to reduced local PKA activities and phosphorylation of phospholamban (PLB) but minimally effected the PKA activities and phosphorylation of RyR2. These changes correlate with slower calcium decay tau in the SR and attenuation of ECC in HFD cardiomyocytes. Selective inhibition of PDE4D3 or PDE4D9 restored PKA activities and phosphorylation of PLB at the SERCA2a complex, recovered calcium decay tau, and increased ECC in HFD cardiomyocytes. Therapies with PDE4 inhibitor roflumilast, PDE4D inhibitor BPN14770 or genetical deletion of PDE4D restored PKA phosphorylation of PLB and cardiac contractile function. CONCLUSION AND IMPLICATIONS The current study identifies upregulation of specific PDE4D isoforms that selectively inhibit SERCA2a function in HFD-induced cardiomyopathy, indicating that this remodelling can be targeted to restore cardiac contractility in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhenduo Zhu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Qiuyun Guan
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Bing Xu
- Department of Pharmacology, University of California, Davis, Davis, California, USA
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, USA
| | - Sherif Bahriz
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Ao Shen
- School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Toni M West
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Yu Zhang
- School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bingqing Deng
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yongsheng Han
- Department of Emergency Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, Davis, California, USA
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, USA
| |
Collapse
|
75
|
Kodama T, Kameshima S, Otani K, Okada M, Yamawaki H. The eukaryotic elongation factor 2 kinase inhibitor, A484954, induces hypoglycaemic and hypotensive effects. Br J Pharmacol 2025; 182:1823-1835. [PMID: 39842419 DOI: 10.1111/bph.17443] [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: 01/21/2024] [Revised: 10/22/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND AND PURPOSE Eukaryotic elongation factor 2 kinase (eEF2K) belongs to the Ca2+/calmodulin-dependent protein kinase family. We previously revealed that A484954, a selective eEF2K inhibitor, caused hypotensive and diuretic effects via the production of nitric oxide (NO) in spontaneously hypertensive rats. Otsuka Long-Evans Tokushima Fatty (OLETF) rats are hypertensive because of obesity and type 2 diabetes. Because an NO synthase inhibitor was reported to increase the expression of sodium glucose co-transporter 2 (SGLT2), we hypothesised that A484954 causes not only hypotensive but also hypoglycaemic effects via NO production in OLETF rats. EXPERIMENTAL APPROACH To test the hypothesis, we examined the effects of A484954 administration on hyperglycaemia and hypertension in OLETF rats. OLETF rats were given an intraperitoneal injection of A484954 (2.5 mg kg-1 day-1) for 7 days. Then, we measured blood and urinary glucose level, urine output, systolic blood pressure and ventricular contractility. We also conducted Western blotting and isometric tension measurements. KEY RESULTS A484954 induced a decrease in blood glucose, an increase in urinary glucose excretion, and a decrease in protein expression of kidney SGLT2. In addition, A484954 induced a decrease in systolic blood pressure, an NO-dependent vasorelaxation, and a diuretic effect. Further, A484954 enhanced left ventricular contractility. CONCLUSION AND IMPLICATIONS We, for the first time, revealed that (1) A484954 caused hypoglycaemic effects through increasing urinary glucose excretion via decreasing SGLT2, (2) A484954 improved diabetic complication, including hypertension, through vasorelaxation and diuresis via NO production, and (3) A484954 had a positive inotropic effect.
Collapse
Affiliation(s)
- Tomoko Kodama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Satoshi Kameshima
- Laboratory of Small Animal Internal Medicine, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Kosuke Otani
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Japan
| |
Collapse
|
76
|
Petrosiute A, Zakšauskas A, Lučiūnaitė A, Petrauskas V, Baranauskienė L, Kvietkauskaitė A, Ščerbavičienė A, Tamošiūnaitė M, Musvicaitė J, Jankūnaitė A, Žvinys G, Stančaitis L, Čapkauskaitė E, Mickevičiūtė A, Juozapaitienė V, Dudutienė V, Zubrienė A, Grincevičienė Š, Bukelskienė V, Schiöth HB, Matulienė J, Matulis D. Carbonic anhydrase IX inhibition as a path to treat neuroblastoma. Br J Pharmacol 2025; 182:1610-1629. [PMID: 39776083 DOI: 10.1111/bph.17429] [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: 06/03/2024] [Revised: 11/12/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Tumour hypoxia frequently presents a major challenge in the treatment of neuroblastoma (NBL). The neuroblastoma cells produce carbonic anhydrase IX (CA IX), an enzyme crucial for the survival of cancer cells in low-oxygen environments. EXPERIMENTAL APPROACH We designed and synthesised a novel high-affinity inhibitor of CA IX. The highest to-date. The affinities were determined for all human catalytically active CA isozymes showing significant selectivity for CA IX over other isozymes. The inhibitor effect on neuroblastoma cancer cell growth was determined in vitro and in vivo via a mice xenograft model. KEY RESULTS The novel designed inhibitor effectively mitigated the acidification induced by CA IX and reduced spheroid growth under hypoxic conditions in the SK-N-AS cell line. It also diminished the secretion of pro-tumour chemokines IL-8 (CXCL2) and CCL2. When we combined this novel CA IX inhibitor with a compound that inhibits the chemokine receptor CCR2 protein activity, we observed a reduction in mouse tumour growth. The combined treatment also prompted tumours to exhibit adaptive resistance by producing higher levels of vascular endothelial growth factor receptors (VEGFR) and other compensatory signals. CONCLUSIONS AND IMPLICATIONS This research underscores the pivotal role of CA IX in cancer and the potential of a novel CA IX inhibitor-based combination intervention therapy for neuroblastoma treatment.
Collapse
Affiliation(s)
- Agne Petrosiute
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Audrius Zakšauskas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Asta Lučiūnaitė
- Department of Immunology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vytautas Petrauskas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Lina Baranauskienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agnė Kvietkauskaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Alvilė Ščerbavičienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Marta Tamošiūnaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Justina Musvicaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Alberta Jankūnaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Gediminas Žvinys
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Laimonas Stančaitis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Edita Čapkauskaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aurelija Mickevičiūtė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vaida Juozapaitienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Virginija Dudutienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Asta Zubrienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Švitrigailė Grincevičienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Virginija Bukelskienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Helgi B Schiöth
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jurgita Matulienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
77
|
Wang Z, Zhang LN, Wu T, Pan X, Li L, Yang X, Zhang M, Liu Y, Liu Y. Actions of dexmedetomidine in regulating NLRP3 in postoperative cognitive dysfunction in aged mice via the autophagy-lysosome pathway. Br J Pharmacol 2025; 182:1683-1703. [PMID: 39815423 DOI: 10.1111/bph.17378] [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: 01/12/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND AND PURPOSE Autophagy-lysosomal pathway dysfunction leads to postoperative cognitive dysfunction (POCD). Dexmedetomidine (Dex) improves POCD, and we probed the effects of Dex on autophagy-lysosomal pathway dysfunction in a POCD model. EXPERIMENTAL APPROACH A POCD mouse model was established and intraperitoneally injected with Dex. Cognitive function was evaluated by Morris water maze/open field test/novel object recognition assay. Levels of neurotransmitters/inflammatory cytokines in hippocampus, and NLRP3/ASC/Cleaved Caspase-1 proteins were determined by ELISA/Western blot. NLRP3 inflammasome-mediated microglial activation/astrocyte A1 differentiation in the hippocampal CA1 region were assessed by immunofluorescence assay. BV-2 cells were treated with lipopolysaccharide (LPS) and Dex and/or the NLRP3 inflammasome activator Nigericin, and transfected with si-TFEB for co-culture with primary reactive astrocytes (RAs) to verify the function of Dex in vitro. KEY RESULTS Dex alleviated cognitive dysfunction in POCD mice and repressed NLRP3 inflammasome-mediated microglial activation and astrocyte A1 differentiation. NLRP3 inflammasome activation partially reversed the protective effect of Dex on the POCD condition. In vitro experiments verified the inhibitory properties of Dex on microglial activation and astrocyte A1 differentiation. Dex induces TFEB nuclear translocation, microglial autophagy and lysosomal biogenesis. By activating the autophagy-lysosome pathway, Dex regulated NLRP3 inflammasome-mediated microglial activation, inhibited astrocyte A1 differentiation and alleviated POCD in vivo. CONCLUSION AND IMPLICATIONS Dex regulates NLRP3 inflammasome-mediated hippocampal microglial activation by promoting TFEB nuclear translocation and activating the autophagy-lysosome pathway and inhibits astrocyte A1 differentiation, thereby alleviating POCD.
