1
|
Yang Y, Ma K, Li S, Xiong T. Multifaceted role of nitric oxide in vascular dementia. Med Gas Res 2025; 15:496-506. [PMID: 40300885 DOI: 10.4103/mgr.medgasres-d-24-00158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 05/01/2025] Open
Abstract
Vascular dementia is a highly heterogeneous neurodegenerative disorder induced by a variety of factors. Currently, there are no definitive treatments for the cognitive dysfunction associated with vascular dementia. However, early detection and preventive measures have proven effective in reducing the risk of onset and improving patient prognosis. Nitric oxide plays an integral role in various physiological and pathological processes within the central nervous system. In recent years, nitric oxide has been implicated in the regulation of synaptic plasticity and has emerged as a crucial factor in the pathophysiology of vascular dementia. At different stages of vascular dementia, nitric oxide levels and bioavailability undergo dynamic alterations, with a marked reduction in the later stages, which significantly contributes to the cognitive deficits associated with the disease. This review provides a comprehensive review of the emerging role of nitric oxide in the physiological and pathological processes underlying vascular dementia, focusing on its effects on synaptic dysfunction, neuroinflammation, oxidative stress, and blood‒brain barrier integrity. Furthermore, we suggest that targeting the nitric oxide soluble guanylate cyclase-cyclic guanosine monophosphate pathway through specific therapeutic strategies may offer a novel approach for treating vascular dementia, potentially improving both cognitive function and patient prognosis. The review contributes to a better understanding of the multifaceted role of nitric oxide in vascular dementia and to offering insights into future therapeutic interventions.
Collapse
Affiliation(s)
- Yi Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Kangrong Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Shun Li
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tianqing Xiong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Key Laboratory of the Jiangsu Higher Education Institutions for Integrated Traditional Chinese and Western Medicine in Senile Diseases Control (Yangzhou University), Yangzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Cho H, Ju H, Ahn Y, Jang J, Cho J, Park E, Kang SM, Lee J, Seo D, Baek MC, Yea K. Engineered extracellular vesicles with surface FGF21 and enclosed miR-223 for treating metabolic dysfunction-associated steatohepatitis. Biomaterials 2025; 321:123321. [PMID: 40209593 DOI: 10.1016/j.biomaterials.2025.123321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disorder with a complex pathogenesis that requires combination therapies rather than monotherapies. Extracellular vesicles (EVs) exhibit inherently efficient delivery to the liver and can be engineered to carry various therapeutic substances, making them promising agents. In this study, EVs were engineered to display fibroblast growth factor 21 (FGF21) on their surface and encapsulate miR-223 (223/F-EVs), aiming to improve steatosis and alleviate inflammation and fibrosis, respectively. Introducing the 223/F-EVs into human liver cell lines significantly reduced both basal and induced levels of lipid storage, inflammation, and fibrosis markers. Furthermore, using an FGF21-blocking antibody or miR-223 inhibitor effectively diminished the efficacy of the 223/F-EVs, confirming the essential roles of FGF21 and miR-223 in these processes. In a Choline-Deficient, l-Amino acid-defined, High-Fat Diet (CDAHFD)-fed mouse model, intravenously administered 223/F-EVs demonstrated liver-preferential delivery and a marked reduction in the MASH phenotype without compromising bone density, unlike conventional FGF21 treatment. Collectively, 223/F-EVs convey FGF21 and miR-223 exclusively to the liver, offering strategic advantages by mitigating MASH progression via multiple pathways. This study lays a solid foundation for further investigation of engineered EVs as a transformative therapeutic approach for treating MASH.
Collapse
Affiliation(s)
- Hanchae Cho
- Department of Biomedical Science, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Hyunji Ju
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Yongdeok Ahn
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhee Jang
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhyeong Cho
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Eunju Park
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Sung-Min Kang
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea.
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea; New Biology Research Center, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 43024, Republic of Korea.
| |
Collapse
|
3
|
Liu Y, Song W, Dong W, Gong X, Dong C, Zhao J, Wang R, Song S, Shuang S. Preparation of mitochondrial targeted near-infrared ratio fluorescent probe and its dual response detection for viscosity and ONOO - and cell imaging. Talanta 2025; 292:127909. [PMID: 40081248 DOI: 10.1016/j.talanta.2025.127909] [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: 11/16/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
The changes in viscosity and the concentration of ONOO- in mitochondria can effectively reflect the physiological and pathological status of cells. Therefore, the development of effective fluorescent probes for the sensing of viscosity and the concentration of ONOO- in mitochondria has great significance. In this article, a mitochondrial targeted fluorescent probe named Mito-RP was synthesized for the dual responsive sensing of viscosity and ONOO- by introducing pyridine ring and phenylboronic acid ester structure into 4-dimethylamino-cinnamaldehyde with long conjugated chain structure as the parent material. Mito-RP exhibits 600 folds fluorescence enhancement of viscosity in the red-light channel at 700 nm, with pyridine cation as the mitochondrial anchoring group. Simultaneously, Mito-RP appears excellent selectivity towards ONOO- using boronic acid esters as response sites. A new ratio fluorescence analysis method was constructed based on the linear correlation between the emission intensity ratio of Mito-RP at 616 nm/700 nm and the concentration of ONOO-. The linear range is 0.05-33 μM and the detection limit is 9.2 nM. Meanwhile, Mito-RP successfully monitored the changes in viscosity during lipopolysaccharide induced inflammation and rapamycin induced mitochondrial autophagy in HeLa cells. In addition, Mito-RP has also achieved visual imaging of intracellular exogenous/endogenous ONOO-. These studies provide a novel method for in-depth investigation of mitochondrial function and its role in diseases.
Collapse
Affiliation(s)
- Yang Liu
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenqiang Song
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenjuan Dong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Xiaojuan Gong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Chuan Dong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Jie Zhao
- Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Shanxi Provincial Integrated TCM and WM Hospital, Taiyuan, 030013, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau
| | - Shengmei Song
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China.
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
4
|
Jiang W, Liu L, Li W, Liu H, Yang J, Wang P. A lysosomal-targeted switchable fluorescent probe for the detection of peroxynitrite in living tumor cells and in vivo. Talanta 2025; 291:127866. [PMID: 40037163 DOI: 10.1016/j.talanta.2025.127866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Peroxynitrite (ONOO-) is a reactive nitrogen species whose abnormal accumulation in the body can lead to various diseases, including those related to oxidative stress. Accurate detection of ONOO- levels is essential for the diagnosis and treatment of these diseases. To address this need, we developed a lysosome-targeted fluorescent probe Lyso-PE for detecting ONOO- in tumors. In the presence of ONOO-, probe Lyso-PE showed a large Stokes shift of 100 nm. The probe exhibited high sensitivity, selectivity, and rapid response toward ONOO-. Additionally, Lyso-PE displayed excellent lysosomal targeting and was successfully employed in imaging the exogenous peroxynitrite in tumor cells. In the 4T1 subcutaneous graft tumor model, the probe could effectively distinguish tumors and normal tissues with the help of fluorescence imaging in vivo. Moreover, Lyso-PE could be used for tumor resection guided by fluorescent signals in vivo. These results suggested that Lyso-PE could enhance our understanding of lysosomal function in disease, identify new therapeutic targets, and aid in developing new diagnostic and therapeutic strategies with significant clinical implications.
Collapse
Affiliation(s)
- Wen Jiang
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Li Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Wenqing Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Huijia Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Jing Yang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China.
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
5
|
Zhou X, Wang J, Li C, Zheng L, Wang H, Sun L. Exploration of material basis: Chemical composition profile and metabolic profile in Xiao Jianzhong Granules. J Pharm Biomed Anal 2025; 260:116793. [PMID: 40068234 DOI: 10.1016/j.jpba.2025.116793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 04/06/2025]
Abstract
Xiao Jianzhong Granules (XJZG), a well-known traditional prescription with protective effects on the gastric mucosa, as documented in the Treatise on Typhoid and Miscellaneous Diseases. Clinical studies have proven that XJZG exhibits significant anti-colitis properties and effectively alleviates duodenal ulcers. However, despite its clinical popularity, comprehensive studies on its chemical composition and in vivo metabolism remain limited. In the present study, gas chromatography coupled with mass spectrometry (GC-MS) and ultra-high-performance liquid chromatography coupled with Q-Exactive Orbitrap mass spectrometry (UHPLC Q-Exactive Orbitrap MS) were used to analyze the chemical composition of XJZG, while it is in vivo metabolic profile was further assessed with UHPLC Q-Exactive Orbitrap MS. Additionally, ultra-high-performance liquid chromatography coupled with triple quadrupole mass spectrometry (UHPLC-MS/MS) was utilized to quantify its primary components. As a result, a total of 51 volatiles were characterized by GC-MS, and 139 compounds were characterized by UHPLC Q-Exactive Orbitrap MS in vitro. In addition, 51 prototype components and 133 metabolites were characterized in vivo. Notably, 5 new compounds were discovered in this process. The main metabolic reactions included oxidation, reduction, hydrolysis, glucuronidation, and sulfate esterification. In quantitative analysis, 17 components were determined and successfully applied for detection by UPLC-MS/MS in multiple reaction monitoring mode. The quantitative methods were validated and met the requirements. Through multivariate statistical analysis, 6 components were selected as potential quality markers for XJZG based on PCA and OPLS-DA. Additionally, our study provides supplementary chemical evidence to further elucidate the material basis of XJZG.
Collapse
Affiliation(s)
- Xuanxuan Zhou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jiaxue Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Caihong Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Ling Zheng
- Pharmacy Department, Fushun Eye Hospital, Fushun, PR China
| | - Hongjin Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China.
| | - Lixin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China.
| |
Collapse
|
6
|
Zhai P, Jiang Y, Hu Z, Guo Y, Zhang H. m6A reader YTHDC1 mediates MAFF nuclear export to induce VMP1 transcription and alleviate I/R-induced oxidative stress injury in hepatocytes. Cell Signal 2025; 131:111719. [PMID: 40054588 DOI: 10.1016/j.cellsig.2025.111719] [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: 01/07/2025] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Hepatic ischemia/reperfusion (I/R) injury occurs after liver resection surgery, trauma, shock, and transplantation. This study aimed to identify and characterize the role of the YTH domain-containing protein 1 (YTHDC1)/MAFF/vacuole membrane protein 1 (VMP1) axis in hepatic I/R injury. YTHDC1, MAFF, and VMP1 were significantly overexpressed in the hepatic tissues of mice with I/R and hepatocytes exposed to hypoxia-reoxygenation (H/R). Knockdown of MAFF exacerbated oxidative stress and inflammatory injury in mice induced with hepatic I/R, which were reversed by overexpression of VMP1. Similarly, I/R-associated injury mitigated by YTHDC1 overexpression was reversed by MAFF knockdown. Mechanistically, YTHDC1 mediated the nuclear export and stability of MAFF mRNA and promoted MAFF translation. Collectively, the findings establish that YTHDC1-mediated m6A-dependent MAFF expression determines hepatocyte oxidative stress via VMP1, providing valuable insights into the potential mechanisms underlying hepatic I/R injury and offering potential therapeutic strategies for its treatment.
Collapse
Affiliation(s)
- Peng Zhai
- Department of General Surgery, The Fifth People's Hospital of Huai'an (Huai'an Hospital Affiliated to Yangzhou University), Huai'an 223000, Jiangsu, PR China.
| | - Yongjun Jiang
- Department of General Surgery, The Fifth People's Hospital of Huai'an (Huai'an Hospital Affiliated to Yangzhou University), Huai'an 223000, Jiangsu, PR China
| | - Zhifeng Hu
- Department of General Surgery, The Fifth People's Hospital of Huai'an (Huai'an Hospital Affiliated to Yangzhou University), Huai'an 223000, Jiangsu, PR China
| | - Yunhu Guo
- Department of General Surgery, The Fifth People's Hospital of Huai'an (Huai'an Hospital Affiliated to Yangzhou University), Huai'an 223000, Jiangsu, PR China
| | - Huaguo Zhang
- Department of General Surgery, The Fifth People's Hospital of Huai'an (Huai'an Hospital Affiliated to Yangzhou University), Huai'an 223000, Jiangsu, PR China.
| |
Collapse
|
7
|
Wang FI, Dixon SJ, Chidiac P. Extracellular ATP and structurally related molecules potentiate adenosine A 2a receptor-stimulated cAMP production. Cell Signal 2025; 131:111711. [PMID: 40044016 DOI: 10.1016/j.cellsig.2025.111711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/18/2025] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Extracellular ATP has been reported to potentiate signalling by several Class B G protein-coupled receptors (GPCRs). The adenosine A2a receptor (A2aR) is a Class A GPCR that regulates many physiological processes, and a potential therapeutic target for many diseases. In vivo, A2aR is exposed transiently to extracellular ATP within the cellular microenvironment under both physiological and pathological conditions. The modulating effects of extracellular ATP seen with Class B GPCRs have not previously been investigated in other classes of GPCRs. In the present study, we investigated the effects of extracellular ATP on A2aR signalling. We also studied the actions of similar molecules to explore the structure-activity relationship. Cyclic 3',5'-adenosine monophosphate (cAMP) levels were monitored following agonist-induced receptor activation in cells co-transfected with plasmids encoding A2aR and a luminescent cAMP biosensor. Extracellular ATP increased the potency of both adenosine and selective A2aR agonists by approximately an order of magnitude. In the absence of agonist, ATP did not activate A2aR, arguing against an effect due to ATP metabolism to adenosine. The potentiating effect of ATP was mimicked by other nucleotides and similarly by phosphorylated sugars. Non-phosphorylated sugars produced comparable effects, but higher concentrations were required to do so. This difference in potency implies that the phosphate group is important for modulating A2aR activity. Here, we present the first evidence that A2aR can be positively modulated by extracellular ATP, thus the effect of ATP is not limited to Class B GPCRs.