Collapse
Affiliation(s)
- Zhi Wang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Li-Na Zhang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Ting Wu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Xu Pan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Le Li
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Xin Yang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Miao Zhang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Ying Liu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Anesthesiology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
78
|
Yoshioka T, Kimiki S, Yamazaki M, Hamano T, Ou M, Ode Y, Ehara R, Kajino K, Kasai S, Yoshizawa K, Saitoh T, Yamada D, Nagase H, Saitoh A. Agonists of the opioid δ-receptor improve irritable bowel syndrome-like symptoms via the central nervous system. Br J Pharmacol 2025; 182:1599-1609. [PMID: 39721072 DOI: 10.1111/bph.17428] [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: 06/18/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND AND PURPOSE Irritable bowel syndrome (IBS) is a common condition that is challenging to treat, and novel drugs are needed for this condition. Previously, a chronic vicarious social defeat stress (cVSDS) mouse model exhibits IBS-like symptoms. Also agonists of the opioid δ-receptor exert anti-stress effects in rodents with minimal adverse effects. Here, we evaluated the effects of δ-receptor agonists on the IBS-like symptoms in cVSDS mice. EXPERIMENTAL APPROACH cVSDS mice (male C57BL/6J mice) were prepared following a 10-day exposure to witness of social defeat stress. Subsequently, intestinal peristaltic motility and abdominal hyperalgesia were evaluated using the charcoal meal test (CMT) and capsaicin-induced hyperalgesia test (CHT), respectively. Extracellular glutamate levels were measured using in vivo brain microdialysis. The drug was singly administrated 30 min before testing. KEY RESULTS In cVSDS mice, systemic (10 mg kg-1) and intracerebroventricular (30 nmol) administration of a δ-receptor agonist regulated intestinal peristalsis in the CMT and relieved abdominal pain in the CHT. Effects of systemic administration were blocked by intracerebroventricular injection of a δ-receptor inhibitor. Local infusion of the δ-receptor agonist (0.6 nmol) into the insular cortex improved cVSDS-induced intestinal hypermotility. The in vivo brain microdialysis study showed that re-exposure to VSDS elevated the extracellular glutamate levels in the IC, which was restored by the δ-receptor agonist. CONCLUSIONS AND IMPLICATIONS We propose that agonists of opioid δ-receptors are potential drugs for the radical treatment of IBS because they can ameliorate IBS-like symptoms via the CNS, specifically the insular cortex.
Collapse
Affiliation(s)
- Toshinori Yoshioka
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Sayaka Kimiki
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Mayuna Yamazaki
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Takumi Hamano
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Mizuki Ou
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yumi Ode
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Rui Ehara
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Keita Kajino
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Satoka Kasai
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kazumi Yoshizawa
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Daisuke Yamada
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Hiroshi Nagase
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Akiyoshi Saitoh
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
79
|
Seaman RW, Galindo DG, Stinson BT, Sulima A, Rice KC, Javors MA, Ginsburg BC, Collins GT. Cardiovascular and locomotor effects of binary mixtures of common 'bath salts' constituents: Studies with methylone, methylenedioxypyrovalerone and caffeine in rats. Br J Pharmacol 2025; 182:1836-1855. [PMID: 39843219 DOI: 10.1111/bph.17444] [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: 01/29/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND AND PURPOSE The use of 'bath salts' drug preparations has been associated with high rates of toxicity and death. Preparations often contain mixtures of drugs, including multiple synthetic cathinones or synthetic cathinones and caffeine. Little is known about the interactions of 'bath salts' constituents and adverse effects often reported by users. EXPERIMENTAL APPROACH This study used adult male Sprague-Dawley rats to characterise the cardiovascular effects, locomotor effects and pharmacokinetics of methylone, methylenedioxypyrovalerone (MDPV) and caffeine, administered alone and as binary mixtures. Dose-addition analyses were used to determine the effect levels of a strictly additive interaction for dose pairs. KEY RESULTS Methylone, MDPV and caffeine increased heart rate (HR) and locomotion, with methylone producing the largest increase in HR, MDPV producing the largest increase in locomotor activity and caffeine being the least effective in stimulating HR and locomotor activity. MDPV and caffeine increased mean arterial pressure (MAP), with caffeine being more effective than MDPV. The nature of the interactions between methylone and MDPV tended towards sub-additivity for all endpoints, whereas interactions between MDPV or methylone and caffeine tended to be additive or sub-additive for cardiovascular endpoints, and additive or supra-additive for increases in locomotion. No pharmacokinetic interactions were observed between individual constituents, but methylone appeared to display nonlinear pharmacokinetics at the largest dose evaluated. CONCLUSION AND IMPLICATIONS These findings demonstrate that 'bath salts' preparations can impact both cardiovascular and locomotor effects and suggest that interactions among constituent drugs could contribute to the 'bath salts' toxidrome reported by human users.
Collapse
Affiliation(s)
- Robert W Seaman
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David G Galindo
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Benjamin T Stinson
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, NIDA and NIAAA, Bethesda, Maryland, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, NIDA and NIAAA, Bethesda, Maryland, USA
| | - Martin A Javors
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Brett C Ginsburg
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Gregory T Collins
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
80
|
Faggin S, Cerantola S, Caputi V, Tietto A, Stocco E, Bosi A, Ponti A, Bertazzo A, Macchi V, Porzionato A, Savarino EV, Giaroni C, Giron MC. Toll-like receptor 4 deficiency ameliorates experimental ileitis and enteric neuropathy: Involvement of nitrergic and 5-hydroxytryptaminergic neurotransmission. Br J Pharmacol 2025; 182:1803-1822. [PMID: 39842456 DOI: 10.1111/bph.17439] [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: 05/22/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND AND PURPOSE Inflammatory bowel disease (IBD) patients display genetic polymorphisms in toll-like receptor 4 (TLR4) genes, contributing to dysregulate enteric nervous system (ENS) circuits with increased levels of 5-HT and alteration of the neuroimmune crosstalk. In this study, we investigated the impact of TLR4 signalling on mouse ENS dysfunction caused by dextran sulphate sodium (DSS)-induced ileitis. EXPERIMENTAL APPROACH Male C57BL/6J (wild-type [WT]) and TLR4-/- mice (10 ± 2 weeks old) received 2% DSS in drinking water for 5 days and then were switched to 3-day regular drinking water. Histological analysis and proinflammatory cytokine mRNA levels were assessed in ileal samples. Gut motility was evaluated by changes in transit of a fluorescent-labelled marker and isometric neuromuscular responses of ileal full-thickness segments to receptor and non-receptor-mediated stimuli. Alterations in ENS architecture were assessed by confocal immunohistochemistry in longitudinal muscle-myenteric plexus whole-mount preparations. KEY RESULTS In WT mice, DSS treatment caused delayed gastrointestinal transit, ileal myenteric neurodegeneration, reactive gliosis and release of proinflammatory cytokines. Enhanced cholinergic and tachykinergic excitatory tone, increased inducible nitric oxide synthase (iNOS)-mediated relaxation, and changes in 5-HT2A and 5-HT3 receptor-mediated responses were observed during ileitis in WT mice. TLR4 deficiency reversed most of the functional and morphological abnormalities. CONCLUSION AND IMPLICATIONS Our results demonstrate that TLR4 activity influences the severity of ileitis, neuroglial plasticity, gut motility, and nitrergic and 5-HTergic neurotransmissions. The neuroimmune interaction between TLR4 and 5-HT observed in our study appears to be a potential pharmacological target to treat ENS dysfunction implicated in IBD onset/progression.