Collapse
Affiliation(s)
- Fang I Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada
| | - S Jeffrey Dixon
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada; Bone and Joint Institute, The University of Western Ontario, London, Canada
| | - Peter Chidiac
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada; Bone and Joint Institute, The University of Western Ontario, London, Canada.
| |
Collapse
|
8
|
Geng S, Zhou Y, Ng G, Fan Q, Cheong S, Mazur F, Boyer C, Chandrawati R. Selenium nanoparticles as catalysts for nitric oxide generation. Colloids Surf B Biointerfaces 2025; 251:114592. [PMID: 40024109 DOI: 10.1016/j.colsurfb.2025.114592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
The critical role of nitric oxide (NO), a potent signalling molecule, in various physiological processes has driven the development of NO delivery strategies for numerous therapeutic applications. However, NO's short half-life poses a significant challenge for its effective delivery. Glutathione peroxidase, a selenium-containing antioxidant enzyme, can catalyse the decomposition of S-nitrosothiols (endogenous NO prodrugs) to produce NO in situ. Inspired by this, we explored selenium nanoparticles (SeNPs) for their enzyme-mimicking NO-generating activity. Stabilised with polyvinyl alcohol (PVA) or chitosan (CTS), SeNPs demonstrated tuneable NO generation when exposed to varying concentrations of NO prodrug, nanoparticles, and glutathione (GSH). In the presence of GSH, a naturally occurring antioxidant in the human body, 0.1 µg mL-1 of SeNPs could catalytically generate 7.5 µM of NO under physiological conditions within 30 min. We investigated the effects of nanoparticle crystallinity and NO prodrug type on NO generation, as well as the stability and sustained NO generation of the catalytic nanoparticles. PVA-stabilised SeNPs were non-toxic to NIH 3T3 cells and effectively dispersed Pseudomonas aeruginosa biofilms upon NO generation. This study broadens the repertoire of nanomaterials for NO generation and highlights SeNPs as a non-toxic alternative for therapeutic NO delivery.
Collapse
Affiliation(s)
- Shu Geng
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Yingzhu Zhou
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Gervase Ng
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia; Cluster for Advanced Macromolecular Design (CAMD), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Qingqing Fan
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Soshan Cheong
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Federico Mazur
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Cyrille Boyer
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia; Cluster for Advanced Macromolecular Design (CAMD), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Rona Chandrawati
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| |
Collapse
|
9
|
Chen T, Zhang H, Shan W, Zhou J, You Y. Liver sinusoidal endothelial cells in hepatic fibrosis: opportunities for future strategies. Biochem Biophys Res Commun 2025; 766:151881. [PMID: 40286764 DOI: 10.1016/j.bbrc.2025.151881] [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: 03/12/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells that form the interface between the hepatic vasculature and parenchymal cells, playing a crucial role in maintaining hepatic homeostasis. Under pathological conditions, LSECs undergo capillarization, marked by the loss of fenestrae and formation of a basement membrane, thereby impairing microcirculation and promoting fibrosis. Beyond capillarization, LSECs experience a spectrum of pathological changes-including angiogenesis, endothelial-to-mesenchymal transition (EndMT), autophagy, and senescence-all of which contribute to fibrogenesis through distinct molecular pathways. Moreover, LSECs orchestrate liver fibrotic remodeling through dynamic crosstalk with hepatic stellate cells (HSCs), hepatocytes, Kupffer cells, and immune cells, exerting both pro- and anti-fibrotic effects. This review comprehensively summarizes LSECs dysfunction in hepatic fibrosis, with a particular focus on intercellular communication and emerging therapeutic strategies. Elucidating the regulatory networks that govern LSECs behavior may uncover new opportunities for the diagnosis and treatment of chronic liver disease.
Collapse
Affiliation(s)
- Ting Chen
- Department of human anatomy, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Huan Zhang
- Department of human anatomy, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Wenqi Shan
- Department of human anatomy, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China.
| | - Yanwen You
- Department of human anatomy, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China.
| |
Collapse
|
10
|
Bilquees A, Saleem D, Aamir M. Constructive feedback on the link between depression and acute myocardial infarction: A response to recent findings. Int J Cardiol 2025; 429:133156. [PMID: 40088952 DOI: 10.1016/j.ijcard.2025.133156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Affiliation(s)
- Adeeba Bilquees
- School of Architecture, VIT Bhopal University, Sehore, India.
| | - Dauood Saleem
- School of Computing Science and Engineering, VIT Bhopal University, Sehore, India.
| | - Mohd Aamir
- School of Architecture, VIT Bhopal University, Sehore, India.
| |
Collapse
|
11
|
Wang P, Ding W, Mo J, Gu C, Ouyang S, Peng K, Zhang Q, Liu G, Lu J, Wang Y, Hu W, Zhu K, Zhang X. A novel adenosine 2A receptor antagonist HZ-086 enhances the efficiency of immunotherapy and alleviates the acquired resistance to PD-L1 by restoration of T cell functions. Eur J Pharmacol 2025; 997:177535. [PMID: 40118325 DOI: 10.1016/j.ejphar.2025.177535] [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: 11/02/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Immunotherapy faces significant challenges due to low clinical response rates and immune escape mechanisms, which ultimately lead to drug resistance. Previous studies suggest that adenosine-2A receptor (A2AR) signaling plays a critical role in immunosuppression and immune escape. However, no potent and selective A2AR inhibitors are currently available for clinical use to address immunotherapy resistance in tumors. In this study, we identified a novel small molecule compound, HZ-086, as a potent and selective inhibitor of A2AR. HZ-086 restored the activation of T-cell signaling which is suppressed by adenosine analogs in vitro. Additionally, HZ-086 enhanced T-cell-mediated cytotoxicity, increased the secretion of cytokines for antitumor and subsequently inhibited growth of tumor cells in vitro and in vivo. Furthermore, HZ-086 inhibited tumor growth, enhances anti-tumor capacity, and reversed PD-L1 resistance in vivo. When combined with FD-L1, a PD-L1 small molecule inhibitor discovered by our lab, HZ-086 achieved over 80 % tumor growth inhibition (TGI) and restored immune response in anti-PD-L1 monoclonal antibody-resistant tumors. This combination treatment also promoted the infiltration and activation of CD8+ T lymphocytes within the tumor microenvironment. Our findings demonstrate that adenosine-A2AR signaling mediates resistance to immunotherapy and discover a novel potent and selective A2AR inhibitor with high efficacy in enhancing antitumor immune responses and reversing PD-L1 resistance. The combination of A2AR inhibitor and PD-L1 inhibitor represents a promising therapeutic strategy for antitumor therapy.
Collapse
Affiliation(s)
- Pengyan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wen Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jianshan Mo
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chenxi Gu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shumin Ouyang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Keren Peng
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Qiyi Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guopin Liu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yandong Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Wenhao Hu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Kai Zhu
- Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xiaolei Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
12
|
Ito T, Suzuki T, Sakai Y, Nishioka K, Itoh Y, Sakamoto K, Ikemura N, Matoba S, Kanda Y, Takagi J, Okamoto T, Tahara K, Hoshino A. Engineered ACE2 decoy in dry powder form for inhalation: A novel therapy for SARS-CoV-2 variants. Mol Ther Methods Clin Dev 2025; 33:101459. [PMID: 40276779 PMCID: PMC12019485 DOI: 10.1016/j.omtm.2025.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
The persistent threat of SARS-CoV-2 and the emergence of new variants has prompted the development of a novel, easily administered modality that can overcome viral mutations. The engineered ACE2 decoy shows neutralizing activity comparable to monoclonal antibodies and is broadly effective against SARS-CoV-2 variants and ACE2-utilizing sarbecoviruses. In addition to intravenous administration, this decoy has shown antiviral efficacy through nebulized aerosol inhalation in murine and primate models, offering a dose-sparing advantage. Clinically, dry powder formulation is ideal for convenience and storage but poses challenges for protein biologics. This study developed a freeze-dried spray formulation of the ACE2 decoy for inhalation. The trehalose and leucine-based excipient maintained neutralizing activity and prevented aggregate formation. The dry powder showed aerodynamic distribution from bronchi to alveoli, aiding protection against SARS-CoV-2 infections. Neutralizing activity, structural stability, and powder dispersibility were preserved after 6 months of storage. In a mouse model of SARS-CoV-2 infection, significant reductions in viral replication and lung pathology were observed with intratracheal administration 24 h post-infection. The ACE2 decoy retained activity against recent JN.1 and current KP.3 strains, confirming its robust efficacy against viral mutations. This ACE2 decoy powder inhalant is a self-administered, next-generation treatment addressing the ongoing immune-evading evolution of SARS-CoV-2.
Collapse
Affiliation(s)
- Takaaki Ito
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Keisuke Nishioka
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kentarou Sakamoto
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Nariko Ikemura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kohei Tahara
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
- Laboratory of Nanofiber Technology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
13
|
Stomberg S, Rühle A, Wittrien T, Sandner P, Behrends S. Discovery of the first isoform-specific sGC activator: Selective activation of GC-1 by runcaciguat. Eur J Pharmacol 2025; 996:177557. [PMID: 40147578 DOI: 10.1016/j.ejphar.2025.177557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Drug research and development programmes targeting soluble guanylyl cyclase (sGC) have been highly successful, leading to the launch of the sGC stimulators riociguat for pulmonary hypertension (2013) and vericiguat for chronic heart failure (2021). As the main receptor for nitric oxide, sGC plays a vital role in various physiological processes. It consists of an alpha and a beta subunit, with two distinct isoforms identified in humans: GC-1 (α1/β1) and GC-2 (α2/β1). Growing evidence indicates that these isoforms engage in different downstream signalling pathways, indicating that isoform-specific approaches could lead to novel therapeutic opportunities and reduce potential side effects. In this study, we performed concentration-response measurements with the sGC activators BAY 60-2770, BI 703704 and runcaciguat (BAY 1101042) in cell systems expressing each isoform and in purified enzymes. We found that runcaciguat selectively activated GC-1, while acting as a competitive antagonist to other activators in GC-2, without interfering with nitric oxide. BAY 60-2770 and BI 703704 activated both isoforms, albeit with varying efficacy. Our findings challenge the historical view that the two sGC isoforms are functionally indistinguishable. In fact, we demonstrate that the isoforms can be activated independently, highlighting their distinct functional profiles. Notably, runcaciguat is the first sGC activator identified to selectively target GC-1 at therapeutic concentrations. This selective targeting of isoforms not only opens avenues for new therapeutic strategies but also provides an alternative to knockout animal models for investigating isoform-specific functions.
Collapse
Affiliation(s)
- Svenja Stomberg
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, Germany.
| | - Anne Rühle
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, Germany.
| | - Theresa Wittrien
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, Germany.
| | - Peter Sandner
- Bayer AG, Pharmaceuticals Drug Discovery, Institute of Cardiovascular and Renal Research, Wuppertal, Germany.
| | - Sönke Behrends
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, Germany; Semmelweiss University Budapest, Asklepios Campus Hamburg, Germany.
| |
Collapse
|
14
|
Mao Q, Zhang P, Lin F, Fu X, Zhang B, Liu C, Liu Z, Chen X, Dai X, Yue X, Shi X, Pang J, Wang S. Design, synthesis and biological evaluation of 2-[1-(pyridin-2-ylmethyl)-1H-pyrazole-3-carboxamido]benzoic acids as promising urate transporter 1 inhibitors with potential nephroprotective efficacy for the treatment of hyperuricemic nephropathy. Eur J Med Chem 2025; 290:117507. [PMID: 40101451 DOI: 10.1016/j.ejmech.2025.117507] [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: 01/05/2025] [Revised: 03/01/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
Hyperuricemic nephropathy (HN) is considered an important risk factor for mortality in patients with hyperuricemia. Reducing serum uric acid (UA) levels and mitigating kidney injury are essential components in the treatment of HN. Thus, UA-lowering drugs that can also protect the kidneys are urgently needed. We identified a urate transporter 1 (URAT-1) inhibitor, T29, with cytoprotective efficacy through screening an internal library against hyperuricemia using a UA-induced HK-2 cell injury model. A bioisosteric strategy was then employed to replace the indole core of T29 with pyrazole moieties; this resulted in a series of 2-[1-(pyridin-2-ylmethyl)-1H-pyrazole-3-carboxamido]benzoic acids. Among them, compound 18 demonstrated the best cytoprotective efficacy (cell viability = 92.2 % vs. model = 31.5 %), and the IC50 value of compound 18 against URAT-1 was 3.36 μM; both of these values exceeded T29. In an HN mice model induced by a 0.75 % adenine diet and intraperitoneal injection of potassium oxonate (400 mg/kg), compound 18 significantly reduced the serum UA levels by inhibiting URAT-1 activity. Furthermore, compound 18 improved kidney function by lowering serum creatinine (CRE) and urea nitrogen (BUN) levels while attenuating tubular dilation and inflammatory cell infiltration in the kidneys. Additionally, it suppressed the release of the proinflammatory cytokines IL-1β and TNF-α and reduced kidney fibrosis by downregulating the expression of α-SMA and TGF-β. In conclusion, compound 18 ameliorated HN by inhibiting URAT-1, alleviating immune-inflammatory responses and mitigating fibrosis; the results from this study demonstrate its potential as a therapeutic agent for HN.
Collapse
Affiliation(s)
- Qing Mao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Peng Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Fengwei Lin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xuefeng Fu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Bing Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Chang Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Ziyuan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xing Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xiwen Dai
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xiaoyi Yue
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xiang Shi
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Shaojie Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
15
|
Huang H, Xiong Y, Zeng J. Silencing RGS7 attenuates atrial fibrillation progression by activating the cGMP-PKG signaling pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167786. [PMID: 40086518 DOI: 10.1016/j.bbadis.2025.167786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/05/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Atrial fibrillation (AF) is a common diagnosed heart disease that needs novel managements. This study aimed to seek potential biomarkers and underlying regulatory pathways associated with AF. METHODS Differential expressed genes (DEGs) were identified from the Gene Expression Omnibus database, followed by a protein-protein interaction (PPI) network to discover hub genes. Principal components analysis (PCA) and receiver operating characteristic (ROC) curves were performed to evaluate the ability of hub genes to discriminate between AF and control. RGS7 was selected as a key hub gene, and genes co-expressed with RGS7 were identified for functional enrichment analysis. Further in vivo and in vitro experiments were conducted to investigate the effects of silencing RGS7 on AF and the potential pathway. RESULTS We identified top 5 hub genes (RGS7, EGFR, RGS4, GNA13 and RGS11) from the PPI network. PCA showed these genes could distinguish between AF and control samples, with 100 % of the area under curve (AUC) values. Silencing RGS7 inhibited cell apoptosis, inflammation and oxidative stress, and increased mitochondrial membrane potential in angiotensin II (AngII)-treated HL-1 cells, while overexpression of RGS7 reversed the inhibitory effects of silencing RGS7 on AF. Additionally, silencing RGS7 improved cardiac function and decreased cardiac fibrosis in AF rats. The cGMP-PKG signaling pathway was screened as a potential signal transduction pathway, and silencing RGS7 increased the expression of PKG1, while KT5823 blocked the process. CONCLUSION Silencing RGS7 attenuates AF by activating the cGMP-PKG signaling pathway, which may offer directions for selecting biomarkers and regulatory pathways for AF.
Collapse
Affiliation(s)
- Hao Huang
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu City, PR China
| | - Yan Xiong
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of, PR China
| | - Jie Zeng
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu City, PR China.
| |
Collapse
|
16
|
Bassett J, Balasubramanian B, Clouse H, Trepakova E. High content imaging of relative ATP levels for mitochondrial toxicity prediction in human induced pluripotent stem cell derived cardiomyocytes. Toxicology 2025; 514:154088. [PMID: 39971086 DOI: 10.1016/j.tox.2025.154088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are increasingly being evaluated in assays aimed at better understanding potential cardiotoxic liability of newly developed therapeutic compounds. Disruption of mitochondria has been implicated in the mechanism of drug-induced cardiotoxicity of some compounds. Therefore, we have developed a high content imaging assay for the investigation of mitochondrial toxicity in hiPSC-CMs using ATP-Red, a fluorescent dye capable of detecting subcellular localization of relative ATP levels in living cells. We demonstrated time-dependent decreases in ATP-Red signal over 6 h treatment with known mitochondrial toxicants antimycin (0.03, 0.1 µM) or oligomycin (3, 10 µM). Concentration-dependent decreases in ATP-Red signal with antimycin (0.001-0.3 µM) and oligomycin (0.003-1 µM) were rescued by restoring glycolysis through glucose supplementation. Decreased ATP levels were also identified in a selection of clinically available drugs with reported association with mitochondrial toxicity but absent in compounds with no known association with mitochondrial dysfunction. ATP measurements using the ATP-Red imaging assay were consistent with orthogonal measurements of whole cell ATP levels in lysed hiPSC-CMs following compound treatment. Similar findings were also obtained with measurement of mitochondrial membrane potential, except amiodarone which had no change despite decreased ATP levels. The developed high throughput imaging assay, assessing subcellular ATP dynamics, could provide mechanistic insights for tested compounds.
Collapse
|
17
|
Schnell A, Schwarz B, Schmidt H, Allabauer I, Schuh W, Regensburger AP, Rauh M, Woelfle J, Hoerning A. Adenosine-generating CD39 + plasmablasts predispose to successful infliximab therapy in pediatric IBD. Life Sci Alliance 2025; 8:e202403055. [PMID: 40199584 PMCID: PMC11979362 DOI: 10.26508/lsa.202403055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
B cells display several immunoregulatory mechanisms including the production of interleukin-10. Ectonucleotidases like CD39 and CD73 influence immune homeostasis by metabolizing eATP and generating immunosuppressive adenosine. The major objective was to examine the expression of those immunoregulatory molecules on B-cell subsets, and, more specifically, to determine their association with an infliximab (IFX) treatment in a pediatric inflammatory bowel disease (IBD) cohort. 42 IBD patients were assessed for IFX response after 12 mo of therapy and compared against 14 healthy controls (HC). Although IL10-producing plasmablasts were decreased in IFX nonresponders (NRS), we detected an up-regulation of CD39 on plasmablasts and increased fractions of CD39/CD73-co-expressing naïve and memory B cells in responding patients (RS). In addition, B cells of responders proved to have superior ATP degradation capacities and adenosine production before therapy initiation compared with NRS and HC. Moreover, IFX nonresponders had a marked deficiency of α4β7hi plasmablasts, whereas both cohorts had fewer CCR9-expressing plasmablasts. Consequently, CD39+ plasmablasts were decreased in biopsies of inflamed mucosal tissues, especially in IFX nonresponders. Our results highlight the regulatory potential of CD39/CD73-expressing B cells in pediatric IBD and suggest CD39+ plasmablasts as a potential determinant of a successful immunosuppressive therapy with IFX.
Collapse
Affiliation(s)
- Alexander Schnell
- Pediatric Gastroenterology and Hepatology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Benedikt Schwarz
- Pediatric Gastroenterology and Hepatology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Hannah Schmidt
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Ida Allabauer
- Pediatric Gastroenterology and Hepatology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang Schuh
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Adrian P Regensburger
- Pediatric Gastroenterology and Hepatology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - André Hoerning
- Pediatric Gastroenterology and Hepatology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
18
|
Aksu M, Kaschke K, Podojil JR, Chiang M, Steckler I, Bruce K, Cogswell AC, Schulz G, Kelly J, Wiseman RL, Miller S, Popko B, Chen Y. AA147 Alleviates Symptoms in a Mouse Model of Multiple Sclerosis by Reducing Oligodendrocyte Loss. Glia 2025; 73:1241-1257. [PMID: 39928347 PMCID: PMC12014361 DOI: 10.1002/glia.70001] [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: 07/25/2024] [Revised: 01/05/2025] [Accepted: 01/28/2025] [Indexed: 02/11/2025]
Abstract
Inflammation-induced oligodendrocyte death and CNS demyelination are key features of multiple sclerosis (MS). Inflammation-triggered endoplasmic reticulum (ER) stress and oxidative stress promote tissue damage in MS and in its preclinical animal model, experimental autoimmune encephalitis (EAE). Compound AA147 is a potent activator of the ATF6 signaling arm of the unfolded protein response (UPR) that can also induce antioxidant signaling through activation of the NRF2 pathway in neuronal cells. Previous work showed that AA147 protects multiple tissues against ischemia/reperfusion damage through ATF6 and/or NRF2 activation; however, its therapeutic potential in neuroinflammatory disorders remains unexplored. Here, we demonstrate that AA147 ameliorated the clinical symptoms of EAE and reduced ER stress, oligodendrocyte loss, and demyelination. Additionally, AA147 suppressed T cells in the CNS without altering the peripheral immune response. Importantly, AA147 significantly increased the expressions of Grp78, an ATF6 target gene, in oligodendrocytes, while enhancing levels of Grp78 as well as Ho-1, an NRF2 target gene, in microglia. In cultured oligodendrocytes, AA147 promoted nuclear translocation of ATF6, but not NRF2. Intriguingly, AA147 altered the microglia activation profile, possibly by triggering the NRF2 pathway. AA147 was not therapeutically beneficial during the acute EAE stage in mice lacking ATF6 in oligodendrocytes, indicating that protection primarily involves ATF6 activation in these cells. Overall, our results suggest AA147 as a potential therapeutic opportunity for MS by promoting oligodendrocyte survival and regulating microglia status through distinct mechanisms.
Collapse
Affiliation(s)
- Metin Aksu
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Kevin Kaschke
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Joseph R. Podojil
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - MingYi Chiang
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - Ian Steckler
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Kody Bruce
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Andrew C. Cogswell
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - Gwen Schulz
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Jeffery Kelly
- The Scripps Research Institute, Department of Chemistry, La Jolla, CA 92037, USA
| | - R. Luke Wiseman
- The Scripps Research Institute, Department of Molecular and Cellular Biology, La Jolla, CA 92037, USA
| | - Stephen Miller
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - Brian Popko
- Northwestern University, Department of Neurology, Chicago, IL 60611, USA
| | - Yanan Chen
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| |
Collapse
|
19
|
Zhang Q, Huang W, Li T, Wang X, Lai X, Hu W, Li Z, Zeng X, Huang J, Zhang R. Δ 9-tetrahydrocannabinol induces blood-brain barrier disruption: Involving the activation of CB1R and oxidative stress. Neuropharmacology 2025; 270:110366. [PMID: 39956317 DOI: 10.1016/j.neuropharm.2025.110366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/13/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Cannabis abuse has increased with the continuous relaxation of cannabis policies. However, the mechanism by which Δ9-tetrahydrocannabinol (THC) negatively affects the central nervous system, especially the blood-brain barrier (BBB), remains unclear. THC exposure models were established in vivo and in vitro. The BBB properties were examined using Western blotting (WB), immunofluorescence staining (IF), transendothelial electrical resistance (TEER), and flux of sodium fluorescein (SF). The oxidative stress regulators were examined using IF and assay kits. The activation of cannabinoid receptor 1 (CB1R) was examined using WB and IF. The THC exposure caused barrier integrity damage and endothelial dysfunction in murine and hCMEC/D3 cells, conclude albumin leakage, increased SF permeability and reduced TEER value. The expression of tight junction proteins, including claudin 5, occludin, and junctional adhesion molecules, was decreased. Additionally, key oxidative stress regulators, including reactive oxygen species, hydrogen peroxide, malonaldehyde levels, and antioxidant enzyme activities, including catalase, glutathione peroxidase, glutathione S-transferase, and superoxide dismutase, and heme oxygenase 1, were increased. Activation of CB1R has been detected in brain microvascular endothelial cells in vivo and in vitro. Furthermore, inhibition of oxidative stress and CB1R could mitigate the aforementioned conditions and BBB damage after THC exposure. The effect of THC on murine and human brain microvascular endothelial cells revealed that THC-induced BBB damage was partly mediated by CB1R activation, triggering the oxidative stress response. This study provides new theoretical insights into the mechanisms of THC-induced BBB damage and offers novel scientific evidence for the potential neurotoxicity and adverse reactions induced by THC.
Collapse
Affiliation(s)
- Qianyao Zhang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Wenxin Huang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Taokun Li
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Xuemei Wang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Ximin Lai
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Wei Hu
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Zhihong Li
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Xiaofeng Zeng
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Jian Huang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Ruilin Zhang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| |
Collapse
|
20
|
Yashaswini C, Kiran NS, Chatterjee A. Zebrafish navigating the metabolic maze: insights into human disease - assets, challenges and future implications. J Diabetes Metab Disord 2025; 24:3. [PMID: 39697864 PMCID: PMC11649609 DOI: 10.1007/s40200-024-01539-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/26/2024] [Indexed: 12/20/2024]
Abstract
Zebrafish (Danio rerio) have become indispensable models for advancing our understanding of multiple metabolic disorders such as obesity, diabetes mellitus, dyslipidemia, and metabolic syndrome. This review provides a comprehensive analysis of zebrafish as a powerful tool for dissecting the genetic and molecular mechanisms of these diseases, focusing on key genes, like pparγ, lepr, ins, and srebp. Zebrafish offer distinct advantages, including genetic tractability, optical transparency in early development, and the conservation of key metabolic pathways with humans. Studies have successfully used zebrafish to uncover conserved metabolic mechanisms, identify novel disease pathways, and facilitate high-throughput screening of potential therapeutic compounds. The review also highlights the novelty of using zebrafish to model multifactorial metabolic disorders, addressing challenges such as interspecies differences in metabolism and the complexity of human metabolic disease etiology. Moving forward, future research will benefit from integrating advanced omics technologies to map disease-specific molecular signatures, applying personalized medicine approaches to optimize treatments, and utilizing computational models to predict therapeutic outcomes. By embracing these innovative strategies, zebrafish research has the potential to revolutionize the diagnosis, treatment, and prevention of metabolic disorders, offering new avenues for translational applications. Continued interdisciplinary collaboration and investment in zebrafish-based studies will be crucial to fully harnessing their potential for advancing therapeutic development.