Collapse
Affiliation(s)
- Sofia Faggin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Silvia Cerantola
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Valentina Caputi
- Poultry Production and Product Safety Research Unit, Agricultural Research Service, United States Department of Agriculture, Fayetteville, Arkansas, USA
| | - Angela Tietto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
- School of Specialization in Clinical Pharmacology and Toxicology, University of Ferrara, Ferrara, Italy
| | - Elena Stocco
- Department of Neuroscience, University of Padua, Padua, Italy
- Department of Women's and Children's Health, University of Padua, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Annalisa Bosi
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Alessandra Ponti
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Antonella Bertazzo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Veronica Macchi
- Department of Neuroscience, University of Padua, Padua, Italy
| | | | - Edoardo V Savarino
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Cristina Giaroni
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
81
|
Göntér K, László S, Tékus V, Dombi Á, Fábián K, Pál S, Pozsgai G, Botz L, Wagner Ö, Pintér E, Hajna Z. New generation capsaicin-diclofenac containing, silicon-based transdermal patch provides prolonged analgesic effect in acute and chronic pain models. Eur J Pharm Sci 2025; 207:107035. [PMID: 39922237 DOI: 10.1016/j.ejps.2025.107035] [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: 11/05/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVE Pain is one of the major public health burdens worldwide, however, conventional analgesics are often ineffective. Capsaicin-the active compound of Capsicum species, being responsible for their pungency-has been part of traditional medicine long ago. Capsaicin is a natural agonist of the Transient Receptor Potential Vanilloid 1 receptor-localized on capsaicin-sensitive sensory neurons and strongly involved in pain transmission-, and has been in focus of analgesic drug research for many years. In this study, we aimed to develop a sustained release transdermal patch (transdermal therapeutic system, TTS) combining the advantages of low-concentration capsaicin and diclofenac embedded in an innovative structure, as well as to perform complex preclinical investigations of its analgesic effect. METHODS Drug delivery properties of the TTS were investigated with Franz cell and flow-through cell tests. Analgesic effect of the TTS was examined in in vivo models of acute postoperative and inflammatory, chronic neuropathic and osteoarthritic pain. RESULTS Modified silicone polymer matrix-based TTS containing low-concentration capsaicin and diclofenac has been developed, releasing both compounds according to zero-order kinetics. Moreover, capsaicin and diclofenac facilitated the liberation of each other. Combined TTS significantly reduced acute postoperative and inflammatory pain, as well as chronic neuropathic and osteoarthritic pain. Interestingly, in acute postoperative and chronic osteoarthritic pain, capsaicin prolonged and potentiated the pain-relieving effect of diclofenac. CONCLUSIONS New generation combined low-concentration capsaicin-diclofenac containing TTS can be an effective therapeutic tool in acute and chronic pain states involving neuropathic and inflammatory components.
Collapse
Affiliation(s)
- Kitti Göntér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; HUN-REN, Chronic Pain Research Group, University of Pécs, Pécs, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, Budapest, 1117, Hungary
| | - Szabolcs László
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111, Budapest, Hungary; HUN-REN, Computation-Driven Chemistry Research Group, Műegyetem rkp. 3, H-1111, Budapest, Hungary
| | - Valéria Tékus
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
| | - Ágnes Dombi
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Katalin Fábián
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Szilárd Pál
- Institute of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Gábor Pozsgai
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus str. 2, H-7624, Pécs, Hungary
| | - Lajos Botz
- Institute of Clinical Pharmacy, Clinical Centre, University of Pécs, Honvéd str. 3, H-7624, Pécs, Hungary
| | - Ödön Wagner
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111, Budapest, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; HUN-REN, Chronic Pain Research Group, University of Pécs, Pécs, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, Budapest, 1117, Hungary.
| | - Zsófia Hajna
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary; HUN-REN, Chronic Pain Research Group, University of Pécs, Pécs, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, Budapest, 1117, Hungary
| |
Collapse
|
82
|
Mei Y, Chen X, Shi S, Lin W, Cheng Z, Fan X, Wu W, Han J, Huang W, Ye B, Dai S. GI-Y2, a novel gasdermin D inhibitor, attenuates sepsis-induced myocardial dysfunction by inhibiting gasdermin D-mediated pyroptosis in macrophages. Br J Pharmacol 2025. [PMID: 40165368 DOI: 10.1111/bph.70040] [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: 06/20/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND AND PURPOSE Myocardial dysfunction is a significant complication associated with sepsis. However, there are currently no specific and effective treatments available. Inhibiting gasdermin D (GSDMD)-mediated pyroptosis has shown promise in mitigating sepsis-induced myocardial dysfunction. The GSDMD inhibitor Y2 (GI-Y2) has been demonstrated to directly bind to GSDMD. Nonetheless, it remains uncertain whether GI-Y2 offers a cardioprotective effect in the context of sepsis-induced myocardial dysfunction. EXPERIMENTAL APPROACH A mouse model of sepsis was created using lipopolysaccharide (LPS), caecal ligation and puncture. Following treatment with GI-Y2 or macrophage membrane-encapsulated GI-Y2 nanoparticles (GI-Y2@MM-NPs), myocardial dysfunction and pyroptosis levels in heart tissues were assessed. Transcriptome sequencing revealed the molecular mechanism of GI-Y2 in treating septic cardiomyopathy. KEY RESULTS We observed that GI-Y2 alleviated myocardial dysfunction and attenuated cardiac inflammation in mice induced by LPS, caecal ligation and puncture. GI-Y2 reduced macrophage pyroptosis and attenuated macrophage-mediated cardiomyocyte injury induced by LPS/nigericin. Concurrently, we confirmed the protective effect of GI-Y2 against LPS-induced cardiac dysfunction was abolished in the absence of GSDMD. Additionally, GI-Y2 attenuated the mitochondrial damage induced by LPS by inhibiting GSDMD in the mitochondria. Furthermore, we developed GI-Y2@MM-NPs to enhance the targeting capability of GI-Y2 towards macrophages in heart tissues and demonstrated its protective effect in vivo. CONCLUSION AND IMPLICATIONS These findings indicate that GI-Y2 alleviates septic myocardial injury and dysfunction by specifically targeting GSDMD, thereby inhibiting GSDMD-mediated pyroptosis and mitochondrial damage. Both GI-Y2 and GI-Y2@MM-NPs may serve as promising therapeutic options for addressing septic myocardial dysfunction.
Collapse
Affiliation(s)
- Yiling Mei
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, Ningbo Hangzhou Bay Hospital, Ningbo, Zhejiang, China
| | - Si Shi
- First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wante Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenfeng Cheng
- Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenqi Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Weijian Huang
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
83
|
Lin L, Sun B, Hu Y, Yang W, Li J, Wang D, Zhang L, Lu M, Li Y, Li Y, Zhang D, Li C. Rhynchophylline as an agonist of sirtuin 3 ameliorates endothelial dysfunction via antagonizing mitochondrial damage of endothelial progenitor cells. Br J Pharmacol 2025. [PMID: 40164963 DOI: 10.1111/bph.70032] [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: 04/28/2024] [Revised: 02/14/2025] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND AND PURPOSE Mitochondrial dysregulation of endothelial progenitor cells (EPCs) has been implicated in endothelial destruction and hypertension. Regulation of silent information regulator 3 (sirtuin 3; SIRT3) in mitochondrial damage of EPCs and the underlying molecular mechanisms remain unclear, and evidence of selective SIRT3 agonists for the treatment of hypertension also is lacking. EXPERIMENTAL APPROACH Here, we discovered a potent SIRT3 agonist, rhynchophylline (Rhy), and explored its underlying action on mitochondrial damage of EPCs and endothelial dysfunction. KEY RESULTS In spontaneously hypertensive rats, Rhy reduced blood pressure and ameliorated vasomotion, paralleling improved EPC function in the peripheral circulation. Moreover, Rhy alleviated mitochondrial damage and inhibited apoptosis via the mitochondrial apoptotic pathway. SIRT3 knockdown interrupted the regulation of mitochondrial homeostasis induced by Rhy, thus abolishing its antagonizing effect on EPC dysfunction and endothelial damage, suggesting that Rhy protection of EPC mitochondria is mediated via the activation of SIRT3. Rhy restrained the production of mitochondrial ROS and improved the activity of superoxide dismutase 2 (SOD2) in a SIRT3-dependent manner, whereas silencing SOD2 eliminated the inhibition by Rhy of oxidative stress and apoptosis, reflecting that SOD2 was indispensable for the regulation of Rhy on mitochondrial dysfunction and the mitochondrial-mediated apoptosis pathway. CONCLUSION AND IMPLICATIONS SIRT3-dependent mitochondrial homeostasis contributes to attenuating hypertension-related EPC dysfunction and endothelial injury, and Rhy itself is a potent and targeted SIRT3 agonist that prevented mitochondrial dysfunction by regulating the SIRT3/SOD2 pathway, which may provide new clues for drug candidates for hypertension therapeutics.