Collapse
Affiliation(s)
- Chandrashekar Yashaswini
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064 India
| | | | - Ankita Chatterjee
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064 India
| |
Collapse
|
21
|
Bounaama A, Djerdjouri B. Matrix metalloproteinase 9 implication during colorectal carcinogenesis. Effect of doxycycline. Fundam Clin Pharmacol 2025; 39:e70012. [PMID: 40273927 DOI: 10.1111/fcp.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/14/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Matrix metalloproteinases (MMPs), including MMP9, play a significant role in colorectal cancer (CRC) progression, mainly by extracellular matrix remodeling. However, little is known about MMP9 role in aberrant crypt foci (ACF) cluster formation, the earliest colon preneoplastic lesions. AIMS AND METHODS We conducted a bioinformatics analysis of MMPs expression in CRC using Gene Expression Profiling Interactive Analysis2 (GEPIA2). Subsequently, we investigated MMP9 expression during the early stage of colon carcinogenesis in mice and assessed the effect of doxycycline (DOX), a global inhibitor of MMPs, on ACF cluster formation. Thus, NMRI mice received two weekly injections of 1,2-Dimethylhydrazine (DMH, 20 mg/kg, subcutaneously), followed or not by DOX (100 mg/kg, orally, from the 4th to the 6th week). RESULTS GEPIA2 analysis indicated that among the 28 identified MMPs with collagenase and doxycycline-sensitive activities, MMPs 1, 3, 7, 9, and 13 were overexpressed in CRC tissues. Moreover, only MMP1 and MMP9 correlated well with collagen expression in colorectal tumors. In vivo, methylene blue-stained DMH-treated colons revealed multiple ACF clusters at week 6, associated with mucosa remodeling and sustained nitrosative stress as attested by enhanced collagen fibers, malondialdehyde level, and nitrotyrosine deposits. Pyrosequencing showed increased methylation at the tenth CpG site of the MMP9 promoter, which was associated with increased MMP9 expression. Interestingly, DOX attenuated the number and size of ACF clusters and mucosa remodeling without rebalancing nitrosative stress. CONCLUSION Overexpression of MMP9 occurs early during colorectal carcinogenesis, and doxycycline may control the pathological remodeling of colon mucosa into ACF clusters by attenuating MMP9 activity.
Collapse
Affiliation(s)
- Abdelkader Bounaama
- Tamayouz_Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria
| | - Bahia Djerdjouri
- Tamayouz_Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria
| |
Collapse
|
22
|
Ali A, Ali MA, Khattak AI, Khattak F, Afridi A, Azeem T, Shabbar Banatwala US, Alam U, Khan A, Jalal U, Moeez A, Khan MW, Collins P, Ahmed R. Outcomes of transcatheter vs surgical aortic valve replacement in pre-existing chronic liver disease patients: A meta-analysis of observational studies. IJC HEART & VASCULATURE 2025; 58:101651. [PMID: 40230501 PMCID: PMC11994331 DOI: 10.1016/j.ijcha.2025.101651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/16/2025]
Abstract
Aortic valve stenosis in patients with chronic liver diseases, particularly liver cirrhosis and End-Stage Liver Disease, poses significant management challenges due to the interplay between cardiovascular and hepatic dysfunction. This systematic review and meta-analysis compared the safety and efficacy of Transcatheter Aortic Valve Replacement (TAVR) and Surgical Aortic Valve Replacement in this high-risk population. An extensive search of PubMed, Embase, and Web of Science (inception to January 5, 2025) identified 11 retrospective studies comprising 19,097 patients. Risk ratios for dichotomous outcomes and mean differences (MD) for continuous outcomes, each with 95% confidence intervals, were calculated using random-effects models. The analysis revealed that TAVR significantly reduced hospital mortality (RR 0.36, 95 % CI: 0.30-0.42; I2 = 7.6 %), acute kidney injury (RR 0.51, 95 % CI: 0.33-0.78; I2 = 57.2 %), bleeding (RR 0.33, 95 % CI: 0.28-0.39; I2 = 0.0 %), stroke (RR 0.35, 95 % CI: 0.23-0.51; I2 = 6.1 %), and blood transfusion (RR 0.48, 95 % CI: 0.40-0.57; I2 = 7.6 %). TAVR was also associated with shorter hospital stays (MD -6.77 days, 95 % CI: -9.17 to -4.38; I2 = 97.5 %). No significant differences were observed in vascular complications requiring surgery or hospital charges and post-operative infections. These findings suggest TAVR offers significant advantages over SAVR in reducing complications such as mortality, acute kidney injury, and bleeding in patients with liver disease. However, further randomized trials are necessary to confirm long-term outcomes and establish optimal treatment strategies for this high-risk population.
Collapse
Affiliation(s)
- Aizaz Ali
- Khyber Medical College, Peshawar, Pakistan
| | | | | | | | | | | | | | - Umama Alam
- Khyber Medical College, Peshawar, Pakistan
| | | | | | | | | | - Peter Collins
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Raheel Ahmed
- National Heart and Lung Institute, Imperial College London, United Kingdom
| |
Collapse
|
23
|
Lee GL, Hu CC, Chen MF, Chang Y, Lin YC, Wu YY, Hsu YH. Inosine from purine metabolism enhances fracture healing by coupling fibrinolysis and angiogenesis of type H vessels. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167818. [PMID: 40157654 DOI: 10.1016/j.bbadis.2025.167818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Fibrinolysis-angiogenesis coupling is crucial for successful fracture healing, in which type H vessels play an indispensable role. However, the metabolic control of fibrinolysis-type H angiogenesis coupling in fracture healing remains unclear. We used a metabolomics approach to gain metabolic insights from mouse fracture models (0.3 mm and 1.0 mm femur defects). Furthermore, human umbilical vein endothelial cells (HUVECs) and MC3T3-E1 cells were employed as in vitro models to examine the effects of identified metabolites on endothelial events and osteogenesis, respectively. CD31 and endomucin (Emcn) were used for detecting type H markers, and tissue-type plasminogen activator (tPA) and plasminogen activator inhibitor-1 (PAI-1) for fibrinolysis. A femur defect of 1.5 mm in mice was included to validate the therapeutic potential of identified metabolites by bone imaging and micro-CT. Purine metabolism is the most significant pathway in fracture healing; three purinergic metabolites, adenosine, adenine, and inosine, are regulated in mouse serum. According to the in vivo results, CD31HiEmcnHi type H vessels are upregulated near the defect site in mouse femur and are associated with tPA expression, but not PAI-1. Additionally, in vitro experiments examining the endothelial functions of HUVECs demonstrated that these three metabolites promote cell migration and tube formation rather than proliferation. Fibrinolytic activity and type H phenotype in HUVECs were induced only by inosine through activation of the adenosine A2A receptor. Inosine, regulated during fracture healing, has the capacity to synchronously induce fibrinolysis and type H phenotype in line with osteogenesis, indicating its role in enhancing fracture healing.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chih-Chien Hu
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Mei-Feng Chen
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yuhan Chang
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Chih Lin
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ying-Yu Wu
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yung-Heng Hsu
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
24
|
Qi J, Lu B, Jin CW, Shang YY, Pan H, Li H, Tong ZJ, Zhang W, Han L, Zhong M. FP receptor inhibits autophagy to aggravate aging-related cardiac fibrosis through PI3K/AKT/mTOR signaling pathway. Arch Gerontol Geriatr 2025; 133:105824. [PMID: 40096796 DOI: 10.1016/j.archger.2025.105824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND F-prostanoid receptor (FP receptor), a receptor for Prostaglandin F2α(PGF2α), is involved in the process of tissue fibrosis, but its exact role in the aging heart remains unclear. METHODS We investigated cardiac function, myocardial fibrosis levels, autophagy levels and related mechanistic pathways in different groups of mice using gene silencing. At the cellular level, we simulated the senescence process of cardiac fibroblasts and investigated the related mechanisms using relevant inhibitors. RESULTS In aging mice, FP receptor and PI3K/AKT/mTOR pathways are increased and autophagy levels are decreased, ultimately leading to cardiac fibrosis. FP receptor gene silencing slows down the above process. We found similar changes at the cellular level. CONCLUSION FP receptor could activate PI3K/AKT/mTOR pathway and inhibit cardiac autophagy, which resulted in aging-related cardiac fibrosis. Thus, the inhibition of FP receptor could improve aging-related cardiac remodeling, implicating its potential therapeutic application to treat cardiovascular diseases associated with aging.
Collapse
Affiliation(s)
- Jia Qi
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China; Department of Cardiology, Zibo Central Hospital, Zibo, Shandong,255000, PR China
| | - Bin Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Cheng-Wei Jin
- Department of Cardiology, Zibo Central Hospital, Zibo, Shandong,255000, PR China
| | - Yuan-Yuan Shang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Hui Pan
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong key Laboratory of Cardiovascular Proteomics, Jinan, Shandong, PR China
| | - Hao Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Zhou-Jie Tong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Wei Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Lu Han
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China; Department of General Practice, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Ming Zhong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China.
| |
Collapse
|
25
|
Kang S, Li H, Li M, Zhao Y, Pang J, Dan J, Sheng M. Erianin alleviates doxorubicin-induced cardiotoxicity by activating the Keap1-Nrf2 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156684. [PMID: 40215822 DOI: 10.1016/j.phymed.2025.156684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Doxorubicin (DOX) has significant toxic side effects on cardiomyocytes, and existing preventive drug dexrazoxane has serious side effects. Therefore, in-depth research on drugs that can enhance the antitumor effect of DOX and simultaneously reduce its cardiotoxicity is of crucial significance. Our study explored the regulatory role of Erianin in DOX-induced cardiotoxicity and the specific molecular mechanism. METHODS In this study, we constructed a myocardial injury model in mice with DOX. The toxic side effects of DOX on the organism were determined by recording the weight changes of the mice and calculating the spleen index and heart-tibia ratio of the mice. The degree of myocardial injury in mice was evaluated by methods such as echocardiography and Sirius red staining. Further in vivo experiments were conducted to verify whether the silencing of Nrf2 could block the protective effect of Erianin on myocardial cells. RESULTS We found Erianin significantly alleviated DOX-induced cardiomyocyte injury (p < 0.0001), increased heart tissue pumping efficiency and contractility (p < 0.001), and reduced myocardial cell fibrosis. Mechanism study showed that Erianin can bind to Keap1, promote its ubiquitination and autophagic degradation, increase the acetylation of lysine 599 site in Nrf2 protein, and activate the antioxidant stress response. CONCLUSIONS Taken together, our study had for the first time elucidated the molecular mechanism by which Erianin alleviated DOX-induced myocardial injury by activating the Keap1-Nrf2 signaling pathway. It provides a theoretical basis for the development of Erianin as a potential protective drug for DOX-induced cardiotoxicity. It has very important clinical application and translational value.
Collapse
Affiliation(s)
- Shiyao Kang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Huimin Li
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Ming Li
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Yuan Zhao
- Department of Oncology, Puer People's Hospital, Puer, Yunnan, 665000, China
| | - Jianyu Pang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China.
| | - Miaomiao Sheng
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China.
| |
Collapse
|
26
|
Ramer R, Hinz B. Effect of cannabinoids on the efficacy and side effects of anticancer therapeutic strategies - Current status of preclinical and clinical research. Pharmacol Ther 2025; 270:108851. [PMID: 40221102 DOI: 10.1016/j.pharmthera.2025.108851] [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: 01/31/2025] [Revised: 03/14/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
Cannabinoids have attracted increasing attention in cancer research in recent decades. A major focus of current preclinical and clinical studies is on the interactions and potential risks when combined with chemotherapeutic agents, targeted therapies and other anticancer strategies. Given the extensive preclinical data on additive, synergistic and, in some cases, antagonistic tumor cell killing effects of chemotherapeutic agents and cannabinoids when co-administered, a critical analysis of these data seems essential. The available data mainly relate to combination treatments for glioblastoma, hematological malignancies and breast cancer, but also for other cancer types. Such an analysis also appears necessary because cannabinoids are used as an option to treat nausea and vomiting caused by chemotherapy, as well as tumor-related pain, and cancer patients sometimes take cannabinoids without a medical prescription. In addition, numerous recent preclinical studies also suggest cannabinoid-mediated relief of other chemotherapy-related side effects such as peripheral neuropathy, nephrotoxicity, cardiotoxicity, cystitis, bladder complications and mucositis. To summarize, the data available to date raise the prospect that cannabinoids may increase the efficacy of chemotherapeutic agents while reducing their side effects. However, preclinical studies on anticancer interactions are mostly limited to cytotoxicity analyses. An equally thorough investigation of the effects of such combinations on the immune system and on the tumorigenic levels of angiogenesis, invasion and metastasis is still pending. On this basis, a comprehensive understanding for the evaluation of a targeted additional treatment of various cancers with cannabinoids could be established.
Collapse
Affiliation(s)
- Robert Ramer
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany.
| |
Collapse
|
27
|
Kim M, Kim J, Tripathi SK, Aswar VR, Gaikwad V, Song J, Kim M, Yu J, Chang TS, Jeong LS. Steric influence of 4'-position substituents and C2-Hexynyl group on A 3AR antagonism in truncated 4'-Thioadenosine derivatives. Bioorg Chem 2025; 159:108359. [PMID: 40101576 DOI: 10.1016/j.bioorg.2025.108359] [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: 01/06/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
A3 adenosine receptor (A3AR) has attracted significant interest due to its therapeutic potential in inflammation, neurodegenerative disorders, and cancer. This study investigates the structure-activity relationships (SAR) of truncated 4'-thioadenosine derivatives with modifications at the 4'- and C2-positions. Alkyl substituents of varying sizes (methyl, ethyl, isopropyl) were introduced at the 4'-position to evaluate steric effects, while C2-propynyl and hexynyl groups were incorporated to explore binding enhancement. The 4'-ethyl derivatives exhibit potent A3AR antagonistic effects, compared to the 4'-methyl series which exhibits partial agonist activity. C2-hexynyl substitution significantly enhanced binding affinity and antagonistic properties. In contrast, 4'-isopropyl derivatives lacked measurable binding, highlighting steric constraints at 4'-position. Docking studies revealed that none of the compounds formed interactions with Thr94, a residue critical for agonistic activity. This lack of interaction likely explains their observed antagonistic behavior, with the 4'-alkyl and C2-hexynyl substitutions appearing to play a significant role in this effect. This work underscores the role of steric bulk at the 4'-position and hydrophobic modifications at the C2-position in modulating A3AR activity, providing valuable insights for designing selective A3AR antagonists.
Collapse
Affiliation(s)
- Minjae Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jina Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sushil K Tripathi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Vikas R Aswar
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Vidyasagar Gaikwad
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jiyoon Song
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Jinha Yu
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Tong-Shin Chang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Future Medicine Co., Ltd, 54 Changup-ro, Sujeong-gu, Seongnam, Gyeonggi-do 13449, Republic of Korea.
| |
Collapse
|
28
|
Yilmaz B, Genc GC, Celik SK, Cinar BP, Acikgoz M, Dursun A. PARP-1 gene promoter region may be associated with progression in multiple sclerosis. Clin Chim Acta 2025; 572:120275. [PMID: 40169083 DOI: 10.1016/j.cca.2025.120275] [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: 03/12/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
Multiple Sclerosis (MS) is a leading cause of disability among young adults. Most cases begin with relapsing-remitting MS (RRMS) and can transition to secondary progressive MS (SPMS) over time. It is known that the inflammatory status of the central nervous system changes during the progression of MS. Poly (ADP-ribose) polymerase-1 (PARP-1) is an enzyme involved in several cellular processes. Our study aimed to investigate the relationship between MS and the PARP-1 gene. We analyzed the PARP-1 gene's missense polymorphism rs1136410, promoter region polymorphism rs7527192, and 3'UTR polymorphism rs8679 in 123 MS patients and 168 healthy controls using the PCR-RFLP method. We examined genotype and allele frequency distributions among case-control groups and clinical subgroups. We observed that the CC genotype of rs7527192 polymorphism was increased in SPMS patients compared to controls. We also found that the CC genotype and C allele frequency were increased in the EDSS score > 3-6 group compared to healthy controls. The C allele frequency was increased in EDSS score > 3-6 compared to those with ≤ 3 and ≥ 6. When the results observed in our study are evaluated with the known effect of PARP-1 on the inflammasome pathway, we suggest that rs7527192 may be effective in the progression process through the activity of the PARP-1 inflammasome pathway.
Collapse
Affiliation(s)
- Busra Yilmaz
- Department of Medical Genetics, Zonguldak Bulent Ecevit University, Zonguldak, Turkey.
| | - Gunes Cakmak Genc
- Department of Medical Genetics, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Sevim Karakas Celik
- Department of Medical Genetics, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Bilge Piri Cinar
- Department of Neurology, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Mustafa Acikgoz
- Department of Neurology, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Ahmet Dursun
- Department of Medical Genetics, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
29
|
Yadav R, Baby K, Nayak Y, Patel D, Viswanathan K, Ghoshdastidar K, Patel A, Patel B. Unveiling the potential of tankyrase I inhibitors for the treatment of type 2 diabetes mellitus: A hybrid approach using network pharmacology, 2D structural similarity, molecular docking, MD simulation and in-vitro studies. Life Sci 2025; 369:123548. [PMID: 40058577 DOI: 10.1016/j.lfs.2025.123548] [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: 01/13/2025] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
AIMS This study explores the association between the Wnt signaling pathway and T2DM, emphasizing the role of Tankyrase1 (TNKS1) in metabolic regulation. Using network pharmacology and computational approaches, it aims to identify potential FDA-approved drugs for repurposing as Wnt inhibitors to improve insulin sensitivity and reduce fat accumulation. MATERIALS AND METHODS Network pharmacology analysis was performed to explore the association between the Wnt pathway and T2DM, identifying Catenin Beta 1 (CTNBB1) as a key hub gene involved in disease progression. A 2D structural similarity search was conducted using reference tankyrase inhibitors (E7449 and XAV939). Potential drug candidates were subjected to molecular docking and 100 ns molecular dynamics (MD) simulations with the Tankyrase I (PDB ID: 4W6E) protein. The shortlisted compounds were further evaluated for Wnt inhibitory activity using the TCF/LEF reporter assay, while their anti-diabetic potential was assessed through a glucose uptake assay in L6 myoblast cells. KEY FINDINGS Niclosamide, Capmatinib, Esomeprazole, and Fenofibrate were identified as promising candidates with strong binding affinities and stable interactions with key amino acids (Gly1185, Ser1221, Tyr1224, Asp1198, Tyr1213, and His1201). Experimental validation through in-vitro Wnt inhibition and glucose uptake assays confirmed that drugs Fenofibrate and Conivaptan exhibited significant Wnt inhibitory activity, suggesting their potential role in modulating T2DM-related pathways. SIGNIFICANCE This study highlights the role of the Wnt signaling pathway in T2DM pathogenesis and identifies potential drug candidates for repurposing as Tankyrase1/Wnt inhibitors. The findings provide a foundation for further in-vivo investigations into the anti-diabetic potential of the identified drugs, paving the way for novel therapeutic strategies in T2DM management.
Collapse
Affiliation(s)
- Ruchi Yadav
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India
| | - Krishnaprasad Baby
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Dhaval Patel
- Gujarat Biotechnology University, Gujarat International Finance Tec-City, Gandhinagar 382355, Gujarat, India
| | - Kasinath Viswanathan
- Zydus Research Centre, Cadila Healthcare Ltd., Sarkhej-Bavla N.H. no. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Krishnarup Ghoshdastidar
- Zydus Research Centre, Cadila Healthcare Ltd., Sarkhej-Bavla N.H. no. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Ankit Patel
- Zydus Research Centre, Cadila Healthcare Ltd., Sarkhej-Bavla N.H. no. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Bhumika Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India.
| |
Collapse
|
30
|
Xie J, Wu M, Li L, Zhu L, Hu L, Li Y, Li W. Integrated bioinformatics and experimental verification to dissect the mechanisms and bioactive ingredients of Radix Rehmanniae in treating multiple sclerosis. Biochem Biophys Res Commun 2025; 763:151790. [PMID: 40233432 DOI: 10.1016/j.bbrc.2025.151790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
Multiple sclerosis (MS), as a primary cause of nontraumatic disability in young adults, has no effective treatment yet. Radix Rehmanniae (RR), a typical Traditional Chinese Medicine (TCM), is commonly used in MS patients as a most frequent herbal item in TCM formulas. Our recent study demonstrated that RR alleviated neurological deficits in an experimental MS model. However, direct evidence regarding the holistic mechanisms and bioactive components of RR for MS remains unclear. In this study, we employed an integrative strategy combining bioinformatics and experimental validation to profile the holistic mechanisms of RR, identify its bioactive components, and investigate their potential targets in MS. First, a network pharmacology approach was used to construct a "compound-target-pathway" network, indicating the action of RR on MS in a multicomponent-multitarget mode, and predicting Echinacoside and Acteoside as the primary bioactive ingredients. Bioinformatics analyses of transcriptomics and single-cell RNA sequencing based on GSE datasets indicated that oxidative stress and inflammatory/immune regulation in microglia might serve as crucial mechanisms of Echinacoside and Acteoside in MS pathology. Then, in vitro assays validated that Echinacoside and Acteoside possessed anti-inflammatory and antioxidant properties by scavenging ONOO- and H2O2 directly, and suppressing microglia-derived ONOO- production through inhibition of NF-κB-mediated iNOS and NADPH oxidase. In addition, molecular docking showed strong affinities between Acteoside and inflammation-related targets TGF-β and SMAD2. These findings provide the scientific evidence for clinical application of RR and bring novel insights into MS drug development.
Collapse
Affiliation(s)
- Jing Xie
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Li Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Lixia Zhu
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Liang Hu
- School of Integrative Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yuzhen Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Wenting Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|
31
|
Li X, Li M, Chen R, Wang Y, Luo G, Gao X. Affinity-purified targets screening facilitates active components discovery of Chinese formula -HuGan tablets as a case. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119703. [PMID: 40188898 DOI: 10.1016/j.jep.2025.119703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alcoholic Liver Disease (ALD), a chronic condition caused by long-term heavy alcohol consumption, can progress to cirrhosis or liver failure. HuGan Tablets (HGT) is a compound preparation made of six Chinese herbs, which is used in clinic for the treatment of chronic hepatitis, with studies demonstrating its efficacy in alleviating alcohol-induced liver injury in rats. However, the active components and therapeutic targets of HGT remain unclear and require further investigation. AIM OF THIS STUDY The aim of this study was to develop a systematic pipeline based on the SPR fishing strategy to identify effective components and therapeutic targets in Chinese formulas, using HGT as a representative case. MATERIALS AND METHODS HRMS was employed to analyze HGT ingredients absorbed in rat blood, while network pharmacology, molecular docking and literature mining were utilized to identify potential targets of HGT for ALD alleviation. A systematic SPR-based fishing system was developed by evaluating protein target coupling efficiency, sample recovery rate, specificity of target-small molecule binding, and LOD, and candidate components screened and identified using this system were further screened by SPR affinity tests. Additionally, therapeutic efficacy of the selected compounds was validated in vitro using an ethanol-induced AML12 model and further confirmed in vivo using a mouse model of ALD by assessing markers such as ALT, AST, and oxidative stress indicators. RESULTS A total of 128 compounds were identified in HGT, with 29 metabolites detected in rat blood. MFN2, SOD2, mTOR, RXRA, and GSTP1 were identified as anti-ALD targets of HGT through integrated network pharmacology, molecular docking, and literature analysis. An SPR-based active component fishing system was successfully developed, capturing 15 candidate compounds. SPR affinity analysis revealed strong binding (KD: 3.41-221.7 μM) between (R,S)-goitrin, chlorogenic acid, saikosaponin B2, schisandrin, schisandrol B, schisandrin A, schisandrin C, and schisantherin A and the target proteins. Except for (R,S)-goitrin, the other seven compounds significantly reduced ALT, AST, TG, ROS, and MDA levels while enhancing SOD and GSH activities in cellular models, with comparable therapeutic effects observed in ALD mice. CONCLUSION This study scientifically established an integrated SPR-based pipeline to systematically characterize active ingredients and therapeutic targets in herbal formulations, which was successfully applied to reveal key therapeutic targets and pharmacodynamic components of HGT for ALD. This study provides a valuable framework for SPR-based screening of bioactive components in traditional formulas, as well as for understanding the material basis and mechanism of action of HGT in the treatment of ALD.
Collapse
Affiliation(s)
- XueJiao Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Miao Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - RuiShu Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Ying Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Gan Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - XiaoYan Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
32
|
Gammal A, Nassar T, Soae Y, Freeman N, Badihi A, Baraghithy S, Nemirovski A, Tam J, Benita S. Synthesis and Pharmacological Characterization of Novel Peripheral Cannabinoid-1 Receptor Blockers Based on a Tricyclic Scaffold. J Med Chem 2025; 68:9431-9445. [PMID: 40258217 DOI: 10.1021/acs.jmedchem.4c03132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
The development of peripherally selective cannabinoid-1 receptor (CB1R) antagonists offers a promising strategy for obesity treatment. Here, we evaluated the efficacy of novel tricyclic CB1R antagonists, focusing on BNS808. Our findings demonstrate that BNS808 exhibits robust CB1R antagonism with notable CB2R selectivity, minimal brain penetration, and potent in vitro and in vivo efficacy. The compound's high plasma protein binding reduces free drug availability for CNS entry, enhancing safety and minimizing drug-drug interactions. In diet-induced obese mice, BNS808 effectively reduced body weight, adiposity, liver triglycerides, and liver enzymes, supporting its peripherally mediated action. These results highlight BNS808 as a promising candidate for obesity treatment. Additionally, our novel library of peripherally selective CB1R antagonists provides a strong foundation for future drug development. With further refinement, BNS808 holds significant clinical potential to address the global obesity epidemic.