Collapse
Affiliation(s)
- Lin Lin
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bowen Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenqing Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Danyang Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengkai Lu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunlun Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
84
|
Kulig P, Brazauskas P, Suffiotti M, Raoult E, Babilonski U, Renault B, Grieder U, Vezzali E, Blattmann P, Martinic MM, Murphy MJ. Efficacy of IDOR-1117-2520, a novel, orally available CCR6 antagonist in preclinical models of skin dermatitis. Br J Pharmacol 2025. [PMID: 40156059 DOI: 10.1111/bph.70025] [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: 10/18/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND AND PURPOSE The chemokine receptor CCR6 guides pathogenic T17 cells, implicated in autoimmune diseases including psoriasis, to sites of inflammation via the chemokine CCL20. Therefor, pharmacological inhibition of CCR6+ immune cell migration provides a novel therapeutic approach. Translatability of such an intervention has not yet been assessed in detail. We evaluated the translatability of the Aldara® mouse model induced skin inflammation to psoriasis, with particular focus on immune cell trafficking and assessed the efficacy of IDOR-1117-2520, a highly selective, potent and orally available CCR6 small inhibitor. EXPERIMENTAL APPROACH Effects of IDOR-1117-2520 were investigated in the Aldara® and IL23 mouse models of skin inflammation using flow cytometry, RNA sequencing and transcriptome-based cell type deconvolution approaches to characterise immune cell migration patterns. These results were compared to human psoriasis transcriptomics data. KEY RESULTS IDOR-1117-2520 dose dependently reduced infiltration of CCR6+ immune cells into inflamed skin, and was equally efficacious as IL-17 and IL-23 inhibition in models of skin inflammation. Pathway analysis showed molecular similarities in the immune response between human psoriasis and the Aldara® mouse model. IL-17/IL-23 pathway genes were expressed in both human psoriasis and the mouse model. CCR6 inhibition modulated multiple pathways associated with inflammation beyond the proximal IL-17/IL-23 pathway. A chemokine-chemokine receptor interaction map implicated CCL20-CCR6 as the dominant axis in recruiting pathogenic T17 cells in both the model and in human psoriasis. CONCLUSION AND IMPLICATIONS IDOR-1117-2520 could provide a promising novel targeted approach to treating psoriasis and, potentially, other autoimmune diseases involving the CCR6/CCL20 axis and the IL-17/IL-23 pathway. IDOR-1117-2520 is currently being evaluated in a clinical phase 1 trial (ISRCTN28892128).
Collapse
Affiliation(s)
- Paulina Kulig
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Pijus Brazauskas
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Madeleine Suffiotti
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Emilie Raoult
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Ulrike Babilonski
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Bérengère Renault
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Ursula Grieder
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Enrico Vezzali
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Peter Blattmann
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Marianne M Martinic
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Mark J Murphy
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| |
Collapse
|
85
|
Michael A, Onisiforou A, Georgiou P, Koumas M, Powels C, Mammadov E, Georgiou AN, Zanos P. (2R,6R)-hydroxynorketamine prevents opioid abstinence-related negative affect and stress-induced reinstatement in mice. Br J Pharmacol 2025. [PMID: 40155780 DOI: 10.1111/bph.70018] [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: 11/22/2024] [Revised: 01/09/2025] [Accepted: 02/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND AND PURPOSE Opioid use disorder (OUD) is a pressing public health concern marked by frequent relapse during periods of abstinence, perpetuated by negative affective states. Classical antidepressants or the currently prescribed opioid pharmacotherapies have limited efficacy to reverse the negative affect or prevent relapse. EXPERIMENTAL APPROACH Using mouse models, we investigated the effects of ketamine's metabolite (2R,6R)-hydroxynorketamine (HNK) on reversing conditioning to sub-effective doses of morphine in stress-susceptible mice, preventing conditioned-place aversion and alleviating acute somatic abstinence symptoms in opioid-dependent mice. Additionally, we evaluated its effects on anhedonia, anxiety-like behaviours and cognitive impairment during protracted opioid abstinence, while mechanistic studies examined cortical EEG oscillations and synaptic plasticity markers. KEY RESULTS (2R,6R)-HNK reversed conditioning to sub-effective doses of morphine in stress-susceptible mice and prevented conditioned-place aversion and acute somatic abstinence symptoms in opioid-dependent mice. In addition, (2R,6R)-HNK reversed anhedonia, anxiety-like behaviours and cognitive impairment emerging during protracted opioid abstinence plausibly via a restoration of impaired cortical high-frequency EEG oscillations, through a GluN2A-NMDA receptor-dependent mechanism. Notably, (2R,6R)-HNK facilitated the extinction of opioid conditioning, prevented stress-induced reinstatement of opioid-seeking behaviours and reduced the propensity for enhanced morphine self-consumption in mice previously exposed to opioids. CONCLUSIONS AND IMPLICATIONS These findings emphasize the therapeutic potential of (2R,6R)-HNK, which is currently in Phase II clinical trials, in addressing stress-related opioid responses. Reducing the time and cost required for development of new medications for the treatment of OUDs via drug repurposing is critical due to the opioid crisis we currently face.
Collapse
Affiliation(s)
- Andria Michael
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Anna Onisiforou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Morfeas Koumas
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chris Powels
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Elmar Mammadov
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Andrea N Georgiou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
86
|
Bruge C, Bourg N, Pellier E, Tournois J, Polentes J, Benabides M, Grossi N, Bigot A, Brureau A, Richard I, Nissan X. High-throughput screening identifies bazedoxifene as a potential therapeutic for dysferlin-deficient limb girdle muscular dystrophy. Br J Pharmacol 2025. [PMID: 40108832 DOI: 10.1111/bph.70017] [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: 04/19/2024] [Revised: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Limb-girdle muscular dystrophy R2 (LGMD R2) is a rare genetic disorder characterised by progressive weakness and wasting of proximal muscles. LGMD R2 is caused by the loss of function of dysferlin, a transmembrane protein crucial for plasma membrane repair in skeletal muscles. This study aimed to identify drugs that could improve the localisation and restore the function of an aggregated mutant form of dysferlin (DYSFL1341P). EXPERIMENTAL APPROACH We developed an in vitro high-throughput assay to monitor the expression and reallocation of aggregated mutant dysferlin (DYSFL1341P) in immortalised myoblasts. After screening 2239 clinically approved drugs and bioactive compounds, the ability of the more promising candidates to improve cell survival following hypo-osmotic shock was assessed. Their protective effects were evaluated on immortalised myoblasts carrying other dysferlin mutations and on dysferlin-deficient muscle fibres from Bla/J mice. KEY RESULTS We identified two compounds, saracatinib and bazedoxifene, that increase dysferlin content in cells carrying the DYSFL1341P mutation. Both drugs improved cell survival and plasma membrane resistance following osmotic shock. Whereas saracatinib acts specifically on misfolded L1341P dysferlin, bazedoxifene shows an additional protective effect on dysferlin KO immortalised myoblasts and mice muscle fibres. Further analysis revealed that bazedoxifene induces autophagy flux, which may enhance the survival of LGMD R2 myofibres. CONCLUSION AND IMPLICATIONS Our drug screening identified saracatinib and bazedoxifene as potential treatments for LGMD R2, especially for patients with the L1341P mutation. The widespread protective effect of bazedoxifene reveals a new avenue toward genotype-independent treatment of LGMD R2 patients.
Collapse
Affiliation(s)
- Celine Bruge
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Nathalie Bourg
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| | - Emilie Pellier
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Johana Tournois
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Jerome Polentes
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Manon Benabides
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Noella Grossi
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Anthony Brureau
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| | - Isabelle Richard
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| | - Xavier Nissan
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| |
Collapse
|
87
|
Bergadà-Martínez A, de Los Reyes-Ramírez L, Martínez-Torres S, Ciaran-Alfano L, Martínez-Gallego I, Maldonado R, Rodríguez-Moreno A, Ozaita A. Sub-chronic administration of AM6545 enhances cognitive performance and induces hippocampal synaptic plasticity changes in naïve mice. Br J Pharmacol 2025. [PMID: 40102206 DOI: 10.1111/bph.70015] [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: 06/19/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND AND PURPOSE There is evidence of crosstalk between the brain and peripheral tissues. However, how the periphery contributes to brain function is not well understood. The cannabinoid CB1 receptor is classically pictured to have a relevant role in cognitive function. We previously demonstrated a novel mechanism where acute administration of the CB1 receptor antagonist AM6545, largely restricted to the periphery, prolonged memory persistence in mice. Here, we have assessed the effects of repeated exposure to AM6545 on cognitive improvements. EXPERIMENTAL APPROACH We evaluated, in young adult male and female mice, the behavioural consequences of sub-chronic treatment with AM6545. An unbiased transcriptomic analysis, as well as electrophysiological and biochemical studies, was carried out to elucidate the central cellular and molecular consequences of such action at peripheral receptors. KEY RESULTS Sub-chronic AM6545 enhanced memory in low and high arousal conditions in male and female mice. Executive function was facilitated after repeated AM6545 administration in male mice. Transcriptional analysis of hippocampal synaptoneurosomes from treated mice revealed a preliminary, sex-dependent, modulation of synaptic transcripts by AM6545. Notably, AM6545 occluded long-term potentiation in CA3-CA1 synapses while enhancing input-output relation in male mice. This was accompanied by an increase in hippocampal expression of Bdnf and Ngf. CONCLUSION AND IMPLICATIONS Our results showed that repeated administration of AM6545 contributed to the modulation of memory persistence, executive function and hippocampal synaptic plasticity in mice, further indicating that peripheral CB1 receptors could act as a target for a novel class of nootropic compounds.