Collapse
Affiliation(s)
- Asaad Gammal
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
- Laboratory of Nano Delivery Systems, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Taher Nassar
- Laboratory of Nano Delivery Systems, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Yael Soae
- BioNanoSim (BNS), Hadassah Ein Kerem Campus, Minrav Building (JBP), Jerusalem 9112101, Israel
| | - Noam Freeman
- BioNanoSim (BNS), Hadassah Ein Kerem Campus, Minrav Building (JBP), Jerusalem 9112101, Israel
| | - Amit Badihi
- BioNanoSim (BNS), Hadassah Ein Kerem Campus, Minrav Building (JBP), Jerusalem 9112101, Israel
| | - Saja Baraghithy
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Alina Nemirovski
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Joseph Tam
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Simon Benita
- Laboratory of Nano Delivery Systems, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| |
Collapse
|
33
|
Guo Y, Huang H, Zhang Q, Wang H, Liu M, Lin W. A novel dual-channel fluorescent probe for the detection of peroxynitrite anions and lipid droplets in epileptic disease. Anal Chim Acta 2025; 1350:343863. [PMID: 40155169 DOI: 10.1016/j.aca.2025.343863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 04/01/2025]
Abstract
Peroxynitrite (ONOO-) and lipid droplets (LDs) are crucial substances essential for maintaining normal physiological functions in biological systems. They play pivotal roles as biomarkers in the initiation and progression of various diseases, such as epilepsy. Therefore, the simultaneous detection of ONOO- and LDs in epilepsy disorders is of great importance. Here, we discovered that the fluorescence probe composed of trifluoromesulfonate and fluorophore can not only be used as the recognition site of ONOO-, but also has the property of LDs targeting. Therefore, we reasonable designed and synthesized a dual-channel fluorescent probe CBT, capable of simultaneously monitoring ONOO- and LDs. CBT exhibited exceptional dual-response properties: firstly, upon specific reaction with ONOO-, the resulting product BHD emitted a robust red fluorescent signal in the near-infrared region (749 nm); secondly, CBT selectively targeted and labeled LDs, emitting green fluorescence at 482 nm for effective LDs tracking. The signals from these two detection channels did not overlap, which significantly enhanced the accuracy and reliability of detection. Based on these characteristics, CBT has been successfully utilized in real-time imaging of ONOO- and LDs in epilepsy models of cells induced by various drugs. Notably, in a pentylenetetrazole (PTZ)-induced chronic epileptic mice model, CBT exhibited excellent efficacy in ONOO- imaging, further confirming its considerable potential for practical applications. In summary, this study validated CBT as an efficient tool capable of simultaneous detection and differentiation of ONOO- and LDs, presenting a novel and promising strategy for the early diagnosis and treatment of diseases such as epilepsy.
Collapse
Affiliation(s)
- Yingxin Guo
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Huawei Huang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Qian Zhang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Hongjian Wang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Miaomiao Liu
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Weiying Lin
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China.
| |
Collapse
|
34
|
Potdar MB, Bhamare RP, Agrawal YO, Belgamwar AV. Crosstalk Between Signaling Stroke Cascade and Therapeutic Receptors PPAR-γ, ROCK, CB1R, and CB2R: From Mechanism to Therapies. Transl Stroke Res 2025:10.1007/s12975-025-01352-2. [PMID: 40338418 DOI: 10.1007/s12975-025-01352-2] [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: 12/14/2024] [Revised: 02/07/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025]
Abstract
Stroke remains a leading cause of disability and mortality worldwide, primarily due to the complex and multifaceted nature of its pathophysiology. This review aims to provide a comprehensive and mechanistic understanding of the crosstalk between key signaling pathways activated during stroke and the therapeutic potential of specific receptors: PPAR-γ, ROCK, CB1R, and CB2R. We delve into the intricate signaling cascades that occur post-stroke, including excitotoxicity, oxidative stress, and inflammation, highlighting the pivotal molecular players involved. PPAR-γ, known for its neuroprotective and anti-inflammatory properties, emerges as a critical modulator in stroke therapy. ROCK, a central component in the Rho/ROCK pathway, is implicated in vascular and neuronal damage, making its inhibition a promising therapeutic strategy. The roles of CB1R and CB2R within the endocannabinoid system are explored, with a focus on their dualistic nature in neuroprotection and neurotoxicity. The review further examines the interconnectivity of these receptors within the stroke signaling network, proposing that their synergistic modulation could enhance therapeutic outcomes. Current therapeutic approaches, including pharmacological and multi-target strategies, are critically evaluated, addressing the challenges in translating mechanistic insights into clinical practice. Additionally, the identification and utilization of biomarkers for stroke diagnosis and therapy monitoring are discussed, offering a glimpse into future prospects. Emerging therapies, novel drug developments, and personalized medicine approaches are presented as potential game-changers in stroke treatment.
Collapse
Affiliation(s)
- Mrugendra B Potdar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Rohit P Bhamare
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Yogeeta O Agrawal
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Aarti V Belgamwar
- Department of Pharmaceutics, Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India.
| |
Collapse
|
35
|
Zaparte A, Cruz FF, de Souza JB, Morrone FB. P2 receptors signaling in the esophagus: from inflammation to cancer. Purinergic Signal 2025:10.1007/s11302-025-10089-4. [PMID: 40338451 DOI: 10.1007/s11302-025-10089-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
The signaling mechanisms of nucleotides and nucleosides have been extensively studied over the past decades in various conditions affecting distinct organs and tissues. It is well-established that purinergic receptors are expressed in healthy tissues, with expression levels often increasing under pathological conditions. These receptors play crucial roles in numerous physiological and pathological processes, including inflammation, tissue repair, and cellular signaling. However, the purinergic context in the esophagus and its associated pathologies remains poorly understood, representing a significant gap in current knowledge. In this review, we compiled and analyzed the available data on the involvement of P2 purinergic receptors in esophageal diseases, such as gastroesophageal reflux disease and esophageal carcinoma. Specifically, we discuss the pharmacological modulation, functional characterization, and expression patterns of these receptors in various esophageal cell lines and immune tissue samples, under both healthy and pathological conditions. Understanding the mechanisms of action and signaling pathways involving P2 purinergic receptors in the esophagus can offer valuable insights into their biological roles and emphasize their potential as therapeutic targets for future clinical applications.
Collapse
Affiliation(s)
- Aline Zaparte
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6690, 90619 - 900, Porto Alegre, RS, Brasil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
| | - Fernanda F Cruz
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
| | - Julia B de Souza
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6690, 90619 - 900, Porto Alegre, RS, Brasil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
| | - Fernanda B Morrone
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6690, 90619 - 900, Porto Alegre, RS, Brasil.
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil.
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil.
| |
Collapse
|
36
|
Tyler J, Park Y, Lu H, Roeder N, Richardson B, Gold MS, Blum K, Pinhasov A, Baron D, Thanos PK. High-intensity interval training exercise decreases brain CB1 receptor levels in male and female adult rats. Neuroscience 2025; 573:254-263. [PMID: 40122443 DOI: 10.1016/j.neuroscience.2025.03.038] [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: 11/08/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
The numerous health benefits of exercise are well-documented, including its efficacy in treating substance use disorders (SUDs). Several exercise regimens have been proposed; however, the most effective regimen for patients with addiction has yet to be elucidated. High-intensity interval training (HIIT) exhibits considerable potential compared to aerobic and resistance exercise. Dopamine signaling is recognized as a key neurobiological mechanism contributing to HIIT's therapeutic potential for SUDs; however, the role of the endocannabinoid system in this context is not well understood. The present study investigated the effects of HIIT exercise on endocannabinoid signaling by measuring cannabinoid receptor 1 (CB1R) binding in the brains of male and female rats using [3H]SR141716A autoradiography. Male and female rats were separated into sedentary and HIIT exercise groups. For six weeks, exercise was completed daily on a treadmill for 30 min (10 3-min intervals) progressively increasing speed to 0.8 mph (21.5 m/min). The HIIT program significantly reduced CB1R binding in both sexes across multiple brain regions, including the striatum, thalamus, and distinct areas of the cortex. Sex differences were observed wherein males exhibited greater CB1R binding than females across brain regions, including the cerebellum, striatum, and parts of the cortex. Males experienced an increase in mean cerebellum CB1R binding due to HIIT, whereas females showed no effect in this region. The results suggest HIIT exercise can modulate endocannabinoid signalling by way of decreased CB1R binding. These findings further support the intensity dependence of endocannabinoid modulation and highlight potential pathways for exercise-induced neurobiological and behavioural change.
Collapse
Affiliation(s)
- John Tyler
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
| | - Youmin Park
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Huy Lu
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Nicole Roeder
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA
| | - Brittany Richardson
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Mark S Gold
- Department of Psychiatry, Washington University in St. Louis Euclid Ave, St. Louis, MO 63110, USA
| | - Kenneth Blum
- Division of Addiction Research and Education, Center for Sports, Exercise, and Mental Health, Western University Health Sciences, Pomona, CA 91766, USA; Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - David Baron
- Division of Addiction Research and Education, Center for Sports, Exercise, and Mental Health, Western University Health Sciences, Pomona, CA 91766, USA; Department of Psychiatry, Stanford University School of Medicine, Palo Alto, CA. 94305-5101 USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
37
|
Mastyugin M, Vlocskó RB, Zsengellér ZK, Török B, Török M. Development of Diaryl Hydrazones for Alleviation of Mitochondrial Oxidative Stress in Preeclampsia. J Med Chem 2025. [PMID: 40333076 DOI: 10.1021/acs.jmedchem.5c00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Preeclampsia is a pregnancy-specific syndrome, linked to oxidative stress, affecting 5-8% of pregnancies, with no effective treatment available. Here, diaryl-hydrazones have been designed, synthesized, and investigated as mitochondria-targeting antioxidants to reduce placental oxidative stress and mitigate preeclampsia symptoms. The design, based on density functional theory studies, revealed that conjugated electron structure with the NH-motif appeared to explain their effect. Thirty compounds were synthesized and tested in three assays, where they exhibited excellent radical scavenging activity, significantly greater than that of the standard, Trolox. Based on the data, eight compounds were selected for cell-based assays. Oxidative stress was induced in human trophoblast cells and assessed whether the compounds reduced downstream antiangiogenic responses using ascorbic acid and MitoTEMPO as standards. The pretreatment with the hydrazones reduced mitochondrial superoxide and sFLT-1 production in H2O2-exposed trophoblast cells, indicating that mitochondrial oxidative stress and the anti-angiogenic response can be alleviated by these compounds.
Collapse
Affiliation(s)
- Maxim Mastyugin
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd., Boston, Massachusetts 02125, United States
| | - R Bernadett Vlocskó
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd., Boston, Massachusetts 02125, United States
| | - Zsuzsanna K Zsengellér
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, United States
- Department of Medicine,Harvard Medical School,Boston,Massachusetts02215,United States
| | - Béla Török
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd., Boston, Massachusetts 02125, United States
| | - Marianna Török
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd., Boston, Massachusetts 02125, United States
| |
Collapse
|
38
|
Szmek J, Englmaierová M, Skřivan M, Pěchoučková E. Skeletal disorders in laying hens: a systematic review with a focus on non-cage housing systems and hemp-based dietary interventions for bone health. Br Poult Sci 2025:1-30. [PMID: 40331968 DOI: 10.1080/00071668.2025.2489059] [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] [Accepted: 03/17/2025] [Indexed: 05/08/2025]
Abstract
1. The poultry sector is possibly the fastest growing and most flexible of all livestock sectors. At present, the main changes to the table egg production system include the gradual abandonment and closure of all cage-housing systems for laying hens, driven by animal welfare concerns and stricter legislation in many countries. In the future, keeping hens in enriched cage systems may be restricted or phased out in response to evolving animal welfare guidelines and public demand. To meet the welfare and behavioural requirements of the hens, it is desirable to choose housing on litter or housing in aviaries as a substitute for housing in enriched cages.2. The objective of this systematic review was to examine non-cage housing systems and hemp-based dietary interventions in relation to skeletal health in laying hens. This review focussed on the risks associated with alternative housing systems, particularly the increased incidence of bone fractures and the potential of nutritional strategies to mitigate skeletal disorders, including osteoporosis.3. The proportion of hens housed in non-cage alternative housing systems is currently increasing sharply but carries certain risks. One of the most significant concerns is skeletal integrity, as hens in aviaries experience a higher rate of keel bone fractures due to collisions, falls and deviations thought to be related to internal pressure. Numerous studies have shown that the incidence of keel bone damage (i.e. fractures and deviations) was greater in aviaries compared to enriched cage systems.4. Optimal skeletal health can be supported through proper nutrition, which plays a crucial role in bone metabolism. Key nutritional elements, including calcium, vitamins D, E and K, polyunsaturated fatty acids and hemp-based products, have been shown to be beneficial in preventing skeletal disorders and associated fractures due to their specific roles in maintaining bone structure and strength.
Collapse
Affiliation(s)
- J Szmek
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague - Suchdol, Czech Republic
- Department of Nutrition Physiology and Animal Product Quality, Institute of Animal Science, Prague - Uhříněves, Czech Republic
| | - M Englmaierová
- Department of Nutrition Physiology and Animal Product Quality, Institute of Animal Science, Prague - Uhříněves, Czech Republic
| | - M Skřivan
- Department of Nutrition Physiology and Animal Product Quality, Institute of Animal Science, Prague - Uhříněves, Czech Republic
| | - E Pěchoučková
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague - Suchdol, Czech Republic
- Department of Nutrition Physiology and Animal Product Quality, Institute of Animal Science, Prague - Uhříněves, Czech Republic
| |
Collapse
|
39
|
Wei Q, Bai Z, Wang L, Wang J, Wang Y, Hu Y, Ding S, Ma Z, Li C, Li Y, Zhuo Y, Li W, Deng F, Liu B, Zhou P, Li Y, Wu Z, Wang J. A high-performance fluorescent sensor spatiotemporally reveals cell-type specific regulation of intracellular adenosine in vivo. Nat Commun 2025; 16:4245. [PMID: 40335490 DOI: 10.1038/s41467-025-59530-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
Adenosine (Ado), a nucleoside bridging intracellular metabolism with intercellular communication, plays an essential role in regulating processes such as sleep and seizure. While the functions of extracellular Ado ("eAdo") are well documented, our knowledge about the distribution and regulatory functions of intracellular Ado ("iAdo") is limited by a lack of methods for detecting iAdo in vivo. Here, we develop HypnoS, a genetically encoded fluorescent sensor for iAdo characterized by its high sensitivity, specificity, spatiotemporal resolution, and rapid response (sub-seconds). HypnoS enables real-time visualization of iAdo dynamics in live cultures, acute brain slices, flies, and freely moving mice. Using HypnoS for dual-color mesoscopic imaging in mice, we show that seizure-induced iAdo waves propagated across the cortex, following calcium signals. Additionally, two-photon imaging reveals that iAdo decays more rapidly in astrocytes than in neurons during seizures. Moreover, by recording iAdo dynamics in the basal forebrain during the sleep-wake cycle, we observe that iAdo signals are present during wakefulness and rapid eye movement (REM) sleep, regulated by equilibrative nucleoside transporters (ENT1/2). Thus, HypnoS is a versatile and powerful tool for investigating the biological functions of iAdo across a range of physiological and pathological states.