Collapse
Affiliation(s)
- Araceli Bergadà-Martínez
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lucía de Los Reyes-Ramírez
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Group in Biology of Cognition, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Sara Martínez-Torres
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laura Ciaran-Alfano
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Group in Biology of Cognition, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Irene Martínez-Gallego
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Programme in Neurosciences, IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain
| | - Andrés Ozaita
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Group in Biology of Cognition, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Programme in Neurosciences, IMIM Hospital del Mar Research Institute, Barcelona, Spain
| |
Collapse
|
88
|
Saleem A, Brindha K, Punnoose AM, Vembu R. An innovative minimally-invasive vaginoscopic approach for intrauterine infusion in rats - an infertility perspective. Lab Anim 2025:236772251317268. [PMID: 40091820 DOI: 10.1177/00236772251317268] [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: 03/19/2025]
Abstract
In fertility research, intrauterine administration in small animals presents significant technical challenges, often necessitating advanced and precise techniques. Historically, surgical methods have been preferred; however, these approaches are complex, invasive and expensive. While less invasive, intravaginal methods are generally performed without direct visualization and lack standardization, which raises the risk of complications and post-procedure mortality. We present a novel, minimally invasive technique that uses video-guided vaginoscopy to overcome these constraints. This technique efficiently eliminates the need for surgical intervention and improves safety and precision by enabling clear visualization and targeted delivery beyond the cervix. To facilitate the intrauterine delivery of agents, the method utilizes a modified 1 ml micropipette tip as a speculum, designed with a 5 mm wide slit as a technical aperture. The vaginoscope, a repurposed otoscope with an integrated camera and optimal focal length, was employed into the opposite end, which was linked to a mobile device enabling real-time visualization. This creative design reduced discomfort for the animal and the researcher while allowing for exact monitoring when the catheter entered the uterine lumen, guaranteeing precise speculum alignment and producing dependable and repeatable results. The protocol has been successfully implemented over 60 times, with all infusions achieving success and no adverse events reported. This minimally invasive intrauterine technique provides a straightforward, sustainable and effective method for delivering drugs or induction agents directly into the vaginal, cervical or uterine regions, making it suitable for applications in cell therapies, gene therapies and embryo transfers in assisted reproduction technologies.
Collapse
Affiliation(s)
- Azeena Saleem
- Department of Reproductive Medicine & Surgery, Sri Ramachandra Institute of Higher Education & Research, Chennai, India
| | - Kalyanaraman Brindha
- Department of Animal Microbiology, Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai, India
| | - Alan M Punnoose
- Stem Cell and Regenerative Biology Laboratory, Sri Ramachandra Institute of Higher Education & Research, Chennai, India
| | - Radha Vembu
- Department of Reproductive Medicine & Surgery, Sri Ramachandra Institute of Higher Education & Research, Chennai, India
| |
Collapse
|
89
|
Hong L, Cai X, Zhan Y, Liu S, Zou P, Chen Y, Shao L. TLR2 activates AP-1 to facilitate CTGF transcription and stimulate doxorubicin-induced myocardial injury. Br J Pharmacol 2025. [PMID: 40097259 DOI: 10.1111/bph.17423] [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: 07/28/2023] [Revised: 10/17/2024] [Accepted: 11/11/2024] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND AND PURPOSE Our study aimed to explore the mechanistic network of toll-like receptor 2 (TLR2)/activator protein-1 (AP-1) combined with SOX10 activation of the mitogen-activated protein kinase (MAPK) pathway via connective tissue growth factor (CTGF) in doxorubicin (Dox)-induced myocardial injury. EXPERIMENTAL APPROACH Rats with Dox-induced myocardial injury were treated with a TLR2 inhibitor or CTGF silencing lentiviral vector. H9c2 cells were treated with genetic vectors or MAPK pathway activators. Cardiac function was tested using echocardiography and serum markers. H&E, Sirius red and TUNEL staining were used to detect myocardial pathological changes, collagen accumulation and apoptosis. Western blot was used to detect proteins related to cardiac hypertrophy, fibrosis, apoptosis and the MAPK pathway. H9c2 cell injury was assessed by testing cell viability, lactate dehydrogenase (LDH) release and mitochondrial membrane potential. KEY RESULTS TLR2 and CTGF were highly expressed in patients with heart failure, and Dox treatment further increased their expression. Inhibiting TLR2 or silencing CTGF improved cardiac function and reduced myocardial fibrosis and apoptosis in Dox-treated rats. Silencing of TLR2 alleviated Dox-induced H9c2 cell injury, which was nullified by CTGF overexpression. TLR2 activated AP-1, which cooperated with SOX10 to promote CTGF transcription. MAPK activation aggravated H9c2 cells against Dox-induced injury. CONCLUSIONS AND IMPLICATIONS TLR2 activates AP-1 which cooperates with SOX10 to promote CTGF transcription and subsequently activate the MAPK pathway, thereby stimulating Dox-induced myocardial injury.
Collapse
Affiliation(s)
- Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yuliang Zhan
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Songtao Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Pengtao Zou
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yanmei Chen
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
90
|
Liu H, Li R, Wang Z, Han W, Sun X, Dong X, Lou H, Xu R, Hu A, Baranenko D, Bai X, Xiao D, Lu W. Drug-likeness evaluation and inhibitory mechanism of the emodin derivative on cardiac fibrosis based on metastasis-associated protein 3. Br J Pharmacol 2025. [PMID: 40083252 DOI: 10.1111/bph.70012] [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: 08/10/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND AND PURPOSE Emodin inhibits cardiac fibrosis through metastasis-associated protein 3 (MTA3), but its limited bioavailability hinders clinical application. To enhance emodin's clinical potential, a new derivative, emodin succinyl ethyl ester, was synthesised by modifying the 3'-OH position. This study assessed its drug-likeness, anti-fibrotic properties and molecular mechanisms involving MTA3. EXPERIMENTAL APPROACH Drug-likeness properties of the emodin derivative were evaluated using computational-aided drug design (CADD). Transverse aortic constriction (TAC)-induced cardiac fibrosis and Angiotensin II (Ang II)-stimulated cardiac fibroblasts were used in vivo and ex vivo, respectively, to determine the effects of the emodin derivative on cardiac fibrosis and fibroblast transdifferentiation. Bioinformatics analysis, CADD, chromatin immunoprecipitation (ChIP), luciferase reporter assays and functional experiments were employed to predict, identify and validate the relationship between MTA3 and its upstream transcription factors. KEY RESULTS The emodin derivative exhibited superior drug-likeness and anti-fibrotic effects compared to emodin by effectively inhibiting cardiac fibroblast transdifferentiation and restored MTA3 expression. E2F1 was identified and validated as a transcriptional regulator, promoting α-SMA and COL1A2 expression, and directly reducing MTA3 expression in cardiac fibroblasts. The emodin derivative demonstrated stronger binding to the E2F1 transcription site than emodin, reducing E2F1 expression and enhancing anti-fibrotic action. CONCLUSIONS AND IMPLICATIONS The emodin derivative shows improved drug-likeness and potent inhibition of cardiac fibrosis by targeting E2F1, disrupting its pro-fibrotic function, restoring MTA3 expression and halting fibrosis progression. This advances emodin derivative's potential as a clinical therapy for cardiac fibrosis and provides insights into its anti-fibrotic mechanisms.
Collapse
Affiliation(s)
- Heng Liu
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Runze Li
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, Heilongjiang, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan, China
| | - Zhixia Wang
- Department of Pharmacy, the fourth people's hospital of Datong, Shanxi, China
| | - Weina Han
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiuxiu Sun
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xinxin Dong
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Han Lou
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Run Xu
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ange Hu
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Denis Baranenko
- School of Life Sciences, Faculty of Ecotechnologies, ITMO University, St. Petersburg, Russia
| | - Xue Bai
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University, Haikou, China
| | - Dan Xiao
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan, China
| | - Weihong Lu
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin, Heilongjiang, China
| |
Collapse
|
91
|
Fehrentz T, Amin E, Görldt N, Strasdeit T, Moussavi-Torshizi SE, Leippe P, Trauner D, Meyer C, Frey N, Sasse P, Klöcker N. Optical control of cardiac electrophysiology by the photochromic ligand azobupivacaine 2. Br J Pharmacol 2025; 182:1125-1142. [PMID: 39543799 DOI: 10.1111/bph.17394] [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: 05/17/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Patients suffering from ischaemic heart disease and heart failure are at high risk of recurrent ventricular arrhythmias (VAs), eventually leading to sudden cardiac death. While high-voltage shocks delivered by an implantable defibrillator may prevent sudden cardiac death, these interventions themselves impair quality of life and raise both morbidity and mortality, which accentuates the need for developing novel defibrillation techniques. EXPERIMENTAL APPROACH Photopharmacology allows for reversible control of biological processes by light. When relying on synthetic and externally applied chromophores, it renders genetic modification of target cells dispensable and may hence be advantageous over optogenetic approaches. Here, the photochromic ligand azobupivacaine 2 (AB2) was probed as a modulator of cardiac electrophysiology in an ex vivo intact mouse heart model. KEY RESULTS By reversibly blocking voltage-gated Na+ and K+ channels, photoswitching of AB2 modulated both the ventricular effective refractory period and the conduction velocity in native heart tissue. Moreover, photoswitching of AB2 was able to convert VA into sinus rhythm. CONCLUSION AND IMPLICATIONS The present study provides the first proof of concept that AB2 enables gradual control of cardiac electrophysiology by light. AB2 may hence open the door to the development of an optical defibrillator based on photopharmacology.