Collapse
Affiliation(s)
- Qingpeng Wei
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zexiao Bai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lei Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Jing Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yipan Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Yufei Hu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Shiyi Ding
- University of Chinese Academy of Sciences, Beijing, China
- Interdisciplinary Center for Brain Information, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhixiong Ma
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chun Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yumo Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yizhou Zhuo
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Fei Deng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Bingjie Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Pengcheng Zhou
- Interdisciplinary Center for Brain Information, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
- National Biomedical Imaging Center, Peking University, Beijing, China.
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- National Biomedical Imaging Center, Peking University, Beijing, China.
| |
Collapse
|
40
|
Karp F, León IE. Cannabinoids: Adaptogens or Not? Cannabis Cannabinoid Res 2025. [PMID: 40332769 DOI: 10.1089/can.2024.0108] [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: 05/08/2025] Open
Abstract
Since ancient times, humanity has been exploring natural substances with the aim of increasing stress resistance, enhancing biochemical homeostasis, and treating different diseases. In this way, the objective of the present review is to compare the biological effects of cannabinoids (CNBs) with adaptogens, this exploration allows us to consider the controversy if they can be classified together considering the effects on the body. First, the work revises different features of adaptogens such as their chemical structure, ligand-receptors properties, and homeostasis-stress capabilities. Also, this review includes an overview of preclinical and clinical studies of the effect of adaptogens considering a broad spectrum of adverse biological, chemical, and physical factors. Then, the work does a review of the CNBs effects on the body including the principal uses for the treatment of several diseases as neurodegenerative disorders, arthritis, cancer, cardiovascular affections, diabetes, anxiety, chronic pain, among others. In addition, the different characteristics of the specific endocannabinoid system are described explaining the wide CNBs body effects. Finally, this review presents a comparative analysis between CNBs and adaptogens properties, expecting to contribute to understanding if CNBs can be classified as adaptogens.
Collapse
Affiliation(s)
- Federico Karp
- Laboratorio de Nanobiomateriales, CINDEFI, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata and CONICET, La Plata, Argentina
| | - Ignacio E León
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
- Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
41
|
Jiang J, Gao Y, Wang J, Huang Y, Yang R, Zhang Y, Ma Y, Wen Y, Luo G, Zhang S, Cao Y, Yu M, Wang Q, Hu S, Wang K, Guo X, Gonzalez FJ, Liu Y, Liu H, Xie Q, Xie C. Hepatic sphingomyelin phosphodiesterase 3 promotes steatohepatitis by disrupting membrane sphingolipid metabolism. Cell Metab 2025; 37:1119-1136.e13. [PMID: 40015281 DOI: 10.1016/j.cmet.2025.01.016] [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: 07/15/2024] [Revised: 12/16/2024] [Accepted: 01/17/2025] [Indexed: 03/01/2025]
Abstract
Metabolic-dysfunction-associated steatohepatitis (MASH) remains a major health challenge. Herein, we identify sphingomyelin phosphodiesterase 3 (SMPD3) as a key driver of hepatic ceramide accumulation through increasing sphingomyelin hydrolysis at the cell membrane. Hepatocyte-specific Smpd3 gene disruption or pharmacological inhibition of SMPD3 alleviates MASH, whereas reintroducing SMPD3 reverses the resolution of MASH. Although healthy livers express low-level SMPD3, lipotoxicity-induced DNA damage suppresses sirtuin 1 (SIRT1), triggering an upregulation of SMPD3 during MASH. This disrupts membrane sphingomyelin-ceramide balance and promotes disease progression by enhancing caveolae-dependent lipid uptake and extracellular vesicle secretion from steatotic hepatocytes to exacerbate inflammation and fibrosis. Consequently, SMPD3 acts as a central hub integrating key MASH hallmarks. Notably, we discovered a bifunctional agent that simultaneously activates SIRT1 and inhibits SMPD3, which shows significant therapeutic potential in MASH treatment. These findings suggest that inhibition of hepatic SMPD3 restores membrane sphingolipid metabolism and holds great promise for developing novel MASH therapies.
Collapse
Affiliation(s)
- Jie Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yuqing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Lingang Laboratory, Shanghai 200444, China
| | - Yan Huang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Rong Yang
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yongxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuandi Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingquan Wen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Gongkai Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shurui Zhang
- Lingang Laboratory, Shanghai 200444, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yutang Cao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Minjun Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinxue Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Shulei Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kanglong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Frank J Gonzalez
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yameng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China.
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China.
| |
Collapse
|
42
|
Kieronska-Rudek A, Zuhra K, Ascenção K, Chlopicki S, Szabo C. The PARP inhibitor olaparib promotes senescence in murine macrophages. GeroScience 2025:10.1007/s11357-025-01679-6. [PMID: 40327273 DOI: 10.1007/s11357-025-01679-6] [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: 04/01/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
Cellular senescence is a multifaceted process involving cell cycle arrest, telomere shortening, and the accumulation of DNA damage associated with aging and cellular stress. It is marked by persistent cell cycle arrest and DNA damage accumulation, and plays an increasingly recognized role in age-related diseases and cancer therapy. Olaparib, a poly (ADP-ribose) polymerase (PARP) inhibitor, is approved for use in ovarian cancer treatment. We hypothesized that olaparib may influence senescence by inhibiting DNA damage repair, and investigated its effects on non-senescent and replicatively senescent murine macrophages (RAW 264.7 cells). Senescent cells exhibited elevated baseline levels of PARP1 expression, PARylation, and DNA damage relative to non-senescent control cells. Olaparib amplified these differences by upregulating senescence markers (SA-β-gal and p21), inhibiting proliferation, and exacerbating DNA damage. Many of its effects were more pronounced in senescent cells. At higher concentrations (10-30 µM), olaparib induced significant cytotoxicity through mixed apoptotic and necrotic mechanisms, with senescent cells exhibiting a predominantly necrotic response. Interestingly, both mitochondrial activity and cellular bioenergetics were elevated in senescent cells at baseline, and were more severely impaired by olaparib compared to non-senescent control cells. These findings underscore olaparib's enhanced cytotoxic and pro-senescent effects in senescent immune cells and suggest potential implications for its use in elderly cancer patients with an increased burden of senescent cells.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Karim Zuhra
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Kelly Ascenção
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Csaba Szabo
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
43
|
Wei X, Fan X, Chai W, Xiao J, Zhao J, He A, Tang X, Li F, Guo S. Dietary limonin ameliorates heart failure with preserved ejection fraction by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis: an integrated transcriptomics and metabolomics analysis. Food Funct 2025; 16:3553-3574. [PMID: 40230319 DOI: 10.1039/d5fo00475f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome characterized by hypertension, metabolic disorders, and impaired diastolic function, with limited therapeutic options. Recent studies have highlighted the role of ferroptosis in the pathogenesis of HFpEF, and the inhibition of ferroptosis occurrence can significantly improve cardiac function. Limonin, a bioactive ingredient derived from citrus fruits, has been confirmed to exert potential anti-inflammatory and antioxidant effects in some cardiovascular diseases. This study aims to investigate the therapeutic effects of limonin on HFpEF and the underlying mechanisms of inhibiting ferroptosis. HFpEF mice were established by a combination of Nω-nitro-L-arginine methyl ester and a high-fat diet for 6 weeks. Subsequently, the HFpEF mice were treated with empagliflozin or limonin via oral gavage for an additional 6 weeks. Limonin curbed body weight gain and improved metabolic disorders and hypertension. Limonin also ameliorated concentric cardiac hypertrophy and diastolic dysfunction. Transcriptomics and metabolomics analyses revealed that limonin regulated ferroptosis-related pathways and lipid peroxidation. In vivo, limonin improved mitochondrial morphology, reduced cardiac Fe2+ levels and ferroptosis markers such as ROS, 4-HNE and MDA, and increased GSH levels, thereby enhancing antioxidant capacity. Mechanistically, limonin regulated the P53/SLC7A11/GPX4 signaling pathway, promoted the nuclear translocation of Nrf2 (its upstream signaling molecule), and subsequently activated its downstream antioxidant elements, ultimately inhibiting ferroptosis. Furthermore, limonin decreased the expressions of ACSL4, COX2, and ALOXs, which reduced the accumulation of lipid peroxides. These results demonstrate that limonin ameliorates HFpEF by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis, providing a novel strategy for HFpEF treatment.
Collapse
Affiliation(s)
- Xiaoqi Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xinyi Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Wangjing Chai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinling Xiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Jiong Zhao
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Aolong He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xianwen Tang
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Fanghe Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
44
|
Montuoro S, Gentile F, Giannoni A. Neuroimmune cross-talk in heart failure. Cardiovasc Res 2025; 121:550-567. [PMID: 39498795 DOI: 10.1093/cvr/cvae236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Heart failure (HF) is characterized by autonomic nervous system (ANS) imbalance and low-grade chronic inflammation. The bidirectional relationship between the ANS and immune system (IS) is named 'neuroimmune cross-talk' (NICT) and is based on common signaling molecules, receptors, and pathways. NICT may be altered in HF, and neuroinflammation seems to be a main driver of HF progression. In HF, heightened sympathetic nerve activity triggers inflammatory cascades that lead to cardiomyocyte death and myocardial interstitial fibrosis. Concurrently, parasympathetic withdrawal may impair the cholinergic anti-inflammatory pathway, with a less effective immune response to infections or inflammatory events. Additionally, microglial activation and inflammatory molecules contribute to autonomic imbalance by acting on central nuclei and peripheral visceral feedbacks, which in turn promote adverse cardiac remodeling, HF decompensation, and potentially life-threatening arrhythmias. Therefore, neuroinflammation has been identified as a potential target for treatment. Pharmacological antagonism of the neurohormonal system remains the cornerstone of chronic HF therapy. While some drugs used in HF management may have additional benefits due to their anti-inflammatory properties, clinical trials targeting inflammation in patients with HF have so far produced inconclusive results. Nevertheless, considering the pathophysiological relevance of NICT, its modulation seems an appealing strategy to optimize HF management. Current research is therefore investigating novel pharmacological targets for anti-inflammatory drugs, and the immunomodulatory properties of denervation approaches and bioelectronic medicine devices targeting NICT and neuroinflammation in HF. A deeper understanding of the complex relationship between the ANS and IS, as outlined in this review, could therefore facilitate the design of future studies aimed at improving outcomes by targeting NICT in patients with HF.
Collapse
Affiliation(s)
- Sabrina Montuoro
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, 56127 Pisa, Italy
| | - Francesco Gentile
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, 56127 Pisa, Italy
- Cardiovascular Medicine Division, Fondazione Toscana G. Monasterio, Via Moruzzi 1, 56126 Pisa, Italy
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà, 33, 56127 Pisa, Italy
- Cardiovascular Medicine Division, Fondazione Toscana G. Monasterio, Via Moruzzi 1, 56126 Pisa, Italy
| |
Collapse
|
45
|
Chen W, Zhuang A, Liu C, He Y, Kaixin Lu, Jiang T, Zhang H, Gao R, Xue X. Mitochondrial Enzyme HIBADH Protects Against Calcium Oxalate Nephrolithiasis by Modulating Oxidative Stress and Apoptosis. Arch Biochem Biophys 2025:110452. [PMID: 40334962 DOI: 10.1016/j.abb.2025.110452] [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: 11/14/2024] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/09/2025]
Abstract
Calcium oxalate (CaOx) nephrolithiasis, as one of the most common types of kidney stones, poses a major threat to human health. This study aimed to investigate the role of 3-hydroxyisobutyrate dehydrogenase (HIBADH) in the pathogenesis of CaOx nephrolithiasis. CaOx nephrolithiasis models were established in rats via 1% ethylene glycol and 2% ammonium chloride induction and in HK-2 cells using calcium oxalate monohydrate (COM, 100 μg/mL). HIBADH expression was modulated through plasmid transfection and siRNA knockdown in vitro, and AAV2/9-mediated gene transfer in vivo. Multiple parameters were assessed, including cell crystal adhesion, apoptosis, cell cycle distribution, oxidative stress markers (SOD, MDA, MitoSOX fluorescence), and mitochondrial function (ATP level, mitochondrial membrane potential), using various techniques such as crystal adhesion assay, flow cytometry, western blot, qRT-PCR, and fluorescence microscopy. Kidney tissues were analyzed through H&E, Von Kossa, and PAS staining. Results demonstrated that HIBADH expression was significantly downregulated in CaOx nephrolithiasis rats and COM-treated HK-2 cells. In vitro, HIBADH overexpression reduced cell crystal adhesion and apoptosis, promoted cell cycle progression, mitigated mitochondria-involved cellular oxidative stress, and enhanced mitochondrial function in COM-induced HK-2 cells. In vivo, AAV2/9-mediated HIBADH overexpression attenuated crystal deposits and tubular injury, reduced apoptosis, and mitigated mitochondria-involved cellular oxidative stress in kidney tissues. The mitochondria-targeted antioxidant Mito-TEMPO counteracted the effects of HIBADH silencing, highlighting the role of mitochondrial function in HIBADH's protective mechanism. This study identifies HIBADH as a critical regulator in CaOx nephrolithiasis, exerting its protective effects through modulation of mitochondrial function and mitochondria-involved cellular oxidative stress, cell crystal adhesion, and apoptosis. Our findings elucidate the link between mitochondrial metabolism and kidney stone formation, positioning HIBADH as a promising therapeutic target for CaOx nephrolithiasis.