Collapse
Affiliation(s)
- Timm Fehrentz
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ehsan Amin
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nicole Görldt
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Strasdeit
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Seyed-Erfan Moussavi-Torshizi
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Leippe
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Dirk Trauner
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christian Meyer
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Division of Cardiology, Angiology and Intensive Care, EVK Düsseldorf, Cardiac Neuro- and Electrophysiology Research Consortium (cNEP), Düsseldorf, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nikolaj Klöcker
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
92
|
Bai L, Wang J, Wang X, Wang J, Zeng W, Pang J, Zhang T, Li S, Song M, Shi Y, Wang J, Wang C. Combined therapy with pirfenidone and nintedanib counteracts fibrotic silicosis in mice. Br J Pharmacol 2025; 182:1143-1163. [PMID: 39546810 DOI: 10.1111/bph.17390] [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: 10/18/2023] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Pneumoconiosis, especially silicosis, is a prevalent occupational disease with substantial global economic implications and lacks a definitive cure. Both pneumoconiosis and idiopathic pulmonary fibrosis (IPF) are interstitial lung diseases, which share many common physiological characteristics. Because pirfenidone and nintedanib are approved to treat IPF, their potential efficacy as antifibrotic agents in advanced silicosis deserves further exploration. Thus, we aimed to evaluate the individual and combined effects of pirfenidone and nintedanib in treating advanced silicosis mice and elucidate the underlying mechanisms of their therapeutic actions via multiomics. EXPERIMENTAL APPROACH We administered monotherapy or combined therapy of pirfenidone and nintedanib, with low and high doses, in silicosis established after 6 weeks and evaluated lung function, inflammatory responses and fibrotic status. Additionally, we employed transcriptomic and metabolomic analyses to uncover the mechanisms underlying different therapeutic strategies. KEY RESULTS Both pirfenidone and nintedanib were effective in treating advanced silicosis, with superior outcomes observed in combination therapy. Transcriptomic and metabolomic analyses revealed that pirfenidone and nintedanib primarily exerted their therapeutic effects by modulating immune responses, signalling cascades and metabolic processes involving lipids, nucleotides and carbohydrates. Furthermore, we experimentally validated both monotherapy and combined therapy yielded therapeutic benefits through two common signalling pathways: steroid biosynthesis and purine metabolism. CONCLUSION AND IMPLICATIONS In conclusion, pirfenidone and nintedanib, either individually or in combination, demonstrate substantial potential in advanced silicosis. Furthermore, combined therapy outperformed monotherapy, even at low doses. These therapeutic benefits are attributed to their influence on diverse signalling pathways and metabolic processes.
Collapse
Affiliation(s)
- Lu Bai
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jiaxin Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xue Wang
- Internal Medicine, Harbin Medical University, Harbin, China
- Department of Respiratory, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jixin Wang
- School of Medicine, Tsinghua University, Beijing, China
| | - Wei Zeng
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Junling Pang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Tiantian Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shengxi Li
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Meiyue Song
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yiwei Shi
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Chen Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| |
Collapse
|
93
|
Teichmann T, Pflüger-Müller B, Giménez VMM, Sailer F, Dirks H, Zehr S, Warwick T, Brettner F, Munoz-Tello P, Zimmer A, Tegeder I, Thomas D, Gurke R, Günther S, Heering J, Proschak E, Geisslinger G, Bibli IS, zu Heringdorf DM, Manucha W, Windbergs M, Knapp S, Weigert A, Leisegang MS, Kojetin D, Brandes RP. The endocannabinoid anandamide mediates anti-inflammatory effects through activation of NR4A nuclear receptors. Br J Pharmacol 2025; 182:1164-1182. [PMID: 39563075 PMCID: PMC11790392 DOI: 10.1111/bph.17366] [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: 04/17/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Endocannabinoids are lipid mediators, which elicit complex biological effects that extend beyond the central nervous system. Tissue concentrations of endocannabinoids increase in atherosclerosis, and for the endocannabinoid N-arachidonoyl-ethanolamine (anandamide, AEA), this has been linked to an anti-inflammatory function. In this study, we set out to determine the anti-inflammatory mechanism of action of AEA, specifically focusing on vascular smooth muscle cells. EXPERIMENTAL APPROACH RNA-sequencing, RT-qPCR, LC-MS/MS, NanoBit, ChIP, microscale thermophoresis, NMR structural footprinting, Gal4 reporter gene assays and loss of function approaches in cell and ex vivo organ culture were used. KEY RESULTS AEA pretreatment attenuated the cytokine-mediated induction of inflammatory gene expression such as CCL2. This effect was also observed in preparations obtained from cannabinoid receptor knockout mice and after pertussis toxin treatment. The anti-inflammatory effect of AEA required preincubation, suggesting an effect through gene induction. AEA increased the expression of the nuclear receptors NR4A1 and NR4A2. Knockdown and knockout of these receptors blocked the AEA-mediated anti-inflammatory effect in cell culture and aortic organ culture, respectively. Conversely, NR4A agonists (CsnB, C-DIM12) attenuated inflammatory gene expression. AEA binds to NR4A, and mutations in NR4A attenuated this effect. The interaction of AEA with NR4A caused recruitment of the nuclear corepressor NCoR1 to the CCL2 promoter, resulting in gene suppression. CONCLUSION AND IMPLICATIONS By binding to NR4A, AEA elicits an anti-inflammatory response in vascular smooth muscle cells. NR4A-binding by AEA analogues may represent novel anti-inflammatory agents.
Collapse
MESH Headings
- Endocannabinoids/pharmacology
- Animals
- Arachidonic Acids/pharmacology
- Anti-Inflammatory Agents/pharmacology
- Polyunsaturated Alkamides/pharmacology
- Mice
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Mice, Knockout
- Humans
- Mice, Inbred C57BL
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Male
- Cells, Cultured
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
Collapse
Affiliation(s)
- Tom Teichmann
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Beatrice Pflüger-Müller
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Virna Margarita Martín Giménez
- Universidad Católica de Cuyo, Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, San Juan, Argentina
| | - Fiona Sailer
- Goethe University Frankfurt, Institute of Biochemistry I, Faculty of Medicine, Frankfurt, Germany
| | - Henrik Dirks
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Simonida Zehr
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Timothy Warwick
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Felix Brettner
- Goethe University Frankfurt, Institute of Pharmaceutical Technology, Frankfurt am Main, Germany
| | - Paola Munoz-Tello
- Vanderbilt University, Department of Biochemistry, Nashville, Tennessee, United States
| | - Andreas Zimmer
- University of Bonn, Institute of Molecular Psychiatry, Medical Faculty, Bonn, Germany
| | - Irmgard Tegeder
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
| | - Dominique Thomas
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Robert Gurke
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Stefan Günther
- Max-Plank-Institute for Heart- and Lung research (MPI-HLR), Bad Nauheim, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Ewgenij Proschak
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
- Goethe University Frankfurt, Institute of Pharmaceutical Chemistry, Frankfurt, Germany
| | - Gerd Geisslinger
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
| | - Iris-S. Bibli
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
- Goethe University Frankfurt, Institute for Vascular Signaling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Dagmar Meyer zu Heringdorf
- Goethe University Frankfurt, Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Frankfurt, Germany
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Mendoza 5500, Argentina
- Universidad Nacional de Cuyo, Departamento de Patología, Área de Farmacología, Facultad de Ciencias Médicas, Mendoza 5500, Argentina
| | - Maike Windbergs
- Goethe University Frankfurt, Institute of Pharmaceutical Technology, Frankfurt am Main, Germany
| | - Stefan Knapp
- Goethe University Frankfurt, Institute of Pharmaceutical Chemistry, Frankfurt, Germany
- Goethe University Frankfurt, Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Frankfurt am Main, Germany
| | - Andreas Weigert
- Goethe University Frankfurt, Institute of Biochemistry I, Faculty of Medicine, Frankfurt, Germany
| | - Matthias S. Leisegang
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Douglas Kojetin
- Vanderbilt University, Department of Biochemistry, Nashville, Tennessee, United States
- Vanderbilt University, Center for Structural Biology and Institute for Chemical Biology, Nashville, Tennessee, United States
| | - Ralf P. Brandes
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| |
Collapse
|
94
|
Jin S, Li T, Liu L, Gao T, Zhang T, Yuan D, Di J, Guo Z, Luo Z, Yuan H, Liu J. V-domain immunoglobulin suppressor of T-cell activation and programmed death receptor 1 dual checkpoint blockade enhances antitumour immunity and survival in glioblastoma. Br J Pharmacol 2025; 182:1306-1323. [PMID: 39626657 DOI: 10.1111/bph.17404] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/19/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The current therapy cannot meet the needs of glioblastoma (GBM). V-domain immunoglobulin suppressor of T-cell activation (VISTA) is significantly up-regulated in GBM patients; however, its therapeutic potential in GBM is still unclear. EXPERIMENTAL APPROACH Flow cytometry was used to detect the expression of VISTA and the co-expression pattern of VISTA and programmed death receptor 1 (PD-1) on brain infiltrating lymphocytes of GBM mice. Monoclonal antibody therapy was used to evaluate the therapeutic effect of α-VISTA monotherapy and α-VISTA combined with α-PD-1 on GBM mice. Transcriptome analysis, flow cytometry, and immunofluorescence were used to detect changes of immune microenvironment in mouse brain tumours. Immunofluorescence and TCGA data analysis were used to further validate the combined treatment strategy on patient data. KEY RESULTS Compared with normal mice, the frequency of VISTA expression and co-expression of VISTA and PD-1 on tumour-infiltrating lymphocytes (TILs) in tumour-bearing mice was increased. Anti-VISTA monotherapy significantly up-regulated multiple immune stimulation-related pathways and moderately prolonged mouse survival time. Blocking the immune checkpoint VISTA and PD-1 significantly prolonged the survival time of mice and cured about 80% of the mice; CD8+ T cells played an important role in this process. In addition, we found that the expression of VISTA and PD-1 was significantly up-regulated in GBM patients by immunofluorescence, and patients with high expression of VISTA and PD-1 were associated with poor overall survival. This combination of blocking the immune checkpoint VISTA and PD-1 may achieve clinical transformation in GBM.
Collapse
Affiliation(s)
- Shasha Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tao Li
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Liu Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ting Gao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Dingyi Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianwen Di
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhanying Guo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhijie Luo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoliang Yuan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
95
|
Zhang X, Zhou Y, Wang C, Ren J, Wang Y, Liu P, Feng W, Li X, Qi M, Yang Y, Zhu C, Wang F, Ma Y, Tang Z, Yu G. S1P/S1PRs-TRPV4 axis is a novel therapeutic target for persistent pain and itch in chronic dermatitis. Br J Pharmacol 2025; 182:1223-1235. [PMID: 39581852 DOI: 10.1111/bph.17393] [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: 12/12/2023] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND AND PURPOSE While pain and itch are both commonly associated with chronic dermatitis (CD), the molecular mechanisms underlying these debilitating symptoms is not well understood. This study aims to identify novel, endogenous compounds that mediate CD-associated pain and itch. EXPERIMENTAL APPROACH Lesional skin of CD model mice was examined using unbiased metabolomic analysis to identify candidate pain or itch inducing compounds in CD. Sphingosine-1-phosphate (S1P) concentration in CD model skin was analysed using UPLC/MS/MS. Behaviour, calcium imaging and immunofluorescence staining were used to determine the pain and itch effects and mechanisms of the identified CD-related compounds. KEY RESULTS In the lesional skin of CD model mice, 136 compounds were significantly changed. These compounds are predominately associated with the sphingolipids metabolism pathway. S1P is significantly increased in the lesional skin . The TRPV4 channel was critical for S1P induced itch and pain. Sphingosine kinase 2 (SPHK2), the key enzyme controlling S1P synthesis, was significantly increased in lesional skin. ABC294640, a SPHK2 inhibitor, significantly decreased S1P concentration in lesional CD model skin, as well as in model associated epidermal hyperplasia and chronic pain and itch. In CD patients, SPHK2 expression and S1P concentration were significantly elevated compared to healthy control skin. CONCLUSION AND IMPLICATIONS Our results indicate that, in CD, increased S1P induces chronic pain and itch partly through TRPV4. Inhibition of S1P synthesis or the S1P/S1P receptor-TRPV4 pathway are promising treatment strategies for CD-associated pain and itch.
Collapse
Affiliation(s)
- Xinyu Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan Zhou
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Changming Wang
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiahui Ren
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yin Wang
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pei Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weimeng Feng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xue Li
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingxin Qi
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Yang
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chan Zhu
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Wang
- Department of Dermatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuxiang Ma
- School of Life Science, China Pharmaceutical University, Nanjing, China
| | - Zongxiang Tang
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guang Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
96
|
Bompierre S, Byelyayeva Y, Mota E, Lefevre M, Pumo A, Kehler J, Castro LRV, Vincent P. Cross-pathway integration of cAMP signals through cGMP and calcium-regulated phosphodiesterases in mouse striatal cholinergic interneurons. Br J Pharmacol 2025; 182:1236-1253. [PMID: 39604216 DOI: 10.1111/bph.17400] [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: 02/05/2024] [Revised: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND AND PURPOSE Acetylcholine plays a key role in striatal function. Firing properties of striatal cholinergic interneurons depend on intracellular cAMP through the regulation of Ih currents. Yet, the dynamics of cyclic nucleotide signalling in these neurons have remained elusive. EXPERIMENTAL APPROACH We used highly selective FRET biosensors and pharmacological compounds to analyse the functional contribution of phosphodiesterases in striatal cholinergic interneurons in mouse brain slices. KEY RESULTS PDE1A, PDE3A and PDE4 appear as the main controllers of cAMP levels in striatal cholinergic interneurons. The calcium signal elicited through NMDA or metabotropic glutamate receptors activates PDE1A, which degrades both cAMP and cGMP. Interestingly, the nitric oxide/cGMP pathway amplifies cAMP signalling via PDE3A inhibition-a mechanism hitherto unexplored in a neuronal context. CONCLUSIONS AND IMPLICATIONS The expression pattern of specific PDE enzymes in striatal cholinergic interneurons, by integrating diverse intracellular pathways, can adjust cAMP responses bidirectionally. These properties eventually allow striatal cholinergic interneurons to dynamically regulate their overall activity and modulate acetylcholine release. Remarkably, this effect is the opposite of the cGMP-induced inhibition of cAMP signals involving PDE2A in striatal medium-sized spiny neurons, which provides important insights for the understanding of signal integration in the striatum.
Collapse
Affiliation(s)
- Ségolène Bompierre
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | | | - Elia Mota
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Marion Lefevre
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Anna Pumo
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | | | - Liliana R V Castro
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Pierre Vincent
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
97
|
Tang C, Tang C, Zhu X, Wang S, Yang Y, Miao Y, Zhao X, Jia L, Yang J, Su Y, Wang L, Wu C. Loss of AXIN1 regulates response to lenvatinib through a WNT/KDM5B/p15 signalling axis in hepatocellular carcinoma. Br J Pharmacol 2025; 182:1394-1409. [PMID: 39653061 DOI: 10.1111/bph.17413] [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: 04/02/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE As a highly heterogeneous cancer, hepatocellular carcinoma (HCC) shows different response rates to the multi-kinase inhibitor lenvatinib. Thus, it is important to explore genetic biomarkers for precision lenvatinib therapy in HCC. EXPERIMENTAL APPROACH The effect and mechanism of AXIN1 mutation on HCC were revealed by cell proliferation assay, long-term clone formation assay, sphere formation assay and small molecule inhibitor library screening. A new therapeutic strategy targeting HCC with AXIN1 mutation was evaluated in humanized models (patient-derived xenograft [PDX] and patient-derived organoid [PDO]). KEY RESULTS Based on The Cancer Genome Atlas (TCGA) data, we screened 6 most frequently lost tumour suppressor genes in HCC (TP53, ARID1A, AXIN1, CDKN2A, ARID2 and PTEN) and identified AXIN1 as the most crucial gene for lenvatinib sensitivity. Further study showed that AXIN1-knockout HCC cells had a more malignant phenotype and lower sensitivity to lenvatinib in vitro and in vivo. Mechanistically, the WNT pathway and its target gene c-Myc were activated when AXIN1 was missing, and the expression of tumour suppressor p15 was inhibited by transcription co-repressors c-Myc and Miz-1, resulting in the exacerbation of the resistant phenotype. Screening of a library of epigenetic-related enzyme inhibitors showed that a KDM5B inhibitor up-regulated p15 expression, leading to increased sensitivity to lenvatinib in vitro and in vivo. CONCLUSION AND IMPLICATIONS AXIN1-deficient patients have a lower response to lenvatinib, which may be associated with suppression of p15 mediated by WNT pathway activation. KDM5B inhibitors can restore p15 levels, resulting in efficient killing of resistant cells in HCC.