Collapse
Affiliation(s)
- Wenwei Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Anni Zhuang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Changyi Liu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Yanfeng He
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Kaixin Lu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Tao Jiang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Hua Zhang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Rui Gao
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Xueyi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
46
|
Wang L, Geng Y, Liu L, Wang J, Chen J, Li Y, Wang J, Song L, Sun K, Yan Y, Zhou S, Tian D, Lin R, Yao H. Synthesis, anti-allergic rhinitis evaluation and mechanism investigation of novel 1,2,4-triazole-enamides as CB1 R antagonist. Eur J Med Chem 2025; 289:117461. [PMID: 40048796 DOI: 10.1016/j.ejmech.2025.117461] [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: 01/12/2025] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
Allergic rhinitis (AR) is a non-infectious inflammatory disease and affects nearly half of the world's population currently, thus becoming a global health problem. In our study, a series of 1,2,4-triazole enamides were designed and used to evaluate the anti-inflammatory activity of AR. We found that compound 11g could significantly reduce the increased expression of interleukin-6 (IL-6), interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in Raw264.7 cells induced by lipopolysaccharides (LPS), and inhibit the expression of inflammation through MAPK pathway and NF-κB pathway by influencing the expression of cannabinoid-1 receptor (CB1 R). In the AR mice model, 11g can significantly reduce the number of inflammatory cells in Nasal lavage fluids (NLF), showing a good effect on the treatment of AR. This study provides a new and effective candidate for treatment of AR.
Collapse
Affiliation(s)
- Lu Wang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Otorhinolaryngology Head and Neck Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Yuexiu District, Dade Road111, Guangzhou, Guangdong, China
| | - Yan Geng
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Lifang Liu
- Department of Biopharmaceutical Sciences, College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China; Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Jun Wang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Jiaxin Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Yunying Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Otorhinolaryngology Head and Neck Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Yuexiu District, Dade Road111, Guangzhou, Guangdong, China
| | - Jingbo Wang
- Department of Biopharmaceutical Sciences, College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Liyan Song
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Kexin Sun
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Yajie Yan
- The Second Clinical College of Guangzhou University of Chinese Medicine, Otorhinolaryngology Head and Neck Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Yuexiu District, Dade Road111, Guangzhou, Guangdong, China
| | - Shiqing Zhou
- The Second Clinical College of Guangzhou University of Chinese Medicine, Otorhinolaryngology Head and Neck Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Yuexiu District, Dade Road111, Guangzhou, Guangdong, China
| | - Dan Tian
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Ran Lin
- Department of Biopharmaceutical Sciences, College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China.
| | - Hongliang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
47
|
Tak KY, Kim J, Park M, Kim W, Lee S, Park N, Kim MJ, Kang JB, Koh Y, Yang HY, Yum MK, Kim I, Yang YR, Jeong WI, Yang J, Lee C, Kim C, Park JE. Quasi-spatial single-cell transcriptome based on physical tissue properties defines early aging associated niche in liver. NATURE AGING 2025:10.1038/s43587-025-00857-7. [PMID: 40325195 DOI: 10.1038/s43587-025-00857-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 03/27/2025] [Indexed: 05/07/2025]
Abstract
Aging is associated with the accumulation of senescent cells, which are triggered by tissue injury response and often escape clearance by the immune system. The specific traits and diversity of these cells in aged tissues, along with their effects on the tissue microenvironment, remain largely unexplored. Despite the advances in single-cell and spatial omics technologies to understand complex tissue architecture, senescent cell populations are often neglected in general analysis pipelines due to their scarcity and the technical bias in current omics toolkits. Here we used the physical properties of tissue to enrich the age-associated fibrotic niche and subjected them to single-cell RNA sequencing and single-nuclei ATAC sequencing (ATAC-seq) analysis and named this method fibrotic niche enrichment sequencing (FiNi-seq). Fibrotic niche of the tissue was selectively enriched based on its resistance to enzymatic digestion, enabling quasi-spatial analysis. We profiled young and old livers of male mice using FiNi-seq, discovered Wif1- and Smoc1-producing mesenchymal cell populations showing senescent phenotypes, and investigated the early immune responses within this fibrotic niche. Finally, FiNi-ATAC-seq revealed age-associated epigenetic changes enriched in fibrotic niche cells. Thus, our quasi-spatial, single-cell profiling method allows the detailed analysis of initial aging microenvironments, providing potential therapeutic targets for aging prevention.
Collapse
Affiliation(s)
- Kwon Yong Tak
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Juyeon Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Myungsun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Wooseok Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seoyeong Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Narae Park
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Min Jeong Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ju-Bin Kang
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Yongjun Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hae Young Yang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Min Kyu Yum
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Injune Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- BioMedical Research Center, KAIST, Daejeon, Republic of Korea
| | - Yong Ryoul Yang
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Won-Il Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jinsung Yang
- Department of Biochemistry and Convergence Medical Science, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Cheolju Lee
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Chuna Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.
- Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
- BioMedical Research Center, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
48
|
Huang Z, Zhou J, Liu S, Zhang Y, Meng J, Zhu X, Du Y. The interplay between systemic inflammation and myopia: A bidirectional Mendelian randomization and experimental validation study. Int Immunopharmacol 2025; 157:114803. [PMID: 40327989 DOI: 10.1016/j.intimp.2025.114803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
PURPOSE Though the pathogenesis of myopia remains unclear, emerging evidence suggests a potential link between the onset of myopia and systemic inflammation. This study aims to elucidate the causal relationships between the two via Mendelian randomization (MR). METHODS We utilized genome-wide association study data on circulating inflammatory proteins (n = 14,824), immune cell traits (n = 3757), and myopia (n cases = 4106, n controls = 394,028) for a standard two-sample bidirectional MR analysis, followed by sensitivity analyses employing diverse approaches. The validation of seven inflammatory molecules was conducted through ELISA analysis of 116 plasma samples from a hospital-based cohort, as well as proteomics data from 3310 participants in the UK Biobank cohort. RESULTS Our analysis identified three inflammatory proteins (CXCL9, CXCL11, and T cell surface glycoprotein CD5) and six immune phenotypes, primarily related to T cells, as risk factors for myopia, and IL-5 and eight traits as protective factors. Meanwhile, we observed that myopia may elevate the levels of two inflammatory agents (TNFRSF9 and IL-24) and 12 peripheral immunophenotypes, predominantly associated with T cells and monocytes. Validation analysis in two independent cohorts further corroborated the proinflammatory state in highly myopic patients manifested by significantly elevated plasma levels of CXCL9, CXCL11, and TNFRSF9. CONCLUSIONS Our study identified a potential bidirectional causal relationship between systemic immune dynamics and myopia, underscoring the importance of considering myopia in the context of systemic condition. Research is warranted to further identify underlying mechanisms.
Collapse
Affiliation(s)
- Zhiqian Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Jitong Zhou
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Shuyu Liu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Ye Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Jiaqi Meng
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Xiangjia Zhu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Yu Du
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| |
Collapse
|
49
|
Ayerra L, Abellanas MA, Vidaurre C, Basurco L, Tavira A, Luquin E, Clavero P, Mengual E, Collantes M, Peñuelas I, de Martin-Esteban SR, Grether U, Hillard CJ, Romero J, Hervás-Stubbs S, Aymerich MS. Activation of central cannabinoid type 2 receptors, but not on peripheral immune cells, is required for endocannabinoid-mediated neuroprotection in Parkinson's disease. Brain Behav Immun 2025; 128:600-611. [PMID: 40320016 DOI: 10.1016/j.bbi.2025.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/03/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025] Open
Abstract
Neuroinflammation is a key feature of Parkinson's disease (PD). The cannabinoid receptor type 2 (CB2R) is expressed by cells of the innate and adaptive immune systems. Inhibition of monoacylglycerol lipase (MAGL) with JZL184 increases the levels of the endocannabinoid 2-arachidonoylglycerol (2-AG), which is neuroprotective for dopaminergic neurons. The aim of this study was to determine whether the neuroprotective effect of MAGL inhibition is mediated by CB2R activation on specific immune cell populations. Experimental parkinsonism was induced by chronic administration of MPTP and probenecid. A specific increase in CD4+ T cell infiltration was detected in the midbrain of parkinsonian mice and was reduced by administration of JZL184. JZL184 had no effect in CB2R KO mice, suggesting that CB2R is required for neuroprotection. In the brain, CB2R expression was restricted to myeloid cells and lymphocytes, and increased in microglia under parkinsonian conditions. Administration of a central CB2R agonist, JWH133, exerted a beneficial effect similar to that of JZL184, whereas the peripheral agonist RO304 lacked neuroprotective activity. These results were confirmed using chimeric mice. In silico analysis, showed that transcripts related to 2-AG biosynthesis are downregulated in the midbrain microglia from PD patients. Our results show that activation of CB2R in the brain prevents nigrostriatal degeneration, CD4+ T cell infiltration and TNFα production in the midbrain of parkinsonian mice. The reduced 2-AG signaling in microglia from PD patients suggests that activation of microglial CB2R may be an interesting strategy for the treatment of PD.
Collapse
Affiliation(s)
- Leyre Ayerra
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; CIMA-Universidad de Navarra, Pamplona, Spain
| | - Miguel Angel Abellanas
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; CIMA-Universidad de Navarra, Pamplona, Spain
| | - Clara Vidaurre
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; CIMA-Universidad de Navarra, Pamplona, Spain
| | - Leyre Basurco
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; CIMA-Universidad de Navarra, Pamplona, Spain
| | - Adriana Tavira
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; CIMA-Universidad de Navarra, Pamplona, Spain
| | - Esther Luquin
- Universidad de Navarra, Facultad de Medicina, Departamento de Patología, Anatomía y Fisiología, Pamplona, Spain
| | - Pedro Clavero
- Servicio de Neurología, Hospital Universitario de Navarra, Pamplona, Spain
| | - Elisa Mengual
- Universidad de Navarra, Facultad de Medicina, Departamento de Patología, Anatomía y Fisiología, Pamplona, Spain
| | - Maria Collantes
- Unidad de Imagen Molecular Traslacional (UNIMTRA), Departamento de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Ivan Peñuelas
- Unidad de Imagen Molecular Traslacional (UNIMTRA), Departamento de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Samuel Ruiz de Martin-Esteban
- Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Uwe Grether
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Julian Romero
- Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Sandra Hervás-Stubbs
- CIMA-Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Maria S Aymerich
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; CIMA-Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.
| |
Collapse
|
50
|
Wang J, Zhang R, Xie H, Yang Y, Chen H, Lin Q. Metal-phenolic epigallocatechin gallate‑zinc antioxidant nanoparticles for cataract treatment. J Control Release 2025; 383:113798. [PMID: 40319915 DOI: 10.1016/j.jconrel.2025.113798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
In cataract, oxidative stress plays an important role. It breaks the balance of the antioxidant defense system by destroying biological macromolecules such as proteins, lipids and nucleic acids in the lens, induces lens opacity, and then leads to the formation of cataract. The antioxidant properties of epigallocatechin gallate (EGCG) are well-established, particularly its ability to scavenge ROS and modulate cellular pathways. However, its therapeutic potential is limited by poor stability and bioavailability. To overcome these challenges, the work is designed to form a metal-phenolic network (MPN) system by coordinating EGCG with zinc ions. MPNs offer distinct advantages for ocular drug delivery, including enhanced structural stability, improved cellular uptake, and the ability to simultaneously target multiple oxidative stress pathways. By doing so, an antioxidant nanoparticle is formed to slow down the turbidity process of the lens induced by oxidative stress and thus prevent the occurrence of cataract. The particle size, zeta potential, ultraviolet absorption spectrum, FTIR, surface morphology and element distribution of EGCG-Zn were determined. In vitro, EGCG-Zn has good biosafety, cell uptake performance and anti-cell damage performance. In addition, from a mechanistic analysis, the particles can scavenge free radicals and reduce the production of intracellular ROS. Similarly, EGCG-Zn can well prevent the damage of reactive oxygen species to intracellular lysosomes, mitochondria, cytoskeleton, DNA synthesis and cell senescence. In the UV-B-induced cataract animal model in rats, EGCG-Zn has good in vivo safety. The transparency of the lens in the experimental group using EGCG-Zn is significantly lower than that in the untreated model group. In conclusion, EGCG-Zn nanoparticles are expected to become an important means in the field of cataract treatment, bringing hope for restoring clear vision to many cataract patients.
Collapse
Affiliation(s)
- Jiahao Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Renjie Zhang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hongying Xie
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yuexin Yang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hao Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| | - Quankui Lin
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|