Collapse
MESH Headings
- Xenograft Model Antitumor Assays
- Organoids
- Tumor Cells, Cultured
- Primary Cell Culture
- Axin Protein/genetics
- Axin Protein/metabolism
- Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors
- Jumonji Domain-Containing Histone Demethylases/metabolism
- Wnt Proteins/metabolism
- Cyclin-Dependent Kinase Inhibitor p15/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Precision Medicine/methods
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Humans
- Animals
- Mice
- Genes, Tumor Suppressor
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic/drug effects
- Male
- Mice, Inbred BALB C
- RNA-Seq
- Loss of Function Mutation
- Down-Regulation
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Drug Synergism
- Adult
- Middle Aged
Collapse
Affiliation(s)
- Chengfang Tang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Chu Tang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Xuanchi Zhu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Simeng Wang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Yuan Yang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Yu Miao
- Clinical Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoyao Zhao
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Lina Jia
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Jingyu Yang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Yang Su
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lihui Wang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| | - Chunfu Wu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, China
| |
Collapse
|
98
|
Aranäs C, Caffrey A, Edvardsson CE, Vestlund J, Schmidt HD, Jerlhag E. Synergistic-like decreases in alcohol intake following combined pharmacotherapy with GLP-1 and amylin in male rats. Br J Pharmacol 2025; 182:1292-1305. [PMID: 39622492 DOI: 10.1111/bph.17406] [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: 07/11/2024] [Revised: 10/08/2024] [Accepted: 10/28/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The limited effectiveness of current pharmacological treatments for alcohol use disorder (AUD) highlights the need for novel therapies. These may involve the glucagon-like peptide-1 receptor or the amylin receptor, as treatment with agonists targeting either of these receptors lowers alcohol intake. The complexity of the mechanisms underlying AUD indicates that combining agents could enhance treatment efficacy. While a combination of amylin receptor and GLP-1 receptor agonists reduced food intake and body weight synergistic-like, its influence on alcohol intake is unknown. EXPERIMENTAL APPROACH Effects of a range of dose-combinations of GLP-1 receptor (dulaglutide) and amylin receptor (salmon calcitonin; sCT) agonists on alcohol intake were explored in male and female rats. We used dose combinations that either lowered alcohol intake as monotherapy (0.1 mg·kg-1 + 5 μg·kg-1), or that did not affect alcohol consumption per se (0.075 mg·kg-1 + 2 μg·kg-1). KEY RESULTS Acute administration of dulaglutide and sCT (0.1 mg·kg-1 + 5 μg·kg-1) reduced alcohol intake in males, but not in females. When higher doses were evaluated in female rats, a decrease in alcohol intake was observed. Furthermore, the low dose combination (0.075 mg·kg-1 + 2 μg·kg-1) decreased, in in a synergistic-like manner, alcohol intake and prevented abstinence-induced drinking without affecting kaolin intake in males. However, tolerance developed during sub-chronic treatment. CONCLUSION AND IMPLICATIONS Collectively, these findings show that the combination of dulaglutide and sCT decreased, in in a synergistic-like manner, alcohol consumption in male rats. Contrarily, higher doses are required for females.
Collapse
Affiliation(s)
- Cajsa Aranäs
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Antonia Caffrey
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christian E Edvardsson
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jesper Vestlund
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
99
|
Caffino L, Targa G, Mottarlini F, Thielens S, Rizzi B, Villers A, Ris L, Gainetdinov RR, Leo D, Fumagalli F. Memantine-induced functional rewiring of the glutamate synapse in the striatum of dopamine transporter knockout rats. Br J Pharmacol 2025; 182:1377-1393. [PMID: 39653030 DOI: 10.1111/bph.17403] [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: 06/11/2024] [Revised: 09/27/2024] [Accepted: 10/29/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Slow-acting biogenic amines, such as dopamine, are known to modulate fast neurotransmitters e.g. glutamate. In the striatum, dopamine (DA) interacts with glutamate, influencing neural excitability and promoting synaptic plasticity. The exact mechanism of such interaction is not fully understood. This study investigates, in detail, how dopamine overactivity in dopamine transporter knockout (DAT-/-) rats, alters the homeostasis of the striatal glutamate synapse from a molecular, behavioural and functional point of view. EXPERIMENTAL APPROACH The expression, localisation, retention and electrophysiological properties of N-methyl-D-aspartate (NMDA) receptors as well as dendritic spine density and morphology were investigated in the striatum of DAT-/- rats, at baseline and after treatment with the non-competitive NMDA receptor antagonist memantine (30 mg kg-1). KEY RESULTS Dopamine overactivity dramatically reorganises the striatal glutamate synapse, redistributing NMDA receptors in the synapse as typified by reduced synaptic availability and reduced expression of NMDA scaffolding proteins, as well as by increased GluN2B-containing NMDA receptors in the extra synapse. Such changes are accompanied by reduced spine density, suggesting dopamine-induced structural rearrangements. These results converge into a compromised plasticity, as shown by the impaired ability to promote long-term depression (LTD) in the striatum of DAT-/-rats. Notably, memantine counteracts hyperlocomotion, reverses spine alterations and abolishes the extrasynaptic movements of NMDA receptors in the striatum of DAT-/- rats, thus restoring functional LTD. CONCLUSION AND IMPLICATIONS A hyperdopaminergic condition seems to alter striatal homeostasis by increasing extrasynaptic NMDA receptors. These findings may be relevant to manipulate disorders characterised by elevated dopaminergic activity.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Sarah Thielens
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Beatrice Rizzi
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
- Center for Neuroscience, University of Camerino, Camerino, Italy
| | - Agnes Villers
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Laurence Ris
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, St. Petersburg, Russia
| | - Damiana Leo
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
100
|
Jedlina L, Paramel G, Soboleva S, Chutna Olin O, Haug M, Fransén K, Lindstam M, Brewinska-Olchowik M, Piwocka K, Grenegård M. Antithrombotic but not anticoagulant activity of the thrombin-binding RNA aptamer Apta-1. Br J Pharmacol 2025; 182:1358-1376. [PMID: 39653034 DOI: 10.1111/bph.17382] [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: 12/18/2023] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Pharmacological intervention of thrombosis is challenging, requiring a fined tune balance between beneficial antithrombotic effect versus risk of major bleeding complications. In this investigation, we elucidated the antithrombotic capacity of the novel 90-mer RNA aptamer Apta-1 and its underlying mechanism of action. EXPERIMENTAL APPROACH We utilized three independent in vivo animal models to establish antithrombotic activity and bleeding risk of Apta-1. Several cellular and molecular techniques were utilized to extensively characterize the effects of Apta-1 on primary and secondary haemostasis. KEY RESULTS Apta-1 significantly reduced thrombus weight in ferric chloride-induced carotid artery thrombosis. A consistent reduction in thrombus weight was also observed in arteriovenous shunt thrombosis in rats, whereas tail bleeding time was unaffected. Cellular and molecular analyses revealed that Apta-1 interacted with thrombin, resulting in significant inhibition of protease-activated receptor (PAR) signalling in platelets. On the other hand, Apta-1 shortened both thrombin generation and thrombin-induced clotting times. CONCLUSIONS AND IMPLICATIONS Apta-1 targets the heparin-binding motif exosite II on thrombin leading to significant suppression of platelet PAR1 and PAR4 signalling. Intriguingly, Apta-1 produces substantial antithrombotic activity without anticoagulant or general antiplatelet properties. In fact, we found that Apta-1 accelerates the formation of blood clots and thus supports haemostasis without exhibiting typical anticoagulant properties. We suggest that Apta-1 may be a promising future drug candidate for treatment of thrombosis in diseases/conditions where there are significant risks of serious bleeding complications.
Collapse
Affiliation(s)
| | - Geena Paramel
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | | | | | - Marianne Haug
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Karin Fransén
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | | | | | - Katarzyna Piwocka
- Nencki Institute of Experimental Biology Polish Academy of Science, Warsaw, Poland
| | - Magnus Grenegård
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|